Ethods IN Olecular Iology
Ethods IN Olecular Iology
Series Editor
                     John M. Walker
                 School of Life Sciences
               University of Hertfordshire
          Hatfield, Hertfordshire, AL10 9AB, UK
Edited by
                              Frank T. Christiansen
    Department of Clinical Immunology, PathWest Laboratory Medicine, Royal Perth Hospital;
School of Pathology and Laboratory Medicine, University of Western Australia, Perth, WA, Australia
                                      Brian D. Tait
 National Transplant Services, Australian Red Cross Blood Service, The Royal Melbourne Hospital,
                                    Melbourne, VIC, Australia
Editors
Frank T. Christiansen                                           Brian D. Tait
Department of Clinical Immunology                               National Transplant Services
PathWest Laboratory Medicine                                    Australian Red Cross Blood Service
Royal Perth Hospital                                            The Royal Melbourne Hospital
Perth, WA, Australia                                            Melbourne, VIC, Australia
School of Pathology and Laboratory
Medicine, University of Western Australia
Perth, WA, Australia
The ability of mice to distinguish “self” from “nonself” and recognize foreign organisms or
tissues (an immune response) was found to be under genetic control by the seminal experi-
ments of Peter Gorer in 1936 and his description of the H-2 system. This was later extended
to the human with the discovery of the HLA system by Jean Dausset in 1958. Since that
time the study and knowledge of the genes controlling such immune responses in the
human has undergone a rapid expansion. These studies have been facilitated by the devel-
opment of a range of molecular DNA techniques and a remarkable degree of international
collaboration.
     The HLA molecules, encoded by a highly polymorphic set of genes located with the
major histocompatibility complex (MHC) on Chromosome 6, are important regulators of
the immune response through mediating antigen presentation and interaction between key
immune mediating cells. They are also the major histocompatibility barriers to transplanta-
tion, which is the clinical paradigm of the self vs. nonself concept. Within the MHC, one of
the best characterized regions of the human genome, are a number of other gene clusters
involved in controlling aspects of the immune response. These encode various accessory
molecules such as TNF, components of the classical complement pathway, and heat shock
proteins. In addition outside the MHC, many other genes encode components which can
act as histocompatibility antigens or are involved in the function of the immune system.
These include genes encoding polymorphic tissue-specific peptides presented within the
context of HLA molecules which have been shown to give rise to a series of minor histo-
compatibility antigens which are barriers to allogeneic transplantation but which can also
be exploited as targets for immunotherapy. Those polymorphic genes encoding the immu-
noglobulin and the T cell receptor molecules, important mediators of humoral and cell-
mediated adaptive immune responses, also play a role in regulation of immune responses.
The immune response is also mediated by a complex of cytokines and cell receptors and
polymorphism in the structural and regulatory elements of the genes have been demon-
strated and shown to have functional correlates. More recently a cluster of genes encoding
receptors on NK cells, including the killer immunoglobulin-like receptors (KIR), which
regulate NK cell activation largely through interaction with ligands encoded within the
MHC, have been described and been shown to be important determinants in transplanta-
tion outcome and in conferring disease susceptibility.
     It is now recognized that this diverse range of gene systems involved in the control of
the immune response has been shown to be important in many aspects of clinical practice.
As a result many new molecular and cellular methods have been developed for identifying
these genes and their polymorphisms, and immunogenetic laboratories specializing in these
methods have developed to support transplantation and other clinical programs. This volume
focuses on such methods. The scope is exclusively for human clinical practice and the
emphasis is on those assays which are of established or potential clinical utility and are likely
to be included in the repertoire of tests provided by a routine diagnostic and service
                                               v
vi       Preface
laboratory. In the tradition of Methods in Molecular Biology series, the methods provide
details of the materials and equipment required and a step-by-step description of the labo-
ratory method. Details of the critical factors in the performance and interpretation of these
assays and other practical tips are provided through as series of Notes which we trust the
reader will find particularly helpful.
     The characterization of the HLA and other genes within the MHC is important to the
routine diagnostic immunogenetics laboratory, particularly in supporting solid organ and
hemopoietic stem cell transplant programs. We have included chapters describing HLA typ-
ing by molecular techniques including sequence-specific oligonucleotide hybridization (SSO)
and sequence-specific priming (SSP) methods, solid phase bead-based assays and by direct
DNA sequencing, and the use of specialist software for HLA allele assignment. An additional
chapter focuses on a number of molecular methods adapted for the typing of specific HLA
alleles which are of diagnostic utility in disease diagnosis and in drug hypersensitivity reac-
tions. In addition to typing of the classical HLA molecules, we have included chapters describ-
ing typing of other MHC genes including the nonclassical HLA molecules HLA-E and
HLA-G, the complement component C4, the MICA genes, and a panel of single nucleotide
polymorphisms (SNPs) throughout the MHC. Methods for detecting the many non-MHC
minor histocompatibility antigens which have been described and for detecting the function-
ally important polymorphisms in a range of cytokine genes are also included.
     The detection and characterization of HLA antibodies directed at epitopes found on
donor HLA molecules and the more recently described antibodies directed at MICA
epitopes is known to be important in organ transplantation, and the recent development of
solid phase bead-based assays to detect these antibodies has led to a new understanding of
the role of HLA and MICA antibodies in organ transplant rejection. Chapters describing
the methods for the detection of donor-specific antibodies using the traditional, but still
gold standard, complement dependent cytotoxicity and the more sensitive flow cytometry-
based crossmatches and bead-based antibody detection assays are included. Assays for the
detection of T cell-mediated reactivity against alloantigens either directly or indirectly
through the cross-reactivity of viral-specific T cells—so-called heterologous immunity—are
also described.
     The highly polymorphic KIR gene cluster, which encodes a family of NK cell receptors
which are important in controlling NK cell function, has generated considerable interest
recently. Methods detecting and characterizing the polymorphism of this complex gene
system including PCR-SSP and direct sequencing and for detecting NK cell alloreactivity
including a flow cytometry-based assay are described. Alloreactive NK cells have the poten-
tial to be effective therapeutic agents, and a detailed method for the clinical production of
such cells is provided.
     In addition to the method chapters we have included a series of overview chapters
highlighting aspects of gene function or various methods available for their study. These
include reviews of the MHC complex, the KIR complex, the human immunoglobulin allo-
types, as well as reviews of the methods for the detection of alloreactive NK cells and the
detection of HLA antibodies by solid phase assays. Because of their expertise and under-
standing of the complexity of the HLA system, many immunogenetics laboratories provide
specialist advice to clinicians in their search for suitable unrelated hemopoietic stem cell
donors. We have therefore included a chapter which provides a detailed and practical guide
to cost-effective strategies for undertaking such searches.
     Immunogenetic data is complex and the genes are highly polymorphic and have been
extensively studied. These features have meant that extensive databases of such genes and
                                                                              Preface       vii
the polymorphisms are now available. Included are chapters on methods for the establish-
ment and management of immunogenetic databases and which describe the use of specialist
HLA, immunoglobulin, and T cell receptor polymorphism databases. In addition two
chapters are included which review the analytical methods available for the study and mea-
surement of human population diversity and the identification, quantitation, and mapping
of disease susceptibility genes respectively. These chapters also provide a number of worked
examples and we trust the readers will find them particularly helpful.
     We would like to thank all the contributors to what we believe is an outstanding collec-
tion of manuscripts which we are sure will be widely read and used by the immunogenetics
community. These contributions represent many hours of work and the sharing of detailed
methods and helpful tips gained by many years of experience in the field. We are delighted
that they have been prepared to share this information with the wider scientific community.
     We would also like to thank John Walker and Humana Press for inviting us to edit this
volume of the very successful Methods in Molecular Biology series and for his excellent edito-
rial guidance. We trust the final product has rewarded his trust in us.
     Finally, we would like to express our special thanks to Natalie Caldwell for her outstand-
ing clerical and editorial assistance. Her enthusiasm and skill has been invaluable as we have
worked through the numerous editorial tasks.
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .     v
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .      xiii
                                                                     ix
x          Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .   681
wwwwwwwwwwwww
Contributors
                                              xiii
xiv     Contributors
STEVEN J. MACK • Center for Genetics, Children’s Hospital and Research Center Oakland,
   Oakland, CA, USA
MARI MALKKI • Fred Hutchinson Cancer Research Centre, Seattle, WA, USA
O. PATRICIA MARTINEZ • Department of Clinical Immunology, PathWest Laboratory
   Medicine, Royal Perth and Fremantle Hospitals, Perth, WA, Australia;
   School of Pathology and Laboratory Medicine (PaLM), University of
   Western Australia, Nedlands, WA, Australia
JAMES MCCLUSKEY • Department of Microbiology and Immunology, University
   of Melbourne, Parkville, VIC, Australia
DAVID H. MCKENNA • Department of Laboratory Medicine and Pathology,
   Cancer Centre and Molecular and Cellular Therapeutics, University of Minnesota,
   Minneapolis, MN, USA
NARINDER MEHRA • Department of Transplant Immunology and Immunogenetics,
   All India Institute of Medical Sciences, New Delhi, India
NICOLE A. MIFSUD • Department of Medicine, Monash University, Alfred Hospital,
   Melbourne, VIC, Australia
JEFFREY S. MILLER • Department of Medicine, University of Minnesota Cancer Centre,
   Minneapolis, MN, USA
MANUELA MORARU • Inmunogenética—HLA, Hospital Universitario Puerta de Hierro,
   Majadahonda, Spain
PHILIPPE MOREAU • Commissariat à l’Energie Atomique, Service de Recherches en Hémato-
   Immunologie, I2BM, Institut Universitaire d’Hématologie, Hôpital Saint-Louis,
   Paris, France
JENNIFER NG • Department of Pediatrics, CW Bill Young Marrow Donor Recruitment
   and Research Program, Georgetown University Medical Centre, Washington, DC, USA
HOLGER NÜCKEL • Department of Haematology and Institute of Pharmacogenetics,
   University Hospital Essen, Essen, Germany
CRISTA OCHSENFARTH • Institute of Pharmacogenetics, University Hospital Essen,
   Essen, Germany
GERHARD OPELZ • Department of Transplantation Immunology, Institute of Immunology,
   University of Heidelberg, Heidelberg, Germany
DAVID ORDÓÑEZ • Inmunogenética—HLA, Hospital Universitario Puerta de Hierro,
   Majadahonda, Spain
MACHTELD OUDSHOORN • Department of Immunohematology and Blood Transfusion,
   Leiden University Medical Centre, Leiden, The Netherlands; Europdonor Foundation,
   Leiden, The Netherlands
EFFIE W. PETERSDORF • Fred Hutchinson Cancer Research Centre, Seattle, WA, USA
RAJA RAJALINGAM • UCLA Immunogenetics Center, Department of Pathology
   and Laboratory Medicine, David Geffen School of Medicine at UCLA,
   University of California, Los Angeles, CA, USA
VERA REBMANN • Institute for Transfusion Medicine, University Hospital Essen, Essen,
   Germany
DAVE L. ROELEN • Department of Immunohematology and Blood Transfusion,
   Leiden University Medical Centre, Leiden, The Netherlands
PÄIVI SAAVALAINEN • Research Program for Molecular Medicine, Department of Medical
   Genetics, Haartman Institute, Biomedicum Helsinki, University of Helsinki,
   Helsinki, Finland
DAVID SAYER • Conexio Pty Ltd, Fremantle, WA, Australia
xvi     Contributors
Abstract
The major histocompatibility complex (MHC) on chromosome 6 is one of the most intensively studied
regions of the human genome and has many features which make it unique. It is the source of much
research interest because of its role in autoimmune and infectious disease susceptibility, and of diagnostic
interest because of its role in transplantation and rejection. It is the most gene-dense and SNP-rich region
of the genome, with large number of complex haplotypes and other features which must be taken into
account when analysing the MHC in the laboratory. This article provides a brief overview of the MHC
highlighting some of the issues that must be considered when developing new methods and assays.
Key words: Major histocompatibility complex, Genetic polymorphism, HLA, Immune genes
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_1, © Springer Science+Business Media New York 2012
                                                           1
2   R.J.N. Allcock
2. Physical
Structure and
Gene Content
                The MHC lies on the short arm of chromosome 6, with the class
                I region at the telomeric end and the class II region at the centro-
                meric end (reviewed in ref. (2)). Between them lies the class III, or
                central MHC, region of approximately 1 Mb. The class II region is
                relatively homogeneous, containing genes predominantly encod-
                ing HLA class II proteins (alpha and beta subunits), as well as a
                small number of genes involved in processing antigens for class I
                proteins (the TAP and PSMB genes). In contrast, the class I region
                encodes the classical (HLA-A, -B, -C) and non-classical (HLA-E,
                -F, -G) proteins, but is littered with the remnants of many evolu-
                tionary relics in the form of pseudogenes. In addition, the class I
                region encodes a significant number of Tripartite-motif (TRIM)
                family and butyrophilin (BTN) family genes, which may well have
                immune-related roles, and a large number of genes with functions
                outside the immune system.
                     The gene families found in the MHC are likely the result of
                multiple duplication events and individual pairs of genes can be
                almost identical (e.g. HLA-B and HLA-C), or highly similar to
                one another (e.g. the MIC family for MHC-class I-like genes—see
                ref. (8)) or much more divergent, sharing little more than broad
                intron–exon structures and having 20–25% sequence identity (e.g.
                TNF, LTA, and LTB—see ref. (9)).
                     The class I and II regions of the MHC are so named primar-
                ily because of the HLA genes encoded within them. Between
                them lies the class III region which has no such defining feature
                and contains a large number of genes of variable function, both
4      R.J.N. Allcock
                        within and outside the immune system. Some areas have been
                        extensively studied (e.g. the complement genes, the TNF cluster
                        and MICA genes), whilst others have been less well studied. The
                        1 Mb class III region has the highest gene density of the three
                        major MHC regions.
3. Genetic
Variation and
Haplotypes
                        The MHC has long been known to harbour large numbers of
                        SNPs, particularly in the MHC class I and II genes. However, it is
                        now recognised that almost all MHC genes are more polymorphic
                        than most genes elsewhere in the genome (2). These variations
                        take the form of point mutations, small indels, large indels, and
                        microsatellites. Within the HLA genes, particularly in the exons
                        encoding the hypervariable regions, there may be 50–100 SNPs
                        located within regions of 100–200 bp, many of which cause amino
                        acid changes (5). Primer-design for PCR is fraught with problems
                        in these regions and therefore many investigators site primers in
                        introns or further afield where there are lower levels of variation.
                        This problem is not limited to the HLA genes however, as variants
                        are found throughout the MHC, sometimes in very close proxim-
                        ity to one another.
                             Initially, only a single MHC sequence, assembled from a vari-
                        ety of sources and individuals, was available (3) and the identification
                        of sequence variations was based around “sample” sequencing of
                        small regions of interest to particular investigators (e.g. see ref.
                        (10)). Subsequently, the “MHC haplotype project” used standard
                        mapping and sequencing techniques to resequence the entire
                        MHC from eight hemi- or homozygous cells lines carrying well-
                        defined and important haplotypes (11). The vast number of cata-
                        logued MHC variants generated by this project has allowed a
                        greater insight into the existence of haplotypes blocks, both within
                        the MHC, but also elsewhere (see refs. (5, 6)). The level of knowl-
                        edge of block boundaries and number of the haplotypes within a
                        given block is far greater in the MHC than elsewhere in the
                        genome, largely as a consequence of the major interest in the
                        region and the large number of variants identified. Whilst particu-
                        lar whole MHC haplotypes based on HLA class I and II combina-
                        tions have been recognised for more than 20 years (12), it is now
                        recognised that much smaller haplotype blocks of some 20–60 kb
                        in length are present. The well-studied TNF block contains more
                        than 100 SNPs but it can be defined by 20–30 SNPs (13), and the
                        SNPs present in each haplotype appear to be found in many popu-
                        lations worldwide, and in similar combinations. Whilst populations
                        differ in the frequency of individual haplotypes, the fundamental
                        observation of a common set of haplotypes remains (14).
                                    1   The Major Histocompatibility Complex…       5
4. Assay Design
and Approaches
                  To date, diagnostic assays have focussed on the amplification of
                  small fragments and either hybridisation to labelled probes or
                  sequencing using standard methods, such as fluorescent dideoxy
                  chain terminators (Sanger sequencing). Diagnostic assays for other
                  parts of the MHC have not been developed to the same extent.
                  Assays for the detection of MICA polymorphisms have been devel-
                  oped and are described in this volume. Studies of the complement
                  region, of interest to some groups, are complicated by a duplicated
                  gene system in which there can be 0–4 genes present on each hap-
                  lotype, all of which may be polymorphic (15). The recent significant
                  interest in copy number variation has led to the development of
                  methods to examine them effectively, and methods incorporating
                  both CNV and SNP analysis are emerging. A similar situation exists
                  at the telomeric end of the HLA class II region, where there is a
                  variable gene content in the HLA-DRB pseudogenes, which once
                  again can contain multiple alleles (16).
                       Arguably, the most complex bioinformatic analysis currently in
                  use in the diagnostic world is the assignment of a pair of HLA
                  alleles to a given individual. Currently, based on Sanger sequencing
                  of the peptide binding-encoding exons, bulk heterozygous
                  sequence is compared to a database of all combinations of known
                  HLA alleles. Sophisticated analysis programmes have been devel-
                  oped for allele assignment and resolution of ambiguities. More
                  recently, next-generation sequencing technologies have been
                  applied to the study of MHC genes. One advantage of next-gener-
                  ation sequencing is the ability to sequence individual DNA strands,
                  thus unambiguously defining the phase of identified SNPs (17).
                  How these high-throughput methods will be applied in diagnostic
                  laboratories is not yet clear, as cost and turn-around times remain
                  significant. However, it is already evident approaches to enrich
                  (capture) the MHC and sequence it in its entirety in a matter of
6        R.J.N. Allcock
5. The Future
Acknowledgements
References
 1. Trowsdale J (2011) The MHC, disease and          5. Horton R, Gibson R, Coggill P et al (2008)
    selection. Immunol Lett. doi:10.1016/j.imlet.       Variation analysis and gene annotation of eight
    2011.01.002                                         MHC haplotypes: the MHC Haplotype Project.
 2. Shiina T, Hosomichi K, Inoko H et al (2009)         Immunogenetics 60:1–18
    The HLA genomic loci map: expression, inter-     6. de Bakker PI, McVean G, Sabeti PC et al (2006)
    action, diversity and disease. J Hum Genet 54:      A high-resolution HLA and SNP haplotype
    15–39                                               map for disease association studies in the
 3. The MHC Sequencing Consortium (1999)                extended      human     MHC.      Nat     Genet
    Complete structure and gene map of a major          38:1166–1172
    histocompatibility complex (MHC). Nature         7. Smith LK, Sayer DC, Whidborne RS et al
    401:921–923                                         (2007) Sequencing-based typing identifies
 4. Marsh SG, Albert ED, Bodmer WF et al (2010)         novel alleles due to single nucleotide polymor-
    Nomenclature for factors of the HLA system.         phisms in ‘conserved’ regions. Tissue Antigens
    Tissue Antigens 75:291–455                          69(suppl 1):56–57
                                                 1     The Major Histocompatibility Complex…              7
 8. Choy MK, Phipps ME (2010) MICA polymor-                 Caucasian population: implications for haplotype
    phism: biology and importance in immunity               tagging. Hum Mutat 24:517–525
    and disease. Trends Mol Med 16:97–106               14. Valente FP, Tan CR, Temple SE et al (2009) The
 9. Posch PE, Cruz I, Bradshaw D et al (2003)               evolution and diversity of TNF block haplotypes
    Novel polymorphisms and the definition of               in European, Asian and Australian Aboriginal
    promoter ‘alleles’ of the tumour necrosis factor        populations. Genes Immun 10:607–615
    and lymphotoxin alpha loci: inclusion in HLA        15. Yang Z, Yu CY (2000) Organisations and gene
    haplotypes. Genes Immun 4:547–558                       duplications of the human and mouse MHC
10. Smith WP, Vu Q, Li SS et al (2006) Toward               complement gene clusters. J Exp Med 191:
    understanding MHC disease associations: partial         2183–2196
    resequencing of 46 distinct HLA haplotypes.         16. Traherne JA, Horton R, Roberts AN et al
    Genomics 87:561–571                                     (2006) Genetic analysis of completely sequenced
11. Stewart CA, Horton R, Allcock RJ et al (2004)           disease-associated MHC haplotypes identifies
    Complete MHC haplotype sequencing for                   shuffling of segments in recent human history.
    common disease gene mapping. Genome Res                 PLoS Genet 2:e9
    14:1176–1187                                        17. Erlich RL, Jia X, Anderson S et al (2011) Next-
12. Degli-Esposti MA, Leaver AL, Christiansen FT            generation sequencing for HLA-typing of class
    et al (1992) Ancestral haplotypes: conserved            I loci. BMC Genomics 12:42–55
    population MHC haplotypes. Hum Immunol              18. Tomazou EM, Rakyan VK, Lefebvre G et al
    34:242–252                                              (2008) Generation of a genomic tiling array of
13. Allcock RJN, Windsor L, Gut IG et al (2004)             the human major histocompatibility complex
    High-density SNP genotyping defines 17                  (MHC) and its application for DNA methylation
    distinct haplotypes of the TNF block in the             analysis. BMC Med Genomics 1:19
                                                                                            Chapter 2
Abstract
Human leukocyte antigen (HLA) typing, utilising the sequence-specific oligonucleotide (SSO) and
sequence-specific primer (SSP) technologies, has been in routine use in many tissue typing laboratories
worldwide for more than 20 years since the development of the polymerase chain reaction. Both methods
are very useful for clinical and research purposes and can provide generic (low resolution) to allelic (high
resolution) typing results. This chapter provides an overview of the SSO and SSP methods in relation to
HLA typing.
1. Introduction
1.1. Sequence-Specific          HLA-SSP typing of the HLA-DR genes was initially developed by
Primer Assay                    Olerup and Zetterquist (1). Further developments of the method
                                quickly followed which enabled sequence-specific primer (SSP)
                                typing of other human leukocyte antigen (HLA) class I and II genes
                                (2–5). In 1995 Bunce et al. developed a comprehensive SSP typing
                                system for the HLA-A, -B, -C, -DRB1, -DRB3, -DRB4, -DRB5,
                                and -DQB1 alleles where all the polymerase chain reactions (PCRs)
                                were designed to work with identical thermocycler conditions (6).
                                SSP typing involves the use of primers complementary to particular
                                HLA allele sequences (e.g. HLA*02:03) or a group of similar alleles
                                (e.g. all the A*02 alleles). Primers are designed so that the polymor-
                                phism to be detected is at the 3¢ end of the primer. If there is a
                                mismatch with the HLA allele at the 3¢ end, the primer will not be
                                extended by Taq DNA polymerase. In the case of HLA-A, -B, and
                                -C typing, most primer sets detect polymorphisms in exons 2 and 3
                                of the class I genes as these regions cover most of the known
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_2, © Springer Science+Business Media New York 2012
                                                           9
10   H. Dunckley
                   Fig. 1. Photo of a sequence-specific primer (SSP) gel. The photo shows amplification of the
                   control gene and human leukocyte antigen (HLA) genes. The control amplicon is present
                   in all lanes and the HLA amplicon is present in lanes 3, 5, and 6. The control amplicon is
                   weaker in lanes 3, 5, and 6.
 Table 1
 SSP result interpretation and HLA typing from the gel in Fig. 1
Well number
1 2 3 4 5 6 7 8 9 10 11 12 13 14
 Control amplicon            +      +    +     +     +       +        +     +     +     +     +     +     +     +
 HLA amplicon                −      −    +     −     +       +        −     −     −     −     −     −     −     −
 DRB1* allele of HLA         01     03   04    07    08      04, 07   09    10    11    12    13    14    15    16
  amplicon                                                     or
                                                               09
“+” Means the amplicon is present; “−” means the amplicon is absent. Based on the positive reactions, the HLA type of
the sample is DRB1*04, 08
                                 and HLA genotyping assignment. The SSP method has the major
                                 advantage of being a very fast method of HLA genotyping, the
                                 process (including DNA extraction) being completed within 2–3 h.
                                 The technique therefore is well suited to the typing of urgent cases,
                                 such as patients requiring HLA-matched platelet transfusions, or
                                 genotyping of donors for organ transplantation. However, because
                                 of the number of PCRs required for each sample, the SSP method
                                 is not suited to the typing of large sample numbers, even with the
                                 use of multi-dispensing pipettes or robotics as there is also the con-
                                 sideration of the number of PCR machines required. Commercial
                                 kits are available which can perform low resolution HLA-A, -B,
                                 -DRB1 typing with 96 PCRs/sample. With the continually increas-
                                 ing large number of HLA alleles, it is becoming more difficult to
                                 resolve the major allele groups with a one-step SSP process.
                                 Additional primer mixes can be added to include newly found
                                 alleles, but there is a limit to the practicality of adding more and
                                 more primer mixes to resolve ambiguities.
1.2. Sequence-Specific           HLA-SSO typing was one of the first PCR-based HLA typing meth-
Oligonucleotide Assay            ods to be developed, initially for the HLA class II loci and then for
                                 the HLA class I genes (7–12). The technique involves extraction of
                                 DNA followed by amplification of a particular HLA gene locus, for
                                 example HLA-A or HLA-B or HLA-DRB1. Primers are selected to
                                 amplify in one PCR tube all known alleles of the particular HLA
                                 locus which requires genotyping. Generally, for HLA class I typing
                                 exons 2 and 3 are co-amplified as one PCR product while for HLA
                                 class II typing only exon 2 is amplified as these are the gene regions
                                 with the majority of the known sequence variation. The amplified
                                 DNA is blotted onto a nylon membrane which is positively charged
                                 permitting the negatively charged DNA to bind strongly. Many sam-
                                 ples can be blotted onto the same membrane and the membrane size
                                 can be increased as sample numbers increase. Multiple membranes
                                 with the amplified DNA samples are prepared, one for each oligo-
                                 nucleotide probe used in the genotyping method.
12   H. Dunckley
But as the SSOs do not cover all regions of the gene, it is still pos-
sible for new alleles to be missed by this method and they would
only be found if the sample was genotyped by another method
such as DNA sequencing.
     While the PCR-SSO method is not a suitable technique for
low sample numbers, there is a method variation known as reverse-
SSO (13, 14) which is well suited to the testing of individual sam-
ples or multiple samples concurrently. In the reverse-SSO
technique, oligonucleotides are individually bound to membranes,
each membrane having all the SSOs bound that are required for a
particular HLA locus or allele group. These membranes are pre-
made so they are available for testing at any time. Sample DNA is
amplified using primers specific for the HLA locus to be typed,
incorporating a label such as biotin. The amplified DNA is then
hybridised to the membrane with the pre-bound oligonucleotides.
Non-specifically bound amplicons are removed by washing and
the hybridisation between the amplicon and membrane bound
oligonucleotide is detected with a chemiluminescent or colour-
based detection system.
     A recently developed variation of the reverse-SSO system
employs Luminex xMAP® technology. The Luminex xMAP® tech-
nique utilises polystyrene microspheres which are dyed internally
with red and infrared fluorophores. The two dyes are incorporated
in differing proportions on different beads allowing 100 unique
sets of microspheres to be developed. Each of the 100 bead sets
can be individually identified with a Luminex® analyser even when
in the same reaction tube. For HLA genotyping, each of the micro-
sphere sets is coated with a different oligonucleotide. Test DNA is
amplified by PCR and hybridised to the bead sets in solution, the
process is carried out in a 96-well PCR plate with one sample
tested/well (each well contains all the different bead sets), allow-
ing 96 samples to be tested simultaneously. Non-hybridised DNA
is removed by washing and amplified DNA which has bound to the
SSOs on the microspheres is detected by a fluorescent dye with a
Luminex® analyser using flow cytometric technology. The analyser
automates the data acquisition and provides quantitative results as
well as HLA assignments. Because 100 different bead sets can be
included in each reaction tube or tray well, it is easier to include a
larger number of SSOs with Luminex xMAP® technology than
other SSO or reverse-SSO methods, enabling a better level of HLA
typing resolution.
     While reverse-SSO typing still has the drawbacks of ambiguity
resolution that the original SSO method has, it is well suited to
genotyping of small sample numbers and there are several com-
mercial kits available utilising this technology. Some commercial
kits incorporate robotics for the hybridisation, washing and detec-
tion steps making the method also suitable for the typing of larger
sample numbers.
14        H. Dunckley
1.3. Interpretation     Because of the large and ever increasing number of HLA alleles,
of Results              data analysis and HLA genotyping assignment now requires the
                        use of software, commercial HLA typing kits generally provide
                        software programs for this purpose. For the SSP, SSO and reverse-
                        SSO methods, most analysis software follows the same principle.
                        The reaction patterns of known alleles are established for the
                        primer sets used in the SSP kit or the oligonucleotides in the SSO
                        or reverse-SSO methods based on known HLA sequence data
                        from the IMGT/HLA database (15, 16). When a sample is tested,
                        the reaction pattern obtained is compared to the reaction patterns
                        of known alleles. The software then provides a list of possible HLA
                        types based on the patterns of known alleles which match the sam-
                        ple being tested. With the SSP and SSO methods, multiple results
                        will sometimes be obtained as it is difficult for these methods to
                        completely resolve all possible heterozygous allele combinations.
                        However, provided the alternative results are within the same
                        allele group and do not cross antigen specificities, this is not usu-
                        ally an issue particularly if a low resolution typing result is satisfac-
                        tory. The SSO and SSP methods can also be used in conjunction
                        with sequencing-based typing (SBT) by providing a generic result
                        which determines the primers to be used in allele level SBT. In
                        some cases, ambiguities can arise with SBT (with heterozygous
                        sequences) in which case the SSP method can be used to help
                        resolve an allele level typing. Because of the continual increase in
                        the identification of new alleles, laboratories must keep all typing
                        methods up to date which requires the databases used for SSO
                        and SSP result interpretation to be updated at least every 6 months.
                        The IMGT/HLA database of officially named HLA alleles is
                        updated every month (15, 16).
                             While molecular HLA typing is more accurate and provides a
                        higher level of resolution than serological tissue typing (17–20),
                        molecular methods will not detect new alleles if a new polymor-
                        phism is not in the region where an amplification primer or SSO
                        matches the gene. In addition, null or non-expressed alleles are
                        usually detected when molecular typing is used in conjunction with
                        serological methods. When only one method is used both serology
                        and molecular techniques sometimes fail to detect null alleles.
                        While null alleles are not common, tissue typing protocols for
                        transplantation should be developed to avoid missing these non-
                        expressed alleles, especially in matching for haematopoietic stem
                        cell transplantation.
1.4. Clinical           The SSP, SSO and reverse-SSO methods are all suitable for use in
Applications            routine tissue typing laboratories as well as for research purposes.
                        With a variety of commercial kits now available, laboratories can
                        choose which method is most suitable for their work, and a combi-
                        nation of kits can be used for different requirements. In laborato-
                        ries testing for clinical purposes like transplantation it is useful to
                                          2   HLA Typing by SSO and SSP Methods          15
                      have more than one method available so that any unusual results
                      can be verified. If dealing with low sample numbers or urgent
                      testing, then SSP is very practical. With low to large sample num-
                      bers reverse-SSO can be used, this method also gives a reasonable
                      turnaround time for testing. For large sample batches and when
                      the test time is less urgent, then SSO typing is a very cost-effective
                      method.
                           As it is now easier to buy commercial HLA-SSP and reverse-
                      SSO kits rather than developing in-house methods, the protocols
                      described below summarise the steps that would be followed when
                      using a commercial kit, some specific details being provided by the
                      kit manufacturer supplier. When using commercial kits, each ship-
                      ment and batch of kits needs to be checked before routine use.
                      This can be accomplished using samples of known HLA type to
                      verify the typing results obtained with the kit.
2. Materials
2.1. HLA-SSP Typing    1. Commercial kits will usually supply the primer sets (both HLA
                          and control primers) already pre-dispensed into microtitre
                          trays or tubes ready for use in a thermocycler. Some trays will
                          also include the dNTPs and PCR buffer with the primers.
                          Sometimes these mixes will be lyophilised, other times they
                          will be in solution but covered with a layer of paraffin oil to
                          avoid evaporation.
                       2. PCR buffer and dNTPs, if not included with the pre-dispensed
                          primer sets, are usually included with the commercial kits as is
                          the gel loading buffer.
                       3. Taq DNA Polymerase—may or may not be included with the
                          commercial kit.
                       4. Tube caps or tray sealers for the PCR trays are usually included
                          with the commercial kits.
                       5. Sterile molecular grade water (dH2O).
                       6. Pipettes and sterile tips to dispense 1–10 μL, 10–200 μL, and
                          200–1,000 μL volume ranges. Electronic multi-dispensing
                          pipettes are better if the sample numbers are high because of
                          the large amount of pipetting involved.
                       7. Thermocycler with a heated lid.
                       8. Agarose horizontal gel tank equipment, gel casting trays, gel
                          combs, and power supply.
                       9. Agarose (DNA grade).
                      10. Electrophoresis buffer as specified by the kit supplier.
                      11. Microwave or heating apparatus to dissolve the agarose.
16       H. Dunckley
2.2. HLA Reverse-SSO    1. A master mix containing biotinylated primers, dNTPs and buf-
Typing                     fer is usually supplied with commercial kits. MgCl2 may be pro-
                           vided in a separate tube. These are normally supplied frozen.
                        2. Taq DNA polymerase may be included in the master mix, or
                           may be provided in a separate tube, or may not be included
                           with a commercial kit.
                        3. Strips with pre-bound oligonucleotides are provided with com-
                           mercial kits.
                        4. Control DNA is usually supplied with a commercial kit.
                        5. Sterile molecular grade water (dH2O).
                        6. Pipettes and sterile tips to dispense 1–10 μL, 10–200 μL, and
                           200–1,000 μL volume ranges. Electronic multi-dispensing
                           pipettes are better if the sample numbers are high because of
                           the large amount of pipetting involved.
                        7. PCR plastic ware—tubes or microtitre trays, caps or tray sealers.
                        8. Thermocycler with a heated lid.
                        9. Agarose horizontal gel tank equipment, gel casting trays, gel
                           combs, and power supply.
                       10. Agarose (DNA grade).
                       11. Electrophoresis buffer as specified by the kit supplier.
                       12. Microwave or heating apparatus to dissolve the agarose.
                       13. Ethidium bromide 10 mg/mL (see Note 1).
                       14. UV transilluminator and photographic equipment—a digital
                           photo allows the photo to be stored electronically.
                       15. Solutions required for denaturation, hybridisation, washing,
                           and probe detection are included with commercial kits (see
                           Note 2).
                       16. Some reverse-SSO kit suppliers provide automated assay equip-
                           ment. Otherwise, the process can be carried out manually and
                           requires a rocking platform, shaking water bath and containers
                           for hybridisation, washing and probe detection steps as well as
                           sterile pipettes (2–50 mL), and adjustable volume dispensers.
                                          2   HLA Typing by SSO and SSP Methods          17
3. Methods
3.1. HLA-SSP Method    1. Remove the PCR tray containing the primer sets and the kit
                          components from the freezer or fridge in the pre-PCR area
                          (see Note 3).
                       2. Following the manufacturer’s instructions, add the required
                          amount of dH2O to the PCR buffer provided in the kit, mix
                          well and then add Taq DNA polymerase. Add the amount
                          specified by the manufacturer to the negative control well of
                          the PCR tray (see Notes 4 and 5).
                       3. Add the required amount of DNA to the remaining buffer/
                          enzyme solution, mix well, and add the amount specified by
                          the manufacturer to each of the test wells of the PCR tray. Seal
                          or cap each tube tightly (see Notes 6–8).
                       4. Ensure the liquid of all the reaction mixes is at the base of the
                          tray—hand flick the tray, or tap the tray gently on the bench,
                          or pulse centrifuge the tray at a low speed (see Note 9).
                       5. In the PCR or post-PCR area, place the tray in the thermocy-
                          cler and perform the PCR according to the kit supplier’s
                          specifications (see Notes 10–13).
                       6. When the PCR program is complete, remove the tray from the
                          thermocycler.
                       7. In the post-PCR area, electrophorese the amplified samples and
                          DNA molecular weight marker on an agarose gel containing
                          ethidium bromide in the buffer recommended by the kit sup-
                          plier. The supplier will provide details on the agarose concentra-
                          tion, gel size, well size, and voltage required (see Notes 14–17).
                       8. Take a photo of the gel under a UV light (see Note 18).
                       9. Check that the negative control is negative (no amplification
                          product present). Score the positive PCRs (checking that the PCR
                          product is the correct size) and note any wells where both the
                          control PCR and HLA PCR failed as these cannot be scored as
                          negative but as an unknown result (see Notes 19–22; Table 2).
                      10. Input the results of the PCRs (positive, negative, or unknown/
                          no result) into the data analysis software program and obtain
                          the HLA typing from the software (see Notes 23–28).
18       H. Dunckley
                       Table 2
                       Interpretation of SSP gel results
3.2. HLA Reverse-SSO   1. In the pre-PCR area, make up the reaction mix as per the man-
Method                    ufacturer’s recommendations (buffer, water, dNTPs, primers,
                          and Taq DNA polymerase). The amount of reaction mix made
                          will depend on the number of samples to be amplified. Aliquot
                          the reaction mix into the PCR tubes or tray (see Notes 3, 5, 7,
                          and 29).
                       2. Add one DNA sample (amount as per the kit supplier
                          specifications) to each reaction mix on the tray checking that
                          the correct sample is added to each well or tube (see Note
                          30).
                       3. Seal or cap each tube tightly, ensure the liquid of all the reac-
                          tion mixes is at the base of the tray—hand flick the tray, or tap
                          the tray gently on the bench, or pulse centrifuge the tray at a
                          low speed (see Notes 8 and 9).
                       4. In the PCR or post-PCR area, immediately place the tray in
                          the thermocycler and perform the PCR according to the kit
                          supplier’s specifications (see Note 31).
                       5. When the PCR program is complete, remove the tray from the
                          thermocycler.
                       6. In the post-PCR area, electrophorese the amplified samples on
                          an agarose gel containing ethidium bromide in the buffer rec-
                          ommended by the kit supplier. The supplier will provide details
                          on the agarose concentration, gel size, well size, and voltage
                          required. Note: the agarose gel step is optional, some commer-
                          cial kits do not include this step or steps 7 and 8 below.
                       7. Photograph the gel under a UV light (see Note 18).
                       8. Check that the negative control is negative (no amplification
                          band present) and the PCR has worked for the test samples
                          (amplification band evident on the gel).
                       9. Denature the amplified DNA sample as per the instructions in
                          the kit and add the denatured DNA to the container with the
                               2   HLA Typing by SSO and SSP Methods         19
4. Notes
24. The presence of more than two specific HLA alleles indicates
    contamination between PCR products or between DNA
    samples during the DNA extraction or PCR preparation
    stages. Make sure the pre- and post-PCR work is kept sepa-
    rate (different rooms, lab coats, gloves, equipment and
    reagents); clean the working area with 10% bleach and ensure
    there is no mixing of PCR amplicons. Check there has been
    no mix up at the DNA extraction stage—check other test
    samples for a possible mix up.
25. False positive SSP reactions can be caused by contamination
    when dispensing reagents to set up the PCR or when loading
    the gel with the amplified product. Take more care when
    pipetting during the PCR setup or when loading the gel.
26. A false positive or false negative reaction may be due to an
    error in loading the gel so that the PCRs have been loaded in
    the wrong order. Check the alignment of the PCR mixes and
    gel lanes.
27. An unexpected positive or negative reaction may sometimes be
    due to the presence of a new allele. Repeat the typing and if the
    same reaction is seen verify the typing by SBT.
28. Persistent false negative or positive reactions with the same
    PCR mix may be due to a kit problem. Double check the kit
    with a control sample and advise the supplier if the control
    sample also gives the same false negative or positive result.
29. If an error is made during the PCR setup, this can lead to inefficient
    amplification and weak probe signals. The PCR must be set up
    exactly as specified in the kit instructions in relation to reagent
    volumes and concentrations. Ensure that pipettes are calibrated.
30. If insufficient test DNA, poor quality or degraded DNA is used,
    there may be inefficient amplification and weak probe signals. If
    this occurs, the volume, concentration, and purity of the test
    DNA should be checked. If required re-precipitate the DNA
    (ethanol and salt step) and resuspend the DNA in dH2O or repeat
    the whole DNA extraction with new reagents. DNA can be
    extracted from nucleated cells by any preferred method (in-house
    or commercial kit, manual or robotic system). If blood samples
    are used the anticoagulant should be EDTA or ACD, not hepa-
    rin which can inhibit the PCR. DNA should be kept in TE buffer
    for long-term storage, the final concentration of the DNA prior
    to amplification should be according to the recommendations of
    the kit supplier as this varies depending on the kit used. The
    OD260/280 of the DNA should be between 1.6 and 1.9.
31. Incorrect PCR conditions can result in a low level of
    amplification or no amplification of the test DNA (and conse-
    quently weak or no hybridisation signals). If this occurs, the
    PCR program should be checked.
24        H. Dunckley
                             32. Always handle the membranes at the end of the strips using
                                 forceps.
                             33. If the water bath, incubator, or wash solution temperature is
                                 too high when removing non-specifically bound probe (strin-
                                 gency wash step), it can result in weak hybridisation signals.
                                 If the wash temperature is too low, there can be false positive
                                 probe signals or high background on the probe support
                                 membrane. It is important to set the temperature with a cali-
                                 brated thermometer and to monitor the temperature during
                                 the stringency wash step.
                             34. If the wash buffer solution is made up incorrectly, this can
                                 cause either weak hybridisation signals, high background on
                                 the probe support membrane or false positive probe signals. It
                                 is critical that wash buffer solutions are prepared carefully and
                                 that salt concentrations are correct.
                             35. High background on the probe support membrane can be
                                 caused by detergent residue in the hybridisation or wash con-
                                 tainers. Do not use detergent when washing the containers or
                                 beakers used for making solutions but rinse the containers very
                                 well with water.
                             36. High background on the probe support membrane can be due
                                 to the conjugate step being performed at an incorrect tem-
                                 perature. Monitor this step with a calibrated thermometer.
                             37. Ensure the substrate incubation time is correct. If the time is
                                 too short, this can lead to weak hybridisation signals and if it is
                                 too long it can result in high background on the probe support
                                 membrane. Also check that the substrate is at room tempera-
                                 ture before it is used.
                             38. If the positive control result is strong but the probe intensity of
                                 the test samples is weak, this is most likely due to a weak PCR
                                 because of insufficient test DNA or poor quality DNA. If this
                                 occurs, the volume, concentration, and purity of the test DNA
                                 should be checked. If required re-precipitate the DNA (etha-
                                 nol and salt step) and resuspend the DNA in dH2O or repeat
                                 the whole DNA extraction with new solutions. Use non-hepa-
                                 rinised blood to extract DNA.
References
 1. Olerup O, Zetterquist H (1992) HLA-DR typ-           2. Olerup O, Aldener A, Fogdell A (1993) HLA-
    ing by PCR amplification with sequence-specific         DQB1 and -DQA1 typing by PCR amplification
    primers (PCR-SSP) in 2 hours: an alternative to         with sequence-specific primers (PCR-SSP) in 2
    serological DR typing in clinical practice includ-      hours. Tissue Antigens 41:119–134
    ing donor-recipient matching in cadaveric            3. Bunce M, Taylor CJ, Welsh KI (1993) Rapid
    transplantation. Tissue Antigens 39:225–235             HLA-DQB typing by eight polymerase chain
                                                  2   HLA Typing by SSO and SSP Methods                   25
    reaction amplifications with sequence-specific    11. Gao X, Jakobsen IB, Serjeantson SW (1994)
    primers (PCR-SSP). Hum Immunol 37:                    Characterization of the HLA-A polymorphism
    201–206                                               by locus-specific polymerase chain reaction
 4. Browning MJ, Krausa P, Rowan A et al (1993)           amplification and oligonucleotide hybridiza-
    Tissue typing the HLA-A locus from genomic            tion. Hum Immunol 41:267–279
    DNA by sequence-specific PCR: comparison of       12. Cereb N, Yang SY (1997) Dimorphic primers
    HLA genotype and surface expression on col-           derived from intron 1 for use in the molecular typ-
    orectal tumor cell lines. Proc Natl Acad Sci          ing of HLA-B alleles. Tissue Antigens 50:74–76
    USA 90:2842–2845                                  13. Erlich H, Bugawan T, Begovich AB et al (1991)
 5. Sadler AM, Petronzelli F, Krausa P et al (1994)       HLA-DR, DQ and DP typing using PCR
    Low-resolution DNA typing for HLA-B                   amplification and immobilized probes. Eur J
    using sequence-specific primers in allele- or         Immunogenet 18:33–55
    group-specific ARMS/PCR. Tissue Antigens          14. Bugawan TL, Apple R, Erlich HA (1994) A
    44:148–154                                            method for typing polymorphism at the HLA-A
 6. Bunce M, O’Neill CM, Barnardo MCNM et al              locus using PCR amplification and immobilized
    (1995) Phototyping: comprehensive DNA typ-            oligonucleotide probes. Tissue Antigens
    ing for HLA-A, B, C, DRB1, DRB3, DRB4,                44:137–147
    DRB5 & DQB1 by PCR with 144 primer mixes          15. Robinson J, Malik A, Parham P et al (2000)
    utilising sequence-specific primers (PCR-SSP).        IMGT/HLA—a sequence database for the
    Tissue Antigens 46:355–367                            human major histocompatibility complex.
 7. Wordsworth BP, Allsopp CE, Young RP et al             Tissue Antigens 55:280–287. http://www.ebi.
    (1990) HLA-DR typing using DNA                        ac.uk/imgt/hla/
    amplification by the polymerase chain reaction    16. Robinson J, Mistry K, McWilliam H et al
    and sequential hybridization to sequence-             (2011) The IMGT/HLA database. Nucleic
    specific oligonucleotide probes. Immunogenetics       Acids Res 39(suppl 1):D1171–D1176
    32:413–418                                        17. Mytilineos J, Scherer S, Dunckley H et al (1993)
 8. Morel C, Zwahlen F, Jeannet M et al (1990)            DNA HLA-DR typing results of 4000 kidney
    Complete analysis of HLA-DQB1 polymorphism            transplants. Transplantation 55:778–781
    and DR-DQ linkage disequilibrium by oligonu-      18. Mytilineos J, Lempert M, Middleton D et al
    cleotide typing. Hum Immunol 29:64–77                 (1997) HLA class I DNA typing of 215 “HLA-
 9. Scharf SJ, Griffith RL, Erlich HA (1991) Rapid        A, -B, -DR zero mismatched” kidney trans-
    typing of DNA sequence polymorphism at the            plants. Tissue Antigens 50:355–358
    HLA-DRB1 locus using the polymerase chain         19. Lorentzen DF, Iwanaga KK, Meuer KJ et al
    reaction and nonradioactive oligonucleotide           (1997) 25% Error rate in serologic typing of
    probes. Hum Immunol 30:190–201                        HLA-B homozygotes. Tissue Antigens
10. Kimura A, Sasazuki T (1992) Eleventh inter-           50:359–365
    national histocompatibility workshop refer-       20. Mytilineos J, Christ U, Lempert M et al (1997)
    ence protocol for the HLA-DNA typing                  Comparison of typing results by serology and
    technique. In: Tsuji K, Aizawa M, Sasazuki T          polymerase chain reaction with sequence-
    (eds) HLA 1991. Oxford University Press,              specific primers for HLA-Cw in 650 individuals.
    Oxford, p 397                                         Tissue Antigens 50:395–400
                                                                                            Chapter 3
Abstract
This chapter describes the application of diagnostic HLA typing for disease association and five methods
used for specific HLA genotypes. The methods utilise a combination of polymerase chain reaction (PCR)
amplification to detect sequence polymorphism by the presence or absence of amplification, nucleotide
sequencing of the PCR product, and hybridisation of the PCR product with labelled probes. The probes
are specific for sequence polymorphism associated with the genotype and are attached to either a Micro
Bead or a Solid Phase. In addition, the detection of single nucleotide polymorphism(s) which “tag” for the
genotype using a real-time PCR is described.
     Key words: HLA, Disease, Association, Hypersensitivity, Methods, B*27, B*57, Coeliac, B*1502,
     Narcolepsy, DQ
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_3, © Springer Science+Business Media New York 2012
                                                          27
28   M.D. Varney et al.
Table 1
Diagnostic HLA disease and hypersensitivity associations
 Table 2
 Coeliac disease associated DQ2 and DQ8 haplotypes and their prediction by the six
 tagging single nucleotide polymorphisms (SNPs)
                                                            Positive        Negative
                                                            predicting      predicting Applied
 DQ                                                         allele (other   allele (other biosystems
 haplotype DRB1           DQA1 DQB1 SNP rs number           allele)         allele)       assay number
2. Materials
2.1. Sequence             1. HLA-B 10× PCR Buffer: To prepare dissolve 8.11 g Trisma-
Specific Priming             Base in 80 mL H2O on a magnetic stirrer and adjust to pH = 8.8
and Sequence-Based           using concentrated HCl. Add 2.19 g (NH4)2 SO4 and dissolve.
Typing                       Make up to 95 mL, filter through a 0.22-mm filter, and add
                             1 mL Tween 20. Aliquot and store frozen.
                          2. SSP Buffer: To prepare 1,500 mL add 500 mL 10× 1.5 mM
                             MgCl2 PCR Buffer (as supplied with Taq DNA Polymerase),
                             500 mL 10× dNTPS, 250 mL Glycerol, 200 mL H2O, 50 mL
                             Cresol Red Solution (10 mg/mL diluted in H2O).
                          3. 10× dNTPS: A stock solution is prepared containing a final
                             concentration of 2 mM of dCTP, dGTP, dATP, and dTTP
                             diluted in H2O.
                          4. B*1502 Master Mix: For each sample a 39-mL Master Mix is
                             prepared containing 5 mL HLA-B 10× PCR buffer, 2 mL
                             dNTPs (40 mM), 2 mL 50 mM MgCl2, 3.25 mL B*1502
                             primer mix, 35.35 mL H2O.
                          5. B*27 Master Mix: For each sample a 39-mL Master Mix is pre-
                             pared containing 5 mL HLA-B 10× PCR buffer, 0.3 mL dNTPs
                             (40 mM), 6 mL B*27 primer mix, 27.7 mL H2O.
                          6. B*57 Master Mix: For each sample a 39-mL Master Mix is pre-
                             pared containing 5 mL HLA-B 10× PCR buffer, 2 mL dNTPs
                             (40 mM), 3 mL 50 mM MgCl2, 15.75 mL B*57 primer mix,
                             13.25 mL H2O.
                          7. Formamide Master Mix: For each sample prepare a formamide
                             mix containing 5 mL Deionised Formamide and 4 mL deionised
                             H2O.
                          8. 2.5 mM MgCL2 DQB PCR Buffer: To prepare sufficient for
                             200 reactions (4,310 mL) add 500 mL 10× PCR Buffer (as sup-
                             plied with Taq DNA Polymerase, no MgCl2), 250 mL 50 mM
                             MgCl2, 500 mL 10× dNTPS, 250 mL DiMethyl Sulphoxide,
                             3,010 mL H2O; 2.0 and 1.2 mM MgCL2 DQB PCR Buffers
                             are prepared by adjusting the proportion of 50 mM MgCl2 and
                             H2O appropriately.
32       M.D. Varney et al.
2.4. TaqMan® Typing           1. TaqMan® (2×) Genotyping Master Mix (Applied Biosystems).
of HLA-Tagging SNPs           2. TaqMan® (20×) SNP Genotyping Assay Mix (Applied
in Coeliac Disease               Biosystems).
                              3. PCR-quality H2O.
                              4. Specialist equipment: ABI 7900HT real-time PCR system
                                 (Applied Biosystems) or equivalent with 84- or 96-well optical
                                 PCR plates and lids.
  3   Methods for Diagnostic HLA Typing in Disease Association and Drug Hypersensitivity    33
3. Methods
3.2. Sequence-Based        1. Select the PCR reaction(s) for the association to be tested
Typing                        using the primer pair specificities listed in Table 4. Additional
                              primer pairs e.g. DQB1*0201/02/03, B CG are described to
                              assist in the resolution of ambiguous results.
                                                                                                                               34
Table 3
SSP primer pair sequences and specificity used in diagnostic HLA disease and hypersensitivity association testing
Table 4
SBT primer pair sequences and specificity used in diagnostic HLA disease and hypersensitivity association testing
3.3. Micro Bead            1. A 10-mL PCR is prepared for each sample containing 6.9 mL
Hybridisation Typing          D-mix, 2.0 mL Primer Set, 0.1 mL Taq DNA Polymerase (not
Assay (Luminex/               supplied) and 1 mL DNA (20 ng/mL). Cycle in a Perkin Elmer
Labtype)                      PCR machine with ramp setting at 9600 for 96°C-180 s ×1
                              cycle, 96°C-20 s, 60°C-20 s, 72°C-20 s ×5 cycles, 96°C-10 s,
                              60°C-15 s, 72°C-20 s ×30 cycles, 72°C-10 min, 4°C hold.
40   M.D. Varney et al.
3.4. Solid Phase           1. Prepare a 45-mL PCR Master Mix for each sample and a nega-
Hybridisation Typing          tive control containing 19 mL ultrapure H2O, 5 mL AmpliTaq
(DELFIA Coeliac               Gold 10× PCR Buffer, 5 mL dNTP (2 mM), 5 mL DQA1fo
Disease)                      (10 pml/mL), 5 mL DQA1re (10 pml/mL), 2.5 mL DQB1fo
                              (10 pml/mL), 2.5 mL DQB1re (10 pml/mL), 1 mL AmpliTaq
                              Gold per sample (see Note 6).
                           2. Pipette 45 mL of the Master Mix to each PCR well/tube and
                              add 5 mL of DNA (optimally 20 ng/mL) and cycle at 95°C-
                              15 min ×1 cycle, 95°C-50 s, 54°C-60 s, 72°C-60 s ×40 cycles,
                              72°C-5 min, 4°C hold. The PCR products may be stored at
                              2–8°C for 2 days or at approximately −20°C for 2–3 weeks
                              prior to assay.
                           3. The PCR products may be visualised and approximate size
                              determined using standard agarose or polyacrylamide gel
                              electrophoresis.
                           4. Binding the PCR products: Dilute the Wash Concentrate
                              (50 + 1,200 mL) and warm half of it in an incubator at 43°C
                              (see Note 7). Dilute the Bio-Hybridization Control to DELFIA
                              Hybridization Buffer (0.5 + 49.5 mL).
                           5. Pipette 10 mL of water to the wells 1–4 (background control
                              for hybridization), 10 mL of diluted Bio-Hybridization Control
                              to the wells 5–8, 10 mL of negative control to the wells
                              9–12,10 mL of each PCR product to 4 wells, 50 mL Hybridization
                              Buffer to each well mentioned above.
                           6. Incubate 30 min at room temperature on a plateshaker.
                           7. Dilute the probes with the Hybridization Buffer, 100 mL is
                              required per well. Prepare two solutions: A including Europium
                              (Eu)-DQB1*0302 (1 ng/well) and Terbium(Tb)-DQB1*02
                              (1 ng/well), and B including Samarium (Sm)-DQA1*05
                              (3 ng/well), Tb-DQB1 Control (3 ng/well), and Eu-DQA1
                              Control (0.5 ng/well). The dilutions must be kept on ice and
                              used within 30 min. The probes must be on ice and returned
                              to a freezer as quick as possible (see Note 8).
                           8. Denaturation and hybridization: Wash the plate four times
                              with 400 mL of prewarmed washing dilution using the auto-
                              matic washer.
42      M.D. Varney et al.
3.5. TaqMan® Typing          1. For a 384-well microplate, a 4-mL PCR is prepared for each
of HLA-Tagging SNPs             sample containing 2.0 mL TaqMan® (2×) Genotyping Master
in Coeliac Disease              Mix (Applied Biosystems), 0.2 mL TaqMan® (20×) SNP
                                Genotyping Assay mix and 1.8 mL DNA (10 ng/reaction)
                                suspended in PCR-quality H2O. Prepare a pre-mix of the
                                Genotyping Master Mix and SNP Genotyping Assay mix for
                                the required number of samples and aliquot 2.2 mL to each
                                well of an empty plate followed by 1.8 mL of DNA using a
                                multichannel pipette. Alternatively, first transfer the DNA
                                using a pipette robot into the wells of six identical 384 plates
                                and let the DNA dry into the bottom of the wells at room
                                temperature or 37°C. After drying, the plates can be stored
                                covered with lid at room temperature. To the plates add the
                                pre-mix, described above but including 1.8 mL PCR-quality
                                H2O instead of DNA, using a multichannel or stepper pipette.
                                To reduce reagent use, the pre-mix may be modified to 1.5 mL
                                Genotyping Master Mix, 0.12 mL SNP Genotyping Assay mix,
                                and 0.58 mL H2O (or 2.38 mL H2O if plate dried DNA is used).
                                This may also improve the amplification of some of the SNPs
                                (see Note 12).
3   Methods for Diagnostic HLA Typing in Disease Association and Drug Hypersensitivity     43
4. Notes
                            11. The hybridization control must be over 100 cps, the DQA1
                                control over 700 cps, and the DQB1 control over 60 cps.
                                Washes must be done very carefully as background signal can
                                easily interfere with quantification.
                            12. In many populations, over 90% of patients with coeliac disease
                                are positive for the DQA1*05-DQB1*02 haplotype. Whenever
                                possible, it is therefore advisable to have both patients and
                                controls within each genotyped 384-well plate to provide a
                                good cluster definition of the three genotypes in the analysis
                                step.
                            13. A long number of cycles may be required to see a clear plateau
                                phase when the SNPs are not working optimally due to the
                                difficult template sequence.
Acknowledgements
                            The DR/DQ SSP, SBT, and micro bead hybridisation methods are
                            a culmination of input by the molecular typing staff of the
                            Transplantation and Immunogenetics Service of the Australian
                            Red Cross Blood Service.
References
    standardised evaluation of SSO, SSP and               induced cutaneous reactions in Han Chinese
    sequencing-based typing results. Tissue               [Erratum in: epilepsia 2008]. Epilepsia
    Antigens 51:587–592                                   48:1015–1018
11. Sayer DC, Goodridge DM, Christiansen FT           18. Kotsch K, Wehling J, Blasczyk R (1999)
    (2004) Assign 2.0: software for the analysis of       Sequencing of HLA class II genes based on the
    Phred quality values for quality control of HLA       conserved diversity of the non-coding regions:
    sequencing-based typing. Tissue Antigens              sequencing based typing of HLA-DRB genes.
    64:556–565                                            Tissue Antigens 53:486–497
12. Rozemuller EH, Ploeger LS, Mulder W (2008)        19. Voorter CE, Kik MC, van den Berg-Loonen
    SBTengine; the ultimate solution for high reso-       EM (1998) High-resolution HLA typing for
    lution HLA typing. Hum Immunol 69:131                 the DQB1 gene by sequence-based typing.
13. Olerup O, Aldener A, Fogdell A (1993) HLA-            Tissue Antigens 51:80–87
    DQB1 and -DQA1 typing by PCR amplification        20. Van Dijk A, Melchers R, Tilanus M, Rozemuller
    with sequence-specific primers (PCR-SSP) in 2         E (2007) HLA-DQB1 sequencing-based typing
    hours. Tissue Antigens 41:119–134                     updated. Tissue Antigens 69(Suppl 1):
14. Olerup O (1994) HLA-B*27 typing by group              64–65
    specific PCR amplification. Tissue Antigens       21. Dunn PP, Day S, Williams S, Bendukidze N
    43:253–256                                            (2005) HLA-DQB1 sequencing-based typing
15. Sayer DC, Cassell HS, Christiansen FT (1999)          using newly identified conserved nucleotide
    HLA-B*27 typing by sequence specific                  sequences in introns 1 and 2. Tissue Antigens
    amplification without DNA extraction. Mol             66:99–106
    Pathol 52(5):300–301                              22. Tilanus MGJ (ed) (2000) IHWG technical
16. Martin AM, Nolan D, Mallal S (2005) HLA-              manual. Genomic analysis of the human MHC.
    B*5701       typing    by     sequence-specific       DNA based typing for HLA Alleles & linked
    amplification: validation and comparison              polymorphisms. Fred Hutchinson Cancer
    with sequence-based typing. Tissue Antigens           Research Centre, Seattle, USA
    65:571–574                                        23. Cereb N, Yang SY (1997) Dimorphic primers
17. Man CB, Kwan P, Baum L, Yu E, Lau KM,                 derived from intron 1 for use in the molecular
    Cheng AS, Ng MH (2007) Association between            typing of HLA-B alleles. Tissue Antigens
    HLA-B*1502 allele and antiepileptic drug-             50:74–76
                                                                                            Chapter 4
Abstract
The LABType® SSO (One Lambda, Inc) and Gen-Probe LIFECODES HLA-SSO HLA Typing tests are
rapid and efficient assays for determining human leukocyte antigens (HLAs). The principle of these assays
involves the hybridization of reverse sequence-specific oligonucleotide probes each attached to a unique
colour coded microsphere to identify HLA class I and class II alleles. Target DNA is polymerase chain
reaction (PCR) amplified using group-specific primers and then biotinylated which allows it to be detected
using R-Phycoerythrin-conjugated Streptavidin. The PCR product is then denatured and allowed to
hybridise to complementary DNA probes conjugated to fluorescently code microsperes. The Luminex®
Flow Analyser achieves detection by determining the fluorescent intensity of PE on each microsphere. The
assignment of HLA alleles is based on the reaction pattern of the various beads compared to patterns with
known HLA alleles.
1. Introduction
                                Over the past few decades, human leukocyte antigen (HLA) typing
                                methods have utilized advances in technology to provide assays
                                with higher resolution whilst reducing cost and time. Historically,
                                HLA typing was first performed by serological typing of HLA anti-
                                gens using antisera with defined specificities (1, 2), but this proce-
                                dure has largely been replaced by DNA typing methods. The
                                polymerase chain reaction (PCR) assay developed by Saiki et al. in
                                1985 (3) was a major breakthrough technology for HLA genotyp-
                                ing, and methods using PCR-amplified DNA are now widely used
                                by HLA laboratories throughout the world. Currently there are
                                numerous DNA-based methods available that can detect HLA
                                alleles from PCR-amplified DNA, such as restriction fragment
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_4, © Springer Science+Business Media New York 2012
                                                          47
48         D. Trajanoski and S.J. Fidler
2. Materials
2.1.2. Materials and          1. Genomic DNA—patient samples and controls (see Note 2).
Reagent Requirements
                              2. Deionised water.
                              3. 70% Ethanol.
                  4   HLA Typing Using Bead-Based Methods        49
2.2.2. Materials and        The same as Subheading 2.1.2 except replace One Lambda SSO
Reagent Requirements        Genotyping kit with:
                            ●      Gen-Probe LIFECODES HLA-SSO Typing Kit (see Note 6)
                                   containing
                                   –     LifeCodes HLA master mix.
                                   –     LifeCodes Probe Mix.
                                   –     Dilution Solution.
                                   –     SAPE.
3. Methods
3.1. LABType® SSO               1. Create a LABType® PCR program on your thermal cycler as
HLA Typing Method                  shown in Table 1.
3.1.1. DNA Amplification
                                Table 1
                                LABType® SSO polymerase chain reaction (PCR) program
    Table 2
    LABType® amplification mixture
                                          Amplification   AmpliTaq
    Number of reactions    D-mix (mL)a    primer (mL)a    polymerase (mL)
     1                        13.8          4              0.2
    10                       138.0         40              2.0
    50                       690.0        200             10.0
    96                     1,491.0        432             21.6
a
As provided in the One Lambda SSO genotyping kit
3.1.2. Denaturation/             1. Turn on thermal cycler and run program to 60°C hold. Make
Neutralisation                      sure lid is heated to optimum temperature before use.
                                 2. Remove reagents listed in Tables 3 and 4 and supplied in the
                                    SSO genotyping kit from −80°C freezer to room temperature
                                    to thaw and then prepare all reagent volumes in an appropri-
    Table 3
    LABType® reagent volumes
     1                     2.5                5                    34                 480             4
    10                    25                50                   340                4,800            40
    20                    50               100                   680                9,600            80
    50                  125                250                 1,700               24,000           200
    96                  240                480                 3,264               46,080           384
a
 Number of tests includes all samples to be tested including appropriate controls and extra volume (see Notes 2 and 9
for comments)
b
 As provided in the One Lambda SSO genotyping kit
                                             4    HLA Typing Using Bead-Based Methods           53
                           Table 4
                           LABType® R-Phycoerythrin-conjugated Streptavidin
                           (SAPE) and SAPE buffer volumes
Number of tests SAPE stock volume (mL) SAPE buffer volume (mL)a
                            1                    0.5                         49.5
                           10                    5.0                       495.0
                           20                 10.0                         990.0
                           50                 25.0                        2,475.0
                           96                 48.0                        4,752.0
                       a
                       As provided in the One Lambda SSO genotyping kit
3.1.5. Sample Acquisition      1. Turn on the system and set up the Luminex® Flow Analyser for
on Luminex® Flow                  sample acquisition and calibration. (Further details of how to
Analyser                          use the Luminex® Flow Analyser can be found in the Luminex®
                                  user’s manual.)
                               2. Verify the levels of sheath fluid and waste fluid (see Note 23).
                               3. Perform Warm up by selecting “Warm Up” button in the
                                  Luminex opening system. The warm up requires no plate or
                                  solution.
                               4. Prime the instrument by selecting “Prime”. Again this step
                                  requires no plate or solution.
                               5. Perform a 70% alcohol flush, followed by four washes with
                                  sheath fluid.
                               6. A calibration should be performed every time the instrument
                                  is turned on or if the temperature of the instrument has
                                  increased or decreased by more than 3°C from the time of
                                  calibration.
                                              4   HLA Typing Using Bead-Based Methods          55
3.1.6. Data Analysis: One    1. Data analysis for the HLA Class I and II assays are performed
Lambda HLA Fusion™              with the software package HLA Fusion™. The LABType®
                                analysis feature of the program analyses Luminex .csv output
                                files and is based on catalogue specifications provided with the
                                software. Import the data files as specified by the manufacturer
                                (see Note 28).
                             2. The analysis software establishes the HLA type by firstly cate-
                                gorising each of the beads via its internal fluorescence, then
                                determining whether a probe is bound or not by the strength of
                                external fluorescence on that bead produced by the SAPE. For a
                                particular sample, the bead/probe fluorescence for each of the
                                beads bearing a particular probe is visualised in the lower left-
                                hand quadrant of the software (Fig. 1) as a bar chart. Any probe
                                can be selected from this bar chart for further scrutiny. The
                                upper left quadrant shows the manufacturer’s QC panel reactiv-
                                ity with the selected probe. In this example, the QC for probe
                                A107 (Bead 512) is shown (see Note 29). The upper right
                                quadrant shows the probe reactivity for all samples in the run
                                being analysed (see Note 30). The lower right quadrant shows
                                the software analysis derived HLA type of the sample based on
                                the positive and negative probe reactions (see Note 31).
56         D. Trajanoski and S.J. Fidler
Fig. 1. HLA-Fusion® analysis software. Quadrant 1 shows the bead/probe fluorescence for each of the beads bearing a
particular probe. Any probe can be selected from this bar chart for further scrutiny. Quadrant 2 shows the manufacturer’s
QC panel reactivity with the selected probe (see Note 29). Quadrant 3 shows the probe reactivity for all samples in the run
being analysed (see Note 30). Quadrant 4 shows the software analysis derived HLA type of the sample based on the
positive and negative probe reactions (see Note 31). Courtesy of One Lambda, Inc. Used with permission.
                                     Table 5
                                     Gen-Probe LIFECODES components for amplification mix
3.2. Gen-Probe                       1. Select the appropriate SSO kit for the HLA locus to be typed.
LIFECODES HLA-SSO                    2. Allow the master mix (containing amplification primers) to
Typing Method                           thaw to room temperature (see Note 8).
3.2.1. DNA Amplification             3. Using Table 5, prepare the components for amplification for
                                        n + 2 reactions using the indicated amount of each component
                                        per reaction (except for DNA) (see Note 33).
                  4   HLA Typing Using Bead-Based Methods        57
 Table 6
 Gen-Probe LIFECODES thermal cycler conditions
 for amplification
                             14. Place the cover on the electrophoresis tank and run at 150 V
                                 for 20 min. Take out gel and place on UV transilluminator to
                                 inspect PCR products. Save photo and check all samples have
                                 correctly amplified (see Note 35).
3.2.2. Hybridisation             1. Warm probe mix in a 55–60°C heat block for at least 5–10 min
                                    to thoroughly solubilise components in probe mixture.
                                 2. Vortex briefly for about 15 s to thoroughly suspend the beads
                                    (see Note 18).
                                 3. Aliquot 5 μL of locus-specific PCR product from Subheading
                                    3.2.1 step 7 into a 96-well PCR plate (see Note 12).
                                 4. Aliquot 15 μL of corresponding probe mix for the locus-
                                    specific product into each well. When aliquoting probe mix to
                                    more than 10 wells, gently vortex probe mix after each set of
                                    ten. Seal plate with film.
                                 5. Hybridize samples under the incubation conditions indicated
                                    in Table 7.
                                 6. While the samples are hybridizing, prepare a 1:200 Dilution
                                    Solution/SAPE mixture. Combine 170 μL Dilution Solution
                                    (DS) and 0.85 μL 1 mg/mL SAPE per sample (follow Table 8
                                    and see Notes 15, 36, and 37).
                                 Table 7
                                 Gen-Probe LIFECODES thermal cycler conditions |for
                                 hybridization
                                 Table 8
                                 Gen-Probe LIFECODES SAPE and dilution solution volumes
Number of samples Dilution solution (DS) mLa SAPE stock volume (mL)b
                                  1                    170                      0.85
                                  5                    850                      4.25
                                 10                  1,700                      8.5
                                 20                  3,400                      17
                                 50                  8,500                      42.5
                             a
                              As provided in the Gen-Probe SSO genotyping kit
                                             4   HLA Typing Using Bead-Based Methods           59
                            7. At the 56°C hold step (i.e. step 4, Table 7), while the tray is on
                               the thermal cycler, dilute each sample with 170 μL of the
                               prepared DS/SAPE mixture. It is critical to dilute all samples
                               within 5 min (following the 10 min 56°C step i.e. step 3,
                               Table 7) (see Note 15).
                            8. Transfer contents of each well to a 96-well V-bottom microplate
                               (reading plate) using an 8-channel pipette. To avoid contami-
                               nation use fresh pipette tips each time.
                               NOTE: The Gen-Probe LIFECODES HLA-SSO assay requires
                               no centrifugation or wash steps.
                            9. Cover reading plate with seal and aluminium foil. Keep in dark
                               and at 4°C until ready to read using LABScan™ 100 Flow
                               Analyser (see Note 22).
3.2.3. Sample Acquisition   The procedure for sample acquisition for the Gen-Probe
on Luminex® Flow            LIFECODES SSO method is the same as that for the ONE
Analyser                    LAMBDA method. Follow Subheading 3.1.5 steps 1–17.
3.2.4. Data Analysis-Gen-   1. Data analysis for the HLA Class I and II assay is performed
Probe LifeMatch Quick-         with the software package LifeMatch™. The Quick-Type™
Type™                          analysis feature of the program analyses Luminex .csv output
                               files and is based on catalogue specifications provided with the
                               software. Import the data files as specified by the manufacturer
                               (see Note 38).
                            2. The analysis software establishes the HLA type by firstly cate-
                               gorising each of the beads via its internal fluorescence, then
                               determining whether a probe is bound or not by the strength
                               of external fluorescence on that bead produced by the SAPE.
                               The probe reactivity thresholds and probe assignments for an
                               individual sample are shown in the table in section A (Fig. 2).
                               In the example shown in Fig. 2, for probe 331 the positive and
                               negative thresholds are 0.624 and 0.511, respectively. For the
                               sample tested, for probe 331 the fluorescence observed is 0.992
                               which is greater than the positive threshold. This is scored
                               positive. An assignment can be changed from positive to nega-
                               tive or vice versa in this table (see Notes 30 and 39). The drop-
                               down in section B allows selection of an individual probe. In
                               this section, the probe reactivity for all samples in the run under
                               analysis can be viewed. In the example in Fig. 2, probe 202 gave
                               a fluorescence above the positive threshold for three of the sam-
                               ples displayed. Section C shows the selected probe reactivity for
                               all historical samples. These local data can be used to allow the
                               user to modify the cut-offs as required. The final HLA type is
                               displayed in section D, including any results from other analyses
                               of the same sample. In this example, the final HLA type is A*01,
                               A*02. The suggested allelic combination for an individual sample
                               is shown in section E. In this example, possible combinations
60         D. Trajanoski and S.J. Fidler
Fig. 2. LIFECODES® Quick-Type™ analysis software. Section A shows the probe reactivity thresholds and probe
assignments for an individual sample. An assignment can be changed from positive to negative or vice versa in this
table (see Notes 30 and 39). The drop-down in section B allows selection of an individual probe. In this section, the
probe reactivity for all samples in the run under analysis can be viewed. Section C shows the selected probe reactivity
for all historical samples. The final HLA type is displayed in section D, including any results from other analyses of the
same sample. The suggested allelic combination for an individual sample is shown in section E. Courtesy of Gen-
Probe, Inc. Used with permission.
4. Notes
Acknowledgments
References
 1. Claas FH, van der Poel JJ, Castelli-Visser R, Pool J,     4. Olerup O (1990) HLA class I typing by digestion
    Chen RB, Xu KY, van Rood JJ (1985) Interaction               of PCR-amplified DNA with allele specific restric-
    between des-Tyr1-gamma-endorphin and HLA                     tion endonucleases. Tissue Antigens 36:83–87
    class I molecules: serological detection of HLA-          5. Sayer D, Whidborne R, Brestovac B et al (2001)
    A2 subtype. Immunogenetics 22:309–314                        HLA-DRB1 DNA sequencing based typing: an
 2. Kojima S, Inaba J, Yoshimi A et al (2001)                    approach suitable for high throughput typing
    Unrelated donor marrow transplantation in chil-              including unrelated bone marrow registry
    dren with severe aplastic anaemia using cyclo-               donors. Tissue Antigens 57:46–54
    phosohamide, anti-thymocyte globulin and total            6. Olerup O, Zetterquist H (1992) HLA-DR typ-
    body irradiation. Br J Haematol 114:706–711                  ing by PCR amplification with sequence-
 3. Saiki RK, Scharf S, Faloona F et al (1985)                   specific primers (PCR-SSP) in 2 hours: an
    Enzymatic amplification of beta-globulin                     alternative to serological DR typing in clinical
    genomic sequences and restriction site analysis              practice including donor-recipient matching in
    for diagnosis of sickle cell anaemia. Science                cadaveric transplantation. Tissue Antigens
    230:1350–1354                                                39:225–235
                                                                                            Chapter 5
Abstract
Sequencing-based typing is a high resolution method for the identification of HLA polymorphisms. The
majority of HLA Class I alleles can be discriminated by their exon 2 and 3 sequence, and for Class II alleles,
exon 2 is generally sufficient. There are polymorphic positions in other exons which may require additional
sequencing to exclude certain alleles with differences outside exon 2 and 3, depending on the clinical
requirement and relevant accredition guidelines. The process involves selective amplification of target
alleles by PCR, agarose gel electrophoresis of the PCR products to assess the quantity and quality, followed
by purification of PCR amplicons to remove excess primer and dNTPs. Cycle sequencing reactions using
Applied Biosystems™ BigDye® Terminator Ready Reaction v1.1 or v3.1 Kit are performed, then purification
of sequence reactions before electrophoresing using Applied Biosystems™ 3730 or 3730XL Genetic
Analyser (or similar). Data is processed by specialised software packages, which compare the sample
sequence to the sequences of all possible theoretical allele combinations to assign an accurate genotype.
Examination of all nucleotides, both at conserved and polymorphic positions enables the direct identification
of new alleles, which may not be possible with techniques such as SSP and SSO typing.
1. Introduction
1.1. Amplification              Sequencing is carried out using a PCR product as the template.
of Target Region                The PCR amplifies a specific region of the gene/locus of interest,
Using PCR                       which is then sequenced from both directions to generate a high-
                                quality DNA sequence. Bi-directional sequencing is required to
                                achieve the desired level of accuracy and confidence in the base
                                calls at critical positions throughout the length of the sequence
                                (see Fig. 1 for overview).
                                     Two of the most common strategies for designing PCR methods
                                for sequencing-based typing (SBT) include:
                                (a) Designing primers with a sequencing “tail” or “tag” (Fig. 2).
                                    The most common tail (or tag) sequence is M13, which was
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_5, © Springer Science+Business Media New York 2012
                                                          67
68         L.K. Smith
Fig. 3. Sequencing with internal sequencing primers and tailed PCR primers.
2. Materials
2.2. PCR Amplification    1. CG-HLA Sequence Based Typing Kits™ (see Table 1 for
of Target DNA                components).
and Agarose Gel           2. Sterile molecular grade or tissue culture grade deionised water.
Electrophoresis
                          3. Adjustable pipettes (manual or electronic, various volumes)
                             and aerosol resistant pipette tips.
                          4. Thermal Cycler with heated lid.
                             Conexio Genomics kits have been tested using the following
                             thermal cyclers: Applied Biosystems™ (by Life Technologies™)
                             GeneAmp PCR system 9700, Biorad DYAD™ (previously MJ
                             Research DNA Engine DYAD), and Eppendorf Mastercycler
                             Pro™. Use of other thermal cyclers will require validation by
                             the user.
                          5. 0.2-mL thin walled reaction tubes either in 8-tube strips or
                             96-well plates. Use those to suit your thermal cycler as not all
                             plates fit all thermal cyclers.
                          6. Strip lids to fit 96-well PCR plates.
                          7. Sterile 1.5-mL tubes.
                          8. Vortex mixer.
                          9. Sterile biological safety cabinet or hood.
                         10. Agarose gel electrophoresis apparatus.
                         11. 1% Agarose gel (molecular biology grade) in 0.5× TBE, con-
                             taining 0.1 g/mL ethidium bromide.
                         12. 0.5× TBE electrophoresis buffer, diluted from 10× TBE which
                             is prepared as follows:
Table 1
Conexio genomics HLA sequencing-based typing kit details
Kit     Catalogue number   No of tests   Reagent            Volume (mL)   Primer name                           Volume (mL)
HLA-A   XH-PD1.1-2(50)      50           DNA Pol            1 × 55        AEX1F, AEX1R, AEX2F, AEX2R, AEX3F,    1 × 110 of each
                                                                           AEX3R, AEX4F, AEX4R
                                         CG-HLA-A mix       1 × 880
        XH-PD1.1-2(100)    100           DNA Pol            1 × 110       AEX1F, AEX1R, AEX2F, AEX2R, AEX3F,    1 × 220 of each
                                                                           AEX3R, AEX4F, AEX4R
                                         CG-HLA-A mix       2 × 880
HLA-B   BS-PD2.1-2(50)      50           DNA Pol            1 × 55        BEX1F, BEX2F, BEX2R, BEX3F, BEX3R,    1 × 110 of each
                                                                            BEX4F, BEX4R
                                         CG-HLA-B mix       1 × 880
        BS-PD2.1-2(100)    100           DNA Pol            1 × 110       BEX1F, BEX2F, BEX2R, BEX3F, BEX3R,    1 × 220 of each
                                                                            BEX4F, BEX4R
                                         CG-HLA-B mix       2 × 880
HLA-C   HH-PD 3.2-2(50)     50           DNA Pol            1 × 55        CEX1F, CEX1R, CEX2F, CEX2R, CEX3F,    1 × 110 of each
                                                                           CEX3R, CEX4F, CEX4R, CEX5F, CEX5R,
                                         CG-HLA-C mix       1 × 880
                                                                           CEX6F, CEX6R, CEX7F, CEX7R, CEX8F
        HH-PD 3.2-2(100)   100           DNA Pol            1 × 1101      CEX1F, CEX1R, CEX2F, CEX2R, CEX3F,    1 × 220 of each
                                                                           CEX3R, CEX4F, CEX4R, CEX5F, CEX5R,
                                         CG-HLA-C mix       2 × 880
                                                                           CEX6F, CEX6R, CEX7F, CEX7R, CEX8F
 DRB1       HH-PD5.2-4(50)           50            DNA Pol                  1 × 18          DRB1EX2F, DRB1EX3R, DRB1EX3R,                      1 × 110 of each
                                                                                             RB-TG344-R
                                                   CG-HLA-DRB1 mix          1 × 920
            HH-PD5.2-4(100)         100            DNA Pol                  1 × 36          DRB1EX2F, DRB1EX3R, DRB1EX3R,                      1 × 220 each
                                                                                             RB-TG344-R
                                                   CG-HLA-DRB1 mix          2 × 920
 DQB1       PQ-PD6.2-1(50)           50            DNA Pol                  1 × 18          DQB1EX2F, DQB1EX2R, DQB1EX3F.                      1 × 110 of each
                                                                                             DQB1EX3R
                                                   CG-HLA-DQB1 mix          1 × 920
            PQ-PD6.2-1(100)         100            DNA Pol                  1 × 36          DQB1EX2F, DQB1EX2R, DQB1EX3F.                      1 × 220 of each
                                                                                             DQB1EX3R
                                                   CG-HLA-DQB1 mix          2 × 920
The PRE-PCR component of each kit consists of a vial/s of a locus-specific PCR mix (e.g. CG-HLA-A mix) consisting of PCR buffer, dNTPs, MgCl2, locus-specific PCR
primers, and a single vial of DNA polymerase (DNA Pol). The POST-PCR kit contains sequencing primers (e.g. AEX1F)
                                                                                                                                                                    5
                                                                                                                                                                    HLA Typing by Direct DNA Sequencing
                                                                                                                                                                    73
74        L.K. Smith
                            Weigh each of the reagents and place into a clean sterile 2-L
                            conical flask. Dissolve in 1.2 L of Milli-Q water with continuous
                            mixing on a magnetic stirrer. When dissolved make up to 2 L
                            with Milli-Q water. Place buffer into 2 × 2 L clean sterile auto-
                            clavable containers. The buffer is divided into two containers
                            so as not to spill over during autoclaving. Autoclave to sterilise.
                            Store in the dark.
                        13. Electrophoresis loading buffer. Prepare as follows:
                            Place 20 mL of Milli-Q water into a clean sterilised glass
                            beaker; add 8 g of sucrose and 0.05 g Bromophenol Blue. Mix
                            using a magnetic stirrer. Once dissolved, make the solution up
                            to 160 mL with Milli-Q water. Store at 4°C for immediate use,
                            or for long term storage at −20°C.
                        14. PCR Marker (“Lambda ladder”) suitable to cover range of
                            300–1,300 bp (e.g. TrackIt 100 bp DNA ladder or TrackIt
                            1 Kb Plus DNA ladder, from Invitrogen™).
                        15. BIO-RAD™ GEL DOC 2000 Transilluminator or similar.
3. Methods
3.1. PCR Amplification     1. For each locus set up one reaction for each sample being
of Target DNA (17)            amplified (see Note 3). Include appropriate positive controls
                              of known genotype (see Note 4) and at least one negative con-
                              trol (see Note 5) for each group of samples being amplified.
                           2. CG-HLA Sequence Based Typing Kits™ are supplied as sepa-
                              rate aliquots of primer mix and polymerase. The polymerase
                              needs to be added to the primer mix just prior to setting up
                              the PCR reactions. Thaw the required number of vials of the
                              appropriate PCR Mix. For example, for a run of 15 samples
                              including controls, make up enough primer/polymerase mix
                              for 16 samples. This allows for slight pipetting inaccuracies
                              which can be due to a small amount of the solution adhering
                              to the outside of the tips. Once thawed, vortex briefly to mix
                              components.
                           3. Calculate the number of samples to be tested and dispense the
                              required amount of PCR mix and DNA polymerase into a
                              sterile tube. Refer to Table 2 for volumes required per sample.
                              Vortex the solution three to four times, for approximately 1 s
                              each time.
                           4. Dispense 17 mL of the primer/polymerase mix prepared in
                              step 3 above into each reaction well of either a 96-well plate or
                              8-tube strips.
                           5. Add 3 mL of sample DNA or appropriate positive control DNA
                              to each reaction well. Add 3 mL of sterile water to the negative
                              control reaction well.
 Table 2
 PCR mastermix volumes—per sample
Locus HLA-A (mL) HLA-B (mL) HLA-C (mL) HLA-DRB1 (mL) HLA-DQB1 (mL)
                              8. When the PCR is completed, remove the plate from the ther-
                                 mal cycler and either proceed directly to gel electrophoresis or
                                 store at 4°C until required. Purification of amplicons by
                                 ExoSap-IT® treatment (see Subheading 3.3) should occur
                                 within 24 h of completion of PCR.
 Table 3
 Expected number of PCR products and respective sizes for each locus
3.3. Purification   PCR products contain excess primers and unconsumed dNTPs
of PCR Products     which require removal before sequencing reactions can be per-
for Sequencing      formed. They may interfere with the efficiency of the sequencing
                    reaction, or can result in background “noise” in the sequence.
                    There are a number of options for purifying PCR products: Spin
                    columns, enzyme digestion, or magnetic bead-based technology.
                    Purification systems other than ExoSAP-IT® (e.g. Agencourt®
                    AMPure® XP or column-based systems) can be used to purify these
                    PCR products. It is strongly recommended that users validate these
                    procedures before proceeding. If ExoSAP-IT® is used; it is recom-
                    mended that users follow the procedure described below (17).
                     1. Prepare a mastermix consisting of 4 mL of ExoSAP-IT® and
                        8 mL of 2 mM MgCl2 per sample. Dispense 12 mL of the
                        mastermix into each PCR sample to be sequenced. (This is
                        determined by the band intensities seen following agarose gel
                        electrophoresis as described in Subheading 3.2. Very weak
                        products are not suitable for sequencing and therefore do not
                        need to be treated with ExoSAP-IT®.) Seal the tubes or plates,
                        vortex or place on shaker for 2 min, and centrifuge as described
                        in Subheading 3.1, step 6 to ensure all reagents are at the
                        bottom of the wells, before placing into the thermal cycler.
                     2. Run the thermal cycler according to the following protocol:
                       37°C-30 min
                       80°C-15 min
                       4°C-hold
                     3. Upon completion, dilute the purified product 1:8 with sterile
                        water. This dilution step will ensure that there is sufficient
                        template to perform the sequencing reactions and ensure that
                        the concentration of the template is sufficient to produce good
                        quality sequence data. Note that weaker PCR products may
                        require a lower dilution factor than 1:8.
                     4. ExoSAP-IT® treated samples may be stored at 4°C until ready
                        for use.
78       L.K. Smith
 Table 4
 Sequencing primers provided for use with each locus
                                          96°C for 10 s
                                          50°C for 5 s             25 cycles
                          Table 5
                          Data collection settings for ABI 3730 or 3730xl
Parameter Setting
3.7. Allele Assignment   Sequence analysis may be performed using Assign SBT™ or Assign
                         ATF™ software. For more details, please refer Chapter 6 or the
                         Conexio Genomics Pty Ltd. Web site (http://www.conexio-
                         genomics.com). See Notes 16–19 for some critical issues in allele
                         assignment.
4. Notes
                96°C    for 10 s
                50°C    for 5 s          25 cycles
                60°C    for 4 min
                Then 4°C indefinitely
References
 1. Applied Biosystems (2000) Automated DNA           8. Voorter CE, Kik MC, van den Berg-Loonen
    sequencing chemistry guide. © Copyright              EM (1998) High-resolution HAL typing for
    2000, Applied Biosystems                             the DQB1 gene by sequence-based typing.
 2. Cereb N, Maye P, Lee S, Kong Y, Yang SY              Tissue Antigens 51(1):80–87
    (1995) Locus-specific amplification of HLA        9. Van der Vlies SA, Voorter CE, van den Ber-
    class I genes from genomic DNA: locus-specific       Loonen EM (1998) A reliable and efficient
    sequences in the first and third introns of          high resolution typing method for HLA-C
    HLA-A, -B and -C alleles. Tissue Antigens            using sequence-based typing. Tissue Antigens
    45(1):1–11                                           52(6):558–568
 3. Petersdorf EW, Hansen JA (1995) A compre-        10. Kurz B, Steiert I, Heuchert G, Muller CA
    hensive approach for typing the alleles of the       (1999) New high resolution typing strategy for
    HLA-B locus by automated sequencing. Tissue          HLA-A locus alleles based on dye terminator
    Antigens 46(2):73–85                                 sequencing of haplotypic group-specific PCR-
 4. Blasczyk R, Wehling J, Weber M, Salama A             amplicons of exon 2 and exon 3. Tissue Antigens
    (1996) Sequence analysis of the 2nd intron           53(1):81–96
    revealed common sequence motifs providing        11. Kotsch K, Wehling J, Blasczyk R (1999)
    the means for a unique sequencing based              Sequencing of HLA class II genes based on the
    typing protocol of the HLA-A locus. Tissue           conserved diversity of the non-coding regions:
    Antigens 47(2):102–110                               sequencing based typing of HLA-DRB genes.
 5. Cereb N, Yang SY (1997) Dimorphic primers            Tissue Antigens 53(5):486–497
    derived from intron 1 for use in the molecular   12. Sayer D, Whidborne R, Brestovac B, Trimboli
    typing of HLA-B alleles. Tissue Antigens             F, Witt C, Christiansen F (2001) HLA-DRB1
    50(1):74–76                                          DNA sequencing based typing: an approach
 6. Kotsch K, Wehling J, Kohler S, Blasczyk R            suitable for high throughput typing including
    (1997) Sequencing of HLA class I genes based         unrelated bone marrow registry donors. Tissue
    on the conserved diversity of the noncoding          Antigens 57(1):46–54
    regions: sequencing-based typing of the HLA-A    13. Sayer DC, Whidborne R, De Santis D,
    gene. Tissue Antigens 50(2):178–191                  Rozemuller EH, Christiansen FT, Tilanus MG
 7. Voorter CE, de Bruyn-Geraets D, van den              (2004) A multicenter international evaluation
    Berg-Loonen EM (1997) High-resolution                of the single-tube amplification protocols for
    HLA typing for the DRB3/4/5 genes by                 sequencing-based typing of HLA-DRB1 and
    sequence-based typing. Tissue Antigens 50(3):        HLA-DRB3,4,5. Tissue Antigens 63(5):
    283–290                                              412–423
86       L.K. Smith
14. Dunn PP, Day S, Williams S, Bendukidze N            HLA Class I and II Loci, Version No: 6.0,
    (2005) HLA-DQB1 sequencing-based typing             Feb 2011
    using newly identified conserved nucleotide     18. Applied     Biosystems     (2002)    BigDye®
    sequences in introns 1 and 2. Tissue Antigens       Terminator v3.1 cycle sequencing kit protocol.
    66(2):99–106                                        © Copyright 2002, Applied Biosystems
15. Van Dijk A, Melchers R, Hilkes Y, Rozemuller    19. Applied Biosystems (2002) 3730/3730xl DNA
    E, Tilanus M (2007) HLA-DRB sequencing-             analyzers user guide. © Copyright 2002.
    based typing: an improved protocol which            Applied Biosystems
    shows complete DRB exon 2 sequences and         20. Rozemuller E (2000) SBT resolution and ambi-
    includes exon 3 of HLA-DRB4/5. Tissue               guities (Chapter 6). In: Tilanus MGJ, Hansen
    Antigens 69(Suppl 1):61–63                          JH, Hurley CK (eds) IHWG technical manual
16. Applied Biosystems (2002) 3730/3730xl DNA           genomic analysis of the human MHC: DNA-
    analyzers sequencing chemistry guide. ©             based typing for HLA alleles and linked
    Copyright 2002, Applied Biosystems                  Polymorphisms. Publication of the 13th
17. Conexio Genomics Pty Ltd (2011) CG-HLA              International    Histocompatibility   Working
    sequence based typing kits™ Instructions for        Group. Fred Hutchinson Cancer Research
    Use, PCR amplification and sequencing of            Centre, Seattle, USA
                                                                                            Chapter 6
Abstract
DNA Sequencing is now a standard frontline high-throughput HLA typing procedure with some
unrelated bone marrow donor registry typing laboratories performing tens of thousands of tests per
year. The advantage of DNA sequencing is that, by definition, sequencing directly identifies all bases in
the DNA template. Alternative molecular-based assays such as the use of sequence-specific PCR primers
(PCR-SSP) and oligonucleotide probes (PCR-SSO) provide information only for those regions to which
the oligos are designed and no information is obtained for the regions between primers and probes.
     The era of routine high-throughput sequencing-based typing (SBT) was made possible by the devel-
opment of locus-specific PCR-based assays and the development of the HLA sequencing-based typing
software, Assign-SBT v3.2.7 by Conexio Genomics. A single PCR per locus simplified the template prepa-
ration stage of the test and Assign-SBT simplified the sequence analysis and allele assignment stage.
Together these developments dramatically simplified the SBT procedure, making SBT cost effective.
     This chapter provides a comprehensive description of Assign-SBT sequence analysis software for use
in a HLA typing laboratory.
Key words: High throughput DNA sequencing, Sequence analysis, Assign-SBT, HLA
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_6, © Springer Science+Business Media New York 2012
                                                          87
88   C. Wirtz and D. Sayer
2. Getting Started
and Using
the Software
                          Assign-SBT v3.6+ is a standalone computer software program and
2.1. Installation         should be installed on the computer on which SBT analysis is
                          performed. It is recommended that Assign is installed by a user
                          with complete administrator access to the computer. The installer
                          package can be acquired by contacting Conexio via their website,
                          http://www.conexio-genomics.com. It is also helpful if the com-
                          puter has access to the Internet to facilitate the system updates
                          with new libraries and other files as needed.
                           1. To install: Double click on the installer file icon and follow the
                              instructions for installation. During installation, a 16-digit
                              computer-specific hardware identifier will be generated. Copy
                              and paste this ID into an e-mail and send to key@conexio-
                              genomics.com to obtain licence keys. The software will not be
                              functional until the key files are successfully installed.
                           2. Reference libraries and NMDP codes, if required, need to be
                              installed. Go to the Conexio Genomics website, http://www.
                              conexio-genomics.com, and click on the Downloads tab on
                              the left side of the screen. Click on the Libraries tab. Click
                              on the library version that you require. A zip file will be down-
                              loaded to your computer. Unzip the file and save the References
                              folder to the desktop or other convenient location. The
                              References folder will contain the gene-specific reference files,
                              P Group file, and G Group file.
                              (a) Next, launch the software. Use the default operator (admin)
                                  and password (cg01) login. Click on Help | Update
                                  System on the top menu bar.
                              (b) Click on the References button and navigate to the
                                  unzipped References folder. Highlight the .xml reference
                6   Data Analysis of HLA Sequencing Using Assign-SBT v3.6+ from Conexio            91
Fig. 1. The Update screen is used to update Keys, Reference files, and NMDP Codes.
2.2. Login and Adding            1. Launch the software by double clicking on the Assign 3.6+
Users                               icon on the desktop. The default operator is “admin” and the
                                    default current password is “cg01”. It is recommended that
                                    the admin password is never changed.
                                 2. Additional users can be added. Enter the admin login and
                                    password then click “More”. Below will be a section to add
                                    additional users. Type in the new operator’s name in the Edit
                                    Operator section. Type in a password for that user. Re-type the
                                    password. Select the Operator Level. Click Add/Update
                                    directly next to the Retype Password box. Repeat for addi-
                                    tional users (Fig. 2).
92         C. Wirtz and D. Sayer
Fig. 2. The Operator Login screen is used to add/edit users and select Settings files.
2.3. Settings              The settings menu enables the user to configure the software for
                           their requirements. Settings can be saved as different settings files
                           to enable the software to be individualized for different users.
                            Fig. 3. The screen display is modified by selecting a colour scheme customized for
                            the user.
2.4. Naming                 In the Advanced tab, the user can enter the parameters that define
Conventions                 the sample name and the locus identifier in sample sequence
                            filename. The sequence filename must be unique for sequencing
                            reaction for a sample and contain the sample name and an identifier
                            that can be used as an alias for the locus being genotyped. If a stan-
                            dard system is used by the operator, analysis will occur automati-
                            cally and different data from different loci can be entered into the
                            same layout (Fig. 4).
2.4.1. Sample Delimiters    Sample Delimiters have been used to separate the components
                            of the sequence-file name.
                                Example:
                                 Sequence-file name A01[12345_DQ2F_A01
                                 Delimiters have been used to separate the components of the
                                 sequence-file name
                                     [ has been used to separate the PCR number (A01) and
                                     the sample name (12345).
                                     _ has been used to separate the sample name and the locus
                                     (HLA-DQB1) and primer (exon 2 Forward).
                                     _ has also been used to separate the locus and well location
                                     (A01).
                                 Set the Naming convention by defining the Sample
                                 Delimiters.
                                In the example above, the sample name begins with [ and
                            the sample name ends with _Enter [ in the Start string box, and
                            enter _ in the End string box.
94         C. Wirtz and D. Sayer
Fig. 4. Settings | Advanced tab is used to customize sample naming conventions and reference aliases, select Nomenclature
output naming, CWD Allele set, and to indicate if genomic references are to be used.
2.4.2. Alias Names               1. In the example above, DQ is used as a symbol for HLA-DQB1.
                                    In Subheading 2.4.7, use the Ref: drop down menu to select
                                    the locus. Next, in the Alias drop down menu select the alias
                                    used (DQ). If the alias is not present, type it into the Alias box,
                                    and then click Update directly to the right of the Ref box.
                                 2. Repeat this for each locus alias you will be using. After all aliases
                                    have been added, click on the Update button in the lower
                                    right-hand corner of the Settings box.
2.4.3. Nomenclature             The HLA nomenclature standards changed in April 2010 from v2
                                to v3.0. In Select the Output Naming Standard, either IMGT/
                                HLA3.0 or 2.0 can be selected for the naming convention of the
                                reported alleles.
                              method used, the DNA Sequencer make and model, and the polymer
                              in the sequencer capillaries.
                               1. Enter the number of base pairs between the end of the primer
                                  and the first usable sequence generated in the Start Gap.
2.4.5. CWD Alleles            The CWD alleles are stored in a text file. A default set of CWD
                              alleles based on those described by Cano et al. (1) are provided
                              with the software. However, laboratories may want to create their
                              own list.
                               1. Set the CWD allele set you will be utilizing, if any.
2.4.6. Extended Analysis      By selecting Genomic References, the sequence analysis will
                              enable the comparison of the sample sequence with the Genomic
                              References provided by the IMGT/HLA database. This analysis
                              improves typing accuracy by enabling those alleles that are charac-
                              terized by polymorphisms to be genotyped. By leaving the
                              Genomic References box unchecked, analysis will be performed
                              against the standard cDNA reference.
                               1. If the laboratory will utilize analysis of the non-coding regions,
                                  put a check in the Use Genomic References box.
2.4.7. Reference Aliases      Reference aliases are used in the sequence filenames to indicate the
                              HARP being used for sequencing.
                               1. Click on the References tab of the Setting menu to establish
                                  alias names for primers used.
                               2. Click on the Load Reference box at the top right of the screen.
                                  Navigate to the References folder and click on the reference
                                  needed. Click OK.
                               3. The reference information will populate the screen.
                               4. To establish a reference alias for the Codon 86 resolution
                                  primer, highlight the DRB1.xml file and click OK.
                               5. In the References tab, in the lower left corner drop down
                                  menu, select the Codon 86 primer. To the right of the primer
                                  will be the alias drop down menu. Click the drop down menu
                                  to determine if your naming alias is present.
                               6. To add an alias, type the alias into this box. For example, DR86
                                  as an alias for the Codon 86 GTG primer. Next, click on Add
                                  Alias directly below the alias drop down menu box. Then, click
                                  on Update in the lower right corner. After all aliases have been
                                  added, click on Done.
2.4.8. Variant Positions      Variant Positions is a tool to draw attention to sequence artefacts
                              that may result in base call errors, or other positions within a
                              sequence where automated base calls may be frequently incorrect.
96         C. Wirtz and D. Sayer
Fig. 5. User Defined Variant positions are established using the References tab in the Update Settings menu.
                            Fig. 6. The Import Files menu is accessed by selecting File | Import | Electropherograms
                            from the top menu bar.
2.6. The Screen             Once the sequence data has been imported, the software screen
Layout, Editing,            becomes populated with sample, sequence, and result information.
and Analysis                There is a predominance of white to red shading used to demon-
                            strate sequence data quality.
98         C. Wirtz and D. Sayer
Fig. 7. Shading within the electropherogram coverage indicates quality of given sequences.
2.6.1. Sample ID Pane          The screen layout shows information for a particular sample.
                               It includes the sample ID, the electropherograms data, the aligned
                               sequences for a sample, and the best-matched allele combina-
                               tions (Fig. 8).
                                    The samples imported are listed on the left side of the screen.
                               Sample names are colour coded to indicate data quality.
                                 1. The sample pane also includes five columns of boxes.
6   Data Analysis of HLA Sequencing Using Assign-SBT v3.6+ from Conexio                      99
Fig. 8. The Sample ID Pane shows information about the samples in the project.
Fig. 10. Sequence Electropherogram information is contained in the Sample layout screen, including the structure of
the gene being sequenced, sample sequence alignments, consensus sequences, and sample sequence data.
Fig. 11. The structure of the gene being sequenced is represented at the top of the screen.
Fig. 12. The sample sequence alignment shows sequence quality shaded within each electropherogram. Lighter electro-
pherograms represent better quality sequence.
Fig. 13. Library Consensus sequence is colour coded depending on region. This sequence represents all alleles within the
reference library.
Fig. 14. Sample Consensus Sequence of the sample being sequenced appears just below the Library Consensus sequence,
just above the electropherogram tracings.
Fig. 15. Sample sequence data, software base calls, and quality indicators are represented within the electropherogram
tracing.
Fig. 17. Amino Acid sequence can be viewed instead of base pair sequence by pressing Ctrl + A in the electropherogram
pane.
2.6.3. Results Pane            The Results pane includes the sample name, the start and stop
                               positions of the test sequence, and the allele combination with the
                               best-matched sequence to the test sequence. Bolded alleles are
                               CWD alleles whilst those unbolded are and rare alleles (Fig. 19).
                                1. Allele 1 and Allele 2 columns display the allele pairs best
                                   matched to the test sequence.
                6   Data Analysis of HLA Sequencing Using Assign-SBT v3.6+ from Conexio                      105
                               Fig. 18. Coloured boxes above the Sample Consensus sequence indicate points of
                               interest in the sequence.
Fig. 19. The Results pane includes sample name, start/stop positions, and best-matched allele combination.
Fig. 20. Non-coding and Indel data are viewed in the N-C and IND columns, respectively.
                               Fig. 21. The Navigator enables editing, review, and movement from sample to sample
                               within the project.
2.7. Navigator                 The navigator enables sequence editing, moving between samples,
                               and moving between positions within a sequence. Importantly, the
                               Navigator is used to Validate automated base calls (Fig. 21).
             6   Data Analysis of HLA Sequencing Using Assign-SBT v3.6+ from Conexio      107
Fig. 22. View options allows the data and reference library data to be viewed in different ways.
2.9. View Options               The View options enable the samples sequence data and the
                                sequence data from the alleles in the library to be viewed in differ-
                                ent ways (Fig. 22).
                                  1. Status Bar shows the status of the project at the bottom of the
                                     screen.
                                  2. Panes
                                     (a) Electropherogram is the default and displays the electro-
                                         pherogram tracings of the sample.
                                     (b) Consensus displays the consensus sequence for all samples
                                         in a project.
                                     (c) Quality displays the consensus sequence shaded according
                                         to the consensus sequence BCS for each base for every
                                         sample in the project.
                                     (d) Alignments displays the consensus sequence for each of the
                                         possible allele combinations for a given sample. Mismatches
                                         with the sample consensus appear highlighted in yellow.
                6    Data Analysis of HLA Sequencing Using Assign-SBT v3.6+ from Conexio        109
2.10. Data Analysis          Log into the Assign 3.6+ software and select the settings file desired
and Editing EPGs             by clicking on Edit then Settings. Select the settings file and
                             click Done.
2.10.1. Logging on
2.10.2. Importing Data        1. Import the desired dataset using either the Browse option to
                                 import an entire folder of data or the Select Files Manually to
                                 selectively import files.
                              2. Imported samples will be displayed as a list on the left side of
                                 the screen.
                              3. The Electropherogram data will be in the centre of the screen.
                              4. The Allele assignments for the active (highlighted in blue)
                                 sample will be displayed at the right side of the screen.
110        C. Wirtz and D. Sayer
2.10.3. Navigation          1. The Navigator box is used to navigate through the data
                               checking the critical bases.
                            2. Set the desired bases to be audited in the Navigator box: BCS,
                               Edits, MM, and variant positions.
                            3. Highlight the sample to be reviewed by clicking on it in the left
                               sample pane.
                            4. Navigate to the first base in the sample by clicking on the left
                               arrow with the bar in the navigator.
                            5. Using the red X button, navigate through the sequence verify-
                               ing all desired bases. As each base is confirmed, the red X will
                               change to a green check and the cursor will move to the next
                               base to be verified. A green box will appear above each base
                               pair that has been verified using this method.
                               (a) There should be at least one allele pair in the Results Pane
                                   on the right that indicates no mismatches in the MM0
                                   column.
                               (b) Once the Master Layer has been reviewed, the Navigator
                                   will take the user to any resolution primer layers that are
                                   present. These layers must also be reviewed before the
                                   sample analysis is complete.
                               (c) Once all priority review bases have been verified, the red X
                                   will change to a green check mark indicating no additional
                                   bases need confirming. In the Sample Pane, the box under
                                   the A column (Audit) will turn green.
                               (d) Clicking on the box under the 1 column will indicate the
                                   sample has been reviewed once.
                               (e) At this point, the project should be saved to prevent any
                                   accidental loss of data review. Click on File then Save As.
                                   Select a file name and location to save the project. The
                                   saved project (.xml format) indicates which electrophero-
                                   grams were used including their saved location, any edits
                                   and confirmations that were done, and information about
                                   the user.
                                   (f) If Genomic References were selected, the N-C and IND
                                       column may be present. Refer to the differences column to
                                       determine if analysis of the non-coding region would be
                                       beneficial. Click on the N-C column to highlight it in yellow.
                                       Navigate to the mismatch positions and make any edits
                                       necessary. Once all review and edits have been made, click
                                       back on the MM0 column to view the allele pairs.
                 6   Data Analysis of HLA Sequencing Using Assign-SBT v3.6+ from Conexio              111
Fig. 24. The MM2 column indicates the presence of a resolution primer to resolve heterozygous ambiguities.
Fig. 25. The Resolving layer is activated to allow review of resolution primer data.
2.10.4. Resolution Primer         1. HARPs are used to resolve heterozygous ambiguities within
Layers                               the regions sequenced. HARPs target one of the alleles present,
                                     creating a hemizygous sequence. In the example below,
                                     multiple ambiguities exist after analysing the master sequence
                                     as indicated in the MM0 column highlighted in yellow.
                                     Heterozygous sequence is present (Fig. 24).
                                  2. Selection of an HARP that will detect only one of the alleles in
                                     the pair will result in a hemizygous sequence in the MM1 layer.
                                     This will eliminate some of the heterozygous ambiguities.
                                     Allele combinations with no mismatches in the MM0 and
                                     MM1 column will be included in the report. Alleles with mis-
                                     matches in the MM1 column have been eliminated by the
                                     HARP sequence (Fig. 25).
2.10.5. Auditing                 All user interaction is logged in the audit trail. The software logs
                                 time and date any edit was made, when the project was saved and
                                 the user performing the action. This information can be printed on
                                 the allele report along with the sample allele assignments.
                6    Data Analysis of HLA Sequencing Using Assign-SBT v3.6+ from Conexio         113
2.11.1. Genotyping Report      The Genotyping report is used to report the allele combinations that
                               have identical sequence to the sequence of the sample (Fig. 26).
                                 1. The Full Report Section enables laboratories to create their
                                    report format. The Sample drop down menus enable the lab to
                                    include or exclude specific items from the report.
                                    (a) The Sample section contains the Auditing, Match Summary,
                                        G Groups, and P Groups reports.
                                    (b) Match Summary will list all the possible allele pair combi-
                                        nations. If the CWD option is selected, the CWD alleles
                                        will appear in bold type in the report (Table 1).
114   C. Wirtz and D. Sayer
                       Table 1
                       Match summary option listing all possible allele pair
                       combinations
                       Sample                      Q950050964
                       Reference                   IMGT/B 3.3.0 2011-01-14
                       The allele pairs listed below are compatible with the consensus sequence
                       B*18:01:01                  B*44:02:01:01
                       B*18:01:01                  B*44:02:01:02S                  Intron 4
                       B*18:01:05                  B*44:02:01:01                   Exon 2
                       B*18:01:05                  B*44:02:01:02S                  Exon 2, Intron 4
                       B*18:01:05                  B*44:27:01                      Exon 2
                       B*18:09                     B*44:09                         Exon 2
                       B*18:12                     B*44:12                         Exon 2
                       B*18:20                     B*44:51                         Exon 3
                       B*18:43                     B*44:55                         Exon 2
                      CWD alleles appear in bold
                Table 2
                G group report
                Sample                                 Q950050964
                Reference                              IMGT/B 3.3.0 2011-01-14
                The allele pairs listed below are compatible with the consensus sequence
                B*18:01:01G                            B*44:02:01G
                B*18:09                                B*44:09
                B*18:12                                B*44:12
                B*18:20                                B*44:51
                B*18:43                                B*44:55
                Table 3
                P group report
                Sample                    Q950050964
                Reference                 IMGT/B 3.3.0 2011-01-14
                The allele pairs listed below are compatible with the consensus sequence
                B*18:01P                  B*44:02P
                B*18:09                   B*44:09
                B*18:12                   B*44:12
                B*18:20                   B*44:51
                B*18:43                   B*44:55
Fig. 27. The heterozygous ambiguity resolving primers (HARPs) report indicates which resolution primers are required
to resolve ambiguities.
2.11.2. HARPs Report            1. The HARPs report indicates to the operator which HARPs are
                                   required to resolve heterozygous ambiguities and which exons
                                   are required to be sequenced to resolve allele ambiguities
                                   (Fig. 27).
                                   (a) The Output Filters can be used to filter for a single sam-
                                       ple/locus or all samples/loci in a project.
                                   (b) The Start Gap under Parameters can be set depending on
                                       the gap between the end of the sequencing primer and
                                       usable data. This is generally around ten for POP6 users
                                       and longer for POP7 users.
                                   (c) The operator can choose between a Full Report and a G
                                       Groups Report under the Type section.
                                   (d) The Full Report will list the HARP resolution for each
                                       allele pair (Table 4).
                                   (e) The G Group Report will group the resolution based on
                                       the broader G Grouping of the allele pairs (Table 5).
                6    Data Analysis of HLA Sequencing Using Assign-SBT v3.6+ from Conexio         117
 Table 4
 Example of a full HARPs report listing resolution for each allele pair and HARPs
 required to resolve the ambiguities
 Sample                                   Q950050964
 Reference                                IMGT/B 3.3.0 2011-01-14
 Use ONE primer from group 1              C1-AC559-R-520            C1-TG539-R-507   C1-CT559-R-494
 Use ONE primer from group 2              C1-GAA309-R-953
 Splits                                   B*18:01:01                B*44:02:01:01
                                          B*18:01:01                B*44:02:01:02S   Intron 4
                                          B*18:01:05                B*44:02:01:01
                                          B*18:01:05                B*44:02:01:02S   Intron 4
                                          B*18:01:05                B*44:27:01       Exon 4
                                          B*18:09                   B*44:09
                                          B*18:12                   B*44:12
                                          B*18:20                   B*44:51
                                          B*18:43                   B*44:55
Unresolved ambiguities remain within exon 4, intron 4
 Table 5
 Example of a G group HARPs report
 Sample                                  Q950050964
 Reference                               IMGT/B 3.3.0 2011-01-14
 Use ONE primer from group 1             C1-AC559-R-520             C1-TG539-R-507   C1-CT559-R-494
 Use ONE primer from group 2             C1-GAA309-R-953
 Splits                                  B*18:01:01G                B*44:02:01G
                                         B*18:01:05                 B*44:02:01G
                                         B*18:09                    B*44:09
                                         B*18:12                    B*44:12
                                         B*18:20                    B*44:51
                                         B*18:43                    B*44:55
Unresolved ambiguities remain within exon 4, intron 4
Resolution is based on the broader G grouping of the allele pairs
118        C. Wirtz and D. Sayer
Fig. 28. The FASTA report enables sequences in FASTA text format to be produced.
2.11.3. FASTA                  The FASTA report enables sequences in FASTA text format to be
                               produced. Selecting the sample, locus, layer, group, and region
                               provides a detailed description of the FASTA file in the FASTA file
                               name (Fig. 28).
2.11.4. Quality Reports        The quality reports utilizes the BCS at each position to create qual-
                               ity control information for a sample, which can then be compared
                               between different samples to create quality control information for
                               the assay. The principle is that if the mean and standard deviation of
                               BCS for a number of sequence positions can be calculated, this will
                               provide a quality score for the region of sequence from which the
                               mean and standard deviation were calculated. This information can
                 6   Data Analysis of HLA Sequencing Using Assign-SBT v3.6+ from Conexio                            119
Fig. 29. The Quality Report utilizes the base caller score at each position to create a comparison between different sam-
ples, providing quality control information for the assay.
Fig. 30. The Quality Report shows the mean Base Call Score (BCS) represented by squares, and the standard deviation in
diamonds for all samples. The dots show the number of edits made for each sample.
3. Notes
4. Conclusion
References
Abstract
The non-classical human leukocyte antigen (HLA)-G plays a crucial role in the induction of tolerance at
the feto–maternal interface as well as in transplantation, cancer, inflammation, and autoimmune diseases.
To understand gene regulation and the impact of polymorphic sites on the function, simple and easy
feasible approaches are needed for the detection of HLA-G variants in coding and non-coding regions.
Here we summarize a set of methods for the identification of variants in the exon 2–4, in the 3¢ untrans-
lated region and in the gene promoter region of the HLA-G gene.
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_7, © Springer Science+Business Media New York 2012
                                                         123
124        H. Nückel et al.
Table 1
HLA-G polymorphisms in exon 1–4 and in 3¢UTR
Rel. to
ATG = 0        234     239 280        292       297     306      324      361        366       372      408      706       726    738 741
G*01:01:01 GCC TCC TAC ACG CAG CGG GCG CAG GAG CCG GCC CAC GGC GGG TCC
G*01:01:02 --- --- --- --- --- --- --- --- --- --A --- --T --- --- ---
G*01:01:03 --- --- --- --- --- --- --- --- --- --A --- --- --- --- ---
G*01:01:04 --- --- --- --- --- --- --- --- --- --A --T --- --- --- ---
G*01:01:05 --- --- --- --- --- --- --- --- --- --- --- --- --- --- ---
G*01:01:06 --- --- --- --- --- --- --- --- --- --- --- --- --- --- ---
G*01:01:07 --- --- --- --- --- --- --- --- --- --A --- --T --- --- ---
G*01:01:08 --- --- --- --- --- --- --- --- --- --A --- --- --- --- ---
G*01:01:09 --- --- --- --- --- --- --- --- --- --- --- --- --- --- ---
G*01:01:11 --- --- --- --- --- --- --- --- --- --- --- --- --- --- ---
G*01:01:12 --- --- --- --- --- --- --T --- --- --A --- --T --- --- ---
G*01:01:13 --- --- --- --- --- --- --- --- --- --A --- --T --- --- ---
G*01:01:14 --- --- --- --- --- --A --- --- --- --A --- --T --- --- ---
G*01:01:15 ---        ---     ---    ---        ---    ---      ---      ---        --A        ---      ---      ---      ---    ---   ---
7 Simple Methods for the Detection of HLA-G Variants in Coding and Non-coding Regions                                    125
748 755 778 814 871 902 394 1590 1659 1681 1734 1799 1827 3741
107 110 117 130 148 159 169 188 211 219 236 258 267
                                                                                                              Frequencies in
G     A     L/I   L/-    E       Y/H H       H      A         R/W/Q A            T/M    p                     Brazilians (2)
GGA CTC GCC CTG GAG TAC CAC CAC GCG CGG GCA ACG CCG −14 bp 39.8
--- --- --- --- --- --- --- --- --- --- --- --- --- +14 bp 19.9
--T --- --- --- --- --- --- --- --- --- --- --- --- +14 bp 5.34
--- --- --- --- --- --- --- *** *** *** *** *** *** *** 0.49
--T --- --- --- --- --- --- --- --- --- --- --- --- −14 bp 0
--- --- --- --- --- --- --- --T --- --- --- --- --- *** 0.97
--T --- --- --- --- --- --- --- --- --- --- --- --- *** 0
--- --- --- --- --- --- --- --- --- --- --- --- --- −14 bp 4.37
--- --- --- --- --A --- --- --- --- --- --- --- --- *** 0
--- --- --G --- --- --- --- --- --- --- --- --- --- *** 0.49
--- --- --- --- --- --- --- --- --- --- --- --- --- *** 0
--- --- --- --- --- --- --T *** *** *** *** *** *** *** 0
--- --- --- --- --- --- --- --- --- --- --- --- --- *** 0
---   ---   ---   ---    ---     ---   ---   ---    ---       ---         ---    ---    ---       ***         0
                                                                                                                  (continued)
126         H. Nückel et al.
 Table 1
 (continued)
 Rel. to
 ATG = 0        234     239 280        292       297     306      324      361        366       372      408      706       726    738 741
G*01:01:16 --G --- --- --- --- --- --- --- --- --A --- --T --- --- ---
G*01:01:17 --- --- --- --- --- --- --- --- --- --A --- --T --T --- ---
G*01:01:18 --- --- --- --- --- --- --- --- --- --A --- --T --- --- ---
G*01:01:19 --- --- --- --- --- --- --- --- --- --A --- --T --- --- ---
G*01:01:20 --- --- --- --- --- --- --- --- --- --- --- --T --- --- ---
G*01:02 --- --- --- --- --- --- --- -G- --- --- --- --- --- --- ---
G*01:03 --- --- --- T-- --- --- --- --- --- --- --- --- --- --- ---
G*01:04:01 --- --- --- --- --- --- --- --- --- --A --- --- --- --- ---
G*01:04:02 --- --- --- --- --- --- --- --- --- --A --- --- --- --- ---
G*01:04:03 --- --- --- --- --- --- --- --- --- --- --- --- --- --- ---
G*01:04:04 --- --- --- --- --- --- --- --- --- --A --- --- --- --- ---
G*01:04:05 --- --- --- --- --A --- --- --- --- --A --- --- --- --- ---
G*01:05N --- --- --- --- --- --- --- --- --- --A --- --T --- --- ---
G*01:06 --- --- --- --- --- --- --- --- --- --A --- --T --- --- ---
G*01:07 --- -T- --- --- --- --- --- --- --- --A --- --- --- --- ---
G*01:08 --- --- --- --- --- --- --- --- --- --A --- --T --- --- ---
G*01:09 --- --- --- --- --- --- --- --- --- --A --- --T --- --- ---
G*01:10 --- --- --- -T- --- --- --- --- --- --- --- --- --- --- ---
G*01:11 --- --- --- -T- --- --- --- --- --- --A --- --- --- --- ---
G*01:12 --- --- C-- --- --- --- --- --- --- --A --- --- --- --- ---
G*01:13N --- --- --- --- --- --- --- T-- --- --A --- --T --- --- ---
G*01:14 --- --- --- --- --- --- --- --- --- --A --- --- -A- --- ---
G*01:15 --- --- --- --- --- --- --- --- --- --A --- --- --- -T- ---
G*01:16 --- --- --- --- --- --- --- --- --- --A --- --T --- --- -G-
 G*01:17       ---     ---     ---    ---        ---    ---      ---      ---        ---        --A      ---      --T      ---    ---   ---
Nucleotide sequence variations observed in exon 1–4 of the coding region of HLA-G according to the IMGT (3) A
alanine; S serin; F phenylalanine; Y tyrosine; T threonine; M methionine; Q glutamine; R arginine; E glutamine;
Table 1 indicates SNP identified by the primer mentioned in Table 2
7 Simple Methods for the Detection of HLA-G Variants in Coding and Non-coding Regions                                       127
748 755 778 814 871 902 394 1590 1659 1681 1734 1799 1827 3741
107 110 117 130 148 159 169 188 211 219 236 258 267
                                                                                                                 Frequencies in
G     A      L/I    L/-     E     Y/H H        H       A         R/W/Q A            T/M    p                     Brazilians (2)
--- --- --- --- --- --- --- --- --- --- --- --- --- *** 0
--- --- --- --- --- --- --- --- --- --- --- --- --- *** 0
--- --- --- --- --- --- --- --- --A --- --- --- --- *** 0
--- --- --- --- --- --- --- --- --- --- --- --A --- *** 0
--- --- --- --- --- --- --- --- --- --- --- --- --- *** ***
--- --- --- --- --- --- --- --- --- --- --C --- --- *** 0
--- --- --- --- --- --- --- --T --- --- --- --- --- +14 bp 8.74
--- A-- --- --- --- --- --- --- --- --- --- --- --- −14 bp 8.25
--- A-- --- --- --- --- --- --T --- --- --- --- --- *** 0
--- A-- --- --- --- --- --- --- --- --- --- --- --- *** 0
--- A-- --- --- --- --- --- --- --- --- --- --- --A *** 3.88
--- A-- --- --- --- --- --- --- --- --- --- --- --- *** 0
--- --- --- ~TG --- --- --- --- --- --- --- --- --- +14 bp 0.97
--- --- --- --- --- --- --- --- --- --- --- -T- --- +14 bp 4.85
--- A-- --- --- --- --- --- --- --- --- --- --- --- *** 0
--- --- --- --- --- --- --- --- --- T-- --- --- --- *** 0
--- --- --- --- --- C-- --- *** *** *** *** *** *** *** 0.49
--- --- --- --- --- --- --- *** *** *** *** *** *** *** 0
--- A-- --- --- --- --- --- *** *** *** *** *** *** *** 0
--- --- --- --- --- --- --- --- --- --- --- --- --- *** 0
--- --- --- --- --- --- --- --- --- --- --- --- --- *** 0
--- --- --- --- --- --- --- --- --- -A- --- -T- --- *** 0
--- A-- --- --- --- --- --- --- --- --- --- --- --- *** 0
--- --- --- --- --- --- --- --- --- --- --- -T- --- *** 0
---   ---    ---    ---     ---   ---    -G-   ***     ***       ***         ***    ***    ***       ***         ***
+14 bp insertion (presence) of 14 bp; −14 bp deletion (absence) of 14 bp; *** not evaluated; ~ deleted; amino acid codes:
H histidine; P proline; G glycine; D asparagine acid; V valine; C cysteine; L leucine; W tryptophan; grey background in
128        H. Nückel et al.
2. Materials
2.1. Materials for             1. 2× Master Mix RED for PCR: 150 mM Tris–HCl, pH 8.5,
HLA-G* Allele Typing              40 mM (NH4)2SO4, 3 mM, MgCl2, 0.2% Tween 20®, 0.4 mM
and the Identification            DNTPs, 0.05 units/mL Ampliqon Taq DNA polymerase, inert
of the 14 bp Fragment             red dye, and a stabilizer (Ampliqon, Skovlunde, Denmark).
ins/del Polymorphism           2. Template DNA: A good quality DNA at a concentration of
in 3 ¢ UTR                        approximately 50 mg/mL.
2.1.1. Polymerase Chain        3. Oligonucleotides primers for PCR (Table 2): All primers are
Reactions                         diluted with sterile and nuclease-free water to a concentration
 Table 2
 Primers to identify SNPs within the coding region and the 14 bp fragment insertion/deletion (ins/del) polymorphism at
 3¢UTR of HLA-G
SE sense; AS anti-sense; BT 5¢biotinylation of the primer at the time of synthesis; seq sequencing primer
130        H. Nückel et al.
2.1.2. Gel Electrophoresis     1. Biozym LE agarose for gel electrophoresis (Biozym Scientific
                                  GmbH, Oldendorf, Germany).
                               2. 10× Tris buffered ethelene-diamine-tetra-acetic acid (EDTA):
                                  108 g Tris base, 55 g boric acid, 7.44 g EDTA to 1 L with
                                  double distilled water.
                               3. Sybr Safe DNA gel stain or ethidium bromide (Invitrogen,
                                  Darmstadt, Germany).
                               4. Electrophoresis chambers and sufficient power supply.
                               5. UV-transilluminator of a Gel Documentation System with UV
                                  light.
                               6. DNA marker—pBR322DNA/AluI              Marker    (Fermentas
                                  GmBH, St. Leon-Rot, Germany).
2.2. Materials           1. DNA polymerase: this protocol was optimized and tested
for the HLA-G Gene          using three different DNA polymerases (a) Platinum Taq
Promoter Region             DNA polymerase (Invitrogen, Carlsbad, CA), (b) PCR Long
Variations                  PCR enzyme mix (Fermentas, Maryland, USA), and (c)
                            Platinum Taq DNA polymerase high-fidelity (Invitrogen,
                            Carlsbad, CA).
                         2. 0.2-mL PCR tubes or plates.
                         3. Ultra-pure deionized and autoclaved water, DNAse and
                            RNAse-free.
                         4. dNTPs: this method was optimized using a 100 mM dNTP set
                            (Invitrogen) but any set of dNTP should do.
                         5. 5 mM dNTP solution: 5 mL of each dNTP from the 100 mM
                            dNTP Set and 80 mL of ultra-pure deionized water, resulting
                            in a solution of 5 mM of each dNTP.
                         6. 50 mM Magnesium chloride or 50 mM magnesium sulphate:
                            typically this is provided with the polymerases described in step 1.
                         7. Primers: Primers used in this protocol are given in Table 3.
                            Each primer was diluted to a final concentration of
                            10 pmol/mL.
                         8. DNA samples diluted to 50 ng/mL.
                         9. This protocol was standardized using the Veriti 96-well
                            Thermal Cycler (Applied Biosystems), but any thermal cycler
                            should do.
                        10. Agarose, horizontal electrophoresis cube apparatus, and ethid-
                            ium bromide to assemble a 1% agarose gel.
                        11. Any non-denaturant load buffer.
                        12. DNA ladder that allows the identification of a 1,852-bp
                            fragment. This protocol was standardized using the 1 Kb
                            GeneRuler Express (Fermentas).
                        13. UV-transilluminator or Gel documentation system with UV
                            light.
                        14. Exonuclease I enzyme (EXO) and Shrimp alkaline phosphatase
                            (SAP).
                        15. Sequencer and reagents for sequencing according to the
                            sequencer available.
                                                                                                                                           132
                                                                                                                                           H. Nückel et al.
    Table 3
    Primers to amplify and sequence the HLA-G promoter region
3. Methods
3.1. HLA-G* Allele        See Table 2 for a detailed outline of individual PCR products and
Typing and the            their representative mutations with codon names. A total of six
Identification of the     PCR products will be produced: Exon 2 (including codons 31, 35,
14 bp Fragment ins/del    and 57), exon 3 (including codons 93, 100, 107, 110), exon 3
Polymorphism in 3¢UTR     codon 130, exon 4 codon 188, and exon 4 codon 258, and 3¢UTR.
                          It is recommended to make a master mix in order to simplify the
3.1.1. Polymerase Chain   pipetting and reduce the chance of any single error (see Note 1).
Reaction
                           1. Exon 2 codons 31, 35, 57 PCR: Each individual PCR reaction
                              has a total volume of 50 mL; this includes 25 mL Ampliqon
                              Master Mix RED, 20 mL high purity water, 2 mL of each primer
                              numbers 1 and 2 (Table 2), and 1 mL DNA. Thermal cycler
                              was set as following: 5 min at 95°C (hot start), 30 s at 95°C
                              (denaturation), 40 s at 65°C (annealing), 30 s at 72°C (exten-
                              sion), and finally, 10 min at 72°C. Steps 2–5 are repeated 38
                              times before the final extension time. The amplified product is
                              208 bp long (see Note 2). This product will be used for pyros-
                              equencing—see Subheading 3.1.3, step 1.
                           2. Exon 3 codons 93, 100, 107, 110 PCR: Each individual reac-
                              tion has a total volume of 30 mL; this includes 15 mL Amplicon
                              Master Mix RED, 12 mL high purity water, 1 mL of each primer
                              numbers 5 and 6 (Table 2), and 1 mL DNA. Thermal cycler
                              conditions are exactly as for Exon 2 PCR. End product should
                              be 278 bp long. All of the 30 mL of PCR product will be for
                              pyrosequencing—see Subheading 3.1.3, step 2 (see Note 2).
                           3. Exon 3 codon 130 PCR: Use the same master mix recipe as for
                              exon 3 (30 mL) (step 2), using primer numbers 9 and 10
                              (Table 2). Thermal cycler conditions are the same as for Exon
                              2 PCR with the following exception: The annealing tempera-
                              ture is changed to 65.5°C and the extension is changed from
                              30 to 15 s. PCR product is 167 bp long. 15 mL of this product
                              will be used for pyrosequencing—see Subheading 3.1.3, step
                              3. The rest can be frozen at −20°C until all genotypes are
                              confirmed.
                           4. Exon 4 codon 188 PCR: Use the same master mix recipe as
                              for exon 3 (30 mL) (step 2), using primer numbers 12 and 13
                              (Table 2). Thermal cycler conditions are the same as for Exon
                              2 PCR with one exception: The annealing temperature is
                              changed to 60°C. PCR product is 58 bp long. 15 mL of this
                              PCR product will be used for pyrosequencing—see
                              Subheading 3.1.3, step 4. The rest can be frozen at −20°C
                              until all genotypes are confirmed.
                           5. Exon 4 codon 258 PCR: Use the same master mix recipe as for
                              exon 3 (30 mL) (step 2), using primer numbers 15 and 16
134        H. Nückel et al.
                                (Table 2). Thermal cycler conditions are the same as for Exon
                                2 PCR with one exception: The annealing temperature is
                                changed to 60°C. PCR product is 267 bp long. 15 mL of this
                                PCR product will be used for pyrosequencing—see
                                Subheading 3.1.3, step 5. The rest can be frozen at −20°C
                                until all genotypes are confirmed.
                              6. 3¢UTR PCR: Use the same master mix recipe as for exon 3
                                 (30 mL), using primer numbers 18 and 19 (Table 2). Thermal
                                 cycler conditions are exactly as for Exon 2 PCR. PCR product
                                 should be 434 bp long. 10 mL of this PCR product will be used
                                 for Gel Electrophoresis—see Subheading 3.1.2, steps 3–5. The
                                 rest can be frozen at −20°C until all genotypes are confirmed.
3.1.2. Electrophoretical      1. To confirm that the PCR of the coding region worked properly,
Separation of the PCR            the PCR products (i.e. products from Subheading 3.1.1, steps
Products of the Coding and       1–5) are applied to a 2% agarose gel. To prepare the gel add
the 3¢UTR Region                 2 g agarose to 100 mL of 0.5× TBE and cook in the micro-
                                 wave for approximately 3–5 min until all agarose is dissolved
                                 (see Note 3). Thereafter Sybr Safe DNA gel stain or ethidium
                                 bromide is added (0.5 mg/mL). Keep in mind that you need
                                 enough wells in the gel for all the samples, plus negative and
                                 positive PCR controls, as well as for the DNA Marker. A nega-
                                 tive PCR control would consist of PCR master mix which was
                                 run in the thermal cycler exactly like the patient samples, only
                                 without template DNA. A positive PCR control would be PCR
                                 master mix which was run in the thermal cycler exactly like the
                                 test samples except that here a DNA sample with known
                                 HLA-G allele (14) would be used being already proven to
                                 function in PCR.
                              2. Place the agarose gel in the gel chamber and make sure it is
                                 sufficiently covered by 0.5× TBE.
                              3. Load the samples completely in the gel and attach the power
                                 supply. Run at 180 V until good separation is acquired.
                              4. Put the gel into a UV-Transilluminator of a Gel Documentation
                                 System with UV light. The presence of a fragment with the
                                 expected bp indicates that the amplification ran properly (see
                                 Note 4).
                              5. For the SNP in the 3¢UTR, 10 mL of the product (i.e. from
                                 Subheading 3.1.1, step 6) is applied to a 3% agarose. This is
                                 prepared by mixing 3 g agarose with 100 mL 0.5× TBE and
                                 cooking for approximately 5 min. Microwaving times may vary.
                                 Thereafter Sybr Safe DNA gel stain or ethidium bromide is
                                 added (0.5 mg/mL).
                              6. After the gel has cooled off, place in the gel chamber, make
                                 sure that it is covered by 0.5× TBE, and load 10 mL of each
7 Simple Methods for the Detection of HLA-G Variants in Coding and Non-coding Regions               135
                            PCR product onto the gel. Run at 180 V until good separation
                            is acquired. Put the gel into a UV-Transilluminator of a Gel
                            Documentation System with UV light. The two different PCR
                            products of 210 and 224 bp should be present.
                         7. A sample with only one band at 210 (deletion 14 bp) or 224
                            (insertion 14 bp) indicates a homozygosity for the given geno-
                            type, for example del/del or ins/ins. A sample with two bands
                            visible (one at 210 and the other at 224) indicates a heterozy-
                            gote del/ins (Fig. 1).
3.1.3. Pyrosequencing   All steps here are to be performed at room temperature unless
                        otherwise stated. In addition, all buffers should be stored at 4°C,
                        but allowed to warm up to room temperature before starting. Also
                        before use, all sequences to be analysed should be entered into the
                        computer as indicated by the software instructions.
                         1. The exon 2 codons 31, 35, 57 PCR product will be used for
                            two individual sequencing reactions. The first, using primer
                            number 3, sequences SNPs in codon 31 and 35. The second,
                            using primer number 4, sequences one SNP in codon 57.
                         2. The exon 3 codons 93, 100, 107, 110 PCR product will also
                            be used for two individual sequencing reactions. The first,
                            using primer number 7, sequences SNPs in codons 93 and
                            100. The second, using primer number 8, sequences SNPs
                            in codons 107 and 110.
                         3. The exon 3 codon 130 PCR product will be used for only one
                            sequencing reaction using primer number 11 for the SNP in
                            codon 188.
                         4. The exon 4 codon 188 PCR product will be used for only one
                            sequencing reaction using primer number 14 for the SNP in
                            codon 188.
                         5. The exon 4 codon 258 PCR product will be used for only one
                            sequencing reaction using primer number 17 for the SNP in
                            codon 258.
136   H. Nückel et al.
3.2. Methods for the     1. Separate the reagents for PCR amplification (DNA polymerase,
HLA-G Gene Promoter         dNTPs, ultra-pure deionized water, and the primers for the
Region Variations           first amplification GPromo.S and GPromo.R—Table 3), thaw
                            and keep them on ice.
3.2.1. HLA-G Promoter
Amplification by PCR     2. On a separate bench, thaw the DNA samples and keep them
                            on ice.
                         3. Prepare a mix of reagents for the amplification. The following
                            protocol is for one reaction and the volumes are adjusted to
                            the reagents in proposed final concentration. Amplification is
                            performed in a final volume of 50 mL containing:
                            (a) 33.7 mL of ultra-pure water (or a quantity enough to a final
                                volume of 50 mL if any of the volumes given below changed);
                            (b) 1× PCR buffer (typically 5 mL of the buffer provided with
                                the polymerases described in Subheading 2);
                            (c) 3 mL of magnesium chloride (for Platinum Taq Polymerase
                                from Invitrogen and Long PCR enzyme Mix from
                                Fermentas) or 3 mL of magnesium sulphate (Platinum Taq
                                Polymerase High-Fidelity from Invitrogen), giving a final
                                concentration of 1.5 mM;
                            (d) 2 mL of the 5 mM dNTP, giving a final concentration of
                                0.2 mM of each dNTP;
                            (e) 2 mL of each primer for the first amplification, giving a final
                                amount of primer of 20 pmol each per reaction;
                            (f) 0.3 mL of DNA polymerase (1.5 units);
                            (g) Gently homogenize this solution by pipetting up and
                                down. Cap the tube.
138       H. Nückel et al.
3.2.2. HLA-G Promoter        In the previous section, the protocol was given to amplify the
Sequencing                   HLA-G promoter region by PCR. Once you have it amplified
                             with a verified clean product of approximately 1,852 bp (see
                             Subheading 3.2.1, step 7), you may sequence this PCR product by
                             using the appropriate primers described in Table 3. Alternatively,
                             you may use restriction endonucleases to screen for variability (not
                             within the scope of this chapter). Depending on the sequencer
                             available, the sequencing protocol must be adjusted for each
                             machine and sequencing reagents used. No sequencing protocol
                             is provided in this chapter since it depends on the infrastructure
                             available. Please refer to your sequencer’s documentation in order
                             to better adjust a sequencing reaction of the PCR product. In this
                             section, a strategy to evaluate the entire promoter by sequencing
                             using the primers listed in Table 3 is given.
                              1. To assure a purified product for sequencing, take 10 mL of the
                                 PCR product in a separate 0.2 mL PCR tube and add 1 mL
                                 of EXO and 1 mL of SAP. Mix the solution by pipetting up
                                 and down. Cap the tube and put it on ice before using the
                                 thermal cycler.
7 Simple Methods for the Detection of HLA-G Variants in Coding and Non-coding Regions                                139
Fig. 2. HLA-G gene promoter amplification products. 1% Agarose gel stained by ethidium bromide. The DNA ladder used
was 1 Kb GeneRuler Express (Fermentas). From the heavier to the lighter fragment, the ladder indicates fragments of 5,000,
3,000, 2,000, 1,500, 1,000, 750, 500, 300, and 100 bp. The expected amplification is approximately 1,852 bp.
Fig. 3. Primer map used to amplify and sequence the HLA-G promoter region.
4. Notes
                              1. For the PCR for the coding region you may want to run a
                                 control on an electrophoresis gel to confirm that the PCR
                                 worked properly. This will mean that you need more than
                                 one PCR reaction for each patient because all of the 50 mL
                                 (or 30 mL) of PCR product will be needed for further analysis.
                                 We suggest this for optimization of DNA concentration as well.
                              2. We use DNA prepared from EDTA-blood. This is stable and of
                                 relative good quality. When, for example, DNA has been
                                 extracted from paraffin slices, it is possible that the amount of
                                 DNA and/or primer concentration would not be enough or
                                 possibly too much.
                              3. When making more than one gel, it is helpful to prepare them
                                 individually. Cooking the gels for too long (which can be nec-
                                 essary when making many gels at once) alters the concentra-
                                 tion of the gel when too much TBE evaporates because of
                                 the boiling.
                              4. After visualization of PCR products on the gel, it is possible
                                 that no PCR product or an incorrect one has been amplified.
                                 It is very likely that this is caused by problems with the tem-
                                 plate DNA. Furthermore make sure that the correct annealing
                                 temperature is used which otherwise might lead to false priming.
                                 Possible causes of no PCR products could be either that simply
                                 DNA was not added to the mixture or that the DNA was pre-
                                 pared from paraffin slices. In this case, a long enough product
                                 cannot be amplified. Suggestions for improvement include
                                 simply trying the PCR again making sure to pipette DNA into
                                 the PCR mix or measuring the concentration of the DNA to
                                 make sure that enough template is present.
                              5. PSQ manufacturers suggest a maximum of 30 mL PCR prod-
                                 uct per reaction. We have optimized, and for our purposes,
                                 with our DNA templates, 15 mL of PCR product is usually
                                 enough. Too much PCR product can also be detrimental for
                                 results.
                              6. Leaving the binding plate to shake for longer than 5 min
                                 (manufacturers recommended time) does not make a differ-
                                 ence in the results. We have allowed the plate to shake for up
                                 to 10 min with no difference in results.
                              7. The sepharose beads sediment quickly, and when they are at
                                 the bottom of the plate, then they are harder to collect with the
                                 vacuum prep tool. It is useful to act quickly and gently to collect
                                 the maximum amount of beads bound to DNA. If too many
                                 beads have been left at the bottom of the plate, it is possible to
                                 add a bit (approximately 20 mL) of high purity water, shake for
7 Simple Methods for the Detection of HLA-G Variants in Coding and Non-coding Regions                 141
                                 a minute, and then aspirate one more time. The vacuum filter
                                 tool should be left ON at all times in order to not to lose
                                 any beads. Be cautious to always hold the filter at the same
                                 point, and not to turn it 180° by accident thereby mixing
                                 the samples!
References
 1. Carosella ED, Favier B, Rouas-Freiss N, Moreau     10. Ober C, Aldrich CL (1997) HLA-G polymor-
    P, Lemaoult J (2008) Beyond the increasing             phisms: neutral evolution or novel function?
    complexity of the immunomodulatory HLA-G               J Reprod Immunol 36:1–21
    molecule. Blood 111:4862–4870                      11. Suarez MB, Morales P, Castro MJ, Fernandez
 2. Donadi EA, Castelli EC, Arnaiz-Villena A,              V, Varela P, Alvarez M, Martinez-Laso J, Arnaiz-
    Roger M, Rey D, Moreau P (2011) Implications           Villena A (1997) A new HLA-G allele (HLA-
    of the polymorphism of HLA-G on its func-              G*0105N) and its distribution in the Spanish
    tion, regulation, evolution and disease associa-       population. Immunogenetics 45:464–465
    tion. Cell Mol Life Sci 68:369–395                 12. Tan Z, Shon AM, Ober C (2005) Evidence of
 3. Robinson J, Waller MJ, Fail SC, Marsh SG               balancing selection at the HLA-G promoter
    (2006) The IMGT/HLA and IPD databases.                 region. Hum Mol Genet 14:3619–3628
    Hum Mutat 27:1192–1199                             13. Berger DS, Hogge WA, Barmada MM, Ferrell
 4. Castelli EC, Mendes-Junior CT, Deghaide NH,            RE (2010) Comprehensive analysis of HLA-G:
    de Albuquerque RS, Muniz YC, Simoes RT,                implications for recurrent spontaneous abor-
    Carosella ED, Moreau P, Donadi EA (2010)               tion. Reprod Sci 17:331–338
    The genetic structure of 3¢untranslated region     14. Rebmann V, van der Ven K, Passler M, Pfeiffer K,
    of the HLA-G gene: polymorphisms and haplo-            Krebs D, Grosse-Wilde H (2001) Association of
    types. Genes Immun 11:134–141                          soluble HLA-G plasma levels with HLA-G
 5. Castelli EC, Mendes-Junior CT, Donadi EA               alleles. Tissue Antigens 57:15–21
    (2007) HLA-G alleles and HLA-G 14 bp poly-         15. Stephens M, Smith NJ, Donnelly P (2001) A
    morphisms in a Brazilian population. Tissue            new statistical method for haplotype recon-
    Antigens 70:62–68                                      struction from population data. Am J Hum
 6. Hviid TV (2006) HLA-G in human reproduc-               Genet 68:978–989
    tion: aspects of genetics, function and preg-      16. Stephens M, Donnelly P (2003) A comparison of
    nancy complications. Hum Reprod Update                 bayesian methods for haplotype reconstruction
    12:209–232                                             from population genotype data. Am J Hum
 7. Moreau P, Contu L, Alba F, Lai S, Simoes R,            Genet 73:1162–1169
    Orru S, Carcassi C, Roger M, Rabreau M,            17. Excoffier L, Slatkin M (1995) Maximum-
    Carosella ED (2008) HLA-G gene polymor-                likelihood estimation of molecular haplotype
    phism in human placentas: possible association         frequencies in a diploid population. Mol Biol
    of G*0106 allele with preeclampsia and miscar-         Evol 12:921–927
    riage. Biol Reprod 79:459–467
                                                       18. Excoffier L, Laval G, Schneider S (2005)
 8. Rizzo R, Hviid TV, Govoni M, Padovan M,
                                                           Arlequin ver. 3.0: an integrated software pack-
    Rubini M, Melchiorri L, Stignani M, Carturan
                                                           age for population genetics data analysis. Evol
    S, Grappa MT, Fotinidi M, Ferretti S, Voss A,
                                                           Bioinform Online 1:47–50
    Laustrup H, Junker P, Trotta F, Baricordi OR
    (2008) HLA-G genotype and HLA-G expres-            19. Excoffier L, Laval G, Balding D (2003) Gametic
    sion in systemic lupus erythematosus: HLA-G            phase estimation over large genomic regions
    as a putative susceptibility gene in systemic          using an adaptive window approach. Hum
    lupus erythematosus. Tissue Antigens 71:               Genomics 1:7–19
    520–529                                            20. Castelli EC, Mendes-Junior CT, Veiga-Castelli
 9. Sipak-Szmigiel O, Cybulski C, Wokolorczyk D,           LC, Pereira NF, Petzl-Erler ML, Donadi EA
    Lubinski J, Kurzawa R, Baczkowski T, Radwan            (2010) Evaluation of computational methods
    M, Radwan P, Ronin-Walknowska E (2009)                 for the reconstruction of HLA haplotypes.
    HLA-G polymorphism and in vitro fertilization          Tissue Antigens 76:459–466
    failure in a Polish population. Tissue Antigens    21. Castelli EC, Mendes-Junior CT, Deghaide
    73:348–352                                             NH, de Albuquerque RS, Muniz YC, Simoes
142        H. Nückel et al.
      RT, Carosella ED, Moreau P, Donadi EA        22. Ober C, Aldrich CL, Chervoneva I, Billstrand
      (2010) The genetic structure of 3¢untrans-       C, Rahimov F, Gray HL, Hyslop T (2003)
      lated region of the HLA-G gene: polymor-         Variation in the HLA-G promoter region
      phisms and haplotypes. Genes Immun 11:           influences miscarriage rates. Am J Hum Genet
      134–141                                          72:1425–1435
                                                                                            Chapter 8
Abstract
Human leukocyte antigen-E (HLA-E) is a non-classical HLA class I gene that shows a limited degree of
polymorphism compared to the classical HLA genes. The HLA-E molecule can bind peptides derived from
the leader sequence of various HLA class I alleles and some viral homologues, including CMV. The HLA-E
peptide complex can act as a ligand for the CD94/NKG2 receptors expressed on the surface of natural
killer cells and T cell subsets. Differences in expression levels between the different HLA-E alleles have
been reported and a role for HLA-E polymorphism in stem cell transplantation has been postulated. This
chapter focuses on routine technologies for HLA-E typing: the sequence-specific primer-PCR method that
uses sequence-specific primers, the PCR sequence-specific oligonucleotides Luminex method, using
sequence-specific probes attached to beads and the sequencing-based typing method, where sequencing of
the alleles is performed.
     Key words: Human leukocyte antigen-E, HLA typing method, Sequence-specific primer-PCR,
     PCR-sequence-specific oligonucleotides, Sequencing-based typing
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_8, © Springer Science+Business Media New York 2012
                                                         143
144        N. Lauterbach et al.
2. Materials
2.2. PCR-SSO Method           1. HLA-E SSO Luminex kit (One Lambda, not commercially
                                 available yet).
2.2.1. PCR, Hybridization,
and Luminex Bead                  – Denaturation buffer, store at RT.
Reactions                         – Neutralization buffer, store at RT.
                                  – Hybridization buffer, store at RT.
                                  – Wash buffer, store at RT.
                                  – SAPE stock (100×), store at 4°C.
                                  – SAPE buffer, store at 4°C.
                                  – Primer Set D-Mix, store at −30°C (mix should be pink or
                                    light purple, when necessary vortex).
                                  – HLA-E specific amplification primers, store at −30°C.
                                  – HLA-E SSO beads (including positive and negative control
                                    beads), store at −30°C (after thawing store in the dark at
                                    4°C, never re-freeze).
                              2. Optional: Biohazard.
                              3. AmpliTaq DNA polymerase (5 Units/mL), (Applied Biosystems).
                                 Store at −20°C.
                              4. 1.5-mL microfuge tubes.
                              5. PCR tubes (micronics) and caps or seal.
                              6. Micropipettes and tips.
                              7. 96-Well, thin walled PCR plate and holder.
                              8. PCR Thermocycler.
                              9. Centrifuge
                                  – Rotor for 1.5-mL microfuge tubes.
                                  – Swing bucket rotor for 96-well microplate.
                             10. Vortex mixer with adjustable speed.
                             11. Luminex fluoranalyser (Luminex Corporation) or Labscan100
                                 (One Lambda).
                             12. Luminex XMap Sheath fluid.
2.3. SBT Method               1. Expand High Fidelity PCR kit, containing 10× Expand High
                                 Fidelity buffer and Expand High Fidelity enzyme mix, (Roche).
2.3.1. Amplification and
Sequencing Reaction           2. dNTP 10 mM each.
                              3. HLA-E amplification primers:
                                    8   Molecular Typing of HLA-E           147
3. Methods
3.1. PCR-SSP                  In the HLA-E PCR-SSP approach, two different PCR reactions are
                              used; one positive for HLA-E*01:01 and one positive for HLA-
                              E*01:03. Internal amplification control primers, located in the
                              growth hormone gene, are included to check for any inconsistencies
                              during the PCR. Analysis is performed by agarose gel electrophore-
                              sis and by the detection of ethidium bromide-stained fragments.
3.1.1. Amplification           1. Prepare the mastermix, the DNA sample and thaw primers
                                  (see Note 2).
                               2. Provide a clean work environment, e.g. a Biohazard and keep
                                  the reaction mixes on ice throughout the entire protocol.
                               3. For each sample, pipette the reaction mix for HLA-E*01:01
                                  and HLA-E*01:03 amplification as described below in two
                                  separate PCR tubes (It is most efficient to make reaction mixes
                                  before starting, see Notes 3 and 4).
                                   1× Reaction mix HLA-E*01:01:
                                              4.6 mL         MM
                                              2 pmol         IC1
                                              2 pmol         IC2
                                              10 pmol        Forward primer: MSSP08079
                                              10 pmol        Reverse primer: MSSP08088
                                              Add aqua dest to a final volume of 8 mL
                                                        8    Molecular Typing of HLA-E   149
                                      4.6 mL           MM
                                      2 pmol           IC1
                                      2 pmol           IC2
                                      10 pmol          Forward primer: MSSP08080
                                      10 pmol          Reverse primer: MSSP08088
                                      Add aqua dest to a final volume of 8 mL
                         4. Add 100 ng of DNA to each mix and centrifuge the PCR tubes
                            (pulse centrifugation until reaching 200 × g).
                         5. Dilute Taq polymerase with AD to obtain 0.33 Unit/mL and
                            add 1 mL per reaction/tube.
                         6. Centrifuge the PCR tubes again.
                         7. Cap or seal the PCR tubes and transfer them to a PCR
                            Thermocycler.
                         8. Run the PCR program:
                                           2 min-96°C
                                           10 cycles               10 s-94°C
                                                                   1 min-65°C
                                           20 cycles               10 s-94°C
                                                                   50 s-61°C
                                                                   30 s-72°C
                                           Hold on 4°C
                              8. Remove the combs from the geltray and put the geltray in the
                                 electrophoresis system.
                              9. Take the PCR tray out of the thermocycler and centrifuge the
                                 tray (pulse centrifugation until reaching 200 × g).
                             10. Load 10 mL of each PCR product in each lane of the gel and
                                 10 mL of 50 bp DNA marker in one lane.
                             11. Close the electrophoresis lid and run the gel for 36 min at
                                 10 V/cm.
                             12. Following electrophoresis transfer the gel to a Geldoc system to
                                 visualize the ethidium bromide-stained bands under UV-light.
3.2. PCR-SSO                 For the SSO method, different probes specific for HLA-E*01:01,
                             *01:03, and *01:04 are bound to fluorescently coded beads, devel-
                             oped in collaboration with Dr J. Lee (One Lambda). HLA-E is
                             amplified using two amplification primers each of which is labelled
                             with biotin. After annealing of the biotinylated DNA amplicon to
                             the beads, the DNA is labelled by addition of streptavidin coated
                             phyco-erythrin. The fluorescence intensity is measured by luminex
                                                                     8   Molecular Typing of HLA-E        151
                                                         3 min-96°C
                                                         5 cycles              20 s-96°C
                                                                               20 s-60°C
                                                                               20 s-72°C
                                                         30 cycles             10 s-96°C
                                                                               15 s-60°C
                                                                               20 s-72°C
                                                         10 min-72°C
                                                         Hold on 4°C
3.2.2. Denaturation/        1. Prepare a crushed ice bath and place a clean 96-well plate in a
Neutralization Procedure       tray holder.
                            2. Transfer 5 mL of PCR product into a well of the plate.
                            3. Add 2.5 mL denaturation buffer per reaction, mix thoroughly, seal
                               or cap the tray and incubate for 10 min at room temperature.
                            4. Vortex the neutralization buffer, add 5 mL to each reaction,
                               mix thoroughly (notice the colour change to clear or pale
                               yellow) and place the plate on the ice bath.
152         N. Lauterbach et al.
3.2.3. Hybridization          1. Make sure that the thermocycler has been turned on and is
Procedure                        pre-warmed at 60°C.
                              2. Prepare hybridization mixture by combining 34 mL of hybrid-
                                 ization buffer with 4 mL of bead mixture per reaction, see Note
                                 10. Protect the mixture with the fluorescently labelled beads
                                 from the light as much as possible.
                              3. Keep the tray on ice to prevent early annealing. Vortex the
                                 hybridization mixture and add 38 mL to each well, mix well by
                                 pipetting up and down.
                              4. Cover tray with tray seal and place PCR plate into the pre-
                                 warmed thermocycler.
                              5. Incubate for 15 min at 60°C.
                              6. Following incubation, place tray in tray holder, remove seal,
                                 and quickly add 100 mL wash buffer to each well.
                              7. Cover tray with tray seal and centrifuge for 5 min at
                                 1,000–1,300 × g.
                              8. Remove wash buffer by flicking (see Note 11).
                              9. Repeat the washing step twice.
                             10. Vortex SAPE stock and prepare 1× SAPE solution during third
                                 centrifugation (see Note 12):
                                   0.5 mL SAPE stock + 49.5 mL SAPE buffer, keep solution in
                                   the dark until use.
3.2.4. Labelling              1. Add 50 mL of 1× SAPE solution to each well, seal tray, and
                                 vortex thoroughly at low speed.
                              2. Place PCR plate in the pre-warmed thermocycler and incubate
                                 for 5 min at 60°C.
                              3. Following incubation, place tray in holder, remove seal, and
                                 quickly add 100 mL wash buffer to each well.
                              4. Cover tray and centrifuge for 5 min at 1,000–1,300 × g.
                              5. Remove supernatant by flicking.
                              6. Add 70 mL wash buffer to each well, mix by pipetting, and
                                 transfer 80 mL to reading plate.
                              7. Read the plate in the Luminex apparatus.
                              8. If tray is not immediately read, keep it in the dark and at 4°C
                                 (see Note 13).
3.2.5. Analysis               1. The luminex output of one reaction will show the fluorescent
                                 intensity (FI) value measured for each bead in the reaction.
                                 There are 6 beads present, bead 35 is an internal negative con-
                                 trol, no DNA must be bound to this bead. Bead 57 is a positive
                                 control, each PCR product must be bound to this bead. Bead 58
                                 contains a probe for HLA-E*01:01, bead 59 for HLA-E*01:03
                                                        8    Molecular Typing of HLA-E      153
                        Table 1
                        FI values of the various beads and the calculated normalized
                        values
Sample 1 2 3
                        Bead 35                        14            12         9
                        Bead 57                        3,137         3,302      3,304
                        Bead 58                        2,942         100        2,414
                        Normalized value bead 58       94            3          73
                        Bead 59                        41            2,552      2,150
                        Normalized value bead 59       1             77         65
                        Bead 60                        5,184         4,965      5,314
                        Normalized value bead 60       166           151        161
                        Bead 61                        436           415        623
                        Normalized value bead 61       14            12         19
                        HLA-E typing result            *01:01        *01:03     *01:01,01:03
                       Sample 1 is typed as HLA-E*01:01 homozygous, sample 2 HLA-E*01:03 homozy-
                       gous, and sample 3 heterozygous HLA-E*01:01,01:03
3.3. Sequencing-       For sequence-based typing, the HLA-E gene is amplified using
Based Typing           primers in the 5¢ UT region and in intron 5, enabling sequencing
                       of exons 1–5 and the intervening introns. The obtained sequences
                       are analysed using the Lasergene software program, or equivalents,
                       by aligning the sequence of a sample with the known HLA-E
                       alleles. Sequence comparison will enable typing of the alleles HLA-
                       E*01:01:01, 01:03:01, 01:03:02, 01:03:03, 01:03:04, 01:04.
3.3.1. Amplification    1. Provide a clean work environment, e.g. a Biohazard. Keep the
                           reaction mixes on ice throughout the entire protocol.
154         N. Lauterbach et al.
                                                    2 min-94°C
                                                    10 cycles         15 s-94°C
                                                                      30 s-65°C
                                                                      2 min-72°C
                                                    10 cycles         15 s-94°C
                                                                      30 s-61°C
                                                                      2.5 min-72°C
                                                    10 cycles         15 s-94°C
                                                                      30 s-61°C
                                                                      3.5 min-72°C
                                                    7 min-72°C
                                                    Hold on 4°C
3.3.2. Purification of the    1. Pipette ExoSAP-IT in a PCR tube (all steps need to be per-
PCR Product                      formed on ice) followed by addition of the PCR product (4 mL
                                 ExoSAP-IT/10 mL of PCR product), mix well and centrifuge
                                 (impulse centrifugation until reaching 200 × g).
                              2. Transfer the PCR tubes to the PCR Thermocycler.
                              3. Run PCR program:
                                                      15 min         37°C
                                                      15 min         80°C
                                                      Hold on 4°C (see Note 17)
                                                       8   Molecular Typing of HLA-E     155
                                           1 min-96°C
                                           25 cycles           10 s-96°C
                                                               5 s-50°C
                                                               4 min-60°C
                                           Hold on 4°C
4. Notes
                   14. Since HLA-E*01:04 has never been detected after its initial
                       identification, it has not been possible to check the specificity
                       of the HLA-E*01:04 bead.
                   15. In case MFI values of the control beads are out of range (too
                       low positive or too high negative control values), we consider it
                       necessary to repeat the procedure. When the MFI value of the
                       positive beads is too low, this could be caused by inadequate
                       PCR amplification, that can be checked by agarose gel electro-
                       phoresis. If a correct strong PCR band is observed, the too low
                       positive control value might be caused by insufficient flicking,
                       resulting in a lower concentration of SAPE. A too high negative
                       control value might be due to some contamination. Repeating
                       the reactions with new reagents might be necessary.
                       SBT Method
                   16. Vortex the primers before adding to the mixes.
                   17. The purified PCR product can be stored for a maximum of
                       1 week at 4°C.
                   18. The amplification primers can also be used as sequencing prim-
                       ers in addition to the described sequencing primers.
                   19. It is important that all fluid has been extracted; otherwise, it
                       can result in a higher background because the remaining
                       labelled dNTPs could interfere with the sequence analysis.
                   20. The sequencing standard can be run to check for any inconsisten-
                       cies during the capillary electrophoresis and analysis procedure.
                   21. The sequencing reaction products can be analysed by any auto-
                       mated DNA sequencer that is able to detect the Big Dye
                       Terminator-labelled nucleotides.
                   22. After DNA sequence analysis, the sequences are stored in .AB1
                       files. DNASTAR software Lasergene contains EDITSEQ, ALIGN,
                       and SEQMAN modules. Individual sequences are selected, these
                       are the sample AB1 files, and HLA-E sequences obtained from the
                       IMGT/HLA database/EBI (www.ebi.ac.uk/imgt/hla) are used
                       as reference. The uploaded reference and sample sequence files are
                       assembled by SEQMAN. From the resulting contig, the nucle-
                       otide peaks can be viewed, checked and if necessary adjusted.
                       Extended sequences obtained as displayed in the contig can be
                       used as reference sequence additionally. Lasergene software is a
                       product from DNASTAR, Madison, USA.
Acknowledgements
                   The authors thank Jarhow Lee (One Lambda, Los Angeles, USA)
                   for the development of the HLA-E SSO kit and reagents and Els
                   Bielen and Timo Olieslagers for their practical assistance.
158       N. Lauterbach et al.
References
 1. Antoun A et al (2009) Ethnic variability in                diversity for the regulation of NK cells. Hum
    human leukocyte antigen-E haplotypes. Tissue               Immunol 61(11):1059–1065
    Antigens 73(1):39–45                                    9. Tamouza R et al (2005) Association of HLA-E
 2. Grimsley C et al (2002) Definitive high resolu-            polymorphism with severe bacterial infection
    tion typing of HLA-E allelic polymorphisms:                and early transplant-related mortality in
    identifying potential errors in existing allele            matched unrelated bone marrow transplanta-
    data. Tissue Antigens 60(3):206–212                        tion. Transplantation 80(1):140–144
 3. Ohya K, Kondo K, Mizuno S (1990)                       10. Tamouza R et al (2006) Homozygous status
    Polymorphism in the human class I MHC locus                for HLA-E*0103 confers protection from acute
    HLA-E         in     Japanese.    Immunogenetics           graft-versus-host disease and transplant-related
    32(3):205–209                                              mortality in HLA-matched sibling hematopoi-
 4. Grimsley C, Ober C (1997) Population genetic               etic stem cell transplantation. Transplantation
    studies of HLA-E: evidence for selection. Hum              82(11):1436–1440
    Immunol 52(1):33–40                                    11. Danzer M et al (2009) Clinical significance of
 5. Geraghty DE et al (1992) Polymorphism at                   HLA-E*0103 homozygosity on survival after
    the HLA-E locus predates most HLA-A and -B                 allogeneic hematopoietic stem-cell transplanta-
    polymorphism. Hum Immunol 33(3):                           tion. Transplantation 88(4):528–532
    174–184                                                12. Ludajic K et al (2009) Association of HLA-E
 6. Ulbrecht M et al (1999) Cell surface expression            polymorphism with the outcome of hematopoi-
    of HLA-E: interaction with human beta2-                    etic stem-cell transplantation with unrelated
    microglobulin and allelic differences. Eur J               donors. Transplantation 88(10):1227–1228
    Immunol 29(2):537–547                                  13. Park KS et al (2007) HLA-E*0101 and HLA-
 7. Strong RK et al (2003) HLA-E allelic variants.             G*010101 reduce the risk of Behcet’s disease.
    Correlating differential expression, peptide               Tissue Antigens 69(2):139–144
    affinities, crystal structures, and thermal stabili-   14. Paquay MM, Schellekens J, Tilanus MG (2009)
    ties. J Biol Chem 278(7):5082–5090                         A high-throughput Taqman(R) approach for
 8. Maier S et al (2000) Implications of HLA-E                 the discrimination of HLA-E alleles. Tissue
    allele expression and different HLA-E ligand               Antigens 74(6):514–519
                                                                                            Chapter 9
Abstract
Classical, alternative, or lectin pathways may activate the complement system cascade. The classical pathway
includes the C4 protein and functions in the prevention of immune complex precipitation and in clearance
of immune complexes.
      Two isotypes of C4—C4A and C4B—are coded by genes located at two loci within the major histo-
compatibility complex (MHC) on chromosome 6. While these isotypes share over 99% amino acid sequence
homology, five nucleotide differences located in exon 26 are responsible for major structural and functional
differences between the C4 isotypes.
      C4A and C4B are highly polymorphic with over 40 alleles, gene duplications, and “null alleles”. C4
genes may be short (14.6 kb) or long (21 kb), due to the absence or presence of an endogenous retroviral
sequence—HERV-K(C4)—in intron 9, respectively. The C4 gene copy number (GCN) can vary from 1–3
per haplotype or 2–6 per diploid genome. The variation in GCN leads to a range of C4 plasma protein
concentrations among healthy subjects. In subjects with equal numbers of C4 genes, subjects with short
genes have C4 plasma levels relatively higher than subjects with long genes.
      Variation of the C4 GCN, the gene size (long or short) and the C4 isotypes (C4A and C4B) may also
lead to susceptibility to autoimmune disease. Therefore, in subjects with autoimmune disease, a low serum
C4 level may be due to ongoing disease activity associated with complement activation and consumption
or it may be due to genetic factors. Distinguishing between these will have clinical implications.
      Exact determination of GCN can be difficult, at least in part due to the high degree of homology
between C4A and C4B and a variety of techniques has been described. This chapter describes a quantitative
TaqMan real-time PCR (qPCR) copy number assay, based on our laboratory experience using this assay.
Key words: Human complement genes, C4A, C4B, Gene copy number, Real-time PCR
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_9, © Springer Science+Business Media New York 2012
                                                         159
160   A.S.L. Castley and O.P. Martinez
          been reported in 1,241 European Americans, with 0–5 C4A and 0–4
          C4B isotypes. The risk of SLE significantly increased in subjects with
          only two copies of C4 and patients with SLE had lower total C4 and
          C4A genes (OR = 6.514; P = 0.00002) (21). Another study has shown
          susceptibility to SLE decreased in subjects with five or more total
          copies of C4 (OR = 0.466; P = 0.016). Both zero copies (OR = 5.267;
          P = 0.001) and one copy (OR = 1.613; P = 0.022) of C4A were risk
          factors for SLE, whereas three or more copies of C4A appeared to be
          protective (OR = 0.574; P = 0.012) (25).
               Polymorphism of C4 can be defined by a number of methods,
          including electrophoretic mobility of the intact protein or its sub-
          units, serologically using the Rodgers (Rg) and Chido (Ch) deter-
          minants (26) and by DNA-based methods (27). Exact determination
          of GCN can be difficult, at least in part due to the high degree of
          homology between C4A and C4B. Furthermore, the polymor-
          phisms are located in the C4d fragment of the a-chain of the C4
          molecule, and only five nucleotide polymorphisms located in exon
          26 are responsible for the major structural and functional differ-
          ences between the C4 isotypes (28).
               A number of approaches have been tried to determine the C4
          GCN, including counting of fragment numbers following restriction
          fragment length polymorphism (RFLP) analysis (25), densitometry
          of products obtained by pulsed field gel electrophoresis (PFGE) and
          Southern blotting (22), and a multiplex ligation-dependent probe
          amplification (MLPA) assay combined with isotype-specific ELISAs
          to determine the level of C4A and C4B (20).
               Wu et al. developed quantitative TaqMan real-time PCR (qPCR)
          assays based on nucleotide sequences specific for the C4A and C4B
          genes, structural characteristics corresponding to long and short C4
          genes, and the breakpoint region of RP-C4-CYP21-TNX (RCCX)
          modular duplication (29). Using cloned C4 genomic DNA (gDNA)
          covering six logs of DNA concentrations for calibration and a relative
          standard curve they described eight HLA haplotypes with single C4
          genes in mono-modular RCCX that are associated with multiple auto-
          immune and infectious diseases and 32 bi-modular, 4 tri-modular, and
          one quadri-modular RCCX (29).
               In this chapter, a qPCR copy number assay based on the
          method of Wu et al. (29) is described.
               In a qPCR, a fluorescence dye is incorporated into the amplified
          DNA as the PCR proceeds. The dye emits light proportional to the
          number of amplified copies. The TaqMan dye chemistry is specific
          and precise for monitoring the kinetics of the amplification process.
          Quantification of the initial amount of DNA present in the reaction
          is based on the number of cycles required to reach a threshold (CT),
          at which the fluorescence is increased significantly and passes an
          arbitrarily defined value. The greater the initial amount of DNA
          template, the fewer cycles that are required for the reaction to reach
          the fluorescence threshold and a smaller CT observed.
162     A.S.L. Castley and O.P. Martinez
2. Materials
3. Methods
3.1. Pre-PCR Set Up           1. Ensure blood has been collected in either ACD or EDTA
                                 collection tubes.
3.1.1. Prepare Genomic DNA
                              2. Extract, purify, and quantify DNA using your usual laboratory
                                 methods.
                              3. Dilute the gDNA. Use sterile deionised water to obtain a con-
                                 centration of 5 ng/mL (see Notes 1 and 2).
                              4. Use the diluted gDNA immediately or keep at 4°C for long-
                                 term storage.
                              5. Vortex the diluted gDNA for 5 s before use.
    Table 1
    Reaction mixture composition – Volume (mL) of reaction mixture components
    required in a C4 assay
3.2. Prepare PCRs               1. Calculate the volume of components needed, based on the
                                   reaction volume and the number of reactions (see Table 1).
                                   Include excess volume to provide for losses that occur during
                                   reagent transfers (equivalent to two extra samples). Thus,
                                   number of reactions = 4 × [number of samples + controls] + 2.
                                2. Thaw the TaqMan® Copy Number and TaqMan® Copy Number
                                   Reference reagents at room temperature, vortex gently for 5 s
                                   and then centrifuge tubes briefly (500 rpm for 10 s).
                                3. Thoroughly mix the TaqMan® Genotyping Master Mix by
                                   flicking and briefly centrifuge (500 rpm for 10 s).
                                4. Combine the required volumes of reaction components in a
                                   microcentrifuge tube (see Table 1). Use separate microcentri-
                                   fuge tubes for the C4A and for the C4B assays and ensure the
                                   tube caps are firmly in place to avoid leakage.
                                5. Mix the contents thoroughly by flicking or inverting the micro-
                                   centrifuge tubes, then centrifuge briefly at 500 rpm for 10 s.
                                6. Pipette 16 mL of reaction mixture into each of the appropriate
                                   wells of a MicroAmp® Optical 96-well reaction plate.
                                7. Briefly vortex the diluted gDNA samples for 5 s (from
                                   Subheading 3.1.1, step 5).
                                8. Using a different pipette tip for each sample, add 4 mL of
                                   gDNA (5 ng/mL) and the control samples to the appropriate
               9      Molecular Analysis of Complement Component C4 Gene Copy Number           165
3.3. Running the PCRs        1. Place the reaction plate on the 7900 qPCR instrument to be
on the Real-Time                used.
Instrument                   2. Under “Instrument” ensure the following parameters are
                                correctly identified on the template.
                                          Parameter                 Setting
                                          Run                       Standard
                                          Reaction plate            96-Well standard
                                          Ramp speed/model          9600 Emulation
                                          Sample volume             20 mL
3.4. Post Real-Time          1. Open the Analysis Settings window of the RT-PCR Instrument
PCR Analysis                    software and set:
                                (a) Manual CT Threshold of 0.2.
                                (b) Autobaseline—On.
                             2. Apply settings and close window.
                             3. Select “Analyse” to analyse experiment and review data.
                             4. Select the VIC and FAM tabs in the analysis programme indi-
                                vidually and make sure that the amplification curves have a dis-
                                tinct exponential amplification phase for (see Figs. 1 and 2):
                                (a) Reference Assay (VIC® dye signal) in all samples.
                                (b) Copy Number Assay (FAM® dye signal) in most wells.
166         A.S.L. Castley and O.P. Martinez
Fig. 1. Principles of the real-time PCR. A fluorescent dye is incorporated into the amplified DNA and emits light proportional
to the number of amplified copies. Quantification of the initial amount of target DNA sequence present in the reaction is
based on the number of cycles required for the measured fluorescence to reach a given threshold. The number of cycles
required to reach that threshold is the CT. The CT value is highly proportional to the copy number of the target sequence in
the template. The greater the copy number present, the fewer cycles of PCR required for the accumulated product to be
detected in the PCR process, and the lower the CT value.
Fig. 2. An amplification profile from a C4B copy number assay from the 7900 (FAM representation). Samples with 1, 2, and
3–4 copy numbers are identified. Zero copy numbers are seen as background fluorescence. The CT values for samples with
one and two C4B copies are 26 and 25 respectively. The CT value for the sample with three C4B copies is just over 24 and
for the sample with four C4B copies is just under 24 (see Figs. 1 and 3).
             9   Molecular Analysis of Complement Component C4 Gene Copy Number       167
4. Notes
Fig. 3. Results generated by the CopyCaller™ software from the C4B amplification profiles shown in Fig. 2. (a) A bar graph
of the C4 copy number calculated by the software for each sample from the results shown in (b). A copy number range bar
(asterisk) indicates the minimum and maximum copy number calculated for the replicates of each sample. (b) The sample
location in the 96-well reaction plate (well), the sample identification (sample), the CT for FAM (C4) and for VIC (reference
sequence) and the difference between the CT values for C4 and the reference (DCT). For example—wells 57–60 contain
replicates of the control that has two copies of C4B (2 CN CTL), with FAM CT values of 24.9362; 24.8713; 24.8222, and
24.9778. The same samples have VIC CT values of 25.5299; 25.3803; 25.4582, and 25.4402, respectively. (c) The results
of the copy number analysis of individual samples. Each row shows the combined values of the replicates used to generate
the results for a single sample, the expected and the observed copy number for each sample and the mean value of the
FAM and VIC CT for C4 and the reference assay, respectively.
References
 1. Schifferli JA, Ng YC, Peters DK (1986) The             component C4 gene is due to an ancient retro-
    role of complement and its receptor in the             viral integration. Exp Clin Immunogenet 12:74
    elimination of immune complexes. N Engl J           8. Awdeh ZL, Alper CA (1980) Inherited struc-
    Med 21:488                                             tural polymorphism of the fourth component
 2. Lachmann PJ (1987) Heberden Oration 1986.              of human complement. Proc Natl Acad Sci
    Complement—friend or foe. Br J Rheumatol               USA 77:3576
    26:409–415                                          9. Ulgiati D, Abraham LJ (1996) Comparative anal-
 3. Prentice HL, Schneider PM, Strominger JL               ysis of the C4B1 gene from the HLA-A1, B8,
    (1986) C4B gene polymorphism detected in a             DR3 haplotype. Exp Clin Immunogenet 13:43
    human cosmid clone. Immunogenetics 23:             10. Law SA, Doods AW, Porter RR (1984) A com-
    274                                                    parison of the properties of two classes, C4A
 4. Schneider PM, Carroll MC, Alper CA et al               and C4B, of the human complement compo-
    (1986) Polymorphism of the human comple-               nent C4. EMBO J 3:1819
    ment C4 and steroid 21-hydroxylase genes:          11. Isenman DE, Young JR (1984) The molecular
    restriction fragment length polymorphisms              basis for the difference in immune hemolysis
    revealing structural deletions, homoduplica-           activity of Chido and Rodgers isotypes of human
    tions, and size variants. J Clin Invest 78:650         complement component C4. J Immunol 132:
 5. Palsdottir A, Fossdal R, Arnason A et al (1987)        3019
    Heterogeneity of human C4 gene size: a large       12. Braun L, Schneider PM, Giles CM et al (1990)
    intron (6.5 kb) is present in all C4A genes and        Null alleles of human complement C4. Evidence
    some C4B genes. Immunogenetics 25:299                  for pseudogenes at the C4A locus and for gene
 6. Dangel AW, Mendoza AR, Baker BJ et al                  conversion at the C4B locus. J Exp Med
    (1994) The dichotomous size variation of the           171:129
    human complement C4 genes is mediated by a         13. O’Neill GJ, Yang SY, Dupont B (1978) Two
    novel family of endogenous retroviruses, which         HLA-linked loci controlling the fourth compo-
    also establish species-specific genomic patterns       nent of human complement. Proc Natl Acad
    among Old World primates. Immunogenetics               Sci USA 75:5165
    40:425                                             14. Mauff G, Brenden M, Braun-Stilwell M et al
 7. Chu X, Rittner C, Schneider PM (1995) Length           (1990) C4 reference typing report. Complement
    polymorphism of the human complement                   Inflamm 7:193
                  9   Molecular Analysis of Complement Component C4 Gene Copy Number                       171
15. Carroll MC, Palsdottir A, Belt KT, Porter RR               temic lupus erythematosus. Cytogenet
    (1985) Deletion of complement C4 and steroid               Genome Res 123(1–4):131
    21-hydroxylase genes in the HLA class III              23. Zhang WJ, Degli-Esposti MA, Cobain TJ et al
    region. EMBO J 4(10):2547                                  (1990) Differences in gene copy number car-
16. Yu CY, Campbell RD (1987) Definitive RFLPs                 ried by different MHC ancestral haplotypes.
    to distinguish between the human complement                Quantitation after physical separation of haplo-
    C4A/C4B isotypes and the major Rodgers/                    types by pulsed field gel electrophoresis. J Exp
    Chido determinants: application to the study of            Med 171:2101
    C4 null alleles. Immunogenetics 25:383                 24. Christiansen FT, McCluskey J, Dawkins RL
17. Uko G, Christiansen FT, Dawkins RL, McCann                 et al (1983) Complement allotyping in SLE:
    VJ (1986) Reference ranges for serum C4                    association with C4A null. Aust N Z J Med
    concentrations in subjects with and without C4             13:483
    null alleles. J Clin Pathol 39:573                     25. Bohme JM, Andersson G, Andersson E et al
18. Rebmann V, Doxiadis I, Kubens BS, Grosse-                  (1985) HLA-DR beta genes vary in number
    Wilder H (1992) Quantitation of the human                  between different DR specificities whereas the
    component C4: definition of C4 Q0 alleles and              number of DQ beta genes is constant.
    C4A duplications. Vox Sang 62:117                          J Immunol 135:2149
19. Yang Y, Chung EK, Zhou B et al (2003)                  26. Giles CM, Uring-Lambert B, Goetz J et al
    Diversity in intrinsic strengths of the human              (1988) Antigenic determinants expressed by
    complement system: serum C4 protein concen-                human C4 allotypes; a study of 325 families
    trations correlate with C4 gene size and poly-             provides evidence for the structural antigenic
    genic variations, haemolytic activities, and               model. Immunogenetics 27:442
    body mass index. J Immunol 171(5):                     27. Schneider PM, Stradmann-Bellinghausen B,
    2734–2745                                                  Rittner C (1996) Genetic polymorphism of the
20. Wouters D, van Schouwenburg P, van der Horst               fourth component of human complement:
    A et al (2009) High-throughput analysis of the             population study and proposal for a revised
    C4 polymorphism by a combination of MLPA                   nomenclature based on genomic PCR typing of
    and isotype-specific ELISA’s. Mol Immunol                  Rodgers and Chido determinants. Eur J
    46(4):592                                                  Immunogenet 23:335
21. Yang Y, Chung EK, Wu YL et al (2007) Gene              28. Yu CY, Belt KT, Giles CM et al (1986)
    copy-number variation and associated polymor-              Structural basis of the polymorphism of
    phisms of complement component C4 in human                 human complement components C4A and
    systemic lupus erythematosus (SLE): low copy               C4B: gene size, reactivity and antigenicity.
    number is a risk factor for and high copy number           EMBO J 5:2873
    is a protective factor against SLE susceptibility in   29. Wu YL, Savelli SL, Yang Y et al (2007) Sensitive
    European Americans. Am J Hum Genet 80:                     and specific real-time polymerase chain reac-
    1037                                                       tion assays to accurately determine copy num-
22. Wu YL, Yang Y, Chung EK et al (2008)                       ber variations (CNVs) of human complement
    Phenotypes, genotypes and disease suscepti-                C4A, C4B, C4-long, C4-short, and RCCX
    bility associated with gene copy number vari-              modules: elucidation of C4 CNVs in 50 con-
    ations: complement C4 CNVs in European                     sanguineous subjects with defined HLA geno-
    American healthy subjects and those with sys-              types. J Immunol 179(5):3012
                                                                                        Chapter 10
Abstract
Real-time quantitative PCR is an efficient method for high-throughput genotyping of single nucleotide
polymorphisms (SNPs). In this chapter, we describe the 5¢ nuclease allelic discrimination assay for geno-
typing biallelic SNPs.
     Key words: Single nucleotide polymorphism, Real-time polymerase chain reaction, TaqMan®
     chemistry, Reporter dye, Fluorescent label
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_10, © Springer Science+Business Media New York 2012
                                                         173
174      M. Malkki and E.W. Petersdorf
2. Materials
2.1. TaqMan Assay        Supplies needed for performing the TaqMan® assay are as follows:
                          1. 96-Well optical plates and adhesive optical covers (Applied
                             Biosystems, [ABI]).
                          2. Splash-free support base for 96-well plates.
                          3. Plate centrifuge.
                          4. 10, 200 and 1,000-mL sterile tips.
                          5. 1–10, 20–200 and 100–1,000-mL single or multiple tip electronic
                             pipets or 10, 20, 200 and 1,000-mL manual pipets.
                          6. Eppendorf tubes.
                          7. TaqMan® Universal Master Mix.
2.2. Real-Time PCR       ABI 7500 Real-Time PCR system (see Note 1).
Instrumentation
10   Genotyping of Single Nucleotide Polymorphisms by 5¢ Nuclease Allelic Discrimination   175
2.3. Design of Probes    Currently, over 160,000 inventoried, ready-to-use SNP assays and
and Primers              over 4.5 million predesigned, made-to-order SNPs assays are avail-
                         able from ABI (https://products.appliedbiosystems.com). The par-
                         ticular SNP of interest can be searched by its rs number (http://
                         www.ncbi.nlm.nih.gov/SNP/) from the ABI Web site. ABI 40× or
                         80× TaqMan® SNP genotyping assays can be ordered for different
                         sample sizes: small scale for 1,500 reactions, medium for 5,000 reac-
                         tions, and large for 12,000 reactions calculated from 5 mL reaction/
                         sample. The ready-to-use assay mixture is preloaded with (1) the
                         forward and reverse primers for amplification of the polymorphic
                         sequence, and (2) the two allele-specific TaqMan® probes descriptive
                         of the SNP of interest, one labeled with VIC® dye and the other with
                         6FAM™ dye. Every assay comes with an information file containing
                         details on the chromosomal location of the SNP, allele frequency
                         (only for validated assays) and the nucleotide sequence to which the
                         VIC/FAM reporter dye is linked. Upon delivery, all assays should be
                         protected from light by wrapping the tube in foil. The 40× and 80×
                         assays should be diluted with 1× TE buffer (10 mM Tris–HCl, 1 mM
                         EDTA, pH 8.0) to 20× working mixture for best stability. This assay
                         mixture should be aliquoted in batches to avoid freeze–thaw cycling.
                         Assay should be stored at −15 to −25°C.
                              For SNPs which do not have ready-made assays available, there
                         are several approaches for the user to design probe and primer sets:
                         (1) use ABI Primer Express Software (see below); (2) use ABI
                         Custom TaqMan® design service; and (3) search of the literature or
                         available databases, such as SNP500Cancer (http://variantgps.nci.
                         nih.gov/cgfseq/pages/snp500.do) for TaqMan® genotyping reagents
                         for the SNP of interest.
                              Should the user require probes and primers to be designed, the
                         ABI Primer Express software v2.0 provides assay design guidelines
                         developed specifically for quantification assays. The general guide-
                         lines are as follows:
                           1. Import the DNA sequence containing the SNP of interest
                              directly from NCBI dbSNP database (http://www.ncbi.nlm.
                              nih.gov/SNP/).
                           2. Design forward and reverse amplification primers each with
                              optimal lengths of 20 nucleotides and with matching Tm
                              (59°C optimal) within 1°C of each other. Strive for an ampli-
                              con length of 75–150 bp. Avoid a guanine residue at the 5¢
                              end of the probe because a G residue adjacent to reporter dye
                              will quench the reporter fluorescent even after cleavage. Target
                              a Tm of 65–67°C for each probe and a minimum length of 13
                              nucleotides. Avoid runs of an identical nucleotide in the probe
                              sequence (especially runs of four or more consecutive Gs
176      M. Malkki and E.W. Petersdorf
3. Methods
3.1. Preparation         Prepare 5–25 ng/mL of DNA in TE buffer (10 mM Tris pH 8.0,
of Genomic DNA           0.1 mM EDTA) or in nuclease-free water. All samples should
                         preferably have the same DNA concentration for genotyping to
                         avoid a wide range of fluorescence activity within a genotype group
                         (see Notes 2 and 3).
3.3. TaqMan®             This can be purchased from ABI. To avoid PCR contamination
Universal PCR            from previously amplified material, “carry-over,” inclusion of
Master Mix               AmpErase® uracil-N-glycosylase (UNG) in the buffer is recom-
                         mended (see Note 4). A single lot of master mix should be used in
                         the same experiment, since lot-to-lot variability can contribute to
                         variation in fluorescence values. The PCR master mix includes a
                         passive reference dye ROX™, which is used to normalize fluorescent
                         signals in individuals wells caused by small differences in PCR reac-
                         tion mixture volume (see Note 5).
3.4. PCR Mixture         For one well, a basic cocktail volume is 3.75 mL containing
                         2.5 mL 2× Taqman universal PCR master mix, 0.25 mL 20× SNP
                         assay mix, and 1 mL of nuclease-free water. For full 96-well plate
                         this cocktail is made as a 105× mixture to allow for dead volume
                         and pipetting errors as follows: 262.5 mL 2× TaqMan® universal
                         PCR master mix, 26.25 mL 20× SNP assay mix, and 105 mL of
10   Genotyping of Single Nucleotide Polymorphisms by 5¢ Nuclease Allelic Discrimination   177
3.5. Amplification         1. Clean up the working surface with ethanol and water.
of Target Sequence         2. Label the 96-well optical plate with assay name and plate
                              identifier and put the plate into a PCR splash-free support
                              base (see Note 6).
                           3. Prepare the PCR mixture for as many plates as needed.
                           4. Pipet 1.3 mL of individual DNAs into the bottom of wells,
                              changing the tip between every DNA sample. Leave two wells
                              in each plate devoid of template DNA as negative controls; for
                              these wells, add 1.3 mL of water in place of DNA.
                           5. Add 3.75 mL of PCR master mix onto wall of each well. Change
                              tips if necessary to prevent carry over from well to well.
                           6. Cover the plate with adhesive optical cover and centrifuge the
                              plates at 1,500 rpm for 1 min to concentrate reagents at the
                              bottom of the wells, and to remove any air bubbles.
                           7. Place the plate/s in ABI 9700 PCR thermal cycler or 7500
                              Real-time PCR system (see Note 1). If a real-time machine is
                              used for the amplification step, please follow the manufacturer’s
                              instructions for plate setups and use of the machine. For MGB
                              quencher probes, program the following cycling conditions:
                              2 min 50°C (this step only if UNG containing buffer is used),
                              10 min 92°C followed by 45 cycles of 30 s 92°C and 1 min 30 s
                              60°C. For TAMRA quencher probes, recommended conditions
                              are: 2 min 50°C (perform this step only if UNG containing
                              buffer is used), 10 min 95°C followed by 45 cycles of 30 s 95°C
                              and 1 min 30 s 60°C. After cycling, keep plates at 4°C.
                           8. Read the plates in 7500 Real-Time PCR system using allelic
                              discrimination with automated allele calling settings for the
                              SDS 2.1 software (please refer to manufacturer’s instructions)
                              (see Note 7).
4. Notes
Fig. 1. Example of “trailing” genotype clusters AA (circle), AB (triangle), and BB (diamond ). Trailing describes the variation
of fluorescence signals among a given genotype that has the appearance of being spread across an imaginary line that
extends from the no template control (square). This phenomenon is often caused by variability in concentration of individual
genomic DNAs.
Fig. 2. Example of a good separation of genotype clusters AA (circle), AB (triangle), and BB (diamond ). The no template
controls (square) are distant from any clusters. Samples within a given cluster are close to the center of the cluster and the
separation of the three clusters is very clear. Contrast this figure with that shown in Fig. 1.
Fig. 3. Example of SNPs assay requiring modification of PCR annealing temperature from 60 to 62°C. (a) At 60°C
annealing, AB (triangle) and BB (diamond ) clusters are not well separated, (b) the same assay with 62°C annealing
temperature showing better separation of AB (triangle) and BB (diamond ) clusters.
Fig. 4. Example of SNPs assay requiring modification of PCR annealing temperature from 60 to 59ºC. (a) At 60°C annealing,
AB (triangle) and AA (circle) clusters are not well separated, (b) the same assay with 59°C annealing temperature showing
better separation of AB (triangle) and AA (circle) clusters.
References
12. The MHC Sequencing Consortium (1999)                  and disease associations: 2004. Tissue Antigens
    Complete sequence and gene map of a human             64:631–649
    major histocompatibility complex. Nature          22. Saiki RK et al (1985) Enzymatic amplification
    401:921–923                                           of beta-globin genomic sequences and restric-
13. Walsh EC et al (2001) An integrated haplotype         tion site analysis for diagnosis of sickle cell
    map of the human major histocompatibility             anemia. Science 230:1350–1354
    complex. Am J Hum Genet 73:580–590                23. Mullis KB, Faloona FA (1987) Specific syn-
14. Horton R et al (2004) Gene map of the extended        thesis of DNA in vitro via a polymerase-cata-
    human MHC. Nat Rev Genet 5:889–899                    lyzed chain reaction. Methods Enzymol
15. Miretti MM et al (2005) A high-resolution             155:335–350
    linkage-disequilibrium map of the human major     24. Higuchi R et al (1993) Kinetic PCR analysis:
    histocompatibility complex and first generation       real-time monitoring of DNA amplification
    of tag single-nucleotide polymorphisms. Am J          reactions.     Biotechnology       (NY)     11:
    Hum Genet 76:634–646                                  1026–1030
16. de Bakker PI et al (2006) A high-resolution       25. Morrison TB, Weis JJ, Wittwer CT (1998)
    HLA and SNP haplotype map for disease asso-           Quantification of low-copy transcripts by con-
    ciation studies in the extended human MHC.            tinuous SYBR Green I monitoring during
    Nat Genet 38:1166–1172                                amplification. Biotechniques 24:954–958;
17. Cree BA et al (2010) A major histocompatibility       960; 962
    Class I locus contributes to multiple sclerosis   26. Piatek AS et al (1998) Molecular beacon
    susceptibility independently from HLA-DRB1*           sequence analysis for detecting drug resistance
    15:01. PLoS One 5:e11296                              in Mycobacterium tuberculosis. Nat Biotechnol
18. Baschal EE et al (2009) Defining multiple             16:359–363
    common “completely” conserved major histo-        27. Marras SA, Kramer FR, Tyagi S (1999)
    compatibility complex SNP haplotypes. Clin            Multiplex detection of single-nucleotide varia-
    Immunol 132:203–214                                   tions using molecular beacons. Genet Anal
19. International MHC and Autoimmunity                    14:151–156
    Genetics Network et al (2009) Mapping of          28. Mhlanga MM, Malmberg L (2001) Using
    multiple susceptibility variants within the MHC       molecular beacons to detect single-nucleotide
    region for 7 immune-mediated diseases. Proc           polymorphisms with real-time PCR. Methods
    Natl Acad Sci USA 106(44):18680–18685                 25:463–471
20. Valdes AM, Thomson G, Type 1 Diabetes             29. Holland PM et al (1991) Detection of specific
    Genetics Consortium (2009) Several loci in the        polymerase chain reaction product by utilizing
    HLA class III region are associated with T1D          the 5¢—3¢ exonuclease activity of Thermus
    risk after adjusting for DRB1-DQB1. Diabetes          aquaticus DNA polymerase. Proc Natl Acad Sci
    Obes Metab 11(suppl 1):46–52                          USA 88:7276–7280
21. Shiina T, Inoko H, Kulski JK (2004) An update     30. Heid CA et al (1996) Real time quantitative
    of the HLA genomic region, locus information          PCR. Genome Res 6:986–994
                                                                                        Chapter 11
Abstract
We have developed a MICA typing method based on polymerase chain reaction (PCR) sequence-based
typing and a computer program that determines the polymorphisms and distinguishes the GCT repeats in
exon 5. One PCR amplification was performed to obtain templates of 2.2 kb, including exons 2, 3, 4, and
5 of MICA to be sequenced with two forward and two reverse primers. Overlay of nucleotide sequencing
signals resulting from presence of different GCT repeats in exon 5 from two different MICA alleles can be
identified by a computer program that analyses the combined signal string containing the 35 bases.
Key words: MICA typing, SBT, External domains, Transmembrane region, Computer analysis
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_11, © Springer Science+Business Media New York 2012
                                                         183
184      Y. Zou and P. Stastny
2. Materials
                           Table 1
                           Sequences of primers
2.3. Supplies              1. Thin-wall 96-well PCR plates (ISC BioExpress, Kaysville, UT,
                              Cat. T3031-21).
                           2. Thermal seal sealing films (ISC BioExpress, Cat. T2417-5).
                           3. Quanti-Marker 100 bp (GeneMate Inc., Kaysville, UT, USA.
                              Cat. QM4-104K).
                           4. MicroAmp Optical Plate (Applied Biosystems, Part No. N801-
                              0560).
                           5. Running Buffer, 10× (Applied Biosystems, Part No. 402824).
                           6. Metal Plate (Applied Biosystems, Cat. MACL09645).
                           7. MultiScreen®-HV (Millipore, Billerica, MA, USA. Cat.
                              MAHVN4550).
                           8. Pipette tips for 1–20, 21–200, and 100–1,000 mL.
                           9. PCR tubes (0.2 thin walled) Applied Biosystems, #N801-
                              0838.
                          10. Genetic analyzer plate Septa 96-wells Applied Biosystems,
                              #4315933.
                          11. Genetic analyzer     plate    Retainer       Applied   Biosystems,
                              #4317241.
                          12. Capillary array 50 cm long, Applied Biosystems, #4315930.
3. Methods
3.1. DNA Preparation       1. DNA can be isolated from acid-citrate-dextrose (ACD) anti-
                              coagulated whole blood, culture cell lines, or buccal epithelial
                              cells. DNA isolation methods vary considerably depending on
                              the starting material. The goal is to prepare high-molecular-
                              weight DNA of sufficient purity to allow HLA and MICA
                              allele-level typing (see Note 1). Preparations of high-quality
                              DNA are critical for amplification of MICA alleles since the
                              length of the amplified fragment is in the range of 2,200 bp.
                              Therefore, a protocol that produces a high-quality DNA prep-
                              aration should be selected. DNA samples should be of high
                              quality as determined by 260/280 nm absorbance ratios (1.7–
                              2.0). Prepare DNA samples with concentration at 20 ng/mL in
                              water. For example, to make a 100 mL DNA sample at 20 ng/mL
                              from a 100 ng/mL DNA sample, add 20 mL of DNA
                              (100 ng/mL) to 80 mL of water.
                          Fig. 1. Strategy for MICA allele-level typing by sequence-based typing (SBT). DNA frag-
                          ments (2.2 kb) containing the polymorphic sites in exons 2, 3, and 4 and the STR in exon
                          5 are amplified with the generic MICA-specific primers MICA6823F and MICA9023R. The
                          PCR primer and sequencing locations and their orientation are shown by the arrows.
                  11    High Resolution MICA Genotyping by Sequence-Based Typing (SBT)               187
                          Fig. 2. PCR products of MICA gene amplification. 4 mL of PCR products from DNA samples
                          were loaded to 2% gel for electrophoreses analysis. Bands with molecular weight at
                          2.2 kb are shown.
3.3. Cleaning the MICA    The PCR products have to be purified before they are used as
DNA Templates             sequencing templates, because residual PCR primers and nucle-
                          otide triphosphates (dNTPs) can interfere with the SBT chemistry
                          resulting in lower data quality. An easy way to perform PCR
                          purification is by using exonuclease I and shrimp alkaline phos-
                          phatase (ExoSAP-IT) that will remove unincorporated primers and
                          dNTPs.
                           1. Take the vial of ExoSAP-IT from the freezer (see Note 4).
                           2. Add 3.6 mL of ExoSAP-IT to the well containing the PCR
                              products.
                           3. Seal the purification tray with micro amp full plate covers or
                              caps.
                           4. Centrifuge at 1,000 × g for 30 s.
                           5. Place in the thermal cycler and run the following profile.
                                                          37°C       30 min
                                                          80°C       15 min
                                                          4°C        Infinite
3.4. Preparation          There are four sequencing primers (Table 1) for SBT.
for MICA Allele
                           1. Prepare the Sequencing Mix. The following table is for each
Sequencing
                              reaction. To the sequencing plate add 7.25 mL of water, 2 mL
                              of purified PCR product, and 2 mL of primers. Then mix with
                              BigDye and buffer.
                 11     High Resolution MICA Genotyping by Sequence-Based Typing (SBT)           189
                                Keep at 4°C.
                             4. Following cycle sequencing, the sequencing reaction products
                                have to be purified to remove non-incorporated dye termina-
                                tors which would otherwise cause sequencing artifacts.
3.5. EDTA/NaOAc/            After Cycle sequencing is complete, remove plate from thermo-
Ethanol Precipitation       cycler and place at 4 or −20°C until ready to precipitate. Do not let
of Cycle-Sequenced          plate sit in thermocycler overnight (low sample volume leads to
Products                    easy evaporation). This protocol assumes a 15-mL cycle-sequencing
                            reaction.
                             1. To each well add 2.5 mL of NaOAc/EDTA (4 part of 3M
                                sodium acetate pH 5.5 plus 1 part of 0.5M EDTA pH 8.0).
                                Mix with pipette tips.
                             2. Add 25 mL 100% EtOH (from freezer) to each. Mix with
                                pipette tips. Replace sealing tape, seal very well and vortex for
                                1 min, and incubate at room temperature for 15 min.
                             3. Centrifuge at 2,500 × g for 30 min.
                             4. Invert plate onto paper towel and spin at 185 × g for 1 min to
                                dry.
                             5. Add 50 mL 80% EtOH to each well and mix with pipette tips.
                             6. Do not vortex and make EtOH fresh to ensure accurate con-
                                centration (see Note 8).
                             7. Centrifuge at 2,500 × g for 15 min.
190        Y. Zou and P. Stastny
                                8. Flick plate contents into sink, invert plate onto paper towel,
                                   and spin at 185 × g for 1 min to dry. Start timer immediately.
                                9. Resuspend samples in 10 mL Hi-Diformamide. Cover plate
                                   with sealing tape, vortex thoroughly (15 s), and denature in
                                   thermocycler (96°C for 3 min).
                              10. Centrifuge for 30 s at 1,000 rpm and place it on the support
                                  base, specific for each analyzer.
                              11. Place a full plate over the tray and follow with the 96-well plate
                                  retainer.
                              12. Link tray map to tray ID and click on the green arrow to start
                                  the run.
                              13. The run module for this procedure takes about 90 min for
                                  each injection (see Note 9) on the 3130XL ABI Sequencer,
                                  when using a 50 cm long capillary array (see Note 10).
3.6. Analysis                 The raw data results are analyzed by the ABI Sequence Analysis
of Allele-Level Typing        program and saved in four electronic files (SampleID_MICA_1F.
by Computer Program           ab1, SampleID_MICA_2R.ab1, SampleID_MICA_3F.ab1, and
                              SampleID_MICA_4R.ab1). The segments of MICA containing
                              exons 2, 3, 4, and 5 are assembled and the aligned sequences (see
                              Note 11), with MICA*001 as consensus, are analyzed with a pro-
                              gram for assignment of MICA alleles (see Note 12, Fig. 3). This
                              software was designed to analyze nucleotide substitutions at poly-
                              morphic sites using an updated MICA allele database (see Note 13).
Fig. 3. Alignment of MICA sequences by SBT. This aligned sequence is generated from four sequencing files (see Note
15). Sequences in exon 2, 3, 4, and 5 are assembled as in this alignment window by a computer program. Their intron
sequences were removed.
           11   High Resolution MICA Genotyping by Sequence-Based Typing (SBT)                  191
                     Table 2
                     Alignment of the STR in the transmembrane region
                     of the MICA genes (5¢, 5¢–3¢)
                     A4        GCTGCTGCTGCTATTTTTGTTATTATTATTTTCTATGT
                     A5        GCTGCTGCTGCTGCTATTTTTGTTATTATTATTTTCTA
                     A5.1      GCTGCTGGCTGCTGCTATTTTTGTTATTATTATTTTCT
                     A6        GCTGCTGCTGCTGCTGCTATTTTTGTTATTATTATTTT
                     A7        GCTGCTGCTGCTGCTGCTGCTATTTTTGTTATTATTAT
                     A9        GCTGCTGCTGCTGCTGCTGCTGCTGCTATTTTTGTTAT
                     A10       GCTGCTGCTGCTGCTGCTGCTGCTGCTGCTATTTTTGT
                    MICA STR is located in the exon 5 with the different number of “GCT” repeat. MICA
                    allele named by WHO HLA nomenclature is based on the sequences of MICA gene
                    including STR type
4. Notes
Fig. 4. The combined signals from two different STR types in one sample. Five different sequencing results of the TM
polymorphisms are shown with the overlay signals matching the combined TM types from A4/A9, A5/A5.1, A5.1/A6, A5/
A6, or A6/A9. Computer typing for MICA TM is based on this algorithm. It is available at this web address: http://www4.
utsouthwestern.edu/stastny-lab/download_page.htm.
11   High Resolution MICA Genotyping by Sequence-Based Typing (SBT)       193
                           Table 3
                           Alleles with identical extracellular domains (exons 2, 3, and 4)
                           and their associated transmembrane types (exon 5)
                           1                           MICA002:01                              A9
                                                       MICA020                                 A10
                                                       MICA023                                 A5.1
                                                       MICA050                                 A7
                           2                           MICA00801                               A5.1
                                                       MICA027                                 A5
                                                       MICA048b                                A5
                           3                           MICA009:01                              A6
                                                       MICA049b                                A6
                           4                           MICA007:01                              A4
                                                       MICA026                                 A6
                       a
                        Groups of alleles with identical sequences in exons 2, 3, and 4. The differences are
                       either in TM type (group 1, 4) or
                       b
                         Different only in exon 6 (group3) or both (group 2)
References
 1. Zwirner NW, Fernandez-Vina MA, Stastny P             7. Petersdorf EW, Shuler KB, Longton GM et al
    et al (1998) MICA, a new polymorphic HLA-               (1999) Population study of allelic diversity in
    related antigen, is expressed mainly by kerati-         the human MHC class I-related MIC-A gene.
    nocytes, endothelial cells, and monocytes.              Immunogenetics 49:605–612
    Immunogenetics 47:139–148                            8. Komatsu-Wakui M, Tokunaga K, Ishikawa Y
 2. Stephens HA (2001) MICA and MICB genes:                 et al (1999) MIC-A polymorphism in Japanese
    can the enigma of their polymorphism be                 and a MIC-A-MIC-B null haplotype.
    resolved? Trends Immunol 22:378–385                     Immunogenetics 49:620–628
 3. Zwirner NW, Marcos CY, Mirbaha F et al               9. Ahmad T, Marshall SE, Mulcahy-Hawes Y et al
    (2000) Identification of MICA as a new poly-            (2002) High resolution MIC genotyping:
    morphic alloantigen recognized by antibodies            design and application to the investigation of
    in sera of organ transplant recipients. Hum             inflammatory bowel disease susceptibility.
    Immunol 61:917–924                                      Tissue Antigens 60:164–179
 4. Cerwenka A, Lanier LL (2003) NKG2D                  10. Ota M, Katsuyama Y, Mizuki N et al (1997)
    ligands: unconventional MHC class I-like mol-           Trinucleotide repeat polymorphism within exon
    ecules exploited by viruses and cancer. Tissue          5 of the MICA gene (MHC class I chain-related
    Antigens 61:335–343                                     gene A): allele frequency data in the nine popu-
 5. Parmar S, Del Lima M, Zou Y et al (2009)                lation groups Japanese, Northern Han, Hui,
    Donor-recipient mismatches in MHC class I               Uygur, Kazakhstan, Iranian, Saudi Arabian,
    chain-related gene A in unrelated donor trans-          Greek and Italian. Tissue Antigens 49:448–454
    plantation lead to increased incidence of acute     11. Zou Y, Han M, Wang Z et al (2006) MICA
    graft-versus-host    disease.      Blood    114:        allele-level typing by sequence-based typing
    2884–2887                                               with computerized assignment of polymorphic
 6. Zou Y, Stastny P, Süsal C et al (2007) Antibodies       sites and short tandem repeats within the trans-
    against MICA antigens and kidney-transplant             membrane region. Hum Immunol 67:
    rejection. N Engl J Med 357:1293–1300                   145–151
                                                                                        Chapter 12
Abstract
In this chapter, we outline some basic principles for the consistent management of immunogenetic data.
These include the preparation of a single master data file that can serve as the basis for all subsequent analyses,
a focus on the quality and homogeneity of the data to be analyzed, the documentation of the coding
systems used to represent the data, and the application of nomenclature standards specific for each immu-
nogenetic system being evaluated. The data management principles discussed here are intended to provide
a foundation for the data analysis methods detailed in Chaps. 13 and 14. The relationship between the data
management and analysis methods covered in these three chapters is illustrated in Fig. 3.
      The application of these data management principles is a first step toward consistent and reproducible
data analyses. While it may take extra time and effort to apply them, we feel that it is better to take this
approach than to assume that low data quality can be compensated for by large sample sizes.
      In addition to their relevance for analytical reproducibility, it is important to consider these data man-
agement principles from an ethical perspective. The reliability of the data collected and generated as part
of a research study should be as important a component of the ethical review of a research application as
the security of those data. Finally, in addition to ensuring the integrity of the data from collection to pub-
lication, the application of these data management principles will provide a means to foster research integ-
rity and to improve the potential for collaborative data sharing.
Key words: Data management, Data standards, High polymorphism, HLA, Immunogenetics, KIR
1. Introduction
1.1. Avoiding the               In recent years, large amounts of genetic data have become avail-
“Garbage-In Garbage             able to the research community, increasing the potential for new
Out” Predicament                findings in ways that have not previously been possible. The need
                                for standardized data management and statistical analysis increases
                                as more data become available in order to mitigate the variability
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_12, © Springer Science+Business Media New York 2012
                                                         197
198      P.-A. Gourraud et al.
2. Data Organization
and Storage
2.1. Organization         The “master data file” is the foundation of a quality data analysis.
of a Master Data File     Ideally, a master data file will contain all the data needed for the
                          analysis in a single electronic document, which can be validated,
                          transferred, and exported. While individual software tools may
                          require data to be organized in different ways (i.e., different data
                            12     Standard Methods for the Management of Immunogenetic Data                                 199
Fig. 1. The benefits of a single master data file. A master data file integrates all data pertinent to a given study in a standard
fashion. Genotype and phenotype data that must be compiled for a research project are shown on the right. These are all
incorporated into a single master data file, which is stored in a database, or as an MS Excel table.
                                  input formats), the master data file should be the source of all data
                                  analyzed regardless of the organization of those data. This first step
                                  crystallizes all the thought that has been put in the study design
                                  and the development of the hypothesis(ses) under investigation.
                                  Study design is beyond the focus of this chapter but sample selec-
                                  tion, phenotype assessment, and required sample size computations
                                  clearly warrant close attention. As shown in Fig. 1, the master data
                                  file gathers demographic, phenotypic, and genotypic information;
                                  see supplementary Table S1 for a sample master data file.
2.2. Characteristics              A typical master data file consists of (1) a series of rows dedicated
of a Master Data File             to each analytical unit of the study and (2) a series of columns each
                                  consisting of either a “primary” or “derived” variable.
2.2.1. Units of Analysis          The master data file is generally structured using a single row dedi-
                                  cated to each analytical unit of the study. The analytical unit is the
                                  entity for which the required items of data have been collected and
                                  compiled. In general, analytical units are individual subjects,
                                  although in some cases the major entity being analyzed in a study
                                  is not an individual; for example, a transplantation study may focus
                                  on donor/recipient pairs, or an epidemiological study may focus on
                                  cases and controls. When nuclear families are analyzed, it may also
                                  be more appropriate to consider the nuclear family itself as the
                                  analytical unit of the study rather than its constituent members.
                                  In these cases, data on more than one individual may be included
                                  on a single row of the master data file. Conversely, when the study
                                  design involves repeated measures (over time or under different
200         P.-A. Gourraud et al.
2.2.2. Variables             Each column (or field) in the master data file should be dedicated to
                             a variable. A variable is an observed or measured characteristic that
                             can have more than one value and to which a numerical measure
                             or a category from a classification can be assigned. It describes a
                             single item in the data set. Where possible, each column should be
                             numerically coded, and missing values must be given a conven-
                             tional code as well. It is often useful to give each variable a short
                             (but informative) name, which is stored in the first row of each
                             column (also known as the column header). These variables are
                             sometime called “raw” or “primary” variables, because they directly
                             encode information; “analysis” or “derived” variables encode
                             information that can be derived from the raw variables, and can
                             be included in the master data file as well. For example, body mass
                             index (BMI) can be calculated from weight and height variables,
                             but it may be useful to include a calculated BMI in the master data
                             file as well. The master data file may include other variable fields
                             that will contain variable data generated as part of the analyses.
                                  In general, the master data file should be designed to exclude
                             redundancies between fields. At this early stage of the analysis, it is
                             also important to identify the type of the variables being included
                             in the master data file; the two most important variable types are
                             “quantitative” and “qualitative” variables, and the statistical
                             method that will be applied will depend on the type of variable
                             being analyzed.
                               1. Quantitative variables
                                       Quantitative variables provide numerical measures of a
                                  given characteristic being observed, and can be either “continu-
                                  ous” or “discrete.” Continuous quantitative variables have an
                                  infinite number of possible values that are not countable,
                                  whereas discrete quantitative variables have either a finite
                                  number of possible values or a countable number of possible
                                  values. For example, in hematopoietic stem cell allogeneic
                                  transplantation, the dose of CD34+ cells (e.g., 4.61 × 106 kg−1
                                  of body weight) is a continuous quantitative variable. When
                                  these values are rounded (e.g., to 5 × 106 kg−1 of body weight),
                                  the variable may be considered as a discrete quantitative
                                  variable. In general, mathematical and statistical operations
                                  can be carried out on quantitative variables.
                               2. Qualitative variables
                                      Qualitative (or categorical) variables allow for classification
                                  of individuals based on a characteristic, and can be either
                                  “nominal” or “ordinal.” Nominal qualitative variables define
                                  unordered or non-hierarchical categories, whereas ordinal
                           12   Standard Methods for the Management of Immunogenetic Data                            201
2.2.3. Data Dictionary          In addition to the numerically encoded variables, a master data file
                                should also contain (or be associated with) a dictionary defining
                                each variable and its associated coded values. The minimal items
                                for this data dictionary are the short name of the variable, a detailed
                                description of its content, a specification of the meaning of each
                                associated numerical code, and the code used for missing values.
                                Any additional information or comment that can be useful for a
                                better characterization of the variable has to be mentioned. This
                                effort will ensure sustainability of the data.
                                     Figure 2 shows an example of the data structure in a master
                                data file: numerically encoded data and data dictionary; see
Fig. 2. Characteristics of a Master Data File and associated Data Dictionary. A data dictionary provides context for the
numerically encoded observations recorded in the master data file. Each field (column) in the master data file corresponds
to a row in the data dictionary, where it is described in a standardized fashion.
202      P.-A. Gourraud et al.
2.3. Storage of Data      The best way to appropriately organize the data usually requires
                          building a database. Because many research groups do not have the
                          information technology (IT) resources to do so, a spreadsheet
                          application (e.g., Microsoft Excel or its equivalent) is often used
                          instead. Although very easy to use, accidental sorting, deletions,
                          and unintended changes to the data can occur with such spread-
                          sheet applications. For example, when HLA allele names are stored
                          in Excel, it is common for allele names that should be treated as
                          text (e.g., “0101” or “01:01”) to be treated as numbers or times
                          (e.g., 101 or 1:01). Such data rearrangements may be difficult to
                          detect and will certainly affect the results of the study. If spread-
                          sheet applications are used to manage allele name data, individual
                          datasets should be stored and transmitted as text-formatted “flat
                          files,” and measures should be taken to ensure the validity of allele
                          names prior to analysis.
3. Nomenclature
Management
                          The high degree of polymorphism observed for immunogenetic
                          genes and loci, coupled with the structural variation of immune
                          genes, has resulted in regularly updated allele name nomenclatures
                          that keep pace with the growing complexity of these systems, but
                          each nomenclature revision brings with it the potential for confu-
                          sion when data generated using different nomenclature versions
                          are analyzed together (the so-called immunogenetic tower of
                          Babel) (1). This is a common problem for HLA and KIR genes,
                          alleles, and haplotypes; MHC microsatellites, single nucleotide
                          polymorphism (SNP), copy number variants (CNVs), and small
                          insertion-deletion changes (indels). It is essential that the data in
                          an analysis share a common nomenclature, and that the specific
                          nomenclature version under which a dataset was generated (or
                          validated if the dataset is compiled from multiple sources) is
                          identified in the master data file.
3.1. HLA Nomenclature     The HLA nomenclature for allele names has changed considerably
Variation                 over the last several decades. Each allele name contains a string of
                          numbers arranged in “domains” that correspond to polymorphisms
                          at the serological, protein, synonymous nucleotide, noncoding
                                                                                                                                                                                          12
                                                                                                                                                                                          Standard Methods for the Management of Immunogenetic Data
Fig. 3. A roadmap to the landscape of statistical techniques. This roadmap is a guide summarizing potential approaches to data analysis. Starting with raw data in the center, analyses
                                                                                                                                                                                          203
proceed first through an assessment of missing data, and then the application of classical descriptive statistical techniques (both numerical and graphical), before a set of final
statistical tests, specific to the type of data and the hypothesis being tested, and which inform the interpretation of the data, are selected.
204        P.-A. Gourraud et al.
3.1.1. Antigen Recognition   Rather than treating each allele as distinct, it is often useful to ana-
Sequence Nomenclature        lyze alleles that share the peptide sequences that constitute the
                             antigen recognition sequence (ARS) or that share the nucleotide
                             sequences that encode the ARS as a combined category. The ARS is
                             alternatively referred to as the peptide-binding domain or peptide-
                             binding region (respectively abbreviated as PBD or PBR). The ARS
                             of class II HLA molecules is encoded by exon 2, and the class I
                             ARS is encoded by exons 2 and 3. While the version 2 HLA nomen-
                             clature was in effect, allele-codes identifying alleles that share the
                             same ARS encoding exon sequences were developed. For example,
                             the “A*010101g” and “A*02 G1” codes have been used to repre-
                             sent all HLA-A alleles that shared exon 2 and 3 nucleotide sequences
                             with the A*010101 allele (4, 5).
                                  Version 3 of the HLA nomenclature includes official codes for
                             identifying alleles that share an ARS or ARS-encoding exon
                             sequence. Alleles that share an identical ARS are included in a P
                             group (e.g., A*01:01P includes all HLA-A alleles that share the
                             ARS with the A*01:01:01:01 alleles), and alleles that share identi-
                             cal exon 2 or exons 2 and 3 nucleotide sequences are included in a
                             G group (e.g., A*01:01:01G includes all HLA-A alleles that share
                             their exon 2 and exon 3 sequences with A*01:01:01:01) (3).
3.1.2. Nomenclature          Several resources are available for the conversion of allele names
Conversion Resources         between nomenclature versions. The ImMunoGeneTics (IMGT)/
                             HLA web site and database provides tables for the conversion of
                             version 2 allele names to version 3 names, and houses a web-based
                             conversion tool for individual allele names at http://www.ebi.
                             ac.uk/imgt/hla/convert_name.html (6).
                                 Entire datasets can be converted from nomenclature version 2
                             to version 3 using either the Allele Name Translation Tool (ANTT)
                             or Update NomenCLature (UNCL), tools available from the
                                                                                                                                                                                         12
    Table 1
    Features of the three nomenclature versions for classical HLA allele names
3.1.3. Allele Name          Because the polymorphic domains that make up an HLA allele
Truncation                  name are arranged in a hierarchical fashion, it is possible to delete
                            domains from the right end of an allele name while retaining
                            important information for analysis. This process of deleting
                            domains is known as truncation (aka right-truncation), and it is
                            important to ensure that truncation has been carried out consis-
                            tently within a dataset. For example, the full-length A*01:01:01:01
                            allele name can be truncated to A*01:01:01, A*01:01, and A*01.
                            In some cases, truncation of allele names may be equivalent to
                            applying a P or G group code, but this is not always the case.
                            Depending on the research question being investigated, different
                            levels of truncation may be appropriate (e.g., it may be appropriate
                            to truncate allele names to the peptide level for a study of peptide
                            presentation, but not for a study of allelic diversity and evolution),
                            but all versions of a given allele name must be truncated to the
                            lowest common level for analysis to avoid spurious results.
3.2. Microsatellite         In general, microsatellite alleles should be identified using both the
Nomenclature                number of repeats and the fragment size (10). In order to compare
(14th Workshop)             the results of several studies, the correspondence between the
                            repeat number and the various fragment lengths (depending on
                            primer pairs used) should be established. This correspondence list
                            must include specific details (e.g., in the form of UniSTS numbers)
                            of the primer pairs used to genotype each microsatellite (http://
                            www.ncbi.nlm.nih.gov/sites/entrez?db=unists). When multiple
                            synonymous names are in use for a given microsatellite allele, the
                            lowest numbered DS6 number should be used as a reference. For
                            example D6S273 should be used to refer to the following synony-
                            mous names—142XH6, AFM142xh6, GC378-D6S273. More
                            details can be found on the NCBI’s MHC database, “dbMHC”
                            (http://www.ncbi.nlm.nih.gov/projects/gv/mhc/).
3.3. Single Nucleotide      As recently discussed (1), multiple distinct identifiers may exist
Polymorphism and            for a given simple genetic marker (e.g., a SNP or indel), some-
Insertion/Deletion          times leading to confusion in the comparison of markers across
Nomenclature                studies. This is partly due to the fact that the unique reference
                            sequences applied to structurally variable and highly polymorphic
                        12   Standard Methods for the Management of Immunogenetic Data         207
3.4. SNPs: RS# and           regions like the KIR cluster and the MHC region are not appro-
HGVS Nomenclature            priate for these markers. One commonly accepted solution to
                             this problem is to use the NCBI’s dbSNP reference SNP
                             (RefSNP) accession ID (rs number or rs#) (11). However, because
                             rs numbers are not necessarily stable identifiers across successive
                             dbSNP releases/builds, all references to an rs# should be accom-
                             panied by dbSNP build number. For example, rs16375 is not in
                             use anymore in build 130 of dbSNP; rs1704 should be used
                             instead.
                                 Many other possibilities can be used as complementary indica-
                             tions which directly refer to the sequence changes (e.g.,
                             rs2306220:A>G). The other acceptable option is to use the Human
                             Genome Variation Society (HGVS) nomenclature (12, 13). For
                             rs16375/rs1704, an accessioned sequence from EBI, GenBank, or
                             DDBJ can be used as the reference sequence NT 007592.14:
                             20656832insATTTGTTCATGCCT, but many different acces-
                             sioned genomic or mRNA sequences can be used.
3.5. KIR Nomenclature        Ideally, the goal in storing and analyzing KIR data is to have
                             some data representation for each chromosome at every locus.
                             The limitations of the typing technology and the variation in
                             KIR haplotype structure dictate that in many cases the second
                             chromosome will be typed as “unknown” (?). Nevertheless, it is
                             important to have data representation for the full genotype in
                             order to facilitate downstream analyses. Each KIR locus typed
                             can have at least one full genotypic record (more in the case of
                             genotypic ambiguity, which is likely for allelic typing results).
                             When a locus is absent from a given haplotype, this absence must
                             be coded in the data (i.e., as an “absent” allele), and should be
                             denoted as “0.”
                                  Where allelic typing is available, a genotype with two alleles
                             represents the simplest case scenario, a heterozygote with the locus
                             present on both haplotypes. However, when we have only one
                             allele detected, e.g., KIR2DL2*002, there are two possible geno-
                             types: “002,002” or “002,0.” At present, most typing systems can-
                             not distinguish these. In these cases, the genotype should be treated
                             as ambiguous, i.e., 002,0/002.
                                  For typing data that is strictly presence/absence, the data can
                             be treated as for a biallelic locus: allele “1” = “present” and allele
                             “0” = “absent.” If the locus is absent, we have the full genotype
                             0,0; however for locus present, we have an ambiguous genotype,
                             which may be either “1,0” or “1,1.” This can also be represented
                             as an ambiguous genotype : 1,0/1. Because this notation does not
                             consider order, a genotype coding of “0,1” is not used, but would
                             be treated the same as “1,0.”
                                  Some KIR loci have particular additional details to consider
                             prior to analyzing data:
208   P.-A. Gourraud et al.
                         1. KIR2DL2/L3:
                                 KIR2DL2 and KIR2DL3 were formerly treated as separate
                            loci. The alleles were not named in a series; therefore, it is
                            important to distinguish between KIR2DL2 and KIR2DL3
                            when the data is recorded. In the case of this locus, since all
                            typing systems are able to detect both KIR2DL2 and KIR2DL3
                            an investigator should always have a full genotype, i.e., no
                            missing data or ambiguity with presence/absence typing.
                         2. KIR2DL5:
                                 KIR2DL5 may be either centromeric or telomeric in the
                            KIR cluster, and possibly on both ends. As such, an individual
                            may have four copies of KIR2DL5; at this time there is no way
                            to distinguish these definitively. Many typing systems will simply
                            type for the presence of KIR2DL5; if typed as “absent,” there is
                            confirmation of genotypes of 0,0 both centromerically and
                            telomerically. However, a typing of ‘present’ gives the genotype
                            1,0/1 on either or both sides of the KIR complex. Some typing
                            systems currently distinguish “KIR2DL5A” and “KIR2DL5B”;
                            all current data suggest that KIR2DL5A corresponds to the
                            telomeric position and that KIR2DL5B corresponds to the
                            centromeric position.
                         3. KIR2DS3/S5:
                                As with KIR2DL5, KIR2DS3/S5 may be either centro-
                            meric or telomeric in the KIR complex, and possibly on both
                            ends. As such, an individual may have four copies of the gene.
                            Previously, KIR2DS3 and KIR2DS5 were thought to be dif-
                            ferent loci, and all current typing systems can distinguish
                            between them. However, an individual who is typed, for exam-
                            ple, as positive for both KIR2DS3 and KIR2DS5 in a pres-
                            ence/absence typing system may have these two alleles on
                            either or both sides of the KIR cluster. Some data is emerging
                            that suggests that particular alleles of each of these may be
                            either centromeric or telomeric, but this is not yet confirmed.
                            Presently, the only way to ascertain definitively that an indi-
                            vidual has both centromeric and telomeric copies of either
                            KIR2DS3 or KIR2DS5 is the case where we have allelic typing
                            with more than two allele calls for the locus.
                         4. KIR3DL1/S1:
                                As above for KIR2DL2/L3, KIR3DL1 and KIR3DS1
                            were formerly treated as separate loci. However, it has been
                            recognized for some time that they are allotypes of the same
                            locus, and in this case alleles have been named in a series. As with
                            KIR2DL2/L3, all typing systems minimally detect KIR3DL1
                            and KIR3DS1, so there should be no missing data/ambiguity
                            in presence/absence typing systems at this locus. If a typing is
                  12   Standard Methods for the Management of Immunogenetic Data          209
Acknowledgments
Glossary
Genetic data            1. Allele: Any of the alternative forms (sets of forms) of DNA
                           sequence at locus. These variants may occur for genes and/or
                           genetic markers.
                           Example: B, HLA-DRB1*01:01:01, HLA-A*01, D6S1666
                                    (184).
                        2. Diplotype: The pair of haplotypes within a given genotype.
                           The chromosomal phase between alleles is always known.
                           Example: HLA (A*01 B*08 DR*17, A*26 B*27 DR*17).
                                    HLA (A*01 B*27 DR*17, A*26 B*08 DR*17).
                           When analyzing data for more than one locus, diplotypic data
                           must be distinguished from genotypic data. Because the alleles
                           at different loci in a given genotype can be combined to make
                           many possible haplotype pairs, a genotype must be considered
                           to correspond to multiple diplotypes. Unfortunately, the term
                           “genotype” is sometimes used to refer to a given pair of haplo-
                           types, especially when familial segregation has been studied.
                        3. Gene: The functional and physical unit of heredity. A gene
                           consists of a DNA segment with a specific sequence. It includes
                           information for the synthesis of mRNA molecules that direct
                           the synthesis of proteins.
                           Example: ABO Glycosyltransferase gene, HLA-DRB1 gene,
                                    KIR-2DS3/S5 gene.
                        4. Genotype: The genetic makeup at one or more loci of an indi-
                           vidual. It refers to a set of alleles carried by an individual
                           (regardless of the expression of those alleles). The chromo-
                           somal phase (chromosomal identity of alleles at different loci)
                           between alleles may not be known.
210     P.-A. Gourraud et al.
                                Example: HLA-A (1, 2); HLA-B (8, 44); HLA-DRB1 (03, 04).
                                         KIR: KIR (A, A).
                                         Microsatellites: D6S273 (134, 136) D6S273
                                         (*(GT)19, *(GT)20).
                                         SNP: RS345336443 (G/G).
                           5. Haplotype: Set of alleles of contiguous loci. They are usually
                              co-transmitted on a parental chromosome.
                                Example: HLA-A*01-B*08-DRB1*03.
                           6. Locus: Literally, “place” in Latin, it is the specific usual physi-
                              cal location of genes, an individual genetic marker, or set of
                              genetic markers in a genome.
                                Example: ABO locus, HLA-DRB locus, KIR-2DS3/S5cen
                                         locus, D6S1666 microsatellite locus.
                           7. Phenotype: The observable expression of alleles as a physical or
                              biochemical trait resulting from the interaction of the genome,
                              the environment, and the experimental settings. In disease
                              studies it may refer to the presence or a manifestation of the
                              disease under study. Disease phenotypes may be reflected in a
                              variety of ways as quantitative or qualitative variables.
                              This term may also refer to a set of alleles (expressed or not)
                              detected by a technique. In codominant or heterozygous situ-
                              ations, phenotypes are noted as pairs of data; each pair is specific
                              to a particular gene and locus.
                                Example: ABO system: [A]. HLA system: [HLA-A (1, 2);
                                         HLA-B (8,44); HLA-DR (3, 4)].
        include the formats and programs used to store and edit the
        data (e.g., a specific spreadsheet program or database system),
        as well as any modifications that were made to the data between
        the generation of the data resulting from the typing assay and
        the inclusion of the data in the master data file. For example, if
        ambiguities were resolved, the approach used to resolve them
        should be documented in the data dictionary; if HLA allele
        data were truncated to a common level, or “binned” into a
        common sequence category (e.g., treating all alleles that
        encode the same peptide-binding region as the same allele)
        this should documented.
     5. Ethnicity: A group of individuals (or populations) sharing a
        common language, culture, or religion, and who are assumed
        to share a common ancestry. Ethnicity should be distinguished
        from geography (e.g., “North American” is not an ethnicity),
        and though ethnicity is often associated with indigenous
        nationality (e.g., “Irish,” “Chinese”) qualifiers are often neces-
        sary to distinguish ethnicity from nationality (e.g., “Han
        Chinese”).
     6. Family: If individuals in the study belong to discrete familial
        groups, a family ID is qualitative variable identifying member-
        ship in a particular pedigree, as well as the relationship to the
        index case (proband).
     7. Geographic region: A specific continental or subcontinental
        area comprised by multiple nations in which the population is
        located, or from which the population was derived, if the popu-
        lation is a migrant population. For example, European Americans
        or European Australians would be assigned to the European
        region, or to a specific subregion of Europe. Conversely North
        America would only pertain to Native American/Amerindian/
        Aleut/Eskimo populations. Populations derived from more
        than one region (admixed populations) can be assigned to a
        specific class for the type of admixture (depending on the
        regions of origin) or included in a single class for all admixed
        populations. The definitions of each region and admixed class
        should be defined in the data dictionary.
     8. Latitude and longitude: Geographic coordinates that specify
        specific locations on the surface of the Earth. Latitude and
        longitude values should be recorded in a decimal format,
        with minutes and seconds indicated as factions of each
        degree value. North latitudes and east longitudes should be
        recorded with positive values, and south latitudes and west
        longitudes should be recorded with negative values. For
        example, 35° 20 min south latitude would be recorded as
        −35.333, and 2° 30 min east longitude should be recorded
        as 2.5 or +2.5.
212      P.-A. Gourraud et al.
References
 1. Gourraud PA, Feolo M (2010) The Babel               Nomenclature for factors of the HLA system,
    Tower      revisited:   SNPs—Indels—CNVs.           2002. Tissue Antigens 60:407–464
    Confusion in naming sequence variant always      3. Marsh SG, Albert ED, Bodmer WF, Bontrop
    rises from ashes Tissue Antigens 75:199–200         RE, Dupont B, Erlich HA, Fernández-Viña M,
 2. Marsh SG, Albert ED, Bodmer WF, Bontrop             Geraghty DE, Holdsworth R, Hurley CK, Lau
    RE, Dupont B, Erlich HA, Geraghty DE,               M, Lee KW, Mach B, Maiers M, Mayr WR,
    Hansen JA, Mach B, Mayr WR, Parham P,               Müller CR, Parham P, Petersdorf EW, Sasazuki
    Petersdorf EW, Sasazuki T, Schreuder GM,            T, Strominger JL, Svejgaard A, Terasaki PI,
    Strominger JL, Svejgaard A, Terasaki PI (2002)      Tiercy JM, Trowsdale J (2010) Nomenclature
                         12   Standard Methods for the Management of Immunogenetic Data               213
      for factors of the HLA system, 2010. Tissue       11. Sherry ST, Ward MH, Kholodov M, Baker J,
      Antigens 75:291–455                                   Phan L, Smigielski EM, Sirotkin K (2001)
 4.   Cano P, Klitz W, Mack SJ, Maiers M, Marsh             dbSNP: the NCBI database of genetic varia-
      SG, Noreen H, Reed EF, Senitzer D, Setterholm         tion. Nucleic Acids Res 29:308–311
      M, Smith A, Fernández-Viña M (2007)               12. den Dunnen JT, Antonarakis SE (2000)
      Common and well-documented HLA alleles:               Mutation nomenclature extensions and sug-
      report of the Ad-Hoc committee of the                 gestions to describe complex mutations: a dis-
      American society for histocompatibility and           cussion. Hum Mut 15:7–12
      immunogenetics. Hum Immunol 68:                   13. den Dunnen J (2010) Nomenclature for the
      392–417                                               description of sequence variants. Human
 5.   Robinson J, Mistry K, Marsh SGE (2010) Exon           Genome Variation Society. http://www.hgvs.
      identity and ambiguous typing combinations.           org/mutnomen/
      Anthony Nolan Research Institute. http://         14. Bodmer JG, Marsh SGE, Parham P, Erlich HA,
      www.ebi.ac.uk/imgt/hla/pdf/ambiguity_                 Albert E, Bodmer WF, Dupont B, Mach B,
      v2280.pdf                                             Mayr WR, Sasasuki T, Schreuder GMT,
 6.   Robinson J, Mistry K, McWilliam H, Lopez R,           Strominger JL, Svejgaard A, Terasaki PI (1990)
      Parham P, Marsh SG (2011) The IMGT/HLA                Nomenclature for factors of the HLA system,
      database. Nucleic Acids Res 39(Database               1989. Tissue Antigens 35(1):1990
      Issue):D1171–D1176                                15. Who Nomenclature Committee (1988)
 7.   Mack SJ, Hollenbach JA (2010) Allele Name             Nomenclature for factors of the HLA system,
      Translation Tool and Update NomenCLature:             1987. Tissue Antigens 32:177–187
      software tools for the automated translation of   16. Bodmer JG, Marsh SG, Albert ED, Bodmer
      HLA allele names between successive nomen-            WF, Dupont B, Erlich HA, Mach B, Mayr WR,
      clatures. Tissue Antigens 75:457–461                  Parham P, Sasazuki T et al (1991) Nomenclature
 8.   Helmberg W, Lanzer G, Zahn R, Weinmayr B,             for factors of the HLA system, 1990. Hum
      Wagner T, Albert E (1998) Virtual DNA                 Immunol 31(3):186–194
      analysis—a new tool for combination and stan-     17. Bodmer JG, Marsh SG, Albert ED, Bodmer
      dardised evaluation of SSO, SSP and sequencing-       WF, Bontrop RE, Charron D, Dupont B,
      based typing results. Tissue Antigens 51:             Erlich HA, Mach B, Mayr WR (1995)
      587–592                                               Nomenclature for factors of the HLA system,
 9.   Helmberg W (2000) Storage and utilization of          1995. Tissue Antigens 46:1–18
      HLA genomic data—new approaches to HLA            18. Bodmer JG, Marsh SG, Albert ED, Bodmer
      typing. Rev Immunogenet 2:468–476                     WF, Bontrop RE, Charron D, Dupont B,
10.   Gourraud PA, Cambon-Thomsen A, Dauber                 Erlich HA, Fauchet R, Mach B, Mayr WR,
      EM, Feolo M, Hansen J, Mickelson E, Single            Parham P, Sasazuki T, Schreuder GM,
      RM, Thomsen M, Mayr WR (2007)                         Strominger JL, Svejgaard A, Terasaki PI (1997)
      Nomenclature for HLA microsatellites. Tissue          Nomenclature for factors of the HLA system,
      Antigens 69(Suppl 1):210–213                          1996. Tissue Antigens 49:297–321
                                                                                        Chapter 13
Abstract
In this chapter, we describe analyses commonly applied to immunogenetic population data, along with
software tools that are currently available to perform those analyses. Where possible, we focus on tools that
have been developed specifically for the analysis of highly polymorphic immunogenetic data. These analytical
methods serve both as a means to examine the appropriateness of a dataset for testing a specific hypothesis,
as well as a means of testing hypotheses. Rather than treat this chapter as a protocol for analyzing any
population dataset, each researcher and analyst should first consider their data, the possible analyses,
and any available tools in light of the hypothesis being tested. The extent to which the data and analyses
are appropriate to each other should be determined before any analyses are performed.
Key words: Data analysis, Highly polymorphic, HLA, Immunogenetics, KIR, Population study
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_13, © Springer Science+Business Media New York 2012
                                                         215
216   S.J. Mack et al.
2. Data Reporting
3. Analyses
                                General
                                statistical
                                software
 Analysis                       (e.g., SAS) MS Excel GenAlExa PyPopb Arlequinc EstiHaplod PHYLIPe Structuref CLUTOg PLINKh R packagei GenePopj
 Carrier frequency              +              +             ++          −                       −              −           −                        −
                                                                                                                                                                                               S.J. Mack et al.
   estimation
 Hardy–Weinberg                 −              −             +           +++        ++           −              −           −                        +                         ++
 Haplotype estimation           −              −             +           +++        ++           +++            −           −             +++        +         haplo.stats
                                                                                                                                                               ++
 Linkage disequilibrium −                      −                         +++        ++           −              −           −             +++        +                         ++
 Measures of selection          −              −             ++          +++        ++           −              −           −             −          −
 Measurement of genetic −                      −             ++          −          ++           −              ++          ++            +++        −         ++              ++
  differentiation
 Principal component            −              −             ++          −                       −              −           −             −          −         ++
   analysis
 Phylogenetic analysis          −              −                         −          −            −              ++          +             +++        −         ++
 Population structure           −              −                         −          ++           −              −           ++            +++        −
   analysis
− = analysis is not possible with that tool; + = analysis is possible, but the tool is not recommended for this analysis; ++ = analysis is possible with this tool; +++ = this tool has been
optimized for the analysis of immunogenetic data
a
  http://www.anu.edu.au/BoZo/GenAlEx/
b
  http://www.pypop.org
c
  http://cmpg.unibe.ch/software/arlequin3/
d
  http://www.methodomics.com/estihaplo
e
  http://evolution.genetics.washington.edu/phylip.html
f
  http://pritch.bsd.uchicago.edu/structure.html
g
  http://glaros.dtc.umn.edu/gkhome/views/cluto/
h
  http://pngu.mgh.harvard.edu/~purcell/plink/
i
 http://www.r-project.org/
j
 http://genepop.curtin.edu.au/
  13   Analytical Methods for Immunogenetic Population Data      219
                         GF = 1 − 1 − CF.
       However, Lynch and Milligan (6) have shown that this
   provides a downwardly biased estimate and suggest that a better
   estimate is obtained by:
              GF = 1 − (x 1/ 2 (1 − (var(x ) / 8x 2 ))−1 ).
   where x = 1−CF, and var(x) = x(1−x)/N, where N is the num-
   ber of sampled individuals.
       Table 2 presents gene frequencies estimated using each of
   these methods (formula a and formula b) in comparison to
   frequencies calculated by direct counting.
3. Confidence intervals
       Any estimated GF^ should be accompanied by a confidence
   interval (CI), a range of values that reveals the precision of the
   estimated frequency. The likelihood that the CI includes the
   actual population GF is given by the confidence level (usually,
   a 95% chance). For GF estimated from a sample of size n, the
   CI has a lower bound of:
    Table 2
    Comparison of methods for determining gene frequencies for present/absent data
3.2. Hardy–Weinberg              The Hardy–Weinberg (HW) principle provides a useful model for
Testing                          primary quality control (QC) verification of the integrity of geno-
                                 type data, as genotyping errors may result in both individual
                                 genotype deviations and overall deviations from HW equilibrium
                                 (HWE) (see Note 1). In addition, HW testing is also useful for
                                 detecting sampling errors (see below) in population samples.
                                 Confidence in the accuracy of Hardy–Weinberg testing is therefore
                                 crucial for confidence in subsequent analyses, as many analytical
                                 methods (e.g., LD and haplotype estimation, Ewens–Watterson
                                 analyses of selection) are predicated on an assumption of HWE in
                                 the data set. In a Hardy–Weinberg test, observed genotype counts
                                 are compared to those expected under Hardy–Weinberg equilib-
                                 rium proportions (HWEP), as calculated by generating a table
                                 of all possible genotypes, using an appropriate statistical method.
                                 The relationship between the allele and genotype frequencies under
                                 HWEP is given as:
f (Ai Ai ) = pi 2
and
                                                             (      )
                                                           f Ai A j = 2 pi p j ,
              No Allele "Lumping"
                                                            Number of Each Genotype Expected
            Allele   Count Frequency                  1        2      3       4       5       6       7       8
             1        12      0.29            1      3.43
             2        10      0.24            2      5.71     2.38
             3        9       0.21            3      5.14     4.29   1.93
             4        5       0.12            4      2.86     2.38   2.14    0.60
             5        2       0.05            5      1.14     0.95   0.86    0.48    0.10
             6        2       0.05            6      1.14     0.95   0.86    0.48    0.19    0.10
             7        1       0.02            7      0.57     0.48   0.43    0.24    0.10    0.10    0.02
             8        1       0.02            8      0.57     0.48   0.43    0.24    0.10    0.10    0.05    0.02
            Total     42       1
Fig. 1. Sparse cells and the “lumping” of alleles into combined classes. The effect of creating a combined, “lumped” allele
class on expected genotype counts is illustrated in the upper and lower halves. In the upper half, expected genotype counts
are calculated for all eight alleles; the expected counts for the ten genotypes comprising alleles 5, 6, 7, and 8 (shaded) are
much less than 1. In the lower half, alleles 5–8 have been lumped into the “5–8” allele class, and no genotype in the table
has an expected count less than 0.6.
                                     2. Exact tests
                                             An exact test for HWEP was developed by Louis and
                                        Dempster (7). This test generated all possible tables of geno-
                                        types (based on observed allele frequencies) when the sample
                                        size and allele frequencies are held constant in accordance with
                                        the exact distribution. The p-value was given by the cumulative
                                        conditional probability of obtaining a table of genotypes (with
                                        sample size and allele frequencies equal to the observed sam-
                                        ple) with a conditional probability less than or equal to that of
                                        the genotypes in the observed sample (8, 9). This test provides
                                        the exact p-value for every sample and it does not require input
                                        parameters that may affect the result. However, the number of
                                        possible tables of genotypes grows exponentially as either the
                                        sample size (n) or the number of distinct alleles (k) increases,
                                        reducing the feasibility of this test when n and k are large.
                                     3. Resampling approximations
                                            Resampling approximations to complete enumeration of all
                                        possible tables were developed for data sets with larger num-
                                        bers of alleles, where the asymptotic chi-square test may be par-
                                        ticularly problematic and exact tests with complete enumeration
                                        were not possible (10–13). While these approximation or resa-
                                        mpling tests are often erroneously referred to as “exact” tests,
                                        they do not perform a true exhaustive search for all possible
                                        tables of genotypes. These methods generally use the Monte
                                        Carlo (MC) simulation method to approximate to the exact
                    13   Analytical Methods for Immunogenetic Population Data      223
3.3. Haplotype   Estimated haplotypes and haplotype frequencies play a central role
Estimation       in most genetic studies. Haplotype-level analyses are important to
                 studies of the etiology of human disease, selective forces acting on
                 populations, and optimal sizes for bone-marrow donor registries
                 (BMDRs). Associations between markers and disease loci that are
                 not evident with a single-marker locus may be identified in mul-
                 tilocus marker analyses using estimated haplotype frequencies
                 (HFs). The design of studies and the recruitment of the samples
                 are dependent on the possibility of identifying haplotypes by segre-
                 gation analysis in families or estimating haplotypes from population
                 samples of phase-unknown unrelated individuals (18). Haplotypes
224       S.J. Mack et al.
3.3.1. Expectation-          Early work on the estimation of haplotype frequencies from unre-
Maximization Algorithm       lated genotype data was based on the expectation-maximization
                             (EM) algorithm with the assumption of HWEP at the locus-level
                             (20–24). Later work refined, explored, and extended aspects of the
                             algorithm (25–30). Application to haplotypes of SNPs (31–33)
                             and Bayesian methods (34, 35) are commonly used. It remains
                             unclear whether the Bayesian algorithms perform better than max-
                             imum likelihood implemented in EM algorithm (35).
                                 Haplotypes can be estimated using a number of software tools,
                             for example, in a standard implementation of the Expectation-
                             Maximization (EM) algorithm in the haplo.stats package for R, the
                             open source language, and environment for statistical computing
                             (1). Although there is a great desire within the immunogenetic
                             community for applications capable of analyzing very large (>mil-
                             lion individuals) data sets, available HF and LD estimation soft-
                             ware are generally limited in their capacity to a few thousands of
                             individuals. For example, precompiled versions of PyPop are cur-
                             rently limited to 7 loci and 5,000 individuals when it comes to
                             estimating haplotypes and calculating LD values. In contrast,
                             haplo.stats can accommodate very large datasets, depending on the
                             number of alleles at each locus; for example, haplo.stats estimates
                             haplotypes for 240,000 individuals over four loci with an average
                             of 25.5 alleles per locus, or 60,000 individuals over 50 loci with
                             the same mean number of alleles. Supplementary Table S6 is a
                             haplo.stats-formatted version of the synthetic genotype data in
                             Supplementary Tables S1–S4. The “master data file” described in
                             Chap. 12 of this volume (Chap. 12 Supplementary Table S1) can
                             also serve as a haplo.stats input file.
                                 Population-level haplotype frequencies are estimated via EM
                             using simultaneous maximum-likelihood estimation of n-locus
                             haplotype frequencies. The expectation step determines the
                             expected number of copies for each haplotype contributing to a
                             given genotype. For a three locus haplotype, this is calculated as:
                                      sample sizes are usually too small to cover the extent of the
                                      haplotype diversity. As a result, the haplotype frequencies and
                                      linkage disequilibrium between alleles are overestimated; such
                                      a bias would occur even if the chromosome phase was known.
                                  3. HF Estimation for KIR
                                          Because some KIR genes are present only on certain haplo-
                                     types, the space of possible KIR haplotypes excludes some locus
                                     combinations that could be generated from the observed geno-
                                     typic data. The EM algorithm for estimating KIR HFs must
                                     be modified to account for this reduced combinatorial space,
                                     e.g., using an a priori list of known/possible haplotypes to
                                     constrain the EM algorithm (39, 40). The user-designated a
                                     priori haplotype list is said to span a set of observed genotypes
                                     if each observed genotype can be generated from at least one
                                     pair of haplotypes in the list. If the list does not span the
                                     observed genotypes, the resulting estimates must be carefully
                                     interpreted.
                                          Several recent KIR HF estimation studies have noted short-
                                     comings in the use of such constraints, imposed by the need to
                                     specify predefined haplotype patterns (Fig. 2). Yoo et al. (40)
                                     found that accuracy measures related to haplotype identification
                                     were particularly low for fewer than 200 individuals and sug-
                                                                          Generate
               HLA                                                       All Possible
                                                                                            KIR Reference
                                                                                             Haplotypes
                                                                             Pairs
                                      Determine
               Observed HLA                                                                 Observed KIR
                                     All Possible
                Genotypes            Haplotypes                         KIR                  Genotypes
                                                2 Classes of
                                                Observed Genotypes:
                                                a) Unambiguous Haplotypes
                                                b) Ambiguous Haplotypes
                                                          E-M
                                                       Algorithm
                                                      Haplotype
                                                      Frequency
                                                       Estimates
Fig. 2. Overview of HLA haplotype estimation and KIR haplotype estimation strategies. The EM algorithm for estimating KIR
haplotype frequencies (HFs) can be modified from the standard approach applied to HLA genotypes (upper-left box) to
account for a reduced combinatorial space using a set of reference haplotypes as an a priori list of known/possible haplo-
types to constrain the algorithm (upper-right box).
                                 13    Analytical Methods for Immunogenetic Population Data       227
                                  where Dmax is the lesser of piqj and (1−pi)(1−qj), when Dij is <0
                                  and pi(1−pi) and qj(1−qj), when Dij is >0.
                               2. r2
                                      The r2 measure is another means of normalizing Dij to
                                  account for differing allele frequencies. This is the square of the
                                  correlation coefficient (r) between the alleles at the p and q loci
                                  (42). Because r is given as:
3.4.2. Global LD Statistics   For loci with more than two alleles, global LD statistics extend the
                              haplotype-level statistics to account for all possible combinations
                              of alleles at each locus (43).
                               1. Wn
                                      Wn is a multiallelic extension of the correlation measure r.
                                  The chi-square value for testing the significance of LD can be
                                  written as W/(2N) where:
228        S.J. Mack et al.
                                                  Wn = W / (min(k, l ) − 1).
                                       The values of Wn fall between 0 and 1, and the significance
                                  of the overall disequilibrium is assessed using the abovemen-
                                  tioned chi-square test. It should be noted that the Wn measure
                                  is always symmetric with respect to two loci, whereas the num-
                                  ber of alleles reported at each locus can differ considerably.
                                  It is therefore important not to overinterpret values of Wn
                                  for locus pairs with highly asymmetric numbers of alleles.
                                  Finally, for biallelic loci, Wn is equivalent to r.
                               2. D¢
                                      D¢ is a second global disequilibrium statistic, which sums
                                  the absolute value of normalized Dij values over all haplo-
                                  types, weighted by the frequencies of the alleles in each
                                  haplotype (46). As with Wn, D¢ values fall between 0 (equilib-
                                  rium) and 1 (linkage). This is given as:
D ′ = ΣΣpi × q j | Dij′ |,
3.4.3. Graphical              The interpretation of LD values between many markers can be facili-
Representation of LD          tated through the graphical representation of LD patterns. Compared
Patterns                      to a tabular presentation of LD values, such visual representations
                              facilitate the identification of patterns and interesting subsets of the
                              data. So-called “heat maps” are a common means of representing
                              pairwise LD values across markers, as a half-matrix in which the
                              strength of the LD (e.g., the log of the p-value) is represented by a
                              color scale. However, most of the software tools developed for
                              graphical LD presentations represent biallelic markers and can there-
                              fore only represent average LD between multiallelic loci. Popular
                              software tools for this purpose include graphical overview of linkage
                              disequilibrium (GOLD) (47), Haploview (48), MIDAS (49), and
                              various packages in R (e.g., LDHeatmap (50)). While PyPop does
                              not generate graphical LD representations, PyPop-generated LD
                              data can be imported directly to R. To our knowledge, only MIDAS
                              will simultaneously represent the interallelic component of LD.
3.7. Measures of             The measures below are used to quantify genetic variation within
Genetic Differentiation      and between populations and to determine subdivisions (subpopu-
                             lations) of a single source (total population).
                              1. FST
                                     FST values quantify levels of population differentiation by
                                assessing the proportion of genetic variance in subpopulations
                                relative to the total genetic variance (58). FST can be calculated
                                based on a partitioning of heterozygosity:
                                                   FST = (H t − H S ) / H t ,
  13     Analytical Methods for Immunogenetic Population Data       231
3.8. Graphical              There are a variety of methods for representing genetic difference
Representations             data between subpopulations in a graphical (as opposed to tabular)
of Genetic                  format. In many cases, the graphical representation can be applied
Difference Data             independently of the measure of differentiation, so that multiple
                            different genetic differentiation measures can be compared using
                            the same graphical representation and multiple graphical represen-
                            tations can be applied to the same genetic differentiation measure.
                            Because the graphical representation usually depends on an addi-
                            tional analysis, we describe some of the commonly used represen-
                            tations here as individual analyses.
                                 In general, these representations should not necessarily be
                            thought of as providing the definitive answer to a question so much
                            as they serve as aids for the interpretation of genetic differentiation
                            data that may be too complex to present in a tabular format (as
                            with LD values). As Ada Lovelace noted, “the Analytical Engine
                            has no pretensions whatever to originate anything.” The results of
                            these methods should always be interpreted critically, and the
                            researcher who uses these methods should develop a set of criteria
                            for accepting or rejecting the results of a method before using that
                            method. Overinterpretation of any of these representations should
                            be avoided when there is no obvious historical, functional, or bio-
                            logical basis for them.
                             1. Principal component analysis
                                     Principal component analysis (PCA) is used for dimension-
                                ality reduction in a data set, identifying those elements that
                                contribute most to its variance, and is particularly useful as an
                                exploratory tool in a complex data set (68). For the representa-
                                tion of genetic differentiation analyses, it is common to present
                                the results of a PCA via multidimensional scaling (MDS),
                                where each range for a given component is presented along a
                                corresponding MDS axis (69, 70). Each data-element (a popu-
                                lation or an individual) is represented by its position relative to
  13   Analytical Methods for Immunogenetic Population Data       233
Victoria
Marie Byrd
                                                                                                      Queen
                                                                                                      Maud
                                                                                Marie Byrd
                                                                                Queen Maud
                                                                                Victoria
Fig. 5. Structure bar plot generated using the supplementary data. Structure analysis of the
synthetic data in Supplementary Table S9 with the number of clusters (K) set to 2 or 4.
Vertical bars represent each individual included in the analysis, and each tone (or color in
the electronic version) indicates the extent to which that individual’s genotype is derived
from one of the K clusters, with each tone (or color) corresponding to a cluster. Because of
the low number of loci and the extensive sharing of alleles between populations, very few
individuals are assigned to a single cluster (tone/color). However, the relative relatedness
of each population can be inferred from the tone/color compositions of their constituents.
Fig. 6. Examples of phylogenetic trees. Three representations of the same phylogeny for
four taxa (A–D). Black dots indicate nodes in each tree. The branches between each taxon
and the nearest node are known as “twigs” or “leaves”. Grey dots in the rooted trees
indicate the position of the root node. Taxa A and C are more similar to each other than
either is to taxon B or D, and taxa B and C are more similar to each other than either is to
taxon A or D. In the unrooted tree and the midpoint rooted tree, A and C are in one clade,
and B and D are in a second clade. In the outgroup rooted tree, A, B, and C are in a single
clade, to the exclusion of D.
     PHYLIP
     The PHYLogeny Inference Package (PHYLIP) (75, 76) is a
     software suite of applications for building phylogenetic trees
     using a variety of methods. Supplementary Table S10 is a PHYLIP
     GENDIST-formatted allele-frequency data file for the synthetic
     population datasets analyzed in this chapter. Figure 7 includes a
     pair of unrooted Neighbor-Joining (NJ) (77) trees generated
     using the data included in Supplementary Tables S7 and S10.
          The tree in Fig. 7a is based on Nei’s standard genetic distances
     (SGD) (78) calculated in PHYLIP, whereas the tree in Fig. 7b
     is based on population-pairwise FST values calculated in Arlequin.
     A genetic distance scale should be included with every tree.
          Steps (ii)–(iv) outlined above were carried out with PHYLIP
     to generate Fig. 7a using the GENDIST (for step ii.), NEIGHBOR
     (for step iii.), and DRAWTREE (for step iv) programs to draw
     unrooted NJ trees based on Nei’s Standard Genetic Distances.
     Figure 7b was generated using the same procedure for steps (iii)
     and (iv), but steps (i) and (ii) were carried out using Arlequin to
     generate population-pairwise FST values.
          PHYLIP’s GENDIST estimates genetic distance with three
     different measures—Nei’s SGD, Cavalli-Sforza’s chord dis-
     tance (79), and Reynold’s genetic distance (65)—and each
     measure is based on implicit assumptions that may not always
     apply to immunogenetic data. For example, all three measures
     assume that population differentiation derives from genetic
     drift, yet the HLA loci have been shown to be under balancing
     selection in numerous studies (4, 80, 81).
238   S.J. Mack et al.
                         Fig. 7. Phylogenetic trees generated using the supplementary data. (a) Unrooted
                         neighbor-joining tree generated in PHYLIP using Nei’s standard genetic distances (included
                         in Supplemental Table S10) generated for the synthetic data in Supplementary Tables
                         S1–S4. Inset bar shows a genetic distance of 0.082. (b). Unrooted neighbor-joining tree
                         generated in PHYLIP using population-pairwise FST distances generated in Arlequin for
                         the synthetic data in Supplementary Table S7. Inset bar shows a population-pairwise FST
                         distance of 0.007.
4. Notes
Acknowledgments
References
 1. R Core Development Team (2008) R: A lan-         3. Lancaster AK, Single RM, Solberg OD, Nelson
    guage and environment for statistical comput-       MP, Thomson G (2007) PyPop update—a soft-
    ing. R Foundation for Statistical Computing,        ware pipeline for large-scale multilocus popula-
    Vienna, Austria                                     tion genomics. Tissue Antigens 69(s1):192–197
 2. Li W, Hu Z, Jiang W (2010) An alphabetic list    4. Solberg OD, Mack SJ, Lancaster AK, Single
    of genetic analysis software. North Shore LIJ       RM, Tsai Y, Sanchez-Mazas A, Thomson G
    Research Institute. http://www.nslij-genetics.      (2008) Balancing selection and heterogeneity
    org/soft/. Accessed 7 Oct 2010                      across the classical human leukocyte antigen
242        S.J. Mack et al.
      loci: a meta-analytic review of 497 population     20. Piazza A (1975) Haplotypes and linkage disequi-
      studies. Hum Immunol 69(7):443–464                     libria from three-locus phenotypes. In: Kissmeyer-
 5.   Peakall R, Smouse PE (2006) GENALEX 6:                 Nielsen F (ed) Histocompatibility testing.
      genetic analysis in Excel. Population genetic          Munskgaard, Copenhagen, pp 923–927
      software for teaching and research. Mol Ecol       21. Dempster AP, Laird NM, Rubin DB (1977)
      Notes 6:288–295                                        Maximum likelihood from incomplete data via
 6.   Lynch M, Milligan BG (1994) Analysis of pop-           the EM algorithm. J R Stat Soc 39:1–38
      ulation genetic structure with RAPD markers.       22. Ott J (1977) Counting methods (EM algorithm)
      Mol Ecol 3:91–99                                       in human pedigree analysis: linkage and segrega-
 7.   Louis EJ, Dempster ER (1987) An exact test             tion analysis. Ann Hum Genet 40:443–454
      for Hardy-Weinberg and multiple alleles.           23. Yasuda N (1978) Estimation of haplotype fre-
      Biometrics 43(4):805–811                               quency and linkage disequilibrium parameter
 8.   Levene H (1949) On a matching problem aris-            in the HLA system. Tissue Antigens
      ing in genetics. Ann Math Stat 20(1):91–94             12:315–322
 9.   Emigh TH (1980) A comparison of tests for          24. Morton NE, Simpson SP, Lew R, Yee S (1983)
      Hardy-Weinberg equilibrium. Biometrics                 Estimation of haplotype frequencies. Tissue
      36(4):627–642                                          Antigens 22(4):257–262
10.   Guo SW, Thompson EA (1992) Performing              25. Hawley ME, Kidd KK (1995) HAPLO: a pro-
      the exact test of Hardy-Weinberg proportion            gram using the EM algorithm to estimate the
      for multiple alleles. Biometrics 48(2):                frequencies       of     multisite    haplotypes.
      361–372                                                J Hered 86:409–411
11.   Huber M, Chen Y, Dinwoodie I, Dobra A,             26. Long JC, Williams RC, Urbanek M (1995) An
      Nicholas M (2006) Monte Carlo algorithms               E-M algorithm and testing strategy for
      for Hardy-Weinberg proportions. Biometrics             multiple-locus haplotypes. Am J Hum Genet
      62:49–53                                               56:799–810
12.   Yuan A, Bonney GE (2003) Exact test of             27. Fallin D, Schork NJ (2000) Accuracy of haplo-
      Hardy-Weinberg equilibrium by Markov chain             type frequency estimation for biallelic loci, via
      Monte Carlo. Math Med Biol 20:327–340                  the expectation-maximization algorithm for
13.   Ebrahimi N, Bilgili D (2007) A new method of           unphased diploid genotype data. Am J Hum
      testing for Hardy-Weinberg equilibrium and             Genet 67:947–959
      ordering populations. J Genet 86:1–7               28. Tishkoff SA, Pakstis AJ, Ruano G, Kidd KK
14.   Metropolis N, Rosenbluth AW, Rosenbluth                (2000) The accuracy of statistical methods for
      MN, Teller AH, Teller E (1953) Equations of            estimation of haplotype frequencies: an example
      state calculations by fast computing machines.         from the CD4 locus. Am J Hum Genet
      J Chem Phys 21:1087–1092                               67:518–522
15.   Chen JJ, Hollenbach JA, Trachtenberg EA,           29. Kirk KM, Cardon LR (2002) The impact of
      Just JJ, Carrington M, Rønningen KS, Begovich          genotyping error on haplotype reconstruction
      A, King MC, McWeeney SK, Mack SJ, Erlich               and frequency estimation. Eur J Hum Genet
      HA, Thomson G (1999) Hardy-Weinberg test-              10:616–622
      ing for HLA class II (DRB1, DQA1, DQB1             30. Single RM, Meyer D, Hollenbeck J, Nelson M,
      and DPB1) loci in 26 human ethnic groups.              Noble JA, Erlich HA, Thomson G (2002)
      Tissue Antigens 54:533–542                             Haplotype frequency estimation in patient
16.   Hernández JL, Weir BS (1989) A disequilib-             populations: the effect of departures from
      rium coefficient approach to Hardy-Weinberg            Hardy-Weinberg proportions and collapsing
      testing. Biometrics 45(1):53–70                        over a locus in the HLA region. Gen Epidemiol
                                                             22:186–195
17.   Chen JJ, Thomson G (1999) The variance for
      the disequilibrium coefficient in the individual   31. Stephens M, Smith NJ, Donnelly P (2001) A
      Hardy-Weinberg            test.      Biometrics        new statistical method for haplotype recon-
      55:1269–1272                                           struction from population data. Am J Hum
                                                             Genet 68:978–989
18.   Barnetche T, Gourraud PA, Cambon-Thomsen
      A (2005) Strategies in analysis of the genetic     32. Niu T, Qin ZS, Xu X, Liu JS (2002) Bayesian
      component of multifactorial diseases; biostatis-       haplotype inference for multiple linked single-
      tical aspects. Transpl Immunol 14(3–4):                nucleotide polymorphisms. Am J Hum Genet
      255–266                                                70:157–169
19.   Guan Y, Stephens M (2008) Practical issues in      33. Qin ZS, Niu T, Liu JS (2002) Partition-
      imputation-based association mapping. PLoS             ligation-expectationmaximization algorithm
      Genet 4(12):e1000279                                   for haplotype inference with singlenucleotide
                                  13   Analytical Methods for Immunogenetic Population Data               243
66. Weir BS, Cockerham CC (1984) Estimating              83. Middleton D, Gonzalez F, Fernandez-Vina M,
    F-statistics for the analysis of population struc-       Tiercy JM, Marsh SG, Aubrey M, Bicalho MG,
    ture. Evolution 38:1358–1370                             Canossi A, Carter V, Cate S, Guerini FR,
67. Hedrick PW (2005) A standardized genetic                 Loiseau P, Martinetti M, Moraes ME, Morales
    differentiation        measure.         Evolution        V, Perasaari J, Setterholm M, Sprague M,
    59:1633–1638                                             Tavoularis S, Torres M, Vidal S, Witt C,
68. Pearson K (1901) “On lines and planes of                 Wohlwend G, Yang KL (2009) A bioinformat-
    closest fit to systems of points in space” (PDF).        ics approach to ascertaining the rarity of HLA
    Phil Mag 2(6):559–572                                    alleles. Tissue Antigens 74:480–485
69. Cox TF, Cox MAA (2001) Multidimensional              84. Cadavid LF, Watkins DI (1997) Heirs of the
    Scaling, 2nd edn. Chapman and Hall, Boca                 jaguar and the anaconda: HLA, conquest and
    Raton, FL                                                disease in the indigenous populations of the
                                                             Americas. Tissue Antigens 6:702–711
70. Borg I, Groenen P (2005) Modern multidi-
    mensional scaling: theory and applications, 2nd      85. Erlich HA, Mack SJ, Bergström T, Gyllensten
    edn. Springer, New York                                  UB (1997) HLA class II alleles in Amerindian
                                                             populations: implications for the evolution of
71. Pritchard JK, Stephens M, Donnelly P (2000)              HLA polymorphism and the colonization of
    Inference of population structure using multilo-         the Americas. Hereditas 127(1–2):19–24
    cus genotype data. Genetics 155(2):945–959
                                                         86. Sokal R, Michener C (1958) A statistical
72. Hubisz MJ, Falush D, Stephens M, Pritchard               method for evaluating systematic relationships.
    JK (2009) Inferring weak population structure            Univ Kansas Sci Bull 38:1409–1438
    with the assistance of sample group informa-
    tion. Mol Ecol Resour 9(5):1322–1332                 87. Murtagh F (1984) Complexities of hierarchic
                                                             clustering algorithms: the state of the art.
73. Rosenberg NA, Pritchard JK, Weber JL, Cann               Comput Stat Quart 1:101–113
    HM, Kidd KK, Zhivotovsky LA, Feldman MW
    (2002) Genetic structure of human populations.       88. Bergström TF, Josefsson A, Erlich HA,
    Science 298(5602):2381–2385                              Gyllensten UB (1997) Analysis of intron
                                                             sequences at the class II HLA-DRB1 locus:
74. Karypis G. (2002) CLUTO: a clustering toolkit.           implications for the age of allelic diversity.
    Technical Report 02-017. University of                   Hereditas 127(1–2):1–5
    Minnesota, Minneapolis, MN
                                                         89. Bergström TF, Josefsson A, Erlich HA,
75. Felsenstein J (1989) PHYLIP—Phylogeny                    Gyllensten U (1998) Recent origin of HLA-
    Inference Package (Version 3.2). Cladistics              DRB1 alleles and implications for human evo-
    5:164–166                                                lution. Nat Genet 18(3):237–242
76. Felsenstein, J. (2005) PHYLIP (Phylogeny             90. Felsenstein J (1985) Confidence limits on phy-
    Inference Package) version 3.6 (Distributed by           logenies: an approach using the bootstrap.
    the author). Department of Genome Sciences,              Evolution 39:783–791
    University of Washington, Seattle
                                                         91. Penny D, Hendry MD (1985) Testing meth-
77. Saitou N, Nei M (1987) The neighbor-joining              ods of evolutionary tree construction. Cladistics
    method: a new method for reconstructing phy-             1:266–278
    logenetic trees. Mol Biol Evol 4(4):406–425
                                                         92. Cavalli-Sforza LL, Menozzi P, Piazza A (1994)
78. Nei M (1972) Genetic distance between popu-              The history and geography of human genes.
    lations. Am Nat 106:283–292                              Princeton University Press, Pinceton, NJ
79. Cavalli-Sforza LL, Edwards AFW (1967)                93. Cavalli-Sforza LL, Feldman MW (2003) The
    Phylogenetic analysis: models and estimation             application of molecular genetic approaches to
    procedures. Am J Hum Genet 19:233–257                    the study of human evolution. Nat Genet
80. Hughes AL, Nei M (1988) Pattern of nucle-                33:266–275
    otide substitution at MHC class I loci reveals       94. Mack SJ, Erlich HA (2007) Population relation-
    overdominant selection. Nature 335:167–170               ships as inferred from classical HLA genes. 13th
81. Hughes AL, Yeager M (1998) Natural selection             International histocompatibility workshop
    at major histocompatibility complex loci of ver-         anthropology/human genetic diversity joint
    tebrates. Ann Rev Genet 32:415–435                       report. In: Hansen JA (ed) Immunobiology of
82. Sanchez-Mazas A, Fernandez-Viña M,                       the human MHC: Proceedings of the 13th
    Middleton D, Hollenbach JA, Buhler S, Di D,              international histocompatibility workshop and
    Rajalingam R, Dugoujon JM, Mack SJ,                      conference, vol I. IHWG, Seattle, pp. 747–757
    Thorsby E (2011) Immunogenetics as a tool in         95. Nei M (1978) Estimation of average heterozy-
    anthropological        studies.      Immunology          gosity and genetic distance from a small num-
    133(2):143–164                                           ber of individuals. Genetics 89:583–590
                                                                                        Chapter 14
Abstract
Disease association studies involving highly polymorphic immunogenetic data may involve analyses at one
or many units of analysis, including amino acid, allele, genotype and haplotype levels, as well as consideration
of gene–gene or gene–environment interactions. The selection of the appropriate statistical tests is critical
and will be dependent on the nature of the dataset (e.g., case-control vs. family data) as well as the specific
research hypotheses being tested. This paper describes the various study and analysis categories used for
such analyses, including the advantages and limitations of such techniques.
Key words: HLA, KIR, Immunogenetic, Data analysis, Disease association, Case-control, Family
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_14, © Springer Science+Business Media New York 2012
                                                         245
246   J.A. Hollenbach et al.
2. Study
and Analysis
Categories
                            Case-control studies are a critical, population-based tool for epide-
2.1. Case-Control           miological studies, including genetic association studies. These
Studies                     studies generally involve samples of unrelated individuals with the
                            (disease) phenotype of interest and a corresponding control sample
                            of unrelated, unaffected (or randomly ascertained) individuals
                            drawn from the same (ethnic and preferably geographic) popula-
                            tion. While a discussion of study design, including power calcula-
                            tions and sample size considerations is out of the scope of this
                            chapter, a general recommendation in immunogenetic studies is to
                            have a minimum of 200 chromosomes (100 individuals) in each of
                            the case and control cohorts, although much larger sample sizes
                            are preferable. Among the advantages of case-control studies is
                            elimination of the necessity to collect data from family members,
                            which is often logistically difficult and costly. However, case-con-
                            trol studies can be very sensitive to population stratification within
                            the sample cohorts, a particularly important issue in immunoge-
                            netic data, where allele frequency distributions can vary consider-
                            ably between human ethnic groups. If the samples are not collected
                            with scrupulous attention to homogeneity of ancestry background,
                            investigators run the risk of misinterpreting genetic difference
                            between cases and controls. In these cases, heterogeneity between
                            cases and controls due to allele frequency differences related to
                            population stratification may be mistaken for association with a
                            particular locus.
                                 Population analyses, including calculations of gene (allele)
                            frequencies, tests for fit to expectations under Hardy–Weinberg
                            Equilibrium (HWE), and estimations of haplotype frequencies,
                            are the first step in any study involving immunogenetic data,
                            and will provide additional means to assess any underlying pop-
                            ulation substructure, as well as additional units of analysis on
                            which association analyses will be performed. These calculations
                            are detailed in Chap. 13. Many computer software programs are
                            available to perform such calculations on genetic data, but as
                            mentioned in Chap. 13, care must be taken to utilize tools
                            either specifically designed for immunogenetic data (e.g., the
                            PYPOP analytical package (http://www.pypop.org)) or those
                            that are able to handle the high levels of polymorphism characteristic
                            of these loci.
2.1.1. Contingency Tables    1. Basic construction and analysis of contingency tables, with par-
                                ticular attention to sparse cells.
                                     A contingency table, or cross-tabulation, is used to test the
                                difference, or independence, of frequency distributions for cat-
                                egorical variables. Tests for heterogeneity between specific
248   J.A. Hollenbach et al.
                   Table 1
                   A 2 ¥ k contingency table for case-control data
                   0101            75                     29                      104
                   0102                5                    5                      10
                   0301            59                     56                      115
                   0401                6                  15                       21
                   0701                9                  25                       34
                   0701                6                  11                       17
                   0801            73                     39                      112
                   1101            39                     35                       74
                   1103                9                    4                      13
                   1104            40                     24                       64
                   Binned          33                     25                       57
                   Total          354                     268                     622
                   Table 2
                   A 2 ¥ 2 contingency table for allele 0101
                   0101                     75                   29               104
                   Others (not 0101)       279                  239               517
                   Total                   354                  268               622
                                                     (Oi - Ei )2
                                            c2 = å               ,
                                                         Ei
                      where
                      Oi = the observed count of allele i
250   J.A. Hollenbach et al.
    Table 3
    JIA-OP HLA-DRB1 allele data ranked by odds ratio (OR)
RPEa Ab Bc DRB1 Patient Control Chi-square (c2) p-Valued Effect OR CIe CIe
2.1.2. Odds Ratios          Odds ratios (OR) are used in disease association studies as means
and Relative Risk           to describe the strength of association between two variables, and
                            reflect the ratio of odds of an outcome (e.g., disease) occurring in
                            one group (e.g., individuals with allele a) to the odds of it occur-
                            ring in another group (individuals without allele a). Relative risk
                            (RR) is a related but different measure that describes the risk of
                            having the outcome of interest relative to exposure (e.g., positive
                            or negative for allele a). In case-control studies, the OR is the pre-
                            ferred measure, due in part to its use in logistic regression (see next
                            section), allowing a more ready comparison of results derived from
                            different methods. Another important advantage to the OR is that
                            it is by its mathematical nature invertible, i.e., the OR for predis-
                            position to disease is the direct inverse of the OR for protection;
                            this is extremely important in terms of immunogenetic studies,
                            where alleles may be associated in either a PRO or PRE role. It is
                            not unusual to find both PRO and PRE alleles in a given disease
                            study for a given locus; the ability to invert ORs allows more direct
14   Analytical Methods for Disease Association Studies with Immunogenetic Data     255
                   Table 4
                   Generic 2 × 2 table
                   Y                        a                           b
                   Not Y                    c                           d
    Table 5
    Example of input variables for logistic regression analysis
                               Covariable
                     Dependent
    Independent      variable  Risk factor:           Risk factor:     Risk factor:        Genetic:
    variable (HLA    (disease  smoking (yes/          smoking (years)- smoking             snp-
    or KIR)a,b       status)c  no)-Binaryd            Quantitativee    (duration)-Ordinalf Categoricalg
    0101,0401        0            1                   15.8                  1                      1
    0401,0501        0            1                   10                    2                      2
    0301,0401        1            1                    2.5                  3                      3
    0101,0501        1            0                    1                    2                      4
a
  Data can be analyzed at the allele, genotype, haplotype, or other user-defined level
b
  User will need to specify the genetic model, e.g., recessive, codominant, etc.
c
  By convention, 0 = control; 1 = case
d
  Here, risk factor (smoking) is analyzed as present or absent. 0 = nonsmoker, 1 = smoker
e
  Here, risk factor (smoking) is analyzed as a continuous quantitative variable, number of years smoked
f
 Here, risk factor (smoking) is analyzed as an ordered discrete variable, number of years smoked categorized 1 = 0–5
years; 2 = 6–10 years; 3 = 11–20 years; 4 = more than 20 years
g
  Here, SNP data categorized by genotype, e.g., 1 = AG; 2 = AT; 3 = GG; 4 = GT
2.1.3. Logistic Regression     Analysis using logistic regression can also be used in case-control
                               studies. Logistic regression is a form of generalized linear modeling
                               for data with a dichotomous, or binary, outcome variable, such as
                               case-control data where the outcome is either “affected” or “unaf-
                               fected.” Among its advantages, logistic regression provides a means
                               to develop association models that include the contribution of
                               both quantitative and qualitative covariables, which can involve
                               cumbersome stratification procedures in contingency table testing;
                               logistic regression is a critical tool in the analysis of complex multi-
                               variate datasets. Example of potential types of input variables for a
                               logistic regression analysis with immunogenetic data are given in
                               Table 5. Attention must be paid to how both quantitative and
                               qualitative variables are coded, because interpretation of the ORs
                               can greatly vary. For example in Table 5, including number of years
                               smoking as a covariate will provide a single OR associated with
                               each increment of 1 year of smoking and assumes that the risk
                               increment is fully proportional to years smoking. This model may
                               not make sense, so in some cases it is more appropriate to recode
                               quantitative variables as binary (e.g., smoking yes/no in Table 5)
                               or discrete, ordinal (e.g., smoking duration, Table 5), rather than
                               continuous, variables.
                                   A disadvantage of logistic regression analysis is a tendency to
                               overestimate Ors (46) in sample sizes <500, possibly leading to
         14    Analytical Methods for Disease Association Studies with Immunogenetic Data      257
2.2. Family-Based           Family studies have long been the bread and butter of genetic epi-
Studies                     demiology. Family studies provide a means to assess genetic linkage
                            and/or association by testing within a nuclear family.
2.2.1. Pedigree Types       The simplest type of family study involves (1) “trios,” sets of
in Family Studies           “simplex” families consisting of a proband (an individual with the
                            disease phenotype of interest) and both of the proband’s parents
                            (Fig. 1a). Additional family collections may include (2) multiplex
                            sib pairs (MSP) pedigrees with two or more affected siblings
                            (Fig. 1c), (3) multiplex parent child (MPC), with one affected par-
                            ent and child (Fig. 1b) and (4) multiplex parent sibs (MPS) with at
258         J.A. Hollenbach et al.
                             Fig. 1. The four family-based ascertainment schemes are selected in the presence of at
                             least: (a) one affected child (S), (b) one affected child and one affected parent (multiplex
                             parent child (MPC)), (c) two affected sibs (multiplex sib pairs (MSP)), and (d) two affected
                             sibs and one affected parent (multiplex parent sibs (MPS)). As noted in the text, additional
                             family members (parent or sib) may be affected within each of these schemes; they occur
                             with frequencies expected under the disease model and are retained in analyses. For the
                             MSP and MPS ascertainment schemes, using the ordered notation of Thomson (55) the
                             first affected sib ascertained is always denoted by genotype AC, with the four possible
                             genotypes for the second affected sib listed.
                             least one affected parent and two or more affected siblings (Fig. 1d).
                             The type of analyses available will depend on the nature of the
                             pedigrees collected.
2.2.2. Affected Sib Pair     When considering related individuals, the loci linked to disease
Method                       susceptibility are expected to be found in the region of the genome
                             inherited from the same ancestors. Affected sib pair methods can
                             be used in the study of HLA-associated diseases. Deviations from
                             the Mendelian random expectations of 25, 50, and 25% that two
                             affected sibs will on average share 2, 1, and 0 parental chromo-
         14    Analytical Methods for Disease Association Studies with Immunogenetic Data     259
2.2.3. Transmission         Originally developed as a test for genetic linkage, the Transmission
Disequilibrium Test and     Disequilibrium Test (TDT) is applicable as an association test as
Affected Family-Based       well, since it always tests a compound hypothesis of linkage and
Controls                    association: only linked markers that are associated with disease can
                            give a significant result. The fundamental basis for the TDT involves
                            the likelihood that if a marker is linked to disease, there will be a
                            bias in transmission from parents to the affected individual. In the
                            TDT test, only parents who are heterozygous for the marker of
                            interest are considered. The TDT test has the advantage of being
                            robust with respect to population structure, thereby sidestepping
                            the possible pitfall of population stratification often encountered in
                            case-control studies. The TDT test looks at deviations from the
                            expected 50:50 ratio of transmitted and nontransmitted alleles
                            from parents heterozygous for the marker alleles.
                                 The use of nuclear family data (two parents and their children)
                            to estimate control marker allele frequencies (and similarly haplo-
                            type frequencies) was introduced, with application to HLA data,
                            by Rubinstein et al. (48), and Falk and Rubinstein (49). In families
                            ascertained for the presence of at least one affected child, termed S
                            (simplex) pedigrees (also referred to as trio families), the two
                            parental marker alleles not transmitted to the affected child (the
                            proband) are used as population (control) alleles (see Fig. 1a).
                            These are referred to as affected family-based controls (AFBACs).
                            This matched design for patient (parental transmitted) and “con-
                            trol” (parental nontransmitted) marker alleles avoids ethnic con-
                            founding in the case of a stratified population (50–52).
                                 Field (53) and Thomson (54)—extended this AFBAC approach
                            to nuclear pedigrees ascertained for the presence of at least two
                            affected sibs, termed MSP pedigrees (although referred to then as
                            MS pedigrees, for multiplex sibs); using the alleles from both par-
                            ents that are never transmitted to either sib in the affected sib pair
                            as the “control” population (Fig. 1c). Additionally, MPC and MPS
                            ascertainment schemes were considered: these extend the S and
                            MSP ascertainment schemes to also include at least one affected
                            parent. The non- or never-transmitted alleles, respectively, from
                            the unaffected parent form the respective AFBAC populations
                            (Fig. 1b, d). Similar extensions apply to other ascertainment
                            schemes, e.g., with ascertainment based on the presence of at least
                            three affected sibs in a nuclear family, the AFBACs are the parental
                            alleles never transmitted to any of the three affected sibs.
                                 AFBAC is similar to the TDT in that it examines the transmis-
                            sion of marker alleles in family trios. The AFBAC method has often
                            mistakenly been equated to a heterogeneity test of association of
260   J.A. Hollenbach et al.
Table 6
Potential units of analysis for killer cell immunoglobulin-like receptors (KIR) data
in disease association
KIR ligands           HLA-C1 carrier; HLA-C1 homozygous; HLA-C2 carrier; HLA-C2 homozy-
                       gous; HLA-Bw4 carrier; HLA-Bw4 homozygous; HLA-Bw6 carrier;
                       HLA-Bw6 homozygous; HLA-Bw4_Thr80 carrier; HLA-Bw4_Ile80 carrier
KIR genes             KIR2DL1; KIR2DP1; KIR2DL2; KIR2DL3; KIR2DL5; KIR3DL1; KIR2DS1;
                        KIR2DS2; KIR2DS3; KIR2DS4; KIR2DS5; KIR3DS1
KIR diplotypes        AA; AA (with no activating); AB; BB; centromeric AA; telomeric AA; centro-
                       meric AB; telomeric AB; centromeric BB; telomeric BB
KIR haplotypes        A; B; B variants
KIR and ligands       KIR2DL1 with HLA-C2; KIR2DL1 without HLA-C2; KIR2DL2 with
                        HLA-C1; KIR2DL2 without HLA-C1; KIR2DL3 with HLA-C1; KIR2DL3
                        without HLA-C1; KIR3DL1 with HLA-Bw4; KIR3DL1 with HLA-Bw4_
                        Ile80; KIR3DL1 with HLA-Bw4 homozygous; KIR2DL3 with HLA-C1;
                        KIR3DS1 with HLA-Bw4; KIR3DS1 with HLA-Bw4_Ile80; KIR3DS1 with
                        HLA-Bw4 homozygous
3. KIR Data:
Special
Considerations
                           While KIR data are in many ways typical of immunogenetic data,
                           with high levels of polymorphism and linkage disequilibrium
                           through the region, as well as a clear history of extensive recombi-
                           nation and gene conversion events, the limitations of most typing
                           techniques mean that in many cases investigators will have to deal
                           with large amounts of missing data. Likewise, most typing tech-
                           niques cannot detect or distinguish between loci that may be dupli-
                           cated on a KIR haplotype. In addition, the relationship between
                           the KIR and their HLA ligands dictates consideration of interac-
                           tion effects between these two important polymorphic regions
                           when analyzing for disease associations. Table 6 illustrates poten-
                           tial units of analysis in disease association studies with KIR data.
                           This list is by no means exhaustive, and the ultimate choice of
                           analyses performed should be determined by the specific research
                           hypotheses being tested in the study.
                                Issues specific to the analysis of KIR data for disease association
                           studies include:
                             1. Treatment of presence/absence or gene-content typing data. As
                                allele level KIR genotyping improves, datasets will contain a
                                mixture of gene-content and allele level typing data. Analysis
                                of KIR data at the gene-content level may at times be more
                                robust than at the allele level, or vice-versa. In gene-content
262     J.A. Hollenbach et al.
4. Data Reporting
Acknowledgments
Glossary
References
 1. Vandiedonck C, Knight JC (2009) The human                Rickards O, Klitz W (2001) Genetic variability
    major histocompatibility complex as a para-              and linkage disequilibrium within the HLA-DP
    digm in genomics research. Brief Funct                   region: analysis of 15 different populations.
    Genomic Proteomic 8(5):379–394                           Tissue Antigens 57:424–439
 2. Mather KA, President M, Eastel S, Hollenbach        8.   Bergstrom TF, Engkvist H, Erlandsson R,
    J, Klitz W, Huttley G, Thomson G (2007)                  Josefsson A, Mack SJ, Erlich HA, Gyllensten U
    Long distance linkage disequilibrium between             (1999) Tracing the origin of HLA-DRB1
    HLA microsatellites in two human popula-                 alleles by microsatellite polymorphism. Am J
    tions. In: Hansen JA (ed) Immunobiology of               Hum Genet 64:1709–1718
    the human MHC: proceedings of the 13th              9.   Bergstrom TF, Mack SJ, Gyllensten U, Ehrlich
    international histocompatibility workshop and            HA (2000) Evolution of HLA-DRB loci,
    congress. IHWG Press, Seattle                            DRB1 lineages, and alleles: analyses of intron-1
 3. Ann B, Begovich WK, Steiner LL, Grams S, Suraj-          and -2 sequences. In: Kasahara M (ed) Major
    Baker V, Hollenbach J, Trachtenberg E, Louie L,          histocompatibility complex: evolution, struc-
    Zimmerman PA, Hill AVS, Stoneking M, Sasazuki            ture and function. Springer, Tokyo
    T, Rickards O, Titanji VPK, Konenkov VI,           10.   Crawford MH, Reddy BM, Martinez-Laso J,
    Sartakova ML (1999) HLA DQ hapotypes in 15               Mack SJ, Erlich HA (2001) Genetic variation
    different populations. In: Kasahara M (ed) Major         among the Golla pastoral caste subdivisions of
    histocompatibility complex: evolution, structure         Andhra Pradesh, India, according to the HLA
    and function. Springer, Tokyo                            system. Hum Immunol 62:1031–1041
 4. Petzel-Erler ML, Gorodezky C, Layrisse Z,          11.   Ellis JM, Mack SJ, Leke RF, Quakyi I, Johnson
    Klitz W, Fainboim L, Vullo C, Bodmer J, Egea             AH, Hurley CK (2000) Diversity is demonstrated
    E, Navarrete C, Infante E, Alaez C, Olivo A,             in class I HLA-A and HLA-B alleles in
    Debaz H, Bautista N, de la Rosa G, Vazquez               Cameroon, Africa: description of HLA-
    MN, Navarro JL, Pujol MJ, Duran C,                       A*03012, *2612, *3006 and HLA-B*1403,
    Schafhauser C, Faucz FR, Janzen M, Maciag P,             *4016, *4703. Tissue Antigens 56:291–302
    Boldt ABW, Souza PSA, Probst CM, da Silva          12.   Erlich HA, Mack SJ, Bergstrom T, Gyllensten
    GF, Makhatadze N, Dominguez E, Montagnani                UB (1997) HLA class II alleles in Amerindian
    S, Matos M, Martinez A, Herrera F, Hollenbach            populations: implications for the evolution of
    J, Thomson G, Pando M, Satz L, Larriba J,                HLA polymorphism and the colonization
    Fernandez G, Pesoa SA, Borosky A, Garavito               of the Americas. Hereditas 127:19–24
    G, Angel L, Brown J, Llop E (1997)
    Anthropology component report for region           13.   Hildesheim A, Apple RJ, Chen CJ, Wang SS,
    Latin-America: Amerindian and admixed pop-               Cheng YJ, Klitz W, Mack SJ, Chen IH, Hsu
    ulations. In: Charron D (ed) Genetic diversity           MM, Yang CS, Brinton LA, Levine PH, Erlich
    of HLA: functional and medical implications.             HA (2002) Association of HLA class I and II
    EDK Press, Paris                                         alleles and extended haplotypes with nasopha-
                                                             ryngeal carcinoma in Taiwan. J Natl Cancer
 5. Trachtenberg EA, Hayes E, Hollenbach JA,
                                                             Inst 94:1780–1789
    Keyeaux G, Bernal J, Klitz W (1997) HLA
    class II variation and linkage disequilibrium in   14.   Johansson A, Ingman M, Mack SJ, Erlich H,
    nine Amerindian and three African American               Gyllensten U (2008) Genetic origin of the
    tribes from Columbia. Results of expedicion              Swedish Sami inferred from HLA class I and
    humana. In: Charron D (ed) Genetic diversity             class II allele frequencies. Eur J Hum Genet
    of HLA: functional and medical implications.             16:1341–1349
    EDK Press, Paris                                   15.   Leffell MS, Fallin MD, Erlich HA, Fernandez-
 6. Trachtenberg E, Vinson M, Hayes E, Hsu YM,               Vijna M, Hildebrand WH, Mack SJ, Zachary
    Houtchens K, Erlich H, Klitz W, Hsia Y,                  AA (2002) HLA antigens, alleles and haplo-
    Hollenbach J (2007) HLA class I (A, B, C) and            types among the Yup’ik Alaska natives: report
    class II (DRB1, DQA1, DQB1, DPB1) alleles                of the ASHI Minority Workshops, Part II.
    and haplotypes in the Han from southern                  Hum Immunol 63:614–625
    China. Tissue Antigens 70:455–463                  16.   Mack SJ, Bugawan TL, Moonsamy PV, Erlich
 7. Begovich AB, Moonsamy PV, Mack SJ,                       JA, Trachtenberg EA, Paik YK, Begovich AB,
    Barcellos LF, Steiner LL, Grams S, Suraj-Baker           Saha N, Beck HP, Stoneking M, Erlich HA
    V, Hollenbach J, Trachtenberg E, Louie L,                (2000) Evolution of Pacific/Asian populations
    Zimmerman P, Hill AV, Stoneking M, Sasazuki              inferred from HLA class II allele frequency
    T, Konenkov VI, Sartakova ML, Titanji VP,                distributions. Tissue Antigens 55:383–400
         14   Analytical Methods for Disease Association Studies with Immunogenetic Data                 265
17. Mack SJ, Erlich HA (1998) HLA class II               27. Williams AP, Bateman AR, Khakoo SI (2005)
    polymorphism in the Ticuna of Brazil: evolu-             Hanging in the balance. KIR and their role in
    tionary implications of the DRB1*0807 allele.            disease. Mol Interv 5:226–240
    Tissue Antigens 51:41–50                             28. Li Y, Zhang T, Ho C, Orange JS, Douglas SD,
18. Mack SJ, Tu B, Lazaro A, Yang R, Lancaster               Ho WZ (2004) Natural killer cells inhibit hep-
    AK, Cao K, Ng J, Hurley CK (2009) HLA-A,                 atitis C virus expression. J Leukoc Biol
    -B, -C, and -DRB1 allele and haplotype fre-              76:1171–1179
    quencies distinguish Eastern European                29. Hollenbach JA, Ladner MB, Saeteurn K, Taylor
    Americans from the general European American             KD, Mei L, Haritunians T, McGovern DPB,
    population. Tissue Antigens 73:17–32                     Erlich HA, Rotter JI, Trachtenberg EA (2009)
19. MacKenzie J, Perry J, Ford AM, Jarrett RF,               Susceptibility to Crohn’s disease is mediated by
    Greaves M (1999) JC and BK virus sequences               KIR2DL2/KIR2DL3 heterozygosity and the
    are not detectable in leukaemic samples from             HLA-C ligand. Immunogenetics 61(10):
    children with common acute lymphoblastic                 663–671
    leukaemia. Br J Cancer 81:898–899                    30. Gaudieri S, Desantis D, McKinnon E, Moore
20. Meyer D, Single RM, Mack SJ, Erlich HA,                  C, Nolan D, Witt CS, Mallal SA, Christiansen
    Thomson G (2006) Signatures of demographic               FT (2005) Killer immunoglobulin-like recep-
    history and natural selection in the human               tors and HLA act both independently and syn-
    major histocompatibility complex loci. Genetics          ergistically to modify HIV disease progression.
    173:2121–2142                                            Genes Immun 6:683–690
21. Solberg OD, Mack SJ, Lancaster AK, Single            31. Khakoo SI, Carrington M (2006) KIR and dis-
    RM, Tsai Y, Sanchez-Mazas A, Thomson G                   ease: a model system or system of models?
    (2008) Balancing selection and heterogeneity             Immunol Rev 214:186–201
    across the classical human leukocyte antigen         32. Kunert K, Seiler M, Mashreghi MF, Klippert
    loci: a meta-analytic review of 497 population           K, Schonemann C, Neumann K, Pratschke J,
    studies. Hum Immunol 69:443–464                          Reinke P, Volk HD, Kotsch K (2007) KIR/
22. Tang TF, Hou L, Chen M, Belle I, Mack S,                 HLA ligand incompatibility in kidney trans-
    Lancaster A, Ho GY, Hwang WY, Alsagoff F,                plantation. Transplantation 84:1527–1533
    Ng J, Hurley CK (2007) HLA haplotypes in             33. Gedil MA, Steiner NK, Hurley CK (2007)
    Singapore: a study of mothers and their cord             KIR3DL2: diversity in a hematopoietic stem
    blood units. Hum Immunol 68:430–438                      cell transplant population. Tissue Antigens 70:
23. Tu B, Mack SJ, Lazaro A, Lancaster A,                    228–232
    Thomson G, Cao K, Chen M, Ling G,                    34. Sun JY, Gaidulis L, Dagis A, Palmer J, Rodriguez
    Hartzman R, Ng J, Hurley CK (2007) HLA-                  R, Miller MM, Forman SJ, Senitzer D (2005)
    A, -B, -C, -DRB1 allele and haplotype frequen-           Killer Ig-like receptor (KIR) compatibility plays
    cies in an African American population. Tissue           a role in the prevalence of acute GVHD in unre-
    Antigens 69:73–85                                        lated hematopoietic cell transplants for AML.
24. Cao K, Hollenbach J, Shi X, Shi W, Chopek M,             Bone Marrow Transplant 36:525–530
    Fernandez-Vina MA (2001) Analysis of the             35. Lanier LL (1999) Natural killer cells fertile
    frequencies of HLA-A, B, and C alleles and               with receptors for HLA-G? Proc Natl Acad Sci
    haplotypes in the five major ethnic groups of            USA 96:5343–5345
    the United States reveals high levels of diversity   36. Moffett A, Hiby SE (2007) How does the
    in these loci and contrasting distribution pat-          maternal immune system contribute to the
    terns in these populations. Hum Immunol                  development of pre-eclampsia? Placenta 28
    62:1009–1030                                             (suppl A):S51–S56
25. Hollenbach JA, Thomson G, Cao K, Fernandez-          37. Hu LP, Tao LX (2009) (Statistical analysis
    Vina M, Erlich HA, Bugawan TL, Winkler C,                using SAS software package for data of RxC
    Winter M, Klitz W (2001) HLA diversity, dif-             contingency table (part two)]. Zhong Xi Yi Jie
    ferentiation, and haplotype evolution in                 He Xue Bao 7:878–882
    Mesoamerican Natives. Hum Immunol 62:
                                                         38. Horstmann GA (1988) [Product of the month:
    378–390
                                                             SPSS statistical package for the sciences].
26. Khakoo SI, Chloe LT, Martin MP, Brooks CR,               Radiologe 28:438–439
    Gao X, Astemborski J, Cheng J, Goedert JJ,
                                                         39. R Development Core Team (2008) R: a lan-
    Vlahov D, Hilgartner M, Cox S, Little AM,
                                                             guage and environment for statistical comput-
    Alexander GJ, Cramp ME, O’Brien S,
                                                             ing. R Foundation for Statistical Computing,
    Rosenberg WMC, Thomas DL, Carrington M
                                                             Vienna
    (2004) HLA and NK cell inhibitory receptor
    genes in resolving hepatitis C virus infection.      40. Thomson G, Valdes AM, Noble JA, Kockum I,
    Science 305:872–874                                      Grote MN, Najman J, Erlich HA et al (2007)
266         J.A. Hollenbach et al.
      Relative predispositional effects of HLA class II   47. Purcell S, Neale B, Todd-Brown K, Thomas L,
      DRB1-DQB1 haplotypes and genotypes on                   Ferreira MA, Bender D, Maller J, Sklar P, de
      type 1 diabetes: a meta-analysis. Tissue Antigens       Bakker PI, Daly MJ, Sham PC (2007) PLINK:
      70:110–127                                              a tool set for whole-genome association and
41.   Hollenbach JA, Thompson SD, Bugawan TL,                 population-based linkage analyses. Am J Hum
      Ryan M, Sudman M, Marion M, Langefeld                   Genet 81:559–575
      CD, Thomson G, Erlich HA, Glass DN (2010)           48. Rubinstein P, Walker M, Carpenter C, Carrier
      Juvenile idiopathic arthritis and HLA class I           C, Krassner J, Falk C, Ginsberg F (1981)
      and class II interactions and age-at-onset              Genetics of HLA disease associations: the use
      effects. Arthritis Rheum 62:1781–1791                   of the haplotype relative risk (HRR) and the
42.   Thomson G, Marthandan N, Hollenbach JA,                 “haplo-delta” (Dh) estimates in juvenile diabetes
      Mack SJ, Erlich HA, Single RM, Waller MJ,               from three racial groups. Hum Immunol 3:
      Marsh SG, Guidry PA, Karp DR, Scheuermann               384
      RH, Thompson SD, Glass DN, Helmberg W               49. Falk CT, Rubinstein P (1987) Haplotype rela-
      (2010) Sequence feature variant type (SFVT)             tive risk: an easy reliable way to construct a
      analysis of the HLA genetic association in              proper control sample for risk calculations. Ann
      juvenile idiopathic arthritis. Pac Symp                 Hum Genet 51:227–233
      Biocomput:359–370                                   50. Khoury MJ, Beaty TH (1994) Applications of
43.   Thomson G, Valdes AM (2007) Conditional                 the case-control method in genetic epidemiol-
      genotype analysis: detecting secondary disease          ogy. Epidemiol Rev 16:134–150
      loci in linkage disequilibrium with a primary       51. Schaid DJ, Sommer SS (1994) Comparison of
      disease locus. BMC Proc 1(suppl 1):S163                 statistics for candidate-gene association studies
44.   Thomson G, Marthandan N, Hollenbach JA,                 using cases and parents. Am J Hum Genet
      Mack SJ, Erlich HA, Single RM, Waller MJ,               55:402–409
      Marsh SGE, Guidry PA, Karp DR, Scheuermann          52. Thomson G (1995) Mapping disease genes:
      RH, Thompson SD, Glass DB, Helmberg W                   family-based association studies. Am J Hum
      (2009) Sequence feature variant type (SFVT)             Genet 57:487–498
      analysis of the HLA genetic association in juve-
      nile idiopathic arthritis. Proceedings of the       53. Field LL (1991) Non-HLA region genes in
      2010 pacific society of biocomputing confer-            insulin dependent diabetes mellitus. Baillieres
      ence. Pac Symp Biocomput:359–370                        Clin Endocrinol Metab 5:413–438
45.   Clopper CJ, Pearson ES (1934) The use of            54. Thomson G (1993) AGFAP method: applica-
      confidence or fiducial limits illustrated in            bility under different ascertainment schemes
      the case of the binomial. Biometrika 26:                and a parental contributions test. Genet
      404–413                                                 Epidemiol 10:289–310
46.   Nemes S, Jonasson JM, Genell A, Steineck G          55. Thomson G (1995) Analysis of complex human
      (2009) Bias in odds ratios by logistic regression       genetic traits: an ordered-notation method and
      modelling and sample size. BMC Med Res                  new tests for mode of inheritance. Am J Hum
      Methodol 9:56                                           Genet 57:474–486
                                                                                        Chapter 15
Abstract
The Collaborative Transplant Study (CTS) was initiated in 1982 in Heidelberg, Germany, and originated
from the need to gain further insight into the complex problems and risks involved in human organ trans-
plantation. Currently, more than 400 transplant centers in 45 countries are contributing to this voluntary
international effort, and from the beginning of the study, the impact on graft outcome of immunological
factors, such as matching for HLA antigens and allosensitization to HLA and non-HLA antigens, have
been areas of interest. Herein, we summarize the recent findings from the CTS on these two topics.
     Key words: HLA matching, HLA antibodies, Crossmatch, Kidney transplantation, Liver transplantation,
     Heart transplantation
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_15, © Springer Science+Business Media New York 2012
                                                         267
268      C. Süsal and G. Opelz
                         compatibility (3, 4). Recent CTS work has focused on factors other
                         than graft survival and showed that better HLA matching in renal
                         transplantation is associated with the administration of lower dos-
                         ages of immunosuppressive agents (5) and a lower incidence of
                         side effects of immunosuppression, such as non-Hodgkin lym-
                         phoma and hip fractures (5, 6). Others have shown that better
                         HLA matching is associated with lower grade sensitization if a
                         patient lost a kidney graft and is relisted for a retransplant (7). A
                         recent CTS analysis indicated that HLA matching might also
                         improve the currently poor long-term outcome in lung transplan-
                         tation (8).
2. HLA Matching
in Kidney
Transplantation
                         In a comparative analysis of more than 130,000 deceased-donor
2.1. HLA Matching,       kidney transplantations performed during two successive decades
Cold Ischemia Time       (1985–1994 and 1995–2004), we were able to demonstrate that—in
and Graft Outcome in     spite of improvements in immunosuppression and a significant
Kidney Transplantation   decline in the length of ischemic preservation—the effect of HLA
                         matching on the survival rate of kidney transplants from deceased
                         donors continues to be influential (4). While numerically the
                         percent improvement in graft survival through HLA matching was
                         smaller in recent years, the relative contribution of HLA mis-
                         matches to the fraction of graft failures remained nearly unchanged
                         (Fig. 1). The main difference between the results obtained in the
                         comparative analysis of two decades was an improvement in the
                         overall graft survival rate. However, when 3 HLA-A + B + DR mis-
                         matches were taken as reference, transplants with no HLA mis-
                         matches showed a relative risk (RR) of failure of 0.76 (95%
                         confidence interval [CI] 0.71–0.80) among transplants performed
                         1985–1994 and 0.83 (CI 0.77–0.89) in the decade 1995–2004,
                         and likewise the increased risk of failure associated with six mis-
                         matches changed only little, from 1.23 (CI 1.14–1.32) in 1985–
                         1994 to 1.16 (CI 1.05–1.27) in 1995–2004. Analysis of transplants
                         performed from 2000 to 2004, during which most recipients pre-
                         sumably received “modern” immunosuppressive treatment, con-
                         tinued to show a strong and statistically significant HLA matching
                         effect.
                              In a separate analysis, cold ischemia up to 18 h was found not to
                         be detrimental for graft outcome, and short cold ischemia did not
                         eliminate the effect of HLA matching (3). These findings altogether
                         indicated that an abandonment of HLA matching in exchange for
                         transplanting with short ischemia times would be detrimental to
                         clinical kidney transplantation. For practical/logistical reasons, we
                         recommend emphasis on the avoidance of poorly matched grafts
                         (4, 5 or 6 HLA-A + B + DR mismatches) which is feasible even in
                         programs with relatively short transplant waiting lists.
                15    Impact of HLA Matching and HLA Antibodies in Organ Transplantation…                               269
Fig. 1. Graft survival rates in relation to the number of HLA-A, -B, -DR mismatches for transplants performed between 1985
and 1994 (left) and between 1995 and 2004 (right). Transplants from deceased donors were analyzed. Numbers of mis-
matched HLA antigens (MM) and numbers of patients studied are indicated together with graft survival rates in percent
observed at 5 years posttransplant. The association of HLA matching with graft survival was statistically significant in both
decades (weighted regression P < 0.001). Reproduced from ref. (4) with permission from Wolters Kluwer Health.
2.2. Impact of HLA               Posttransplant non-Hodgkin lymphoma is a rare but serious compli-
Mismatching                      cation after organ transplantation which is associated with high mor-
on Incidence                     tality. In a recent analysis of more than 150,000 deceased-donor
of Posttransplant                kidney transplant recipients reported to the CTS study, we found
Non-Hodgkin                      that one and two HLA-DR mismatches were a significant risk factor
Lymphoma After                   for posttransplant non-Hodgkin lymphoma, particularly for the loca-
Kidney Transplantation           tion kidney and central nervous system, whereas two HLA-B mis-
                                 matches increased the risk for the development of lymphoma of the
                                 kidney (5). An independent analysis of some 9,000 pediatric kidney
                                 transplant recipients showed that two HLA-DR mismatches were
                                 associated with a twofold higher risk of developing non-Hodgkin
                                 lymphoma, which led us to conclude that two HLA-DR mismatches
                                 should be avoided altogether in pediatric renal transplantation (9).
2.3. Association                 Bone fractures are a frequent complication after kidney transplanta-
of Mismatches                    tion for which various predisposing factors, such as older age, female
for HLA-DR with                  gender, diabetes (10–12), extended duration of ESRD disease, (11,
Incidence of                     13) and prolonged pretransplant dialysis (10, 11), have been
Posttransplant Hip               identified. Analysis of some 20,000 kidney transplant recipients
Fracture in Kidney               reported to CTS showed that there was a significant association
Transplant Recipients            between the number of HLA-DR mismatches and the occurrence
                                 of osteoporosis within 5 years after transplantation (6). Importantly,
                                 the risk of hip fracture increased for grafts with one HLA-DR mis-
                                 match to a hazard ratio (HR) of 1.85 (95% CI 1.18–2.89, P = 0.007)
270       C. Süsal and G. Opelz
3. Impact of HLA
Compatibility on
Lung Transplant
Survival                    The impact of HLA compatibility on graft survival was examined
                            in some 8,000 deceased-donor lung transplants performed during
                            1989–2009 and reported to CTS (8). While graft survival rates
                            showed a stepwise decrease as the combined number of
                            HLA-A + B + DR mismatches increased from one to six, it was strik-
                            ing that lung transplants with “perfect” matches, that is zero HLA
                            mismatches, showed an extremely poor outcome with half of all
                            grafts failing during the first year. It thus appears that compatibility
                            at all three HLA loci represents a particular constellation that can
                            override the beneficial effect of HLA matching in terms of amelio-
                            rating immunologic rejection, probably by increasing the risk of
                            severe lung transplant damage due to HLA-restricted immune
                            phenomena. Donor lung preservation for up to 12 h was not asso-
                            ciated with inferior graft survival vs. shorter preservation times,
                            thereby providing a window for efforts at HLA matching. These
                            results indicate that it may be appropriate to consider HLA match-
                            ing of donor lungs within restricted geographical areas to achieve
                            a moderate level of matching in order to help to improve the current
                            poor long-term outcomes after lung transplantation.
4. HLA Matching
and Presensitization
in Organ
Transplantation             Meier-Kriesche et al., who examined some 16,000 renal transplant
                            candidates in the USA that were relisted after loss of a primary
4.1. Poor HLA Match in      kidney transplant, found that with increasing numbers of HLA
Previous Transplant as a    mismatches of the first transplant there was a significant increase in
                            panel reactive antibody (PRA) at relisting (7). Only 10% of the
Cause of Presensitization
             15   Impact of HLA Matching and HLA Antibodies in Organ Transplantation…          271
4.3. Presensitization    Although graft survival rates improved greatly during the last 25
as a Major Problem in    years, sensitization of the recipient at the B-cell level still represents
Organ Transplantation    a major problem in kidney transplantation. Dependent on the anti-
                         body detection method, approximately 20–40% of patients on the
                         kidney transplant waiting list possess serum alloantibodies prior to
                         transplantation, a phenomenon called presensitization. Mainly
                         three events can lead to presensitization: blood transfusion, preg-
                         nancy, and previous transplantation. Although there are great indi-
                         vidual variations, generally the probability that a patient will
                         develop antibodies increases with the frequency of exposure to for-
                         eign antigen. According to older literature, 15–25% of women
                         develop antibodies after the first pregnancy and this rate increases
                         to 50–60% after the second pregnancy. Lymphocytotoxic B-cell
                         reactivity was detectable in 20% of waiting list patients who were
                         treated with 1 blood transfusion, 40% after 10 transfusions, and
                         70% after 20 transfusions (17). Using an ELISA screening assay,
                         we found HLA antibodies of the IgG isotype in 17% of patients
                         waiting for a first transplant, 64% for a second, 84% for a third,
                         and 92% for a fourth graft. Using the more sensitive Luminex
                         technology, El-Awar et al. reported that almost all patients waiting
272      C. Süsal and G. Opelz
                         for a regraft had HLA antibodies of the IgG, IgM, or IgA isotypes
                         in their serum (18). At our own center, retransplant recipients cur-
                         rently make up 14% of the kidney waiting list.
                              An additional critical issue is antibody development against
                         allogeneic antigen systems on graft endothelium other than HLA
                         that are not necessarily detected in routine antibody testing. The
                         influence of high PRA on graft outcome in transplants between
                         HLA-identical siblings (19), the anti-MICA reactivity in eluates
                         from rejected kidney allografts described by Zou et al. (20), the
                         report by Terasaki and Ozawa who found that lymphocytotoxicity
                         correlated better with graft outcome than solid phase flow cytom-
                         etry or ELISA (which exclude non-HLA reactivity) (21), the recent
                         results by Breimer et al. who observed, in the absence of HLA
                         antibodies, significantly more rejections in patients with a positive
                         flow cytometric endothelial crossmatch (22), and the work by
                         Reinsmoen et al. who found a strong association between the pres-
                         ence of high antibody binding to Angiotensin type 1 receptor and
                         antibody-mediated rejection in recipients whose sera did not con-
                         tain antibody to donor HLA or MICA (23), all indicate that anti-
                         bodies directed against non-HLA antigens are important. In our
                         Heidelberg algorithm for transplantation of broadly sensitized
                         high risk patients, we use additional measures, such as peritrans-
                         plant apheresis, to reduce such presumed undetected antibody
                         activity below clinically relevant threshold levels (24).
4.4. Disadvantages       Presensitized kidney recipients have two major disadvantages: (a)
Associated with          they are less likely to receive a transplant due to a high number of
Presensitization         HLA antigens defined as unacceptable because they would cause a
                         positive pretransplant crossmatch; positive crossmatches occur
                         especially if the antibodies are directed against a wide range of
                         HLA specificities (high panel reactive antibodies [PRA]); (b) they
                         reject their grafts more readily even if the pretransplant crossmatch
                         is negative, possibly due to the presence of weak donor-specific
                         antibodies that were overlooked in the crossmatch test. Alternatively,
                         it has been suggested that high PRA reactivity may be an indicator
                         of a generally high responder status against foreign alloantigens. It
                         is an important task of the HLA laboratory to identify a suitable
                         donor against whom the recipient is not sensitized.
                              According to Fuggle and Martin, presensitized patients remain,
                         dependent on the grade of PRA, two or three times longer on the
                         waiting list than nonsensitized patients (25). Actual data from
                         the CTS indicate that, despite recent developments in the manage-
                         ment of presensitized patients, patients who are positive in cyto-
                         toxic PRA testing as well as patients who are negative in cytotoxic
                         PRA testing but positive in ELISA testing for HLA class I antibod-
                         ies continue to show an inferior graft outcome (2, 26).
                              Positive crossmatches in the complement-dependent lym-
                         phocytotoxic test were shown in the 1960s to be associated with
          15   Impact of HLA Matching and HLA Antibodies in Organ Transplantation…        273
5. Analysis of
Positive Kidney,
Heart, and Liver
Transplant            Currently, with improved methods of immunosuppression, desen-
Crossmatches          sitization, and antibody detection, some “weak” positive cross-
Reported to the       match reactions are no longer considered a contraindication at
                      some centers. Virtual crossmatches gained ground (29) and conse-
Collaborative
                      quently the mandatory performance of a pretransplant crossmatch
Transplant Study
                      is not anymore an iron rule. Sometimes, a crossmatch test per-
                      formed after the transplant operation unexpectedly can turn out
                      positive. For these reasons, a number of kidney transplantations
                      were performed and reported to the CTS despite a positive cross-
                      match against donor T lymphocyte targets. On the other hand,
                      positive crossmatches against B-cell targets have generally not been
                      considered a contraindication to transplantation because investiga-
                      tors disagreed on the clinical relevance of this test (30–33). Some
                      20 years ago, flow cytometry crossmatching was introduced and
                      claimed to be superior to the lymphocytotoxicity assay because of
                      higher technical sensitivity (34). Others claimed that the flow
                      crossmatch excludes too many patients from transplantation based
                      on false positive results (35). Therefore, at some centers, comple-
                      ment-dependent lymphocytotoxic crossmatching and flow cytom-
                      etry crossmatching was performed in parallel, and the flow
                      crossmatch was not necessarily considered the determining test.
                      This resulted in positive flow crossmatches reported to CTS. In
                      this manner, over the years a sizable number of positive crossmatch
                      kidney transplants were recorded in the CTS database and became
                      available for analysis. Most heart and liver transplants reported to
                      CTS are done without a pretransplant crossmatch test. At some
                      centers, crossmatches are performed retrospectively for study pur-
                      poses, and the results have been compiled in the CTS database.
                           Our analysis showed that, overall, high rates of hyperacute
                      rejection were not observed with any type of positive crossmatch
                      or type of organ transplant (28), probably because immunosup-
                      pressive treatment has improved. When first kidney transplant
274   C. Süsal and G. Opelz
Fig. 2. Impact of positive crossmatches on survival of deceased-donor kidney transplants in the time period between 1995 and
2008. Lymphocytotoxic crossmatches using T-cell (left) or B-cell targets at warm incubation temperature (middle) were
analyzed (right). Results obtained with flow cytometry. Numbers of patients studied and P values (log rank) are indicated.
(a) First transplants; (b) retransplants.
6. Conclusion
Acknowledgments
References
 1. Mange KC et al (2001) A comparison of the              phoma: a collaborative transplant study report.
    survival of shipped and locally transplanted           Transplantation 90:292–297
    cadaveric renal allografts. N Engl J Med 345:      10. Nikkel LE et al (2009) Risk of fractures after
    1237–1242                                              renal transplantation in the United States.
 2. Collaborative Transplant Study, University of          Transplantation 87:1846–1851
    Heidelberg, Germany, Website. www.ctstrans-        11. O’Shaughnessy EA et al (2002) Risk factors
    plant.org. Accessed 29 Nov 2010                        for fractures in kidney transplantation.
 3. Opelz G, Döhler B (2007) Multicenter analysis of       Transplantation 74:362–366
    kidney preservation. Transplantation 83:247–253    12. Vautour LM et al (2004) Long-term fracture
 4. Opelz G, Döhler B (2007) Effect of human               risk following renal transplantation: a popula-
    leukocyte antigen compatibility on kidney graft        tion-based study. Osteoporos Int 15:160–167
    survival: comparative analysis of two decades.     13. Patel S et al (2001) Prevalence and causes
    Transplantation 84:137–143                             of low bone density and fractures in kidney
 5. Opelz G, Döhler B (2010) Impact of HLA mis-            transplant patients. J Bone Miner Res 16:
    matching on incidence of posttransplant non-           1863–1870
    Hodgkin lymphoma after kidney transplantation.     14. Opelz G et al (1999) HLA compatibility and
    Transplantation 89:567–572                             organ transplant survival. Collaborative Trans-
 6. Opelz G, Döhler B (2010) Association of mis-           plant Study. Rev Immunogenet 1:334–342
    matches for HLA-DR with incidence of post-         15. Süsal C, Pelzl S, Opelz G (2003) Strong human
    transplant hip fracture in kidney transplant           leukocyte antigen matching effect in nonsensi-
    recipients. Transplantation 91:65–69                   tized kidney recipients with high pretransplant
 7. Meier-Kriesche HU et al (2009) A lifetime ver-         soluble CD30. Transplantation 76:1231–1232
    sus a graft life approach redefines the impor-     16. Süsal C, Döhler B, Opelz G (2009) Presensitized
    tance of HLA matching in kidney transplant             kidney graft recipients with HLA class I and II
    patients. Transplantation 88:23–29                     antibodies are at increased risk for graft failure:
 8. Opelz G et al (2010) Impact of HLA compati-            a Collaborative Transplant Study report. Hum
    bility on lung transplant survival and evidence        Immunol 70:569–573
    for an HLA restriction phenomenon: a collab-       17. Opelz G et al (1981) Lymphocytotoxic anti-
    orative transplant study report. Transplantation       body responses to transfusions in potential kid-
    90:912–927                                             ney transplant recipients. Transplantation 32:
 9. Opelz G, Döhler B (2010) Pediatric kidney              177–183
    transplantation: analysis of donor age, HLA        18. El-Awar N et al (2002) Almost all patients who
    match, and posttransplant non-Hodgkin lym-             are waiting for a regraft of a kidney transplant
               15   Impact of HLA Matching and HLA Antibodies in Organ Transplantation…                     277
Abstract
Antibodies against MICA have been found in organ transplant recipients and were found to be associated
with decreased survival of kidney allografts. The MICA antibody screening assay is a Luminex-based solid
phase immunoassay designed to detect IgG antibodies binding to beads pre-coated with recombinant
preparations of MICA alleles. These beads coated with soluble MICA recombinant proteins including 11
common alleles have been produced in our laboratory and similar preparations have been available from
commercial sources. Here, we describe the procedure of MICA antibody screening with a prepared kit, in
which all the reagents were optimized and standardized. We also review how to document the quality of
single MICA antigen beads using MICA-specific monoclonal antibodies, as well as quality control of the
procedure and data analysis.
     Key words: Single antigen MICA beads, Antibodies, Luminex, MICA epitopes, Positive and negative
     control sera
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_16, © Springer Science+Business Media New York 2012
                                                         279
280     Y. Zou and P. Stastny
2. Materials
                          1. Serum Samples.
                             Freshly obtained serum is the specimen of choice. A sterile clot-
                             ted blood sample with no anticoagulant is preferred (see Note 1).
                             The following reagents are calculated for 25 tests:
                          2. Bead Mixture: (500 mL): We use a mixture of 11 beads coated
                             with single MICA glycoproteins and two control beads. The
                             storage buffer is optimized and the preparation is ready for
                             testing human sera. Do not dilute or concentrate these beads.
                             Protect the beads from light by keeping exposure to light to a
                             minimum. It is possible to store at 2–8°C in the dark for 4
                             weeks or to store them at −80°C in the dark for 6 months.
                             Ensure the beads are resuspended before dispensing.
                          3. Sample Dilution Buffer 5× (500 mL): This sample dilution buf-
                             fer is used to reduce the background signals with human serum.
                             The sample dilution buffer must be diluted to a working solu-
                             tion (1×) with 2 mL of PBS pH = 7.4 containing 0.05% Tween-
                             20 (prepared by user) and can be stored at 2–8°C.
                          4. Positive Control Serum (40 mL ready to use): This serum or
                             serum mixture is obtained by combining sera which contain
                             MICA-specific antibodies to all the MICA antigens on the
                             beads. Store at 2–8°C (see Note 1) (5). Negative Control
                             Serum (40 mL ready to use): This serum is obtained from an
                             individual known to have no antibodies against MICA anti-
                             gens. Store at 2–8°C (see Note 2).
                          5. Secondary Antibody-PE: (20 mL) Goat anti-Human IgG con-
                             jugated to phycoerythrin in PBS. Must be diluted by adding
                             appropriate amount of sample dilution buffer (1×) before use
                             (we use 1,630 mL). Store at 2–8°C (see Note 3).
                          6. Wash Solution: 0.05% Tween-20/PBS (PBST): Not provided
                             in the kit and should be prepared by the user.
                          7. Luminex beads with recombinant MICA antigens (Table 1) in
                             the storage buffer (0.05% Tween-20/PBS plus 0.05% Azide).
                          8. Luminex 100 System (Luminex, Austin, TX).
                 16   Screening for Antibodies Against MICA by Luminex Flow Cytometry   281
                                Table 1
                                Luminex beads and MICA antigens attached
                                to them (see Note 4)
3. Methods
3.1. The Microbead        1. Collect serum samples to be tested and centrifuge the samples
Assay for MICA               for 10 min at 1,300 × g (see Note 6).
Antibody Detection        2. Prepare working solution (1×) from sample dilution buffer
                             (5×, 500 mL) in a new tube by adding 2 mL of PBS pH7.4,
                             05% tween-20 (prepared by user).
                          3. Add 25 mL of the working solution (1×) to each well that is
                             going to be used for the assay in a 96-well black Whatman tray.
                             Save remaining buffer for the secondary antibody dilution.
                          4. Add 15 mL of each centrifuged serum sample to each appropri-
                             ately assigned well and mix with the preloaded working solu-
                             tion 1× (see Note 7). Cover the plate with Thermowell sealing
282   Y. Zou and P. Stastny
3.2. Quality Controls                 1. Positive control and negative control sera should be included
                                         with each test. The MFI values of each bead with the control
                                         sera indicate if the test is valid (see Note 9).
                                      2. Different reagent lots in kits must never be mixed.
                                      3. Monoclonal antibodies against MICA can be used to assess
                                         conjugation of MICA antigens to the beads.
                                      Current lot Luminex beads are coated with 11 preparations of
                                  single MICA antigens, including MICA*001, *002, *004, *006,
                                  *007, *008, *009, *012, *017, *018, and MICA*019, representing
 Table 2
 Data analysis form for Luminex
Fig. 1. Reaction of single MICA antigen beads with MICA-specific monoclonal antibodies. The beads were tested with mAb
6B3 (black bars) or 3.2H3 (gray bars) or normal mouse IgG (mIgG, open bars) on 11 single antigen beads named as
rMICA*001,*002, *004, *006, *007, *008, *009, *012, *017, *018, and *019. Results are given as mean fluorescence
intensities (MFI).
                               the 11 most common MICA alleles. All of these beads give strong
                               signals with the monomorphic MICA-specific monoclonal anti-
                               body 6B3, which is a monoclonal antibody recognizing native
                               MICA proteins. All are positive also with 3.2H3, except for
                               MICA*001, which is known not to be recognized by this anti-
                               body. The mean fluorescence intensities vary between 6,000 and
                               10,000. Normal mouse IgG and beads without antigen show very
                               low background (Fig. 1).
3.3. Analysis of Results         1. Data files are generated with the bead numbers and their
                                    median fluorescence intensities (MFI) in spreadsheet format.
                                    The raw MFI data obtained by Luminex flow cytometry are
                                    converted to the relative amounts of MICA antibodies by sub-
                                    tracting the background. The threshold is determined for each
                                    bead using the means of the relative amount plus threefold SD
                                    of IgG binding in serum from normal subjects (n > 20) as
                                    shown in Table 3. Based on the relative amount of each MICA
                                    allele-specific antibody and the threshold for each antigen bead
                                    (see Note 10), scores are assigned as follows: 1 = negative;
                                    2 = doubtful, ³threshold and < threshold + 600; 4 = weak posi-
                                    tive, ³threshold + 600 and < threshold + 1,800; 6 = positive,
                                    ³threshold + 1,800 and < threshold + 5,400; 8 = _ strong posi-
                                    tive, ³threshold + 5,400.
                  16    Screening for Antibodies Against MICA by Luminex Flow Cytometry     285
                            Table 3
                            Threshold values above which results are considered positive
                            (score ≥ 4)
 Table 4
 Common serologic patterns of antibodies against MICA
ID Pattern *001 *002 *004 *006 *007 *008 *009 *012 *017 *018 *019
  1   MICA-G1            +         +                  +                  +      +     +
  2   MICA-G2                            +     +            +      +                         +
  3   MICA-G3            +               +     +      +     +      +     +            +      +
  4   MICA-G4            +         +     +     +      +            +     +      +     +
  5   MICA-G5            +                            +                  +            +
  6   MICA-0217                    +                                            +
  7   MICA-469                           +     +                         +
  8   MICA-0819                                             +
  9   MICA-46919                         +     +                   +                         +
 10   MICA-11218         +                                               +            +
 11   MICA-01            +
 12   MICA-04                            +
 13   MICA-06                                  +
 14   MICA-12                                                            +
4. Notes
         2. Negative control serum gives a low MFI with all beads except
            the positive control beads.
         3. Since the fluorescence of diluted Ab2-PE may change with
            time, make dilution immediately before use. The optimal con-
            centration of Ab2-PE was determined by titration. It will be
            different with each new batch of second antibody or new
            kit lot.
         4. MICA single antigen beads were developed in our laboratory.
            Commercial MICA single antigen beads are available from
            Tepnel (6) and One Lambda (3). Validation of each kit of
            beads is needed before use in testing. Serum exchange, parallel
            testing, and antibody absorption studies are useful to deter-
            mine the sensitivity, specificity, and stability of the MICA anti-
            body test kit.
         5. Positive control beads (#03) were pre-coated with the purified
            normal human IgG. Negative control beads (#02) were pre-
            pared in the same way as other beads, but no antigen was
            added. High MFI reactivity with bead #03 indicates that the
            appropriate anti-human IgG secondary antibody was added to
            each of the positive control sera, negative control serum, and
            patient serum assay wells.
         6. If there are suspended particles on the surface of the serum
            after centrifugation, do not include the suspended particles
            when adding serum samples for testing.
         7. Pipetting accuracy is important, since dilution of patient serum
            is performed as part of the assay. Do not pipet 15 mL patient
            serum with a 200-mL pipetting device.
         8. Complete washing is important and necessary before you add
            the secondary antibodies. A common error when using the
            assay is incomplete washing. This could result in a low MFI in
            the positive control beads, and could cause false negative
            results.
         9. The positive control serum supplied in the kit should give a
            high MFI for each of the antigen beads. The negative control
            serum in the kit should not give an MFI of more than 1,000
            except the MFI from positive control (beads #03).
        10. Each laboratory should determine thresholds for positive tests
            similar to the methods described here for defining cutoff
            values.
        11. A few patient sera will exhibit high background binding. They
            may require special treatment in order to lower the background
            nonspecific binding so that the assay can be correctly inter-
            preted. Antibody absorption and elution can be used to resolve
            questionable results in most cases.
288      Y. Zou and P. Stastny
References
 1. Bahram S, Bresnahan M, Geraghty DE et al            and MICA antibodies on kidney graft survival.
    (1994) A second lineage of mammalian major          Am J Transplant 7:408–415
    histocompatibility complex class I genes. Proc   4. Zou Y, Stastny P, Süsal C et al (2007) Antibodies
    Natl Acad Sci 91:6259–6263                          against MICA antigens and kidney-transplant
 2. Zou Y, Heinemann FM, Grosse-Wilde H et al           rejection. N Engl J Med 357:1293–1300
    (2006) Detection of anti-MICA antibodies         5. Zou Y, Qin Z, Silveus A et al (2009)
    in patients awaiting kidney transplantation,        Polymorphisms of MICA recognized by human
    during the post-transplant course, and in elu-      alloantibodies. Immunogenetics 61:91–100
    ates from rejected kidney allografts by          6. Duquesnoy RJ, Mostecki J, Hariharan J et al
    Luminex flow cytometry. Hum Immunol 67:             (2008) Structurally based epitope analysis of
    230–237                                             major histocompatibility complex class I-related
 3. Terasaki PI, Ozawa M, Castro R (2007) Four-         chain A (MICA) antibody specificity patterns.
    year follow-up of a prospective trial of HLA        Hum Immunol 69:826–832
                                                                                        Chapter 17
Abstract
Solid phase immunoassays for the detection and characterization of HLA-specific antibodies provide
greatly increased sensitivity, specificity, and time and reagent efficiency, compared to the traditionally used
cell-based methods. Testing is performed using commercially available test kits. The assays are of two gen-
eral types: enzyme-linked immunosorbent assays and multianalyte bead. The types vary in both sensitivity
and equipment requirements.
      While these assays afford great improvement over the cell-based assays, they can be confounded by
interference from substances within the serum that result in high background reactivity. The high sensitiv-
ity of the assays also makes them more susceptible to environmental factors and operator variability. The
user must be aware of the capabilities of the various formats, the factors that can affect test results, and lot
to lot variability of any single product. Knowledge of the characteristics of each product and thorough and
accurate analysis of the results are essential to the utility of these assays.
     Key words: Enzyme-linked immunosorbent assays, HLA antibody, HLA phenotype panel, Luminex®,
     Multianalyte bead assays, Pooled antigen panel, Single antigen panel, Solid phase immunoassays
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_17, © Springer Science+Business Media New York 2012
                                                         289
290        A.A. Zachary et al.
1.1. Commercially           Kits for the detection and characterization of HLA-Ab are available
Available Assays            in three formats: pooled HLA antigens, panels of individual HLA
                            phenotypes, and panels of single HLA antigens (4, 5). Pooled anti-
1.1.1. Formats
                            gen kits, as the name implies, consist of a pool or mixture of vari-
                            ous HLA antigens and generally include the most common HLA
                            specificities found in several racial groups. The pools may be derived
                            through isolation of antigens from the cells of hundreds of indi-
                            viduals or by mixing antigens prepared in transfected cell lines or
                            baculovirus systems. The pools may be comprised of class I (cI)
                            antigens only, class II (cII) only, or a mixture of cI and cII if the
                            platform used can distinguish the individual antigen classes in a
                            mixture. Phenotype panels are comprised of an array of individual
                                    17   HLA Antibody Detection and Characterization…        291
1.1.2. Assay Types,       Two general types of assays are available, multiplex or multianalyte
Matrices, and Platforms   bead assays and enzyme-linked immunosorbent assays (ELISA) (4,
                          5, 7). For both types of assay, commercial kits provide the matrix
                          with bound soluble HLA antigens. The bead assays are performed
                          on either a conventional flow cytometer or a Luminex® fluoroanalyzer,
                          depending on the kit. In these assays, polystyrene beads are impreg-
                          nated with fluorescent dyes and beads bearing different pools of
                          antigens, phenotypes, or single antigens are differentiated by the
                          fluorescence of the bead. After incubation with control or test sera,
                          a fluorochrome-labeled anti-human globulin is added to detect
                          beads that have antibody bound. When tested in a conventional
                          flow cytometer, beads of different fluorescence are identified on the
292      A.A. Zachary et al.
2. Materials
                     4. Calibration beads.
                     5. Microcentrifuge.
                     6. Microfuge tubes.
                     7. Millipore filter plates or microtiter plates.
                     8. Multiscreen vacuum manifold (kit dependent).
                     9. Plate holder for centrifuge.
                    10. Rotator or plate shaker.
                    11. Various adjustable and multichannel pipettes.
                    12. Vortex mixer.
                    13. Computer.
3. Methods
4. Notes
                              control bead values (<50 MFI), with unusually low test bead
                              values (<100 MFI) (together) may indicate that serum was not
                              added to the assay.
                         18. Test plates must be covered during incubations to prevent
                             evaporation.
                         19. Inadequate washing before addition of antiglobulin will result
                             in reduced specific reactivity due to blocking of the antiglobu-
                             lin reagent by residual immunoglobulin.
                         20. Inadequate washing before addition of substrate will increase
                             non-specific reactivity due to reactivity between free conju-
                             gated antiglobulin and substrate.
                         21. Colorimetric enzyme substrate and substrate buffer are light
                             sensitive and should be mixed immediately prior to use.
                         22. Time and temperature of the substrate incubation are critical
                             and failure to adhere to manufacturer’s specifications may lead
                             to increased background.
                         23. Optimal reactivity requires that stopping solution be added at
                             the required time.
                         24. Plate readers should be calibrated according to manufacturer’s
                             recommendations or whenever problems arise. If trays are
                             stored for rereading, OD values will diminish over time.
5. Data Analysis
and Interpretation
                         Analysis software is available for all products and the manufacturers
                         also provide calculations for performing analyses manually. The ini-
                         tial calculations performed by the software are to determine posi-
                         tivity and negativity. These calculations take into account the
                         overall reactivity of the assay as determined from the negative con-
                         trol bead(s) and background noise due to individual bead variabil-
                         ity. The values for background noise are lot specific. Also, different
                         assays within and between manufacturers may yield different negative
                         and positive control bead values. Thresholds for positivity can be
                         altered by the user. The software can also perform analyses to
                         determine specificity, correlation coefficients, chi-squared values,
                         two-by-two analysis data for each specificity, and values normalized
                         for antigen density. The analysis software can: provide customized
                         reports designed by the user and lot-specific updates for back-
                         ground noise; allow the user to view all data for a particular patient;
                         search by sample, patient, or test run; and generate single patient
                         or batch reports in a variety of document formats.
                              Specificities may be analyzed by antigen, allele, or cross-reac-
                         tive antigen groups.
          17   HLA Antibody Detection and Characterization…        299
6. Quality Control
                         Each lot of kits should be tested with human serum from a healthy,
                         non-sensitized male or a pool of sera from such individuals. The
                         serum should be known to lack HLA-Ab and should give unques-
                         tionable negative reactions with all or nearly all targets, individual
                         beads, or wells (except the internal positive control if one is included
                         in the kit). Higher than expected reactions with any target may
                         indicate the likelihood of non-specific reactivity and/or higher
                         than expected reaction strength with that target in the presence of
                         antibody specific for the target. Reactivity with the negative con-
                         trol serum also provides information about the different back-
                         ground reactivities of the targets, although this should be accounted
                         for by the manufacturer’s software.
                              Each new lot of kits should be tested with several sera contain-
                         ing different, well-characterized, HLA-Ab to check the sensitivity
                         and specificity of the lot.
                              Each run should include positive and negative control sera to
                         determine the validity of the run.
                              Ongoing monitoring of each target should be performed to
                         assess the possibility of inappropriately low reaction strength or
                         increased background. The data should also be used to evaluate
                         lot-to-lot differences in reaction strength.
                              Positive and negative control targets should be monitored for
                         consistency and level of reactivity.
                              Whenever new targets are introduced into a new lot, their
                         reactivity should be evaluated with antibody specific for the HLA
                         antigen(s) of that target.
7. Technical Issues
Fig. 1. Test serum vs. positive control. Run-to-run variability must be accounted for when interpreting results of HLA anti-
body testing. The strength of HLA-Abs from sequential serum specimens from the same patient is plotted against the
strength of the positive control. The left side of the plot shows an unmistakable increase in HLA-Abs with a much more
limited change in the reaction strength of the positive control. However, beginning with week 6 (dotted vertical line), the
HLA-Ab strength and that of the positive control are parallel suggesting that apparent changes in the HLA-Ab strength most
likely reflect day-to-day differences in test sensitivity.
8. Applications
9. Considerations
and Recommend-
ations
                    The review of the capabilities and shortcomings of various formats
                    and platforms suggests that complete and accurate information
                    about a patient’s HLA-Abs is best obtained with the use of more
                    than one assay. While this may appear to be cost prohibitive, by
                    customizing the use of multiple assays according to the antibodies
                    of a patient, we have been able to maintain test costs in the lowest
                    quartile of independent US laboratories.
                         There has been much recent discussion about two issues sur-
                    rounding SPI: results reporting and standardization. There is inter-
                    est in having some quantitative measure of sensitization using test
                    values expressed as MFIs or OD ratios. As we have noted above,
                    the values are affected by several factors, including: the antibody
                    specificity, with antibodies to enhanced antigens yielding higher
                    values; by the format used, with higher values obtained with single
                    antigen panels than with phenotype panels; by the sensitivity of the
                    particular lot of reagents used; by the presence of interference
                    causing high background and reduced test values; and by the day-
                    to-day variability inherent in serologic assays. All of these factors
                    must be taken into consideration for a meaningful interpretation of
                    the test results. The trend of DSA and the crossmatch reactivity
                    that would be predicted from the SPI results are likely to be more
                    meaningful clinically. However, this implies that thorough and
                    accurate analysis of the test results by the laboratory scientist and
                    technologist is absolutely necessary to providing such meaningful
                    interpretation.
                         Standardization of test results would be extremely useful for
                    interpreting data from different laboratories. However, there are
                    several hurdles to overcome to achieve this goal. Most impor-
                    tantly, at present there are no reference sera with known amounts
                    of antibody to use as standards. Monoclonal antibodies are not
                    representative of the polyclonal nature of antibodies present in
                    most patients and even mixtures of such antibodies would not
                    replicate the nature of patient’s sera which contains other factors
                    that could affect test results. Second, while some laboratories treat
                    sera to remove interfering factors, at present this is not done
                    universally. Third, the high degree of sensitivity of SPI and its
                    susceptibility to variability suggests that standardization might
                    only be achieved by considering ranges of test values. Fourth,
                    standardization cannot be readily achieved until manufacturer-
                    provided kits are standardized from lot to lot. The question is,
                    how much of a problem is this. A perusal of the literature reveals
                    that multiple variations of the cytotoxicity crossmatch have been
                    used. Differences in incubation times and temperatures, the use of
                    an antiglobulin, the source and specificity of the antiglobulin, and
306       A.A. Zachary et al.
References
 1. Oh SK et al (1975) Soluble HL-A antigens in          8. Zeevi A, Girnita A, Duquesnoy R (2006) HLA
    serum. I. Isolation and purification. Eur J             antibody analysis: sensitivity, specificity and
    Immunol 5:161–166                                       clinical significance in solid organ transplanta-
 2. Buelow R et al (1995) Detection of panel-               tion. Immunol Res 36:255–264
    reactive anti-HLA class I antibodies by enzyme-      9. Phelan D et al (2009) Living donor renal
    linked       immunosorbent          assay     or        transplantation in the presence of donor-
    lymphocytotoxicity. Results of a blinded, con-          specific human leukocyte antigen antibody
    trolled multicenter study. Hum Immunol                  detected by solid-phase assay. Hum Immunol
    44:1–11                                                 70:584–588
 3. Zachary AA et al (1995) Evaluation of HLA           10. Leffell MS, Montgomery RA, Zachary AA
    antibodies with the PRA-STAT test. An ELISA             (2005) The changing role of antibody testing
    test using soluble HLA class I molecules.               in transplantation. Clin Transpl 259–271
    Transplantation 60:1600–1606                        11. Bartel G et al (2007) Determinants of the
 4. Zachary AA, Leffell MS (2008) Detecting and             complement-fixing ability of recipient presen-
    monitoring human leukocyte antigen-specific             sitization against HLA antigens. Transplantation
    antibodies. Hum Immunol 69:591–604                      83:727–733
 5. Tait BD (2009) Solid phase assays for HLA           12. Smith JD et al (2007) C4d fixing, Luminex
    antibody detection in clinical transplantation.         binding antibodies—a new tool for prediction
    Curr Opin Immunol 21:573–577                            of graft failure after heart transplantation. Am
 6. Zachary AA et al (2009) Using real data for a           J Transplant 7:2809–2815
    virtual     crossmatch.     Hum         Immunol     13. Wahrmann M et al (2009) Clinical relevance of
    70:574–579                                              preformed C4d-fixing and non-C4d-fixing
 7. Fuggle SV, Martin S (2008) Tools for human              HLA single antigen reactivity in renal allograft
    leukocyte antigen antibody detection and their          recipients. Transpl Int 22:982–989
    application to transplanting sensitized patients.   14. Chin C et al (2011) Clinical usefulness of a
    Transplantation 86:384–390                              novel C1q assay to detect immunoglobulin G
                                           17   HLA Antibody Detection and Characterization…              307
42. Zachary AA et al (2003) Specific and durable     48. Bielmann D et al (2007) Pretransplant risk
    elimination of antibody to donor HLA anti-           assessment in renal allograft recipients using
    gens in renal-transplant patients. Transplant-       virtual crossmatching. Am J Transplant
    ation 76:1519–1525                                   7:626–632
43. Locke JE et al (2009) Proinflammatory events     49. Aubert V et al (2009) Low levels of human
    are associated with significant increases in         leukocyte antigen donor-specific antibodies
    breadth and strength of HLA-specific anti-           detected by solid phase assay before transplan-
    body. Am J Transplant 9:2136–2139                    tation are frequently clinically irrelevant. Hum
44. Zachary AA, Leffell MS (2010) Barriers to            Immunol 70:580–583
    successful transplantation of the sensitized     50. Terasaki PI, Cai J (2008) Human leukocyte
    patient. Expert Rev Clin Immunol 6:449–460           antigen antibodies and chronic rejection: from
45. Verghese PS et al (2010) Impaired graft sur-         association to causation. Transplantation
    vival in pediatric renal transplant recipients       86:377–383
    with donor-specific antibodies detected by       51. El-Awar N et al (2009) Epitopes of HLA-A, B,
    solid phase assays. Pediatr Transplant               C, DR, DQ, DP, and MICA antigens. Clin
    14:730–734                                           Transpl 295–321
46. Gibney EM et al (2006) Detection of donor-       52. Cano P, Fernández-Viña M (2009) Two
    specific antibodies using HLA-coated micro-          sequence dimorphisms of DPB1 define the
    spheres: another tool for kidney risk                immunodominant serologic epitopes of
    stratification. Nephrol Dial Transplant              HLA-DP. Hum Immunol 70:836–843
    21:2625–2629                                     53. Marrari M et al (2010) Human monoclonal
47. Leffell MS, Zachary AA (2010) Anti-allograft         antibody reactivity with human leukocyte anti-
    antibodies: some are harmful, some can be            gen class I epitopes defined by pairs of mis-
    overcome, and some may be beneficial. Discov         matched eplets and self-eplets. Transplantation
    Med 9:478–484                                        90(12):1468–1472
                                                                                        Chapter 18
Abstract
T cell alloreactivity is responsible for much of the morbidity and mortality associated with tissue transplan-
tation and graft versus host disease. Immunoassays for ex vivo monitoring and quantitation of alloreactive
T cells are being increasingly utilised to provide valuable information for individualised clinical manage-
ment of transplant recipients. Here we describe detailed methodologies for both traditional and novel
assays utilised for the detection, quantitation, and functional characterisation of alloreactive T cells and
highlight the key advantages and disadvantages of each system.
     Key words: Allogeneic, Alloreactivity, 51Cr release assay, Enzyme-linked immunospot, Graft versus
     host disease, Intracellular cytokine staining, T cells, T cell receptors, Tetramer staining
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_18, © Springer Science+Business Media New York 2012
                                                         309
310      M. Bharadwaj et al.
1.1. 51Chromium (Cr)     Despite the increasing reluctance to utilise radioactive assay systems,
Release Assays           more than 4 decades after it was first developed, the 51Cr release
                         assay is still considered as the gold standard for measuring the
                         cytolytic efficiency of CD8+ T cells for both alloreactivity (8, 9) and
                         anti-viral T cell responses (10, 11). Host PBMCs or alloreactive T
                         cells proliferating in an LDA or mixed lymphocyte culture are co-
                         incubated with 51Cr labelled, allogeneic stimulators (targets), and
                         the 51Cr released in the culture medium is measured after 4–5 h.
                         This provides a measurement of the specific lysis of the 51Cr labelled
                         targets by the alloreactive T cells. Whilst a direct correlation has
                         been demonstrated between precursor frequencies determined by
                         an LDA and clinical data on prediction and outcomes of GVHD
                         (12–15), there are also reports suggesting that the enumeration of
                         allo-specific T cell precursor frequencies by an LDA may not cor-
                         relate with human transplant outcomes (16–18). Furthermore,
                         studies based on anti-viral T cells in Epstein–Barr virus (EBV),
                         Influenza virus, and Lymphocytic Choriomeningitis virus infection
                         have highlighted inherent biological and technical problems asso-
                         ciated with the LDA (12, 19–22). Overall, the LDA may only
                         assess the precursor frequency of a single dominant alloantigen
                         rather than accurately reflecting the complexity of the alloresponse
                         generated by all of the mismatched HLA antigens.
                           18    Detection and Characterisation of Alloreactive T Cells                  311
1.3. Peptide/MHC         The design and application of soluble MHC tetrameric complexes
Tetramer Staining        for investigating anti-viral CD8+ T cells was first described in
                         1996 (32). Subsequently, tetrameric complexes have revolu-
                         tionised the quantitation and dissection of antigen-specific T cell
                         responses (32–35). The generation of pMHC tetramers is
                         described in a number of publications (7, 23, 32). The HLA/b2m/
                         peptide tetrameric complex is conjugated to a fluorochrome of
                         interest, such as phycoerythrin (PE), fluorescein isothiocyanate
                         (FITC), or allophycocyanin (A-PC), and used as a staining reagent
                         for flow cytometry. MHC class I tetramers have been extensively
                         used for the detection of antigen-specific CD8+ T cells in both
                         viral infections (10) and alloreactive conditions (9). However,
                         more recently the development of MHC class II tetramers
                         ( 36– 38) have further advanced the analysis of CD4+ T cells in
                         the fields of autoimmunity (39–41), tumour antigens (42), and
                         virology (43).
                              It is noteworthy that CD8+ T cell frequencies quantitated
                         by tetramer staining have been shown to be higher than those
                         detected by conventional cytotoxicity assays (44). This is probably
                         due to the ability of tetramers to detect different T cell pheno-
                         types including naïve, memory, and effector cells and the ten-
                         dency for memory T cells to undergo apoptosis upon activation
                         (activation-induced cell death) resulting in a loss of reactive T cells
                         in culture (45). Compared to the traditional assays, the pMHC
                         complexes (1) are quantitative and sensitive (1:50,000), (2) do
                         not involve radioactivity, (3) require minimal material (blood,
                         tissue), (4) are a fast approach, (5) permit batch processing for
                         large numbers, (6) can be used in combination with other cell
                         surface antibodies used for T cell phenotyping (10, 19, 26, 32,
                         46, 47), and (7) do not kill the T cells allowing the cells to be
                         sorted for use in other assays (such as ELISPOT and intracellular
                         cytokine staining [ICS]). Recent studies investigating the anti-
                         viral T cells (10) and/or cross-reactive virus-specific T cells recog-
                         nising specific HLA alloantigens have demonstrated the power in
                         combining both pMHC tetramers and the ICS assay (10, 48, 49)
                         (described below). The only major limitation of this assay is that
                         both the peptide epitope and MHC-restriction elements must be
                         known for the detection of specific T cells.
                              18   Detection and Characterisation of Alloreactive T Cells    313
1.4. Intracellular       The activation of T cells initiates the production of cytokines that
Cytokine Staining        can be detected and quantitated using ICS (7, 50, 51). Briefly,
                         T cells are activated through co-incubation with a stimulus for a total
                         of 6 h. An inhibitor of intracellular transport is added after the first
                         2 h to arrest the transport of newly synthesised proteins, including
                         cytokines, from the endoplasmic reticulum into the Golgi. Post-
                         incubation, the T cells are stained with a cocktail of specific mono-
                         clonal antibodies (mAbs) for cell surface phenotyping. The T cells
                         are then paraformaldehyde fixed, permeabilised in mild detergent,
                         and stained with anti-cytokine antibodies. Phenotypic analysis is
                         facilitated by flow cytometry.
                              The T cell frequencies detected by ICS have been shown to
                         correlate with frequencies obtained by pMHC tetramers in HIV
                         patients (52) and with ELISPOT in an Influenza study of healthy
                         individuals (53). Conversely, no correlation was demonstrated
                         between the frequencies detected by ICS and pMHC tetramers in
                         a metastatic melanoma study (54), and the enumeration of EBV-
                         specific T cells and alloreactive T cells by ICS yielded higher fre-
                         quencies than that obtained by the LDA-based 51Cr release assay
                         (55). Notwithstanding the discrepancies in the frequency of T cells
                         enumerated by the different assay systems, ICS has been used for
                         the detection of cytokine producing CD4+ T cells in the peripheral
                         blood of individuals with HIV (56) and quantification of activated
                         allo-specific CD8+ T cells in healthy controls (9, 57) and solid
                         organ transplant recipients (58). It has various advantages over the
                         other assays described above, including (1) analysis of large num-
                         bers of cells, (2) multiparameter analysis using combinations of
                         intracellular and cell surface antibodies, (3) parallel measurement
                         of T cell cytokine profiles, and (4) measurement of cytotoxic poten-
                         tial using a degranulation marker.
2. Materials
3. Methods
                         Table 1
                         Selection of cell culture plates
3.1. Isolation of PBMC    1. Collect peripheral blood in appropriate blood collection tubes
and In Vitro Expansion       containing anti-coagulant (see Note 7).
of Allo-Specific CD8+     2. To a 50 mL tube, add 5–20 mL of peripheral blood and an
T Cells                      equal volume of RPMI to achieve a 1:2 dilution of the periph-
                             eral blood.
                          3. To another 50 mL tube, add 10 mL of Ficoll-Paque PLUS
                             (see Note 8).
                          4. Using a 25 mL serological pipette gently overlay the diluted
                             peripheral blood onto the Ficoll-Paque PLUS reagent using
                             the slow setting on the pipette gun (see Note 9).
                          5. Centrifuge for 20 min at 931 × g, room temperature with the
                             brake setting = OFF (see Note 10).
                          6. Blood components will be fractionated and plasma can be col-
                             lected using a 1 mL transfer pipette and stored at −80°C if
                             required (see Fig. 2).
                          7. Using a sterile 1 mL transfer pipette, carefully collect lympho-
                             cyte/monocyte/platelet layer (interface) into a 30 mL
                             V-bottom tube. Wash cells in a final volume of 20 mL of RPMI.
                          8. Centrifuge for 15 min at 524 × g, room temperature.
                          9. A cell pellet should be visible at the bottom of the tube, discard
                             supernatant by aspiration, leaving approximately 1 mL of resid-
                             ual volume and resuspend cell pellet.
                         10. Transfer the cell pellet to a 10 mL tube containing 8 mL of
                             RPMI and centrifuge for 10 min at 524 × g, room temperature.
                             Repeat wash by resuspending the cell pellet in 8 mL of RPMI
                             and centrifuging for 10 min at 524 × g, room temperature.
320         M. Bharadwaj et al.
Fig. 2. Isolation of PBMCs. Peripheral blood is layered over the Ficoll-Paque and, following centrifugation (with brake OFF),
the blood components are separated into (1) plasma, (2) lymphocytes, monocytes, platelets, and (3) granulocytes, erythro-
cytes. The lymphocytes, monocytes, and platelets layer can be extracted, centrifuged to remove platelets with the residual
cells representing the peripheral blood mononuclear cell population.
Fig. 3. In vitro expansion of allo-specific and antigen-specific CD8+ T cells. (a) PBMC from
a healthy individual was stimulated for up to 16 days with allogeneic PBMC that enabled
generation of allo-specific CD8 +T cells against all three mismatched HLA-A1, -B8, and
-B57 allotypes. The maximal CD8+ T cell response, measured by the production of IFN-g,
was observed at 12–13 days post-stimulation. (b) PBMC from a HLA-B8 healthy individual
was stimulated with autologous PBMC pulsed with EBV peptide RAKFKQLL (RAK) for up to
13 days. The maximal response, measured in HLA-B8/RAK tetramer+CD8+ T cells by the
production of IFN-g, was observed at 12–13 days post-stimulation.
322        M. Bharadwaj et al.
3.2. Dissection and          1. Preparation of responder (effector) cells: Count effector cells
Quantitation of the             and serially dilute in 2 mL of RF10 in a series of 10 mL tubes
Alloreactive CD8+               to achieve a cell concentrations of 2 × 106, 1 × 106, and 5 × 105
T Cell Response                 cells/mL. The aim is to achieve multiple replicates of an
                                effector:target (E:T) ratio of 100:1, 50:1, and 25:1 in the assay.
3.2.1. 51Cr Release Assay
                             2. Preparation of reporter (target) cells—51Cr labelling: Wear
                                protective eyewear, gown, radiation badge, and double gloves.
                                Scan the area in which you are planning to perform 51Cr label-
                                ling of target cells with a Geiger-Muller counter to assess if
                                there is a pre-existing background level of radiation. Use the
                                Geiger counter when using the isotope to monitor spills and
                                exposure (see Note 15).
                                 (a) Count allogeneic reporter or target cells and aliquot 3 × 106
                                     cells/10 mL tube into two tubes for test sample and posi-
                                     tive (viral) control (traditionally viral controls are target
                                     cells pulsed with 10 mg/mL of peptide antigen derived
                                     from EBV/Influenza/CMV, for 1 h at 37°C).
                                 (b) Centrifuge, both test and viral control tubes for 7 min at
                                     335 × g at room temperature to pellet the cells.
                                 (c) Discard supernatant by aspiration and resuspend the cell
                                     pellet in 100 mL of RPMI.
                                 (d) Label target cells with 51Cr by adding 50–100 mL of
                                     Na51CrO4 to each 10 mL tube using a 1-mL syringe (see
                                     Note 15).
                                 (e) Mix each tube gently and incubate for 1 h at 37°C.
                                 (f) Wash the 51Cr-labelled target cells in a final volume of
                                     10 mL in cold RF10 and centrifuge for 7 min at 800 × g at
                                     room temperature.
                                 (g) Repeat the wash step an additional two times.
                                 (h) Resuspend the 51Cr-labelled target cells in 1 mL of RF10.
                                 (i) Count cells and resuspend to 2 × 104 target cells/mL in
                                     RF10.
                             3. Add 100 mL of serially diluted responder (effector) cells
                                (2 × 106, 1 × 106, and 5 × 105 cells/mL—as described in step 1)
                                to wells of a 96-well U-bottom plate. Cells at each different
                                concentration (titrated for different E:T ratios) should be
                                added into three wells of a 96-well U-bottom plate (see Fig. 4).
                             4. Add 100 mL of 51Cr-labelled target cells (2 × 103 target cells) as
                                described in step 2 to each well of the 96-well U-bottom
                                plate.
                             5. Negative controls (media only): 3–6 wells containing 100 mL
                                of 2 × 103 target cells/well (without responders) plus an addi-
                                tional 100 mL RF10/well is used to calculate the spontaneous
                                release of 51Cr from the target cells.
       18    Detection and Characterisation of Alloreactive T Cells                        323
     Target 1
     Target 2
     Target 3
     Target 4
     Target 5
     Target 6
     Target 7
     Target 8
Fig. 4. Proposed plan for setting up the 51Cr release assay at an E:T ratio of 100:1 and
50:1. Other ratios may be tested similarly in different plates. Target 1–8 represent the
different cell lines (BLCL or transfected C1R) that need to be tested for allogeneic reactivity
to the responders (or effectors), the untransfected parent C1R cell line and a positive control
(reporter cell line pulsed with a viral peptide). All targets are resuspended in the assay media
at a set concentration (e.g. 2 × 104 target cells/mL) and the effectors or responder cells
are serially diluted to achieve an E:T ratio of 100:1, 50:1, and 25:1 (and more if required).
                           14. Using the mean plus 3 standard deviations (SD) cut-off, the
                               number of negative wells is determined for each dilution.
                               The relationship between the number of negative wells and the
                               mean number of precursors are plotted, and a frequency of
                               alloreactive T cells is obtained (3, 60, 61).
3.2.2. ELISPOT Assay       Procedures on day 0 and day 1 are to be performed under sterile
                           conditions in a Biohazard class II cabinet to maintain sterility of
                           plate and reagents.
                           Day 0: Coat Plate
                             1. Prepare capture mAb as per manufacturer’s instructions. For
                                example: dilute anti-IFN-g mAb (1-DIK anti-IFN-g mouse
                                IgG1) to 10 mg/mL in 0.1 M sodium bicarbonate buffer pH
                                9.6 or 1× PBS and dispense 100 mL to each well of the
                                ELISPOT plate (see Notes 18 and 19). Incubate plate with lid
                                overnight (approximately 18 h) at 4°C (refrigerator) on a level
                                surface.
                           Day 1:
                             2. Plate blocking: After overnight incubation, retrieve test plate
                                from 4°C incubation and wash wells by flicking off coating
                                mAb solution into discard tray. Then add 200 mL sterile PBS
                                per well and flick off into discard tray. Repeat washing another
                                five times. After the final wash add 200 mL per well of sterile
                                PBS with 5% FCS (blocking solution) to test plate, incubate
                                for 1 h at 37°C (or at room temperature if to be left longer).
                             3. Prepare effectors: PBMC (fresh or cryopreserved) or in vitro
                                expanded alloreactive T cells can be used as effector cells for
                                assessing the alloreactive response (see Note 20). Perform cell
                                count and adjust cell concentration to 2 × 106 cells/mL to seed
                                at a final concentration of 2 × 105 cells/well/100 mL in the
                                ELISPOT plate (see Note 19). If using in vitro expanded
                                alloreactive T cells, the cell concentration should be between
                                1 × 103–5 × 104cells/well.
                             4. Prepare targets: Allogeneic PBMC depleted of CD3+ T cells
                                (B cell enriched) are utilised (see Note 21). Relevant B-LCLs or
                                reporter cell lines (see Subheading 3 introduction) can also be
                                used although they may give a high background. Seed target
                                cells in limiting dilution (5 × 104, 2.5 × 104, and 1 × 104 cells/
                                well) in triplicate in a total volume of 100 mL to appropriate
                                wells.
                             5. Positive control: A final concentration of 10 mg/mL of either
                                phytohemagglutinin (PHA) or a known anti-viral peptide
                                (EBV, CMV, or Influenza A) restricted to the same HLA mol-
                                ecules expressed by the effector cells, in a total volume of 5 mL
                                serves as positive control.
     18   Detection and Characterisation of Alloreactive T Cells   325
                           18. Tease away the bottom plastic covering gently and wash the
                               bottom of the ELISPOT plate. Allow the plates to dry over-
                               night in open air on bench top. Once dry, the plate is ready to
                               be analysed using an ELISPOT reader.
                           19. Results are presented as mean values of spots detected in triplicate
                               wells containing effectors plus stimulator cells or antigen, after
                               subtracting the response of wells with responder cells or effector
                               cells alone (normally less than 10 spots per 200,000 cells).
3.2.3. Intracellular       Before an ICS is conducted, individual mAbs (to be used for phe-
Cytokine Staining Assay    notyping and functional analysis) should be titrated to determine
                           amounts for optimal staining (see Table 2). Prior to the assay,
                           alloreactive/responder T cells derived from an MLR are restimu-
                           lated by specific reporter cells (i.e. transfected cell lines or B-LCLs),
                           and the antigen-specific CD8+ T cells are restimulated by appropri-
                           ate reporter cells pulsed with the relevant peptide. Reporter cells
                           for alloreactive T cells should carry HLA allotypes corresponding
                           to the allogeneic stimulator (PBMC utilised for the initial stimula-
                           tion). For example, for dissecting T cells from an MLR in which
                           responder PBMC were expanded with an allogeneic stimulator
                           carrying: HLA-A*0101, A*0201; HLA-B*0801, B*5701, the
                           following reporter cell lines would be required:
                           Transfected cell line panel: C1R parental, C1R.A*0101,
                              C1R.A*0201, C1R.B*0801, C1R.B*5701.
                           B-LCL panel: Cell lines expressing individual HLA allotypes-
                              A*0101, A*0201, B*0801, or B*5701.
                             1. Calculate the total number of T cells and reporter cells required
                                for the assay (for the test sample, negative controls, and com-
                                pensation controls) considering that you will require 2 × 105
                                T cells/100 mL of assay media (RF10 + 20 U/mL of IL-2) per
                                well and 1 × 105 reporter cells/100 mL of assay media
                             Table 2
                             Selection of monoclonal antibodies used for characterising
                             cell phenotype and function
 Table 3
 Selection of pMHC tetramers used to quantify and dissect antigen-specific T cells
Fig. 5. Gating strategy for allo-specific CD8+ T cells. Identification of lymphocytes is achieved using a forward scatter (FSC)
vs. side scatter (SSC) dot plots. From the lymphocyte population, the percentage of CD8+ T cells can be measured using a
CD8 (FL3 channel) vs. SSC dot plot. Functional analysis of the CD8+ T cells can be quantitated using a CD107a (cytolytic
potential; FL1 channel) vs. IFN-g (cytokine production; FL4 channel).
Fig. 6. Gating strategy for antigen-specific CD8+T cells. Identification of lymphocytes is achieved using a FSC vs. SSC dot
plots. From the lymphocyte population, the percentage of CD8+ T cells can be measured using a CD8 (FL3 channel) vs. SSC
dot plot. Antigen-specific CD8+T cells are identified using a HLA-B8/FLRGRAYGL (FLR) Tetramer (FL2 channel) vs. SSC.
Functional analysis of the CD8+ T cells can be quantitated using a CD107a (cytolytic potential; FL1 channel) vs. IFN-g
(cytokine production; FL4 channel).
Fig. 7. Detection of circulating EBV-specific CD8+ T cells using pMHC tetramers. Ex vivo PBMC isolated from a HLA-B8
healthy individual were co-incubated with an anti-CD8 antibody in the presence of either the HLA-B8/RAK or HLA-B8/FLR
tetramer. The corrected frequencies, following the subtraction of the background value, for both RAK- and FLR-specific
CD8+ T cells detected were 1.1 and 0.6%, respectively.
4. Notes
Acknowledgements
References
 1. Burrows SR, Khanna R, Burrows JM, Moss DJ                dynamics of antigen-specific CD8+ cytotoxic T
    (1994) An alloresponse in humans is domi-                lymphocytes following primary Epstein–Barr
    nated by cytotoxic T lymphocytes (CTL) cross-            virus infection. Blood 98(8):2588–2589
    reactive with a single Epstein–Barr virus CTL        12. Kaminski E, Hows J, Man S, Brookes P,
    epitope: implications for graft-versus-host dis-         Mackinnon S, Hughes T et al (1989) Prediction
    ease. J Exp Med 179(4):1155–1161                         of graft versus host disease by frequency analy-
 2. Brunner KT, Mauel J, Cerottini JC, Chapuis B             sis of cytotoxic T cells after unrelated donor
    (1968) Quantitative assay of the lytic action of         bone marrow transplantation. Transplantation
    immune lymphoid cells on 51-Cr-labelled allo-            48(4):608–613
    geneic target cells in vitro; inhibition by isoan-   13. Schwarer AP, Jiang YZ, Deacock S, Brookes
    tibody and by drugs. Immunology 14(2):                   PA, Barrett AJ, Goldman JM et al (1994)
    181–196                                                  Comparison of helper and cytotoxic antirecipi-
 3. Lefkovits I (1972) Induction of antibody-                ent T cell frequencies in unrelated bone mar-
    forming cell clones in microcultures. Eur J              row transplantation. Transplantation 58(11):
    Immunol 2(4):360–366                                     1198–1203
 4. Lefkovits I, Waldmann H (1984) Limiting dilu-        14. Roelen DL, Stobbe I, Young NT, van Bree SP,
    tion analysis of cells of the immune system I.           Doxiadis II, Oudshoorn M et al (2001)
    The clonal basis of the immune response.                 Permissible and immunogenic HLA-A mis-
    Immunol Today 5:265–268                                  matches: cytotoxic T-cell precursor frequencies
 5. Waldmann H, Lefkovits I (1984) Limiting dilu-            reflect graft survival data. Hum Immunol
    tion analysis of cells of the immune system II:          62(7):661–667
    what can be learnt? Immunol Today 5:295–298          15. Dolezalova L, Vrana M, Dobrovolna M,
 6. Sharrock CE, Kaminski E, Man S (1990)                    Loudova M, Cukrova V, Vitek A et al (2002)
    Limiting dilution analysis of human T cells: a           Cytotoxic T lymphocyte precursor frequency
    useful clinical tool. Immunol Today 11(8):               analysis in the selection of HLA matched unre-
    281–286                                                  lated donors for hematopoietic stem cell trans-
 7. Hickling JK (1998) Measuring human                       plantation: the correlation of CTLp frequency
    T-lymphocyte function. Expert Rev Mol Med                with HLA class I genotyping and aGVHD
    1:1–20                                                   development. Neoplasma 49(1):26–32
 8. Kaminski E, Hows J, Brookes P, Mackinnon S,          16. Fussell ST, Donnellan M, Cooley MA, Farrell
    Hughes T, Avakian O et al (1989) Alloreactive            C (1994) Cytotoxic T lymphocyte precursor
    cytotoxic T-cell frequency analysis and HLA              frequency does not correlate with either the
    matching for bone marrow transplants from                incidence or severity of graft-versus-host dis-
    HLA matched unrelated donors. Transplant                 ease after matched unrelated donor bone mar-
    Proc 21(1 Pt 3):2976–2977                                row        transplantation.      Transplantation
 9. Macdonald WA, Chen Z, Gras S, Archbold JK,               57(5):673–676
    Tynan FE, Clements CS et al (2009) T cell            17. Eberspacher ML, Otto G, Herfarth C, Kabelitz
    allorecognition     via    molecular     mimicry.        D (1994) Frequency analysis of donor-reactive
    Immunity 31(6):897–908                                   cytotoxic T lymphocyte precursors in in allograft
10. Bharadwaj M, Thammanichanond D, Aitken                   recipients. Lack of correlation with clinical out-
    CK, Moneer S, Drummer HE, Tracy S et al                  come.           Transplantation           57(12):
    (2009) TCD8 response in diverse outcomes of              1746–1752
    recurrent exposure to hepatitis C virus.             18. Wang XN, Taylor PR, Skinner R, Jackson GH,
    Immunol Cell Biol 87(6):464–472                          Proctor SJ, Hedley D et al (2000) T-cell fre-
11. Bharadwaj M, Burrows SR, Burrows JM, Moss                quency analysis does not predict the incidence
    DJ, Catalina M, Khanna R (2001) Longitudinal             of graft-versus-host disease in HLA-matched
336       M. Bharadwaj et al.
    sibling bone marrow transplantation. Trans-           30. Sherritt MA, Bharadwaj M, Burrows JM,
    plantation 70(3):488–493                                  Morrison LE, Elliott SL, Davis JE et al (2003)
19. Tan LC, Gudgeon N, Annels NE, Hansasuta P,                Reconstitution of the latent T-lymphocyte
    O’Callaghan CA, Rowland-Jones S et al (1999)              response to Epstein–Barr virus is coincident
    A re-evaluation of the frequency of CD8+ T                with long-term recovery from posttransplant
    cells specific for EBV in healthy virus carriers. J       lymphoma after adoptive immunotherapy.
    Immunol 162(3):1827–1835                                  Transplantation 75(9):1556–1560
20. Lalvani A, Brookes R, Hambleton S, Britton            31. Gazagne A, Claret E, Wijdenes J, Yssel H,
    WJ, Hill AV, McMichael AJ (1997) Rapid effec-             Bousquet F, Levy E et al (2003) A Fluorospot
    tor function in CD8+ memory T cells. J Exp Med            assay to detect single T lymphocytes simultane-
    186(6):859–865                                            ously producing multiple cytokines. J Immunol
21. Murali-Krishna K, Altman JD, Suresh M,                    Methods 283(1–2):91–98
    Sourdive DJ, Zajac AJ, Miller JD et al (1998)         32. Altman JD, Moss PA, Goulder PJ, Barouch
    Counting antigen-specific CD8 T cells: a                  DH, McHeyzer-Williams MG, Bell JI et al
    reevaluation of bystander activation during viral         (1996) Phenotypic analysis of antigen-specific
    infection. Immunity 8(2):177–187                          T lymphocytes. Science 274(5284):94–96
22. Kuzushima K, Hoshino Y, Fujii K, Yokoyama             33. McMichael AJ, O’Callaghan CA (1998) A new
    N, Fujita M, Kiyono T et al (1999) Rapid                  look at T cells. J Exp Med 187(9):1367–1371
    determination of Epstein–Barr virus-specific          34. Ogg GS, McMichael AJ (1998) HLA-peptide
    CD8(+) T-cell frequencies by flow cytometry.              tetrameric complexes. Curr Opin Immunol
    Blood 94(9):3094–3100                                     10(4):393–396
23. Hernandez-Fuentes MP, Warrens AN, Lechler             35. Miles JJ, Thammanichanond D, Moneer S,
    RI (2003) Immunologic monitoring. Immunol                 Nivarthi UK, Kjer-Nielsen L, Tracy SL et al
    Rev 196:247–264                                           (2011) Antigen-driven patterns of TCR bias
24. Miyahira Y, Murata K, Rodriguez D, Rodriguez              are shared across diverse outcomes of human
    JR, Esteban M, Rodrigues MM et al (1995)                  hepatitis C virus infection. J Immunol 186(2):
    Quantification of antigen specific CD8+ T cells           901–912
    using an ELISPOT assay. J Immunol Methods             36. James EA, LaFond R, Durinovic-Bello I, Kwok
    181(1):45–54                                              W (2009) Visualizing antigen specific CD4+ T
25. Czerkinsky C, Andersson G, Ekre HP, Nilsson               cells using MHC class II tetramers. J Vis Exp
    LA, Klareskog L, Ouchterlony O (1988)                     (25) pii:1167
    Reverse ELISPOT assay for clonal analysis of          37. Landais E, Romagnoli PA, Corper AL, Shires J,
    cytokine production. I. Enumeration of                    Altman JD, Wilson IA et al (2009) New design
    gamma-interferon-secreting cells. J Immunol               of MHC class II tetramers to accommodate
    Methods 110(1):29–36                                      fundamental principles of antigen presentation.
26. Elliott SL, Suhrbier A, Miles JJ, Lawrence G,             J Immunol 183(12):7949–7957
    Pye SJ, Le TT et al (2008) Phase I trial of a         38. Vollers SS, Stern LJ (2008) Class II major his-
    CD8+ T-cell peptide epitope-based vaccine for             tocompatibility complex tetramer staining:
    infectious mononucleosis. J Virol 82(3):                  progress, problems, and prospects. Immunology
    1448–1457                                                 123(3):305–313
27. Scheibenbogen C, Romero P, Rivoltini L, Herr          39. Raddassi K, Kent SC, Yang J, Bourcier K,
    W, Schmittel A, Cerottini JC et al (2000)                 Bradshaw EM, Seyfert-Margolis V et al (2011)
    Quantitation of antigen-reactive T cells in               Increased frequencies of myelin oligodendro-
    peripheral blood by IFNgamma-ELISPOT                      cyte glycoprotein/MHC class II-binding
    assay and chromium-release assay: a four-centre           CD4 cells in patients with multiple sclerosis. J
    comparative trial. J Immunol Methods 244                  Immunol 187(2):1039–1046
    (1–2):81–89                                           40. Laughlin E, Burke G, Pugliese A, Falk B,
28. Pelfrey CM, Rudick RA, Cotleur AC, Lee JC,                Nepom G (2008) Recurrence of autoreactive
    Tary-Lehmann M, Lehmann PV (2000)                         antigen-specific CD4+ T cells in autoimmune
    Quantification of self-recognition in multiple            diabetes after pancreas transplantation. Clin
    sclerosis by single-cell analysis of cytokine pro-        Immunol 128(1):23–30
    duction. J Immunol 165(3):1641–1651                   41. Bonvalet M, Wambre E, Moussu H, Horiot S,
29. Bestard O, Nickel P, Cruzado JM,                          Kwok WW, Louise A et al (2011) Comparison
    Schoenemann C, Boenisch O, Sefrin A et al                 between major histocompatibility complex
    (2008) Circulating alloreactive T cells corre-            class II tetramer staining and surface expres-
    late with graft function in longstanding renal            sion of activation markers for the detection of
    transplant recipients. J Am Soc Nephrol 19(7):            allergen-specific CD4(+) T cells. Clin Exp
    1419–1429                                                 Allergy 41(6):821–829
                                  18   Detection and Characterisation of Alloreactive T Cells              337
42. Ayyoub M, Dojcinovic D, Pignon P, Raimbaud             cally infected adults and children. J Exp Med
    I, Schmidt J, Luescher I et al (2010) Monitoring       192(12):1819–1832
    of NY-ESO-1 specific CD4+ T cells using            53. Asemissen AM, Nagorsen D, Keilholz U,
    molecularly defined MHC class II/His-tag-              Letsch A, Schmittel A, Thiel E et al (2001)
    peptide tetramers. Proc Natl Acad Sci USA              Flow cytometric determination of intracellular
    107(16):7437–7442                                      or secreted IFNgamma for the quantification of
43. Cusick MF, Yang M, Gill JC, Eckels DD (2011)           antigen reactive T cells. J Immunol Methods
    Naturally occurring CD4+ T-cell epitope vari-          251(1–2):101–108
    ants act as altered peptide ligands leading to     54. Whiteside TL, Zhao Y, Tsukishiro T, Elder EM,
    impaired helper T-cell responses in hepatitis C        Gooding W, Baar J (2003) Enzyme-linked
    virus       infection.      Hum        Immunol         immunospot, cytokine flow cytometry, and
    72(5):379–385                                          tetramers in the detection of T-cell responses to
44. Sun Y, Iglesias E, Samri A, Kamkamidze G,              a dendritic cell-based multipeptide vaccine in
    Decoville T, Carcelain G et al (2003) A system-        patients with melanoma. Clin Cancer Res
    atic comparison of methods to measure HIV-1            9(2):641–649
    specific CD8 T cells. J Immunol Methods            55. Koehne G, Smith KM, Ferguson TL, Williams
    272(1–2):23–34                                         RY, Heller G, Pamer EG et al (2002)
45. McMichael AJ, Ogg G, Wilson J, Callan M,               Quantitation, selection, and functional charac-
    Hambleton S, Appay V et al (2000) Memory               terization of Epstein–Barr virus-specific and
    CD8+ T cells in HIV infection. Philos Trans R          alloreactive T cells detected by intracellular
    Soc Lond B Biol Sci 2355(1395):363–367                 interferon-gamma production and growth of
46. Ogg GS, Jin X, Bonhoeffer S, Dunbar PR,                cytotoxic precursors. Blood 99(5):1730–1740
    Nowak MA, Monard S et al (1998) Quantitation       56. Waldrop SL, Pitcher CJ, Peterson DM, Maino
    of HIV-1-specific cytotoxic T lymphocytes and          VC, Picker LJ (1997) Determination of
    plasma load of viral RNA. Science                      antigen-specific memory/effector CD4+ T cell
    279(5359):2103–2106                                    frequencies by flow cytometry: evidence for a
47. Callan MF, Tan L, Annels N, Ogg GS, Wilson             novel, antigen-specific homeostatic mechanism
    JD, O’Callaghan CA et al (1998) Direct visual-         in HIV-associated immunodeficiency. J Clin
    ization of antigen-specific CD8+ T cells during        Invest 99(7):1739–1750
    the primary immune response to Epstein–Barr        57. Mifsud NA, Purcell AW, Chen W, Holdsworth
    virus In vivo. J Exp Med 187(9):1395–1402              R, Tait BD, McCluskey J (2008) Immu-
48. Mifsud NA, Nguyen TH, Tait BD, Kotsimbos               nodominance hierarchies and gender bias in
    TC (2010) Quantitative and functional diversity        direct T(CD8)-cell alloreactivity. Am J
    of cross-reactive EBV-specific CD8+ T cells in a       Transplant 8(1):121–132
    longitudinal study cohort of lung transplant       58. Allen KJ, Mifsud NA, Williamson R, Bertolino
    recipients. Transplantation 90(12):1439–1449           P, Hardikar W (2008) Cell-mediated rejection
49. Nguyen TH, Sullivan LC, Kotsimbos TC,                  results in allograft loss after liver cell transplan-
    Schwarer AP, Mifsud NA (2010) Cross-                   tation. Liver Transpl 14(5):688–694
    presentation of HCMV chimeric protein enables      59. Elkington R, Walker S, Crough T, Menzies M,
    generation and measurement of polyclonal T             Tellam J, Bharadwaj M et al (2003) Ex vivo
    cells. Immunol Cell Biol 88(6):676–684                 profiling of CD8+-T-cell responses to human
50. Pala P, Hussell T, Openshaw PJ (2000) Flow             cytomegalovirus reveals broad and multispecific
    cytometric measurement of intracellular cytok-         reactivities in healthy virus carriers. J Virol
    ines. J Immunol Methods 243(1–2):107–124               77(9):5226–5240
51. Prussin C, Metcalfe DD (1995) Detection of         60. Taswell C (1981) Limiting dilution assays for
    intracytoplasmic cytokine using flow cytometry         the determination of immunocompetent cell
    and directly conjugated anti-cytokine antibod-         frequencies. I. Data analysis. J Immunol
    ies. J Immunol Methods 188(1):117–128                  126(4):1614–1619
52. Goulder PJ, Tang Y, Brander C, Betts MR,           61. Strijbosch LW, Buurman WA, Does RJ, Zinken
    Altfeld M, Annamalai K et al (2000)                    PH, Groenewegen G (1987) Limiting dilution
    Functionally inert HIV-specific cytotoxic T            assays. Experimental design and statistical anal-
    lymphocytes do not play a major role in chroni-        ysis. J Immunol Methods 97(1):133–140
                                                                                        Chapter 19
Abstract
The ability to directly measure virus-specific lymphocytes using fluorochrome-labeled tetrameric complexes
has proven a great advancement for the transplantation field. Viral peptide/HLA tetrameric complexes allow
the rapid generation of virus-specific clones using single cell sorting apparatus, permitting the determination
of alloreactivity from a single TCR with known specificity. When combined with new target “detector” cells
called single HLA antigen-transfected K562 cells (SALs), the human alloresponse can for the first time be
examined specifically and reliably. Here we describe a method for detection of “heterologous immunity”
from virus-specific memory T-cells using single HLA expressing cell lines as allogeneic targets.
Key words: Heterologous immunity, Memory T-cells, Viral immunity, Alloreactivity, SALs
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_19, © Springer Science+Business Media New York 2012
                                                         339
340      L.J. D’Orsogna et al.
2. Materials
2.1. Virus-specific        1. Viral peptide/HLA tetrameric complex of interest (see Note 1).
T-Cell Cloning Using       2. PBMCs from donor known to have viral peptide/HLA
Single Cell Sorting           tetramer complex binding T-cells in the peripheral blood (or at
                              least known to be serologically positive for virus of interest)
                              (see Notes 2–3).
                           3. Iscoves Modified Dulbecco Medium (IMDM) with 1%
                              l-glutamine.
                           4. Fetal Calf Serum (FCS) made to 1 and 10% solutions in
                              IMDM.
                           5. Human Serum (HS) made to 5% HS and 5% FCS in IMDM.
                           6. Phytohaemagglutinin (PHA) 800 ng/mL.
                           7. Interleukin-2 (IL-2).
                           8. Freshly collected human PBMCs to be used as feeder cells
                              (see Note 4).
                                          19   Detection of Allo-HLA Cross-Reactivity...   341
3. Methods
3.1. Virus-specific CD8     1. Perform all steps at 4°C (see Note 8).
T-Cell Cloning Using        2. Ensure cell donor for single cell sorting is serologically positive
Single Cell Sorting            for the virus and has a population of the relevant tetramer
                               binding T-cells (see Notes 2 and 9).
                            3. Count the ficoll hypaque separated PBMCs ensuring you have
                               at least 1 × 106 cells. Wash twice in medium containing 1%
                               FCS/IMDM by spinning down the cells at 700 G for 8 min.
                            4. After second wash, resuspend the pellet in the residual medium
                               and transfer to a FACS tube (see Note 9).
                            5. Add 1 μL tetramer-PE to the FACS tube on ice (see Note 1).
                            6. Add markers CD45Ra/CD4/CD14-FITC (see Note 10).
                 19   Detection of Allo-HLA Cross-Reactivity...   343
3.2. Generation            1. Culture K562 until you have 10 × 106 cells per transfection in
of Single HLA-                IMDM supplemented with 10% FCS and penicillin/streptomycin.
Transfected K562 Cells     2. Spin down the cells and wash two times in IMDM/10%FCS.
Lines (SALs)
                           3. Count the cells and resuspend the cells in IMDM/10%FCS at
                              a concentration of 10 × 106 cells per mL.
                           4. Mix 1 mL of the cells with 10 μg of the plasmid DNA contain-
                              ing the HLA-cDNA construct of the HLA specificity
                              required.
                           5. Put 1 mL of the cell/DNA mixture in a 0.4-cm cuvette and
                              store on ice for a minimum of 5 min.
                           6. Mix and pulse (960 μF, 230 V).
                           7. Store on ice for 15 min.
                           8. Add a few drops of IMDM/10%FCS/pen/strep medium to
                              the cuvette and transfer the cells to a culture flask containing
                              9 mL of the same medium.
                           9. Culture the cells for 2 days at 37°C.
                          10. Add selection antibiotic at the correct final concentration (see
                              manufacturers protocol) (see Note 5).
                          11. Culture for another week at 37°C. If culture flasks become full
                              then split flasks and add new medium.
                          12. Perform a flow cytometry test to see whether there are HLA-
                              positive cells (incubate 105 cells with w6/32-PE for 30 min on
                              ice, wash twice, take up in 100 μL PBS/1% paraformaldehyde,
                              read in FACS calibur and analyze) (see Note 17).
                          13. If positive cells are present, separate positive cells from nega-
                              tive cells (described in steps 14–31) (see Notes 18 and 19).
                          14. Take 20–25 mL of your cultured cells (leave some in the flask
                              and continue culturing them).
                          15. Spin down, remove supernatant, and resuspend the cell pellet.
                          16. Add 0.5 mL unlabeled-w6/32 antibody and vortex.
                          17. Incubate on ice for 30 min.
                          18. Wash three times with ice cold medium (IMDM/10%FCS) in
                              a 15-mL tube.
                          19. Resuspend the cells and add 30 μL Sheep anti-Mouse dynabeads.
                          20. Incubate on a rollerbench in 4°C room for 30 min.
                          21. Put the tube in the magnet, add 10 mL ice cold medium.
                          22. Wait for 5 min.
                          23. Discard the medium with the nonbound cells.
                          24. Repeat from step 21 twice.
                          25. Add 10 mL ice cold medium and resuspend the cells bound to
                              the beads, put them in a small culture flask, and culture at 37°C.
                                         19    Detection of Allo-HLA Cross-Reactivity...   345
                        26. Wait for the cells to grow and expand. Check daily (see Notes
                            19 and 20).
                        27. When the cells have expanded, remove the beads as follows.
                        28. Put the cells in a 15-mL tube within the dynal magnet.
                        29. Wait for 5 min.
                        30. Collect the medium with the cells into a culture flask leaving
                            the beads in the tube.
                        31. Add 10 mL of fresh medium (10%FCS/IMDM) to the tube.
                        32. Repeat twice from step 28 and on the last removal of beads
                            transfer the cells to a culture flask, add 10% of fresh culture
                            medium.
                        33. Culture at 37oC.
                        34. After a few days growth, test the class I expression of the cells
                            using HLA-specific monoclonal antibodies and FACS analysis
                            (step 12).
                        35. If there still are cells with no expression at all, repeat the bead
                            sorting (repeat from step 14 onwards).
                        36. If the cells all have a good expression, they can be used in the
                            IFNγ assay (see Subheading 3.3) or cytotoxicity assay (see
                            Subheading 3.4). Alternatively the cells can be frozen in sev-
                            eral aliquots for subsequent use in these assays.
                        37. Test the cells with a flow cytometry test against a panel of anti-
                            HLA monoclonal antibodies to confirm the HLA expression
                            of the K562 cell (see Note 17).
3.3. IFN g ELISA         1. Spin down the virus-specific T-cell clone (generated using
Using Responder             method in Subheading 3.1), resuspend and count (see Notes
Virus-specific T-Cell       21 and 22).
Clones and SAL           2. Make a T-cell clone solution of 0.1 × 106 T-cells/mL in T-cell
Stimulator Cells            medium (see Subheading 3.1, step 10).
                         3. Dilute the SALs and the K562 line in T-cell medium to a con-
                            centration of 0.25 × 106 cells/mL.
                         4. Add 50 μL/well of the T-cell clone solution to a sterile 96-well
                            plate.
                         5. Add 100 μL/well of the different SALs in duplicate to the
                            T-cell clone. Include:
                            (a) A SAL transfected with the restricting HLA molecule
                                loaded with the viral peptide that is recognized by the
                                clone as a positive control. To load peptides onto SALs
                                first incubate harvested cell pellet with 100 μg/mL of
                                peptide for 30 min at room temperature, then wash twice
                                and resuspend to required concentration of 0.25 × 106
                                cells/mL.
346       L.J. D’Orsogna et al.
3.4. Cytotoxicity Assay     1. Spin down the SALs and the nontransferred K562 cell line (see
Using Effector                 Subheading 3.2, step 36) direct from the culture medium or
Virus-specific T-Cell          following thawing. Do not resuspend. The cell pellets must
Clones and SAL                 first be labeled with chromium (see Note 28). Include:
Target Cells                      (a) A SAL transfected with the restricting HLA molecule
                                      loaded with the viral peptide that is recognized by the
                                      clone as a positive control (see Notes 23–25).
                                  (b) A SAL transfected with the restricting HLA molecule
                                      loaded with the control peptide that is not recognized by
                                      the clone as a negative control.
                                  (c) A nontransfected K562 as a negative control.
                            2. Add the appropriate amount of sodium chromate to the cell
                               pellets (see Note 29)
                            3. Incubate the cells in a 37°C water bath for 1 h.
                            4. Meanwhile spin down, resuspend, and count the T-cell clone
                               and make the necessary dilutions of the clone (see Note 30).
                               Add 100 μL/well to a 96-well plate.
                            5. Wash the chromium-labeled SAL and K562 cells three times
                               with 4 mL IMDM + 1%FCS.
                            6. Add 100 μL of the labeled target cells (concentration of 5,000
                               cells/100 μL) to each of the wells already containing the T-cell
                               clone.
                            7. Make a control plate for spontaneous and maximum release.
                               For each target make 3 wells with 100 μL TCM and 3 wells
                               100 μL 1% Triton X-100. Add 100 μL target suspension per
                               well.
                            8. Spin the plates for 1 min at 550 × g.
                            9. Incubate the plates for 4 h at 37°C, 5%CO2.
                           10. Spin the plates for 1 min at 550 × g.
                           11. Harvest the supernatants.
                            19   Detection of Allo-HLA Cross-Reactivity...   347
           12. Measure the chromium release and calculate the specific lysis
               (see Note 31).
4. Notes
                            26. For easy transfer of your supernatants, make the layout of your
                                culture plate the same as the layout of the ELISA plate and
                                leave wells open for your standard dilutions and blank.
                            27. When the supernatants have to be stored for a longer period
                                (>1 month), it is better to store them in small siliconized
                                vials.
                            28. Cytotoxicity assays are best performed at day 7–8 after stimula-
                                tion of the T-cell clone.
                            29. The half life off sodium chromate is short, be sure to use the
                                correct amount according to the manufacturers instructions.
                            30. The effector target ratios commonly used are 30:1, 10:1, 1:1,
                                and 0.1:1. Therefore, given that there are 5,000 target cells/
                                well then the number of effector cells to be added are 150,000,
                                50,000, 5,000, and 500 (each in 100 μL), i.e., cell concentra-
                                tions 1.5 × 106, 0.5 × 106, 0.05 × 106, and 0.005 × 106/mL
                                respectively.
                            31. % specific lysis = ((test release–spontaneous release) (maximum
                                release–spontaneous release)) × 100%. For more details on cal-
                                culations of % specific lysis, see ref. (2).
References
1. Amir A, D’Orsogna L, Roelen D, van Loenen                 HLA-specific antibodies. Hum Immunol 66:
   M, Haagedoorn R, de Boer R et al (2010) Allo-             519–525
   HLA reactivity of viral specific memory T-cells        4. Mulder A, Zoet Y, Eijsink C. Single MHC anti-
   is common. Blood 115:3146–3157                            gen expressing cell lines for the definition of
2. D’Orsogna LJA, Amir A, Zoet Y, van der Meer-              monoclonal antibody specificities. In: Hansen
   Prins P, van der Slik A, Kester M et al (2009)            JA(ed) Histocompatibility testing 2002, HLA
   New tools to monitor the impact of viral infec-           2002, Munksgaard, 2005
   tion on the alloreactive T-cell repertoire. Tissue     5. van der Stoep N, Biesta P, Quinten E, van den
   Antigens 74:290–297                                       Elsen P (2002) Lack of IFNγ mediated induc-
3. Zoet Y, Eijsink C, Kardol M, Franke-van Dijk              tion of the class II transactivator (CIITA)
   M, Wilson G, de Paus R et al (2005) The                   through promoter methylation is predomi-
   single antigen expressing lines (SALs) con-               nantly found in developmental tumor cell Lines.
   cept: an excellent tool for screening for                 Int J Cancer 97:501–507
                                                                                        Chapter 20
Abstract
Spleen and lymph node retrieved post-mortem from deceased organ donors are a rich source of lympho-
cytes. Storage of lymphocytes separated from these sources can be valuable where post-transplant testing
(crossmatching) is required. DNA extraction from stored lymphocytes also allows further genetic testing
where required, for example additional HLA typing not performed at the time of transplant for donor-
specific antibody monitoring. Methods for the isolation and freezing of such cells is described.
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_20, © Springer Science+Business Media New York 2012
                                                         351
352       G. Tassone and S.J. Fidler
2. Materials
2.1. Isolation of Spleen    1. RPMI medium 1640 (Gibco Invitrogen) (see Note 1).
and Lymph Node Cells        2. Sections of fresh lymph node or spleen (see Note 2).
                            3. Ficoll-Paque (GE Healthcare Bioscience) (see Note 1).
                            4. Sterile Petri dishes (Sarstedt) (see Note 1).
                            5. Disposable sterile scalpels (Swann- Morton).
                            6. 30-mL round bottom capped tubes (Sarstedt).
                            7. 30-mL syringes (Becton Dickinson).
                            8. Cannulae (Maerski Medical).
                            9. Size 7 and/or size 9 sterile plastic pasteur pipettes (Samco
                               Scientific).
                           10. Centrifuge (Beckman TJ-6 or equivalent).
                           11. 50-mL Cellstar tubes (Greiner).
                           12. 20% Foetal calf serum (FCS)/RPMI (see Note 3).
3. Methods
3.1. Isolation of Spleen    1. Place an approximate 2 cm3 section of spleen onto a sterile
and Lymph Node Cells           Petri dish and using a sterile scalpel, trim the capsule from the
                               spleen and discard.
                            2. In a separate Petri dish, place the lymph nodes and remove any
                               fatty tissue from around the nodes.
     20   Separation and Cryopreservation of Lymphocytes from Spleen and Lymph Node         353
4. Notes
Fig. 1. The Neubauer chamber, indicating sample introduction point, and counting chamber.
                                 7. This centrifugation will spin down any tissue debris but is short
                                    and gentle enough to leave the cells in suspension. This step
                                    can be repeated if necessary.
                                 8. Remove the syringe plunger from the barrel and discard. Place
                                    a cannula on the end (where a hypodermic needle is usually
                                    placed) and carefully place the cannula into the cell suspension
                                    so the end sits on the bottom of the tube. Pour 10 mL of den-
                                    sity gradient into the syringe barrel and lift the syringe slightly
                                    to allow flow of gradient.
                                 9. If the tubes are accidently knocked or dropped resulting in
                                    mixing of the gradient/cell suspension interface, the samples
                                    can be recovered by underlaying the gradient/cell mix with a
                                    further 10 mL of density gradient.
                               10. Cell concentration determination using a Neubauer chamber
                                   (see Fig. 1):
                                    ●     Clean lens thoroughly prior to use.
                                    Ensure cover slip is also thoroughly cleaned.
                                    ●     Place cover slip over the counting surface. Add the cell
                                          suspension using a pipette. (The area under the cover slip
                                          fills by capillary action).
                                    ●     Place the counting chamber on the microscope stage and
                                          bring the counting grid into focus at low power (40×).
                                    ●     There are 9 large squares, each of which contains 16
                                          medium squares, each of which contains 25 small squares.
                                          One large square has 0.1 μL volume (see Fig. 2).
                                    ●     Count the number of lymphocytes in 5 of the medium
                                          squares and multiply by 5 (=number of cells/0.1 μL).
356   G. Tassone and S.J. Fidler
                      Fig. 2. Diagrammatic representation of the Neubauer counting chamber. One large square
                      have 0.1 μL volume.
References
Crossmatching by Complement-Dependent
Lymphocytotoxicity
Samantha J. Fidler
Abstract
The presence of preformed donor-specific HLA antibodies detected by Complement-dependent cyto-
toxicity (CDC) crossmatch assay is associated with a high incidence of hyperacute or accelerated rejection
and remains one of the gold standard tests pre-transplant. The standard CDC crossmatch detects IgG1,
IgG3, and IgM antibody, i.e. complement fixing, bound to the native viable cell surface of lymphocytes.
The crossmatch can be enhanced with the addition of anti-human-globulin to detect non-complement
fixing antibodies (IgG2 and IgG4), and sensitivity can be improved with prolonged incubation times.
1. Introduction
                                Since the landmark paper by Patel and Terasaki (1) was published
                                in the late 1960s, a positive (T cell) lymphocytotoxic crossmatch
                                has been considered a contraindication to renal transplantation.
                                The crossmatch assay detects high levels of recipient antibodies
                                against donor HLA antigens, and a positive crossmatch is associ-
                                ated with hyperacute and accelerated acute rejection, and long-
                                term graft failure. The complement-dependent lymphocytotoxicity
                                (CDC) crossmatch remains to this day the gold standard as the
                                final pre-transplant check for donor-specific anti-HLA antibodies.
                                Since the 1960s a number of modifications, such as the AHG-
                                CDC crossmatch and treatment of sera to remove IgM reactivity,
                                have been made to improve sensitivity and specificity. The barrier
                                of a positive crossmatch to transplantation has somewhat dimin-
                                ished in the current era of desensitisation protocols and effective
                                treatment for rejection episodes. However, it is clear that the
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_21, © Springer Science+Business Media New York 2012
                                                         359
360       S.J. Fidler
2. Materials
2.2. Standard CDC           1. 1 and 5-μL single dispense pipettes (e.g., Hamilton, Robbins).
Crossmatch                  2. 60- or 72-well microlymphocytotoxicity (Terasaki) trays
                               (containing 5 μL mineral oil per well).
                            3. Recipient sera (see Notes 7 and 8).
                            4. Donor lymphocytes—refer to Subheading 2.1.
                            5. Fluorescent stain     (Haemoglobin/ethidium      bromide    or
                               alternative).
                            6. Pooled rabbit complement—refer to Subheading               3.5:
                               Evaluation of complement—(PelFreeze) (see Note 9).
                            7. Citrated PBS pH 7.4.
                            8. PBS pH 7.4.
                            9. AB Negative serum (see Note 10)—negative control.
                           10. Anti-thymocyte globulin (ATG) (see Note 11)—positive
                               control.
                           11. B cell positive control (see Note 12).
                           12. Inverted fluorescence microscope.
                           13. 37°C waterbath.
                           14. 22°C incubator (see Note 6).
                           15. 1- and 5-μL automatic dispensers (e.g. Greiner, OLI, Robbins).
                           16. 1% Kaolin (Sigma).
3. Methods
3.1. Separation of T     1. Into a 15-mL round bottomed tube add 4 mL ACD blood and
and B Lymphocytes           4 mL of 1× RBC lysis buffer. Mix by gently inverting.
                         2. Add 200 μL of positive selection cocktail (Human B cell WB
                            positive selection for B cells or Human CD3 WB positive selec-
                            tion for T cells) to the whole blood/lysis mixture. Incubate at
                            room temperature for 15 min (see Note 6).
                         3. Mix the EasySep beads to ensure uniform suspension by
                            pipetting up and down vigorously at least five times (DO NOT
                            VORTEX).
                         4. Add 200 μL of the mixed EasySep beads to the respective
                            tubes. Incubate for 10 min at room temperature.
                         5. Add 2 mL PBS pH 7.4 and mix gently by pipetting up and
                            down.
                         6. Remove cap from tube and place on EasySep magnet. T cells
                            require 5 min, B cells require 10 min on the magnet.
                         7. Pick up magnet and in one continuous motion pour contents
                            out of the 15-mL tube, leave inverted for 2–3 s (DO NOT
                            SHAKE OR BLOT ANY DROPS REMAINING ON THE
                            MOUTH OF THE TUBE).
                         8. Remove tube from magnet and add 10 mL PBS pH 7.4. Place
                            on magnet for another 5 min (both T and B cells).
                         9. Pick up magnet and in one continuous motion pour contents
                            out leave inverted for 2–3 s as per step 7 (DO NOT SHAKE
                            OR BLOT ANY DROPS REMAINING ON THE MOUTH
                            OF THE TUBE).
                        10. Remove the 15-mL tube from magnet add 0.5 mL CFDA
                            solution drop wise directly onto the cells in the 15-mL tube and
                            incubate for 5 min at room temperature (mix the tube gently by
                            hand from time to time during this period).
                        11. Place the 15-mL tube into the magnet and fill the tube to top
                            of magnet with PBS pH 7.4. Incubate for 2 min.
                        12. Pick up magnet and in one continuous motion pour contents
                            out leave inverted for 2–3 s (DO NOT SHAKE OR BLOT
                            ANY DROPS REMAINING ON THE MOUTH OF THE
                            TUBE).
                        13. Fill CTS tube to top of magnet with PBS pH 7.4 for 2 min.
                           21   Crossmatching by Complement-Dependent Lymphocytotoxicity            363
3.2. Standard CDC                1. Add 1 μL of ATG dilutions and AB negative sera to an oiled
Crossmatch                          Terasaki tray using a Hamilton syringe (see Note 18, Fig. 1).
                                    3A—ATG50, 3B—AB Neg, 3C—ATG500, 3D—ATG200,
3.2.1. Tray Set-Up
                                    3E—AB Neg, 3F—B Cell Pos, 4A—ATG400, 4B—ATG25,
                                    4C—AB Neg, 4D—ATG100, 4E—ATG800, 4F—ATG1000.
                                 2. Add 1 μL recipient serum to wells 1D, 1E, and 1F and 1 μL
                                    AB negative serum to wells 1A, 1B, and 1C using a Hamilton
                                    syringe. Historical peak sera can be added as appropriate in
                                    triplicate to wells 2A to 2F (see Note 8). Trays should be made
                                    at least in duplicate for both T and B cell crossmatch, i.e. at
                                    least four trays in total (see Note 19).
                               Table 1
                               Cytotoxicity scores
                               0                           A       1
                               1–2                         B       1
                               3–5                         C       1
                               6–10                        D       1
                               11–15                       1       1
                               15–25                       2       1
                               25–35                       3       2
                               35–45                       4       2/4
                               45–55                       5       4
                               55–65                       6       4/6
                               65–75                       7       6
                               75–85                       8       8
                               85–100                      9       8
                               Empty well or unreadable    X       0
                               Insufficient cell numbers   0       0
Fig. 2. (a) Autologous crossmatch (The circle denotes B cell crossmatch results, the triangle denotes T cell crossmatch
results). Crossmatches are read according to Table 1. The AB serum is read as a background control, i.e. contains no HLA
antibodies. The autologous crossmatch should be interpreted in relation to the crossmatch results of this control. The AB
Neg is read as a carry-over control—this AB serum is located next to strongly positive ATG controls. The ATG controls can
be used to monitor the sensitivity of the assay. The laboratory should determine its own QC criteria for example an assay
might only pass QC if the ATG 400 dilution crossmatch result is >AB serum control. In this example, the autologous T and
B cell crossmatches are negative. (b) Allogeneic crossmatch (the circle denotes B cell crossmatch results, the triangle
denotes T cell crossmatch results). Crossmatches are read according to Table 2. The AB serum is read as a background
control, i.e. contains no HLA antibodies. The allogeneic crossmatch should be interpreted in relation to the crossmatch
results of this control. Other controls on this crossmatch tray follow the same criteria as those in the autologous cross-
match (a). In this example, both the T and B cell allogeneic crossmatches are positive.
366        S.J. Fidler
Table 2
Interpretation of CDC crossmatch results
Table 3
Interpretation of T cell crossmatch results taking account of the presence or absence
of donor-specific antibodies (DSA)
Table 3
(continued)
Table 4
Interpretation of B cell crossmatch results taking account of the presence
or absence of DSA
 Table 4
 (continued)
3.3.2. Crossmatch Method          1. Follow Subheading 3.2.2 to step 3 (see Note 27).
                                  2. Wash trays with RPMI. Add 10 μL RPMI to each well.
                                     Centrifuge for 1 min at 1,000 rpm. Quickly flick out the RPMI
                                     in one swift movement.
                                  3. Repeat step 2 two more times, then add 1 μL AHG to each
                                     well. Leave for 1 min.
                                  4. Follow Subheading 3.2.2 from step 4 until the end.
                             21   Crossmatching by Complement-Dependent Lymphocytotoxicity         369
3.4.1. Preparation of Sera         1. Pre-warm the waterbath to 60°C (see Note 29).
                                   2. Aliquot 200 μL of patient crossmatch sera and control sera
                                      (AB Neg, IgG, and IgM controls) into suitably labelled
                                      Beckman tubes.
                                   3. Place the tubes into the waterbath ensuring that the sera are
                                      fully submerged (see Note 30).
                                   4. Incubate the tubes for exactly 13 min (see Note 31).
                                   5. Remove the samples from the waterbath and immediately
                                      microfuge at 10,000 rpm for 1 min.
                                   6. Remove the supernatant into a clean Beckman tube, taking
                                      care not to disturb the gelatinous protein precipitate.
3.4.2. Tray Set-Up                 1. Add ATG dilutions as per step 1 of Subheading 3.2.1.
                                   2. Add 1 μL AB Negative serum to wells 1A and 1B and 1 μL
                                      heat treated (HT) AB negative serum to wells 1C and 1D using
                                      a Hamilton syringe.
                                   3. Add 1 μL IgG control serum to wells 1E and 1F and 1 μL HT
                                      IgG control serum to wells 2E and 2F.
                                   4. Add 1 μL IgM control serum to wells 2C and 2D and 1 μL
                                      HT IgM control serum to wells 2A and 2B.
                                   5. Add 1 μL patient serum to wells 5A and 5B and 1 μL HT
                                      patient serum to wells 5C and 5D. Historical peak sera can be
                                      added as appropriate in duplicate to wells 5E to 6F (see Note 8).
                                      Trays should be made in duplicate for both T and B cell cross-
                                      match, i.e. four trays in total.
3.4.3. Crossmatch Method          The standard CDC crossmatch as described in Subheading 3.2.2,
                                  or the AHG crossmatch as described in Subheading 3.3.2 may be
                                  performed.
Table 5
Interpretation of heat-treated T cell crossmatch results
Table 6
Interpretation of heat-treated T cell crossmatch results
3.5. Complement                 Pooled rabbit serum is used as a source of complement (see Note 9).
Evaluation
3.5.1. Tray Set-Up               1. Serially dilute the complement N/2 with McCoys medium
                                    (see Note 32).
                                 2. Serially dilute antisera with well-characterised reactivity N/2
                                    in AB negative serum (see Note 33).
                                 3. Add 1 μL of serum to a Terasaki tray from Neat to 1/512 (see
                                    Fig. 3).
3.5.2. Crossmatch Set-Up         1. Add 1 μL T or B cell suspension (refer to Subheading 3.1) using
                                    an automated cell dispenser to avoid cross-contamination (see
                                    Note 34). Ensure all wells are thoroughly mixed (see Note 20).
                                 2. Add 5 μL of diluted complement according to the format in
                                    step 3. Incubate at room temperature for 45 min (see Note 21).
                                 3. Add 5 μL ethidium bromide/haemoglobin stain and allow to
                                    rest for 20 min (see Note 3).
                                 4. Score the percentage cell death under the fluorescent micro-
                                    scope according to Table 1 (see Notes 22 and 23).
Fig. 4. (a) Autologous crossmatch (the circle denotes B cell crossmatch results, the triangle denotes T cell crossmatch
results). Crossmatches are read according to Table 1. The AB serum is read as a background control, i.e. contains no
372         S.J. Fidler
Fig. 4. (continued) HLA antibodies. The autologous crossmatch should be interpreted in relation to the crossmatch results
of this control. The IgG control should not reduce with heat treatment (HT). Reduction may be due to over-incubation of
serum or incubation at the incorrect temperature (>63°C) leading to denaturation of IgG antibody. The IgM control should
reduce with HT. Failure to reduce may be due to under-incubation of serum or incubation at the incorrect temperature
(<60°C). In this example, the autologous T and B cell crossmatches are positive, and reduce with HT indicating the pres-
ence of IgM autoantibody. (b) Allogeneic crossmatch (the circle denotes B cell crossmatch results, the triangle denotes T
cell crossmatch results). Crossmatches are read according to Table 2. The AB serum is read as a background control, i.e.
contains no HLA antibodies. The allogeneic crossmatch should be interpreted in relation to the crossmatch results of this
control. The IgG control and IgM controls follow the same criteria as in the autologous crossmatch (a). In this example,
the allogeneic T and B cell crossmatches are positive, and reduce with HT consistent the presence of IgM autoantibody.
Fig. 5. (a) Example of crossmatch results of a homozygous cell against neat antiserum where the circle is the current comple-
ment, the triangle is evaluation complement 1, the rectangle is evaluation complement 2 and the star is evaluation complement
                          21   Crossmatching by Complement-Dependent Lymphocytotoxicity                            373
Fig. 5. (continued) 3. Evaluation complement 2 has low reactivity and should be discarded. Complement 1 appears to have
reactivity similar to the current complement. Complement 3 has increased reactivity, this may be due to high background
or stronger complement activity (see (b)). (b) Example of crossmatch results of a negative cell against neat antiserum
where the circle is the current complement, the triangle is evaluation complement 1, the rectangle is evaluation comple-
ment 2 and the star is evaluation complement 3. Complement 5 has high reactivity and should be discarded. Complement
1 appears to have consistent reactivity with the current complement.
4. Notes
                           29. The precise temperature is critical for this assay. We prefer to use
                               a waterbath as the temperature can be constantly monitored
                               and adjusted as necessary.
                           30. Place tubes in polystyrene floats so that the serum is fully sub-
                               merged in the waterbath. These floats can be easily made by
                               cutting the bottom from polystyrene cups and making small
                               holes in which the tubes are held snugly.
                           31. The precise timing and incubation temperature should be vali-
                               dated in your laboratory to ensure IgM antibody is reduced
                               and IgG reactivity is not affected.
                           32. Perform the evaluation in parallel with the current batch of
                               complement. It is reasonable to request evaluation samples
                               from several different commercial companies for comparison.
                           33. Well-characterised class I and class II antisera previously tested
                               by CDC crossmatch should be selected. Monospecific sera, or
                               sera against well-defined epitopes present in several antigens
                               (e.g. Bw4 or Bw6) are ideal for this evaluation.
                           34. A number of cells should be chosen with the following charac-
                               teristics: (1) homozygous for the antigen against which the
                               antiserum is reactive; (2) heterozygous for the antigen against
                               which the antiserum is reactive; and (3) negative for the antigen
                               against which the antiserum is reactive.
References
Abstract
The flow cytometric lymphocyte crossmatch is a standard technique for evaluating the compatibility of
potential kidney transplant recipients and donors. Recipient serum is incubated with donor lymphocytes
and the latter are analysed in a flow cytometer for the presence of bound IgG antibodies. An increase in
the level of IgG binding compared to a negative control indicates the presence of donor-specific antibodies
which may lead to deleterious graft function. Described here is a method to perform T and B lymphocyte
crossmatching in the same tube to detect IgG donor-reactive antibodies.
     Key words: Flow cytometry, Lymphocyte crossmatch, Kidney transplantation, Human leukocyte
     antigen, Pronase treatment
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_22, © Springer Science+Business Media New York 2012
                                                         379
380     J. Downing
2. Materials
3. Methods
3.1. Isolation            Peripheral blood lymphocytes (PBL) are the main target of the
of Peripheral Blood       crossmatch test. Anti-coagulated blood samples should be stored
Mononuclear Cells         at room temperature and should be no older than 48 h before
                          isolating the lymphocytes, where possible.
                           1. Transfer 10 mL of anti-coagulated peripheral blood into two
                              labelled 50-mL centrifuge tubes.
                           2. Dilute with PBS to a total volume of 30 mL.
                           3. Underlay diluted blood with Ficoll-Paque™ Plus taking care
                              not mix the blood-Ficoll interface.
                           4. Centrifuge for 10 min at 1,600 × g.
                           5. Carefully aspirate the mononuclear cell layer at the Ficoll—
                              plasma interface from each 50-mL centrifuge tube into clean
                              labelled 15-mL centrifuge tubes and top up to 15 mL with
                              PBS.
                           6. Centrifuge for 3 min at 650 × g.
                           7. Decant the supernatant and re-suspend the cell button in
                              15 mL of PBS.
                           8. Centrifuge for 3 min at 650 × g.
                           9. Decant the supernatant.
                         10. Pool and re-suspend the cell buttons from the two 15-mL cen-
                             trifuge tubes into 1 mL of RPMI cell medium (see Note 6).
                         11. Adjust the mononuclear cell count to 8 × 106 cells/mL (see
                             Note 7).
382      J. Downing
3.2. Cleavage of Fc   T and B lymphocyte cell surface Fc receptors can bind IgG molecules
Receptors Using       non-specifically and lead to a high anti-IgG FITC measurement on
Pronase               the cell. This can lead to difficulty in discriminating between nega-
                      tive and positive crossmatch results. Cleavage of Fc receptors using
                      Pronase at can lead to a reduction in the incidence of false negative
                      crossmatch due to the presence of excess irrelevant IgG bound to
                      Fc receptors (15) (see Note 8).
                       1. Add 0.5 mL of 1 mg/mL Pronase to 1 mL of donor or recipient
                          cells.
                       2. Incubate for 30 min at 37°C.
                       3. After incubation top up to 15 mL with RPMI.
                       4. Centrifuge for 3 min at 650 × g.
                       5. Decant the supernatant and re-suspend the cell button in
                          0.5 mL RPMI.
                       6. Re-adjust the mononuclear cell count to 8 × 106 cells/mL.
3.3. Crossmatch       PBLs should be prepared from the donor sample for the allo-cross-
Procedure             match and from the patient sample where an auto-crossmatch test
                      is required (see Note 9).
                       1. Label 7× 3-mL flow cytometry tubes in the following sequence:
                          3 negative control serum vs. donor cells; 3 patient serum vs.
                          donor cells; 1 positive control serum vs. donor cells. Additionally,
                          if required, label a series of flow tubes for an auto control cross-
                          match to include a negative control serum vs. patient cells and
                          at least two replicates of patient serum vs. patient cells (see
                          Note 10).
                       2. In each tube, add 50 μL of appropriate serum to 50 μL of
                          appropriate cells, mix by gentle vortexing and incubate at room
                          temperature for 30 min.
                       3. After incubation, wash each tube by adding approximately
                          2 mL of cold PBS-azide (see Note 11) and centrifuging for 2 min
                          at 1,800 × g. Decant the supernatant and gently re-suspend the
                          cell button. Alternatively an automated cell washer can be
                          employed.
                       4. Repeat the wash process twice. After the third wash remove as
                          much supernatant as possible and gently re-suspend the cell
                          button (see Note 12).
                       5. Add 50 μL of diluted anti-human-IgG FITC to each tube, mix
                          by gentle vortexing, and incubate for 15 min in the dark at 5°C
                          (see Note 13).
                       6. Wash once as described in step 3 above.
                       7. To each tube add 2 μL each of anti-CD3 and anti-CD19
                          reagents (see Note 14). Mix by gentle vortexing and incubate
                          for 15 min in the dark at 5°C.
            22    The Lymphocyte Crossmatch by Flow Cytometry for Kidney Transplantation                              383
3.4. Flow Cytometric            The flow cytometer instrument must be set up correctly before
Analysis                        use. The manufacturer’s daily start-up and shut-down procedures
                                must be followed and a daily calibration using commercial stan-
                                dard reagents performed. In addition, optimisation (see Note 16)
                                and colour compensation must be performed (see Note 17).
                                  1. Collect data on 1,200 B lymphocyte events as follows, see
                                     Fig. 1 (see Note 18).
                                  2. Use the cytometer acquisition and analysis software to create a
                                     1,024 channel 4-decade log plot of forward scatter (FSC) vs.
                                     side scatter (SSC) (Fig. 1a).
                                  3. Draw a polygon gate around the lymphocyte population and
                                     use this gate to create a dot plot to show FL2 (PE) vs. FL3
                                     (PerCP) to discriminate the B and T lymphocytes, respectively
                                     (Fig. 1b) (see Note 19).
                                  4. Draw a rectangular gate around each of the lymphocyte popu-
                                     lations, and use these to plot an FL1 (FITC) vs. cell count
                                     histogram for each population (Fig. 1c, d). Draw a histogram
Fig. 1. Flow cytometer analysis histograms. Flow analysis histograms created using Becton Dickinson Cell Quest Pro analysis
software. Lymphocytes are displayed by plotting forward scatter (FSC) against side scatter light on a 1,024 channel log-
scale plot (a). A gate is drawn around the lymphocytes and gated data used to populate a CD19-PE against CD3-PERCP
plot to distinguish the B and T population, respectively (b). Rectangular gates are drawn around each lymphocyte popula-
tion and gated data used to plot anti-IgG FITC on a log scale against count on a linear scale for T lymphocytes (c) and B
lymphocytes (d).
384        J. Downing
4. Notes
References
 1. Garovoy MR et al (1983) Flow cytometry anal-        3. Patel R, Terasaki PI (1969) Significance of the
    ysis: a high technology crossmatch technique           positive crossmatch test in kidney transplanta-
    facilitating transplantation. Transplant Proc          tion. N Engl J Med 280:735–739
    15:1939–1944                                        4. Chapman JR et al (1985) Analysis of flow
 2. Bray RA, Lebeck LK, Gebel HM (1989) The                cytometry and cytotoxic crossmatches in renal
    flow cytometric crossmatch: dual-colour analy-         transplantation. Transplant Proc 17:2480–2481
    sis of T and B cell reactivities. Transplantation   5. Ogura K et al (1993) The significance of a posi-
    48:834–840                                             tive flow cytometry crossmatch test in primary
390       J. Downing
    kidney transplantation. Transplantation 56:           for renal transplantation. Exp Mol Pathol 85:
    294–298                                               59–63
 6. Sridhar NR et al (1992) Evaluation of             12. Gebel HM, Bray RA, Nickerson P (2003) Pre-
    flowcytometric crossmatching in renal allograft       transplant assessment of donor-reactive. HLA-
    recipients. Nephron 62:262–266                        specific antibodies in renal transplantation:
 7. Lazda VA (1994) Identification of patients at         contraindication vs. risk. Am J Transplant
    risk for inferior renal allograft outcome by a        3:1488–1500
    strongly positive B cell flow cytometry cross-    13. Taylor CJ et al (2009) Back to the future: appli-
    match. Transplantation 57:964–969                     cation of contemporary technology to long-
 8. Talbot D et al (1995) Flow cytometric cross-          standing questions about the clinical relevance
    matching in renal transplantation—the long            of human leukocyte antigen-specific alloanti-
    term outcome. Transpl Immunol 3:352–355               bodies in renal transplantation. Hum Immunol
 9. Graff RJ et al (2009) The role of positive flow       70:563–568
    cytometry crossmatch in late renal allograft      14. Riethmuller S et al (2010) Donor-specific anti-
    loss. Hum Immunol 70:502–505                          body levels and three generations of cross-
10. Lefaucher C et al (2008) Clinical relevance of        matches to predict antibody mediated rejection
    preformed HLA donor-specific antibodies in            in kidney transplantation. Transplantation
    kidney transplantation. Am J Transplant               90:160–167
    8:324–331                                         15. Vaida S et al (2001) Improved flow cytometric
11. Delgado JC, Eckels DD (2008) Positive B-cell          detection of HLA alloantibodies using pronase.
    only flow cytometric crossmatch: implications         Transplantation 71:422–428
                                                                                        Chapter 23
Abstract
Natural killer (NK) cells are more than simple killers and have been implicated in control and clearance of
malignant and virally infected cells, regulation of adaptive immune responses, rejection of bone marrow
transplants, and autoimmunity and the maintenance of pregnancy. Human NK cells largely use a family of
germ-line encoded killer cell immunoglobulin-like receptors (KIR) to respond to the perturbations from
self-HLA class I molecules present on infected, malignant, or HLA-disparate fetal or allogenic transplants.
Genes encoding KIR receptors and HLA class I ligands are located on different chromosomes, and both
feature extraordinary diversity in the number and type of genes. The independent segregation of KIR and
HLA gene families produce diversity in the number and type of KIR-HLA gene combinations inherited in
individuals, which may determine their immunity and susceptibility to diseases. This chapter provides an
overview of NK cells and their unprecedented phenotypic and functional diversity within and between
individuals.
Key words: NK cells, Innate immunity, HLA, KIR, Polymorphism, Immune genes
1. Natural Killer
Cells
                                Natural killer (NK) cells are the third population of lymphocytes
                                defined by CD3−, CD56+ cell surface phenotype, and they represent
                                5–25% of the mononuclear cell fraction of normal human periph-
                                eral blood (1, 2). NK cells share several features with CD8+ cytolytic
                                T lymphocytes (CTL) in their development, morphology, cell-
                                surface phenotypes, killing mechanism, and cytokine production
                                (3). Compared to T and B lymphocytes, NK cells are larger and
                                contain preformed cytolytic granules (granzymes and perforin) as
                                well as an exponential amount of constitutively expressed tran-
                                scripts for interferon-g (IFN-g) and certain cytokines that activate
                                other immune cell types. The fundamental function of NK cells is
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_23, © Springer Science+Business Media New York 2012
                                                         391
392     R. Rajalingam
2. Natural Killer
Cell Receptors
                        NK cells have a highly specific and complex target cell recognition
                        receptor system arbitrated by the integration of signals triggered by a
                        multitude of inhibitory and activating receptors, which trigger cyto-
                        toxicity and the secretion of chemokines and cytokines (14, 15).
                        Unlike T and B lymphocytes, NK cells do not express receptors that
                        require somatic gene rearrangements to generate receptor diversity
                        and specificity. Instead, NK cells express a wide array of conventional
                        germline-encoded receptor families with inhibitory or activating
                        functions, including (i) killer cell immunoglobulin-like receptors
                        (KIR), (ii) killer cell lectin-like receptors (KLR) such as CD94/
                        NKG2, (iii) leukocyte immunoglobulin-like receptors (LILR), and
                        (iv) natural cytotoxicity receptors (NCR), such as NKp46, NKp44,
                        and NKp30, and 2B4 (2, 16–18). Most of these receptors are
                        expressed in stochastic, variegated combinations of activating and
                        inhibitory receptors, resulting in many subsets of functionally distinct
23   Overview of the Killer Cell Immunoglobulin-Like Receptor System                           393
      Fig. 1. Natural killer (NK) cell response against healthy and unhealthy cells. NK cells
      express both inhibitory and activating receptors. Inhibitory receptors recognize self HLA
      class I molecules and trigger signals that stop the natural lytic function of NK cells. By
      expressing normal levels of multiple HLA class I molecules, the healthy cells are resistant
      to NK cell attack (a). Downregulation of HLA class I expression due to tumor transforma-
      tion or viral infection relieves the inhibitory influence on NK cells (b), permitting NK cells
      to lyse these unhealthy target cells, a phenomenon first described as the “missing-self”
      hypothesis. Upon transformation or infection, the unhealthy cells express ligands for
      activating receptors, which could be either “induced-self” (MICA, MICB, ULBP), “altered-
      self” (HLA-class I loaded with foreign peptide), and/or “non-self” (pathogen-encoded mol-
      ecules). Upon recognizing these ligands, the activating receptors trigger signals that
      augment NK cell lysis of unhealthy targets.
394     R. Rajalingam
                        distinct HLA class I molecules (HLA-A, -B, -C, and -E) on normal
                        healthy cells provide ligands for a variety of inhibitory receptors of
                        NK cells, and consequently are resistant to NK cell attack.
                        Downregulation of HLA class I expression due to certain viral
                        infections, neoplastic transformations or other forms of stress,
                        relieves the inhibitory influence on NK cells, permitting NK cells
                        to eliminate these unhealthy target cells, a phenomenon originally
                        described as the “missing-self” hypothesis (22). In addition to the
                        “missing-self” mechanism, the expression of ligands for activating
                        receptors on target cell surface might also contribute to NK cell
                        attack (Fig. 1). The ligands identified for activating receptors are
                        either “induced-self” that are structurally related to HLA class I
                        molecules (such as, major histocompatibility complex [MHC] class
                        I-like chains A and B [MICA and MICB] and unique long binding
                        proteins [ULBP]), “altered-self” (HLA class I molecule loaded
                        with foreign peptide) or pathogen encoded “non-self” (molecules
                        associated with infection and tumor transformation).
2.1. Killer Cell        Compared to other NK cell receptors, the KIR receptors are con-
Immunoglobulin-Like     sidered to be the key receptors that control the development and
Receptors               function of human NK cells (16, 23–26). The following rationale
                        substantiates the importance of KIR receptors: (i) KIR receptor
                        system is totally absent in rodents, the basic model for clinical
                        research, and therefore the mouse and human NK cell biology do
                        not converge in many crucial aspects (27, 28); (ii) species compari-
                        son suggests that the KIR system originated in mammals, and
                        evolved rapidly to keep up with species-specific evolution and adap-
                        tation, and thus only a small minority of KIR genes is shared by
                        humans and chimpanzees (29); (iii) KIR receptors are encoded by
                        a polygenic and polymorphic gene family that produces remarkable
                        diversity within individuals and in populations in the number and
                        type of genes (30, 31); (iv) KIR receptors recognize an array of
                        polymorphic HLA class I molecules that evolved rapidly to mount
                        effective adaptive immune response against rapidly evolving viruses
                        and pathogens; (v) genes encoding KIR receptors and HLA class I
                        ligands are located on different chromosomes, and their indepen-
                        dent segregation results in variable KIR-HLA combinations in
                        individuals, which may contribute to the individual’s health and
                        disease. Consistent with this theory, combinations of certain KIR-
                        HLA genes have been associated with diseases as diverse as autoim-
                        munity, infection, cancer, and reproductive failure (32, 33).
                             Fourteen KIR receptors triggering either inhibition (3DL1-3,
                        2DL1-3, 2DL5) or activation (3DS1, 2DS1-5), or both (2DL4)
                        have been identified in humans (Fig. 2) (34–36). KIR receptors
                        possess two or three extracellular Ig-like domains involved in ligand
                        binding and either a long or short cytoplasmic tail involved in sig-
                        naling. KIRs are named on the basis of their number of Ig domains
                        and type of cytoplasmic tail (37). The first digit following the
23   Overview of the Killer Cell Immunoglobulin-Like Receptor System                          395
      Fig. 2. Killer cell immunoglobulin-like receptors (KIR). Fourteen distinct KIR receptors have
      been characterized in humans that comprise either two (2D) or three (3D) extracellular
      Ig-like domains and either a long (L) or short (S) cytoplasmic tail. Six KIR receptors are
      activating types and the remaining KIR are inhibitory types. The ITIM motifs in the cyto-
      plasmic tails of inhibitory KIRs are shown as boxes, and positively charged residues in the
      transmembrane regions of activating KIRs are shown as circles. The inhibitory KIR recep-
      tors bind to distinct HLA class I allotypes and the ligands for most activating KIR receptors
      are unknown.
2.2. Ligands for KIR             KIR receptors recognize specific motifs of HLA class I molecules,
Receptors                        which are the products of highly polymorphic genes of the MHC
                                 located on chromosome 6 (39–41) (Figs. 2 and 3). KIR binds in a
                                 nearly orthogonal orientation across the a1 and a2 helices of HLA
                                 class I molecules, and the KIR footprint on HLA overlaps with but
                                 is distinct from that of the T cell receptor (42). The six loops of D1
                                 and D2 interdomain hinge region of KIR2DL as well as KIR3DL
                                 form the HLA class I binding site, while the membrane-distal D0
                                 domain of KIR3D enhances HLA class I ligand binding (43). Since
                                 the KIR binds to the polymorphic peptide binding groove of HLA
                                 class I molecules, it is constantly subjected to changes driven by
                                 rapidly evolving viruses. Therefore, the KIR binding motifs are
                                 often variable between individuals and their distribution differs
                                 substantially between populations (39, 44, 45).
                                      HLA-C is the dominant HLA class I locus that provides ligands
                                 for many KIR receptors (Fig. 2). Amino acid residues 76 and 80 on
                                 HLA-C allotypes determine KIR binding epitopes (46–48) (Fig. 3).
                                 All HLA-C allotypes carry valine (V) at position 76, while position
Fig. 3. KIR receptor binding region of HLA class I molecule. Locations of amino acids (position 73, 76–83, and 90) in the a1
domain of HLA class I molecules (shown in dark shade) involved in KIR receptor binding.
                   23   Overview of the Killer Cell Immunoglobulin-Like Receptor System     397
3. Extraordinary
Diversity of KIR
Receptor System
                         The unique feature of the KIR system that distinguishes it from
                         other types of NK cell receptors is its substantial diversity, which is
                         contributed by various factors such as individual-specific KIR gene
                         content, nucleotide sequence polymorphism of each KIR gene, and
                         stochastic and variegated expression of KIR receptor repertoires on
                         individual NK cell clones.
398        R. Rajalingam
3.1. Gene Content                KIR receptors are encoded by a family of tightly clustered genes on
Diversity of KIR Gene            the leukocyte receptor complex (LRC) that spans a region of about
Complex                          150 kb on chromosome 19q13.4 (34, 35). The number and type
                                 of KIR genes vary substantially between haplotypes (Fig. 4). Over
                                 30 distinct KIR haplotypes with distinct gene content have been
                                 characterized to date by sequencing genomic clones and haplotype
                                 segregation analysis in families (34, 69–74). The most commonly
                                 occurring haplotype in most human populations is conventionally
                                 called the “group-A haplotype,” which carries a fixed gene content
                                 comprising KIR3DL3-2DL3-2DP1-2DL1-3DP1-2DL4-3DL1-
                                 2DS4-3DL2 (Fig. 4; haplotype 1) (31, 34). Remaining KIR hap-
                                 lotypes are collectively referred to as “group-B haplotypes,” which
                                 have variable gene content comprising several genes that are not
                                 part of the A haplotype (Fig. 4; haplotypes 2–22). Particularly,
                                 KIR2DS1, 2DS2, 2DS3, 2DS5, 2DL2, 2DL5, and 3DS1 are asso-
                                 ciated only with group-B haplotypes, and thus B haplotypes gener-
                                 ally encode more activating KIR receptors than the A haplotype
                                 that encodes a single activating receptor, KIR2DS4 (34, 69, 73, 74).
                                 Despite such extraordinary gene-content diversity, four KIR genes,
                                 2DL4, 3DL2, 3DL3, and 3DP1, are present on virtually all KIR
Fig. 4. KIR haplotypes differ by gene content. Map of KIR haplotypes as determined by family segregation analysis. Haplotype
1 represents group-A KIR haplotype and the remainder group-B haplotypes. The framework genes, present in all haplotypes
are shown in dark boxes; genes encoding activating KIR are in white boxes ; and those for inhibitory receptors are in gray
boxes. The KIR2DP1 and 3DP1 are pseudogenes that do not express a receptor. Maps are not drawn to scale.
                         23    Overview of the Killer Cell Immunoglobulin-Like Receptor System                           399
Fig. 5. KIR gene content differs between individuals. The number and type of KIR genes can be substantially variable
between individuals, even within a family. (a) Is the gel picture indicating the extreme difference in the KIR gene content of
two siblings. Sibling-1 has fewer genes and sibling-2 has all known genes. Arrows indicate internal positive control bands
specific to an invariant gene. The pseudogenes KIR2DP1 and 3DP1 are not typed. (b) Shows the possible KIR haplotype
composition of each sibling.
400         R. Rajalingam
Fig. 6. Ten most commonly occurring KIR genotypes in populations. Genotyping studies revealed a significant ethnic difference
in the distribution of KIR genotypes. Listed are the ten most frequently occurring KIR genotypes in 51 distinct human popu-
lations. The presence of a gene is indicated by a shaded box while the absence of a gene is indicated by a white box.
Fig. 7. Cetromeric and telomeric halves of KIR haplotypes. A stretch of 14 kb enriched with L1 repeats upstream of KIR2DL4
divides the KIR haplotype into two halves. The centromeric half is delimited by 3DL3 and 3DP1 while the telomeric half is
delimited by 2DL4 and 3DL2. Multiple reciprocal meiotic recombination events between 3DP1 and 2DL4 shuffled the
centromeric (c) and telomeric (t) motifs, and thus diversify gene content KIR haplotypes across individuals and populations.
Most of the KIR gene content haplotypes published to date can be explained by the recombination of these 10 centromeric
and 10 telomeric gene content motifs. The framework genes, present in all haplotypes are shown in dark boxes; genes
encoding activating KIR are in white boxes; and those for inhibitory receptors are in gray boxes. The KIR2DP1 and 3DP1
are pseudogenes that do not express a receptor. Letter “A” in gene-content motif identification indicates parts of group-A
haplotypes while “B” indicates parts of group-B haplotypes.
                                 are present on all KIR haplotypes and thus occur in 100% of all
                                 populations (75). In contrast, the existence of the other 12 KIR
                                 genes is considerably variable. The inhibitory receptors KIR2DL2
                                 and 2DL3 segregate as alleles of a single locus at the centromeric
                                 half. Similarly, the inhibitory KIR3DL1 and activating KIR3DS1
                                 behave as alleles of the same locus at the telomeric half. Almost all
23   Overview of the Killer Cell Immunoglobulin-Like Receptor System   401
      haplotypes contain these two loci, such that virtually every one has
      either 2DL2 or 2DL3, and 3DL1 or 3DS1 within their KIR genome.
      KIR2DL1, 2DL2, 2DL3, and 2DS2 are specific to the centromeric
      half while KIR3DL1, 3DS1, 2DS1, and 2DS4 are specific to the
      telomeric half. Three KIR genes, 2DL5, 2DS3, and 2DS5, are
      found in both centromeric and telomeric locations (70, 74). For
      genes within each half, there is significant linkage disequilibrium,
      but much less for genes in the two different halves (73, 77).
           Multiple reciprocal recombination events at the center of the
      KIR complex, between 3DP1 and 2DL4, presumably diversify
      gene content for KIR haplotypes across individuals and popula-
      tions (75, 78). Most of the KIR gene content haplotypes published
      to date can be explained by the recombination of 10 centromeric
      and 10 telomeric gene content motifs (Figs. 4 and 7). For example,
      haplotype 1 listed in Fig. 4 is a recombinant of the centromeric half
      cA01 and telomeric half tA01; similarly, haplotype 8 is a recombinant
      of the centromeric half cB02 and telomeric half tB02. The recipro-
      cal recombination also results haplotypes carrying both group-A
      and group-B haplotype-specific motifs. For instance, haplotype 6
      listed in Fig. 4 is a recombinant of the centromeric half cA01 and
      telomeric half tB04, and haplotype 9 is a recombinant of the cen-
      tromeric half cB01 and telomeric tA01. Recombination events
      also are reported outside the region between 3DP1 and 2DL4.
      These are generally nonallelic crossovers generating several unusual
      haplotypes, including truncated haplotypes that are missing some
      framework genes (70, 77) or elongated haplotypes that contain
      duplicated genes (79–81). It is important to remark here that the
      current genotyping methods used to detect the presence and
      absence of individual KIR gene will not identify the copy number
      of each KIR gene within a given individual. Therefore, more pre-
      cise typing methods, such as a yet to be developed quantitative
      real-time PCR assay are necessary to investigate the KIR gene copy
      number and their impact on human disease.
           All human populations have both group-A and B haplotypes,
      but their distribution varies considerably across distinct popula-
      tions. Individuals carrying both A and B haplotypes are more com-
      mon in Caucasians and Africans, and therefore A and B haplotypes
      are approximately equally distributed in these populations (31,
      75). In contrast, the prevalence of inhibitory haplotypes (group-A)
      dominates over the activating haplotypes (group-B) in one popula-
      tion and vice versa in others. Individuals carrying homozygous
      group-A KIR haplotypes (AA genotypes) are common in Northeast
      Asians (Chinese, Japanese, and Koreans) (72, 82, 83). Conversely,
      individuals carrying AB or BB genotypes are common in the natives
      of America (84, 85), Australia (86), and India (87–89). The NK
      cells from AA homozygous individuals can express a maximum of
      four inhibitory KIRs (2DL1, 2DL3, 3DL1, and 3DL2) and one
      activating KIR (2DS4). In contrast, individuals carrying AB or BB
402         R. Rajalingam
3.2. Nucleotide                   In addition to gene content diversity, each KIR gene itself exhibits
Polymorphism                      considerable nucleotide sequence polymorphism (37, 73, 77,
of KIR Genes                      90, 91). Allelic sequence variants of KIR genes are named in a
                                  fashion analogous to that used for HLA alleles (37) (Fig. 8). After
                                  the gene name, an asterisk is used as a separator before a numerical
                                  allele designation. The first three digits of the numerical designa-
                                  tion indicate alleles that differ in the sequences of their encoded
                                  proteins. The next two digits distinguish alleles that differ only by
                                  synonymous (noncoding) differences within the coding sequence.
                                  The final two digits distinguish alleles that only differ by substitu-
                                  tions in an intron, promoter, or other noncoding region of the
                                  sequence.
                                       To date, 614 KIR nucleotide sequences encoding 321 distinct
                                  KIR proteins have been deposited in IPD-KIR database (Release
                                  2.4.0, 15 April 2011), the database that provides a centralized
                                  repository for human KIR sequences (http://www.ebi.ac.uk/ipd/
                                  kir/). Inhibitory KIR receptors are more polymorphic than the
                                  activating KIR receptors. The highest allelic polymorphisms are
                                  reported for 3DL1, 3DL2, and 3DL3 loci, which is probably due
                                  to the subjective sequencing analyses of large cohorts by certain
                                  laboratories that focus on these loci (92–95).
                                       The functional consequence of sequence polymorphism has
                                  been studied in great detail for the KIR3DL1 locus. The amino
Fig. 8. Nomenclature of KIR alleles. Allelic sequence variants of KIR genes are named in a fashion analogous to that used for
HLA alleles. After the gene name, an asterisk is used as a separator before a numerical allele designation. The first three digits
of the numerical designation indicate alleles that differ in the sequences of their encoded proteins. The next two digits distin-
guish alleles that only differ by synonymous (noncoding) differences within the coding sequence. The final two digits distinguish
alleles that only differ by substitutions in an intron, promoter, or other noncoding region of the sequence.
                       23   Overview of the Killer Cell Immunoglobulin-Like Receptor System     403
3.3. KIR Receptor            In addition to the gene content variation and sequence polymor-
Repertoire Diversity         phism, the level of mRNA expression varies among KIR genes and
of NK Cell Clones            alleles. For instance, the KIR3DL3 transcripts are detected at low
                             levels in peripheral blood when compared to other KIR genes
                             (24, 105). Further, alternatively spliced mRNA are reported for
                             most KIR genes, and such isoforms can hinder the cell surface
404        R. Rajalingam
Fig. 9. Sequence polymorphism of KIR receptors. The number of nucleotide sequences (black bars) and their predicted
protein variants (gray bars) characterized to date for each KIR locus are shown. This data was extracted from IPD-KIR
database (http://www.ebi.ac.uk/ipd/kir/stats.html; Release 2.4.0; April 2011) that provides a centralized repository for
human KIR sequences. Null alleles identified for each locus are given in the parenthesis.
4. Diversity
of KIR-HLA
Compound
Genotypes and         KIR and HLA loci are both highly polymorphic and they map to
Relavance in          distinct human chromosomes (Chromosomes 19 and 6, respec-
Disease               tively). KIR and HLA gene families segregate independently, yield-
                      ing many individuals who express KIR receptors for which they
                      lack HLA class I ligands, and vice versa, thus creating human diver-
                      sity in the number and type of KIR-HLA inherited gene pairs (99).
                      Nevertheless, at the population level there is compelling evidence
406   R. Rajalingam
5. Alloreactive
NK Cells in
Hematopoietic
Stem Cell               HLA-matched allogeneic hematopoietic stem cell transplantation
Transplantation         (HSCT) is an effective treatment for hematologic malignancies,
                        including leukemia, lymphoma, and inherited hematopoietic stem
                        cell diseases (133). Donor T cells in the allograft are critical for pro-
                        moting engraftment and eradicating malignant cells. Unfortunately,
                        alloreactive T cells also cause graft-versus-host disease (GVHD),
                        which is an attack on recipient tissues, primarily the gastrointestinal
                        tract, liver, and skin. T cell depletion prevents GVHD but increases
                        the risk of graft rejection and leukemic relapse.
                             Following allogeneic HSCT, the progeny of the donor stem
                        cells repopulate the entire hematopoietic system of the recipient.
                        NK cells are the first lymphocyte population to appear in peripheral
                        blood shortly after HSCT. KIR receptor repertoires of NK cells
408   R. Rajalingam
Acknowledgment
References
  1. Caligiuri MA (2008) Human natural killer          12. Manaster I, Mandelboim O (2008) The unique
     cells. Blood 112:461–469                              properties of human NK cells in the uterine
  2. Vivier E et al (2011) Innate or adaptive              mucosa. Placenta 29(suppl A):S60–S66
     immunity? The example of natural killer cells.    13. Colonna M (2009) Interleukin-22-producing
     Science 331:44–49                                     natural killer cells and lymphoid tissue
  3. Colucci F et al (2003) What does it take to           inducer-like cells in mucosal immunity.
     make a natural killer? Nat Rev Immunol                Immunity 31:15–23
     3:413–425                                         14. Bryceson YT, Long EO (2008) Line of attack:
  4. Lee SH et al (2007) Keeping NK cells in               NK cell specificity and integration of signals.
     highly regulated antiviral warfare. Trends            Curr Opin Immunol 20:344–352
     Immunol 28:252–259                                15. Lanier LL (2003) Natural killer cell receptor
  5. Smyth MJ et al (2002) New aspects of                  signaling. Curr Opin Immunol 15:308–314
     natural-killer-cell surveillance and therapy of   16. McQueen KL, Parham P (2002) Variable
     cancer. Nat Rev Cancer 2:850–861                      receptors controlling activation and inhibi-
  6. Sun JC et al (2010) Immune memory                     tion of NK cells. Curr Opin Immunol 14:
     redefined: characterizing the longevity of nat-       615–621
     ural killer cells. Immunol Rev 236:83–94          17. Moretta L et al (2000) Human NK-cell recep-
  7. Cooper MA et al (2001) Human natural killer           tors. Immunol Today 21:420–422
     cells: a unique innate immunoregulatory role      18. Lanier LL (2005) NK cell recognition. Annu
     for the CD56 (bright) subset. Blood                   Rev Immunol 23:225–274
     97:3146–3151                                      19. Lanier LL (2008) Up on the tightrope: natu-
  8. Freud AG, Caligiuri MA (2006) Human                   ral killer cell activation and inhibition. Nat
     natural killer cell development. Immunol Rev          Immunol 9:495–502
     214:56–72                                         20. Valiante NM et al (1997) Functionally and
  9. Jacobs R et al (2001) CD56bright cells differ         structurally distinct NK cell receptor reper-
     in their KIR repertoire and cytotoxic features        toires in the peripheral blood of two human
     from CD56dim NK cells. Eur J Immunol                  donors. Immunity 7:739–751
     31:3121–3127                                      21. Biron CA (2001) Interferons alpha and beta
 10. Fehniger TA et al (2003) CD56bright natural           as immune regulators—a new look. Immunity
     killer cells are present in human lymph nodes         14:661–664
     and are activated by T cell-derived IL-2: a       22. Ljunggren HG, Karre K (1990) In search of
     potential new link between adaptive and               the ‘missing self’: MHC molecules and NK cell
     innate immunity. Blood 101:3052–3057                  recognition. Immunol Today 11:237–244
 11. Cooper MA et al (2004) NK cell and DC             23. Lanier LL (1998) NK cell receptors. Annu
     interactions. Trends Immunol 25:47–52                 Rev Immunol 16:359–393
410      R. Rajalingam
24. Long EO et al (2001) Inhibition of natural            41. Klein J, Sato A (2000) The HLA system. First
    killer cell activation signals by killer cell immu-       of two parts. N Engl J Med 343:702–709
    noglobulin-like receptors (CD158). Immunol            42. Boyington JC et al (2000) Crystal structure
    Rev 181:223–233                                           of an NK cell immunoglobulin-like receptor
25. Moretta A et al (2001) Activating receptors               in complex with its class I MHC ligand.
    and coreceptors involved in human natural                 Nature 405:537–543
    killer cell-mediated cytolysis. Annu Rev              43. Khakoo SI et al (2002) The D0 domain of
    Immunol 19:197–223                                        KIR3D acts as a major histocompatibility
26. Vilches C, Parham P (2002) KIR: diverse,                  complex class I binding enhancer. J Exp Med
    rapidly evolving receptors of innate and adaptive         196:911–921
    immunity. Annu Rev Immunol 20:217–251                 44. Prugnolle F et al (2005) Pathogen-driven
27. Lanier LL (2008) Evolutionary struggles                   selection and worldwide HLA class I diversity.
    between NK cells and viruses. Nat Rev                     Curr Biol 15:1022–1027
    Immunol 8:259–268                                     45. Parham P, Ohta T (1996) Population biology
28. Parham P (2003) Immunogenetics of killer-                 of antigen presentation by MHC class I mol-
    cell immunoglobulin-like receptors. Tissue                ecules. Science 272:67–74
    Antigens 62:194–200                                   46. Colonna M et al (1992) Alloantigen recogni-
29. Abi-Rached L et al (2010) Human-specific                  tion by two human natural killer cell clones is
    evolution and adaptation led to major qualita-            associated with HLA-C or a closely linked
    tive differences in the variable receptors of             gene. Proc Natl Acad Sci USA 89:
    human and chimpanzee natural killer cells.                7983–7985
    PLoS Genet 6:e1001192                                 47. Mandelboim O et al (1996) Protection from
30. Parham P (2005) MHC class I molecules and                 lysis by natural killer cells of group 1 and 2
    KIRs in human history, health and survival.               specificity is mediated by residue 80 in human
    Nat Rev Immunol 5:201–214                                 histocompatibility leukocyte antigen C alleles
31. Uhrberg M et al (1997) Human diversity in                 and also occurs with empty major histocom-
    killer cell inhibitory receptor genes. Immunity           patibility complex molecules. J Exp Med
    7:753–763                                                 184:913–922
32. Khakoo SI, Carrington M (2006) KIR and                48. Mandelboim O et al (1997) The binding site
    disease: a model system or system of models?              of NK receptors on HLA-C molecules.
    Immunol Rev 214:186–201                                   Immunity 6:341–350
33. Bashirova AA et al (2006) The killer                  49. Colonna M et al (1993) HLA-C is the inhibi-
    immunoglobulin-like receptor gene cluster:                tory ligand that determines dominant resis-
    tuning the genome for defense. Annu Rev                   tance to lysis by NK1- and NK2-specific
    Genomics Hum Genet 7:277–300                              natural killer cells. Proc Natl Acad Sci USA
34. Wilson MJ et al (2000) Plasticity in the orga-            90:12000–12004
    nization and sequences of human KIR/ILT               50. Wagtmann N et al (1995) Molecular clones of
    gene families. Proc Natl Acad Sci USA 97:                 the p58 NK cell receptor reveal
    4778–4783                                                 immunoglobulin-related molecules with
35. Trowsdale J (2001) Genetic and functional                 diversity in both the extra- and intracellular
    relationships between MHC and NK receptor                 domains. Immunity 2:439–449
    genes. Immunity 15:363–374                            51. Winter CC, Long EO (1997) A single amino
36. Steffens U et al (1998) Nucleotide and amino              acid in the p58 killer cell inhibitory receptor
    acid sequence alignment for human killer cell             controls the ability of natural killer cells to dis-
    inhibitory receptors (KIR), 1998. Tissue                  criminate between the two groups of HLA-C
    Antigens 51:398–413                                       allotypes. J Immunol 158:4026–4028
37. Marsh SG et al (2003) Killer-cell immuno-             52. Moesta AK et al (2008) Synergistic polymor-
    globulin-like receptor (KIR) nomenclature                 phism at two positions distal to the ligand-
    report, 2002. Tissue Antigens 62:79–86                    binding site makes KIR2DL2 a stronger
38. Lanier LL (2009) DAP10- and DAP12-                        receptor for HLA-C than KIR2DL3.
    associated receptors in innate immunity.                  J Immunol 180:3969–3979
    Immunol Rev 227:150–160                               53. Winter CC et al (1998) Direct binding and
39. Marsh SG et al (2011) Nomenclature for fac-               functional transfer of NK cell inhibitory recep-
    tors of the HLA system, 2010. Tissue Antigens             tors reveal novel patterns of HLA-C allotype
    75:291–455                                                recognition. J Immunol 161:571–577
40. Klein J, Sato A (2000) The HLA system. Second         54. Stewart CA et al (2005) Recognition of
    of two parts. N Engl J Med 343:782–786                    peptide-MHC class I complexes by activating
                      23    Overview of the Killer Cell Immunoglobulin-Like Receptor System            411
82. Jiang K et al (2005) Distribution of killer cell          predicts a soluble KIR molecule analogous to
    immunoglobulin-like receptor genes in the                 the KIR1D molecule observed in the rhesus
    Chinese Han population. Tissue Antigens                   monkey. Tissue Antigens 60:254–258
    65:556–563                                          99.   Du Z et al (2007) Receptor-ligand analyses
83. Yawata M et al (2002) Predominance of                     define minimal killer cell Ig-like receptor
    group A KIR haplotypes in Japanese associ-                (KIR) in humans. Immunogenetics 59:1–15
    ated with diverse NK cell repertoires of KIR       100.   Witt CS et al (2000) Detection of KIR2DL4
    expression. Immunogenetics 54:543–550                     alleles by sequencing and SSCP reveals a com-
84. Gendzekhadze K et al (2006) High KIR                      mon allele with a shortened cytoplasmic tail.
    diversity in Amerindians is maintained using              Tissue Antigens 56:248–257
    few gene-content haplotypes. Immunogenetics        101.   Goodridge JP et al (2003) KIR2DL4
    58:474–480                                                (CD158d) genotype influences expression
85. Ewerton PD et al (2007) Amazonian                         and function in NK cells. J Immunol 171:
    Amerindians exhibit high variability of KIR               1768–1774
    profiles. Immunogenetics 59:625–630                102.   Vilches C et al (2000) Gene structure and
86. Toneva M et al (2001) Genomic diversity of                promoter variation of expressed and nonex-
    natural killer cell receptor genes in three pop-          pressed variants of the KIR2DL5 gene.
    ulations. Tissue Antigens 57:358–362                      J Immunol 165:6416–6421
87. Rajalingam R et al (2008) Distinct diversity of    103.   Pando MJ et al (2003) The protein made
    KIR genes in three southern Indian popula-                from a common allele of KIR3DL1
    tions: comparison with world populations                  (3DL1*004) is poorly expressed at cell sur-
    revealed a link between KIR gene content and              faces due to substitution at positions 86 in Ig
    pre-historic          human          migrations.          domain 0 and 182 in Ig domain 1. J Immunol
    Immunogenetics 60:207–217                                 171:6640–6649
88. Rajalingam R et al (2002) Distinctive KIR          104.   VandenBussche CJ et al (2006) A single poly-
    and HLA diversity in a panel of north Indian              morphism disrupts the killer Ig-like receptor
    Hindus. Immunogenetics 53:1009–1019                       2DL2/2DL3 D1 domain. J Immunol 177:
89. Kulkarni S et al (2008) Comparison of the                 5347–5357
    rapidly evolving KIR locus in Parsis and natives   105.   Trompeter HI et al (2005) Three structurally
    of India. Immunogenetics 60:121–129                       and functionally divergent kinds of promoters
90. Garcia CA et al (2003) Human KIR sequences                regulate expression of clonally distributed
    2003. Immunogenetics 55:227–239                           killer cell Ig-like receptors (KIR), of
                                                              KIR2DL4, and of KIR3DL3. J Immunol
91. Rajalingam R et al (2001) Identification of
                                                              174:4135–4143
    seventeen novel KIR variants: fourteen of
    them from two non-Caucasian donors. Tissue         106.   Dohring C et al (1996) Alternatively spliced
    Antigens 57:22–31                                         forms of human killer inhibitory receptors.
                                                              Immunogenetics 44:227–230
92. Norman PJ et al (2007) Unusual selection on
    the KIR3DL1/S1 natural killer cell receptor        107.   Raulet DH et al (2001) Regulation of the
    in Africans. Nat Genet 39:1092–1099                       natural killer cell receptor repertoire. Annu
                                                              Rev Immunol 19:291–330
93. Gardiner CM et al (2001) Different NK cell
    surface phenotypes defined by the DX9 anti-        108.   Shilling HG et al (2002) Genetic control of
    body are due to KIR3DL1 gene polymor-                     human NK cell repertoire. J Immunol
    phism. J Immunol 166:2992–3001                            169:239–247
94. Jones DC et al (2006) Nature of allelic            109.   Santourlidis S et al (2002) Crucial role of
    sequence polymorphism at the KIR3DL3                      DNA methylation in determination of clon-
    locus. Immunogenetics 58:614–627                          ally distributed killer cell Ig-like receptor
                                                              expression patterns in NK cells. J Immunol
95. Hou L et al (2007) Seventeen novel alleles                169:4253–4261
    add to the already extensive KIR3DL3 diver-
    sity. Tissue Antigens 70:449–454                   110.   Chan HW et al (2003) DNA methylation
                                                              maintains allele-specific KIR gene expression
96. O’Connor GM et al (2007) Functional poly-                 in human natural killer cells. J Exp Med
    morphism of the KIR3DL1/S1 receptor on                    197:245–255
    human NK cells. J Immunol 178:235–241
                                                       111.   Andersson S et al (2009) KIR acquisition
97. Carr WH et al (2005) KIR3DL1 polymor-                     probabilities are independent of self-HLA
    phisms that affect NK cell inhibition by                  class I ligands and increase with cellular KIR
    HLA-Bw4 ligand. J Immunol 175:5222–5229                   expression. Blood 114:95–104
98. Maxwell LD et al (2002) A common                   112.   Schonberg K et al (2011) Analyses of HLA-
    KIR2DS4 deletion variant in the human that                C-specific KIR repertoires in donors with
                       23    Overview of the Killer Cell Immunoglobulin-Like Receptor System                413
       group A and B haplotypes suggest a ligand-                mediated by fetal HLA-C2. J Clin Invest
       instructed model of NK cell receptor aquisi-              120:4102–4110
       tion. Blood 117:98–107                             128.   Terme M et al (2008) Natural killer cell-
113.   Anfossi N et al (2006) Human NK cell educa-               directed therapies: moving from unexpected
       tion by inhibitory receptors for MHC class I.             results to successful strategies. Nat Immunol
       Immunity 25:331–342                                       9:486–494
114.   Kim S et al (2005) Licensing of natural killer     129.   Levinson RD et al (2010) Killer cell immuno-
       cells by host major histocompatibility com-               globulin-like receptor gene-cluster 3DS1-
       plex class I molecules. Nature 436:709–713                2DL5-2DS1-2DS5 predisposes susceptibility to
115.   Cooley S et al (2007) A subpopulation of                  Vogt-Koyanagi-Harada syndrome in Japanese
       human peripheral blood NK cells that lacks                individuals. Hum Immunol 71:192–194
       inhibitory receptors for self MHC is develop-      130.   Lopez-Vazquez A et al (2005) Protective
       mentally immature. Blood 110:578–586                      effect of the HLA-Bw4I80 epitope and the
116.   Yokoyama WM, Kim S (2006) How do natu-                    killer cell immunoglobulin-like receptor 3DS1
       ral killer cells find self to achieve tolerance?          gene against the development of hepatocel-
       Immunity 24:249–257                                       lular carcinoma in patients with hepatitis C
117.   Raulet DH, Vance RE (2006) Self-tolerance                 virus infection. J Infect Dis 192:162–165
       of natural killer cells. Nat Rev Immunol           131.   Bonagura VR et al (2010) Activating killer
       6:520–531                                                 cell immunoglobulin-like receptors 3DS1 and
118.   Bix M et al (1991) Rejection of class I MHC-              2DS1 protect against developing the severe
       deficient haemopoietic cells by irradiated                form of recurrent respiratory papillomatosis.
       MHC-matched mice. Nature 349:329–331                      Hum Immunol 71:212–219
119.   Furukawa H et al (1999) Tolerance of NK and        132.   Carrington M et al (2005) Hierarchy of resis-
       LAK activity for HLA class I-deficient targets            tance to cervical neoplasia mediated by com-
       in a TAP1-deficient patient (bare lymphocyte              binations of killer immunoglobulin-like
       syndrome type I). Hum Immunol 60:32–40                    receptor and human leukocyte antigen loci.
                                                                 J Exp Med 201:1069–1075
120.   Yu J et al (2007) Hierarchy of the human
       natural killer cell response is determined by      133.   Appelbaum FR (2003) The current status of
       class and quantity of inhibitory receptors for            hematopoietic cell transplantation. Annu Rev
       self-HLA-B and HLA-C ligands. J Immunol                   Med 54:491–512
       179:5977–5989                                      134.   Ruggeri L et al (2002) Effectiveness of donor
121.   Chang C et al (1995) Molecular characteriza-              natural killer cell alloreactivity in mismatched
       tion of human CD94: a type II membrane                    hematopoietic transplants. Science 295:
       glycoprotein related to the C-type lectin                 2097–2100
       superfamily. Eur J Immunol 25:2433–2437            135.   Giebel S et al (2003) Survival advantage with
122.   Uhrberg M et al (2001) The repertoire of                  KIR ligand incompatibility in hematopoietic
       killer cell Ig-like receptor and CD94:NKG2A               stem cell transplantation from unrelated
       receptors in T cells: clones sharing identical            donors. Blood 102:814–819
       alpha beta TCR rearrangement express highly        136.   Davies SM et al (2002) Evaluation of KIR
       diverse killer cell Ig-like receptor patterns.            ligand incompatibility in mismatched unre-
       J Immunol 166:3923–3932                                   lated donor hematopoietic transplants. Killer
123.   Single RM et al (2007) Global diversity and               immunoglobulin-like receptor. Blood 100:
       evidence for coevolution of KIR and HLA.                  3825–3827
       Nat Genet 39:1114–1119                             137.   Kawase T et al (2009) HLA mismatch combi-
124.   Gendzekhadze K et al (2009) Co-evolution                  nations associated with decreased risk of
       of KIR2DL3 with HLA-C in a human popu-                    relapse: implications for the molecular mecha-
       lation retaining minimal essential diversity of           nism. Blood 113:2851–2858
       KIR and HLA class I ligands. Proc Natl Acad        138.   Clausen J et al (2007) Impact of natural killer
       Sci USA 106:18692–18697                                   cell dose and donor killer-cell immunoglobulin-
125.   Khakoo SI et al (2004) HLA and NK cell                    like receptor (KIR) genotype on outcome fol-
       inhibitory receptor genes in resolving hepati-            lowing human leucocyte antigen-identical
       tis C virus infection. Science 305:872–874                haematopoietic stem cell transplantation. Clin
                                                                 Exp Immunol 148:520–528
126.   Hiby SE et al (2004) Combinations of mater-
       nal KIR and fetal HLA-C genes influence the        139.   Willemze R et al (2009) KIR-ligand incom-
       risk of preeclampsia and reproductive success.            patibility in the graft-versus-host direction
       J Exp Med 200:957–965                                     improves outcomes after umbilical cord blood
                                                                 transplantation for acute leukemia. Leukemia
127.   Hiby SE et al (2011) Maternal activating                  23:492–500
       KIRs protect against human reproductive failure
414       R. Rajalingam
140. Pende D et al (2009) Anti-leukemia activity            registry. Biol Blood Marrow Transplant 12:
     of alloreactive NK cells in KIR ligand-                876–884
     mismatched haploidentical HSCT for pediat-        143. Cooley S et al (2010) Donor selection for
     ric patients: evaluation of the functional role        natural killer cell receptor genes leads to supe-
     of activating KIR and redefinition of inhibi-          rior survival after unrelated transplantation
     tory KIR specificity. Blood 113:3119–3129              for acute myelogenous leukemia. Blood 116:
141. Lowe EJ et al (2003) T-cell alloreactivity             2411–2419
     dominates natural killer cell alloreactivity in   144. Venstrom JM et al (2010) Donor activating
     minimally T-cell–depleted HLA-non-identical            KIR3DS1 is associated with decreased acute
     paediatric bone marrow transplantation. Br             GVHD in unrelated allogeneic hematopoietic
     J Haematol 123:23–326                                  stem cell transplantation. Blood 115:
142. Farag SS et al (2006) The effect of KIR ligand         3162–3165
     incompatibility on the outcome of unrelated       145. Sivori S et al (2011) Natural killer cells
     donor transplantation: a report from the cen-          expressing the KIR2DS1-activating receptor
     ter for international blood and marrow trans-          efficiently kill T-cell blasts and dendritic cells:
     plant research, the European blood and                 implications in haploidentical HSCT. Blood
     marrow transplant registry, and the Dutch              117:4284–4292
                                                                                        Chapter 24
Abstract
The killer-cell immunoglobulin-like receptors (KIR), which enable NK cells to detect allogeneic target
cells and abnormalities in the expression of self-HLA molecules, are encoded by genes that display extensive
copy number variation. These variations in the KIR genotype are relevant for multiple aspects of human
health, including therapy of cancer. PCR with sequence-specific primers (SSP) is simplest and most widely
used among techniques for studying KIR genotypes. Here, we present a protocol that details the critical
steps of a method for KIR genotyping by PCR-SSP.
     Key words: Alloreactivity, Copy number variation, Electrophoresis, Genotyping, Killer-cell immuno-
     globulin-like receptors, Hemopoietic transplant, HLA, Natural killer cells, PCR, Quality assurance
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_24, © Springer Science+Business Media New York 2012
                                                         415
416         D. Ordóñez et al.
1.2. Role of KIR                NK cells kill target cells that lack HLA ligands for the inhibitory
Genotyping in the               receptors of the NK cell, as it often happens in tumours and infected
Clinical Context                cells. Killing can also occur when an NK cell meets an allogeneic
                                target that lacks one or more of the polymorphic HLA epitopes
1.2.1. Exploiting NK-Cell
                                that “educated” or “licensed” the NK cell. This phenomenon has
Alloreactivity Against
                                been proposed to mediate beneficial effects on patient survival, and
Cancer
                                on graft vs. host and leukaemia-relapse rates after hemopoietic
                                transplant from non-HLA identical donors (5). Similarly, adoptive
                                transfer of allogeneic NK cells is a promising form of immuno-
                                therapy against cancer. In both clinical settings, selection of the
                                appropriate donors requires identification of the HLA epitopes
                                recognised by the inhibitory KIR 2DL1, 2DL2/2DL3, 3DL1,
                                and 3DL2 (lysine-80 of HLA-C; asparagine-80 of HLA-C; the
                                Bw4 motif of HLA-B and HLA-A; and alleles HLA-A*03 and
                                -A*11, respectively). Furthermore, because NK-cell alloreactivity
                                is based on the presence in the allogeneic NK cells of certain inhib-
                                itory KIR, optimal donor selection for transplant or adoptive
                                NK-cell therapy should include KIR genotyping (and, if possible,
                                surface KIR phenotyping by flow cytometry) to ensure that the
                                necessary receptors are included in the donor KIR repertoire.
                                     Independently of the predictable NK-cell alloreactivity deter-
                                mined by mismatch between the HLA epitopes of donors and
                                recipients, some transplant centres have observed that patients
                                transplanted from donors carrying certain combinations of KIR
                                genes have better outcomes than patients who received hemopoi-
                                etic transplants from donors that lacked those genes (6). According
                                to this, KIR genotyping should be part of the protocol for donor
                                selection.
1.2.2. Other Clinical           KIR polymorphisms can influence multiple aspects of human
Conditions                      health, including at least: (a) reproduction and its complications;
                                (b) chronic infection; (c) inflammatory disease and autoimmunity;
                                (d) solid organ transplant; and (e) tumours. In none of these clinical
                                settings is KIR genotyping yet unanimously accepted as a part of
                                decision making for patient care. Further research is needed to
                                understand how KIR participate in the physiopathology of those
                                processes, which should allow for predictive models of clinical out-
                                comes based on the patient KIR genotype, and also for therapies
                                based on a more comprehensive knowledge of NK-cell physiology.
                                In addition, subsequent studies will likely identify new diseases in
                                which risk or clinical course is influenced by the diversity of KIR
                                genotypes.
1.3. KIR Typing Using           The diversity of KIR genotypes was discovered by means of a poly-
a PCR-SSP Method                merase-chain reaction with sequence-specific primers (PCR-SSP)
                                technique that detected distinctive motifs of each KIR gene (7).
                                Different PCR-SSP methods are still most widely used for exploring
                                the genetic diversity of KIR. Here, we provide a detailed protocol for
                                performing a PCR-SSP method for KIR genotyping that amplifies
                                       24   KIR Typing by Non-Sequencing Methods…         417
2. Materials
2.1. Special Equipment    1. Electronic repetitive pipettors (e.g. EDP-1 or -3 from Rainin)
                             can be programmed to dispense multiple aliquots of the desired
                             volume after a single aspiration step. They are of extraordinary
                             help for dispensing primer mixes and liquid wax into typing
                             sets; DNA:buffer:Taq mixes for PCR setup; and the loading
                             dye to processed reactions before electrophoresis.
                          2. Thermal cycler with a 96-well block. We have used models
                             PTC-100 and PTC-200 from MJR, and models 2720 and
                             Veriti 96-Well from Applied Biosystems.
                          3. Electrophoresis system. A horizontal electrophoresis chamber
                             with a tray that accommodates six 17- or 18-well combs enables
                             genotyping six DNAs (96 reactions) in a single run (16 reac-
                             tions and 1 or 2 size markers per row). Moreover, multichan-
                             nel pipette-compatible combs greatly speed up the loading of
                             a full gel (see Notes 1 and 2).
                          4. A multichannel pipettor in the 0.5–10.0 mL range (e.g. CAPP)
                             for gel loading (see Note 3).
2.2. Consumables          1. Strips of eight attached 0.2-mL PCR microtubes are the most
and Reagents                 convenient format. The primer set for typing one DNA will
                             comprise two such strips (16 reactions).
                          2. The oligonucleotides primers used in this method have been
                             described previously (8). Standard quality (desalted) oligonu-
                             cleotides are sufficient.
                          3. Polymerases: regular Taq polymerase is fine; hot start is not
                             necessary (see Note 4). Proof-reading polymerases with 3¢–5¢
                             exonuclease activity must not be used for PCR-SSP.
                          4. Other speciality reagents: Chill-out red Liquid Wax (Bio-Rad)
                             for storage of aliquoted primer mixes; store at room tempera-
                             ture or 4°C.
3. Methods
3.1. Prepare Primer       1. These primer sets (Tables 1 and 2), designed in 2007 (8),
Mixes                        detected all 168 KIR alleles of the KIR-IPD database release
                             1.4.0 (http://www.ebi.ac.uk/ipd/kir/). Current release of
                             the database (2.3.0) includes 614 alleles: 608 are still detected
418       D. Ordóñez et al.
    Table 1
    Chart for preparing primer mixes for KIR genotyping
    Mix Gene        Water (mL) IPC283 (mL)       Name        Volume (mL) Name       Volume (mL)
    1    KIR2DL1    925.0        50              Fa517a      12.5         Rc621     12.5
    2    KIR2DL2    825.0        50              Fcon750     62.5         Rt854     62.5
    3    KIR2DL3    825.0        50              Fcon1254    62.5         Rt1375    62.5
    4    KIR3DL1    762.5        50              Ft624       62.5         Rt697     62.5
                                                 Ftt624      62.5
    5    KIR3DL2    932.5        50              Fg864       12.5         Rc962      5.0
    6    KIR2DS1    762.5        50              Fg621       62.5         Rcon682   62.5
                                                 Fa621b      62.5
    7    KIR2DS2    825.0        50              Fa546       62.5         Rcon621   62.5
                                                         a
    8    KIR2DS3    887.5        50              Ft803       31.25        Ra925     31.25
    9    KIR2DS4    825.0        50              Fat781      62.5         Rca877    62.5
    10   KIR2DS5    825.0        50              Fc551       62.5         Rcon662   62.5
    11   KIR3DS1    762.5        50              Fg624       62.5         Rt697     62.5
                                                 Fg624b      62.5
    12   KIR2DP1    918.8        50              Fc567       15.62        Rdel674   15.62
    13   KIR3DL3    700.0        50              Fg510       62.5         Rta669b 62.5
         KIR3DX1                                 Fma920      62.5         Rdel967b 62.5
    14   KIR2DL4    825.0        50              Ftgc157b    62.5         Rga250b   62.5
    15   KIR2DL5    887.5        50              Fag843      31.25        Rc953     31.25
    16   KIR3DP1    856.3        IPC608: 50 mL   Fi2–89      31.25        Rg287     31.25
                                                 Fa–97b      31.25
a
See Note 5
Table 2
Oligonucleotide primers for KIR typing by PCR-SSP (8)
(primers sequences reproduced with permission from
John Wiley and Sons)
Fa–97b                            gtacgtcaccctcccatgatgta
Fi2–89                            gtgtggtaggagccttag
Ftgc157b                          tcaggacaagcccttctgc
Fg510                             aatgttggtcagatgtcag
Fa517                             gttggtcagatgtcatgtttgaa
Fa546                             tgcacagagaggggaagta
Fc551                             agagaggggacgtttaacc
Fc567                             cgacactttgcacctcac
Fg621                             tctccatcagtcgcatgag
Fa621b                            tctccatcagtcgcatgaa
Fg624                             catcggttccatgatgcg
Fg624b                            catcagttccatgatgcg
Ft624                             ccatyggtcccatgatgct
Ftt624                            tccatcggtcccatgatgtt
Fcon750                           aaaccttctctctcagccca
Fat781                            ggttcaggcaggagagaat
Ft803                             cttgtcctgcagctcct
Fag843                            atctatccagggaggggag
Fg864                             catgaacgtaggctccg
Fma920                            tttctgtgggccgtgcaa
Fcon1254                          agaccctcaggaggtga
Rga250b                           ggacagggaccccatctttc
Rg287                             gaaaacggtgtttcggaatac
Rc621                             cctgccaggtcttgcg
Rcon621                           ccctgcaaggtcttgca
Rcon662                           ctgatagggggagtgagt
Rta669b                           gcygacaactcatagggta
Rdel674                           gggagctgacaactgatg
Rcon682                           ggtcactgggagctgac
                                                      (continued)
420      D. Ordóñez et al.
                                 Table 2
                                 (continued)
3.2. Prepare Sets            A stock of ready-to-use primer mixes, pre-aliquoted into two
of Ready-to-Use              8-tube strips, will allow you to determine quickly the KIR geno-
PCR Primers in Strips        types of any number DNA samples as soon as you receive them.
                             1. Prepare an appropriate number of 0.2 mL, 8-tube strips
                                (e.g. 24–48, see Note 6) and set them in 96-well trays (empty
                                tip cases are handy). Mark the left side of each strip with one
                                dot or line—this tube will hold reaction 1. Mark an identical
                                number of strips with two dots or lines—the labelled tube will
                                be reaction 9.
                              2. Thaw primer mixes 1–8, and homogenise them by inversion.
                              3. Using an electronic repetitive pipettor, dispense 4 mL primer
                                 mix 1 in tube 1 of all strips. Follow with reactions 2–8.
                              4. Using an electronic repetitive pipettor, overlay the mixes with
                                 5–7 mL Chill-out red Liquid Wax (see Note 7 and Fig. 1).
                    24    KIR Typing by Non-Sequencing Methods…                         421
 5. Cap one of every eight strips, stack groups of eight strips, and
    lay them into one or more boxes (again, empty cases of 1,000-
    mL tip racks are handy) (Fig. 2 and video in http://www.
    yotube.com/watch?v=8wU55yu-BgA). You can hold the strips
Fig. 1. A few microliters of liquid wax laid over the aliquoted primer mixes avoid their
evaporation during long-term storage in the freezer.
Fig. 2. Strips of primer mixes, overlaid with liquid wax, stacked for frozen storage.
422      D. Ordóñez et al.
3.3. Prepare PCR             1. If you purchase individual dNTPs, make a 20 mM mix (e.g.
Solutions                       100 mL H2O, and 100 mL each 100 mM dATP, dCTP, dGTP,
                                and dTTP). Store everything at −20°C or below. Do not leave
                                nucleotide-containing solutions at room temperature for long
                                periods.
                             2. Prepare one or more vials of a 2×-concentrated PCR solution
                                (enough for 12 DNAs): add 923 mL H2O, 260 mL 10× Taq
                                polymerase buffer (Mg-free), 104 mL 50 mM MgCl2 (see
                                Note 8), and 13 mL 20 mM dNTPs. Mix well by inversion,
                                make 12 single-use, 100 mL aliquots, and store them at −20°C
                                or below.
3.4. Setup a Typing          1. Thaw the DNAs and a corresponding number of 2× PCR-
Assay                           solution aliquots, and strips with pre-dispensed primer mixes.
                             2. Add 20 mL DNA (containing 0.2–3.0 mg, see Note 9) and
                                1.2 mL Taq (5 U/mL) to a 100 mL aliquot of 2× PCR solution,
                                and mix thoroughly. Prepare as many similar mixes as DNAs
                                you have to analyse (six is a convenient number).
                             3. Using a repetitive pipettor, dispense 6 mL aliquots of this solu-
                                tion onto each primer mix, and cap the strips tightly. Make
                                sure that reagents have fallen to the tube bottoms, transfer
                                the strips to the thermocycler, and start cycling (denaturation at
                                95°C, 2 min; 10 cycles of 94°C, 10 s, and 65°C, 40 s; and 20
                                cycles of 94°C, 20 s, 61°C, 20 s, and 72°C, 30 s).
                             4. Prepare a 2% agarose gel in your preferred electrophoresis
                                buffer.
                             5. When thermocycling has finished, add 10 mL of a 2×-concen-
                                trated loading dye (see Note 10) to the wall of each tube using
                                an electronic repetitive pipettor, and let the dye fall to the bot-
                                tom. The same pipette tip can be used for all reactions.
                             6. Load 10 mL of each reaction in the agarose gel, using an
                                8-channel pipettor (see Notes 11–13 and video in http://
                                www.youtube.com/watch?v=8wU55yu-BgA). Leave an empty
                                well between strips 1 and 2, and load in it ~300 ng of a size
                                marker that covers the range 100–600 bp. Save the remaining
                                reaction volume for KIR2DS4 subtyping (see Note 14) or for
                                possible reanalysis.
                             7. Set the power supply to constant 200 V (or 9 V/cm if your
                                chamber is shorter than 20 cm) with no current limit, and let
                                your samples run 10–15 min (bromophenol blue will migrate
                                ~1.5–2.0 cm).
                                        24   KIR Typing by Non-Sequencing Methods…       423
3.5. Quality Control      1. Contamination tests. Before you use your 2× PCR solution
                             and KIR-typing sets, you should verify that they are not con-
                             taminated with DNA. To that end, run one typing assay in
                             which you substitute 20 mL H2O for the DNA, and make sure
                             that you see no amplification bands (other than possible primer
                             multimers). If you prepare single-use aliquots of all reagents
                             (namely, the 2× PCR solution, and the primer-mix strips), you
                             only have to test periodically that your Taq polymerase is not
                             contaminated. To that end, you can perform a single-tube
                             PCR using the 283 bp IPC primers: these will detect contami-
                             nation with genomic DNA or with any product derived from
                             the 16 reactions of the KIR typing set.
                          2. Reference samples. It is advisable to control that the method is
                             working properly on reference DNAs with known KIR geno-
                             types (9). You can obtain reference DNAs from another lab, or
                             purchase them from a repository of well-characterised cell lines
                             (e.g. http://www.hpacultures.org.uk/collections/ecacc.jsp).
                             In addition, you can test your proficiency by participating in a
                             DNA exchange, such as those organised by UCLA (http://
                             www.hla.ucla.edu/cellDna.htm).
                          Table 3
                          Amplicon sizes (8)
                          1              KIR2DL1                          142
                          2              KIR2DL2                          142
                          3              KIR2DL3                          156
                          4              KIR3DL1                          108/109
                          5              KIR3DL2                          131
                          6              KIR2DS1                           96
                          7              KIR2DS2                          110
                          8              KIR2DS3                          158
                          9              KIR2DS4 (canonical)              133
                                         KIR2DS4 (exon 5-deleted)         111
                          10             KIR2DS5                          147
                          11             KIR3DS1                          107
                          12             KIR2DP1                          141
                          13             KIR3DL3                          196
                                         KIR3DX1                           88
                          14             KIR2DL4                          131
                          15             KIR2DL5                          147
                          16             KIR3DP1 (canonical)              279
                                         KIR3DP1 (exon 2-deleted)         398
                          1–15           Internal positive control        283
                          16             Internal positive control        608
4. Notes
Acknowledgements
References
14. Alves LG, Rajalingam R, Canavez F (2009) A        18. Nong T et al (2007) KIR genotyping by reverse
    novel real-time PCR method for KIR genotyp-           sequence-specific oligonucleotide methodology.
    ing. Tissue Antigens 73:188–191                       Tissue Antigens 69(suppl 1):92–95
15. Thompson A et al (2006) An improved RT-PCR        19. Gonzalez A et al (2009) Killer cell immuno-
    method for the detection of killer-cell immuno-       globulin-like receptor allele discrimination by
    globulin-like receptor (KIR) transcripts.             high-resolution melting. Hum Immunol 70:
    Immunogenetics 58:865–872                             858–863
16. Crum KA et al (2000) Development of a PCR-        20. Witt CS, Martin A, Christiansen FT (2000)
    SSOP approach capable of defining the natural         Detection of KIR2DL4 alleles by sequencing and
    killer cell inhibitory receptor (KIR) gene            SSCP reveals a common allele with a shortened
    sequence repertoires. Tissue Antigens 56:             cytoplasmic tail. Tissue Antigens 56:248–257
    313–326                                           21. Houtchens KA et al (2007) High-throughput
17. Halfpenny IA et al (2004) Investigation of            killer cell immunoglobulin-like receptor geno-
    killer cell immunoglobulin-like receptor gene         typing by MALDI-TOF mass spectrometry
    diversity: IV. KIR3DL1/S1. Hum Immunol                with discovery of novel alleles. Immunogenetics
    65:602–612                                            59:525–537
                                                                                        Chapter 25
Abstract
DNA sequencing is a powerful technique for identifying allelic variation within the natural killer cell
immunoglobulin-like receptor genes. Because of the relatively large size of the KIR genes, each locus is
amplified in two or more overlapping segments. Sanger sequencing of each gene from a preparation
containing one or two alleles yields a sequence that is used to identify the alleles by comparison with a
reference database.
Key words: Natural killer cell, Killer immunoglobulin-like receptor, DNA sequencing, Alleles
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_25, © Springer Science+Business Media New York 2012
                                                         431
432         L. Hou et al.
1.1. Overview                    This protocol describes the amplification and sequencing of each
of Methods                       KIR gene from genomic DNA. The polymerase chain reaction is
                                 used to obtain two or more overlapping amplicons covering all or
                                 most of each gene (Fig. 1). The nucleotide sequences of the exons
                                 carried by each amplicon are determined using Sanger sequencing
                                 (2) with primers that anneal in the introns and flank each exon.
                                 Both alleles of a locus, if present, are sequenced concurrently and
                                 the allele assignments made by comparison to a KIR reference data-
                                 base. Some loci (KIR2DL1, KIR2DL2, KIR2DL3, KIR2DL5,
                                 KIR2DS4) require special steps in order to obtain unambiguous
                                 sequences as described in Table 1. An initial survey of the KIR genes
                                 present or absent in the sample using sequence-specific priming will
                                 provide the information necessary to determine the additional steps
                                 required to obtain allele assignments.
1.2. Use of Methods              The impact of genetic variation in the KIR gene complex on the
in Clinical Practice             functional activity of NK cells is yet to be fully understood. The
                                 presence of specific KIR genes has been associated with susceptibil-
                                 ity or resistance to infectious and autoimmune diseases and to
                                 malignancy (1, 3). In hematopoietic progenitor cell transplanta-
                                 tion for acute myelogenous leukemia, a decreased frequency of
                                 relapse and infection has been noted in transplants with donors
                                 carrying haplotypes with increased numbers of activating KIR
Fig. 1. Amplification of overlapping amplicons covering the KIR2DL1 coding region sequence. KIR genes have eight to nine
exons. PCR amplification primers are designed to generate two or more overlapping amplicons. The figure shows the
three amplicons, A, B, and A2, that cover the coding sequence of the KIR2DL1 gene. If the sample does not contain KIR2DS1,
the laboratory needs only to generate the A and B amplicons for sequencing as described in Table 1. Amplicon A will allow the
sequence determination from nucleotide 11 of exon 1 through nucleotide 632 of exon 5; amplicon B will cover nucleotide 332
in exon 4 through the last nucleotide of exon 9. If the sample contains the KIR2DS1 gene, the laboratory will perform instead
three amplifications generating amplicons A, B, and A2. The A2 amplicon will contain only KIR2DL1 and will provide the
sequence covering nucleotide 1 in exon 1 through nucleotide 330 of exon 4. The A amplicon which contains DNA from both
KIR2DL1 and KIR2DS1 genes will provide sequence information covering the region where the A2 antisense and the B sense
primers anneal, i.e., around nucleotide 331 in exon 4. The small arrows under the exons denote the positions of sequencing
primers that anneal in the introns and that provide the sequence of both sense and antisense DNA strands for each exon.
Tables 2 and 3 list the amplification and sequencing primers for all the KIR loci and describe their annealing sites.
        25    Killer Cell Immunoglobulin-Like Receptors (KIR) Typing by DNA Sequencing           433
Table 1
Summary of amplification protocols for 15 KIR Locia
KIR2DL1      Amplicon A—General PCR in Subheading 3.2 with genomic DNA. If KIR2DS1 is
              present, it will coamplify with this amplicon. When KIR2DS1 is present, amplicon A
              should be characterized to obtain DNA sequence in the area where the antisense A2
              and sense B primers anneal (i.e., in region of nucleotide 331)
             Amplicon A2—General PCR in Subheading 3.2 with genomic DNA. In cells carrying
              KIR2DS1, coamplification of KIR2DS1 is eliminated in this additional reaction. This
              amplification is not required if the cell does not carry KIR2DS1
             Amplicon B—General PCR in Subheading 3.2 with genomic DNA
KIR2DL2      Amplicon A—General PCR in Subheading 3.2 with genomic DNA
             Amplicon B—General PCR in Subheading 3.2 with genomic DNA with the following
              exception. For those cells shown to carry KIR2DL1 or KIR2DS2 or both
              KIR2DL2 and KIR2DL3, use haplotype-specific extraction with probe KIR2DL2-
              999T as described in Subheading 3.4 prior to general PCR in Subheading 3.2 to isolate
              KIR2DL2
             Amplicon C— General PCR in Subheading 3.2 with genomic DNA with the following
              exception. For those cells shown to carry KIR2DL1 or KIR2DS2 or both
              KIR2DL2 and KIR2DL3, use haplotype-specific extraction with probe KIR2DL2-
              999T as described in Subheading 3.4 prior to general PCR in Subheading 3.2
             Amplicon D—If KIR2DL2 and KIR2DL3 are both present in the cell, perform nested
              PCR with these primers using the amplicon B template to eliminate the highly
              homologous KIR2DL3 gene as described in Subheading 3.3
KIR2DL3      Amplicon A—If the cell is KIR2DL2 negative, follow the general PCR protocol in
              Subheading 3.2 beginning with Bc1I digested genomic DNA as described in
              Subheading 3.5. Cleavage of KIR2DP1 with the restriction enzyme BclI eliminates its
              coamplification. If the cell is KIR2DL2 positive, follow the general PCR protocol in
              Subheading 3.2 beginning with haplotype-specific extraction with the KIR2DL3-
              1316T probe as described in Subheading 3.4
             Amplicon B1—If the cell is KIR2DL2 negative, follow the general PCR protocol in
              Subheading 3.2 beginning with the Bc1I digested genomic DNA as described in
              Subheading 3.5. Cleavage of KIR2DP1 with the restriction enzyme BclI eliminates its
              coamplification. If the cell is KIR2DL2 positive, do not prepare the B1 amplicon but
              instead use the amplicon B2 primers described below
             Amplicon B2—If the cell is KIR2DL2 positive, use this primer pair and follow the
              general PCR protocol in Subheading 3.2 beginning with haplotype-specific extraction
              with the KIR2DL3-1316T probe as described in Subheading 3.4. If the cell is
              KIR2DL2 negative, do not prepare the B2 amplicon but instead use the amplicon B1
              primers described above
             Amplicon C1—General PCR in Subheading 3.2 with genomic DNA
             Amplicon C2—General PCR in Subheading 3.2 with genomic DNA. Together, the
              information provided by the C1 and C2 amplicons produces more robust sequence
              results
             Amplicon D—This is a nested PCR of amplicon A required to clarify the sequence in this
              region
KIR2DL4      Amplicon A—General PCR in Subheading 3.2 with genomic DNA
             Amplicon A2—General PCR in Subheading 3.2 with genomic DNA. This amplicon will
              allow characterization of exon 1
             Amplicon B—General PCR in Subheading 3.2 with genomic DNA
                                                                                         (continued)
 Table 1
 (continued)
                          genes (4, 5). Less is known about the impact of KIR allelic
                          polymorphism on the immune response. Allelic variation alters the
                          level of protein expression and the affinity of ligand binding as
                          demonstrated for KIR2DL2/KIR2DL3 (6) and KIR3DL1 (7, 8).
                          For example, in HIV infection, allotypic variation of KIR3DL1
                          influences disease progression and levels of the pathogen in plasma
                          (9). Thus, as we learn more about their impact, identification of
                          KIR alleles may be used to predict the response of an individual to
                          a disease or to therapy and to select optimal stem cell donors for
                          patients with some malignancies.
2. Materials
2.1. DNA Preparation       1. Whole blood drawn into a standard blood tube containing the
                              anti-coagulant acid citrate dextrose (ACD) (see Note 1).
                           2. QIAamp® DNA Blood Mini Kit (QIAGEN, Valencia, CA,
                              USA): The kit contains buffers AL, AW1, AW2, protease
                              and solvent for protease, spin columns, collection tubes, and
                              instruction manual. The buffers in the kit, AW1 and AW2, are
                              provided as concentrates. When opening a new bottle, add
                              the appropriate amount of 96–100% ethanol (as written on the
                              label). To reconstitute the protease, add the supplied solvent
                              to the protease powder and invert the bottle several times to
                              mix. Store for 2 months at 4°C after preparation.
                           3. 96–100% ethanol.
                           4. Phosphate buffered saline (PBS).
                           5. 1.5 mL microcentrifuge tubes.
                           6. Pipettor (5–200 mL) and tips.
                           7. Heat block or water bath at 56°C.
                           8. Vortex mixer.
                           9. Centrifuge capable of holding 1.5 mL tubes with a maximum
                              speed of 20,000 × g (14,000 rpm).
                                                               Annealing
                                                               sites: sense/   Amplicon PCR reaction         PCR reaction components
                                                               antisenseb      size (bp) conditions          (50 mL)
                                                                                        Annealing
                                                                                        temp
                                                                                        (°C)—initial/                 DMSO/
                                                                                        secondary Extension MgSO4 Betaine
KIR locus Amplicon Sense primer         Antisense primer                                cycles        time (min) (mL) (mL)     Taq
KIR2DL1 A          TGTAAAACGACGGC       CAAGCAGTGGG            10T/633G        5,605    64/61          5     2.0    2.5/−      High
                    CAGTGGCAGCAC         TCACTTGAC                                                                              Fidelity
                    CATGTCGCTCT
         A2        ATAACATCCTG          GGGTCACTGGG            5UTR/331G       3,825    66/64          5     1.5    3.0/−      High
                     TGCGCTGCT           AGCTGACAC                                                                              Fidelity
         B         ACTCACTCCC           TGTTGACTCCC            331G/3UTR 10,282         62/59      10        2.0    −/−        High
                    CCTATCAGG            TAGAAGACG                                                                              Fidelity
                                                       Annealing
                                                       sites: sense/   Amplicon PCR reaction         PCR reaction components
                                                       antisenseb      size (bp) conditions          (50 mL)
                                                                                Annealing
                                                                                temp
                                                                                (°C)—initial/                 DMSO/
                                                                                secondary Extension MgSO4 Betaine
KIR locus Amplicon Sense primer     Antisense primer                            cycles        time (min) (mL) (mL)    Taq
KIR2DL3 A          TGTAAAACGACGGC   GCCCTGCAGAG        10A/505C         5,385   62/58      5         3.0    −/10       High
                    CAGTGGCAGCAC     AACCTACG                                                                           Fidelity
                    CATGTCGCTCA
         B1        GTTCTGTTACTC     CTCTCCTCTG         325T/IN5         2,131   62/58      5         3.0    −/10       High
                    ACTCCCCCT        GGTCTCTCC                                                                          Fidelity
                                     TGACCG
         B2        CGTTCTGCACAG                        194A/IN5         2,262   62/58      5         3.0    −/10       High
                    AGAAGGGAAc                                                                                          Fidelity
         C1        TCAAGACAGTGGG    CTTCGTGAGAC        IN6/809G         3,344   62/58      5         3.0    −/10       High
                    CGTCACATACA      TTACTTT TTT                                                                        Fidelity
                                     TGTTGC
         C2        ACACCTGCATGT     GCAGGAGACAAC       746G/1024T        879    62/58      5         3.0    −/10       High
                    TCTGATTGG        TTTGGATCA                                                                          Fidelity
         D         AGCAAGGGGAAGCC   CCAATGACAA         IN2/IN4           419    62/57      1.5       -      −/10       Taq/10×
                    TCACTCATTC       TGAGAATG                                                                            PCR
                                                       Nested Ex4
                                                                                                                         buffer
                                                                                                                         with
                                                                                                                         MgCl2
KIR2DL4 A       CACCCACGGTC      CCCTTTCSCTG      28C/IN6     5,378    64/57   6    2.0   2.0/−   High
                 ATCATCC          TTGGAGTGT                                                        Fidelity
       A2       TCCTGGCAGCAG     GGAAAGAGCC       5UTR/581G   2,564    64/57   5    2.0   2.0/−   High
                 AAGCTGCACC       GAAGCATC                                                         Fidelity
       B        CATGTTCTAGG      TGGGCTAAGCA      666T/3UTR   5,420    64/57   6    2.0   2.0/−   High
                 AAACCCTTCT       AAGGAGTGT                                                        Fidelity
KIR2DL5 A       ATCTTGTGTTC      TCATAGGGTGA      5UTR/589C   3,274    64/62   5    2.0   2.0/−   High
                  GGGAGGTTG       GTCATGGAG                                                        Fidelity
       B        GAGGGGAGGG       GGAAGAGCGAT      491C/3UTR   6,193    64/62   7    2.0   2.0/−   High
                 CCCATGAACC       CCCCTAAGA                                                        Fidelity
       A*001+   CTCCCGTGATGTGG   TCATAGGGTG       5UTR/589C   3,109    64/62   5    2.0   2.0/−   High
                 TCAACATGTAAA     AGTCATGGAG                                                       Fidelity
       B*002+   CTCCCATGATGTA    TCATAGGGTG       5UTR/589C   3,109    64/62   5    2.0   2.0/−   High
                 GTCAACATGTAAG    AGTCATGGAG                                                       Fidelity
KIR2DS1 A       GGCAGCACCAT      GCATCTGTAGG      10A/576T    5,540    64/60   7    1.5   1.0/−   High
                 GTCGCTCA         TCCCTCCA                                                         Fidelity
       B        TCTCCATCAGT      GGGTGTCTTG       272R/3UTR 10,227     64/60   10   2.0   1.0/−   High
                 CGCATGAR         GGCCTCTC                                                         Fidelity
KIR2DS2 A       ATCCTGTGCGCTG    CACGCTCTCT       5UTR/418T   5,239    62/58   7    1.5   2.0/−   High
                  CTGAGCTGAG      CCTGCCAA                                                         Fidelity
       B        CTTCTGCACAGAGA   TTATGCGTATGACA   197A/893A   10,253   62/58   10   1.5   2.0/−   High
                 GGGGAAGTA         CCTCCTGAT                                                       Fidelity
                                                                                                  (continued)
Table 2
(continued)
                                                       Annealing
                                                       sites: sense/   Amplicon PCR reaction         PCR reaction components
                                                       antisenseb      size (bp) conditions          (50 mL)
                                                                                Annealing
                                                                                temp
                                                                                (°C)—initial/                 DMSO/
                                                                                secondary Extension MgSO4 Betaine
KIR locus Amplicon Sense primer     Antisense primer                            cycles        time (min) (mL) (mL)    Taq
KIR2DS3 A          ATCCTGTGCGCTGC   GCATCTGTAG         5UTR/576A        5,919   64/61          7     2.0    −/−        High
                     TGAGCTGAG       GTTCCTCCT                                                                          Fidelity
         B         GACATGTAC        TTATGCGTATGAC      485C/888G        8,427   60/57      10        2.0    −/−        High
                    CATCTATCCAC       ACCTCCTGA                                                                         Fidelity
                                      TGGTCC
KIR2DS4 A          CATGTCGCTCA      ACACTCTCACCT       20T/360G         5,122   64/58          7     2.0    −/−        High
                    TGGTCATCAT       ATGATCACC                                                                          Fidelity
         B         ATCCTGCAA        TTATGCGTATGACAC    153G/893A       10,299   64/58      10        1.5    −/−        High
                     TGTTGGTCG        CTCCTGAT                                                                          Fidelity
         C         CGCAGTGACCC      GTGACGGAAACAA      360G/642T        1,875   62/57          1.5   -      −/10       Taq/10×
                    TCTGGACATGc      GCAGTGGA                                                                            PCR
                                                       Nested Ex 5
                                                                                                                         buffer
                                                                                                                         with
                                                                                                                         MgCl2
KIR2DS5 A          CCATCATGATCTT    CCTCCGTGGGTG       35C/563A         4,541   62/58          5     2.0    −/−        High
                    TCTTTCCAGC       GCAGGGT                                                                            Fidelity
         B         CATTGATGGGGT     TTATGCGTATGACACC   248G/888G       10,188   62/58      10        2.0    −/−        High
                    CTCCAAGGG         TCCTGATGGTCC                                                                      Fidelity
    KIR3DL1 A              TGTCKRCACC                   TAGGTCCCTGC                     5UTR/560T          3,454     60/57           5           1.5       2.0/−        High
                            GGCAGCACC                     AAGGGCAA                                                                                                       Fidelity
              B            CCATCGGTCC                   GACAACTTTGGA                    560T/1303Y 10,365            60/57         11            -         −/−          Expand
                            CATGATGCT                    TCTGGGCTY                                                                                                        Long/
                                                                                                                                                                          Buffer 3
              M            CAARCCCTTCCT                 GAGAGAGAAG                      100T/659C          3,265     60/57           5           1.5       2.0/−        High
                            GTCTGCCT                     GTTTCTCATATG                                                                                                    Fidelity
    KIR3DL2 A              GTCGTCAGCA                   TGCATCCAAGG                     30C/IN6            8,706     60/57           8           1.5       2.0/−        High
                            TGGCGTGC                     CTTCCACC                                                                                                        Fidelity
              A2           TGTCTGCACCG                  GACCACACG                       5UTR/898C          5,421     60/57           5           2.0       −/10         High
                            GCAGCACC                     CAGGGCAG                                                                                                        Fidelity
              B            TCACATCTC                    GGCTGTTGTC                      IN5/1362T          7,693     60/57           8           1.5       2.0/−        High
                            TCCTGTCCCG                   TCCCTAGAAA                                                                                                      Fidelity
    KIR3DL3 A              TTTCCAGGGTTCT                TGACCCTCAG                      49G/799A           4,415     62/60           5           3.0       2.0/−        High
                            TCTTGCTGG                    CACYGCAGT                                                                                                       Fidelity
              A2           TGTCTGCACCG                  CCGACAACTC                      5UTR/605T          3,361     62/60           5           3.0       −/10         High
                            GCAGCACC                     ATAGGGTA                                                                                                        Fidelity
              B            CCCGGAGCTTG                  AGAAGACAAC                      756T/3UTR          6,569     58/54           7           3.0       −/−          High
                            TTTGACATT                    TTTGGATCTGC                                                                                                     Fidelity
    KIR3DS1 A              TGTCKRCACCG                  CTGTGACCAT                      5UTR/A337          2,116     60/57           3           1.0       1.5/−        High
                            GCAGCACC                     GATCACCAT                                                                                                       Fidelity
              B            GGCAGAATAT                   AGAGCGATGCC                     235G/3UTR 12,324             60/57         11            -         −/−          Expand
                            TCCAGGAGG                    CTAAGATGA                                                                                                        Long/
                                                                                                                                                                          Buffer 3
a
 Some of the primers have been previously described (19–23)
b
  UTR, untranslated region and/or other 5¢ or 3¢ noncoding sequences; IN, intron. The designations such as 10T/633G indicate the nucleotide at the annealing site of the 3¢ end of the
sense/antisense primers. Position 1 is defined as the first nucleotide of the ATG codon in exon 1 according to the IPD/KIR database (http://www.ebi.ac.uk/ipd/kir/). The numbering of
KIR2DS4 is based on an allele that does not contain the deletion
c
 Primer sequence is not identical to KIR gene sequence; a substitution was added to avoid the primer from self-annealing
442      L. Hou et al.
2.5. Restriction         1. Genomic DNA from cells carrying KIR2DL3. Table 1 describes
Enzyme Digestion for        the use of restriction enzyme digestion to eliminate a highly
the KIR2DL3 Locus           homologous gene when present in the sample.
                         2. Restriction endonuclease BclI (15 U/mL) and 10× NE Buffer
                            3 (New England BioLabs, Ipswich, MA, USA).
                         3. Reagent grade water.
                         4. Phenol:chloroform:isoamyl alcohol 25:24:1,V/V/V (e.g.,
                            UltraPure™ phenol:chloroform:isoamyl alcohol, Invitrogen)
                            (see Note 7).
                         5. 3M sodium acetate (Sigma-Aldrich).
                         6. 70% ethanol in water (Warner-Graham Company) at −20°C.
                         7. Heating block at 50°C.
           25      Killer Cell Immunoglobulin-Like Receptors (KIR) Typing by DNA Sequencing       443
                               8. −20°C freezer.
                               9. Supplies and equipment described in Subheading 2.2.
2.7. Long Template             1. Genomic DNA from samples carrying KIR3DL1 or KIR3DS1.
PCR for KIR3DL1/                  Table 1 summarizes the strategies used to obtain amplicons for
KIR3DS1 Amplicon B                specific KIR genes.
                               2. Expand Long Template PCR System with Taq DNA poly-
                                  merase and 10× Expand Long Template buffer 3 (Roche,
                                  Mannheim, Germany).
                               3. 10 mM dNTP mixture (Roche, Mannheim, Germany).
                               4. KIR locus PCR primer solutions for KIR3DL1 and KIR3DS1
                                  B amplicons: 10 mM of each oligonucleotide primer in water.
                                  Primers are listed in Table 2.
                               5. Reagent grade water.
                               6. Supplies and equipment described in Subheading 2.2.
2.8. DNA Sequencing            1. Amplified DNA purified with AMPure from Subheading 3.2.
                               2. BigDye Terminator v1.1 diluted 1:1 with 5× sequencing buf-
                                  fer (Applied Biosystems, Foster City, CA, USA).
                               3. KIR locus sequencing primers: 1.5 mM of each oligonucleotide
                                  primer in water. Store at −20°C (see Table 3) (see Note 8).
                               4. DMSO.
                               5. Agencourt CleanSEQ kit (Beckman Coulter, Beverly, MA,
                                  USA).
                                                                                                                       444
Table 3
DNA sequencing primers for KIR loci
                                                                                             (continued)
                                                                                                           445
                                                                                                                     446
Table 3
(continued)
Table 3
(continued)
                                                                                                                  L. Hou et al.
Table 3
(continued)
2.9. Sequence Analysis    1. Analysis software: Assign SBT 3.2.7 (Conexio Genomics,
Including Preparation        Applecross, Western Australia), HLA Librarian (Conexio
of Locus-Specific KIR        Genomics), Sequencher 4.6 (Ann Arbor, MI, USA) with
Libraries                    manuals (see Note 9).
                          2. KIR nucleotide sequence databases: IPD-KIR curated coding
                             region sequence database at http://www.ebi.ac.uk/ipd/kir/
                             index.htmL; leukocyte receptor complex (LRC) database
                             alignment viewer for genomic sequences at http://www.ncbi.
                             nlm.nih.gov/gv/lrc/.
3. Methods
3.1. DNA Preparation      1. Label the appropriate number of 1.5 mL microcentrifuge tubes
                             and QIAamp spin columns with sample identifier (see Note 10
                             on laboratory).
                          2. Add 200 mL whole blood sample to the tube (see Note 11). If
                             the sample volume is less than 200 mL, add PBS to bring
                             sample to volume.
                          3. Pipet 20 mL protease into the blood sample in the tube.
                          4. Add 200 mL buffer AL to the sample (see Note 12). Immediately
                             mix by vortexing for 15 s.
                          5. Incubate at 56°C for 10 min.
                          6. Briefly centrifuge the microcentrifuge tube to remove conden-
                             sation drops from the inside of the lid (see Note 13).
                          7. Add 200 mL 96–100% ethanol to the sample and mix again by
                             vortexing for 15 s. Again briefly centrifuge the microcentrifuge
                             tube.
          25   Killer Cell Immunoglobulin-Like Receptors (KIR) Typing by DNA Sequencing     453
3.2. Polymerase Chain      1. See Table 1 for a listing of those KIR loci that should be
Reaction Amplification        amplified following this protocol (see Note 16).
of Individual KIR Loci:    2. Thaw 10× High Fidelity PCR buffer, 50 mM MgSO4, dNTP
General                       mix, primer solutions, and DMSO or 5M betaine solution (see
                              Note 17). Mix the solutions thoroughly before use.
                           3. Prepare the reaction mix in a 1.5-mL tube as described in
                              Table 4.
                           4. Vortex the reaction mix and dispense 45 mL volumes into each
                              well of a semi-skirted PCR tray.
                           5. Add 5 mL of genomic DNA (50–200 ng), purified as described
                              in Subheading 3.1, to each well containing reaction mix (see
                              Note 18).
                           6. Set up positive and negative amplification control wells. The
                              positive control for each primer pair is 5 mL DNA (50–200 ng)
                              from a cell carrying that KIR locus. The negative control for
                              each primer pair is 5 mL DNA (50–200 ng) from a cell lacking
                              that KIR gene. For primers amplifying framework genes
                              (KIR2DL4, KIR3DL2, and KIR3DL3), use 5 mL water as a
                              negative control instead of DNA.
                           7. Place tape seal over entire tray and quick spin the plate in the
                              centrifuge to ensure all the liquid is at the bottom of the wells.
                              Place in the thermal cycler.
454        L. Hou et al.
 Table 4
 Composition of reaction master mix for Platinum Taq DNA Polymerase High Fidelity
 Table 5
 Polymerase chain reaction amplification conditions
                                                                                  Long Template
                        General PCR conditions              Nested PCR (see       PCR (see
                        (see Subheading 3.2)                Subheading 3.3)       Subheading 3.7)
 Denaturation           95°C for 5 min                      92°C for 4 min        92°C for 2 min
 Initial cycles         10 cycles                           10 cycles:            10 cycles
                         95°C for 20 s                       92°C for 45 s        92°C for 10 s
                         58–66°C for 30 s (see Table 2)      62°C for 45 s        60°C for 30 s
                         68°C for 3–10 min (see Table 2)     72°C for 1.5 min     68°C for 11 min
 Secondary cycles       30 cycles                           30 cycles             30 cycles
                         95°C for 20 s                       92°C for 45 s        92°C for 15 s
                         52–64oC for 30 s (see Table 2)      57°C for 45 s        57°C for 30 s
                         68°C for 3–10 min (see Table 2)     72°C for 1.5 min     68°C for 11 min
 Final extension        68°C for 10 min                     72°C for 10 min       68°C for 10 min
 Final hold             4°C                                 4°C                   4°C
                              14. With the PCR plate on the magnet, aspirate the cleared solu-
                                  tion with a pipet and discard.
                              15. Keeping the PCR plate on the magnet, dispense 200 mL of 70%
                                  ethanol to each well. Allow to sit at least 30 s at room tempera-
                                  ture. Aspirate the wash solution with a pipet, discard and repeat.
                                  Be sure to remove as much ethanol as possible to shorten the
                                  drying time. Dry at room temperature for 10 min.
                              16. To elute the purified DNA, add 30–50 mL (see Note 22) of
                                  reagent grade water to each well and mix well by pipetting up
                                  and down. Place the plate back on the magnet.
                              17. Remove the eluate containing the amplified DNA to a clean
                                  96-well plate to begin the DNA sequencing reactions (see
                                  Subheading 3.8).
3.3. Nested PCR for            1. See Table 1 for a listing of those KIR loci that should be
KIR2DL2 Amplicon B,               amplified following this protocol.
KIR2DL3 Amplicon A,            2. Thaw Taq DNA Polymerase, 10× PCR buffer with MgCl2,
and KIR2DS4                       dNTP mix, 5M betaine solution, and appropriate primer solu-
Amplicon B                        tions (see Table 2). Mix the solutions thoroughly before use
                                  (see Note 23).
                               3. Prepare the nested PCR reaction master mix as shown in
                                  Table 6.
                               4. Aliquot 45 mL of master mix into each well of a semi-skirted
                                  PCR tray.
456     L. Hou et al.
                            Table 6
                            Composition of reaction master mix for nested polymerase
                            chain reaction amplification
                               Table 7
                               HaploPrep reaction master mix
                               Hybridization buffer H                    15 mL
                               HaploPrep Extraction Probe                2 mL
                                2DL2-999T or 2DL3-1316T
                               Water                                     8 mL
                               Genomic DNA                               5 mL (added at a later step)
                              a
                               The volume for a single reaction is 25 mL without the DNA added so multiply
                              the number of reactions desired by 25 to determine how much reaction
                              master mix to make. Always make more than you need to account for losses
                              during pipetting
3.5. Isolation            1. This protocol is performed for some cells carrying KIR2DL3
of KIR2DL3 Locus:            as described in Table 1.
Restriction Enzyme        2. Prepare the restriction enzyme reaction mix according to
Digestion                    Table 8.
                          3. Mix the reaction thoroughly and dispense indicated volume
                             from Table 8 into a 1.5-mL tube.
                          4. Add 20 mL genomic DNA (approximately 2 mg) to each tube
                             containing reaction mix. Incubate at 50°C for 1 h.
458      L. Hou et al.
                                       Table 8
                                       Restriction enzyme reaction master mix
                                       10× NE Buffer 3         20 mL
                                       Bc1I                    3 mL
                                       Genomic DNA             20 mL (added at a later step)
                                       Water                   Bring final volume excluding
                                                                 DNA to 180 mL
                                   a
                                    The volume for a single reaction is 200 mL so multiple the
                                   number of digestion reactions desired by 200 to determine
                                   how much reaction master mix to make. Always make more than
                                   you need to account for losses during pipetting. If you need
                                   to add more or less volume of DNA to the reaction mix,
                                   adjust volume of water so that the final volume for each sample
                                   is 200 mL
3.6. KIR2DS4 Allele       1. Cloning is required only for PCR amplicons containing both a
Isolation by Cloning         full-length allele and an allele with a deletion (see Note 29).
                             Prepare a nested KIR2DS4 amplicon by PCR as described in
                             Subheading 3.3.
                          2. Verify amplified products on a 1.5% agarose gel with 1Kb DNA
                             ladder as described in Subheading 3.2, step 9.
          25   Killer Cell Immunoglobulin-Like Receptors (KIR) Typing by DNA Sequencing   459
3.7. Long Template         1. Amplification of long segments of DNA from KIR3DL1 and
PCR for KIR3DL1 B             KIR3DL2 will require this protocol (see Table 1). Thaw 10×
and KIR3DS1 B                 Expand Long Template buffer 3, dNTP mix, and primer solu-
Amplicons                     tions for KIR3DL1 B and KIR3S1 B amplicons (see Table 2).
                              Vortex the solutions thoroughly before use (see Note 32).
                           2. Assemble the reaction mix for the Expand Long Template
                              PCR System as described in Table 9.
                           3. Vortex the reaction mix thoroughly and dispense 45 mL vol-
                              umes into each well of semi-skirted PCR tray.
                           4. Add 5 mL template DNA (100–200 ng) to each well contain-
                              ing reaction mix (see Note 33).
460     L. Hou et al.
                            Table 9
                            Composition of reaction master mix for Expand Long
                            Template PCR reaction
3.8. DNA Sequencing     1. Sequence the amplicons using KIR loci sequencing primers
                           (see Table 3). For each locus, both sense and antisense primers
                           are used to cover the complete sequence of the exons (Fig. 1)
                           (see Note 34).
                        2. To each well, add 2 mL of diluted Big Dye Terminator, 1 mL of
                           the appropriate primer (see Table 3), and 3 mL of the purified
                           PCR product. For exon 1 sequences for all KIR loci, add
                           0.3 mL DMSO to the reaction (see Note 35).
                        3. Place tape seal over entire tray and quick spin the plate in the
                           centrifuge to ensure all liquid is at the bottom of the wells.
                           Place in the thermal cycler.
          25   Killer Cell Immunoglobulin-Like Receptors (KIR) Typing by DNA Sequencing    461
                                        Table 10
                                        DNA sequencing reaction conditions
                                        30 cycles             30 cycles
                                        96°C for 10 s         96°C for 10 s
                                        50°C for 5 s          60°C for 1 min
                                        60°C for 4 min        Hold at 4°C
                                        Hold at 4°C
3.9. Sequence Analysis     1. Locus-specific KIR libraries must be created prior to analysis of
Including Preparation         KIR sequencing data. Go to the IPD-KIR database downloads
of Locus-Specific             and open up the FTP directory. Obtain the nucleotide coding
KIR Libraries                 region sequences of all known alleles at each KIR locus as nuc.
                              fasta files (e.g., KIR2DL1_nuc.fasta; one file for each locus).
                              Create two separate libraries for KIR2DS4, one library with
                              the full-length allele sequences and a second library with the
462        L. Hou et al.
4. Notes
                      13. The speed of the quick spin should be above 1,000 rpm. Set
                          the speed to 8,000 rpm; press the button for 5 s and release to
                          achieve this speed.
                      14. DNA should be stored in a neutral to slightly basic buffered
                          solution to prevent degradation. Tris–EDTA (TE) buffer can
                          be used for storage. TE contains EDTA which has a high
                          affinity towards divalent ions like Ca+2 and Mg+2. These ions
                          are cofactors for many enzymes including nucleases that digest
                          DNA molecules. Since repeated access to a tube of genomic
                          DNA may introduce nucleases, TE buffer will protect DNA
                          from degradation during long-term storage. However, since
                          EDTA can bind divalent ions, it can inhibit Taq polymerase in
                          the PCR reaction. If DNA is stored in deionized water which
                          is often at an acidic pH, DNA degradation can occur by acid
                          hydrolysis.
                      15. Refer to the QIAamp® DNA Mini Kit handbook for trouble-
                          shooting problems.
                      16. It is helpful to initially assay for the presence or absence of KIR
                          genes using a sequence-specific priming assay as described in
                          Chap. 24. This will facilitate the selection of protocols to use
                          to isolate KIR genes for sequencing as described in Table 1.
                          Methods described in this chapter have been published (10–13)
                          (Hou, in preparation).
                          Some KIR haplotypes include fusion genes. For example,
                          KIR3DL1/KIR3DL2 hybrid alleles have been found in popu-
                          lations of recent African origin (13, 14). These alleles carry the
                          first five exons of KIR3DL1 and exons 6–9 of KIR3DL2. The
                          KIR3DL1 primer pairs in this protocol will amplify this chime-
                          ric gene. When sequencing amplicon B of KIR2DL4, be alert
                          for a single nucleotide deletion that removes the last nucle-
                          otide (811) of exon 7 in some alleles (e.g., KIR2DL4*008).
                          When sequencing KIR2DL5, it is possible that a cell may carry
                          three or four alleles, i.e., two alleles of KIR2DL5A and two
                          alleles of KIR2DL5B are potentially possible. An additional
                          two primer pairs listed in Table 1 will assist in clarifying the
                          allele calls in this situation. These pairs are each specific for a
                          subset of KIR2DL5 alleles. Sequencing primers used with
                          KIR2DL5 amplicon A will anneal to these two amplicons.
                      17. The polymerase and buffer used in the PCR reaction vary for
                          different loci and are described in Table 2. DMSO or 5M
                          betaine solution can improve and enhance the specificity of the
                          polymerase chain reaction. The volumes in each reaction of
                          MgSO4, DMSO, and 5M betaine solution are provided in
                          Table 2.
                      18. It is critical to have high-quality DNA for the PCR reaction. To
                          quantify the DNA and to determine its purity, read its optical
25   Killer Cell Immunoglobulin-Like Receptors (KIR) Typing by DNA Sequencing     465
References
 1. Bashirova AA et al (2006) The killer                    13. Jiang B et al (2010) The profile of KIR3DL1
    immunoglobulin-like receptor gene cluster:                  and KIR3DS1 alleles in an African American
    tuning the genome for Defense. Annu Rev                     population resembles that found in African
    Genomics Hum Genet 7:277–300                                populations. Tissue Antigens 76:64–66
 2. Sanger F, Nicklen S, Coulson AR (1977) DNA              14. Norman PJ et al (2009) Meiotic recombina-
    sequencing with chain-terminating inhibitors.               tion generates rich diversity in NK cell receptor
    Proc Natl Acad Sci USA 74:5463–5467                         genes, alleles, and haplotypes. Genome Res
 3. Khakoo SI, Carrington M (2006) KIR and dis-                 19:757–769
    ease: a model system or system of models?               15. Vilches C, Gardiner CM, Parham P (2000)
    Immunol Rev 214:186–201                                     Gene structure and promoter variation of
 4. Tomblyn M et al (2010) Decreased infections                 expressed and nonexpressed variants of the
    in recipients of unrelated donor hematopoietic              KIR2DL5 gene. J Immunol 165:6416–6421
    cell transplantation from donors with an acti-          16. Ordonez D et al (2008) Duplication, mutation
    vating KIR genotype. Biol Blood Marrow                      and recombination of the human orphan gene
    Transplant 16:1155–1161                                     KIR2DS3 contribute to the diversity of KIR
 5. Cooley S et al (2010) Donor selection for natu-             haplotypes. Genes Immun 9:431–437
    ral killer cell receptor genes leads to superior sur-   17. Gomez-Lozano N et al (2005) The silent
    vival after unrelated transplantation for acute             KIR3DP1 gene (CD158c) is transcribed and
    myelogenous leukemia. Blood 116:2411–2419                   might encode a secreted receptor in a minority
 6. Moesta AK et al (2008) Synergistic polymor-                 of humans, in whom the KIR3DP1, KIR2DL4
    phism at two positions distal to the ligand-                and KIR3DL1/KIR3DS1 genes are dupli-
    binding site makes KIR2DL2 a stronger                       cated. Eur J Immunol 35:16–24
    receptor for HLA-C than KIR2DL3.                        18. Martin MP et al (2003) Cutting edge: expan-
    J Immunol 180:3969–3979                                     sion of the KIR locus by unequal crossing over.
 7. Yawata M et al (2008) MHC class I-specific                  J Immunol 171:2192–2195
    inhibitory receptors and their ligands structure        19. Uhrberg M et al (1997) Human diversity in
    diverse human NK-cell repertoires toward a                  killer cell inhibitory receptor genes. Immunity
    balance of missing self-response. Blood                     7:753–763
    112:2369–2380                                           20. Vilches C et al (2007) Facilitation of KIR geno-
 8. Sharma D et al (2009) Dimorphic motifs in D0                typing by a PCR-SSP method that amplifies
    and D1 + D2 domains of killer cell Ig-like recep-           short DNA fragments. Tissue Antigens
    tor 3DL1 combine to form receptors with                     70:415–422
    high, moderate, and no avidity for the complex          21. Gomez-Lozano N, Vilches C (2002) Geno-
    of a peptide derived from HIV and HLA-                      typing of human killer-cell immunoglobulin-
    A*2402. J Immunol 183:4569–4582                             like receptor genes by polymerase chain reaction
 9. Martin MP et al (2007) Innate partnership of                with sequence-specific primers: an update.
    HLA-B and KIR3DL1 subtypes against HIV-                     Tissue Antigens 59:184–193
    1. Nat Genet 39:733–740                                 22. Murdoch S et al (2006) Detailed gene and
10. Hou L et al (2010) African Americans exhibit a              allele content analysis of three homozygous
    predominant allele in the midst of extensive                KIR haplotypes. Tissue Antigens 68:72–77
    KIR2DL1 allelic diversity. Tissue Antigens              23. Sun JY, Oki A, Senitzer D (2008) Alleles and
    76:31–34                                                    intron polymorphism of KIR3DL1 shown
11. Hou L et al (2010) Thirty allele-level haplo-               by combination of allele group-specific prim-
    types centered around KIR2DL5 define the                    ers and sequencing. Tissue Antigens 72:
    diversity in an African American population.                578–580
    Immunogenetics 62:491–498                               24. Vilches C, Pando MJ, Parham P (2000) Genes
12. Hou L et al (2009) In contrast to other stimu-              encoding human killer-cell Ig-like receptors
    latory natural killer cell immunoglobulin-like              with D1 and D2 extracellular domains all con-
    receptor loci, several KIR2DS5 alleles predom-              tain untranslated pseudoexons encoding
    inate in African Americans. Hum Immunol                     a third Ig-like domain. Immunogenetics
    70:733–737                                                  51:639–646
                                                                                        Chapter 26
Abstract
Donor-vs.-recipient NK cell alloreactivity has been established as a key therapeutic element in HLA
haplotypemismatched hematopoietic transplants in acute myeloid leukemia. NK cell allotherapy for leukemia
is deployed through stem cell transplantation and ensuing NK cell reconstitution across KIR ligand
mismatches. It is effected by functional NK cells which express inhibitory killer cell immunoglobulin-like
receptor(s) (KIRs) for self-class I ligand(s), sense missing expression of donor KIR ligand(s) in the recipient,
and mediate alloreactions. Donor-vs.-recipient NK cell alloreactivity is evaluated by integrating genetic,
phenotypic, and functional features.
     Key words: HLA, KIR, Alloreactivity, Cytotoxicity, Acute leukemia, Haploidentical hematopoietic
     transplantation
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_26, © Springer Science+Business Media New York 2012
                                                         469
470   A. Velardi
Fig. 1. Posttransplant regeneration of donor-vs.-recipient alloreactive NK cell repertoires. Top: absolute numbers of
potentially alloreactive NK cells (as evaluated by immune-fluorescence), i.e., of NK cells that express, as their only inhibi-
tory receptor for self, the KIR for which there is no ligand in the recipient (see Subheading 2.3.4). Bottom: frequencies of
alloreactive NK clones across each one of the three KIR ligand mismatches (C1, C2, Bw4), as documented by limiting
dilution cloning and cytotoxicity assays (see Subheading 2.3.5).
2. Methods
and Key Reagents
2.1. Haploidentical              Conditioning of the recipient consists of 8 Gy total-body irradia-
Donor HSCT                       tion on day 9 in a single fraction at an instantaneous dose-rate of
Transplantation                  0.16 Gy/min; lungs shielded to receive 0.04 Gy; thiotepa (5 mg/
Procedures                       kg daily) on days 8 and 7; fludarabine (40 mg/m2 daily) from day
                                 7 to day 3; rabbit anti-thymocyte globulin (ATG) is given at 5 mg/
                                 kg daily from days 5 to 2 (4, 8, 10). No immune suppression is
                                 given after transplantation as GvHD prophylaxis; no G-CSF is
                                 administered posttransplantation.
2.2. Graft Processing            Peripheral-blood hematopoietic progenitor cells from the donor
                                 are mobilized and collected as previously described (4, 8, 10).
                                 CD34 positive cells are selected using the CliniMacs one-step pro-
                                 cedure (Miltenyi Biotech, Bergisch Gladbach, Germany). Flow
                                 cytometric analysis quantifies CD34, CD3, and CD20 positive
                                 cells before and after selection. G-CSF stem cell mobilization and
                                 graft processing. The stem cell “megadose” (i.e., 10 × 106 cells/kg
                                 body weight) is routinely achieved by administering a short course
                                 of granulocyte-colony stimulating factor (G-CSF) to the donor,
                                 followed by leukapheresis and immunomagnetic-based CD34+
                                 stem cell selection (see Note 1).
472         A. Velardi
2.3.2. Identification         Candidates are assessed for HLA compatibility by HLA typing at
of Suitable Donor-Recipient   HLA-A, -B, -C, -DR, -DQ, and -DP (see elsewhere in this volume
Pairs                         for detailed methods). First, the recipient is HLA typed. Those
                              who express class I alleles belonging to all three class I groups
                              recognized by KIRs (HLA-C group 1, HLA-C group 2, and
                              HLA-Bw4 alleles) will block all NK cells from every donor and will
                              be resistant to alloreactive NK killing (approximately 1/3 of the
                              population). Patients who express only one or two of these allele
                              groups may find NK alloreactive donors. HLA typing on the family
                              will identify those haploidentical family member(s) who expresses
                              the class I group(s) missing in the recipient and who have, there-
                              fore, the potential to be a donor whose NK cells exert alloreactivity
                              against the recipient.
2.3.3. KIR Genotyping         Donor KIR genotyping is performed to ensure the donor possesses
                              the relevant inhibitory KIR gene to exert alloreactivity (see Note
                              2). Moreover, KIR genotyping identifies donors who also possess
                              activating KIR genes (approximately 2/3 of Caucasian popula-
                              tion). Studies suggest activating KIRs may play a role in NK cell
                              alloreactivity and haploidentical transplantation (see Note 3).
                              Detailed methods for KIR typing are provided elsewhere in this
                              volume. In our laboratory, KIR typing is performed using a low
                              resolution PCR-SSP assay (KIR Genotyping Kit, Invitrogen, USA)
                              following the manufacturer’s instructions. This kit is designed to
                              identify 14 KIR genes (2DL1, 2DL2, 3DL1, 2DL4, 2DL5, 2DS1,
                              2DS2, 2DS3, 2DS4, 2DS5, 3DL1, 3DL2, 3DL3, 3DS1), 2 pseudo-
                              genes (2DP1 and 3DP1) and the common variants of KIR2DL5
                              (KIR2DL5A, KIR2DL5B), the KIR2DS4 allele (*001/002 and
                              *003), and KIR3DP1 allele (*001/002 and *003).
2.3.5. NK Cell Cloning      This analysis aims to confirm donor NK alloreactivity against the
and Cytotoxicity Assay      recipient by formally identifying donor NK cells which kill alloge-
                            neic recipient targets, such as PHA lymphoblasts and/or leukemia
                            cells (see Note 4). Large numbers of donor NK clones are gener-
                            ated by limiting dilution and cytotoxicity assays against recipient
                            target cells are used to detect the frequency of alloreactive NK
                            clones. Peripheral blood mononuclear cells are depleted of T cells
                            by negative anti-CD3 immunomagnetic selection (Miltenyi), are
                            plated under limiting-dilution conditions, activated with phytohe-
                            magglutinin (PHA; Biochrom KG, Berlin, Germany), and cultured
                            with interleukin-2 (Chiron BV, Amsterdam, Netherlands) and irra-
                            diated feeder cells. Feeder cells are obtained by pooling buffy coats
                            from 5 to 9 healthy donors. Such donors are not HLA typed as
                            PHA + IL2 activation allows efficient NK cell repertoire cloning
                            regardless of feeder cell HLA type. Cloning efficiencies ranges from
                            1 in 5 to 1 in 10 plated NK cells. Cloned NK cells are screened for
                            alloreactivity by standard 51-Cromium release cytotoxicity at an
                            effector-to-target ratio of 10:1 against recipient PHA lymphoblasts
                            and leukemic cells. Approximately 100 NK clones from each
                            person are screened. Clones exhibiting greater than 30% lysis are
                            scored as alloreactive. The assay is considered positive when the
474        A. Velardi
                        frequency of lytic clones was more than 1 in 50. The same assays
                        are used to monitor posttransplant reconstitution of alloreactive
                        NK cell clones (Fig. 1).
3. Notes
References
 1. Reisner Y, Kapoor N, Kirkpatrick D et al (1983)        with one fully mismatched HLA haplotype. N
    Transplantation     for    severe    combined          Engl J Med 339:186–193
    immunodeficiency with HLA-A, B, D, DR             5.   Ciccone E, Pende D, Viale O et al (1992)
    incompatible parental marrow cells fraction-           Evidence of a natural killer (NK) cell repertoire
    ated by soybean agglutinin and sheep red blood         for (allo) antigen recognition: definition of five
    cells. Blood 61:341–348                                distinct NK-determined allospecificities in
 2. Bachar-Lustig E, Rachamim N, Li HW et al               humans. J Exp Med 175:709–718
    (1995) Nat Med 1:1268–1273                        6.   Colonna M, Brooks EG, Falco M et al (1993)
 3. Aversa F, Tabilio A, Terenzi A et al (1994)            Generation of allospecific natural killer cells by
    Successful engraftment of T-cell-depleted hap-         stimulation across a polymorphism of HLA-C.
    loidentical “three-loci” incompatible trans-           Science 260:1121–1124
    plants in leukemia patients by addition of        7.   Ruggeri L, Capanni M, Casucci M et al (1999)
    recombinant human granulocyte colony-stim-             Role of natural killer cell alloreactivity in HLA-
    ulating factor-mobilized peripheral blood pro-         mismatched hematopoietic stem cell transplan-
    genitor cells to bone marrow inoculum. Blood           tation. Blood 94:333–339
    84:3948–3955                                      8.   Ruggeri L, Capanni M, Urbani E et al (2002)
 4. Aversa F, Tabilio A, Velardi A et al (1998)            Effectiveness of donor natural killer cell allore-
    Treatment of high-risk acute leukemia with             activity in mismatched hematopoietic trans-
    T-cell-depleted stem cells from related donors         plants. Science 295:2097–20100
476      A. Velardi
 9. Kärre K (2002) A Perfect Mismatch. Science       15. Pende D, Marcenaro S, Falco M et al (2009)
    295:2029–2031                                        Anti-leukemia activity of alloreactive NK cells
10. Ruggeri L, Mancusi A, Capanni M et al (2007)         in KIR ligand-mismatched haploidentical
    Donor natural killer cell allorecognition of         HSCT for pediatric patients: evaluation of the
    missing self in haploidentical hematopoietic         functional role of activating KIR and
    transplantation for acute myeloid leukemia:          redefinition of inhibitory KIR specificity. Blood
    challenging its predictive value. Blood 110:         113:3119–3129
    433–440                                          16. Witt CS, Christiansen FT (2006) The relevance
11. Velardi A (2008) Role of KIRs and KIR ligands        of natural killer cell human leucocyte antigen
    in hematopoietic transplantation. Curr Opin          epitopes and killer cell immunoglobulin-like
    Immunol 20:581–587                                   receptors in bone marrow transplantation. Vox
12. Christiansen FT, Velardi A (2009) Progress in        Sang 90:10–20
    understanding and exploiting the immune          17. Foley BA, De Santis D, Van Beelen E et al
    response in solid organ and hemopoietic stem         (2008) The reactivity of Bw4+ HLA-B and
    cell transplantation. Curr Opin Immunol              HLA-A alleles with KIR3DL1: implications for
    21:522–524                                           patient and donor suitability for haploidentical
13. Velardi A, Ruggeri L, Mancusi A et al (2009)         stem cell transplantations. Blood 112:435–443
    Natural killer cell allorecognition of missing   18. Foley B, De Santis D, Lathbury L (2008)
    self in allogeneic hematopoietic transplanta-        KIR2DS1-mediated           activation   overrides
    tion: a tool for immunotherapy of leukemia.          NKG2A-mediated inhibition in HLA-C C2-
    Curr Opin Immunol 21:525–530                         negative individuals. Int Immunol 20:555–563
14. Stern M, Ruggeri L, Mancusi A (2008) Survival    19. Chewning JH, Gudme CN, Hsu KC (2007)
    after T cell-depleted haploidentical stem cell       KIR2DS1-positive NK cells mediate allore-
    transplantation is improved using the mother         sponse against the C2 HLA-KIR ligand group
    as donor. Blood 112:2990–2995                        in vitro. J Immunol 179:854–868
                                                                                        Chapter 27
Abstract
Natural killer (NK) cell alloreactivity can be exploited in haploidentical (one haplotype mismatched)
haematopoietic stem cell transplantation (HSCT) to prevent leukaemia relapse, rejection, and graft-vs-host
disease (GVHD) (Blood 94:333–339; Science 295:2097–2100). If NK cell alloreactivity is to be exploited
in HSCT, it is important to be able to reliably select donors who have NK alloreactivity towards the
patient. The detection of donor NK alloreactivity towards patient target cells has traditionally been evalu-
ated by NK cell cloning and 51Cr-release cytotoxicity assay. This approach is complex and time consuming
with results taking up to 6 weeks. Here, we detail a novel flow cytometric CD107a-based assay capable of
detecting NK cell alloreactivity in 14 days.
     Key words: Natural killer cells, Killer immunoglobulin-like receptors, Alloreactivity, CD107a, Flow
     cytometry
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_27, © Springer Science+Business Media New York 2012
                                                         477
478       D. De Santis et al.
1.2. Role of NK             In a series of studies Ruggeri and colleagues showed that donor-
Alloreactivity              vs-recipient NK cell alloreactivity resulted in less relapse, rejection,
in the Clinical Setting     GVHD, and increased survival in human haploidentical transplants
                            and murine transplant models (11, 12). Most importantly in
                            haploidentical transplants, NK cells were shown to mediate GVL
                            effects without GVHD (11). Since the initial studies of Ruggeri
                            et al., a number of retrospective studies have been performed in
                            haploidentical and unrelated HSCT. However, many of the studies
                            failed to confirm the findings published by Ruggeri et al. The
                            extensive T-cell depletion used in Perugia compared to the other
                            studies has been considered as a possible explanation for the differ-
                            ences observed. However, other differences in transplant proto-
                            cols, such as conditioning regimen or stem cell source, or
                            heterogeneous cohorts with differences in the proportions of dis-
                            ease type could also be responsible for the differences observed
                            between the studies. If the findings of Ruggeri et al. can be
                            confirmed in other transplant settings using the same transplant
                            protocol allowing NK cell alloreactivity to be exploited in stem cell
                            transplants, it will be important to be able to reliably select donors
                            who are alloreactive towards the patient.
                                 Donor NK alloreactivity towards a recipient can largely be
                            predicted if the recipient lacks one of the C1, C2, or Bw4 epitopes
                            that is present in the donor. However, this assumes that all HLA-C
                            and HLA-B alleles interact with the appropriate inhibitory KIR
                            receptors as predicted by the presence of the relevant amino acid
                            and that all alleles of the inhibitory KIR receptors interact the same
                            way with the HLA epitopes. Recently, these assumptions have been
                            shown not to be always true. The Bw4-associated HLA-B13 allele
                            has been shown not to interact with KIR3DL1 (13) while KIR2LD2
                            and KIR2DL3 may interact with both C1- and C2-group HLA-C
                            alleles (14–16). As there has been no comprehensive study to
                            determine which of the HLA alleles interact with KIR receptors, it
                            is not possible to reliably predict alloreactivity from the HLA and
                            KIR genotype. A functional assay is therefore required to confirm
                            NK alloreactivity.
              27   The Detection of NK Cell Alloreactivity by Flow Cytometric CD107a Assay     479
1.3.2. The Flow Cytometric   NK cells have a large number of preformed cytolytic granules in
CD107a Assay                 their cytoplasm. Following activation, NK cells release these gran-
                             ules to the site of cell–cell contact (immunological synapse). These
                             granules contain cytolytic proteins, such as perforin and granzyme,
                             that are involved in inducing target cell death (17). Lining the
                             membrane of the granules is the lysosomal-associated membrane
                             protein, LAMP-1 or CD107a (18). CD107a is a glycoprotein
                             representing approximately 50% of the proteins in the lysosomal
                             membrane. During degranulation, CD107a on the luminal mem-
                             brane is exposed in the immunological synapse and therefore acces-
                             sible for antibody binding. Betts et al. (19) showed CD107a
                             expression on the cell surface was a marker of cytotoxic CD8+ T
                             cell degranulation. Alter et al. (20) showed that CD107a can also
                             be detected on the surface of NK cells following stimulation with
                             target cells lacking HLA ligands. By modifying the method
                             described by Alter et al. we have developed a rapid flow cytometric
                             assay to detect KIR-dependent alloreactive NK cells. Here, we pro-
                             vide a detailed protocol for the rapid, efficient detection of donor
                             NK alloreactivity by a flow cytometric, CD107a-based assay.
2. Materials
                            Table 1
                            HLA types and KIR epitopes of BLCL used as target cells
                            used in the CD107a assay
KIR epitopes
 Table 2
 Antibodies used in the CD107a experiments
3. Methods
3.1. Preparation            1. PBMC are isolated from ten random buffy coats. From the ten
of Peripheral Blood            random donors, centrifuge 10 mL of acid citrate dextrose
Lymphocytes Feeder             (ACD) blood at 300 × g for 10 min and harvest 1 mL of buffy
Cells                          coat from each donor (see Note 3).
                            2. Dilute each buffy coat 1:3 in PBS containing 2% HIFCS
                               (see Note 2), overlay diluted buffy coat onto Ficoll-paque™
                               PLUS and centrifuge at 1,200 × g for 20 min with no brake
                               (see Note 4).
                            3. Harvest the cells from the interface and wash 3 times with
                               PBS/2% HIFCS at 300 × g for 7 min.
                            4. Resuspend the cell pellet in 1 mL RPMI/10% HIFCS, count
                               and adjust to 106/mL.
482      D. De Santis et al.
3.3. Preparation           EBV-transformed BLCL expressing the HLA alleles that encode
of EBV-Transformed         the presence or absence of the C1, C2, and Bw4 epitopes are used
BLCL Target Cells          as target cells in the CD107a assay. The 721.221 cell line, a class I
            27   The Detection of NK Cell Alloreactivity by Flow Cytometric CD107a Assay   483
3.4. CD107a                1. Before use in the CD107a assay, 12-day cultured polyclonal
Cytotoxicity Assay            NK cells are activated by further culture for 48 h with NK
                              medium supplemented with 400 IU IL-2/mL (see Note 10).
                              To each well of a 96-well plate, add 100 μL of the cultured
                              polyclonal NK cells from Subheading 3.2, step 8 and 100 μL
                              of IL-2 supplemented NK medium. Incubate for 48 h at 37°C
                              5% CO2.
                           2. After the 48-h incubation, pool cells from each well, count and
                              adjust to 1 × 106/mL in fresh NK cell culture medium.
                           3. Activated cultured polyclonal NK cells from each donor should
                              be incubated alone (background control), with the HLA class
                              I negative 721.221 target (positive control), a target express-
                              ing all epitopes (negative control), target cells expressing C2
                              and Bw4 and lacking only C1 (C1−), target cells expressing
                              C1 and Bw4 and lacking only C2 (C2−), and target cells
                              expressing both C1 and C2 but not Bw4 (Bw4−) and if avail-
                              able transformed EBV-BLCL from the recipient of HSCT. In
                              duplicate wells, add 100 μL of 1 × 106/mL EBV-BLCL target
                              cells to wells of a 96-well culture plate followed by 100 μL of
                              1 × 106/mL activated cultured polyclonal NK cells (1:1 ratio).
                           4. To the effector/target mix, add 5 μL of anti-CD107a-FITC
                              (see Table 2) antibody and incubate at 37°C in 5% CO2 for 1 h.
                           5. After the hour incubation, add 20 μL of monensin (6 μg/mL)
                              at 1/15 dilution to the cells and further incubate for 5 h at
                              37°C in 5% CO2 (see Note 11).
                           6. Following the incubation, wash the cells by adding 100 μL of
                              flow buffer to each well followed by centrifugation at 200 × g
                              for 5 min and then incubate with an antibody cocktail of a
                              1/40 dilution of anti-CD56-PECy7 and 1/20 dilution of the
                              appropriate KIR antibodies (see Tables 2 and 3) to a total
                              volume of 50 μL in the dark for 15 min at 4°C (see Note 12).
                           7. Wash cells twice in 100 μL flow buffer by centrifugation at
                              200 × g for 5 min.
                           8. Finally, resuspend the cells in 180-μL flow buffer containing
                              1% paraformaldehyde and analyse.
484      D. De Santis et al.
 Table 3
 KIR antibody combinations and BLCL target cells to be used to detect NK
 alloreactivity by the flow cytometric CD107a assay
3.5. Data Analysis         The analysis method is critical in the detection of NK alloreactivity.
                           To minimise the effect of irrelevant receptors, it is important to
                           restrict the analysis to only NK cells expressing the inhibitory KIR
                           receptor relevant to the missing epitope against the relevant target
                           cell. An example of the analysis method used for the detection of
                           potential C1 alloreactive NK cells in a C1+, C2+ NK donor tested
                           against a C1− target is shown in Fig. 1. The analysis method is
                           described below with reference to Fig. 1.
                               1. Analyse the duplicate tubes on a BD FACSCanto™ flow cyto-
                                  meter using BD FACSDiva software (or similar). The final
                                  percentage of cells is an average of the duplicate tubes.
                               2. Gate events initially on forward (FSC) and side scatter (SSC)
                                  to identify lymphocytes. In the example shown in Fig. 1a,
                                  16,217 out of 96,301 events were identified as viable lympho-
                                  cytes (P1).
                               3. Then, gate CD56 vs. SSC to acquire at least 10,000 CD56+
                                  cells. Fig. 1b identifies 11,084 events corresponding to CD56
                                  cells (P2).
                               4. Further gate these CD56+ cells and examine for KIR expres-
                                  sion to identify cells expressing the KIR phenotype of interest,
                                  e.g. CD158a+/158b− or CD158b+/158a−. In the example
                                  shown in Fig. 1c gate P4 (upper left quadrant) identifies cells
                                  expressing CD158b but not CD158a (2,129 events, 19.2%)
                                  while gate P5 identifies cells expressing CD158a but not
                                  CD158b (1,410 events, 12.7%).
                               5. To determine the percentage of cells with the KIR phenotype
                                  of interest that also express CD107a+, use the gates generated
                                  in step 4 (P4 and P5) and analyse in separate bivariate plots the
                           27     The Detection of NK Cell Alloreactivity by Flow Cytometric CD107a Assay                                                                                        485
a All Events b P1
                                         200 250
                                          (⫻ 1.000)
                                                                                                                          105
                                                                                                          CD56 PE-Cy7-A
                                                                                                                          104
                                                                                                                                           P2
                                        FSC-A
                                                150
                                                                                                                          103
                                                100
                                                50
                                                                                                                          102
                                                       50    100      150              200 250                                       50   100     150                200 250
                                                              SSC-A                     (⫻ 1.000)                                          SSC-A                      (⫻ 1.000)
 c                                                            d                                                                            e
                                  P2                                                                P5                                                                            P4
                                                                                                                                                               105
                                                                                 105
                    105
Q1-2 Q2-2
                                                                                                                                                CD158BA PC-A
     CD158BA PC-A
                          P4
                                                                   CD158A PE-A
                                                                                                                                5%                                                         27.4%
                    104
                                                                                                                                                               104
                                                                                 104
                                                                                                     Q2-1
                                                                                 103
                    103
                                                                                                                                                               103
                                                                                                                                                                      Q3-2        Q4-2
                    102
                                                                                                                                                               102
                                                                                 102
P5 Q3-1 Q4-1
                          102     103       104       105                                 102       103                   104    105                                    102       103    104   105
                                CD158A PE-A                                                  CD107A FITC-A                                                                 CD107A FITC-A
Fig. 1. Flow cytometric histograms showing the detection of potential C1 alloreactive NK cells in a C1+, C2+ NK donor
tested against a C1− target. (a) Data gated on forward (FSC) and side scatter (SSC) to identify lymphocytes (16,217 events).
(b) Data gated on CD56 vs. SSC to acquire at least 10,000 CD56+ cells (11,084 events). (c) CD56+ cells expressing the KIR
phenotype of interest, e.g. CD158a+/158b− or CD158b+/158a−. Gate P4 (upper left quadrant ) identifies cells expressing
CD158b but not CD158a (2,129 events, 19.2%) while gate P5 identifies cells expressing CD158a but not CD158b (1,410
events, 12.7%). (d, e) The number of cells in gates P4 and P5, respectively, that express CD107a. The number of CD158a+/
CD158b−/CD107a+ cells is 71 (5%) (d, Q2-1) while the number of CD158b+/CD158a−/CD107a+ cells is 583 (27.4%)
(e, Q2-2). The frequency of C1 alloreactive NK cells is calculated by dividing the number of CD158b+/CD158a−/CD107a+
cells (583) by the total number of CD56+ cells (11,084), giving a frequency of 0.0526 (5.26%), followed by the subtraction
of the percentage of such cells present in the tube that lacks a target, i.e. the background control (0.16%, data not shown).
The frequency of C1 alloreactive NK cells in the example shown in figure is therefore 5.1%. The frequency of C2 alloreac-
tive NK cells against this target (C1−) is calculated in the same way, as expected the frequency of C2 alloreactive NK cells
in this example is low, 0.0056 (0.56%).
4. Notes
References
 1. Valiante NM, Uhrberg M, Shilling HG,                       Lanier LL, Parham P (1997) Human diversity
    Lienert-Weidenbach K, Arnett KL, D’Andrea                  in killer cell inhibitory receptor genes.
    A, Phillips JH, Lanier LL, Parham P (1998)                 Immunity 7:753–763
    Functionally and structurally distinct NK cell       10.   Kim S, Poursine-Laurent J, Truscott SM,
    receptor repertoires in the peripheral blood of            Lybarger L, Song YJ, Yang L, French AR,
    two human donors. Immunity 7:739–751                       Sunwoo JB, Lemieux S, Hansen TH, Yokoyama
 2. Colonna M, Spies T, Strominger JL, Ciccone                 WM (2005) Licensing of natural killer cells by
    E, Moretta A, Moretta L, Pende D, Viale O                  host major histocompatibility complex class I
    (1992) Alloantigen recognition by two human                molecules. Nature 436:709–713
    natural killer cell clones is associated with        11.   Ruggeri L, Capanni M, Casucci M, Volpi I,
    HLA-C or a closely linked gene. Proc Natl                  Tosti A, Perruccio K, Urbani E, Negrin RS,
    Acad Sci USA 89:7983–7985                                  Martelli MF, Velardi A (1999) Role of natural
 3. Colonna M, Brooks EG, Falco M, Ferrara GB,                 killer cell alloreactivity in HLA-mismatched
    Strominger JL (1993) Generation of allospecific            hematopoietic stem cell transplantation. Blood
    natural killer cells by stimulation across a poly-         94:333–339
    morphism        of    HLA-C.      Science    260:    12.   Ruggeri L, Capanni M, Urbani E, Perruccio K,
    1121–1124                                                  Shlomchik WD, Tosti A, Posati S, Rogaia D,
 4. Moretta A, Vitale M, Bottino C, Orengo AM,                 Frassoni F, Aversa F, Martelli MF, Velardi A
    Morelli L, Augugliaro R, Barbaresi M, Ciccone              (2002) Effectiveness of donor natural killer cell
    E, Moretta L (1993) P58 molecules as putative              alloreactivity in mismatched hematopoietic
    receptors for major histocompatibility complex             transplants. Science 295:2097–2100
    (MHC) class I molecules in human natural             13.   Foley BA, De Santis D, Van Beelen E, Lathbury
    killer (NK) cells. Anti-p58 antibodies reconsti-           LJ, Christiansen FT, Witt CS (2008) The reac-
    tute lysis of MHC class I-protected cells in NK            tivity of Bw4+ HLA-B and HLA-A alleles with
    clones displaying different specificities. J Exp           KIR3DL1: implications for patient and donor
    Med 178:597–604                                            suitability for haploidentical stem cell trans-
 5. Vales-Gomez M, Reyburn HT, Erskine RA,                     plantations. Blood 112:435–443
    Strominger J (1998) Differential binding to          14.   Winter CC, Gumperz JE, Parham P, Long EO,
    HLA-C of p50-activating and p58-inhibitory                 Wagtmann N (1998) Direct binding and func-
    natural killer cell receptors. Proc Natl Acad Sci          tional transfer of NK cell inhibitory receptors
    USA 95:14326–14331                                         reveal novel patterns of HLA-C allotype recog-
 6. Cella M, Longo A, Ferrara GB, Strominger JL,               nition. J Immunol 161:571–577
    Colonna M (1994) NK3-specific natural killer         15.   Moesta AK, Norman PJ, Yawata M, Yawata N,
    cells are selectively inhibited by Bw4-positive            Gleimer M, Parham P (2008) Synergistic poly-
    HLA alleles with isoleucine 80. J Exp Med                  morphism at 2 positions distal to the ligand-
    180:1235–1242                                              binding site makes KIR2DL2 a stronger
 7. Litwin V, Gumperz JE, Parham P, Phillips JH,               receptor for HLA-C than KIR2DL3.
    Lanier LL (1994) NKB1: a natural killer cell               J Immunol 180:3969–3979
    receptor involved in the recognition of poly-        16.   Pende D, Marcenaro S, Falco M et al (2008)
    morphic HLA-B molecules. J Exp Med                         Anti-leukemia acitivity of alloreactive NK cells
    180:537–543                                                in KIR ligand-mismatched haploidentical
 8. Gumperz JE, Litwin V, Phillips JH, Lanier LL,              HSCT for pediatric patients: evaluation of the
    Parham P (1995) The Bw4 public epitope of                  functional role of activating KIR and
    HLA-B molecules confers reactivity with natural            re-definition of inhibitory KIR specificity.
    killer cell clones that express NKB1, a putative           Blood 113:3119–3129
    HLA receptor. J Exp Med 181:1133–1144                17.   Djeu JY, Jiang K, Wei S (2002) A view to a kill:
 9. Uhrberg M, Valiante NM, Shum BP, Shilling                  signals triggering cytotoxicity. Clin Cancer Res
    HG, Lienert-Weidenbach K, Corliss B, Tyan D,               8:636–640
             27   The Detection of NK Cell Alloreactivity by Flow Cytometric CD107a Assay                 489
18. Fukuda M (1991) Lysosomal membrane glyco-            20. Alter G, Malenfant JM, Altfeld M (2004)
    proteins. Structure, biosynthesis, and intracellu-       CD107a as a functional marker for the
    lar trafficking. J Biol Chem 266:21327–21330             identification of natural killer cell activity. J
19. Betts MR, Brenchley JM, Price DA, De Rosa                Immunol Methods 294:15–22
    SC, Douek DC, Roederer M, Koup RA (2003)             21. De Santis D, Foley BA, John E, Senitzer D,
    Sensitive and viable identification of antigen-          Christiansen FT, Witt CS (2010) Rapid, flow
    specific CD8+ T cells by a flow cytometric assay         cytometric assay for NK alloreactivity reveals
    for degranulation. J Immunol Methods 281:                exceptions to rules governing alloreactivity. Biol
    65–78                                                    Blood Marrow Transplant 16(2):179–191
                                                                                        Chapter 28
Abstract
Recent advances have improved our understanding of natural killer (NK) cell-mediated alloreactivity after
hematopoietic cell transplantation (HCT) or with adoptive transfer. NK cells contribute to a graft-versus-
leukemia effect and may play a role in preventing graft-versus-host disease or controlling infectious diseases
after allogeneic HCT. New discoveries in NK cell biology, including characterization of NK cell receptors
and their interactions with self-HLA molecules and a better understanding of the mechanism of NK cell
education have led to the development of novel strategies to exploit NK cell alloreactivity against tumors.
While early studies using autologous NK cells lacked efficacy, the use of adoptively transferred NK cells to
treat hematopoietic malignancies has been expanding. The production of allogeneic donor NK cells
requires efficient removal of T- and B cells from clinical-scale leukapheresis collections. The goal of this
chapter is to review NK cell biology, NK cell receptors, the use of NK cells as therapy and then to discuss
the clinical decisions resulting in our current good manufacturing practices processing and activation of
human NK cells for therapeutic use.
Key words: NK cells, Immunotherapy, Cell processing, Current good manufacturing practices
1. Introduction
1.1. NK Cell Biology            NK cells are large granular lymphocytes that were first described in
                                the 1970s for their ability to lyse virally infected and tumor target
                                cells without MHC-restriction or prior sensitization (1, 2). Human
                                NK cells are found in the bone marrow, spleen, lymph nodes, and
                                peripheral blood (PB), where they comprise ~10–15% of the lym-
                                phocyte pool. NK cells play an important role in immune surveil-
                                lance and response to infections and link the innate and adaptive
                                immune systems (3, 4). Activated NK cells produce cytokines and
                                chemokines, including granulocyte colony stimulatory factor
                                (G-CSF), TNF, interferon gamma (IFN-γ), and transforming
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_28, © Springer Science+Business Media New York 2012
                                                         491
492      D.H. McKenna et al.
1.2. NK Cell Receptors   In 1985, Ljunggren and Karre described the phenomenon of “miss-
Control Function,        ing self recognition,” whereby the loss of MHC class I expression
Self-Tolerance,          renders autologous targets more sensitive to NK-mediated killing.
and Alloreactivity       The mechanism by which these innate killer cells recognize class I
                         loss in tumors or virally infected cells (13) was explained with the
                         discovery of killer-cell immunoglobulin-like receptors (KIR). Human
                         KIR gene content can be simplified into two haplotypes, each of
                         which contain the three framework genes (KIR3DL3, KIR2DL4,
                         and KIR3DL2), and a variable number of activating and inhibitory
                         genes from the centromeric (Cen) or telomeric (Tel) ends. KIR A
                         haplotypes contain five inhibitory receptors (2DL1, 2DL3, 3DL1,
                         3DL2, 3DL3), and just one activating receptor (2DS4). KIR B hap-
                         lotypes have variable gene content and contain more activating KIR.
                         Binding of higher affinity inhibitory KIR to their cognate ligands,
                         self-class I HLA, suppresses NK cell effector responses, including
                         cell-mediated lysis and cytokine release (14). KIR2DL1, KIR2DL2/
                         KIR2DL3, and KIR3DL1 bind HLA class I C2, C1 and Bw4 alleles,
                         respectively. In contrast, the natural ligands for activating KIR remain
                         unknown.
                              Several other families of activating and inhibitory receptors affect
                         NK cell function. The NKG2 family of C-type lectin receptors can be
                         inhibitory (NKG2A) or activating (NKG2C/E), and recognize non-
                         classical HLA-E (15). NKG2D recognizes stress-induced molecules,
                         such as MHC class I polypeptide-related sequence A/B (MICA and
                         MICB) or viral particles (16). Other receptors include the natural
                         cytotoxicity receptors (NCR) NKp30, NKp46 and NKp44, DNAM-
                         1, and Nectin-2 (CD122), 2B4, leukocyte-associated immunoglobu-
                         lin-like receptor-1 (LAIR-1) (17, 18), and Ig-like (ILT) receptors
                         (19–23). NK cell function is determined by the net sum of signals
                         delivered through inhibitory and activating NK cell receptors.
                              The mechanism by which NK cells acquire self-tolerance and
                         alloreactivity has been referred to as “NK cell education.” Several
                         models have been proposed to explain the integration of inhibitory
                         receptor expression with the acquisition of effector functions.
   28   Clinical Production and Therapeutic Applications of Alloreactive Natural Killer Cells   493
1.4. Clinical Factors     Studies using autologous NK cells did not require extensive pro-
Influencing               cessing because the non-NK cells (T cells, B cells, and monocytes),
Development of an         also autologous to the patient, were not felt to pose a significant
NK Cell Product           risk even after ex vivo IL-2 activation. However, when crossing
28   Clinical Production and Therapeutic Applications of Alloreactive Natural Killer Cells   495
2. Materials
3. Methods
3.1. cGMP Production         In general, clinical production and administration of novel cell
of NK Cells                  therapies in the USA requires submission to and approval by the
                             FDA for an investigational new drug (IND) application. Further,
   28   Clinical Production and Therapeutic Applications of Alloreactive Natural Killer Cells   497
3.3. Cell Processing       The apheresis product is T cell (CD3)-depleted and B cell (CD19)-
                           depleted using the Miltenyi Biotec CliniMACS® Cell Selection
                           System and CD3 and CD19 MicroBeads and reagent (Miltenyi
                           Biotec, Bergisch Gladbach, Germany). Up to 2.50 × 1010 total
                           nucleated cells (TNC) may be labeled with the MicroBeads and
                           separated on an LS column (Depletion Program 2.1) which is
                           placed in the magnetic field of the CliniMACS® device. Cells are
                           resuspended at 2 × 106 cells/mL in X-VIVO 15, without gentami-
                           cin and phenol red (Cambrex BioScience, Walkersville, Maryland),
                           supplemented with 1,000 U/mL IL-2 (Chiron Corporation,
                           Emeryville, CA) and 10% human AB serum, heat-inactivated
                           (Valley Biomedical Products and Services, Inc., Winchester, VA) in
                           VueLife™ Teflon® (FEP) bags (American Fluoroseal Corporation,
                           Gaithersburg, MD). Cells are then incubated overnight for 8–16 h
                           (37°C/5% CO2). Cells are washed twice with a 5% HSA, and the
                           final formulation consists of nucleated cells in approximately
                           80–150 mL of 5% HSA solution (<100 × 106 NC/mL). The target
                           dose for infusion is £ 8 × 107 NC/kg.
498        D.H. McKenna et al.
3.4.3. Overnight Culture     1. Inspect appropriately sized culture bags (FEP) for expiry date
in X-VIVO 15/IL-2               and integrity
                             2. Transfer cells into culture bags and incubate in 37°C/5% CO2
                                incubator for 8–16 h
                                   (f) Open the hemostat on the cell product line and transfer
                                       cells to the COBE processing bag
                                   (g) Volume reduce product at 1,500 rpm (300 × g) for 3 min
                                       per spin; at the end of super out, expel as much super-
                                       natant as possible into the waste bag
                                   (h) Fill the processing bag with 5% HSA; remove air from pro-
                                       cessing bag; wash product twice at 1,500 rpm (300 × g)
                                       for 3 min per spin; expel as much supernatant as possible
                                       into the waste bag
                                   (i) Heat seal and remove the processing bag from the COBE
                                       2991; detach and discard the remainder of the processing
                                       set
                                   (j) Transfer the cells into an appropriately sized transfer bag;
                                       rinse the processing bag with 5% HSA; and add to the
                                       transfer bag
                                   (k) Dilute the final product to approximately 100 mL total
                                       volume with 5% HSA
                              3. Dose adjust (if necessary)
                                   (a) Calculate available dose and determine if a portion of cells
                                       needs to be removed
3.6. Sampling for QC/         1. All lot release testing is performed on the final product prior to
Lot Release Testing              release of the product for infusion. Lot release criteria are
                                 shown in Table 1.
 Table 1
 Natural killer cells: lot release criteria
3.7. Final Labeling and     1. Label appropriately and include the following statement:
Release                        “Caution: New Drug—Limited by Federal Law to Investigational
                               Use” per 21 CFR 312.6
                            2. Release NK cell product once lot release testing has been
                               completed, passed, and reviewed by Quality Assurance unit.
4. Notes
                               7. Lot release for NK cell purity has been used as 20% in non-
                                  transplant settings of NK cell infusion.
                               8. The average T-cell content of the final product in a recent non-
                                  transplant trial contained a median-infused T-cell dose of
                                  5.6 × 104 CD3+ T cells/kg. Caution should be used in setting
                                  this parameter depending on the clinical setting relative to the
                                  risk of transfusion-associated graft-versus-host disease. For
                                  example, the threshold for T-cell dose may need to be lower
                                  when NK cell products are used as a part of haploidentical
                                  transplant. Safe T-cell doses may vary based on the clinical set-
                                  ting where the NK cell product is administered.
                               9. Absolute safe thresholds for B cell content relative to uncom-
                                  mon complications associated with product B cells have not
                                  been firmly established. These thresholds may vary depending
                                  on the immune competence of the recipient.
                             10. An upper dose limit has been set but is rarely reached based on
                                 the processing of a single apheresis product (15 L processing
                                 volume; roughly 5 h collection). Larger or multiple collection
                                 above this threshold have not been tested for safety.
References
 1. Herberman RB, Ortaldo JR (1981) Natural                9. Carson WE, Giri JG, Lindemann MJ, Linett
    killer cells: their roles in defenses against dis-        ML, Ahdieh M, Paxton R et al (1994)
    ease. Science 214(4516):24–30                             Interleukin (IL) 15 is a novel cytokine that acti-
 2. Kiessling R, Petranyi G, Klein G, Wigzel H                vates human natural killer cells via components
    (1975) Genetic variation of in vitro cytolytic            of the IL-2 receptor. J Exp Med 180(4):
    activity and in vivo rejection potential of non-          1395–1403
    immunized semi-syngeneic mice against a mouse         10. Trinchieri G (2003) Interleukin-12 and the
    lymphoma line. Int J Cancer 15(6):933–940                 regulation of innate resistance and adaptive
 3. Moretta L, Ferlazzo G, Bottino C, Vitale M,               immunity. Nat Rev Immunol 3(2):133–146
    Pende D, Mingari MC et al (2006) Effector and         11. Young HA, Ortaldo J (2006) Cytokines as criti-
    regulatory events during natural killer-dendritic         cal co-stimulatory molecules in modulating the
    cell interactions. Immunol Rev 214:219–228                immune response of natural killer cells. Cell Res
 4. Walzer T, Dalod M, Robbins SH, Zitvogel L,                16(1):20–24
    Vivier E (2005) Natural-killer cells and den-         12. Singh SM, Yanagawa H, Hanibuchi M, Miki T,
    dritic cells: “l’union fait la force”. Blood              Okamura H, Sone S (2000) Augmentation by
    106(7):2252–2258                                          interleukin-18 of MHC-nonrestricted killer
 5. Cooper MA, Fehniger TA, Caligiuri MA (2001)               activity of human peripheral blood mononu-
    The biology of human natural killer-cell subsets.         clear cells in response to interleukin-12. Int J
    Trends Immunol 22(11):633–640                             Immunopharmacol 22(1):35–43
 6. Fehniger TA, Caligiuri MA (2001) Interleukin          13. Ljunggren HG, Karre K (1990) In search of the
    15: biology and relevance to human disease.               ‘missing self’: MHC molecules and NK cell rec-
    Blood 97(1):14–32                                         ognition. Immunol Today 11(7):237–244
 7. Caligiuri MA (2008) Human natural killer cells.       14. Vales-Gomez M, Reyburn HT, Mandelboim
    Blood 112(3):461–469                                      M, Strominger JL (1998) Kinetics of interac-
 8. Rayner AA, Grimm EA, Lotze MT, Chu EW,                    tion of HLA-C ligands with natural killer cell
    Rosenberg SA (1985) Lymphokine-activated                  inhibitory receptors. Immunity 9(3):337–344
    killer (LAK) cells. Analysis of factors relevant to   15. Houchins JP, Yabe T, McSherry C, Bach FH
    the immunotherapy of human cancer. Cancer                 (1991) DNA sequence analysis of NKG2, a
    55(6):1327–1333                                           family of related cDNA clones encoding type II
   28   Clinical Production and Therapeutic Applications of Alloreactive Natural Killer Cells             505
    integral membrane proteins on human natural             reconstitution is altered by T cells in the graft
    killer cells. J Exp Med 173(4):1017–1020                and correlates with clinical outcomes after unre-
16. Lopez-Botet M, Angulo A, Guma M (2004)                  lated      donor      transplantation.       Blood
    Natural killer cell receptors for major histocom-       106(13):4370–4376
    patibility complex class I and related molecules    28. Bottino C, Castriconi R, Moretta L, Moretta A
    in cytomegalovirus infection. Tissue Antigens           (2005) Cellular ligands of activating NK recep-
    63(3):195–203                                           tors. Trends Immunol 26(4):221–226
17. Moretta L, Bottino C, Pende D, Castriconi R,        29. Soriani A, Zingoni A, Cerboni C, Iannitto ML,
    Mingari MC, Moretta A (2006) Surface NK                 Ricciardi MR, Di Gialleonardo V et al (2009)
    receptors and their ligands on tumor cells.             ATM-ATR dependent up-regulation of
    Semin Immunol 18(3):151–158                             DNAM-1 and NKG2D ligands on multiple
18. Meyaard L (2008) The inhibitory collagen                myeloma cells by therapeutic agents results in
    receptor LAIR-1 (CD305). J Leukoc Biol                  enhanced NK cell susceptibility and is associ-
    83(4):799–803                                           ated with a senescent phenotype. Blood
19. Riteau B, Menier C, Khalil-Daher I, Martinozzi          113(15):3503–3511
    S, Pla M, Dausset J et al (2001) HLA-G1 co-         30. Stein MN, Shin J, Gudzowaty O, Bernstein
    expression boosts the HLA class I-mediated              AM, Liu JM (2006) Antibody-dependent cell
    NK lysis inhibition. Int Immunol 13(2):                 cytotoxicity to breast cancer targets despite
    193–201                                                 inhibitory KIR signaling. Anticancer Res
20. Riteau B, Rouas-Freiss N, Menier C, Paul P,             26(3A):1759–1763
    Dausset J, Carosella ED (2001) HLA-G2, -G3,         31. Pende D, Spaggiari GM, Marcenaro S, Martini
    and -G4 isoforms expressed as nonmature cell            S, Rivera P, Capobianco A et al (2005) Analysis
    surface glycoproteins inhibit NK and antigen-           of the receptor-ligand interactions in the natu-
    specific CTL cytolysis. J Immunol 166(8):               ral killer-mediated lysis of freshly isolated myel-
    5018–5026                                               oid or lymphoblastic leukemias: evidence for
                                                            the involvement of the Poliovirus receptor
21. Shiroishi M, Tsumoto K, Amano K, Shirakihara
                                                            (CD155) and Nectin-2 (CD112). Blood
    Y, Colonna M, Braud VM et al (2003) Human
                                                            105(5):2066–2073
    inhibitory receptors Ig-like transcript 2 (ILT2)
    and ILT4 compete with CD8 for MHC class I           32. Re F, Staudacher C, Zamai L, Vecchio V, Bregni
    binding and bind preferentially to HLA-G.               M (2006) Killer cell Ig-like receptors ligand-
    Proc Natl Acad Sci USA 100):8856–8861                   mismatched, alloreactive natural killer cells lyse
                                                            primary solid tumors. Cancer 107(3):640–648
22. Cerboni C, Achour A, Warnmark A, Mousavi-
    Jazi M, Sandalova T, Hsu ML et al (2006)            33. Armeanu S, Bitzer M, Lauer UM, Venturelli S,
    Spontaneous mutations in the human CMV                  Pathil A, Krusch M et al (2005) Natural killer
    HLA class I homologue UL18 affect its binding           cell-mediated lysis of hepatoma cells via specific
    to the inhibitory receptor LIR-1/ILT2/CD85j.            induction of NKG2D ligands by the histone
    Eur J Immunol 36(3):732–741                             deacetylase inhibitor sodium valproate. Cancer
                                                            Res 65(14):6321–6329
23. Kirwan SE, Burshtyn DN (2005) Killer cell
    Ig-like receptor-dependent signaling by Ig-like     34. Guerra N, Tan YX, Joncker NT, Choy A,
    transcript 2 (ILT2/CD85j/LILRB1/LIR-1). J               Gallardo F, Xiong N et al (2008) NKG2D-
    Immunol 175(8):5006–5015                                deficient mice are defective in tumor surveil-
                                                            lance in models of spontaneous malignancy.
24. Anfossi N, Andre P, Guia S, Falk CS, Roetynck           Immunity 28(4):571–580
    S, Stewart CA et al (2006) Human NK cell edu-
    cation by inhibitory receptors for MHC class I.     35. Brenner CD, King S, Przewoznik M, Wolters I,
    Immunity 25(2):331–342                                  Adam C, Bornkamm GW et al (2010)
                                                            Requirements for control of B-cell lymphoma
25. Cooley S, Xiao F, Pitt M, Gleason M, McCullar           by NK cells. Eur J Immunol 40(2):494–504
    V, Bergmann TL et al (2007) A subpopulation
                                                        36. Ruggeri L, Capanni M, Urbani E, Perruccio K,
    of human peripheral blood NK cells that lacks
                                                            Shlomchik WD, Tosti A et al (2002)
    inhibitory receptors for self MHC is develop-
                                                            Effectiveness of donor natural killer cell allore-
    mentally immature. Blood 110:578–586
                                                            activity in mismatched hematopoietic trans-
26. Shilling HG, McQueen KL, Cheng NW,                      plants. Science 295(5562):2097–2100
    Shizuru JA, Negrin RS, Parham P (2003)
                                                        37. Miller JS, Soignier Y, Panoskaltsis-Mortari A,
    Reconstitution of NK cell receptor repertoire
                                                            McNearney SA, Yun GH, Fautsch SK et al
    following HLA-matched hematopoietic cell
                                                            (2005) Successful adoptive transfer and in vivo
    transplantation. Blood 101(9):3730–3740
                                                            expansion of human haploidentical NK cells in
27. Cooley S, McCullar V, Wangen R, Bergemann               patients    with     cancer.     Blood      105(8):
    TL, Spellman S, Weisdorf DJ et al (2005) KIR            3051–3057
506        D.H. McKenna et al.
38. Davies SM, Ruggieri L, DeFor T, Wagner JE,              48. Farrell HE, Vally H, Lynch DM, Fleming P,
    Weisdorf DJ, Miller JS et al (2002) Evaluation              Shellam GR, Scalzo AA et al (1997) Inhibition
    of KIR ligand incompatibility in mismatched                 of natural killer cells by a cytomegalovirus MHC
    unrelated donor hematopoietic transplants.                  class I homologue in vivo. Nature 386(6624):
    Killer immunoglobulin-like receptor. Blood                  510–514
    100(10):3825–3827                                       49. Giebel S, Locatelli F, Lamparelli T, Velardi
39. Farag SS, Bacigalupo A, Eapen M, Hurley C,                  A, Davies S, Frumento G et al (2003) Survival
    Dupont B, Caligiuri MA et al (2006) The effect              advantage with KIR ligand incompatibility in
    of KIR ligand incompatibility on the outcome                hematopoietic stem cell transplantation from
    of unrelated donor transplantation: a report                unrelated donors. Blood 102(3):814–819
    from the center for international blood and             50. Beelen DW, Ottinger HD, Ferencik S,
    marrow transplant research, the European                    Elmaagacli AH, Peceny R, Trenschel R et al
    blood and marrow transplant registry, and the               (2005) Genotypic inhibitory killer immuno-
    Dutch registry. Biol Blood Marrow Transplant                globulin-like receptor ligand incompatibility
    12(8):876–884                                               enhances the long-term antileukemic effect of
40. Miller JS, Cooley S, Parham P, Farag SS,                    unmodified allogeneic hematopoietic stem cell
    Verneris MR, McQueen KL et al (2007) Missing                transplantation in patients with myeloid leuke-
    KIR ligands are associated with less relapse and            mias. Blood 105(6):2594–2600
    increased graft-versus-host disease (GVHD)              51. Lowe EJ, Turner V, Handgretinger R, Horwitz
    following unrelated donor allogeneic HCT.                   EM, Benaim E, Hale GA et al (2003) T-cell
    Blood 109(11):5058–5061                                     alloreactivity dominates natural killer cell allore-
41. Shlomchik WD, Couzens MS, Tang CB, McNiff                   activity in minimally T-cell-depleted HLA-non-
    J, Robert ME, Liu J et al (1999) Prevention of              identical paediatric bone marrow transplantation.
    graft versus host disease by inactivation of host           Br J Haematol 123(2):323–326
    antigen-presenting             cells.         Science   52. Clausen J, Kircher B, Auberger J, Schumacher
    285(5426):412–415                                           P, Ulmer H, Hetzenauer G et al (2010) The
42. Lundqvist A, McCoy JP, Samsel L, Childs R (2007)            role of missing killer cell immunoglobulin-like
    Reduction of GVHD and enhanced antitumor                    receptor ligands in T cell replete peripheral
    effects after adoptive infusion of alloreactive Ly49-       blood stem cell transplantation from HLA-
    mismatched NK cells from MHC-matched donors.                identical siblings. Biol Blood Marrow Transplant
    Blood 109(8):3603–3606                                      16(2):273–280
43. Russell JH, Ley TJ (2002) Lymphocyte-                   53. Linn YC, Phang CY, Lim TJ, Chong SF, Heng
    mediated cytotoxicity. Annu Rev Immunol                     KK, Lee JJ et al (2010) Effect of missing killer-
    20:323–370                                                  immunoglobulin-like receptor ligand in recipients
44. Murphy WJ, Keller JR, Harrison CL, Young                    undergoing HLA full matched, non-T-depleted
    HA, Longo DL (1992) Interleukin-2-activated                 sibling donor transplantation: a single institution
    natural killer cells can support hematopoiesis              experience of 151 Asian patients. Bone Marrow
    in vitro and promote marrow engraftment                     Transplant 45(6):1031–1037
    in vivo. Blood 80(3):670–677                            54. Gagne K, Busson M, Bignon JD, Balere-Appert
45. Siefer AK, Longo DL, Harrison CL, Reynolds                  ML, Loiseau P, Dormoy A et al (2009) Donor
    CW, Murphy WJ (1993) Activated natural killer               KIR3DL1/3DS1 gene and recipient Bw4 KIR
    cells and interleukin-2 promote granulocytic                ligand as prognostic markers for outcome in
    and megakaryocytic reconstitution after synge-              unrelated hematopoietic stem cell transplanta-
    neic bone marrow transplantation in mice.                   tion. Biol Blood Marrow Transplant 15(11):
    Blood 82(8):2577–2584                                       1366–1375
46. Chen C, Busson M, Rocha V, Appert ML,                   55. Triplett BM, Horwitz EM, Iyengar R, Turner
    Lepage V, Dulphy N et al (2006) Activating                  V, Holladay MS, Gan K et al (2009) Effects of
    KIR genes are associated with CMV reactiva-                 activating NK cell receptor expression and NK
    tion and survival after non-T-cell depleted                 cell reconstitution on the outcomes of unre-
    HLA-identical sibling bone marrow transplan-                lated donor hematopoietic cell transplantation
    tation for malignant disorders. Bone Marrow                 for hematologic malignancies. Leukemia
    Transplant 38(6):437–444                                    23(7):1278–1287
47. Stern M, Elsasser H, Honger G, Steiger J,               56. Cooley S, Trachtenberg E, Bergemann TL,
    Schaub S, Hess C (2008) The number of acti-                 Saeteurn K, Klein J, Le CT et al (2009) Donors
    vating KIR genes inversely correlates with the              with group B KIR haplotypes improve relapse-
    rate of CMV infection/reactivation in kidney                free survival after unrelated hematopoietic cell
    transplant recipients. Am J Transplant 8(6):                transplantation for acute myelogenous leuke-
    1312–1317                                                   mia. Blood 113(3):726–732
   28   Clinical Production and Therapeutic Applications of Alloreactive Natural Killer Cells              507
57. Cooley S, Weisdorf DJ, Guethlein LA, Klein JP,           in childhood acute myeloid leukemia. J Clin
    Wang T, Le CT et al (2010) Donor selection for           Oncol 28(6):955–959
    natural killer cell receptor genes leads to supe-    67. Lugli E, Goldman CK, Perera LP, Smedley J,
    rior survival after unrelated transplantation for        Pung R, Yovandich JL et al (2010) Transient
    acute myelogenous leukemia. Blood 116(14):               and persistent effects of IL15 on lymphocyte
    2411–2419                                                homeostasis in nonhuman primates. Blood
58. Burns LJ, Weisdorf DJ, DeFor TE, Vesole DH,              116(17):3238–3248
    Repka TL, Blazar BR et al (2003) IL-2-based          68. Barao I, Hanash AM, Hallett W, Welniak LA,
    immunotherapy after autologous transplanta-              Sun K, Redelman D et al (2006) Suppression
    tion for lymphoma and breast cancer induces              of natural killer cell-mediated bone marrow
    immune activation and cytokine release: a                cell rejection by CD4+ CD25+ regulatory T
    phase I/II trial. Bone Marrow Transplant                 cells. Proc Natl Acad Sci USA 103(14):
    32(2):177–186                                            5460–5465
59. Dudley ME, Wunderlich JR, Robbins PF, Yang           69. Bordin JO, Heddle NM, Blajchman MA (1994)
    JC, Hwu P, Schwartzentruber DJ et al (2002)              Biologic effects of leukocytes present in trans-
    Cancer regression and autoimmunity in patients           fused cellular blood products. Blood
    after clonal repopulation with antitumor lym-            84(6):1703–1721
    phocytes. Science 298(5594):850–854                  70. McKenna DH Jr, Sumstad D, Bostrom N,
60. Bachanova V, Burns LJ, McKenna DH,                       Kadidlo DM, Fautsch S, McNearney S et al
    Curtsinger J, Panoskaltsis-Mortari A, Lindgren           (2007) Good manufacturing practices produc-
    BR et al (2010) Allogeneic natural killer cells          tion of natural killer cells for immunotherapy: a
    for refractory lymphoma. Cancer Immunol                  six-year single-institution experience. Transfusion
    Immunother 59(11):1739–1744                              47(3):520–528
61. Geller MA, Cooley S, Judson PL, Ghebre R,            71. Miller JS, Oelkers S, Verfaillie C, McGlave P
    Carson LF, Argenta PA et al (2010) A phase II            (1992) Role of monocytes in the expansion of
    study of allogeneic natural killer cell therapy to       human activated natural killer cells. Blood
    treat patients with recurrent ovarian and breast         80(9):2221–2229
    cancer. Cytotherapy 13(1):98–107                     72. Godal R, Bachanova V, Gleason M, McCullar
62. Cooley S, Gada P, McKenna D, McCullar V,                 V, Yun GH, Cooley S, Verneris MR, McGlave
    Fautsch S, Verneris MR et al (2008) Successful           PB, Miller JS (2010) Natural killer cell kill-
    haploidentical hematopoietic cell engraftment            ing of AML and ALL blasts by killer cell
    using a non-myeloablative preparative regimen            immunoglobulin-like receptor negative nat-
    including natural killer (NK) cells. In: American        ural killer cells after NKG2A and LIR-1
    Society of Hematology 50th annual meeting,               blockade. Biol Blood Marrow Transplant
    San Francisco, CA: Blood, p 827                          16(5): 612–621
63. Pfeiffer MM, Feuchtinger T, Teltschik HM,            73. Curtis RE, Travis LB, Rowlings PA, Socie G,
    Schumm M, Muller I, Handgretinger R et al                Kingma DW, Banks PM et al (1999) Risk of
    (2010) Reconstitution of NK cell receptors               lymphoproliferative disorders after bone mar-
    influences NK activity and relapse rate after            row transplantation: a multi-institutional study.
    haploidentical transplantation of T and B cell           Blood 94(7):2208–2216
    depleted grafts in children. Haematologica           74. Reed M, Yearsley M, Krugh D, Kennedy MS
    95(8):1381–1388                                          (2003) Severe hemolysis due to passenger lym-
64. Passweg JR, Tichelli A, Meyer-Monard S, Heim             phocyte syndrome after hematopoietic stem cell
    D, Stern M, Kuhne T et al (2004) Purified                transplantation from an HLA-matched related
    donor NK-lymphocyte infusion to consolidate              donor. Arch Pathol Lab Med 127(10):
    engraftment after haploidentical stem cell trans-        1366–1368
    plantation. Leukemia 18(11):1835–1838                75. Hummon D, Zantek ND, Sumstad D, Miller
65. Koehl U, Esser R, Zimmermann S, Tonn T,                  JS, McKenna DH (2009) Transfusion-associated
    Kotchetkov R, Bartling T et al (2005) Ex vivo            graft-versus-host disease: a perspective from a
    expansion of highly purified NK cells for immu-          cell therapy laboratory. Transfusion 49(5):
    notherapy after haploidentical stem cell trans-          1018–1019
    plantation in children. Klin Padiatr 217(6):         76. McKenna DH, Kadidlo DM, Miller JS, Orchard
    345–350                                                  PJ, Wagner JE, McCullough J (2005) The
66. Rubnitz JE, Inaba H, Ribeiro RC, Pounds S,               Minnesota molecular and cellular therapeutics
    Rooney B, Bell T et al (2010) NKAML: a pilot             facility: a state-of-the-art biotherapeutics engi-
    study to determine the safety and feasibility of         neering laboratory. Transfus Med Rev 19(3):
    haploidentical natural killer cell transplantation       217–228
                                                                                        Chapter 29
Abstract
In HLA-matched stem-cell transplantation (SCT), minor H antigens are key molecules driving allo-
immune responses in both graft-versus-host disease (GvHD) and in graft-versus-leukemia (GvL) reactivity.
Dissection of the dual function of minor H antigens became evident through their different modes of
tissue and cell expression, i.e., hematopoietic system restricted or broad. Broadly expressed minor H
antigens are the targets of immune responses in both arms of graft-versus-host (GvH) responses, i.e.,
both GvHD and GvL, whereas the immune responses against the hematopoietic system-specific minor H
antigens are restricted to the GvL arm of SCT. Evidently, it is this latter group of minor H antigens that
can function as curative tools for stem-cell (SC)-based immunotherapy of hematological malignancies and
disorders. The HLA-matched patient/donor combinations, incompatible for one of the hematopoietic-
specific minor H antigens, are suitable for minor H antigen immunotherapy (Goulmy, Immunol Rev
157:125–140, 1997). Information on the minor H antigen phenotype is therefore needed. Hereto,
genomic typing for minor H antigens has been implemented in many HLA laboratories. Here, we firstly
summarize the relevance of minor H antigens particularly in hematopoietic SCT. Secondly, we describe
a method for typing the various polymorphic minor H antigens molecularly identified to date by
DNA sequencing.
1. Introduction
1.1. Background                 Minor H antigens are peptides derived from polymorphic self-proteins
                                that can differ both between unrelated individuals and between
                                related individuals. Minor H peptides are presented by the various
                                MHC class I and class II molecules. Their immune recognition is a
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_29, © Springer Science+Business Media New York 2012
                                                         509
510      E. Spierings and E. Goulmy
1.2. Minor H Antigen     Autosomally encoded minor H antigens show different phenotype
Immunogenetics           frequencies; the latter also differ in the various ethnic populations.
                         Evidently, the number of patients eligible for minor H antigen-
                         based immunotherapy depends on their phenotype frequencies in
                         the relevant ethnic population (12). The high phenotype frequency
                         of HLA-A2 in combination with a relatively high disparity rate for
                         HA-1 in all ethnic populations studied, currently mark HA-1 as
                         the most favorable minor H antigen for clinical application in all
                         ethnic populations. Similar results, however limited to only one or
                         two ethnic groups, were obtained for the minor H antigens ACC-
                         1, ACC-2, and HB-1Y. Based upon frequencies, the ACC-1 minor
                         H antigen is applicable in the Asian/Pacific population, with 6.5%
                         disparity in sibling pairs and 12.1% in MUD pairs. Similarly, ACC-2
                         and HB-1Y can only be applied in a significant proportion of the
                         pairs in the Caucasian and Mulatto population. Finally, the chance
                         of having at least one mismatch for one of the eight hematopoietic
                         system-restricted minor H antigen systems studied herein was cal-
                         culated as 21.2 and 33.6% for Caucasian sibling and MUD pairs,
                         respectively. In the black population, these numbers were 7.4 and
                         11.9%, respectively. In all these estimations, the HLA allele
                         frequencies of the respective populations were taken into account.
                       29   Minor Histocompatibility Antigen Typing by DNA Sequencing…           511
 Table 1
 Y-chromosome-encoded minor H antigens order by HLA-restriction molecule
1.3. Minor H Antigen        The Y-chromosome encoded HY antigens (Table 1), most of which
Tissue Distribution         are broadly expressed, are reported to clinically contribute to graft-
in Relation to Their        versus-leukemia (GvL) activity (13). Among others, CTLs specific
Clinical Relevance          for the broadly expressed HY antigen can be detected in associa-
                            tion with leukemia remission (14).
                                The involvement of broadly expressed minor H antigens in the
                            GvHD arm of SCT was first shown by in vitro functional assays.
                            Namely, broadly expressed minor H antigens, such as the HY anti-
                            gens, are, among others, expressed on cell types of organs affected
                            by GvHD, such as fibroblasts, melanocytes, and keratinocytes (15).
                            This indicates that CTLs directed to broadly expressed minor H
                            antigens are particularly relevant for the development of GvHD.
                            To further address this issue, skin sections were incubated with
                            CTLs specific for the broadly expressed minor H antigen, HY, or
                            for the hematopoietic system-restricted minor H antigens HA-1
                            and HA-2. CTLs specific for the HY minor H antigen induced
                            severe GvH reactions of grades III–IV, while CTLs specific for
                            HA-1 and HA-2 induced no or weak GvH reactions (16). We
                            recently developed specific T-cell staining reagents to directly study
512      E. Spierings and E. Goulmy
1.4. Minor H Antigens    Minor H antigens with expression limited to cells of the hematopoi-
as Treatment Modality    etic system are especially relevant for the GvL activity (21–23). The
                         latter type of minor H antigens can be exploited as curative tools
                         for SC-based immunotherapy of hematological malignancies and
                         disorders (Goulmy, Immunol Rev 157:125–140, 1997). Hereto,
                         HLA-matched minor H antigen-mismatched recipient/donor
                         combinations are selected. Two treatment modalities currently
                         exploit minor H antigen disparity between SC donor and recipient.
                         One modality is the adoptive transfer of in vitro generated SC
                         donor-derived minor H antigen-specific CTLs. The other, a more
                         practical and far less laborious strategy, is post-SCT “vaccination.”
                         Both strategies, which are briefly discussed below, are currently
                         ongoing in clinical phase I/II studies.
                              Adoptive transfer of human minor H antigen-specific CTLs
                         has been shown to be successful in the treatment of human leuke-
                         mia and solid tumors in an experimental nonobese diabetic/severe
                         combined immunodeficiency mouse model (24, 25). Translation
29   Minor Histocompatibility Antigen Typing by DNA Sequencing…           513
2. Materials
Minor H antigen Allele Forward primer Reverse primer Amplicon size (bp)
                                                                                                   (continued)
                                                                                                                 515
 Table 2
                                                                                                                                           516
(continued)
Minor H antigen Allele Forward primer Reverse primer Amplicon size (bp)
Table 3
Oligonucleotide primers for gene-specific PCR
Minor H                                                                                 Size
antigen       Forward primer                      Reverse primer                        (bp)
UGT2B17
  5¢ region   GGCAGTATCTTGCCAATGT                 AGACTCCAAGTGCCAGTT                    341
  Exon 1a     TGTTGGGAATATTCTGACTATAA             CCCACTTCTTCAGATCATATGC                352
  Exon 1b     AAATGACAGAAAGAAACAA                 GCATCTTCACAGAGCTTATAT                 443
  Exon 6      GAATTCATCATGATCAACCG                ACAGGCAACATTTTGTGATC                  201
HY (SRY)      TGGCGATTAAGTCAAATTCGC               CCCCCTAGTACCCTGACAAT                  136
                                                   GTATT
Table 4
Oligonucleotide primers for sequencing-based typing
Minor H                                                                           Amplicon
antigen          Type of primer         Primer sequence                           size (bp)
HA-1             Forward PCR            AAGGACCGTTGTTTGTGAAAGC
                 Reverse PCR            CAGGAAACAGCTATGACCCGTT                    523
                                         GTCATGCCTTCATCCTAC
                 Forward sequence       AAGGACCGTTGTTTGTGAAAGC
                 Reverse sequence       CAGGAAACAGCTATGACC
HA-2             Forward PCR            CCCCTTCTTCCTTCTCCACT
                 Reverse PCR            CAGGAAACAGCTATGACCAGCC                    526
                                         AGGTTCGCTCTCTTA
                 Forward sequence       CACAGGAGATGCTCTAATGG
                 Reverse sequence       TGTGGGTGCCTGTAAG
HA-3             Forward PCR            GTGACTGACCCACAGGGAGT
                 Reverse PCR            CCCAGGTGGGAAAGTACTCA                      498
                 Forward sequence       TACCTCTTGATTGGGAGAAA
                 Reverse sequence       CTTGACTTGTTCGATGACAG
HA-8             Forward PCR            GCTACCTCTTCTGGGCAACA
                 Reverse PCR            CAGGAAACAGCTATGACGCCTG                    402
                                         GCTCTTTAAATGCAC
                 Forward sequence       GCCATTGGAGTTAGAATCTG
                 Reverse sequence       TCAGCTAATCAGGTTACTCC
                                                                                 (continued)
518       E. Spierings and E. Goulmy
Table 4
(continued)
Minor H                                                         Amplicon
antigen            Type of primer      Primer sequence          size (bp)
HB-1               Forward PCR         CCTGAGCCTTCTGACCTCAC
                   Reverse PCR         AAACTCTGGGCTTCGAGTCA     215
                   Forward sequence    CACAGTGGAGAAACCAGAAC
                   Reverse sequence    AAGAGCTGTGCCTCAGATAC
ACC-1              Forward PCR         CATTGCCTCAACAGCTTCAA
 and-2
                   Reverse PCR         CAGGAAACAGCTATGACAGCCT   478
                                        CCGTTTTGCCTTATC
                   Forward sequence    CTCAAGACTTTGCTCTCCAC
                   Reverse sequence    CATTATGAACTCCGCAAC
PANE1              Forward PCR         GCAGGCCACGGAGAT
                   Reverse PCR         GTCAAGCCCTGACTGGACAT     308
                   Forward sequence    GGAAGGAGGAACAGC
                   Reverse sequence    ACCCTCACAGGTCCTCCA
SP110              Forward PCR         CCATCACCTAAGGTCACCAA
                   Reverse PCR         TGAAGAGTGGGCCTTGATTT     573
                   Forward sequence    CAAGATTGGGAGATCAATGT
                   Reverse sequence    TAAAATTCAACCCTCCACAG
CTSH               Forward PCR         GAGAACCCCCGCCTATAATG
                   Reverse PCR         GCCGACAGGTGAACTAGAGC     507
                   Forward sequence    TCTGCGTACCTAAGGAGTTC
                   Reverse sequence    TCCCAGTTGACGCTC
 LRH-1             Forward PCR         CCTTCATTCTGCCAGGAGAG
                   Reverse PCR         CAGGAAACAGCTATGACCCTCA   378
                                        GTGCCTCTCTGGTTC
                   Forward sequence    AGAATAGGGGTTCTTCTTCG
                   Reverse sequence    GGGTTCTTCTTCGCTT
                                                                (continued)
                             29    Minor Histocompatibility Antigen Typing by DNA Sequencing…          519
 Table 4
 (continued)
 Minor H                                                                                        Amplicon
 antigen                 Type of primer             Primer sequence                             size (bp)
 LB-ADIR-1               Forward PCR                CTCCTCGTCCTCGTCACAGT
                         Reverse PCR                CAGGAAACAGCTATGACAGG                        447
                                                     ACCCCAGACAGGGTTT
                         Forward sequence           GCTGAAGAGCGTCCAGTA
                         Reverse sequence           CCACAGCAACACCAAT
 CD19                    Forward PCR                CTCTCTCTGCGGGTCTTTGT
                         Reverse PCR                CAGGAAACAGCTATGACTTCA                       557
                                                     GCTCTAGGCTCAGCAA
                         Forward sequence           GTGTCTCTGCATTTGGTTCT
                         Reverse sequence           CACAGGACAGCCAGAG
Sequences in bold represent the M13-reverse primer sequence
2.2. Special Equipment             1. Standard micropipets ranging from 0.2 μL to 1.0 mL.
2.2.1. Pre-PCR Area                2. Filter tips (0.5–10; 10–100; 100–1,000 μL).
                                   3. Thin-walled PCR tubes, strips, or individual tubes (Eppendorf).
2.2.2. Thermal Cycler Area         1. Thermal cycler with a 96-well block. We have successfully used
                                      the Dyad Disciple (MJ-Research) and the models 2720 and
                                      9700 from Applied Biosystems.
3. Methods
3.1. DNA Preparation           1. Different methods for isolation of genomic DNA are available.
                                  For optimal results, high-quality DNA is required. The
                                  described protocols assume a DNA concentration of 30 ng/μL.
                                  If required, dilute the DNA stock solution in HPLC-grade
                                  water. Higher or lower concentrations of DNA may lead to
                                  unsuccessful amplification.
                               2. Control DNA can be obtained from the IHW cell panel.
                                  A large number of the cell lines in this panel has been typed for
                                  minor H antigens (38). See Note 1 for preventing carryover
                                  contamination.
3.5. Purification            1. Add Sephadex to the matrix plate (Millipore), fill each matrix
of Sequencing                   well to the rim.
Reaction Products            2. Place the Millipore Multiscreen plate upside down on top of
                                the filled matrix plate.
                             3. Transfer the Sephadex from the matrix plate to the Millipore
                                Multiscreen plate by turning the matrix plate to the top
                                position.
522      E. Spierings and E. Goulmy
3.7. Sequence Analysis   Analyses can be performed manually with general sequencing anal-
                         ysis software like BioEdit, or automated with SBTEngine®, which
                         includes data files for a number of minor H antigens. An example
                         of automated allele assignments is depicted in Fig. 1. Homozygosity
                         can be confirmed with PCR-SSP (see Note 6).
                           29    Minor Histocompatibility Antigen Typing by DNA Sequencing…                        523
Fig. 1. Sequencing-based typing results for minor H antigen HA-1. The cell-lines used in this validation protocol were
previous typed by PCR-SSP. Analyses were performed with SBTEngine© (GenDx BV). Left panel shows a reference sample
that is homozygous for the HA-1H allele. Numbers at top indicate the codon position. The sequence underneath the codon
position displays the potential nucleic acids and their position, based upon the known HA-1 alleles. The position of the
nonsynymous SNP at position 416 has been marked with a circle. The right panel displays the result for the HA-1R refer-
ence sample. In the middle, a heterozygous reference sample has been depicted. Both the heterozygous synonymous SNP
at position 412 and the nonsynonymous SNP at position 416 have been underlined.
4. Notes
Table 5
Autosomally encoded minor H antigens, ordered alphabetically
Acknowledgments
References
 1. Goulmy E (1997) Human minor histocompat-           4. Goulmy E, Gratama JW, Blokland E, Zwaan
    ibility antigens: new concepts for marrow trans-      FE, van Rood JJ (1983) A minor transplanta-
    plantation and adoptive immunotherapy.                tion antigen detected by MHC-restricted cyto-
    Immunol Rev 157:125–140                               toxic T lymphocytes during graft-versus-host
 2. Goulmy E, Termijtelen A, Bradley BA, van              disease. Nature 302:159–161
    Rood JJ (1976) Alloimmunity to human H-Y.          5. van Els CA, D’Amaro J, Pool J, Blokland E,
    Lancet 2:1206                                         Bakker A, van Elsen PJ et al (1992)
 3. Goulmy E, Bradley BA, Lansbergen Q, van               Immunogenetics of human minor histocom-
    Rood JJ (1978) The importance of H-Y incom-           patibility antigens: their polymorphism and
    patibility in human organ transplantation.            immunodominance. Immunogenetics 35:
    Transplantation 25:315–319                            161–165
                        29   Minor Histocompatibility Antigen Typing by DNA Sequencing…                      527
 6. Wang W, Meadows LR, den Haan JM, Sherman           16. Dickinson AM, Wang XN, Sviland L, Vyth-
    NE, Chen Y, Blokland E et al (1995) Human              Dreese FA, Jackson GH, Schumacher TN et al
    H-Y: a male-specific histocompatibility antigen        (2002) In situ dissection of the graft-versus-host
    derived from the SMCY protein. Science                 activities of cytotoxic T cells specific for minor his-
    269:1588–1590                                          tocompatibility antigens. Nat Med 8:410–414
 7. Goulmy E, Pool J, van den Elsen PJ (1995)          17. Kim YH, Faaij CM, van Halteren AG, Schrama E,
    Interindividual conservation of T-cell receptor        de Jong TA, Schøller J, Egeler RM, Pavel S, Vyth-
    beta chain variable regions by minor histocom-         Dreese FA, van Tol MJ, Goulmy E, Spierings E.
    patibility    antigen-specific   HLA-A*0201-           In situ detection of HY-specific T cells in acute
    restricted cytotoxic T-cell clones. Blood 85:          graft-versus-host disease-affected male skin after
    2478–2481                                              sex-mismatched stem cell transplantation. Biol
 8. den Haan JM, Sherman NE, Blokland E,                   Blood Marrow Transplant. 2012 Mar;18(3):
    Huczko E, Koning F, Drijfhout JW et al (1995)          381–7. Epub 2011 Nov 4.
    Identification of a graft versus host disease-     18. van der Harst D, Goulmy E, Falkenburg J,
    associated human minor histocompatibility              Kooij-Winkelaar Y, van Luxemburg-Heijs S,
    antigen. Science 268:1476–1480                         Goselink H et al (1994) Recognition of minor
 9. Verdijk RM, Kloosterman A, Pool J, Van De              histocompatibility antigens on lymphocytic and
    KM, Naipal AM, Van Halteren AG et al (2004)            myeloid leukemic cells by cytotoxic T-cell
    Pregnancy induces minor histocompatibility             clones. Blood 83:1060–1066
    antigen-specific cytotoxic T cells: implications   19. Marijt W, Heemskerk M, Kloosterboer F,
    for stem cell transplantation and immunother-          Goulmy E, Kester M, van der Horn M et al
    apy. Blood 103:1961–1964                               (2003) Hematopoiesis-restricted minor histo-
10. Mommaas B, Stegehuis-Kamp JA, Van Halteren             compatibilty antigens HA-1- or HA-2-specific
    AG, Kester M, Enczmann J, Wernet P et al               T cells can induce complete remissions of
    (2004) Cord blood comprises antigen-                   relapsed leukemia. Proc Natl Acad Sci USA
    experienced T cells specific for maternal minor        100:2742–2747
    histocompatibility antigen HA-1. Blood 105:        20. Kim YH, Vyth-Dreese FA, Schrama E, Pavel S,
    1823–1827                                              Bajema I, Goulmy E et al (2010) Exogenous
11. Van Halteren AG, Jankowska-Gan E, Joosten              addition of minor H antigen HA-1+ dendritic
    A, Blokland E, Pool J, Brand A et al (2009)            cells to skin tissues ex vivo causes infiltration
    Naturally acquired tolerance and sensitization         and activation of HA-1 specific cytotoxic T
    to minor histocompatibility antigens in                cells. Biol Blood Marrow Transplant 17(1):
    healthy family members. Blood 114:                     69–77
    2263–2272                                          21. Mutis T, Goulmy E (2002) Hematopoietic
12. Spierings E, Hendriks M, Absi L, Canossi A,            system-specific antigens as targets for cellular
    Chhaya S, Crowley J et al (2007) Phenotype             immunotherapy of hematological malignancies.
    frequencies of autosomal minor histocompati-           Semin Hematol 39:23–31
    bility antigens display significant differences    22. Klein CA, Wilke M, Pool J, Vermeulen C,
    among populations. PLoS Genet 3:e103                   Blokland E, Burghart E et al (2002) The
13. Gratwohl A, Hermans J, Niederwieser D, van             hematopoietic system-specific minor histocom-
    Biezen A, van Houwelingen HC, Apperley                 patibility antigen HA-1 shows aberrant expres-
    J (2001) Female donors influence transplant-           sion in epithelial cancer cells. J Exp Med
    related mortality and relapse incidence in male        196:359–368
    recipients of sibling blood and marrow trans-      23. Fujii N, Hiraki A, Ikeda K, Ohmura Y, Nozaki
    plants. Hematol J 2:363–370                            I, Shinagawa K et al (2002) Expression of
14. Takami A, Sugimori C, Feng X, Yachie A,                minor histocompatibility antigen, HA-1, in
    Kondo Y, Nishimura R et al (2004) Expansion            solid tumor cells. Transplantation 73:
    and activation of minor histocompatibility             1137–1141
    antigen HY-specific T cells associated with        24. Hambach L, Nijmeijer BA, Aghai Z, Schie ML,
    graft-versus-leukemia response. Bone Marrow            Wauben MH, Falkenburg JH et al (2006)
    Transplant 34:703–709                                  Human cytotoxic T lymphocytes specific for a
15. de Bueger MM, Bakker A, Van Rood JJ, Van               single minor histocompatibility antigen HA-1
    der Woude F, Goulmy E (1992) Tissue distri-            are effective against human lymphoblastic leu-
    bution of human minor histocompatibility anti-         kaemia in NOD/scid mice. Leukemia
    gens ubiquitous versus restricted tissue               20:371–374
    distribution indicates heterogeneity among         25. Hambach L, Vermeij M, Buser A, Aghai Z, Van
    human cytotoxic T lymphocyte-defined non-              der KT, Goulmy E (2008) Targeting a single
    MHC antigens. J Immunol 149:1788–1794                  mismatched minor histocompatibility antigen
528       E. Spierings and E. Goulmy
Abstract
The optimal donor of hematopoietic progenitor cells shares alleles of the major histocompatibility genes
with the recipient. This chapter describes the strategies aimed at identifying such a matched donor from
registries of volunteers or from umbilical cord blood banks.
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_30, © Springer Science+Business Media New York 2012
                                                         531
532      C.K. Hurley et al.
2. Materials
2.1. Web Sites Useful          1. Bone Marrow Donors Worldwide (BMDW): This Web site
in Search                         (http://www.bmdw.org/) provides a comprehensive listing of
                                  almost all registries and cord blood banks in the world and
                                  their contact information. It also provides the tools to perform
                                  an initial search of a single worldwide database of donors to
                                  assess the likelihood of identifying a matched donor (5). This
                                  database does not substitute, however, for a search of a specific
                                  registry or cord blood bank because it is only a snapshot of
                                  each registry/bank at one recent time and does not directly
                                  allow access to specific donors/cord blood units for further
                                  testing.
                               2. HLA nomenclature and allele designations: The Web site,
                                  http://hla.alleles.org/nomenclature/, is the World Health
                                  Organization-designated HLA nomenclature Web site and it
                                  includes both current and previous HLA designations (6, 7).
                                  The IMmunoGeneTics/HLA Web site (http://www.ebi.ac.
                                  uk/imgt/hla/) is a source of information on each HLA allele
                                  including whether the sequence has been reported more than
                                  once (i.e., has been confirmed) and the race/ethnicity of the
                                  individuals in whom the allele was first identified (8). It pro-
                                  vides tools to compare HLA sequences and provides a listing
                                  of alleles and genotypes that cannot be distinguished from one
                                  another (i.e., are ambiguous) based on the nucleotide sequence
                                  of their most variable exons. It also provides a tool to link sero-
                                  logic and DNA-based HLA assignments based on an HLA
                                  “dictionary” (9).
                               3. Allele and haplotype frequencies: The frequencies of alleles and
                                  haplotypes (see Note 1) within the US National Marrow
                                  Donor Program (NMDP) registry can be found at http://
                                  bioinformatics.nmdp.org/ (see Note 2) (10). A list of rare
                                  alleles identifies alleles that have never been observed or have
                                  seldom been observed within the NMDP registry. This site
                                  also includes a tool to identify the alleles that are contained
                                  within the letter codes used to report HLA types (e.g.,
                                  DRB1*04:AB where AB indicates 01 or 02 defining the assign-
                                  ment DRB1*04:01 or DRB1*04:02) (7). Two sources of
                                         30   Donor Registries and Search Strategies      533
3. Methods
3.1. HLA Typing         1. Obtain a biological sample from the patient and from nuclear
of Patient and Family      family members if transplantation is being considered as a
                           potential treatment option (see Note 3) and perform HLA
                           typing (see Note 4). If available, include parents of the patient,
                           siblings, and biological children. The patient should be typed
                           for HLA-A,-B, -C, -DR, -DQ (see Note 5) at allele or high
                           resolution (see Note 6). The resolution of the typing of the
                           family members should be sufficient to clearly distinguish the
                           HLA types segregating in the family (see Note 7).
                        2. If sufficient family members are available, confirm the HLA
                           assignments of the patient through observing the same assign-
                           ments in other family members, identify the HLA types carried
                           by each haplotype segregating in the family (see Note 8), and
                           determine if there is a family member who is HLA matched.
                           The latter should be further evaluated to determine if they
                           might serve as an HPC donor (see Note 9).
                        3. Perform an unrelated donor and umbilical cord blood search
                           as described below so that these options for treatment of the
                           patient can be considered.
Table 1
Example of how a patient typing is evaluated
3.4. Reviewing           1. Obtain a search report from the registries and banks contacted
a Search Report             (see Note 22). Most registries will display adult donor matches
to Select Potential         based on HLA-A,-B,-DRB1 typing and will list both 6/6 and
Adult Volunteer             5/6 matches. The selection of umbilical cord blood units is
Donors for Additional       described in Subheading 3.6.
HLA Testing              2. Ensure that the correct match criteria are known for the patient
                            (whether an 8/8 or 10/10 match is preferred; which and how
                            many mismatches are allowed; and any other donor-specific
                            characteristics that need to be considered, such as gender, age,
                            or CMV status). This will guide the selection of the best poten-
                            tial donors and help to increase the likelihood of a successful
                            transplantation (see Note 23).
                         3. Expand any donor typing results (see Note 24) that may have
                            letter codes (e.g., A*01:CRY) as necessary to understand the
                            full set of alleles in the assignments. See Note 25 for a tool to
                            assist with this task.
                         4. Evaluate the donor typing results for completeness according to
                            the transplant center match criteria. For example, if an 8/8 match
536       C.K. Hurley et al.
3.5. Refining the               1. When the search report or the analyses performed above does
Search Strategy                    not yield donors who potentially match the patient, a strategy
to Identify a                      to locate the best potential mismatch may need to be imple-
Mismatched or                      mented. In these instances, a donor with only one mismatch
Mistyped Donor                     (7/8 or 9/10) may be considered as an alternative depending
                                   on the transplant center protocol (see Note 32). The search
                                   reports from most registries include 5/6 matched donors (i.e.,
                                   with one mismatch at one of the three loci, HLA-A, -B,
                                   -DRB1). A longer list of potential donors may be needed if the
                                   search was initiated through EMDIS since some registries limit
                                   the search results to either 50 or 100 donors.
                                2. Evaluate the patient for the presence of IgG antibodies directed
                                   to specific HLA antigens (see Note 33).
                                3. Evaluate the methodology that may have been utilized to gen-
                                   erate the HLA data of the potential donors. Assignments made
                                   using older serologic typing techniques have a higher likeli-
                                   hood of appearing falsely disparate than typing techniques that
Table 2
Examples of how potential donor typing is evaluated for patient in Table 1
                                                                  frequency of the alleles in the code, are likely to be matched (i.e., DRB1*13:03
                                                                  is more common than DRB1*13:33 or DRB1*13:66)
Determine likelihood for      HaploStats                        Predict higher resolution and likelihood of allele match with patient based on
 allele match: serologic                                          ethnicity. A*02:01 is more frequent than A*02:05; it is unlikely that the donor
                              HLA explorer
 typing at HLA-A and -B                                           carries A*68:04. B*35:01 is common and may be present in B35 typed indi-
                              DNA dictionary                      vidual although other B*35 alleles (B*35:02, B*35:03) are possible; B*53:01 is
                                                                  likely to be present
Evaluate typing for           http://bioinformatics.nmdp.org/   Predict HLA-C alleles that might be present. If B*35:01 and B*53:01 are present,
  completeness: HLA-C           index.html, haplotype table       both B alleles are frequently found with C*04:01
  not typed
Is resolution sufficient                                        The donor is likely an HLA-A mismatch (i.e., not A*68:04). A donor matched at
   to make a determination                                        7/8 or 9/10 may be acceptable to the transplant center. Depending on other
   of match?                                                      donors available on the search report, determine whether this donor should be
                                                                  typed at higher resolution
                                                                                                                                                     Donor Registries and Search Strategies
                                                                                                                                                     537
538      C.K. Hurley et al.
3.6. Searching                1. Review the cord blood units that have been listed on the
for a Cord Blood Unit            patient’s search report (see Note 41).
                              2. Identify units with greater than a minimal required cell dose
                                 based on the transplant center protocol (see Note 42).
                              3. Identify units that may meet the transplant center’s HLA match-
                                 ing criteria for a cord blood unit (see Note 43). The units will
                                 likely be HLA typed at a low resolution and higher resolution
                                 testing may be required for optimal donor selection.
                              4. Select one or more units that have both an acceptable cell dose
                                 and an acceptable HLA match. Request that the Registry/
                                 Cord Blood Bank initiate further HLA testing of the selected
                                 unit or request a sample for typing at your local HLA labora-
                                 tory (see Note 44). Also request a Unit Report that gives more
                                 information about the unit plus the maternal health history.
                              5. Consider whether the unit has attached segments for verification
                                 typing (see Note 45) in making your decision about whether
                                 to request the unit for transplant.
                              6. Request that the Registry/Cord Blood Bank reserve the unit
                                 for your patient until you have completed your evaluation.
                            30   Donor Registries and Search Strategies       539
4. Notes
    that there are alleles that may be rare or uncommon in all but
    one ethnic group. In this case, finding a matched donor can be
    greatly facilitated by selecting potential donors from the ethnic
    group in which your patient’s allele is less rare or even
    common.
       Furthermore, it may be very difficult to find donors if the
    patient has a rare HLA association (e.g., HLA-B + HLA-C
    allele association) and this needs to be identified by using HLA
    haplotype frequency tables. For example, if the patient has a
    rare HLA-B + HLA-C association and no potentially identical
    donors are present, one can substitute the HLA-B allele for
    another allele that is commonly associated with the patient’s
    HLA-C allele. See Subheading 3.5 for more information on
    search strategies to find a mismatched donor.
13. Preferentially, family typing is available and the patient’s haplo-
    types can be established through family segregation analyses.
    If family data are not available, there are haplotype frequency
    tables that can be used to predict which haplotypes the patient
    might carry and also to provide information whether or not
    the patient has common haplotypes. Understanding common
    haplotype or allele associations can be useful for predicting
    which alleles are most likely to be present in donors who have
    only low resolution typing information and no family data to
    define haplotypes. Two useful Web sites for haplotype analysis
    are http://bioinformatics.nmdp.org and http://www.hlaex-
    plorer.net.
14. Registries and banks have guidelines for who can submit a
    search request. Usually, search requests must come from a
    physician or from a transplant center.
15. Typically, searches are performed in two steps. In the prelimi-
    nary search, a physician will receive a summary indicating
    whether or not there are potentially matching donors in a
    registry or cord blood bank. If the registry appears to include
    suitable donors, a transplant center will formalize the search.
    In a formal search, the HLA types of each potentially matching
    donor/unit will be listed individually and the transplant center
    can request biologic samples from these donors/units for
    further HLA and infectious disease testing.
16. It is important to confirm the patient’s typing using a second
    biological sample to ensure that no errors are made in the HLA
    assignments to be used in the unrelated donor/umbilical cord
    blood search.
17. It is recommended that the search identify the best unrelated
    donor and the best umbilical cord blood unit to provide alter-
    natives for the physician. An urgent search might focus only on
    umbilical cord blood units because they are more rapidly
    available.
542   C.K. Hurley et al.
                           18. The level of matching required for optimal outcome has been
                               discussed in the literature, e.g., (13, 14, 16, 17).
                           19. Access to the search tool in BMDW requires that a transplant
                               center register with BMDW. The search is submitted online.
                               Matches identified in the BMDW worldwide registry will iden-
                               tify one or more countries where potential donors or cord
                               blood units can be found for the patient. The transplant center
                               can then focus the search on registries and cord blood banks
                               from those countries. Contact information for these registries
                               and banks can be found on the BMDW Web site.
                           20. The national registry in the country of the transplant center
                               caring for the patient may also assist in contacting multiple
                               registries and banks. For example, a search of the NMDP in
                               the USA will also interrogate the inventory of the BMDW.
                               Any physician may request a search of the NMDP on behalf of
                               a patient through the Office of Patient Advocacy (http://www.
                               bethematch.org). There are also several publicly available Web
                               sites that provide summarized search results (e.g., MatchView
                               at http://www.marrow.org). When using the NMDP, staff will
                               perform all of the contacts and assist with HLA expertise to
                               guide the process of selecting the best potential donor or cord
                               blood unit.
                           21. Search requests often go from registry to registry using the
                               European Marrow Donor Information System (EMDIS).
                               Transplant centers may have the ability to directly enter the
                               search information into a software application.
                           22. Searches performed electronically using BMDW or NMDP
                               generate donor lists immediately. Most registries and cord
                               blood banks will provide a search report within 24 h when
                               using the EMDIS system. The matches displayed usually
                               include 6/6 (HLA-A,-B,-DRB1) and 5/6 matched donors
                               but the criteria for the donors displayed on a search should be
                               confirmed with the registry.
                           23. Non-HLA factors that are often considered in donor selection
                               have been reviewed by Confer et al. (18).
                           24. The donors/units listed on the report are sorted by the extent
                               of matching with the patient. It is possible, however, that the
                               optimal donor may not appear at the top of the list so it is criti-
                               cal to examine all of the donors on the report.
                           25. An HLA typing result may include a two or more letter code
                               following the first colon in the allele name (e.g., A*02:AF
                               where AF is a code standing for 01 or 09 indicating that an
                               individual carries A*02:01 or A*02:09). The Web site, http://
                               bioinformatics.nmdp.org, has a tool to define alleles included
                               in any letter codes. Further information on HLA assignments
                               can be found at http://hla.alleles.org and http://www.ebi.ac.
                               uk/imgt/hla/.
                  30   Donor Registries and Search Strategies    543
26. There are several approaches that can be used to predict the
    most likely missing data. The HaploStats tool and the HLA
    Explorer will generate a table of the most frequently seen
    HLA-C types associated with a known HLA-B and the most
    frequently seen HLA-DQB1 types associated with a known
    DRB1 antigen sorted by likelihood and ethnicity. The complete
    list of HLA-A, -B, -C, -DRB1, and -DQB1 haplotypes found on
    http://bioinformatics.nmdp.org can be used by searching on
    the known HLA-A, -B, and DRB1 haplotype and then viewing
    the possibilities of full haplotype combinations considering the
    frequencies of each and the racial/ethnic group of the donor
    (if known).
27. The HaploStats and HLA Explorer tools will each display the
    possible high resolution types given any low resolution input.
    The difference in the two tools is that HaploStats requires all
    known loci to be entered and HLA Explorer will provide data
    on any single low resolution type. They both sort by frequency
    within racial/ethnic populations.
28. High or allele resolution typing should be performed to identify
    those donors who carry the same HLA alleles as the patient.
    This extended typing should include all loci to be considered for
    matching. Verification typing using a fresh biological sample
    from the donor should be performed in order to ensure that this
    individual was the one with the HLA assignment appearing on
    the search report. The resolution of verification typing should
    be sufficient to confirm the original HLA assignments.
29. It is recommended that more than one donor be selected for
    additional HLA testing. This is because some donors may not
    carry the same alleles as the patient following higher resolution
    testing or because some volunteers will be unwilling or unable
    to donate or cannot be located. The number of donors to
    select will depend on the likelihood of the patient’s alleles and
    haplotypes being present in the donors. For common alleles
    and haplotypes, 3–5 donors might be selected for further HLA
    testing. Uncommon alleles and haplotypes may require 10 or
    more donors to be selected for extended HLA typing. The
    registry may have information on the rate of donor availability
    to help with this decision.
30. The donor’s registry or the cord blood bank will request that
    your extended HLA typing results be provided to them to
    update the donor/unit file. Follow the policy of the registry
    for the loci and level of resolution to be tested when reporting
    the results for each donor.
31. If there are multiple suitable matched donors, HLA assignments
    at other HLA loci, not routinely considered by the transplant
    center in determining the match, can be used to select among
    the donors. For example, if the transplant center requests an
    8/8 match and there are several 8/8 matched donors, matching
544   C.K. Hurley et al.
                              45. It is important to insure that the cord blood unit is the one
                                  with the HLA typing found on the search report. This
                                  verification typing should be performed on a biological sample
                                  found in an aliquot physically attached to the unit.
                              46. The specifications for the resolution of HLA typing should be
                                  established by the transplant center in discussion with the his-
                                  tocompatibility laboratory. The resolution of the patient’s typ-
                                  ing assignment will determine which donors appear on a search
                                  report and may vary among registries. For example, donors
                                  listed on a search report for a patient typed as C*04:01 may
                                  differ from the donors listed for a patient typed as C*04:01:01G
                                  (which     includes     alternative     alleles   C*04:01:01:01,
                                  C*04:01:01:02, C*04:01:01:03, C*04:01:01:04, C*04:09N,
                                  C*04:28, C*04:30, C*04:41, C*04:79, C*04:82, C*04:84)
                                  or typed as C*04:FEAS (which includes alternative alleles
                                  C*04:01, C*04:09N, C*04:28, C*04:30, C*04:41).
References
 1. Copelan EA (2006) Hematopoietic stem-             10. Maiers M, Gragert L, Klitz W (2007) High-
    cell transplantation. N Engl J Med 354:               resolution HLA alleles and haplotypes in the
    1813–1826                                             United States population. Hum Immunol
 2. Hurley CK et al (2007) Overview of registries,        68:779–788
    HLA typing and diversity, and search algo-        11. Middleton D et al (2003) New allele frequency
    rithms. Tissue Antigens 69(Suppl 1):3–5               database. Tissue Antigens 61:403–407. http://
 3. Foeken LM et al (2010) Monitoring the inter-          www.allelefrequencies.net
    national use of unrelated donors for transplan-   12. Solberg OD et al (2008) Balancing selection
    tation: the WMDA annual reports. Bone                 and heterogeneity across the classical human
    Marrow Transplant 45:811–818                          leukocyte antigen loci: a meta-analytic review
 4. Welte K et al (2010) International exchange of        of 497 population studies. Hum Immunol
    cord blood units: the registry aspects. Bone          69:443–464
    Marrow Transplant 45:825–831                      13. Lee SJ et al (2007) High-resolution donor-
 5. Oudshoorn M et al (1994) Bone Marrow                  recipient HLA matching contributes to the suc-
    Donors Worldwide: a successful exercise in            cess of unrelated donor marrow transplantation.
    international cooperation. Bone Marrow                Blood 110:4576–4583
    Transplant 14:3–8                                 14. Bray RA et al (2008) National marrow donor
 6. Marsh SG et al (2010) Nomenclature for fac-           program HLA matching guidelines for unrelated
    tors of the HLA system, 2010. Tissue Antigens         adult donor hematopoietic cell transplants. Biol
    75:291–455                                            Blood Marrow Transplant 14:45–53
 7. Bochtler W et al (2007) World Marrow Donor        15. Schipper RF, D’Amaro J, Oudshoorn M (1996)
    Association guidelines for use of HLA nomen-          The probability of finding a suitable related
    clature and its validation in the data exchange       donor for bone marrow transplantation in
    among hematopoietic stem cell donor registries        extended families. Blood 87:800–804
    and cord blood banks. Bone Marrow Transplant      16. Petersdorf EW (2008) Optimal HLA matching
    39:737–741                                            in hematopoietic cell transplantation. Curr
 8. Robinson J et al (2009) The IMGT/HLA                  Opin Immunol 20:588–593
    database. Nucleic Acids Res 37:D1013–D1017        17. Shaw BE et al (2010) Diverging effects of
 9. Holdsworth R et al (2009) The HLA diction-            HLA-DPB1 matching status on outcome fol-
    ary 2008: a summary of HLA-A, -B, -C,                 lowing unrelated donor transplantation depend-
    -DRB1/3/4/5, and -DQB1 alleles and their              ing on disease stage and the degree of matching
    association with serologically defined HLA-A,         for other HLA alleles. Leukemia 24:58–65
    -B, -C, -DR, and -DQ antigens. Tissue Antigens    18. Confer DL et al (2010) Selection of adult
    73:95–170                                             unrelated hematopoietic stem cell donors:
                                                  30    Donor Registries and Search Strategies            547
    beyond HLA. Biol Blood Marrow Transplant                of HLA mismatches on bone marrow transplant
    16:S8–S11                                               outcomes in the United States. Biol Blood
19. Spellman S et al (2010) The detection of donor-         Marrow Transplant 15:971–981
    directed, HLA-specific alloantibodies in recipi-    25. Kawase T et al (2007) High-risk HLA allele
    ents of unrelated hematopoietic cell                    mismatch combinations responsible for severe
    transplantation is predictive of graft failure.         acute graft-versus-host disease and implication
    Blood 115:2704–2708                                     for    its   molecular      mechanism.        Blood
20. Ottinger HD et al (2002) Positive serum cross-          110:2235–2241
    match as predictor for graft failure in HLA-        26. Wade JA et al (2007) HLA mismatching within
    mismatched allogeneic blood stem cell                   or outside of cross-reactive groups (CREGs) is
    transplantation. Transplantation 73:1280–1285           associated with similar outcomes after unrelated
21. Lau M, Park MS, Terasaki PI (1997)                      hematopoietic stem cell transplantation. Blood
    International cell exchange 1974–1996, a                109:4064–4070
    23-year documentation. (eds. Terasaki PI and        27. Duquesnoy R et al (2008) HLAMatchmaker-
    Gjertson DW. It was published by the UCLA               defined triplet matching is not associated with
    Tissue Typing Laboratory in Los Angeles                 better survival rates of patients with class I HLA
    California) 85–124                                      allele mismatched hematopoietic cell trans-
22. Noreen HJ et al (2001) Validation of DNA-               plants from unrelated donors. Biol Blood
    based HLA-A and HLA-B testing of volunteers             Marrow Transplant 14:1064–1071
    for a bone marrow registry through parallel         28. Spellman S, Klein J, Haagenson M, Askar M,
    testing with serology. Tissue Antigens                  Baxter-Lowe LA, He J, et al (2011) Scoring
    57:221–229                                              HLA Class I Mismatches by HistoCheck Does
23. Xiao Y et al (2009) Evaluating the potential            Not Predict Clinical Outcome in Unrelated
    impact of mismatches outside the antigen rec-           Hematopoietic Stem Cell Transplantation. Biol
    ognition site in unrelated hematopoietic stem           Blood Marrow Transplant 2011 Sep 29
    cell transplantation: HLA-DRB1*1454 and             29. Kamani N et al (2008) State of the art review:
    DRB1*140101. Tissue Antigens 73:595–598                 HLA matching and outcome of unrelated
24. Baxter-Lowe LA et al (2009) HLA-A dispari-              donor umbilical cord blood transplants. Biol
    ties illustrate challenges for ranking the impact       Blood Marrow Transplant 14:1–6
                                                                                        Chapter 31
Abstract
In recent years, several functional polymorphisms, particularly, SNPs have been identified in cytokines and
their receptor genes that regulate levels of cytokine expression. These have been implicated as immune
prognostic markers in diseases, including differential response to therapy and as biomarkers of graft out-
come following organ and stem cell transplantation. Population distribution of cytokine gene polymor-
phisms (CGPs) reveals significant variations in allele frequencies in different ethnic groups and this might
explain, to some extent, the observed differences in SNP associations with various diseases and immune-
pathologies. A number of molecular methods are available for defining CGPs. These include PCR-SSP,
AFLP, Taqman probe assays as well as sequencing based typing. Of these, the PCR-based sequence-specific
primer based test is the most widely accepted technique. This chapter describes steps involved in this pro-
cedure along with sources for procuring essential reagents. An important aspect of CGP analyses is the
correct interpretation of results particularly determination of their multilocus haplotypes.
Key words: Cytokinesm, Single nucleotide polymorphisms, PCR, Population diversity, SSP primers
1. Introduction
1.1. What Are                   Cytokines (Greek cyto-, cell; and -kinos, movement) are pleiotro-
Cytokines?                      pic immuomodulatory proteins produced by a wide range of cell
                                types through antigen-specific and nonspecific stimuli. The T-cell
                                subset paradigm and its immunological cytokine networking have
                                attracted much attention over the last decade. With the recent
                                knowledge on cytokine gene polymorphisms (CGPs), differences
                                between individuals have been discovered that influence not only
                                cytokine gene expression, but also susceptibility to diseases, their
                                progression, severity, and clinical outcomes. Ethnic differences
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_31, © Springer Science+Business Media New York 2012
                                                         549
550      G. Kaur and N. Mehra
1.2. Cytokine Genes     Genetic polymorphisms are frequently encountered in the 5¢ and
Polymorphisms           3¢ regulatory regions or introns of a cytokine and/ or its receptors
                        (Fig. 1) and these have been shown to play an important role in
                        regulating the extent of gene expression through one or more of
                        the following possible mechanisms: (1) by altering the structure of
                        transcription factor binding sites within promoters, (2) manipulat-
                        ing the structure of enhancers or silencers within introns or at dis-
                        tant regions, (3) altering the binding sites within nuclear matrix for
                        architectural transcription factors that modulate promoter activity
                        (1, 5, 6).
                             Cytokines and their receptor genes are generally conserved in
                        their exonic sequences with a few exceptions like IL4 receptor,
                        TNFb, TGFb, GM-CSF, and IL2 receptor-g gene (6). Most of the
                        observed mutations are synonymous, i.e., do not affect the protein
                        sequences, although they might influence cellular levels of the
                        cytokine expression. Broadly, three different approaches have been
                        employed for the study of CGPs. These include case-control
     31   Cytokine Gene Polymorphisms: Methods of Detection and Biological Significance                             551
Fig. 1. Commonly studied polymorphisms in human type 1, type II, and other cytokines and their receptors. The chromo-
somal localization, db SNP names, positions of functional polymorphisms in relation to gene start site and the nucleotide
substitutions are given.
1.3. Genotype–                  Recent reports have established that some CGPs correlate with the
Phenotype                       levels of cytokine expression and their phenotypic balance helps to
Correlations                    determine and/or predict the type of immune response. Table 1
                                summarizes the established cytokine genotype–phenotype correla-
                                tions determined largely on the basis of their in vitro levels of
                                expression (14–24).
1.4. Relevance                  Some genetic polymorphisms in cytokine genes affect their tran-
of Cytokine Gene                scription levels. Therefore, these polymorphisms may collectively
Polymorphisms                   modulate the net magnitude of cytokine-mediated immune
                                responses following transplantation or as a result of infection. This
                                underlies the importance of studying SNPs in relation to CGPs in
                                a variety of situations. Although CGPs play an important role in
552   G. Kaur and N. Mehra
                         Table 1
                         Established cytokine alleles/genotypes/haplotypes that
                         correlate with their in vitro expression levelsa
1.5. Methods of                  Several assays are available for defining CGPs or their effects on
Detecting Cytokine               cytokine expression and these can be prioritized depending on the
Gene Polymorphisms               study design and requirements. Their absolute or relative expres-
                                 sion correlates can be estimated using ELISA, CELISA, ELISPOT,
                                 real-time RT-PCR, Luminex monitoring, western blotting and
                                 immunohistochemistry in conjunction with densitometry, flow
                                 cytometry, and microarrays.
                                      Similarly, several techniques are available for studying the bial-
                                 lelic CGPs. These include PCR-SSP (or ARMS PCR), AFLP,
                                 Taqman probe assays, microarrays, and sequencing-based typing.
                                 Of these, the PCR-SSP is the most widely used and well-established
                                 procedure that has been adopted by numerous studies, including
                                 the 13IHWC as well as by the CTS, Heidelberg. Cytokine geno-
                                 typing SSP kits can be procured on request from the CTS,
                                 University of Heidelberg or from commercial vendors like One
                                 lambda, USA. Primers can also be designed individually using soft-
                                 ware like the Primer express and other online packages and in-house
                                 SSP assays can be developed.
                                      Figure 2 describes the basic principle of PCR-SSP procedure.
                                 The PCR primers are designed deliberately to include a perfect
                                 match with a single allele (i.e., a corresponding mismatch with the
                                 other allele). As a result of stringent PCR conditions, allele-specific
                                 amplification is initiated with only the perfectly matched primers
                                 and continued exponentially during the subsequent PCR cycles.
                                      The amplicons are resolved in agarose gels and interpreted. It
                                 is pertinent to include an internal control of reference for
                                 amplification in order to rule out amplification failures, if any. An
                                 internal control is also needed to check amplification related errors
                                 or variations, e.g., quality of template, carryover of salts and other
Fig. 2. Principle underlying the PCR-SSP test: (a) a perfectly matched primer leads to amplification while in (b) a mismatch
at the 3’end of the primer results in no amplification.
554      G. Kaur and N. Mehra
2. Materials
2.1. Extraction of DNA   Salting out procedure is one of the most commonly employed
(Salting-Out Procedure   methods of extraction of DNA and is described in this chapter
Using Ammonium           although a number of commercial kits are also available for DNA
Acetate)                 extraction from peripheral blood and other tissues.
                          1. Source of DNA—(see Note 1).
                          2. Table top Centrifuge.
                          3. ND-1000 Spectrophotometer (NanoDrop).
                          4. Incubator/water bath.
                          5. Vortex mixer.
                          6. Microfuge.
                          7. Autopippettes and tips.
                          8. 1.5-mL polypropylene microfuge tubes.
                          9. pH meter.
                         10. Autoclave.
                         11. Speed-Vac (optional).
                         Reagents
                          1. Preparation of red cell lysis buffer (RCLB).
                             (a) Prepare 1 M ammonium chloride (NH4Cl) Stock Solution:
                                 Dissolve 53.49 g of NH4Cl (M.W. 53.49) in 900 mL of
                                 sterile molecular grade H2O (Milli Q) and make up the
                                 final volume to 1 L.
                             (b) Prepare 1 M sodium bicarbonate (NaHCO3) Stock Solution:
                                 Dissolve 84.01 g of NaHCO3 (M.W. 84.01) in 900 mL of
                                 sterile H2O and bring the final volume to 1 L.
                             (c) Mix 144 mL of 1 M NH4Cl and 1 mL of 1 M NaHCO3
                                 and 855 mL of MilliQ H2O to obtain a final volume of 1 L
                                 and label as RCLB.
    31   Cytokine Gene Polymorphisms: Methods of Detection and Biological Significance   555
3. Methods
3.1.3. Nucleus Lysis        1. Resuspend the pellet obtained in Subheading 3.2, steps 1 and
and Protein Digestion          5 in 3 mL NLB (see Note 3).
                            2. Add 200 mL of 10% SDS followed by 20 mL (10 mg/mL) of
                               Proteinase-K solution. Mix thoroughly (see Note 4).
                            3. The mixture is kept at 37–42°C for 6 h to overnight. This is
                               the optimal temperature for maximum digestion. After
                               proper digestion, the solution becomes visibly clear with no
                               debris.
3.1.6. Storage of DNA           1. DNA is usually stored at 4°C for daily use and at −80°C for
                                   several months to years.
3.2. PCR-SSP Using             Each 96-well tray supplied by the CTS for cytokine genotyping
CTS Tray Kit                   contains prepipetted lyophilized primer mixes for two DNA , i.e.,
                               48 wells per individual. The PCR mix positions corresponding to
                               every locus specificities in the tray are shown in Fig. 3 and Table 2.
                               A separate master mix (CYT) is also supplied with the kit and con-
                               tains the reaction buffer but no Taq polymerase. The reader is
                               advised to refer to the procedure manual that is supplied along
                               with the kit and take a note of lot specific differences if any.
                                1. Program the thermal cycler (ABI 9700) as per following
                                   conditions and save the program (see Note 6).
558       G. Kaur and N. Mehra
Fig. 3. An example of a 2% agarose gel electrophoregram showing PCR-SSP genotyping for cytokine and receptor genes
using the CTS trays obtained from University of Heidelberg, Germany.
Table 2
List of various CGPs, their gene location, allele specificities, and amplicon sizes obtained using CTS PCR-SSP kits
                      3. Each CTS tray contains primers for genotyping two DNA sam-
                         ples. Use scissors to cut the tray into two halves. The PCR
                         reaction mix can be prepared in volumes depending on the
                         number of samples to be tested (see Notes 7–9).
                      4. Remove the CTS tray from the 4°C refrigerator. The lower
                         edge of the tray is marked with a black line. The starting posi-
                         tion in the tray is H1 and the series is oriented from H1 (bot-
                         tom) followed by G1, F1, etc., to A1 (top) until the last well
                         A6. The second set of the kit begins from H7 going on to A7
                         and so on until A12. See Table 2 for all the corresponding
                         positions on the tray.
                      5. Transfer 10 mL of reaction mix from Subheading 3.2, step 2
                         into each of the 48 wells (see Note 10).
                      6. Seal the PCR tray with Strip caps provided with the kit and
                         place in a thermal cycler that preprogrammed as in step 1
                         above. Ensure the caps completely seal the wells to prevent
                         evaporation.
                      7. Start thermal cycling, after completion, remove the tray and
                         proceed with gel electrophoresis.
                      8. A 2% agarose gel can be prepared and kept ready while the
                         PCR is running. To avoid contamination with PCR amplification
                         products, it is necessary to perform the test in two separate
                         working areas: the preamplification and the postamplification
                         areas.
564         G. Kaur and N. Mehra
3.3. Gel Electrophoresis    1. While the PCR is running (Subheading 3.2, step 7), a 2% agarose
                               gel can be poured and kept ready. For this, boil 25 agarose in
                               0.5× TBE, cool to ~60°C and add 5 mL ethidium bromide
                               (0.5 mg/mL final concentration). Mix and pour into gel tray
                               prearranged with combs.
                            2. Allow the gel to set at room temperature for 1 h.
                            3. After the PCR is over, remove the strip caps carefully from the
                               tray.
                            4. Remove the combs and ensure that wells are not broken. Place
                               the gel inside and just under the surface of electrophoresis run-
                               ning buffer (0.5× TBE). Load 10 mL amplicons starting from
                               H1 towards A1 as described in Fig. 3.
                            5. Connect the electrodes in the power-pack and run the electro-
                               phoresis at 100 V until just before the tracking dye exits the
                               gel.
                            6. Visualize the gel under UV illumination (312 nm) and save the
                               image or take a picture for interpretation and documentation.
3.4. Interpretation        Each of the primer mixes contain internal control primer pairs
of Results                 corresponding to either b-globin (90 bp) or CRP gene (440 bp).
                           Note the presence of these internal control bands in all the lanes
3.4.1. Positive Internal
                           as per their expected correct band sizes (Table 2 and Fig. 3). The
Control Amplification
                           440 bp band should be visible in lanes corresponding to IL1a,
                           IL1b, IL1R, IL1RA, IL4Ra, IL12, gIFN, TGFb, and TNFa.
                           Similarly, the 90 bp band should be observed in lanes correspond-
                           ing to IL 2, IL 4, IL 6, and IL 10.
3.4.2. Allele-Specific      1. Record the amplification patterns obtained for each of the
Amplification                  primer pairs in every well after confirmation of the correct
                               band sizes as shown in Table 2. The wells where allele-specific
                               amplicon is present may show weak or sometimes no internal
                               control band (see Note 11).
                            2. Some of the cytokine genotyping reagents in the kit allow
                               detection of certain haplotypes depending on characteristic
                               amplification patterns. Refer Table 2 for deducing correct
                               haplotypes.
                            3. Disregard any products of incorrect size whether strong or
                               weak (see Note 12).
                            4. A primer dimer diffused band of <80 bp may be observed
                               occasionally but can be distinguished from the allele-specific
                               amplifications which are relatively sharper and usually 80 bp or
                               more in size.
                            5. False negatives, if any, may be caused by inefficient amplification
                               or poor quality of DNA or variations in thermal cycler and can
                               be ruled out individually.
     31   Cytokine Gene Polymorphisms: Methods of Detection and Biological Significance           565
3.4.3. An Example         The first observation to look for is an internal control amplification
of Interpretation         of 90 bp (Fig. 3, lanes 43–48). Now, refer to Table 2 for expected
of Allele-Specific        band sizes for allele-specific amplifications relating to IL10 mixes
Amplification             1–6 corresponding to the same lanes 43–48. As can be seen in
for IL 10                 Fig. 3, there is a positive band of 305 bp (mix 1) in lane number
                          43 and a band of size 530 bp (mix 2) in lane 44 while all the
                          remaining lanes 45–48 do not show any allele-specific
                          amplification(s) for IL 10. A combined interpretation of GC*
                          and G*C as suggested against lanes 43 and 44 (Table 2) hence
                          suggests that the individual tested is homozygous for the GCC
                          haplotype.
4. Notes
Acknowledgements
References
 1. Hollegaard MV, Bidwell JL (2006) Cytokine              Greek Cypriot population. Eur J Immmunogenet
    gene polymorphism in human disease: on-line            30:341–343
    databases, supplement 3. Genes Immun               13. Hoffmann SC, Stanley EM, Cox ED et al
    7:269–276                                              (2002) Ethnicity greatly influences cytokine
 2. Kaur G, Mehra N (2009) Genetic determinants            gene polymorphism distribution. Am J
    of HIV-1 infection and progression to AIDS:            Transplant 2(6):560–567
    immune response genes. Tissue Antigens             14. Dudler J, Li J, Pagnotta M et al (2006) Gene
    74:373–385                                             transfer of programmed death ligand-1.Ig pro-
 3. Ryan AW, Mapp J, Myna S et al (2006) Levels            longs cardiac allograft survival. Transplantation
    of interpopulation differentiation among dif-          82(12):1733–1737
    ferent functional classes of immunologically       15. Hahn AB, Kasten-Jolly JC, Conctantino DM
    important genes. Genes and Immunity                    et al (2001) TNF-alpha, IL-6, IFN-gamma,
    7:179–183                                              and IL-10 gene expression polymorphisms and
 4. Mytilineos J, Laux G, Opelz G (2004) Relevance         the IL-4 receptor alpha-chain variant Q576R:
    of IL10, TGFb1, TNFa and IL4Ra gene poly-              effects    on     renal    allograft     outcome.
    morphisms in kidney transplantation: a collab-         Transplantation 72(4):660–665
    orative transplant study report. Am J Transplant   16. Pociot F, Mølvig J, Wogensen L et al (1992) A
    4:1684–1690                                            TaqI polymorphism in the human interleukin-1
 5. Bidwell J, Keen L, Gallagher G et al (1999)            beta (IL-1 beta) gene correlates with IL-1 beta
    Cytokine gene polymorphism in human dis-               secretion in vitro. Eur J Clin Invest 22(6):
    ease: on-line databases. Genes Immun 1:3–19            396–402
 6. Bidwell J, Keen L, Gallagher G et al (2001)        17. Awad MR, El-Gamel A, Hasleton P et al (1998)
    Cytokine gene polymorphism in human dis-               Genotypic variation in the transforming growth
    ease: on-line databases, supplement 1. Genes           factor-beta1 gene: association with transform-
    Immun 2:61–70                                          ing growth factor-beta1 production, fibrotic
 7. Kaur G, Rapthap CC, Kumar N et al (2007)               lung disease, and graft fibrosis after lung trans-
    Frequency distribution of cytokine gene poly-          plantation. Transplantation 66(8):1014–1020
    morphisms in the healthy North Indian popula-      18. Pociot F, Wilson AG, Nerup J et al (1993) No
    tion. Tissue Antigens 69:113–120                       independent association between a tumor
 8. Pyo CW, Hur SS, Kim YK et al (2003)                    necrosis factor-alpha promotor region poly-
    Polymorphisms of IL1B, IL1RN, IL2, IL4,                morphism and insulin-dependent diabetes mel-
    IL6, IL10 and IFNg genes in the Korean popu-           litus. Eur J Immunol 23(11):3050–3053
    lation. Hum Immunol 64:979–989                     19. Fishman D, Faulds G, Jeffery R (1998) The
 9. Kurzawski M, Pawlik A, Czerny B et al (2005)           effect of novel polymorphisms in the interleu-
    Frequencies of the common promoter poly-               kin-6 (IL-6) gene on IL-6 transcription and
    morphisms in cytokine genes in a polish popu-          plasma IL-6 levels, and an association with
    lation. Int J Immunogenet 32:285–291                   systemic-onset juvenile chronic arthritis. J Clin
10. Uboldi de Capei MU, Dametto E, Fasano ME               Invest 102(7):1369–1376
    et al (2003) Genotyping for cytokine               20. Terry CF, Loukaci V, Green FR (2000)
    polymorphisms: allele frequencies in the Italian       Cooperative influence of genetic polymor-
    population. Eur J Immunogenet 30:5–10                  phisms on interleukin 6 transcriptional regula-
11. Trajkov D, Arsov T, Petlichkovski A et al (2005)       tion. J Biol Chem 275(24):18138–18144
    Cytokine gene polymorphisms in population of       21. Turner DM, Williams DM, Sankaran D et al
    ethnic Macedonians. Croat Med J 46(4):685–692          (1997) An investigation of polymorphism in
12. Costeas PA, Koumas L, Koumouli A et al (2003)          the interleukin-10 gene promoter. Eur J
    Cytokine polymorphism frequencies in the               Immunogenet 24(1):1–8
568       G. Kaur and N. Mehra
22. Crawley E, Woo P, Isenberg DA et al (1999)         25. Wang K, Li M, Hakonarson H (2010) Analysing
    Single nucleotide polymorphic haplotypes of            biological pathways in genome-wide associa-
    the interleukin-10 5¢ flanking region are not          tion studies. Nat Rev Genet 11:843–854
    associated with renal disease or serology in
    Caucasian patients with systemic lupus erythe-
    matosus. Arthritis Rheum 42(9):2017–2018
                                                       Useful Website Links
23. van der Pouw Kraan TC, van Veen A, Boeije
    LC et al (1999) An IL-13 promoter polymor-         h t t p : // w w w 3 . n c b i . n l m . n i h . g o v / O m i m /
    phism associated with increased risk of allergic         searchomim.html
    asthma. Genes Immun 1(1):61–65
24. Mullighan CG, Marshall SE, Bunce M et al           http://www.immport.org/immportWeb/que-
    (1999) Variation in immunoregulatory genes             ryref/geneListSummary.do
    determines the clinical phenotype of common        http://snpper.chip.org/
    variable immunodeficiency. Genes Immun             http://genome.cse.ucsc.edu/
    1:137–148                                          http://cmbi.bjmu.edu.cn/cmbidata/cgf/
                                                                                        Chapter 32
Abstract
IMGT/V-QUEST is the highly customized and integrated online IMGT® tool for the standardized analysis
of the immunoglobulin (IG) or antibody and T cell receptor (TR) rearranged nucleotide sequences. The
analysis of these antigen receptors represents a crucial challenge for the study of the adaptive immune response
in normal and disease-related situations. The expressed IG and TR repertoires represent a potential of 1012
IG and 1012 TR per individual. This huge diversity results from mechanisms that occur at the DNA level dur-
ing the IG and TR molecular synthesis. These mechanisms include the combinatorial rearrangements of the
variable (V), diversity (D) and joining (J) genes, the N-diversity (deletion and addition at random of nucle-
otides during the V-(D)-J rearrangement) and, for IG, somatic hypermutations. IMGT/V-QUEST identifies
the V, D, J genes and alleles by alignment with the germline IG and TR gene and allele sequences of the
IMGT reference directory. The tool describes the V-REGION mutations and identifies the hot spot posi-
tions in the closest germline V gene. IMGT/V-QUEST integrates IMGT/JunctionAnalysis for a detailed
analysis of the V-J and V-D-J junctions and IMGT/Automat for a complete annotation of the sequences
and also provides IMGT Collier de Perles. IMGT/HighV-QUEST, the high-throughput version of IMGT/
V-QUEST, implemented to answer the needs of deep sequencing data analysis from Next Generation
Sequencing (NGS), allows the analysis of thousands of IG and TR sequences in a single run. IMGT/V-
QUEST and IMGT/HighV-QUEST are available at the IMGT® Home page, http://www.imgt.org.
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_32, © Springer Science+Business Media New York 2012
                                                         569
570   E. Alamyar et al.
2. IMGT/V-QUEST
2.1. IMGT/V-QUEST                 1. In the IMGT/V-QUEST Welcome page (Fig. 1), locate the
Welcome Page                         section that corresponds to the receptor type (IG or TR) of
                                     your sequences (see Note 1).
                                  2. Locate the species or taxon that corresponds to the IMGT ref-
                                     erence directory against which you want your sequences to be
                                     analyzed (see Note 2).
                                  3. Click on the species (for instance “Human”) or taxon. This
                                     will give access to the IMGT/V-QUEST Search page (Fig. 2).
2.2.2. Selection              The user can choose between three types of display for the results:
of the Result Display         “Detailed view,” “Synthesis view,” or “Excel file” and the para-
                              meters of the analysis can be customized with a large choice of
                              options in “Advanced parameters” (Fig. 2).
572        E. Alamyar et al.
Fig. 1. IMGT/V-QUEST Welcome page. The two upper sections correspond to the immunoglobulin (IG) or antibody and T cell
receptor (TR) analyses, respectively. The lowest section is for download of the IMGT reference directory sets.
                                 1. Detailed view.
                                         “Detailed view” provides the results for each sequence,
                                    individually. They may be displayed in HTML (by default) or
                                    Text format, with a user-defined number of nucleotides per
                                    line in alignments (60 by default). There is a choice of 14 dif-
                                    ferent result displays for the analysis of the V-DOMAIN. The
                                    results the most commonly requested are selected by default.
                                    You can check or uncheck the result displays as needed:
                                    (a) Alignment for V-GENE.
                                    (b) Alignment for D-GENE.
                                    (c) Alignment for J-GENE.
                                    (d) “Results of IMGT/JunctionAnalysis” (9) (with or without
                                        full list of eligible D-GENEs).
                                    (e) Sequence of the JUNCTION (“nt” and “AA”).
                            32   IMGT® Tools for the Nucleotide Analysis of Immunoglobulin…                   573
Fig. 2. The IMGT/V-QUEST Search page. The Search page is that for IG: “Analyse your immunoglobulin (IG) or antibody
sequences.” Your species (or taxon) selection from the IMGT/V-QUEST Welcome page is recalled at the top of the page
(here “Human”).
574   E. Alamyar et al.
                         (c) nt-sequences.
                         (d) IMGT-gapped-AA-sequences.
                         (e) AA-sequences.
                         (f) Junction.
                         (g) V-REGION-mutation-and-AA-change-table.
                         (h) V-REGION-nt-mutation-statistics.
                         (i) V-REGION-AA-change-statistics.
                         (j) V-REGION-mutation-hot-spots.
                         (k) Parameters.
2.2.3. Advanced    Default values of “Advanced parameters” have been set for classical
Parameters         analysis. They should be modified for more sophisticated queries
                   or for unusual sequences (e.g., with insertions or deletions). The
                   options include:
                       1. “Selection of IMGT reference directory set” with a choice of
                          four sets:
                         (a) F + ORF.
                         (b) F + ORF + in frame P (by default).
                         (c) F + ORF including orphons.
                         (d) F + ORF + in frame P including orphons.
                              Where F is functional, ORF is open reading frame, P is
                          pseudogene. This allows sequences to be compared with only
                          relevant gene sequences (e.g., orphon sequences are relevant
                          for genomic but not for expressed repertoire studies). The
                          selected set can also be chosen either “With all alleles” or
                          “With allele *01 only” (see Note 4).
                       2. “Search for insertions and deletions in V-REGION” (“Yes” or
                          “No”): somatic hypermutations by nucleotide insertions and
                          deletions in the V-REGION are rare events that are known to
                          occur in normal and malignant cells (16). By default, IMGT/
                          V-QUEST does not search for insertions and deletions. If “Yes”
                          is selected, the analysis is slower and the number of submitted
                          sequences in a single run is limited to ten (see Note 5).
                       3. Parameters for IMGT/JunctionAnalysis:
                          (a) “Nb of accepted D-GENE”: this selection is provided for
                              the IGH junctions (for the IG) and for the TRB and TRD
                              junctions (for the TR).
                          (b) “Nb of accepted mutations” in 3¢V-REGION,
                              D-REGION, and 5¢J-REGION (by default, no mutation
                              is accepted for the TR junctions. For IGH, 2, 4, and 2
                              mutations are accepted in the 3¢V-REGION, D-REGION,
                              and 5¢J-REGION, respectively. For the IGK and IGL loci,
576        E. Alamyar et al.
2.3. IMGT/V-QUEST              The top of “Detailed view” results are displayed in “A. Detailed
Output for Detailed            results for the IMGT/V-QUEST analyzed sequences.” The top of
View                           this page indicates the number of analyzed sequences with links to
                               individual results. Use the link associated with a sequence name to
                               go directly to its individual result that comprises the sequence and
                               “Result summary” followed by the different displayed results as
                               selected in the Search page.
2.3.1. Sequence                The sequence and “Result summary” are shown at the top of each
and Result Summary             individual result (Fig. 3). The IMGT reference directory set against
                               which the sequence was analyzed (e.g., human IG set) is indicated
Fig. 3. IMGT/V-QUEST “Detailed view” result page: Sequence and “Summary.” The upper part of an individual result shows
the sequence and “Result summary.” The seq1 is the human IGH rearranged sequence with the accession number
AB012909 in the IMGT/LIGM-DB database (34).
                              32   IMGT® Tools for the Nucleotide Analysis of Immunoglobulin…    577
2.3.3. Alignments for V, D,    The alignments for V, D, J genes and alleles, if selected in the
and J Genes and Alleles        Search page, display the alignments with the five closest V, D, and
                               J gene alleles, respectively, with their alignment score and their
578        E. Alamyar et al.
Fig. 4. IMGT/V-QUEST “Detailed view” result page: Sequence and “Result summary” with the option “Search for insertions
and deletions.” An insertion of 9 nt was detected by IMGT/V-QUEST. The analysis is then performed after having removed
the insertion.
                               identity percentage (Fig. 5). The alignment for D-GENE and allele
                               should be considered with caution since it may show discrepancies
                               with the results obtained by IMGT/JunctionAnalysis (9) (see
                               Note 12).
Fig. 5. IMGT/V-QUEST “Detailed view” result page: “Alignment for V-GENE and allele identification” and “Alignment for J-GENE and allele identification.” Dashes indicate identical
nucleotides. Dots indicate gaps. FR-IMGT and CDR-IMGT are delimited according to the IMGT unique numbering (19–21). The seq1 is the human IGH rearranged sequence with the
                                                                                                                                                                                     579
Fig. 6. IMGT/V-QUEST “Detailed view” result page: “Results of IMGT/JunctionAnalysis” (with “Analysis of the JUNCTION,”
“Eligible D genes” and “Translation of the JUNCTION”) and “Sequence of the JUNCTION (‘nt’ and ‘AA’).” The seq1 is the
human IGH rearranged sequence with the accession number AB012909 in the IMGT/LIGM-DB database (34).
                        32    IMGT® Tools for the Nucleotide Analysis of Immunoglobulin…     581
2.3.5. “V-REGION         The results provide three displays of the V-REGION (Fig. 7):
Alignment,” “V-REGION
Translation,” and            1. The “V-REGION alignment according to the IMGT unique
“V-REGION Protein               numbering” displays the nt sequences with the FR-IMGT and
Display”                        CDR-IMGT delimitations according to the IMGT unique
                                numbering (19–21).
582        E. Alamyar et al.
Fig. 7. IMGT/V-QUEST “Detailed view” result page: Three displays of the V-REGION. The seq1 is the human IGH rearranged
sequence with the accession number AB012909 in the IMGT/LIGM-DB database (34). Only a part of each of the three
displays is shown.
                             32    IMGT® Tools for the Nucleotide Analysis of Immunoglobulin…    583
2.3.6. Analysis of the        The analysis of the mutations in the V-REGION is performed by
Mutations: “V-REGION          comparison of the analyzed sequence with the closest germline V
Mutation and AA Change        gene and allele.
Table,” “V-REGION
                                  1. The “V-REGION mutation and AA change table” (Fig. 8)
Mutation and AA Change
                                     lists the nt mutations and the corresponding AA changes if the
Statistics,” and “V-REGION
                                     mutations are not silent. They are described for each FR-IMGT
Mutation Hot Spots”
                                     and CDR-IMGT, with their nt and codon position according
                                     to the IMGT unique numbering (19–21) and for the AA
                                     changes according to the IMGT AA classes (18). For example,
                                     c1 > g, Q1 > E (++−) means that the nt mutation (c > g) at nt 1
                                     leads to an AA change (Q > E) at codon 1 with the same
                                     hydropathy (+) and volume (+), but with different physico-
                                     chemical properties (−) classes. It is the first time that such
                                     qualification of amino acid replacement is provided. This has
                                     led to identify four types of AA changes: very similar (+++),
                                     similar (++−, +−+), dissimilar (−−+, −+−, +−−), and very dis-
                                     similar (−−−).
                                  2. The “V-REGION mutation and AA change statistics” (Fig. 8)
                                     comprises two tables, the first one for “Nucleotide (nt) muta-
                                     tions” described as silent/nonsilent, transitions/transversions,
                                     and the second one for “Amino acid (AA) changes.” For both
                                     tables, results are given for the V-REGION and for FR-IMGT
                                     and CDR-IMGT. Statistics are calculated up to the 3¢ end of
                                     the V-REGION identified in the input sequence (this includes
                                     the 3¢ last two identical nucleotides with the closest germline
                                     V-REGION). The numbers in parentheses, in the V-REGION
                                     and CDR3-IMGT columns, correspond to the statistics calcu-
                                     lated up to the 3¢ end of the closest germline V-REGION and
                                     therefore may include nt and AA differences due to the junc-
                                     tion diversity.
                                  In order to avoid to count sequence differences due to the 5¢
                              primer in “V-REGION mutation and AA change table” and in
                              “V-REGION mutation and AA change statistics,” before launching
                              the analysis it is possible to exclude in 5¢ of the V-REGION, a
                              given number of nucleotides, defined by the user, in “Advanced
584        E. Alamyar et al.
Fig. 8. IMGT/V-QUEST “Detailed view” result page: “V-REGION mutation and AA change table” and “V-REGION mutation and
AA change statistics.” Results are for the human IGH rearranged sequence AB012909 of the IMGT/LIGM-DB database (34)
by comparison with the germline IGHV3-30*04.
Fig. 9. IMGT/V-QUEST “Detailed view” result page: “V-REGION mutation hot spots (motifs and localization of hot spots in
germline V-REGION).” Mutation hot spots are those of the human IGHV3-30*04 allele.
2.3.7. Sequences of V-,        This result provides nt and AA sequences with gaps according to
V-J- or V-D-J-REGION           the IMGT unique numbering (19–21) and includes (Fig. 10) the
(“nt” and “AA”) with Gaps      “V-REGION nucleotide sequence in FASTA format” with access
in FASTA and Access            to the IMGT/PhyloGene tool (22), “V-REGION amino acid in
to IMGT/PhyloGene              FASTA format,” “V-REGION amino acid on one line,” V-J or
for V-REGION (“nt”)            V-D-J-REGION nt and AA sequences in FASTA format, and V-J or
                               V-D-J-REGION AA sequence on one line. In that option, the J read-
                               ing frame of sequences with an out-of-frame junction is not restored
                               (the sequence does not include “#”) and a note alerts the user.
2.3.8. Annotation by IMGT/     The results of the analysis of the input sequence by IMGT/
Automat                        Automat (23, 24) provide a full automatic annotation for the V-J-
                               REGION or V-D-J-REGION using IMGT® standardized labels
                               (Fig. 11).
586        E. Alamyar et al.
Fig. 10. IMGT/V-QUEST “Detailed view” result page: V-REGION and V-(D)-J-REGION sequences with IMGT gaps. Sequences
(“nt” and “AA”) are for the human IGH rearranged sequence AB012909 of the IMGT/LIGM-DB database (34). The 3¢ end of
the “on one line” sequences are not shown owing to the figure format.
2.3.9. IMGT Colliers           The “IMGT Collier de Perles” (25–27) can be displayed either as
de Perles                      a “link to IMGT/Collier-de-Perles tool” or as a direct “IMGT
                               Collier de Perles (for a number of sequences <5)” representation
                               integrated in IMGT/V-QUEST results (Fig. 12), depending on the
                               user selection in the IMGT/V-QUEST Search page (see Note 16).
2.4. IMGT/V-QUEST              The “Synthesis view” results are displayed in “B. Synthesis for the
Output for “Synthesis          IMGT/V-QUEST analyzed sequences.” At the top of the page,
View”                          the number of analyzed sequences is indicated.
2.4.1. Summary Table           The “Summary table” (Fig. 13) displays one row for each input
                               sequence with the corresponding results, including: (1) the name
                               of the sequence (Sequence ID), (2) the name of the closest
                          32    IMGT® Tools for the Nucleotide Analysis of Immunoglobulin…               587
Fig. 11. IMGT/V-QUEST “Detailed view” result page: “Annotation by IMGT/Automat.” Results are for the human IGH
rearranged sequence AB012909 of the IMGT/LIGM-DB database (34). Only a part of the annotation is shown.
                             V-GENE and allele (see Note 7), (3) the functionality of the
                             sequence (when found, the presence of stop codons is indicated),
                             (4) the V-REGION score (see Note 8), (5) the V-REGION per-
                             centage of identity with, between parentheses, the ratio of number
                             of identical nucleotides (nt)/number of aligned nt (see Note 17),
                             (6) the name of the closest J-GENE and allele (see Note 18), (7)
                             and, provided according to the IMGT/JunctionAnalysis results,
                             the D-GENE and allele name, the D reading frame, the CDR-
                             IMGT lengths, the AA JUNCTION, and the JUNCTION frame.
                             In the absence of results of IMGT/JunctionAnalysis, only the AA
                             JUNCTION defined by IMGT/V-QUEST is displayed.
2.4.2. Results of IMGT/      Below the “Summary table,” use the links associated to the locus
JunctionAnalysis             name(s) (e.g., IGH) to display “Results of IMGT/JunctionAnalysis”
                             for sequences identified by the tool as belonging to the same locus
588        E. Alamyar et al.
Fig. 12. IMGT/V-QUEST “Detailed view” result page: IMGT Collier de Perles. The IMGT Collier de Perles was obtained with
the option “IMGT Collier de Perles (for a nb of sequences <5)” of the human IGH rearranged sequence AB012909 of the
IMGT/LIGM-DB database (34). Anchor positions are shown as squares. In FR-IMGT, the hydrophobic amino acids (hydropa-
thy index with positive value) and tryptophan (W) found at a given position in more than 50% of sequences are displayed
(online with a blue background color).
                               (Fig. 14). Results are displayed for the IGH, IGK, or IGL locus for
                               IG sequences, and for the TRA, TRB, TRG, or TRD locus for TR
                               sequences. They are similar to those obtained for individual
                               sequences detailed in Subheading 2.3.4.
2.4.3. Alignment with the      The synthesis results provide six different displays (if all were
Closest Alleles                selected): “Alignment for V-GENE,” “V-REGION alignment
                               according to the IMGT unique numbering” and “V-REGION
                               translation” (Fig. 15), and “V-REGION protein display” in three
                               different formats (Fig. 16).
2.4.4. V-REGION Most           This section shows, for each FR-IMGT and CDR-IMGT, and for
Frequently Occurring AA        each position according to the IMGT unique numbering (19–21),
Per Position and Per           the most frequently occurring amino acid (AA) (Fig. 17).
FR-IMGT and CDR-IMGT
                                                                                                                                                                        32
Fig. 13. IMGT/V-QUEST “Synthesis view” result page: Top of the page: The seq1, seq2, seq3, seq4, seq5, seq6, and seq7 correspond to the DQ100777, AB021524, AB012909,
AB021511, AB021516, AB021514, and AB021539 accession numbers, respectively, from the IMGT/LIGM-DB database (34).
                                                                                                                                                                        IMGT® Tools for the Nucleotide Analysis of Immunoglobulin…
                                                                                                                                                                        589
                                                                                                                                                                                590
                                                                                                                                                                                E. Alamyar et al.
Fig. 14. IMGT/V-QUEST “Synthesis view” result page: Results of IMGT/JunctionAnalysis. The results show the “Analysis of the JUNCTIONs” and the “Translation of the JUNCTIONs”
for the IGH sequences of the submitted analysis. Owing to the figure format, Vmut, Dmut, Jmut, Ngc, molecular mass, and pI are not shown.
Fig. 15. IMGT/V-QUEST “Synthesis view” result page: “Alignment for V-GENE,” “V-REGION alignment according to the IMGT
unique numbering,” and “V-REGION translation.” Displayed sequences are those of the input set, which have been identified
as using the same closest germline V gene and allele (here IGHV3-30*04). Dashes indicate identical nucleotides. Dots
indicate gaps. FR-IMGT and CDR-IMGT are delimited according to the IMGT unique numbering (19–21). The sequence of
the V-REGION of the closest germline V is shown on the first line and the input sequences are aligned, with nt mutations
indicated by comparison. Underlined nucleotides in the closest germline V correspond to mutation hot spot motif localizations.
Only a part of each of the three displays is shown.
                                                                                                                                                                                    592
                                                                                                                                                                                    E. Alamyar et al.
Fig. 16. IMGT/V-QUEST “Synthesis view” result page. V-REGION protein displays: Three different displays are shown. “V-REGION protein display,” “V-REGION protein display with
colored AA according to the AA IMGT classes,” (online colored AA) and “V-REGION protein display (only AA changed displayed).” The first line shows the AA sequence of the closest
V-REGION allele against which are aligned the translations of the input sequences identified by IMGT/V-QUEST as using the same V gene and allele (here IGHV3-30*04).
                                                                                                                                                                                     32
                                                                                                                                                                                     IMGT® Tools for the Nucleotide Analysis of Immunoglobulin…
Fig. 17. IMGT/V-QUEST “Synthesis view” result page: “V-REGION most frequently occurring AA per position and per FR-IMGT and CDR-IMGT.” Results are for a set of four sequences
expressing IGHV3-30*04 (accession numbers AB012909, AB021511, AB021516, AB021514 from IMGT/LIGM-DB database (34)). For each set of user sequences that express the
same closest germline V gene and allele, the most frequently occurring amino acid is shown per amino acid (or codon) position and per FR-IMGT and CDR-IMGT. The display highlights
the conserved as well as the polymorphic positions.
                                                                                                                                                                                     593
594       E. Alamyar et al.
2.5. IMGT/V-QUEST             “Excel file” allows the users to open and save a spreadsheet includ-
Output for Excel File         ing the results of the IMGT/V-QUEST analysis. The file contains
                              11 sheets (if all were selected in the IMGT/V-QUEST Search
                              page, see Subheading 2.2.2). The “Summary” and “Parameters”
                              are always selected.
                                  The content of the eleven sheets is detailed in the IMGT/
                              HighV-QUEST “Download” (Subheading 3.4).
3. IMGT/
HighV-QUEST
                              IMGT/HighV-QUEST, the high-throughput version of IMGT/
                              V-QUEST, can analyze large numbers of IG and TR-rearranged
                              sequences (up to 150,000) in a single run.
3.1. IMGT/HighV-              As for the other IMGT® databases and tools, IMGT/HighV-
QUEST Welcome Page            QUEST is freely available for academics. However, the IMGT/
                              HighV-QUEST Welcome page requires user identification and
                              provides, for new users, a link to register. User identification has
                              been set to avoid nonrelevant use and overload of the server and to
                              contact the user if needed.
3.3. IMGT/HighV-             From their account, the user can check the status of his/her analyses
QUEST “Analysis              at any time by displaying the “Analysis history” (Fig. 19) (available
History Page”                from a link from the top of the Search page). The table displays
                             each analysis with its title, the user name, the status of the analysis,
                             the submission date with a predicted completion time, the number
                             of submitted sequences, the species and the receptor type or locus
                             (as selected by the user), and the actions that can be performed by
                             the user. A user may cancel a queued or a running analysis at any
                             time. When the analysis is completed, the user can download the
                             results as a single file in ZIP format. The size of the ZIP file and the
                             number of included files are indicated in the table (Fig. 19). If
                             the user removes a completed analysis, all related files (sequences
                             and results) are definitively deleted from the server.
3.4. IMGT/HighV-             The downloaded ZIP file (Fig. 20) contains a main folder with
QUEST “Download”             eleven files (equivalent to the eleven sheets of the excel file pro-
                             vided by the classical IMGT/V-QUEST) in CSV format, and one
                             subfolder with individual files, in Text, for each sequence (provid-
                             ing “Detailed view” results). Text and CSV formats have been cho-
                             sen in order to facilitate statistical studies for further interpretation
                             and knowledge extraction. The eleven files (the “Summary” and
                             “Parameters” files are always provided) comprise:
                              1. The “Summary” file (25–29 columns) provides the synthesis
                                 of the analysis (the sequence names, the sequence functional-
                                 ity, the names of the closest V, D, and J genes and alleles with
                                 identity percentage, FR-IMGT and CDR-IMGT lengths,
                                 amino acid JUNCTION, the description of insertions and
                                 deletions if any …).
                              2. The “IMGT-gapped-nt-sequences” file (18 columns) includes
                                 the sequence identification (name, functionality, names of the
                                 closest V, D, and J genes and alleles), the nucleotide sequences
                                 that have been gapped according to the IMGT unique num-
                                 bering (19–21) for V-D-J-REGION, V-J-REGION,
                                 V-REGION, FR1-IMGT, CDR1-IMGT, FR2-IMGT, CDR2-
                                 IMGT, FR3-IMGT, and the nucleotide sequences of CDR3-
                                 IMGT, JUNCTION, J-REGION, and FR4-IMGT.
                              3. The “nt-sequences” file (63–78 columns) includes the sequence
                                 functionality, the names of the closest V, D, and J genes and
                                 alleles, and the nucleotide sequences of all labels that can be
                                 automatically annotated by IMGT/Automat (23, 24) in the
                                 analyzed sequences.
32   IMGT® Tools for the Nucleotide Analysis of Immunoglobulin…                             597
4. Notes
Acknowledgements
References
 1. Lefranc M-P, Lefranc G (2001) The immuno-          12. Lefranc M-P, Giudicelli V, Regnier L, Duroux
    globulin FactsBook. Academic Press, London,            P (2008) IMGT, a system and an ontology that
    UK, pp 1–458                                           bridge biological and computational spheres in
 2. Lefranc M-P, Lefranc G (2001) The T cell               bioinformatics. Brief Bioinform 9:263–275
    receptor FactsBook. Academic Press, London,        13. Duroux P, Kaas Q, Brochet X, Lane J, Ginestoux
    UK, pp 1–398                                           C, Lefranc M-P, Giudicelli V (2008) IMGT-
 3. Giudicelli V, Chaume D, Lefranc M-P (2004)             Kaleidoscope, the formal IMGT-ONTOLOGY
    IMGT/V-QUEST, an integrated software pro-              paradigm. Biochimie 90:570–583
    gram for immunoglobulin and T cell receptor        14. Alamyar E, Giudicelli V, Duroux P, Lefranc
    V-J and V-D-J rearrangement analysis. Nucleic          M-P (2010) IMGT/HighV-QUEST: a high-
    Acids Res 32:W435–W440                                 throughput system and web portal for the anal-
 4. Lefranc M-P (2004) IMGT, the international             ysis of rearranged nucleotide sequences of
    ImMunoGeneTics          information      system.       antigen receptors-high-throughput version of
    Methods Mol Biol 248:27–49                             IMGT/V-QUEST. Journées Ouvertes en
 5. Giudicelli V, Lefranc M-P (2005) Interactive           Biologie, Informatique et Mathématiques,
    IMGT on-line tools for the analysis of immu-           September 7-9, 2010, Montpellier, France
    noglobulin and T cell receptor repertoires. In:        (JOBIM 2010), Poster #60, http://www.
    Veskler BA (ed) New research on immunology.            jobim2010.fr/?q=fr/node/55. Accessed 31
    Nova Science Publishers, Inc., New York, pp            Jan 2011
    77–105                                             15. Giudicelli V, Chaume D, Lefranc M-P (2005)
 6. Brochet X, Lefranc M-P, Giudicelli V (2008)            IMGT/GENE-DB: a comprehensive database
    IMGT/V-QUEST: the highly customized and                for human and mouse immunoglobulin and T
    integrated system for IG and TR standardized           cell receptor genes. Nucleic Acids Res
    V-J and V-D-J sequence analysis. Nucleic Acids         33:D256–D261
    Res 36:W503–W508                                   16. Belessi C, Davi F, Stamatopoulos K, Degano
 7. Giudicelli V, Lefranc M-P (2008) IMGT®                 M, Andreou TM, Moreno C, Merle-Béral H,
    standardized analysis of immunoglobulin rear-          Crespo M, Laoutaris NP, Montserrat E,
    ranged sequences. In: Ghia P, Rosenquist R,            Caligaris-Cappio F, Anagnostopoulos AZ, Ghia
    Davi F (eds) Immunoglobulin gene analysis in           P (2006) IGHV gene insertions and deletions
    chronic lymphocytic leukemia (chapter 2).              in chronic lymphocytic leukemia: “CLL-biased”
    Wolters Kluwer Health, Italy, pp 33–52                 deletions in a subset of cases with stereotyped
                                                           receptors. Eur J Immunol 36:1963–1974
 8. Lefranc M-P, Giudicelli V, Ginestoux C,
    Jabado-Michaloud J, Folch G, Bellahcene F,         17. Ghia P, Stamatopoulos K, Belessi C, Moreno
    Wu Y, Gemrot E, Brochet X, Lane J, Regnier             C, Stilgenbauer S, Stevenson F, Davi F,
    L, Ehrenmann F, Lefranc G, Duroux P (2009)             Rosenquist R (2007) ERIC recommendations
    IMGT®, the international ImMunoGeneTics                on IGHV gene mutational status analysis in
    information system®. Nucleic Acids Res                 chronic lymphocytic leukemia. Leukemia 21:1–3
    37:D1006–D1012                                     18. Pommié C, Levadoux S, Sabatier R, Lefranc G,
 9. Yousfi Monod M, Giudicelli V, Chaume D,                Lefranc M-P (2004) IMGT standardized crite-
    Lefranc M-P (2004) IMGT/JunctionAnalysis:              ria for statistical analysis of immunoglobulin
    the first tool for the analysis of the immuno-         V-REGION amino acid properties. J Mol
    globulin and T cell receptor complex V-J and           Recognit 17:17–32
    V-D-J        JUNCTIONs.           Bioinformatics   19. Lefranc M-P (1997) Unique database number-
    20:i379–i385                                           ing system for immunogenetic analysis.
10. Giudicelli V, Lefranc M-P (1999) Ontology for          Immunol Today 18:509
    immunogenetics: the IMGT-ONTOLOGY.                 20. Lefranc M-P (1999) The IMGT unique num-
    Bioinformatics 15:1047–1054                            bering for Immunoglobulins, T cell receptors
11. Lefranc M-P, Giudicelli V, Ginestoux C, Bosc           and Ig-like domains. Immunologist 7:
    N, Folch G, Guiraudou D, Jabado-Michaloud              132–136
    J, Magris S, Scaviner D, Thouvenin V, Combres      21. Lefranc M-P, Pommié C, Ruiz M, Giudicelli V,
    K, Girod D, Jeanjean S, Protat C, Yousfi-              Foulquier E, Truong L, Thouvenin-Contet V,
    Monod M, Duprat E, Kaas Q, Pommié C,                   Lefranc G (2003) IMGT unique numbering
    Chaume D, Lefranc G (2004) IMGT-                       for immunoglobulin and T cell receptor vari-
    ONTOLOGY for immunogenetics and immu-                  able domains and Ig superfamily V-like
    noinformatics. In Silico Biol 4:17–29                  domains. Dev Comp Immunol 27:55–77
604       E. Alamyar et al.
22. Elemento O, Lefranc M-P (2003) IMGT/                      T cell receptors report. Immunogenetics
    PhyloGene: an on-line tool for comparative                59:899–902
    analysis of immunoglobulin and T cell receptor      32.   Lefranc M-P (2008) WHO-IUIS nomenclature
    genes. Dev Comp Immunol 27:763–779                        subcommittee for immunoglobulins and T cell
23. Giudicelli V, Chaume D, Jabado-Michaloud J,               receptors report August 2007, 13th International
    Lefranc M-P (2005) Immunogenetics sequence                Congress of Immunology, Rio de Janeiro,
    annotation: the Strategy of IMGT based on                 Brazil. Dev Comp Immunol 32:461–463
    IMGT-ONTOLOGY. Stud Health Technol                  33.   Maglott D, Ostell J, Pruitt KD, Tatusova T
    Inform 116:3–8                                            (2011) Entrez gene: gene-centered information
24. Giudicelli V, Protat C, Lefranc M-P (2003)                at NCBI. Nucleic Acids Res 39:D52–D57
    The IMGT strategy for the automatic annota-         34.   Giudicelli V, Duroux P, Ginestoux C, Folch G,
    tion of IG and TR cDNA sequences: IMGT/                   Jabado-Michaloud J, Chaume D, Lefranc M-P
    Automat. In: INRIA (DISC/Spid), Paris,                    (2006) IMGT/LIGM-DB, the IMGT com-
    DKB-31. Proceedings of the European                       prehensive database of immunoglobulin and T
    Conference on Computational Biology (ECCB                 cell receptor nucleotide sequences. Nucleic
    2003), pp 103–104                                         Acids Res 34:D781–D784
25. Ruiz M, Lefranc M-P (2002) IMGT gene                35.   Lefranc M-P (1998) IMGT (ImMunoGeneTics)
    identification and Colliers de Perles of human            locus on focus. A new section of experimental
    immunoglobulins with known 3D structures.                 and clinical immunogenetics. Exp Clin
    Immunogenetics 53:857–883                                 Immunogenet 15:1–7
26. Kaas Q, Lefranc M-P (2007) IMGT Colliers de         36.   Lefranc M-P (2011) Antibody nomenclature:
    Perles: standardized sequence-structure repre-            from IMGT-ONTOLOGY to INN definition.
    sentations of the IgSF and MhcSF superfamily              MAbs 3:1
    domains. Curr Bioinform 2:21–30                     37.   Kaas Q, Ruiz M, Lefranc M-P (2004)
27. Kaas Q, Ehrenmann F, Lefranc M-P (2007)                   IMGT/3Dstructure-DB and IMGT/Structural
    IG, TR and IgSF, MHC and MhcSF: what do                   Query, a database and a tool for immunoglobu-
    we learn from the IMGT Colliers de Perles?                lin. T cell receptor and MHC structural data.
    Brief Funct Genomic Proteomic 6:253–264                   Nucleic Acids Res 32:D208–D210
28. Lefranc M-P (2001) Nomenclature of the              38.   Ehrenmann F, Kaas Q, Lefranc M-P (2010)
    human immunoglobulin genes. In: Coligan JE,               IMGT/3Dstructure-DB            and      IMGT/
    Bierer BE, Margulies DH, Shevach EM,                      DomainGapAlign: a database and a tool for
    Strober W (eds) Current protocols in immu-                immunoglobulins or antibodies, T cell recep-
    nology. Wiley, Hoboken, NJ, USA, pp                       tors, MHC, IgSF and MhcSF. Nucleic Acids
    A.1P.1–A.1P.37                                            Res 38:D301–D307
29. Lefranc M-P (2001) Nomenclature of the human        39.   Lefranc M-P (2009) Antibody databases and
    T cell receptor genes. In: Coligan JE, Bierer BE,         tools: the IMGT® experience. In: Zhiqiang An
    Margulies DH, Shevach EM, Strober W (eds)                 (ed) Therapeutic monoclonal antibodies: from
    Current protocols in immunology. Wiley,                   bench to clinic (chapter 4). Wiley, Hoboken,
    Hoboken, NJ, USA, pp A.1O.1–A.1O.23                       NJ, USA, pp 91–114
30. Wain HM, Bruford EA, Lovering RC, Lush              40.   Ehrenmann F, Duroux P, Giudicelli V, Lefranc
    MJ, Wright MW, Povey S (2002) Guidelines                  M-P (2010) Standardized sequence and struc-
    for human gene nomenclature. Genomics                     ture analysis of antibody using IMGT®. In:
    79:464–470                                                Kontermann R, Dübel S (eds) Antibody engi-
31. Lefranc M-P (2007) WHO-IUIS nomencla-                     neering (chapter 2), vol 2, Springer. Berlin,
    ture subcommittee for immunoglobulins and                 Heidelberg, pp 11–31
                                                                                        Chapter 33
Abstract
IMGT/DomainGapAlign is the online tool of IMGT®, the international ImMunoGeneTics information
system®, for the analysis of amino acid sequences and two-dimensional (2D) structures of domains. IMGT/
DomainGapAlign allows the analysis of the closest variable (V) and constant (C) domains of immuno-
globulins (IG) or antibodies, T cell receptors (TR), and immunoglobulin superfamily (IgSF) proteins, and
of the groove (G) domains of major histocompatibility (MH; in humans, HLA for human leukocyte anti-
gen) and MH superfamily proteins. IMGT/DomainGapAlign aligns the user own sequences against the
IMGT domain reference directory, displays amino acid changes, creates IMGT gaps, and delimits the
domain strands and loops (and helix for G domain) according to the IMGT unique numbering. IMGT/
DomainGapAlign is coupled to the IMGT/Collier-de-Perles tool that draws standardized IMGT Colliers
de Perles. The analysis is based on the IMGT-ONTOLOGY concepts of identification, classification,
description, and numerotation generated from the axioms of the Formal IMGT-ONTOLOGY or IMGT-
Kaleidoscope. IMGT/DomainGapAlign provides an invaluable help for antibody engineering and anti-
body humanization as it precisely defines the standardized framework regions (FR-IMGT) and
complementarity determining regions (CDR-IMGT) to be grafted. IMGT/DomainGapAlign is freely
available at http://www.imgt.org.
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_33, © Springer Science+Business Media New York 2012
                                                         605
606       F. Ehrenmann and M.-P. Lefranc
1.2. Basic Information    Basic information related to domain types, IMGT domain refer-
for IMGT/Domain-          ence directory, IMGT unique numbering, and IMGT Collier de
GapAlign                  Perles, necessary for an effective use of IMGT/DomainGapAlign,
                          is briefly reviewed.
    Table 1
    V domain strands and loops, IMGT positions and lengths, based on the IMGT unique
    numbering for V domain (V-DOMAIN and V-LIKE-DOMAIN) (7–9)
                                   (see Table 1). The sheets are closely packed against each other
                                   through hydrophobic interactions giving a hydrophobic core
                                   and joined together by a disulfide bridge between 1st-CYS at
                                   position 23 in the B-STRAND in the first sheet and the 2nd-
                                   CYS 104 in the F-STRAND in the second sheet. The V domain
                                   type (8, 9) includes the V-DOMAIN of the IG and TR, which
                                   corresponds to the V-J-REGION or V-D-J-REGION encoded
                                   by V-(D)-J rearrangements (12, 13), and the V-LIKE-
                                   DOMAIN of the IgSF other than IG and TR (19–21).
                               2. C domain.
                                      A C domain (10) comprises about 100 amino acids and is
                                  made of seven antiparallel beta strands (A, B, C, D, E, F, and
                                  G) linked by beta turns, a transversal strand (CD) and loops
                                  (BC and FG), on two sheets (see Table 2). A C domain has a
                                  topology and a three-dimensional structure similar to that of a V
608         F. Ehrenmann and M.-P. Lefranc
    Table 2
    C domain strands, turns, and loops, IMGT positions and lengths, based on the IMGT
    unique numbering for C domain (C-DOMAIN and C-LIKE-DOMAIN) (10)
                                   domain, but without the C¢ and C″ strands and the C¢C″ loop.
                                   The C domain type (10) includes the C-DOMAIN of the IG
                                   and TR (12, 13) and the C-LIKE-DOMAIN of the IgSF other
                                   than IG and TR (19–23).
                               3. G domain.
                                      A G domain (11) comprises about 90 amino acids and is
                                  made of four antiparallel beta strands linked by turns and a
                                  helix that sits on the beta strands, its axis forming an angle of
                                  about 40° with the strands (see Table 3). Two G domains are
                                  needed to form the MhSF groove made of a “floor” and two
                                  “walls.” Each G domain contributes groove structure by its
                                  four strands and turns to half of the groove floor and by its
                                  helix to one wall of the groove (11). The G domain type
                                  includes the G-DOMAIN of the MH (11) and the G-LIKE-
                                  DOMAIN of the MhSF other than MH (22, 24).
1.2.2. IMGT Domain           The IMGT domain reference directory is the IMGT reference
Reference Directory          directory for V, C, and G domains (2). It is manually curated and
                               33   IMGT/DomainGapAlign: The IMGT® Tool for the Analysis…                   609
    Table 3
    G domain strands, turns, and helix, IMGT positions and lengths, based on the IMGT
    unique numbering for G domain (G-DOMAIN and G-LIKE-DOMAIN) (11)
1.2.4. IMGT Colliers       IMGT Colliers de Perles are standardized IMGT 2D graphical
de Perles                  representations of protein domains (15–17) that can be obtained
                           using the IMGT/Collier-de-Perles tool available from the IMGT®
                           Home Page at http://www.imgt.org (for sequences already
                           gapped according to the IMGT unique numbering) or using the
                           IMGT/Collier-de-Perles tool integrated in IMGT/V-QUEST
                           (25) (starting from V-(D)-J nucleotide sequences) or integrated in
                           IMGT/DomainGapAlign (2) (starting from V, C, or G domain
                           amino acid sequences).
2. IMGT/
DomainGapAlign
2.1. IMGT/Domain-           1. The IMGT/DomainGapAlign Welcome page is accessed by
GapAlign Welcome               clicking the link IMGT/DomainGapAlign (“IMGT tools”
Page                           section) in the IMGT® Home page at http://www.imgt.org.
                            2. In the IMGT/DomainGapAlign Welcome page (Fig. 1), locate
                               the text area and paste your amino acid sequences in FASTA
                               format. Alternatively, you can upload a file. A precise delimita-
                               tion of the domain sequences is not required; however, if the
                               sequence contains several domains, the sequence should be
                               split between the different domains. Several domain amino
                               acid sequences can be analyzed simultaneously (up to 50), but
                               each sequence must have a distinct name and belong to the
                              33   IMGT/DomainGapAlign: The IMGT® Tool for the Analysis…          611
2.2.1. Top of the Results   The top of the results page for V domain (Fig. 2), C domain
Page                        (Fig. 3), and G domain (Fig. 4) displays:
                             1. The “Sequence name” (as provided by the user).
                             2. The “Closest reference gene and allele(s) from the IMGT domain
                                directory” section. The domain type (V, C or G) and the species
                                as selected by the user are indicated in the section title (online in
                                orange). The following results are displayed: “Gene and allele
                                name” (see Note 2), “Species,” “Domain number,” “Smith-
                                Waterman Score” (online in orange), label of the domain (online
                                in color) as identified in the closest reference gene and allele
                                domain with its “percentage of identity” and “Overlap” score. If
                                several closest gene and alleles are displayed, the user can select
                                “Align your sequence with” to display the corresponding align-
                                ment. For V-DOMAIN (Fig. 2a), the closest reference gene and
                                alleles section shows the results for the V-REGION and
                                J-REGION of the V and J genes and alleles, respectively.
                             3. The Alignment(s) with the domain of the closest gene and
                                allele from the IMGT domain directory. The domain type (V,
                                C or G) and the species as selected by the user as indicated in
                                                                                                                                                                                  33
Fig. 2. IMGT/DomainGapAlign top of the results page for a V domain. (a) V-DOMAIN. The user amino acid (AA) sequence is aligned with the closest germline V-REGION and J-REGION,
with IMGT gaps and delimitations of the FR-IMGT and CDR-IMGT according to the IMGT unique numbering (8, 9). In this example, the user sequence is the V-DOMAIN of the heavy
chain (VH) of the monoclonal antibody (mAb) alemtuzumab (2). The V-REGION and J-REGION of the alemtuzumab (2) VH is identified as having 73 and 92.9% identity with the Homo
                                                                                                                                                                                  IMGT/DomainGapAlign: The IMGT® Tool for the Analysis…
sapiens IGHV4-59*01 and IGHJ4*01, respectively. The alemtuzumab domain, an antibody humanized before the IMGT standardized delimitations of the FR-IMGT and CDR-IMGT,
shows 14 AA changes in the FR-IMGT, which most probably explains the immunogenicity of that antibody. (b) V-LIKE-DOMAIN. The CD58 domain is a V-LIKE-DOMAIN. The domain AA
sequence of the 3D structure 1ci5 from IMGT/3Dstructure-DB (2) shows four AA changes compared to the CD58*01 allele.
                                                                                                                                                                                  613
614   F. Ehrenmann and M.-P. Lefranc
                                       Fig. 2. (continued)
                                                                                                                                                                                33
Fig. 3. IMGT/DomainGapAlign top of the results page for a C domain. (a) C-DOMAIN. The C domain of an IG lambda chain (or C-LAMBDA) is a C-DOMAIN. The domain AA sequence of
the 3D structure 1aqk from IMGT/3Dstructure-DB (2) shows 3 AA changes with the closest allele IGLC3*03. (b) C-LIKE-DOMAIN. The beta2 microglobulin domain is a C-LIKE-DOMAIN.
The AA sequence of the 3D structure 2d4d from IMGT/3Dstructure-DB (2) shows 3 AA changes compared to the B2M*01 allele.
                                                                                                                                                                                IMGT/DomainGapAlign: The IMGT® Tool for the Analysis…
                                                                                                                                                                                615
616   F. Ehrenmann and M.-P. Lefranc
                                       Fig. 3. (continued)
                                                                                                                                                                                     33
Fig. 4. IMGT/DomainGapAlign top of the results page for a G domain. (a) G-DOMAIN. The first G domain of a MH1 chain is a G-DOMAIN (more precisely G-ALPHA1) (11). In this example,
the user sequence MamuA is the G-ALPHA1 of a MH1 protein of the rhesus monkey Macaca mulatta (2) aligned with the G-ALPHA1 of the closest human HLA-A allele. The query was
with a selection of “Homo sapiens” as a species. (b) G-LIKE-DOMAIN. MICA is a member of the MhSF (11). The alignment identifies the closest domain as being [D2] (G-ALPHA2-LIKE)
of MICA*65 (24).
                                                                                                                                                                                     IMGT/DomainGapAlign: The IMGT® Tool for the Analysis…
                                                                                                                                                                                     617
618   F. Ehrenmann and M.-P. Lefranc
                                       Fig. 4. (continued)
                         33   IMGT/DomainGapAlign: The IMGT® Tool for the Analysis…      619
2.2.2. Results Summary   The Results summary (by comparison with the closest gene and
and AA Changes           allele) and AA changes for V domain (Fig. 5), C domain (Fig. 6),
                         and G domain (Fig. 7) are shown as tables:
                          1. The Results summary table has three columns that are com-
                             mon to the V, C, and G domains; and four additional columns
                             for the V domain. The three common columns are:
                              –   Sequence name.
                              –   Domain (V-REGION for V-DOMAIN, V-LIKE-
                                  DOMAIN,     C-DOMAIN,      C-LIKE-DOMAIN,
                                  G-DOMAIN, G-LIKE-DOMAIN) identity percentage.
                              –   Total number of AA changes in the domain.
Fig. 5. IMGT/DomainGapAlign Results summary for a V domain. (a) V-DOMAIN. The results summary provides sequence name, V-REGION identity percentage, CDR-IMGT lengths,
number of different AA in CDR1- and CDR2-IMGT, FR-IMGT lengths, number of different AA in FR-IMGT, and total number of AA changes. AA changes are shown for strands and loops,
and for FR-IMGT and CDR-IMGT (8, 9). (b) V-LIKE-DOMAIN. The results summary provides sequence name, V-LIKE-DOMAIN, identity percentage, loop lengths, number of different AA
in loops, strand lengths, number of different AA in strands, and the total number of AA changes in V-LIKE-DOMAIN. Loop lengths are those of [BC.C¢C″.FG] that correspond to the
CDR-IMGT of the V-DOMAIN. Strand lengths are those of [(A + B).(C + C').(C″ + D + E + F).G] that correspond to the FR-IMGT of the V-DOMAIN (8, 9). AA changes are shown for strands
                                                                                                                                                                                      IMGT/DomainGapAlign: The IMGT® Tool for the Analysis…
Fig. 6. IMGT/DomainGapAlign Results summary for a C domain. Results summary are similar for a C-DOMAIN or a C-LIKE-DOMAIN. A C-DOMAIN (1aqk_L_CL, same sequence as in
Fig. 3a) is shown as an example. The Results summary provides sequence name, C-DOMAIN (or C-LIKE-DOMAIN), identity percentage, and total number of AA changes. AA changes
are shown for strands (A, B, C, CD, D, E, F, and G), turns (AB, DE, EF), and loops (BC and FG) (10).
                                                                                                                                                                            33
Fig. 7. IMGT/DomainGapAlign Results summary for a G domain. Results summary are similar for a G-DOMAIN or a G-LIKE-DOMAIN. A G-DOMAIN (G-ALPHA, same sequence as in
Fig. 4a) is shown as an example. The results summary provides sequence name, G-DOMAIN (or G-LIKE-DOMAIN), identity percentage, and total number of AA changes. AA changes
are shown for strands (A, B, C and D), turns (AB, BC and CD), and helix (11).
                                                                                                                                                                            IMGT/DomainGapAlign: The IMGT® Tool for the Analysis…
                                                                                                                                                                            623
624         F. Ehrenmann and M.-P. Lefranc
Fig. 8. IMGT Collier de Perles for V domain. IMGT Collier de Perles on one and two layers are on the left hand side and right
hand side of the figure, respectively. IMGT Colliers de Perles on one and two layers with AA changes (online pink border)
are shown in the lower section of the figure. (a) V-DOMAIN. In an IG or TR V-DOMAIN, the G-STRAND (or FR4-IMGT) is the
C-terminal part of the J-REGION. The FR4-IMGT is at least composed of 9 or 10 amino acids beyond the phenylalanine F
(J-PHE 118) or tryptophan W (J-TRP 118) of the motif F/W-G-X-G that characterizes the J-REGION. The example is the VH
of alemtuzumab (as in Figs. 2a and 5a). The CDR-IMGT lengths are [8.10.12]. (b) V-LIKE-DOMAIN. The example is the
V-LIKE-DOMAIN of Homo sapiens CD58 (same sequence as in Figs. 2b and 5b). The loop lengths are [5.4.7].
Fig. 8. (continued)
Fig. 9. IMGT Collier de Perles for C domain. IMGT Collier de Perles on one and two layers are on the left hand side and right
hand side of the figure, respectively. IMGT Colliers de Perles on one and two layers with AA changes (online pink border)
are shown in the lower section of the figure. (a) C-DOMAIN. The example is the Homo sapiens C-LAMBDA of 1aqk (1aqk_L_
CL) as in Figs. 3a and 6. (b) C-LIKE-DOMAIN. The example is the B2M C-LIKE-DOMAIN of 2d4d (same sequence as in
Fig. 3b).
                      33   IMGT/DomainGapAlign: The IMGT® Tool for the Analysis…   627
Fig. 9. (continued)
Fig. 10. IMGT Collier de Perles for G domain. IMGT/DomainGapAlign works per domain. IMGT Collier de Perles of the MH groove are shown separately (2). (a) G-DOMAIN. The example
is a MH1 groove. The domain G-ALPHA1 ([D1] of the MH1-ALPHA chain) is automatically located by the tool in the upper part of the IMGT Collier de Perles groove representation (the
example is G-ALPHA1 of MamuA, 3jtt_A_MamuA, left hand side of the figure), whereas the domain G-ALPHA2 ([D2] of the MH1-ALPHA chain) is automatically located in the lower
part of the groove (the example is G-ALPHA2 of MamuA, 3jtt_A_MamuA, right hand side of the figure). (b) G-LIKE-DOMAIN. The domain G-ALPHA1-LIKE ([D1] of the MH1LIKE-ALPHA
chain) is automatically located in the upper part (G-ALPHA1-LIKE, 1b3j_A_MICA, left hand side of the figure) where the domain G-ALPHA2-LIKE ([D2] of the MH1LIKE-ALPHA chain)
is automatically located in the lower part (G-ALPHA2-LIKE, 1b3j_A_MICA, right hand side of the figure) (same sequence as in Fig. 4b). Note that in IMGT/3Dstructure-DB (2), the two
                                                                                                                                                                                      IMGT/DomainGapAlign: The IMGT® Tool for the Analysis…
3. Conclusion
4. Notes
References
 1. Lefranc M-P, Giudicelli V, Ginestoux C,               receptors and Ig-like domains. Immunologist
    Jabado-Michaloud J, Folch G, Bellahcene F,            7:132–136
    Wu Y, Gemrot E, Brochet X, Lane J, Regnier       9.   Lefranc M-P, Pommié C, Ruiz M, Giudicelli V,
    L, Ehrenmann F, Lefranc G, Duroux P (2009)            Foulquier E, Truong L, Thouvenin-Contet V,
    IMGT®, the international ImMunoGeneTics               Lefranc G (2003) IMGT unique numbering
    information system®. Nucleic Acids Res                for immunoglobulin and T cell receptor vari-
    37:D1006–D1012                                        able domains and Ig superfamily V-like
 2. Ehrenmann F, Kaas Q, Lefranc M-P (2010)               domains. Dev Comp Immunol 27:55–77
    IMGT/3Dstructure-DB           and      IMGT/    10.   Lefranc M-P, Pommié C, Kaas Q, Duprat E,
    DomainGapAlign: a database and a tool for             Bosc N, Guiraudou D, Jean C, Ruiz M, Da
    immunoglobulins or antibodies, T cell recep-          Piedade I, Rouard M, Foulquier E, Thouvenin
    tors, MHC, IgSF and MhcSF. Nucleic Acids              V, Lefranc G (2005) IMGT unique numbering
    Res 38:D301–D307                                      for immunoglobulin and T cell receptor con-
 3. Giudicelli V, Lefranc M-P (1999) Ontology for         stant domains and Ig superfamily C-like
    immunogenetics:           IMGT-ONTOLOGY.              domains. Dev Comp Immunol 29:185–203
    Bioinformatics 15:1047–1054                     11.   Lefranc M-P, Duprat E, Kaas Q, Tranne M,
 4. Lefranc M-P, Giudicelli V, Ginestoux C, Bosc          Thiriot A, Lefranc G (2005) IMGT unique
    N, Folch G, Guiraudou D, Jabado-Michaloud             numbering for MHC groove G-DOMAIN and
    J, Magris S, Scaviner D, Thouvenin V, Combres         MHC        superfamily    (MhcSF)     G-LIKE-
    K, Girod D, Jeanjean S, Protat C, Yousfi              DOMAIN. Dev Comp Immunol 29:917–938
    Monod M, Duprat E, Kaas Q, Pommié C,            12.   Lefranc M-P, Lefranc G (2001) The immuno-
    Chaume D, Lefranc G (2004) IMGT-                      globulin FactsBook. Academic Press, London,
    ONTOLOGY for Immunogenetics and                       UK, pp 1–458
    Immunoinformatics. In Silico Biol 4:17–29.      13.   Lefranc M-P, Lefranc G (2001) The T cell
    http://www.imgt.org                                   receptor FactsBook. Academic Press, London,
 5. Lefranc M-P (2004) IMGT-ONTOLOGY and                  UK, pp 1–398
    IMGT databases, tools and web resources for     14.   Giudicelli V, Chaume D, Lefranc M-P (2005)
    immunogenetics and immunoinformatics. Mol             IMGT/GENE-DB: a comprehensive database
    Immunol 40:647–660                                    for human and mouse immunoglobulin and T
 6. Duroux P, Kaas Q, Brochet X, Lane J,                  cell receptor genes. Nucleic Acids Res
    Ginestoux C, Lefranc M-P, Giudicelli V (2008)         33:D256–D261
    IMGT-Kaleidoscope, the formal IMGT-             15.   Ruiz M, Lefranc M-P (2002) IMGT gene
    ONTOLOGY paradigm. Biochimie 90:                      identification and Colliers de Perles of human
    570–583                                               immunoglobulins with known 3D structures.
 7. Lefranc M-P (1997) Unique database number-            Immunogenetics 53:857–883
    ing system for immunogenetic analysis.          16.   Kaas Q, Lefranc M-P (2007) IMGT Colliers de
    Immunol Today 18:509                                  Perles: standardized sequence-structure repre-
 8. Lefranc M-P (1999) The IMGT unique                    sentations of the IgSF and MhcSF superfamily
    numbering for immunoglobulins, T cell                 domains. Curr Bioinform 2:21–30
                              33   IMGT/DomainGapAlign: The IMGT® Tool for the Analysis…                633
17. Kaas Q, Ehrenmann F, Lefranc M-P (2007)              27. Lefranc M-P (2009) Antibody databases and
    IG, TR and IgSf, MHC and MhcSF: what do                  tools: The IMGT® experience. In: Zhiqiang An
    we learn from the IMGT Colliers de Perles?               (ed) Therapeutic monoclonal antibodies: from
    Brief Funct Genomic Proteomic 6:253–264                  bench to clinic (chapter 4). John Wiley & Sons,
18. Pommié C, Levadoux S, Sabatier R, Lefranc M-P            Inc., Hoboken, NJ, USA, pp 91–114
    (2004) IMGT standardized criteria for statistical    28. Ehrenmann F, Duroux P, Giudicelli V, Lefranc
    analysis of immunoglobulin V-REGION amino                M-P (2010) Standardized sequence and struc-
    acid properties. J Mol Recognit 17:17–32                 ture analysis of antibody using IMGT®. In:
19. Duprat E, Kaas Q, Garelle V, Lefranc G,                  Kontermann R, and Dübel S (eds) Antibody
    Lefranc M-P (2004) IMGT standardization for              engineering (chapter 2), vol 2, Springer-Verlab,
    alleles and mutations of the V-LIKE-DOMAINs              Berlin, Heidelberg, pp 11–31
    and C-LIKE-DOMAINs of the immunoglob-                29. Kaas Q, Ruiz M, Lefranc M-P (2004)
    ulin superfamily. In: Pandalai SG (ed) Recent            IMGT/3Dstructure-DB and IMGT/Structural-
    research developments in human genetics                  Query, a database and a tool for immunoglobu-
    Research Signpost, Trivandram, Kerala, vol 2.            lin, T cell receptor and MHC structural data.
    pp 111–136                                               Nucleic Acids Res 32:D208–D210
20. Garapati VP, Lefranc M-P (2007) IMGT                 30. Kaas Q, Lefranc M-P (2005) T cell receptor/
    Colliers de Perles and IgSF domain standard-             peptide/MHC molecular characterization and
    ization for T cell costimulatory activatory              standardized pMHC contact sites in
    (CD28, ICOS) and inhibitory (CTLA4,                      IMGT/3Dstructure-DB. In Silico Biol 5:
    PDCD1 and BTLA) receptors. Dev Comp                      505–528
    Immunol 31:1050–1072                                 31. Kaas Q, Duprat E, Tourneur G, Lefranc M-P
21. Bernard D, Hansen JD, du Pasquier L, Lefranc             (2008) IMGT standardization for molecular
    M-P, Benmansour A, Boudinot P (2005)                     characterization of the T cell receptor/
    Costimulatory receptors in jawed vertebrates:            peptide/MHC complexes. In: Schoenbach C,
    conserved CD28, odd CTLA4 and multiple                   Ranganathan S, Brusic V (eds) Immunoin-
    BTLAs. Dev Comp Immunol 31:255–271                       formatics. Immunomics Reviews, Series of
22. Duprat E, Lefranc M-P, Gascuel O (2006) A                Springer Science and Business Media LLC
    simple method to predict protein binding from            (chapter 2). Springer, New York, USA,
    aligned sequences-application to MHC super-              pp 19–49
    family and beta2-microglobulin. Bioinformatics       32. Lefranc M-P (2009) Antibody databases:
    22:453–459                                               IMGT®, a French platform of world-wide
23. Bertrand G, Duprat E, Lefranc M-P, Marti J,              interest. Bases de données anticorps: IMGT®,
    Coste J (2004) Characterization of human                 une plate-forme française d’intérêt mondial
    FCGR3B*02 (HNA-1b, NA2) cDNAs and                        “Anticorps monoclonaux en thérapeutique”.
    IMGT standardized description of FCGR3B                  Med Sci 25:1020–1023
    alleles. Tissue Antigens Cover 64:119–131            33. Lefranc M-P (2011) Antibody nomenclature:
24. Frigoul A, Lefranc M-P (2005) MICA: standard-            from IMGT-ONTOLOGY to INN definition.
    ized IMGT allele nomenclature, polymorphisms             MAbs 3:1–2
    and diseases. In: Pandalai SG (ed) Recent research   34. Jefferis R, Lefranc M-P (2009) Human immu-
    developments in human genetics, vol 3. Research          noglobulin allotypes: possible implications for
    Signpost, Trivandrum, Kerala, pp 95–145                  immunogenicity. MAbs 1(4):332–338
25. Brochet X, Lefranc M-P, Giudicelli V (2008)          35. Lefranc M-P, Lefranc G (2012) Human Gm,
    IMGT/V-QUEST: the highly customized and                  Km and Am allotypes and their molecular
    integrated system for IG and TR standardized             characterization: a remarkable demonstration
    V-J and V-D-J sequence analysis. Nucleic Acids           of polymorphism. In: Tait B, Christiansen F
    Res 36:W503–W508                                         (eds) Methods in Molecular Biology –
26. Lefranc M-P, Giudicelli V, Regnier L, Duroux P           Immunogenetics, Chapter 34
    (2008) IMGT®, a system and an ontology that          36. Smith TF, Waterman MS (1981) Identification
    bridge biological and computational spheres in           of common molecular subsequences. J Mol
    bioinformatics. Brief Bioinform 9:263–275                Biol 147(1):195–197
                                                                                        Chapter 34
Abstract
Human immunoglobulin allotypes are antigenic determinants (or “markers”) determined serologically,
classically by hemagglutination inhibition, on the human immunoglobulin (IG) heavy and light chains.
The allotypes have been identified on the gamma1, gamma2, gamma3, and alpha2 heavy chains (they are
designated as G1m, G2m, G3m, and A2m allotypes, respectively), and on the kappa light chain (Km allo-
types). Gm-Am allotypes are inherited in fixed combinations, or Gm-Am haplotypes, owing to the linkage
of the human IGHC genes (IGHG3, IGHG1, IGHA1, IGHG2, IGHG4, IGHE, and IGHA2 from 5¢ to
3¢ in the IGH locus on chromosome 14). Gm and Am allotypes have been one of the most powerful tools
in population genetics and very instrumental in molecular characterization of the human IGHC genes
(gene conversion, copy number variation, gene order). They represent a major system for understanding
immunogenicity of the polymorphic IG chains, in relation with amino acid and conformational changes.
The correlation between G3m allotypes and amino acid changes has been possible with the sequencing of
many alleles of the IGHG3 gene, from individuals from different populations and with known allotypes.
In this chapter, we integrate genetics and sequence data and provide an updated overview of the Gm-Am
haplotypes and Km allotypes. We propose, for the first time, a complete elucidation of the G3m allotypes,
illustrated by the “IMGT G3m allele butterfly” concept that allows a graphical representation of the
G3m alleles (variants of a gene expressing a given set of allotypes). Knowledge of allotypes is important in
antibody engineering and humanization of monoclonal antibodies to improve immunotherapy.
     Key words: IMGT, Immunogenetics, Allotype, Haplotype, Gm, Km, Am, Gm-Am, Immuno-
     informatics, Immunoglobulin, Antibody, Allelic polymorphism, IMGT-ONTOLOGY
1. Introduction
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9_34, © Springer Science+Business Media New York 2012
                                                         635
636   M.-P. Lefranc and G. Lefranc
2. Basic
Information
on Allotypes
and Isoallotypes         Allotypy within IgG was first described by Grubb who showed
                         that certain human sera would agglutinate erythrocytes sensitized
2.1. Human IG Allotype   with human “incomplete” anti-Rh antibody (20, 21) (see Note 1).
Discovery                Polymorphism of the C region of human IG heavy gamma and
                         alpha and light kappa chains was subsequently recognized by sero-
                         logical typing using the classical reaction of inhibition of hemag-
                         glutination (20–46). Thus, the discovery of this polymorphism
                         demonstrated that exposure of an individual to IgG or IgA of a
                         nonself allotype can induce an anti-allotype response.
2.2. Gm, Am, and Km      Allotypes of IG are unique antigenic determinants recognized by
Allotype and Allele      specific antibodies. They are IG markers that, in terms of immuno-
Definition               genicity, represent B cell epitopes. Allotypes correspond to sero-
                         logically detected amino acid changes that characterize the
                         polymorphism of a chain within a given isotype. By definition, allo-
                         types are shared among individuals within populations. Allotypes
                         have been identified on the C region of the human IG heavy
                         gamma1, gamma2, gamma3, and alpha2 chains of the IgG1, IgG2,
                         IgG3, and IgA2 subclasses, respectively, and on the C region of the
                         human IG light kappa chains (see Note 2). They are designated as
                         Gm (Gamma marker), Am (Alpha marker), and Km (Kappa
                         marker), with a number for the subclass: G1m, G2m, and G3m for
                         allotypes of the gamma1, gamma2, and gamma3 chains, and A2m
                         for allotypes of the alpha2 chains (1).
638        M.-P. Lefranc and G. Lefranc
2.3. Gm-Am Haplotype       Gm allotypes are inherited in fixed combinations called “Gm haplo-
Definition                 types” (see Subheading 7) or “Gm-Am haplotypes” (if A2m allo-
                           types are tested), owing to the linkage of the IGHC genes within a
                           cluster in the IGH locus (47–53). Haplotypes have a low frequency
                           of crossovers; however, crossover events and gene conversions
                           (100, 102, 103, 106) have occurred during evolution resulting in
                           characteristic haplotypes present in diverse populations, hence the
                           usefulness of the allotype system in population studies. Equal or
                           unequal crossovers, the later generating gene duplication (or
                           expansion) or, in contrast, gene deletion (or contraction) in the
                           IGH locus has been demonstrated (97–99, 102, 105, 107, 108,
                           110, 111) and they are now commonly named CNV.
2.4. Allotype              Allotypes are determined by serological typing using a classical reac-
Determination              tion of inhibition of hemagglutination. The methodology uses
                           human O Rh+ erythrocytes (red blood cells) sensitized (coated)
2.4.1. Hemagglutination
                           with “incomplete” anti-Rh IgG antibodies of known allotypes
Inhibition Methodology
                           (e.g., G1m1), and human reagents (polyclonal IgG specific for a
                           given allotype, e.g., anti-G1m1). The polyclonal IgG reagents are
                           obtained from multiparous women, multiple transfused individuals,
                           and normal blood donors. If the tested serum is G1m1-negative, the
                           anti-G1m1 reagent binds the G1m1-positive antibodies coating
                           the erythrocytes and the hemagglutination occurs. In contrast, if
                           the tested serum is G1m1-positive, the anti-G1m1 reagent binds the
                           G1m1-positive antibodies contained in the serum, and the hemag-
                           glutination is inhibited. The reactions are performed with different
                           dilutions of the reagents and of the tested sera (47, 48, 52, 53). The
                           determination of some allotypes (G2m23, A2m1, and A2m2) is
                           more delicate owing to the rarity of the reagents and, in particular,
                           the absence of available anti-Rh antibodies for the coating. In those
                           cases, myeloma proteins are coupled using chromic chloride (123).
                           It is highly recommended to provide the list and source of the anti-
                           Rh, myeloma proteins, polyclonal IgG, and eventually their dilu-
                           tions, the list of tested allotypes, and the number of individuals tested
                           for each allotype, for allowing standardized result comparison.
2.4.3. Other Methodologies     To compensate for the rarity of some reagents, attempts were made
                               to obtain monoclonal antibodies; however, the main difficulty
                               resides in the characterization of their specificity (124, 125).
                               Molecular biology was first used for the determination of A2m2 by
                               RFLP (101) (see Subheading 8.3). This method is particularly
                               interesting given the rarity of the reagents and was the first proto-
                               col to determine allotypes in the absence of serum (e.g., from cell
                               lines). Polymerase chain reaction (PCR) amplification methods,
                               using allele-specific oligonucleotides (ASO) or specific restriction
                               sites, were subsequently developed for the determination of the
                               Km allotypes (126) and of some Gm allotypes (4, 127–130), but
                               their application remains limited.
2.5.2. Allotype Localization   The localization of the allotypes has been determined by inhibition
                               studies with Fab and Fc fragments (obtained by papain digestion),
                               and with pF’c fragments (obtained by pepsin digestion) of
                               Gm-positive or Gm-negative myeloma proteins (131–136). Amino
                               acid sequence analysis of peptides obtained from Fd (part of the heavy
                               chain from a Fab) and from pF’c have revealed amino acid changes,
                               allowing establishment of correlations between serologically defined
                               allotypes and amino acid sequences. Except for G1m3 and G1m17
                               located on the CH1 of gamma1, all other Gm allotypes are localized
                               on the Fc (on CH2 or on CH3) (Table 1). Thus, for example, the
                               G3m21 allotype was detectable on the Fc fragment of gamma3 chains
                               but not on isolated CH3 domains, and therefore was localized on the
                               CH2 (134). G3m10, G3m11, and G3m13 were localized on the
                               CH3 domain of the gamma3 chain (134). G3m28 was localized on
                               the CH3 domain of a G3m28 myeloma protein (38). However, the
                               first detailed correlation between G3m allotypes and amino acid
                               changes has only been possible following the complete nucleotide
                               sequencing of many IGHG3 alleles from individuals homozygous for
                               well-characterized G3m alleles (104, 106, 113). G3m allotypes and
                               their localization, and correspondence with G3m alleles, illustrated
                               by the “IMGT G3m allele butterfly” representation, are further
                               defined in this chapter (see Subheading 5.2).
640     M.-P. Lefranc and G. Lefranc
Table 1
Nomenclature of the Gm and Km allotypes
Localizationa Nomenclature
    Table 1
    (continued)
Localizationa Nomenclature
    Table 2
    Nomenclature and distribution of the isoallotypes
    WHO               Previous
    nomenclature      designation       gamma1       gamma2        gamma3         gamma4        alpha1     alpha2
    nG1m1             nG1m(a)           Allo         Iso           Iso            Isoa          –          –
    nG1m17            nG1m(z)           Allo         –             Iso            Iso           –          –
    nG3m5             nG3m(b1)          Isob         Iso           Allo           –             –          –
    nG3m11            nG3m(b0)          Iso          Iso           Allo           –             –          –
    nG3m21            nG3m(g)           –            Iso           Allo           –             –          –
    nG4m(a)           nG4m(a)           Iso          –             Iso            Allo          –          –
    nG4m(b)           nG4m(b)           –            Iso           –              Allo          –          –
    nA2m3c            nA2m(2)           –            –             –              –             Iso        Allo
a
 Isoallotype only detected by some antisera
b
  In Negroid populations, the G1m28 allotype can be expressed instead of nG3m5 (see Subheading 5.1.7)
c
  nA2m3 has been renamed as this isoallotype is located on the CH3 domain and is antithetical to the A2m3 allele
(it is not antithetical to A2m2 located on the CH1) (see Subheading 8.5)
642       M.-P. Lefranc and G. Lefranc
2.6. Isoallotypes         By definition, allotypes are found on chains within one IG isotype
                          (encoded by one given IG gene). However, the same amino acids
                          may be found in chains of other isotypes (encoded by other IG
                          genes), but without being polymorphic in these isotypes. If these
                          amino acids are detected in vitro by antibody reagents, they are
                          referred as “isoallotypes” (designated with the letter “n” preced-
                          ing the allotype name, e.g., nG1m1). Seven isoallotypes have
                          been identified for the gamma chains (137–141) and one for the
                          alpha chains identified on the CH3 domain (142) (Table 2) (see
                          Note 5).
2.7. Protein Displays     Protein displays are standardized IMGT representations of the amino
and IMGT Colliers de      acid sequences of the coding regions of the genes (11). Protein dis-
Perles                    plays of C domains show the sequences per domain, using the IMGT
                          unique numbering for C domain (14). They allow a standardized
                          localization of the amino acids involved in the allotypes in relation to
                          the strands and loops of the domains. Figure 1 provides the Protein
                          displays of the C domains (CH1, CH2, and CH3) of the IGHG and
                          IGHA genes (5, 6, 17). Only the first allele is shown. Other alleles are
                          available in IMGT/DomainDisplay (5, 6) and in IMGT/GENE-DB
                          (15) (IMGT®, http://www.imgt.org). IMGT/DomainGapAlign
                          (17) allows gaps to be inserted in C domains of the IGHG, IGHA,
                          and IGKC genes according to the IMGT unique numbering (14).
                          Correspondences with other numberings are available in the IMGT
                          Scientific chart (http://www.imgt.org). Standardized representa-
                          tions or IMGT Colliers de Perles of the C domains can be obtained
                          using the IMGT/Collier-de-Perles tool (18, 19).
3. G1m
3.1. G1m Allotypes        G1m1 (previously G1m(a)) was the first discovered allotype (20).
and nG1m Allotypes        In 1956, Grubb noticed that 60% of normal human sera could
                          inhibit the agglutination of human O Rh+ erythrocytes sensitized
3.1.1. G1m1 and nG1m1
                          by means of certain “incomplete” anti-Rh sera, the factor respon-
                          sible for the inhibition was called Gm(a) (now G1m1 allotype).
                          Grubb and Laurel demonstrated that Gm(a) was transmitted as a
                          dominant autosomal Mendelian trait (21).
                               By inhibition studies with IgG1 fragments, it was established
                          that G1m1 is located on the CH3 domain and was associated with
                          aspartate 356 and leucine 358 (Eu numbering) (131). According
                          to the IMGT unique numbering for C domain (14), the G1m1
                          allotype corresponds to IGHG1 CH3 Asp D12 and Leu L14
                          (Table 3) (Figs. 2 and 3).
                               In the G1m1-negative gamma1 chains and in the gamma
                          chains of the other IgG subclasses, glutamate (E) and methionine
                          (M) are found, respectively, at positions 12 and 14 of the CH3
                                                                                                                                                                                34
                                                                                                                                                                                Human Gm, Km, and Am Allotypes and Their Molecular Characterization…
Fig. 1. Protein display of the human IGHG and IGHA constant (C) domains. (a) Human IGHG CH domains. (b) Human IGHA CH domains. Only allele *01 is shown (see Note 3). Other
alleles are available in IMGT/DomainGapAlign (5, 6) and in IMGT/GENE-DB (15) (IMGT®, http://www.imgt.org). The alignments are based on the IMGT unique numbering for C domain
(14). Hinge and CHS regions are not shown. Domains are numbered with [D1] being the variable domain (not shown).
                                                                                                                                                                                643
     Table 3
     G1m allotypes and isoallotypes
Fig. 2. IMGT Collier de Perles of the IGHG1 CH3 domain. The CH3 domain is from the b12 antibody (IMGT/3Dstructure-DB,
code PDB:1hzh, http://www.imgt.org) (16, 17). The IMGT Collier de Perles is shown on two layers, with hydrogen bonds
shown as green lines online. Hatched positions correspond to gaps according to the IMGT unique numbering (14). The
aspartate D12 and leucine L14 (strand A) correspond to G1m1, whereas E12 and methionine M14 (not shown) correspond
to nG1m1. In G1m2-negative chain, as that of b12, there is an alanine at position 110. A glycine at position 110 would
correspond to G1m2. The amino acids glycine (G)-lysine (K) at positions 129 and 130 represent the CHS in secreted IG.
              34   Human Gm, Km, and Am Allotypes and Their Molecular Characterization…                       645
                            Fig. 3. Three-dimensional structure of the IGHG1 CH3 domain. The CH3 domain is from the
                            b12 antibody (IMGT/3Dstructure-DB, code PDB:1hzh, http://www.imgt.org) (16, 17).
                            Positions 12 and 14 of the G1m1/nG1m1 allotype, and position 110 of the G1m2/- allotype
                            in the IGHG1 CH3 domain are shown. The aspartate D12 and leucine L14 correspond to
                            G1m1, whereas alanine A110 corresponds to nG1m2. Glutamate E12 and methionine M14
                            correspond to nG1m1, whereas a glycine G110 corresponds to G1m2.
3.1.2. G1m2                 The G1m2 allotype was discovered in 1959 by Harboe and Lundevall
                            (22). The G1m2 allotype was detected on the CH3 domain (134).
                            It corresponds to a glycine at position 110 (IGHG1 CH3 Gly
                            G110), whereas the absence of the allotype correlates to alanine at
                            that position (145) (see Note 6) (Table 3) (Figs. 2 and 3).
646        M.-P. Lefranc and G. Lefranc
Fig. 4. IMGT Collier de Perles of the IGHG1 CH1 domain. The CH1 domain is from the b12 antibody (IMGT/3Dstructure-DB,
code PDB:1hzh, http://www.imgt.org) (16, 17). The IMGT Collier de Perles is shown on two layers, with hydrogen bonds
shown as green lines online. Hatched positions correspond to gaps according to the IMGT unique numbering (14). The
lysine at position 120 (K120) corresponds to the G1m17 allotype. The isoleucine I103 is specific for the gamma1 chain
isotype. If an arginine is expressed at position 120 (R120), the simultaneous presence of R120 and I103 corresponds to the
expression of the G1m3 allotype.
3.1.3. G1m3, G1m17,             G1m3 was first identified by Steinberg and Wilson in 1963 (23),
and nG1m17                      then further characterized by Gold et al. (24, 25). G1m3, located
                                on the CH1 domain, corresponds to an arginine at position 120
                                (IGHG1 CH1 Arg R120) (132) (Figs. 4 and 5). The expression of
                                G1m3 requires the presence of an amino acid specific for gamma1,
                                because the CH1 Arg R120 is also present on the gamma3 and
                                gamma4 chains (Fig. 1a). It seems likely from the analysis of 3D
                                structures of Fab and of the b12 antibody, the only complete
                                human IG so far crystallized (IMGT/3Dstructure-DB, code
                                PDB:1hzh) (16, 17) (see Note 7) that the isoleucine at position
                                103 (IGHG1 CH1 Ile I103) is the amino acid involved in the
                                expression of the G1m3 allotype (Figs. 4 and 5).
                                     G1m17 (26), located in the CH1 domain, corresponds to
                                lysine at position 120 (IGHG1 CH1 Lys K120) (132). G1m17 is
           34   Human Gm, Km, and Am Allotypes and Their Molecular Characterization…                       647
                         Fig. 5. Three-dimensional structure of the IGHG1 CH1 domain. The CH1 domain is from the
                         b12 antibody (IMGT/3Dstructure-DB, code PDB:1hzh, http://www.imgt.org) (16, 17).
                         The lysine K120 (strand G) and the isoleucine I103 (strand F) are shown. The K120
                         corresponds to the G1m17 allotype. The simultaneous presence of I103 (specific of
                         the gamma1 isotype) and of arginine R120 corresponds to the G1m3 allotype. The R120
                         corresponds to the nG1m17 isoallotype.
3.1.4. G1m27 and G1m28   G1m27 and G1m28 have only been demonstrated to be present
                         on the gamma1 chains in Negroid populations (96); however, it is
                         not excluded that these “surnumerary” allotypes may explain some
                         uncommon haplotypes found in other populations (52, 53, 94, 95,
                         102). G1m27 most probably corresponds to IGHG1 CH3 Ileu
                         I101 (resulting from an amino acid change V101 > I), and G1m28
                         to IGHG1 CH3 Arg R115, Tyr Y116 (with an amino acid change
                         H115 > R) (see Subheading 5.1.7).
648        M.-P. Lefranc and G. Lefranc
Fig. 6. G1m allotypes localizations on gamma1 chains. The CH1, CH2, and CH3 domains of the b12 gamma1 chains are
shown (IMGT/3Dstructure-DB, code PDB:1hzh, http://www.imgt.org) (16, 17), with the positions involved in the G1m
allotypes. The CH2 position 45.1 is not related to the G1m allotypes, but indicates the amino acid position that should be
responsible for the G2m23 allotype, or of its absence (G2m..), on a gamma2 chain.
3.2. Correspondence             The heavy gamma1 chains of IgG1 may express four typical G1m
Between G1m Alleles             alleles (combinations of G1m allotypes): G1m3, G1m3,1, G1m17,1,
and IGHG1 Alleles               and G1m17,1,2 (and three additional G1m alleles, Gm17,1,27,
                                Gm17,1,28, and Gm17,1,27,28, the last two identified in Negroid
                                populations (96)). The C region of the G1m3,1, G1m17,1, and
                                G1m17,1,2 chains differs from that of the G1m3 chains by two,
                                three, and four amino acids, respectively. The structural correlations
                                with amino acids are illustrated in Fig. 6. The correspondence between
                                the G1m alleles and IGHG1 alleles is shown in Table 4. Thus,
                                IGHG1*01 and IGHG1*02 (see Note 3) are G1m17,1, IGHG1*03
                                is G1m3, IGHG1*04, IGHG1*05, and IGHG1*06 are G1m17,1,27,
                                G1m17,1,28, and G1m17,1,27,28, respectively, IGHG1*07 is
                                G1m17,1,2, and IGHG1*08 is G1m3,1. In Table 4, amino acids
                                corresponding to G1m allotypes are shown in bold. The nG1m1
Table 4
Correspondence between the G1m alleles and IGHG1 alleles
d
  IGHG1*07p and IGHG1*08p amino acids, and codons between parentheses, are expected (GL and MPL) (11). The letter “p” indicates that these alleles have not yet been
sequenced at the nucleotide level, and therefore are not shown in IMGT Repertoire, Alignments of alleles: Homo sapiens IGHG1 (http://www.imgt.org) (5, 6)
                                                                                                                                                                           649
650      M.-P. Lefranc and G. Lefranc
4. G2m
4.1. G2m Allotype        G2m23 (27) is the only allotype shown on the IgG2 heavy chains,
                         and the gamma2 chains are either G2m23 or G2m.. (two dots
                         indicate that a specimen was tested and found to be negative for
                         G2m23 (48, 52, 53)) (see Note 8). G2m23 is localized on the
                         CH2 domain (detectable on the Fc of G2m23 myeloma proteins
                         but not on isolated CH3 domains) (134). Amino acid sequence
                         and 3D structure comparisons show that the G2m23 allotype is
                         correlated with methionine 45.1 (‘.1’ for first position in the trans-
                         verse CD strand (14)) in the CH2 (IGHG2 CH2 Met M45.1),
                         Fig. 7. Three-dimensional structure of the IGHG2 CH2 domain. CH2 position 45.1 (first
                         position of the transversal CD strand) corresponds to the G2m23/G2m.. allotype. Valine
                         V45.1 corresponds to G2m.., whereas a methionine corresponds to G2m23.
             34   Human Gm, Km, and Am Allotypes and Their Molecular Characterization…                   651
    Table 5
    Correspondence between the G2m alleles and IGHG2 alleles
                                                    IMGT                                 45.1
                                                    Exon                                 (52)
                                                    Eu                                   282
                                                                                         G2m23/G2m..
    G2m23          IGHG2*02                                                              Met M45.1 atg
    G2m..          IGHG2*01, IGHG2*03, IGHG2*04, IGHG2*05, IGHG2*06                      Val V45.1 gtg
a
Positions in bold are according to the IMGT unique numbering for C domain (14); between parentheses, exon
numbering (11), and in italics, Eu numbering (correspondence between C numberings, in IMGT Scientific chart,
http://www.imgt.org) (5, 6)
4.2. Correspondence         The G2m alleles are characterized by the presence or absence of
Between the G2m             the G2m23 allotype. Only the IGHG2*02 allele is G2m23. The
Alleles and IGHG2           other alleles IGHG1*01, IGHG2*03, IGHG2*04, IGHG2*05
Alleles                     and IGHG2*06 are G2m23-negative (or G2m..) (Table 5) (see
                            Note 9).
5. G3m
5.1. G3m Allotypes          The G3m allotypes make the gamma3 chain the most polymorphic
                            IG chains in humans. Thirteen G3m allotypes are characterized:
5.1.1. G3m Allotypes
                            G3m5, G3m6, G3m10, G3m11, G3m13, G3m14, G3m15,
and IGHG3 Sequences
                            G3m16, G3m21, G3m24, G3m26, G3m27, G3m28 (Table 1).
                            Three isoallotypes (nG3m5, nG3m11, and nG3m21) have also
                            been characterized (Table 2).
652      M.-P. Lefranc and G. Lefranc
5.1.2. G3m6, G3m10,      A first mosaic of G3m allotypes on the CH3 can be defined around
G3m11, nG3m11, G3m13,    G3m11. G3m11 is characterized by a serine at position 44 of the
G3m24, G3m27             CH3 (IGHG3 CH3 Ser S44), whereas nG3m11 depends on an
                         asparagine at the same position (IGHG3 CH3 Asn N44) (113).
                              Haplotype analysis shows that four allotypes, G3m6, G3m10,
                         G3m13, and G3m24, depends on the presence of G3m11 (49–53).
                              Extensive analysis of the sequence (104, 106, 113) and genetic
                         data demonstrate that:
             34   Human Gm, Km, and Am Allotypes and Their Molecular Characterization…                           653
Fig. 8. G3m allotypes localizations on gamma3 chains. G3m allotypes are described in Subheading 5.1. G3m16 (tryptophan
Trp W83) and G3m21 (leucine Leu L82), nG3m21 (proline Pro P82) are located on the CH2. The other G3m allotypes form
two mosaics on the CH3. G3m26 (R115), G3m5 (R115, F116), G3m28 (R115, Y116), nG3m5 (H115, Y116), G3m14
(M84, R115, F116) and G3m15 (M39, H115, Y116) form a first mosaic. G3m11 (S44), nG3m11 (N44), G3m10 (S44, I101),
G3m24 (S44, V101), G3m27 (I101), G3m6 (S44, E98), G3m13 (S44, Q98) form a second mosaic.
5.1.3. G3m5, nG3m5,       The second mosaic, also observed on the CH3 domain of IGHG3,
G3m28, G3m26              defines three G3m allotypes (G3m5, G3m28, G3m26) and one
                          isoallotype (nG3m5). Extensive analysis of sequences (104, 106,
                          113) and genetic data (49–53, 89–96, 102) demonstrate that:
                          –    G3m5 corresponds to the simultaneous expression of arginine
                               at position 115 (IGHG3 CH3 Arg R115) and phenylalanine
                               at position 116 (IGHG3 CH3 Phe F116).
                          –    The nG3m5 isoallotype corresponds to the simultaneous
                               expression of histidine at position 115 (IGHG3 CH3 His
                               H115) and tyrosine at position 116 (IGHG3 CH3 Tyr Y116).
                          –    G3m28 corresponds to the simultaneous expression of argin-
                               ine at position 115 (IGHG3 CH3 Arg R115) and tyrosine at
                               position 116 (IGHG3 CH3 Tyr Y116). This observation is
                               in agreement with the genetic data that show that G3m5 and
                               G3m28 are antithetical and mutually exclusive on the gamma3
                               chains.
                              These data confirm that G3m26 corresponds to arginine at
                          position 115 (IGHG3 CH3 Arg R115). This explains the high
                          frequency of G3m26, present on all Gm5-positive (R115, F116)
                          and G3m28-positive (R115, Y116) gamma3 chains, and its
                          absence, the arginine being replaced by a histidine, on G3m15-
                          positive gamma3 chains which are nG3m5 (H115,Y116).
5.1.4. G3m14 and G3m15    G3m14 has been the subject of discussion concerning its localiza-
                          tion on either the CH2 or CH3 domain with contradictory sero-
                          logical data (discussed in (50)). Extensive analysis of previously
                          published data of usual and uncommon haplotypes, supported by
                          familial studies (47–53, 89–95, 102), led us to postulate that
                          G3m14 corresponds to the simultaneous presence on CH3 of a
                          methionine at position 84 (IGHG3 CH3 Met M84) with the
                          G3m5 mosaic (CH3 Arg R115, Phe F116).
             34   Human Gm, Km, and Am Allotypes and Their Molecular Characterization…      655
5.1.5. G3m16 and G3m21     Two allotypes, G3m16 and G3m21, are localized on the CH2
                           domain. G3m16 correlates with a tryptophane at position 83
                           (IGHG3 CH2 Trp W83), whereas G3m16-negative chains have
                           an arginine at that position (113). G3m21 correlates with a leucine
                           at position 82 (IGHG3 CH2 Leu L82), whereas nG3m21 corre-
                           lates with Pro P82 (113) (IMGT Repertoire, Alignments of alleles,
                           Homo sapiens IGHG3, http://www.imgt.org (5, 6)).
5.1.6. “Silent” G3m        Among data obtained by Dard et al. (113), one sequence was
Allotypes                  noted as unusual as it presents an asparagine (N44) (IGHG3*08 in
                           IMGT Repertoire, Alignments of alleles, Homo sapiens IGHG3,
                           http://www.imgt.org (5, 6)), whereas a serine (S44) was expected,
                           given the G3m5* phenotype. The individual, Mand114, is
                           heterozygous for a normal G3m5* haplotype associated to the
                           unusual haplotype (the one that was sequenced) (113). We postu-
                           late that the unusual haplotype corresponds to G3m5,14,26, with
                           an absence of G3m (10,11,13), as previously demonstrated in the
                           Tunisian family 275 (95, 102). Two nucleotide substitutions in
                           codon 44 (as a result of mutations or of a genic conversion) are
                           the most probable explanation for these silent G3m (10, 11,13)
                           allotypes.
                                      Table 7
                                      The six most prevalent G3m alleles
                                      G3m5,10,11,13,14,26,27        G3m5*
                                      G3m5,6,10,11,14,26,27         G3m6*
                                      G3m5,6,11,24,26               G3m24*
                                      G3m10,11,13,15,27             G3m15*
                                      G3m10,11,13,15,16,27          G3m16*
                                      G3m21,26,27,28                G3m21*
5.1.8. IGHG3 Hinge CNV    For the gamma3 chains, an additional polymorphism results from
Exon Polymorphism         differing numbers (CNV) of hinge exons. The hinge is encoded by
                          2–5 exons, depending on the alleles (104, 106, 112, 113). Thus,
                          the hinge region can vary from 27 to 83 amino acids and can
                          influence structural conformations.
5.2. G3m Alleles          The thirteen G3m allotypes are inherited in different combinations
and the “IMGT G3m         or G3m alleles. The six most prevalent G3m alleles are shown in
Allele Butterfly”         Table 7 and illustrated in Fig. 9, as “IMGT G3m alleles butterfly”
Representation            representation. For convenience, these most common G3m alleles
                          can be written in a simplified form indicated with an asterisk (see
                          Note 10 for correspondence between the WHO/IUIS/IMGT
                          nomenclature and previous designation).
5.3. Correspondence       The correspondence between the G3m alleles and IGHG3 alleles
Between the G3m           is shown in Table 8.
Alleles and IGHG3
Alleles
658        M.-P. Lefranc and G. Lefranc
Fig. 9. “IMGT G3m allele butterfly” representation. The six most prevalent G3m alleles (Table 7) are shown. The first G3m
mosaic on the CH3 domain of IGHG3 (detailed in Subheading 5.1.2) is illustrated by the top part of each butterfly represen-
tation and corresponds to G3m11/nG3m11, G3m27, and the antithetical G3m10/G3m24, and G3m13/G3m6 allotypes. The
second G3m mosaic on the CH3 domain of iGHG3 (detailed in Subheading 5.1.3) is illustrated by the bottom part of each
butterfly representation and corresponds to G3m26, and the antithetical G3m5/nG3m5/G3m28 allotypes. Amino acids
involved in the allotype expression (Table 6) and their position according to the IMGT unique numbering for C domain (14)
are indicated. Two allotypes are on the CH2 domain, G3m16 (Trp W83) and G3m21 (Leu L82). For each G3m allele butterfly,
G3m alleles are indicated with the simplified form (e.g., G3m5*) and the full nomenclature (Table 7). Haplotypes to which
the G3m alleles belong are indicated between square brackets (Table 10) (see Note 11).
    Table 8
    Correspondence between the G3m alleles and IGHG3 alleles
6. nG4m
Isoallotypes
6.1. nG4m(a)                No allotype has been defined for the gamma4 chains of the IgG4
and nG4m(b)                 subclass. The only serologically defined polymorphism corresponds
                            to the isoallotypes nG4m(a) and nG4m(b), described on the CH2
                            domain (138). These antithetical determinants of the gamma4
                            chains behave as allotypes in the IgG4 subclass, but they are pres-
                            ent on the other subclasses and therefore must be considered as
                            isoallotypes.
                                It has been postulated that nG4m(a) was correlated to leucine
                            309 (IGHG4 CH2 Leu L92) and isoallotype nG4m(b) to a dele-
                            tion at that position (152). However, comparison with the transla-
                            tion of IGHG4 sequences did not confirm that deletion and instead
                            showed that it was an amino acid change of leucine into valine
                            (IGHG4 CH2 Val V92) which explained the “disappearance” of
                            the leucine and was responsible for the expression of nG4m(b)
                            chains (1, 104). The nG4m(a) epitope (IGHG4 CH2 Leu L92) is
                            expressed on the gamma1 and gamma3 chains, whereas the
                            nG4m(b) epitope (IGHG4 CH2 Val V92) is expressed on the
                            gamma2 chains (Table 2).
           Table 9
           Correspondence between the nG4m alleles and IGHG4 alleles
                                                                    IMGT                       92
                                                                    Exon                       (79)
                                                                    Eu                         309
                                                                    nG4m(a)/nG4m(b)
           nG4m(a)       IGHG4*01, IGHG4*03, IGHG4*04               Leu L92 ctg
           nG4m(b)       IGHG4*02                                   Val V92 gtg
       a
        Positions in bold are according to the IMGT unique numbering for C domain (14); between paren-
       theses, exon numbering (11), and in italics, Eu numbering (correspondence between C numberings,
       in IMGT Scientific chart, http://www.imgt.org (5, 6))
                34   Human Gm, Km, and Am Allotypes and Their Molecular Characterization…                           661
7. Gm Haplotypes
7.1. Description                The G1m, G2m, and G3m alleles are inherited in fixed combina-
of the Main Gm                  tions or Gm haplotypes. Table 10 shows the eleven most prevalent
Haplotypes                      Gm haplotypes. The nomenclature of the Gm haplotypes takes
                                into account the IGHG gene order in the locus (97, 114, 115).
                                The Gm allotypes are written in the linkage order of the IGHG
                                subclass genes, i.e., IGHG3, IGHG1, and IGHG2, with semico-
                                lons separating the subclasses and comas separating the allotypes;
                                G2m23 is the only allotype defined on gamma 2, two dots being
                                used to indicate that a specimen was tested and found to be nega-
                                tive for G2m23 (47, 48, 52, 53).
    Table 10
    Prevalent Gm haplotypes
Prevalent Gm haplotypesa
7.2. Prevalent Gm         The eleven most prevalent Gm haplotypes are differently repre-
Haplotypes in Different   sented in the Negroid, Khoisan, Caucasoid, and Mongoloid popu-
Populations               lations (Table 10). Thus, Gm5*;3;23 [A] and Gm5*;3;.. [B] are
                          only typical of the Caucasoid populations. Two haplotypes
                          Gm21*;17,1;.. [C] and Gm 21*;17,1,2;.. [D] are shared by the
                          Caucasoid and Mongoloid populations. A unique set of haplotypes
                          characterizes the Negroid populations, Gm5*;17,1,.. [E],
                          Gm6*;17,1;.. [F], and Gm24*;17,1,.. [G]. The haplotype
                          Gm15*;17,1;.. [H] (with or without G3m16) is characteristic of
                          the Khoisan population of southern Africa. Three Gm haplotypes
                          only occur in the Mongoloid populations, Gm16*;17,1;.. [I]
                          largely in the northern hemisphere, Gm5*;3,1;23 [J] and
                          Gm5*;3,1;.. [K] in the southern hemisphere.
                               Haplotypes E, F, and G in the Negroid populations have been
                          found with G1m27 and G1m28 on gamma1 (demonstrated on
                          isolated chains of Lat-IV-5 (haplotype G) and Sno (haplotype F))
                          (96) (see Subheading 5.1.7).
                  Table 11
                  Gm phenotypes observed in the Tunisian population
                  and deduced Gm haplotypes
    Table 12
    Gm haplotype frequencies among Middle Eastern, European, African, and Asian populations
Gm haplotypes
                                           Caucasoid
                        Caucasoid          Mongoloid                            Negroid                             Khoisan         Mongoloid
                                                                                                                                                                         M.-P. Lefranc and G. Lefranc
8. A2m Allotypes
8.1. A2m1 and A2m2       Two allotypes of the IGHA2 gene have been described, A2m1,
                         identified by two groups, independently (39, 40), and A2m2 (41).
                         A2m1 and A2m2 are antithetical and located on the CH1 domain.
                         A2m1 corresponds to a proline at position 115 (IGHA2 CH1 Pro
                         P115) and is also correlated to a proline at position 124 (IGHA2
                         CH1 Pro P124) (153) (Fig. 1b). A2m2 corresponds to a serine
                         at position 115 (IGHA2 CH1 Ser S115) and is also correlated to
                         an arginine at position 124 (IGHA2 CH1 Arg R124) (154, 155)
                         (Fig. 10a).
8.2. A2m1 and A2m2       The frequency of the A2m1 and A2m2 allotypes varies a lot
Allotype Frequency       between and within most populations (53, 77, 82, 90, 94). In
                         Caucasoid populations, almost all individuals are homozygous for
                         the A2m1 allotype. The A2m2 allotype, rare in Caucasoid popula-
                         tions, is present with a frequency of 0.40–0.75 in Mongoloid
                         populations (78) and 0.60–0.85 in Negroid populations (79).
8.3. A2m1 and A2m2       A2m1 and A2m2 allotypes can be determined at the DNA level by
Allotype Determination   RFLP using appropriate restriction enzymes (101). The amino
by RFLP                  acid responsible for the A2m2 allotype, serine 115, is encoded by
                         nucleotides which are part of an EcoRI restriction site (101)
                         (Table 13). Due to a nucleotide substitution, this site is absent in
                         the A2m1 allele. Thus, whereas the restriction enzyme PstI yields
                         two fragments containing the IGHA1 (1.2 kb) and IGHA2 (2 kb)
                         genes (97, 114) when DNA samples are probed with a Ca probe,
                         double digests EcoRI–PstI show two different patterns: one simi-
                         lar to the PstI one for the A2m1 allele and the other with a new
                         0.9 kb band for the A2m2 allele due to the existence of the EcoRI
                         site (101). The determination of A2m alleles by RFLP is particu-
                         larly useful when reagents are not readily available for the sero-
                         logical determination of the allotypes. Moreover, it is the only way
                         to identify A2m alleles when no serum is available (for instance
                         cell lines).
8.4. Sequence Identity   The sequence of the A2m1 chain is identical to the sequence of
and Conformational       the alpha 1 chain at positions 115 (CH1 Pro P115) and 124 (CH1
Difference for A2m1      Pro P124) (109) (Fig. 1b). However, the IGHA2 A2m1 allotype
and the Alpha1           and the IGHA1 isotypic epitope are recognized by different anti-
Isotypic Epitope         bodies (Erna van Loghem, GL, MPL), due to a difference in the
                         disulfide bridge. Indeed, in IgA1 (as in IgA2 A2m2), the cystein
                         (CH1 Cys C123) is normally involved in a heavy-light (H-L)
                         disulfide bridge. In contrast, in IgA2 A2m1, there is no H-L
                         disulfide bridge to the cysteine (CH1 Cys C123) due to a confor-
                         mational hindrance of the Pro P124, and the two light chains are
                         directly linked to each other (102).
666   M.-P. Lefranc and G. Lefranc
             34    Human Gm, Km, and Am Allotypes and Their Molecular Characterization…                                 667
8.5. A2m3 Allotype               The C-terminal region (CH3 domain and/or CHS region) of the
Antithetical to nA2m3            IGHA2*02 allele (TOU II-5) (11) (Fig. 10b) was shown to be
                                 immunogenic and to correspond to a new allotype-designated
                                 A2m3, as it was antithetical to the nA2m3 isoallotype (previously
                                 called nA2m(2)) (unpublished data, Erna van Loghem, GL and
                                 MPL). One or several of the following IGHA2 amino acid changes
                                 may be involved in the A2m3 allotype/nA2m3 isoallotype expres-
                                 sion, Tyr Y85.2/Phe F85.2, Glu E100/Asp D100, Met M124/Leu
                                 L124 (in the CH3), Ileu I134/Val V134, or Ala A143/Val V143
                                 (in the CHS region) (IMGT Repertoire, Alignments of alleles, Homo
                                 sapiens IGHA2, http://www.imgt.org (5, 6)) (Fig. 1b).
8.6. Correspondence              The correspondence between A2m alleles and IGHA2 alleles is
Between A2m Alleles              shown in Table 13. Three A2m alleles, A2m1, A2m2,3, and A2m2,
and IGHA2 Alleles                are defined based on their allotypes. A2m1 corresponds to the
                                 IGHA2*01 allele, A2m2,3 to the IGHA2*02 (TOU II-5) allele,
                                 and A2m2 to the IGHA2*03 allele.
9. Km Allotypes
9.1. Definition                  Allotypes have been identified for the human IGKC gene and are
                                 designated as Km (for “kappa marker”) (previously Inv) (42, 43).
                                      There are three kappa chain allotypes designated Km1, Km2,
                                 and Km3 that define three alleles. In 1961, Ropartz described the
                                 first IG light chain allotype Inv(a) (42), now called Km2. One year
                                 later, the Inv(l) allotype, now called Km1, was described by the
                                 same group (43). In 1962, Steinberg et al. described the third Km
                                 allotype, Inv(b) (44), now called Km3. They found that sera nega-
                                 tive for Km1 and Km2 were always positive for Km3, and that sera
                                 negative for Km3 were always positive for Km1 and mostly also for
                                 Km2. Thus, the three Km allotypes define three alleles, Km3,
                                 Km1,2, and Km1 (detailed in Subheading 9.3)
Fig. 10. IMGT Colliers de Perles of the IGHA2*02 CH1 and CH3 domains. The CH1 and CH3 domains are from IGHA2*02
(IMGT/DomainDisplay, IMGT/GENE-DB (11, 15)) (IMGT®, http://www.imgt.org). The IMGT Colliers de Perles are shown on
two layers. Hatched positions correspond to gaps according to the IMGT unique numbering (14). (a) CH1 domain. The
serine at position 115 (S115) corresponds to A2m2, with the arginine at position 124 (R124) also correlating to that allotype
(11, 101, 155). A proline at positions 115 (P115) and 124 (P124) would correspond to the A2m1 allotype (11, 101, 155).
(b) CH3 domain. One or several of the following amino acids in the CH3 domain, tyrosine Tyr at position 85.2 (Y85.2),
glutamate Glu at position 100 (E100), methionine Met at position 124 (M124), and/or in the CHS (not shown), isoleucine
Ileu I134, alanine Ala A143 may be involved in the A2m3 allotype (11, 15, 155). One or several of the following amino acids,
in the CH3 domain, phenylalanine Phe F85.2, aspartate Asp D100, leucine Leu L124, and/or in the CHS (not shown), valine
Val V134 and valine Val V143 are involved in the antithetical nA2m3 allotype (11, 15, 155).
                                                                                                                                                                668
    Table 13
    Correspondence between A2m alleles and IGHA2 alleles
9.2. Km Allotype         The three Km allotypes, Km1, Km2, and Km3, are determined by
Determination, Km        the hemagglutination inhibition technique (47, 50, 51, 53). Given
Phenotypes, and Km       the rarity of anti-Km2 and anti-Km3 reagent antibodies, only the
Genotypes                Km1 allotype is tested in most of the population studies. Km geno-
                         types and observed phenotypes depend on the number of tested
                         allotypes. In Table 14, are shown:
                         –     The six Km genotypes defined by their Km alleles.
                         –     The five Km phenotypes observed when sera are tested for the
                               three allotypes Km1, Km2, and Km3.
                         –     The three Km phenotypes observed when sera are only tested
                               for allotypes Km1 and Km2.
                         –     The two Km phenotypes observed when sera are only tested
                               for allotype Km1.
9.3. Km Phenotypes       The two allotypes Km1 and Km2 mostly cooccur and are present
and Km Allele            in 10–20% of Caucasoid (47, 53, 54, 67), 40–60% of Negroid
Frequencies in           (79), and 30–60% of Mongoloid (78) populations (Table 15) (1).
Different Populations    Km allele frequency in the different populations is given in Table 16.
                         Details on extensive Km analysis in populations from Lebanon
                           Table 14
                           Km genotypes and observed Km phenotypes depending
                           on the tested Km allotypes
Km phenotypes
                           Table 15
                           Km phenotype
Populations
                          Table 16
                          Km allele frequency
Populations
10. Notes
    Table 17
    Correspondence between Km alleles and IGKC alleles
                                 A                Gm5*;3;23                           Gmb*;f;n
                                 B                Gm5*;3;..                           Gmb*;f;..
                                 C                Gm21*;17,1;..                       Gmg*;z,a;..
                                 D                Gm21*;17,1,2;..                     Gmg*;z,a,x;..
                                 E                Gm5*;17,1;..                        Gmb*;z,a;..
                                 F                Gm6*;17,1;..                        Gmc3*;z,a;..
                                 G                Gm24*;17,1;..                       Gmc5*;z,a;..
                                 H                Gm15*;17,1;..                       Gms*;z,a;..
                                 I                Gm16*;17,1;..                       Gmt*;z,a;..
                                 J                Gm5*;3,1;23                         Gmb*;f,a;n
                                 K                Gm5*;3,1;..                         Gmb*;f,a;..
                                 L                Gm5*;17,1,2;..                      Gmb*;z,a,x;..
                                 M                Gm21*;17,1;23                       Gmg*;z,a;n
                             a
                              24* was previously designated as 6,24*, and 16* as 15,16*
                             b
                              c5* was previously designated as c3,c5*, and t* as s,t*
674      M.-P. Lefranc and G. Lefranc
Acknowledgments
References
  1. Lefranc M-P, Lefranc G (1990) Molecular               that bridge biological and computational
     genetics of immunoglobulin allotype expres-           spheres in bioinformatics. Brief Bioinform
     sion. In: Shakib F (ed) The human IgG sub-            9:263–275
     classes: molecular analysis of structure,         10. Lefranc M-P (2000) Nomenclature of the
     function and regulation (chapter 4). Pergamon,        human immunoglobulin genes. In: Coligan JE,
     Oxford, UK, pp 43–78                                  Bierer BE, Margulies DE, Shevach EM, Strober
  2. Lefranc M-P (2011) Antibody nomenclature:             W (eds) Current protocols in immunology.
     from IMGT-ONTOLOGY to INN definition.                 Wiley, Hoboken, NJ, pp A.1P.1–A.1P.37
     MAbs 3(1):1–2                                     11. Lefranc G, Lefranc M-P (2001) The immuno-
  3. Jefferis R, Lefranc M-P (2009) Human immu-            globulin FactsBook. Academic Press, London,
     noglobulin allotypes: possible implications for       UK, pp 1–458
     immunogenicity. MAbs 1(4):332–338                 12. Lefranc M-P (2007) WHO-IUIS nomencla-
  4. Magdelaine-Beuzelin C, Vermeire S, Goodall            ture subcommittee for immunoglobulins and
     M, Baert F, Noman M, Van Assche G,                    T cell receptors report. Immunogenetics
     Ohresser M, Degenne D, Dugoujon JM,                   59:899–902
     Jefferis R, Rutgeerts P, Lefranc M-P, Watier H    13. Lefranc M-P (2008) WHO-IUIS nomenclature
     (2009) IgG1 heavy chain-coding gene poly-             subcommittee for immunoglobulins and T cell
     morphism (G1m allotypes) and development              receptors report August 2007, 13th Inter-
     of antibodies-to-infliximab. Pharmacogenet            national Congress of Immunology, Rio de Janeiro,
     Genomics 19:383–387                                   Brazil. Dev Comp Immunol 32:461–463
  5. Lefranc M-P (2008) IMGT®, the international       14. Lefranc M-P, Pommié C, Kaas Q, Duprat E,
     ImMunoGeneTics information system® for                Bosc N, Guiraudou D, Jean C, Ruiz M, Da
     immunoinformatics. Methods for querying               Piédade I, Rouard M, Foulquier E, Thouvenin
     IMGT® databases, tools and Web resources in           V, Lefranc G (2005) IMGT unique number-
     the context of immunoinformatics. Mol                 ing for immunoglobulin and T cell receptor
     Biotechnol 40:101–111                                 constant domains and Ig superfamily C-like
  6. Lefranc M-P, Giudicelli V, Ginestoux C,               domains. Dev Comp Immunol 29:185–203
     Jabado-Michaloud J, Folch G, Bellahcene F,        15. Giudicelli V, Chaume D, Lefranc M-P (2005)
     Wu Y, Gemrot E, Brochet X, Lane J, Regnier            IMGT/GENE-DB: a comprehensive database
     L, Ehrenmann F, Lefranc G, Duroux P (2009)            for human and mouse immunoglobulin and T
     IMGT®, the international ImMunoGeneTics               cell receptor genes. Nucleic Acids Res
     information system®. Nucleic Acids Res                33:D256–D261
     37:D1006–D1012                                    16. Kaas Q, Ruiz M, Lefranc M-P (2004)
  7. Giudicelli V, Lefranc M-P (1999) Ontology             IMGT/3Dstructure-DB             and    IMGT/
     for immunogenetics: IMGT-ONTOLOGY.                    StructuralQuery, a database and a tool for
     Bioinformatics 15:1047–1054                           immunoglobulin, T cell receptor and MHC struc-
  8. Duroux P, Kaas Q, Brochet X, Lane J,                  tural data. Nucleic Acids Res 32:D208–D210
     Ginestoux C, Lefranc M-P, Giudicelli V            17. Ehrenmann F, Kaas Q, Lefranc M-P (2010)
     (2008) IMGT-Kaleidoscope, the formal                  IMGT/3Dstructure-DB              and   IMGT/
     IMGT-ONTOLOGY paradigm. Biochimie                     DomainGapAlign: a database and a tool for
     90:570–583                                            immunoglobulins or antibodies, T cell recep-
  9. Lefranc M-P, Giudicelli V, Regnier L, Duroux          tors, MHC, IgSF and MhcSF. Nucleic Acids
     P (2008) IMGT®, a system and an ontology              Res 38:D301–D307
          34   Human Gm, Km, and Am Allotypes and Their Molecular Characterization…                 675
18. Kaas Q, Lefranc M-P (2007) IMGT Colliers         33. Natvig JB (1966) Gm(g)–a “new” gamma-
    de Perles: standardized sequence-structure           globulin factor. Nature 211:318–319
    representations of the IgSF and MhcSF super-     34. van Loghem E, Martensson L (1967) Genetic
    family domains. Curr Bioinform 2:21–30               (Gm) determinants of the g2c (Vi) subclass of
19. Kaas Q, Ehrenmann F, Lefranc M-P (2007)              human IgG immunoglobulins. A study with
    IG, TR and IgSf, MHC and MhcSF: what do              special reference to Gm(c3) and Gm(c5), and
    we learn from the IMGT Colliers de Perles?           their relationship with the Gm(b) determinants.
    Brief Funct Genomic Proteomic 6:253–264              Vox Sang 13:369–392
20. Grubb R (1956) Agglutination of erythrocytes     35. van Loghem E, Grobbelaar BG (1971) A new
    coated with “incomplete” anti-Rh by certain          genetic marker of human IgG3 immunoglob-
    rheumatoid arthritic sera and some other sera.       ulin. Evolutionary dissociation of Gm allo-
    The existence of human serum groups. Acta            types. Vox Sang 21:405
    Pathol Microbiol Scand 39:195–197                36. Schanfield MS, Fudenberg HH (1974) Gm
21. Grubb R, Laurell AB (1956) Hereditary sero-          (Ray), a new allotypic marker on human IgG3.
    logical human serum groups. Acta Pathol              Vox Sang 26:133–140
    Microbiol Scand 39:390–398                       37. Blanc M, Dugoujon JM, Ruffié J (1976) [A
22. Harboe M, Lundevall J (1959) A new type in           new genetic marker for human IgG3 usually
    the Gm system. Acta Pathol Microbiol Scand           associated with Gm (21) factor found in the
    45:357–370                                           caucasian and mongolian populations]. In
23. Steinberg AG, Wilson JA (1963) Studies on            French. Un nouveau marqueur génétique des
    hereditary gamma globulin factors: evidence          IgG3 humaines habituellement associé au fac-
    that Gm (b) in Whites and Negroes is not the         teur Gm(21) dans les populations caucasoides
    same and that Gm-like is determined by an            et mongoloides. C R Acad Sci Paris
    allele at the Gm locus. Am J Hum Genet               283:1461–1463
    15:96–105                                        38. van Loghem E, Blanc M, de Lange G (1977)
24. Gold ER, Ropartz C, Rivat L, Rousseau PY             Human IgG3 allotypes, with special reference
    (1965) Gm(f)–a determinant of human                  to a new allotype related to G3m(g)
    gamma-globulin. Vox Sang 10:299–302                  (G3m(21)). J Immunogenet 4:371–383
25. Gold ER, Mandy WJ, Fudenberg HH (1965)           39. Vyas GN, Fudenberg HH (1969) Am(1), the
    Relation between Gm(f) and the structure of          first genetic marker of human immunoglobu-
    the gamma-globulin molecule. Nature                  lin A. Proc Natl Acad Sci USA
    207(5001):1099–1101                                  64:1211–1216
26. Litwin SD, Kunkel HG (1966) A gamma-             40. Kunkel HG, Smith WK, Joslin FG, Natvig JB,
    globulin genetic factor related to Gm(a) but         Litwin SD (1969) Genetic marker of the
    localized to a different portion of the same         gamma-A2 subgroup of gA immunoglobulins.
    heavy chains. Nature 210:866                         Nature 223:1247–1248
27. Kunkel HG, Yount WJ, Litwin SD (1966)            41. van Loghem E, Wang AC, Shuster J (1973) A
    Genetically determined antigen of the Ne sub-        new genetic marker of human immunoglobu-
    group of gamma-globulin: detection by pre-           lins determined by an allele at the a2 locus.
    cipitin analysis. Science 154(752):1041–1043         Vox Sang 24:481–488
28. Harboe M (1959) A new hemagglutinating           42. Ropartz C, Lenoir J, Rivat L (1961) A new
    substance in the Gm system, anti-Gmb. Acta           inheritable property of human sera: the Inv
    Pathol Microbiol Scand 47:191–198                    factor. Nature 189:586–587
29. Steinberg AG, Giles BD, Stauffer R (1960) A      43. Ropartz C, Rivat L, Rousseau PY (1962) Deux
    Gm-like factor present in Negroes and rare or        nouveaux facteurs dans les systèmes héréditaires
    absent in whites: its relation to Gma and Gmx.       de gammaglobuline: le Gm(e) et l’Inv(l).
    Am J Hum Genet 12:44–51                              Proceedings of the 9th Society of blood
30. Ropartz C, Rivat L, Rousseau PY (1963) Le            Transfusion. Karger Baset, Mexico, p 455
    Gm(b) et ses problèmes. Vox Sang 8:717–723       44. Steinberg AG, Wilson JA, Lanset S (1962) A
31. Steinberg AG, Goldblum R (1965) A genetic            new human gamma globulin factor deter-
    study of the antigens associated with the            mined by an allele at the Inv locus. Vox Sang
    Gm(b) factor of human g-globulin. Am J Hum           7:151–156
    Genet 17:133–147                                 45. WHO (1976) Review of the notation for the
32. Martensson L, van Loghem E, Matsumoto H,             allotypic and related markers of human immu-
    Nielsen J (1966) Gm(s) and Gm(t): genetic            noglobulins. WHO meeting on human immu-
    determinants of human gamma-globulin. Vox            noglobulin allotypic markers. J Immunogenet
    Sang 11:393–418                                      3:357–362
676      M.-P. Lefranc and G. Lefranc
46. WHO. Review of the notation for the allo-             the Inv and the ISf systems in Danes. Hum
    typic and related markers of human immuno-            Hered 20:275–280
    globulins (1976) WHO meeting on human             59. Jenkins T, Zoutendyk A, Steinberg AG (1970)
    immunoglobulin allotypic markers, held July           Gammaglobulin groups (Gm and Inv) of vari-
    16–19, 1974, Rouen, France. Report amended            ous Southern African populations. Am J Phys
    June 1976. Eur J Immunol 6:599                        Anthropol 32:197–218
47. Lefranc G, Loiselet J, Rivat L, Ropartz C         60. Mayr WR, Mickerts D (1970) Der menschliche
    (1976) Gm, Km and ISf allotypes in the                Gammaglobulin-Polymorphismus. Acta Biol
    Lebanese population. Acta Anthropogenet               Med Germ 25:475–482
    1:34–45                                           61. Bonné B, Godber M, Ashbel S, Mourant AE,
48. Lefranc G, Rivat L, Rivat C, Loiselet J,              Tills D (1971) South-Sinai Beduin. A prelimi-
    Ropartz C (1976) Evidence for “deleted” or            nary report on their inherited blood factors.
    “silent” genes homozygous at the locus cod-           Am J Phys Anthropol 34:397–408
    ing for the constant region of the g3 chain.      62. Ropartz C, Rivat L, Rousseau PY, Legueult
    Am J Hum Genet 28:51–61                               LC (1972) Frequency of Gm, Inv and ISf
49. Lefranc G, Rivat L, Salier JP, van Loghem E,          phenotypes in the population of 4 Yugoslavian
    Aydenian H, Zalzal P, Chakhachiro L, Loiselet         villages. Hum Hered 22:508–518
    J, Ropartz C (1977) Recombination, muta-          63. Salzano FM, Steinberg AG, Tepfenhart MA
    tion, or constitutive expression at a Gm locus        (1973) Gm and Inv allotypes of Brazilian Cayapo
    and familial hypergammaglobulinemia. Am J             Indians. Am J Hum Genet 25:167–177
    Hum Genet 29:523–536
                                                      64. Steinberg AG (1973) Contribution of the Gm
50. Lefranc G (1978) Allotypes et haplotypes des          and Inv allotypes to the characterization of
    immunoglobulines dans les communautés                 human populations. Israel J Med Sci
    libanaises: intérêt exceptionnel en immunogé-         9:1249–1256
    nétique et séro-anthropologie [in French].
                                                      65. Steinberg AG (1973) Gm and Inv allotypes of
    PhD Thesis. Université de Rouen, France
                                                          some Sidamo Ethiopians. Am J Phys Anthropol
51. Lefranc G (1978) Immunoglobulin allotypes             39:403–408
    and haplotypes in the Lebanese communities:
                                                      66. Wiebecke D (1973) Die frequenczen von Gm
    discussion of unusual genetic events and pop-
                                                          (1,2,3,5) und Inv (1) in der Bevölkerung der
    ulation structure. PhD Thesis. Université de
                                                          Bundesrepublik Deustchland. Humangenetik
    Rouen, France, pp 1–137
                                                          18:175–180
52. Lefranc G, Rivat L, Serre JL, Lalouel JM, Pison   67. Fraser GR, Volkers WS, Bernini LF, van
    G, Loiselet J, Ropartz C, de Lange G, van             Loghem E, Meera Khan P, Nijenhuis LE
    Loghem E (1978) Common and uncommon                   (1974) Gene frequencies in a Dutch popula-
    immunoglobulin haplotypes among Lebanese              tion. Hum Hered 24:435–448
    communities. Hum Genet 41:197–209
                                                      68. Daveau M, Rivat L, Langaney A, Afifi N, Bois E,
53. Lefranc G, de Lange G, Rivat L, Langaney A,           Ropartz C (1975) Gm and Inv allotypes in
    Lefranc M-P, Ellouze F, Sfar G, Sfar M, van           French Guiana Indians. Hum Hered 25:88–92
    Loghem E (1979) Gm, Am and Km immuno-
    globulin allotypes of two populations in          69. Schanfield MS, Gergely J, Fudenberg HH
    Tunisia. Hum Genet 50:199–211                         (1975) Immunoglobulin allotypes of European
                                                          populations. I. Gm and Km(Inv) allotypic mark-
54. Ropartz C, Rivat L, Rousseau PY, Baitsch H,           ers in Hungarians. Hum Hered 25:370–377
    van Loghem J (1963) The Gm and Inv systems
    in Europe. Acta Genet Stat Med 13:109–123         70. Schanfield MS, Herzog P, Fudenberg HH
                                                          (1975)       Immunoglobulin     allotypes    of
55. Ropartz C, Rousseau PY, Rivat L (1963) Les            European populations. II.Gm, Am and
    groupes de gamma-globulines Gm et Inv :               Km(Inv) allotypic markers in Czechoslovakians.
    Etude génétique et anthropologique. Bull Soc          Hum Hered 25:382–392
    Anthropol Paris 4(Xie ser.):458–469
                                                      71. Steinberg AG, Jenkins T, Nurse GT,
56. Steinberg AG, Matsumoto H (1964) Studies              Harpending HC (1975) Gammaglobulin
    on the Gm, Inv, Hp and Tf serum factors of            groups of the Khoisan peoples of Southern
    Japanese populations and families. Hum Biol           Africa: evidence for polymorphism for a
    36:77–85                                              Gm1,5,13,14,21 haplotype among the San. Am J
57. Taleb N, Ruffie J (1968) Hémotypologie des            Hum Genet 27:528–542
    populations jordaniennes. Bull Mém Soc            72. Lalouel JM, Loiselet J, Lefranc G, Chaiban D,
    Anthrop Paris 3(XIIe sér.):269–282                    Chakhachiro L, Rivat L, Ropartz C (1976)
58. Ropartz C, Rivat L, Rousseau PY, Lauridsen            Genetic differentiation among Lebanese com-
    UB, Persson I (1970) A survey of 9 Gm-factors,        munities. Acta Anthropogenet 1:15–33
           34   Human Gm, Km, and Am Allotypes and Their Molecular Characterization…                  677
73. Piazza A, van Loghem E, de Lange G, Curtoni         85. Sukernik RI, Osipova LP, Schanfield MS
    ES, Ulizzi L, Terrenato L (1976)                        (1992) Distribution of GM allotypes and
    Immunoglobulin allotypes in Sardinia. Am J              deleted IGHG1 haplotypes in the Sel’kups in
    Hum Genet 28:77–86                                      comparison to the Forest Nentsi of north-
74. Gershowitz H, Neel JV (1978) The immuno-                western Siberia. Exp Clin Immunogenet
    globulin allotypes (Gm and Km) of twelve                9:15–23
    Indian tribes of Central and South America.         86. Schanfield MS (1992) Immunoglobulin
    Am J Phys Anthropol 49:289–301                          allotypes (GM and KM) indicate multiple
75. Rivat L, Rivat C, Cook CE, Steinberg AG                 founding populations of Native Americans:
    (1978) Gm(28). Un nouvel allotype du sys-               evidence of at least four migrations to the New
    tème Gm présent sur les IgG3: Intérêt particu-          World. Hum Biol 64:381–402
    lier de son étude dans les populations négroides.   87. Yacoubi Loveslati B, Sanchez-Mazas A,
    Ann Immunol Inst Pasteur 129:33–45                      Ennafaa H, Marrakchi R, Dugoujon J-M,
76. Steinberg AG, Cook CE (1981) The distribu-              Lefranc M-P, Ben Ammar Elgaaied A (2001)
    tion of the human immunoglobulin allotypes.             A study of Gm allotypes and immunoglobulin
    Oxford University Press, Oxford                         heavy gamma chain IGHG genes in Berbers,
77. Chaabani H, Helal AN, van Loghem E,                     Arabs and sub-Saharan Africans from Jerba
    Langaney A, Benammar Elgaaied A, Rivat                  Island, Tunisia. Eur J Immunogenet
    Peran L, Lefranc G (1984) Genetic study of              28:531–538
    Tunisian Berbers. I. Gm, Am and Km immu-            88. Geha RS, Malakian A, Lefranc G, Chaiban D,
    noglobulin allotypes and ABO blood groups.              Serre JL (1976) Immunologic reconstitution
    J Immunogenet 11:107–113                                in severe combined immunodeficiency follow-
78. de Lange G, Zhong FM, Henke J, Feng ZC,                 ing transplantation with parental bone mar-
    Bernhardt R, van Leeuwen F, van Eede P                  row. Pediatrics 58:451–455
    (1985) Immunoglobulin allotypes in a Chinese        89. van Loghem E, Natvig JB (1970) Uncommon
    population: comparison of haplotype frequen-            Gm gene complexes. Vox Sang 18:421–434
    cies with other Asian groups. J Immunogenet         90. van Loghem E, Natvig JB, Matsumoto H
    12:191–195                                              (1970) Genetic markers of immunoglobulins
79. van Loghem E (1986) Immunoglobulin mark-                in Japanese families. Inheritance of associated
    ers. In: Cavalli-Sforza LL (ed) African pygmies         markers belonging to one IgA and three IgG
    (chapter 14). Academic Press, Orlando, p 201            subclasses. Ann Hum Genet 33:351–359
80. Helal AN, Rivat-Peran L, van Loghem E, de           91. Lefranc G, Dumitresco SM, Salier JP, Rivat L,
    Lange G, Langaney A, Lefranc G (1988)                   de Lange G, van Loghem E, Loiselet J (1979)
    Further contribution of common Gm-Am                    Familial lack of the IgG3 subclass: gene
    haplotypes and Km alleles in the characteriza-          elimination or turning off expression and neu-
    tion of the Tunisian population. Exp Clin               tral evolution in the immune system. J
    Immunogenet 5:1–14                                      Immunogenet 6:215–221
81. Helal AN, de Lange G, van Loghem E,                 92. Salier JP, Rivat L, Daveau M, Lefranc G,
    Lefranc G (1988) Uncommon Gm haplotypes                 Breton P, De Menibus CH, Henocq A,
    in the Tunisian population: further contribu-           Fudenberg HH (1980) Quantitative studies
    tion to the genetics of the IgG immunoglobu-            of Gm allotypes. V. Simultaneous presence of
    lins. Exp Clin Immunogenet 5:24–38                      latent Gm allotypes and deficient Gm genes in
82. Soua Z, Ghanem N, Salem B, Lefranc G,                   a family with hypogammaglobulinaemic
    Lefranc M-P (1989) Frequencies of the human             probands. J Immunogenet 7:123–135
    immunoglobulin            IGHA2*M1           and    93. van Loghem E, Sukernik RI, Osipova LP,
    IGHA2*M2 alleles corresponding to the                   Zegers BJM, Matsumoto H, De Lange G,
    A2m(1) and A2m(2) allotypes in the French,              Lefranc G (1980) Gene deletion and gene
    Lebanese, Tunisian and Black African popula-            duplication within the cluster of human heavy
    tions. Nucleic Acids Res 17:3625                        chain genes. Selective absence of IgG sub-
83. Zhao TM, Lee TD (1989) Gm and Km allo-                  classes. J Immunogenet 7:285–299
    types in 74 Chinese populations: a hypothesis       94. Lefranc M-P (1980) Les allotypes Gm, Am et
    of the origin of the Chinese nation. Hum                Km des immunoglobulines humaines dans
    Genet 83:101–110                                        deux échantillons de la population Tunisienne
84. Blanc M, Sanchez-Mazas A, Van Blyenburgh                (Mahdia et Sfax): contribution à la connais-
    NH, Sevin A, Pison G, Langaney A (1990)                 sance séro-anthropologique de cette popula-
    Interethnic genetic differentiation: Gm poly-           tion et à la génétique des immunoglobulines
    morphism in eastern Senegal. Am J Hum                   [in French] Thesis, Série 3° cycle n°16,
    Genet 46:383–392                                        Université Paris Sud, France, pp 1–108
678       M.-P. Lefranc and G. Lefranc
 95. Lefranc G, Lefranc M-P, Helal AN, Boukef K,            (Gmb0, b1, c3, c5, u) with an IGHG4 con-
     Chaabani H, Gandoura MS, van Loghem E                  verted region and three hinge exons.
     (1982) Unusual heavy chains of human IgG               Immunogenetics 30:250–257
     immunoglobulins: rearrangements of the CH         107. Keyeux G, Lefranc G, Lefranc M-P (1989) A
     domain exons. J Immunogenet 9:1–9                      multigene deletion in the human IGH constant
 96. van Loghem E, de Lange G, van Leeuwen                  region locus involves highly homologous hot
     AM, van Eede PH, Nijenhuis LE, Lefranc                 spots of recombination. Genomics 5:431–441
     M-P, Lefranc G (1982) Human IgG allotypes         108. Lefranc M-P, Hammarström L, Smith CI,
     co-occurring in more than one IgG subclass.            Lefranc G (1991) Gene deletions in the
     Vox Sang 43:301–309                                    human immunoglobulin heavy chain constant
 97. Lefranc M-P, Lefranc G, Rabbitts TH (1982)             region locus: molecular and immunological
     Inherited deletion of immunoglobulin heavy             analysis. Immunodefic Rev 2:265–281
     chain constant region genes in normal human       109. Bensmana M, Chuchana P, Lefranc G, Lefranc
     individuals. Nature 300:760–762                        M-P (1991) Sequence of the CH1 and hinge-
 98. Lefranc M-P, Lefranc G, de Lange G, Out TA,            CH2 exons of the human immunoglobulin
     van den Broek PJ, van Nieuwkoop J, Radl J,             IGHA2 A2m(2) allele: comparison with the
     Helal AN, Chaabani H, van Loghem E,                    nonallelic and allelic IGHA genes. Cytogenet
     Rabbitts TH (1983) Instability of the human            Cell Genet 56:128
     immunoglobulin heavy chain constant region        110. Lefranc M-P, Hammarström L, Smith CIE,
     locus indicated by different inherited chromo-         Lefranc G (1993) Gene deletions in the
     somal deletions. Mol Biol Med 1:207–217                human immunoglobulin heavy chain constant
 99. Lefranc G, Chaabani H, van Loghem E,                   region locus: molecular and immunological
     Lefranc M-P, de Lange G, Helal AN (1983)               analysis. In: Rosen FS, Seligmann M (eds)
     Simultaneous absence of the human IgG1,                Immunodeficiencies (chapter 5). Harwood
     IgG2, IgG4 and IgA1 subclasses: immuno-                Academic Publishers, Newark, New Jersey,
     logical and immunogenetical considerations.            USA, pp 99–109
     Eur J Immunol 13:240–244                          111. Wiebe V, Helal A, Lefranc M-P, Lefranc G
100. Flanagan JG, Lefranc M-P, Rabbitts TH                  (1994) Molecular analysis of the T17 immu-
     (1984) Mechanisms of divergence and con-               noglobulin CH multigene deletion (del
     vergence of the human immunoglobulin                   A1-GP-G2-G4-E). Hum Genet 93:520–528
     alpha1 and alpha2 constant region gene            112. Dard P, Huck S, Frippiat JP, Lefranc G,
     sequences. Cell 36:681–688                             Langaney A, Lefranc M-P, Sanchez-Mazas A
101. Lefranc M-P, Rabbitts TH (1984) Human                  (1997) The IGHG3 gene shows a structural
     immunoglobulin heavy chain A2 gene allo-               polymorphism characterized by different
     type determination by restriction fragment             hinge lengths: sequence of a new 2-exon hinge
     length polymorphism. Nucleic Acids Res                 gene. Hum Genet 99:138–141
     12:1303–1311                                      113. Dard P, Lefranc M-P, Osipova L, Sanchez-
102. Lefranc M-P (1984) Recombinaisons, délé-               Mazas A (2001) DNA sequence variability of
     tions et conversions de gènes de la région con-        IGHG3 alleles associated to the main G3m
     stante des chaînes lourdes des immunoglobulines        haplotypes in human populations. Eur J Hum
     humaines [in French]. PhD Thesis. Université           Genet 9:765–772
     Montpellier 2, France                             114. Flanagan JG, Rabbitts TH (1982) Arrangement
103. Lefranc M-P, Helal AN, de Lange G, Chaabani            of human immunoglobulin heavy chain con-
     H, van Loghem E, Lefranc G (1986) Gene                 stant region genes implies evolutionary dupli-
     conversion in human immunoglobulin g locus             cation of a segment containing g, e and a
     shown by unusual location of IgG allotypes.            genes. Nature 300:709–713
     FEBS Lett 196:96–102                              115. Rabbitts TH, Flanagan JG, Lefranc M-P
104. Huck S, Fort P, Crawford DH, Lefranc M-P,              (1983) Flexibility and change within the
     Lefranc G (1986) Sequence of a human immu-             human immunoglobulin gene locus. In:
     noglobulin g3 heavy chain constant region              Chater KF et al (eds) Genetic rearrangement.
     gene: comparison with the other human Cg               Croom Helm, London, UK, pp 143–154
     genes. Nucleic Acids Res 14:1779–1789             116. Ghanem N, Lefranc M-P, Lefranc G (1988)
105. Lefranc M-P, Lefranc G (1987) Human immu-              Definition of the RFLP alleles in the human
     noglobulin heavy-chain multigene deletions in          immunoglobulin IGHG gene locus. Eur
     healthy individuals. FEBS Lett 213:231–237             J Immunol 18:1059–1065
106. Huck S, Lefranc G, Lefranc M-P (1989) A           117. Ghanem N, Dugoujon JM, Bensmana M,
     human immunoglobulin IGHG3 allele                      Huck S, Lefranc M-P, Lefranc G (1988)
            34   Human Gm, Km, and Am Allotypes and Their Molecular Characterization…                    679
     Restriction fragment haplotypes in the human        128. Balbin M, Grubb A, de Lange GG, Grubb R
     immunoglobulin IGHG locus and their cor-                 (1994) DNA sequences specific for Caucasian
     relation with the Gm polymorphism. Eur J                 G3m (b) and (g) allotypes: allotyping at the
     Immunol 18:1067–1072                                     genomic level. Immunogenetics 39:187–193
118. Ghanem N, Dugoujon JM, Lefranc M-P,                 129. Brusco A, de Lange GG, Boccazzi C, Carbonara
     Lefranc G (1989) BstEII restriction fragment             AO (1995) Molecular characterization of G2m
     alleles and haplotypes of the human IGHG                 (n+) and G2m (n-)allotypes. Immunogenetics
     genes with reference to the BamHI/SacI                   42:414–417
     RFLPs and to the Gm polymorphism. Exp               130. Hougs L, Svejgaard A, Barington T (2001)
     Clin Immunogenet 6:39–54                                 The first constant-domain (CH1) exon of
119. Ghanem N, Bensmana M, Dugoujon JM,                       human IGHG2 is polymorphic and in strong
     Constans J, Lefranc M-P, Lefranc G (1989)                linkage disequilibrium with the (CH2) exon
     BamHI and SacI RFLPs of the human immuno-                polymorphism encoding the G2m(n+) allotype
     globulin IGHG genes with reference to the Gm             in Caucasians. Immunogenetics 52:242–248
     polymorphism in African people. Evidence for a      131. Frangione B, Franklin EC, Fudenberg HH,
     major polymorphism. Hum Genet 83:37–4418                 Koshland ME (1966) Structural studies of
120. Hammarström L, Ghanem N, Smith CIE,                      human gG myeloma proteins of different anti-
     Lefranc G, Lefranc M-P (1990) RFLP of                    genic subgroups and genetic specificities.
     human immunoglobulin genes. Exp Clin                     J Exp Med 124:715–732
     Immunogenet 7:7–19                                  132. Press EM, Hogg NM (1970) The amino-acid
121. Dard P, Sanchez-Mazas A, Dugoujon JM, de                 sequences of the Fd fragments of two human
     Lange G, Langaney A, Lefranc M-P, Lefranc                g1 heavy chains. Biochem J 117:641–660
     G (1996) DNA analysis of the immunoglobu-           133. Burton RM, Deutsch HF (1970) The struc-
     lin IGHG loci in a Mandenka population from              ture of peptides of Fc-fragments from gG-
     eastern Senegal: correlation with Gm haplo-              globulins of known allotypic specificity.
     types and hypotheses for the evolution of the            Immunochemistry 7:145–156
     Ig CH region. Hum Genet 98:36–47                    134. Natvig JB, Turner MW (1971) Localization of
122. Osipova LP, Posukh OL, Wiebe VP, Miyazaki                Gm markers to different molecular regions of the
     T, Matsumoto H, Lefranc G, Lefranc M-P                   Fc fragment. Clin Exp Immunol 8:685–700
     (1999) BamHI-SacI RFLP and Gm analysis of           135. Michaelsen TE, Frangione B, Franklin EC
     the immunoglobulin IGHG genes in the                     (1977) The amino acid sequence of a human
     Northern Selkups (west Siberia): new haplo-              immunoglobulin G3m(g) pFc’ fragment.
     types with deletion, duplication and triplica-           J Immunol 119:558–563
     tion. Hum Genet 105:530–541
                                                         136. Pardo AG, Rosenwasser ES, Frangione B
123. Gold ER, Fudenberg HH (1967) Chromic                     (1978) The primary structure of a human
     chloride: a coupling reagent for passive hemag-          immunoglobulin G2 (IgG2) pFc’fragment.
     glutination reactions. J Immunol 99(5):                  J Immunol 121:1040–1044
     859–866
                                                         137. Natvig JB, Kunkel HG, Joslin FG (1969)
124. de Lange G (1988) Monoclonal antibodies                  Delineation of two antigenic markers, “non a”
     against human immunoglobulin allotypes.                  and “non g” related to the genetic antigens of
     PhD Thesis. London, Amsterdam, pp 1–175                  human gamma globulins. J Immunol
125. Zelaschi D, Newby C, Parsons M, van West B,              102:611–617
     Cavalli-Sforza     LL,    Herzenberg        LA,     138. Kunkel HG, Joslin FG, Penn GM, Natvig JB
     Herzenberg LA (1983) Human immuno-                       (1970) Genetic variants of gG4 globulin. A
     globulin allotypes: previously unrecognized              unique relationship to other classes of gG
     determinants and alleles defined with mono-              globulin. J Exp Med 13:508–520
     clonal antibodies. Proc Natl Acad Sci USA
     80:3762–3766                                        139. van Loghem E, de Lange G (1972) ‘non-b’,
                                                              antigenic determinant of IgG. Vox Sang
126. Kurth JH, Bowcock AM, Erlich HA, Nevo S,                 22:193–199
     Cavalli-Sforza LL (1991) Km typing with
                                                         140. Gaarder PI, Natvig JB (1972) Two new anti-
     PCR: application to population screening. Am
                                                              gens of human IgG, “non b0” and “non b1”,
     J Hum Genet 48(3):613–620
                                                              related to the Gm system. J Immunol
127. Balbin M, Grubb A, Abrahamson M, Grubb R                 108:617–621
     (1991) Determination of allotypes G1m (f) and
                                                         141. van Loghem E, de Lange G (1976) The first
     G1m (z) at the genomic level by subclass-specific
                                                              example of an isoallotype of human IgG
     amplification of DNA and use of allele-specific
                                                              located on the Fd fragment. Scand J Immunol
     probes. Exp Clin Immunogenet 8:88–95
                                                              5:1015–1019
680       M.-P. Lefranc and G. Lefranc
142. van Loghem E, de Lange G, Koistinen J             154. Toraño A, Putnam FW (1978) Complete
     (1976) The first isoallotype of human IgA              amino acid sequence of the alpha 2 heavy
     proteins. An antigenic determinant occurring           chain of a human IgA2 immunoglobulin of
     as allotype in the IgA2 subclass and as isotype        the A2m (2) allotype. Proc Natl Acad Sci USA
     in the IgA1 subclass. Scand J Immunol                  75(2):966–969
     5:161–164                                         155. Flanagan JG, Lefranc M-P, Rabbitts TH
143. van Loghem E (1971) Formal genetics of the             (1984) Mechanisms of divergence and con-
     immunoglobulin systems. Ann N Y Acad Sci               vergence of the human immunoglobulin alpha
     190:136–149                                            1 and alpha 2 constant region gene sequences.
144. Wang AC, Shuster J, Fudenberg HH (1969)                Cell 36(3):681–688
     Evolutionary origin of the Gm“a” peptide of       156. Baglioni C, Alescio Zonta L, Cioli D, Carbonara
     immunoglobulins. J Mol Biol 41:83–86                   A (1966) Allelic antigenic factor Inv(a) of the
145. Cook CE, Steinberg AG (1979) An amino                  light chains of human immunoglobulins:
     acid substitution in the g1 chain of human             chemical basis. Science 152:1517–1519
     immunoglobulin G associated with the Gm           157. Steinberg AG, Milstein CP, McLaughlin CL,
     (2) allotype. Mol Immunol 16:555–558                   Solomon A (1974) Structural studies of an
146. Wolfenstein-Todel C, Frangione B, Prelli F,            Inv (1, -2) kappa light chain. Immunogenetics
     Franklin EC (1976) The amino acid sequence             2:1–10
     of “heavy chain disease” protein ZUC. Structure   158. Milstein CP, Steinberg AG, McLaughlin CL,
     of the Fc fragment of immunoglobulin G3.               Solomon A (1974) Amino acid sequence
     Biochem Biophys Res Commun 71:907–914                  change associated with genetic marker Inv(2)
147. Frangione B, Rosenwasser E, Prelli F, Franklin         of     human      immunoglobulin.        Nature
     EC (1980) Primary structure of human g3                248:160–161
     immunoglobulin deletion mutant: g3 heavy-         159. van Loghem E, Aalberse RC, Matsumoto H
     chain disease protein Wis. Biochemistry                (1984) A genetic marker of human IgE heavy
     19:4304–4308                                           chains, Em(1). Vox Sang 46:195–206
148. Alexander A, Steinmetz M, Barritault D,           160. Ghanem N, Dariavach P, Bensmana M,
     Frangione B, Franklin EC, Hood L, Buxbaum              Chibani J, Lefranc G, Lefranc M-P (1988)
     JN (1982) Gamma heavy chain disease in                 Polymorphism of immunoglobulin lambda
     man: cDNA sequence supports partial gene               constant region genes in populations from
     deletion model. Proc Natl Acad Sci USA                 France, Lebanon and Tunisia. Exp Clin
     79(10):3260–3264                                       Immunogenet 5:186–195
149. Recht B, Frangione B, Franklin E, van Loghem      161. Lefranc M-P, Lefranc G (1992) Les
     E (1981) Structural studies of a human g3              Immunoglobulines humaines. In: Breton-
     myeloma protein (GOE) that binds Staph                 Gorius J, Reyes F, Rochant H, Rosa J, Vernant
     protein A. J Immunol 127:917–923                       JP (eds) L’hématologie de Bernard Dreyfus
150. Matsumoto H, Ito S, Miyazaki T, Ohta T                 (chapter 20). Médecine-Sciences Flammarion,
     (1983) Structural studies of a human g3                pp 197–254
     myeloma protein (JIR) bearing the allotypic       162. Lefranc M-P, Pallarès N, Frippiat J-P (1999)
     marker Gm(st). J Immunol 131: 1865–1870                Allelic polymorphisms and RFLP in the human
151. Matsumoto H, Ito S, Miyazaki T, Kawai N                immunoglobulin lambda light chain locus.
     (1986) Structural studies of another human             Hum Genet 104:361–369
     IgG3 myeloma protein (Kam) carrying the           163. Lefranc M-P, Lefranc G (2004) Immunoglo-
     allotypic markers Gm(s,t) and its alteration           bulin lambda (IGL) genes of human and
     induced by chemical modification. In:                  mouse. In: Honjo T, Alt FW, Neuberger MS
     Brinkmann B, Henningsen K (eds) Advances               (eds) Molecular biology of B cells. Academic
     in forensic haemogenetics, Springer Verlag,            Press, Elsevier Science Amsterdam, The
     Berlin-Heidelberg, Germany vol I. p 153                Netherlands, pp 37–59
152. Abel CA, Despont JP (1974) An amino acid          164. Goetze AM, Zhang Z, Liu L, Jacobsen FW,
     deletion associated with the IgG4b allotype of         Flynn GC (2011) Rapid LC-MS screening for
     human      IgG4      myeloma      proteins.   J        IgG Fc modifications and allelic variants in
     Immunogenet 1:79–82                                    blood. Mol Immunol 49:338–352
153. Tsuzukida Y, Wang CC, Putnam FW (1979)            165. Liu YS, Low TL, Infante A, Putnam FW
     Structure of the A2m(1) allotype of human              (1976) Complete covalent structure of a
     IgA. A recombinant molecule. Proc Natl Acad            human IgA1 immunoglobulin. Science
     Sci USA 76:1104–1108                                   193(4257):1017–1020
                                                                                  INDEX
Frank T. Christiansen and Brian D. Tait (eds.), Immunogenetics: Methods and Applications in Clinical Practice,
Methods in Molecular Biology, vol. 882, DOI 10.1007/978-1-61779-842-9, © Springer Science+Business Media New York 2012
                                                                                        681
    IMMUNOGENETICS: METHODS AND APPLICATIONS IN CLINICAL PRACTICE
682 Index
GeneRuler Express (Fermentas™) .......................... 131, 139                   Heterozygous ambiguity resolving primers
Genetic analysis in Excel (GenAlEx) ..................... 218, 219,                             (HARPs)..............................78, 94–95, 105, 107,
           233, 234, 240                                                                        109, 112, 113, 115–118
Genetic polymorphism .................................... 224, 550–552               HLA. See Human leukocyte antigen (HLA)
Genome Diagnostics® (BV, Utrecht,                                                    HLA dictionary ........................................532, 538, 544, 545
           Netherlands) .................................. 156, 520, 523             HLA disease associations
Genotyping.............................2, 3, 10, 11, 13, 14, 28, 30, 32,                ankylosing spondylitis .................................................. 29
           42, 49–53, 56, 58, 75, 84, 88, 113–115, 121, 133,                            coeliac disease .............................................................. 29
           137, 144, 162, 164, 170, 173–181, 183–195, 199,                              drug hypersensitivity-Abacavir, Carbamazepine.......... 29
           212, 217, 220, 221, 248, 261, 399–401, 415–418,                              narcolepsy .................................................................... 29
           472, 478, 493, 497, 519, 532, 550, 554, 557, 558,                            rheumatoid arthritis ..................................................... 29
           563, 564, 566, 638                                                           type 1 diabetes ............................................................. 29
Gen-Probe® LIFECODES HLA-SSO™ ............ 50, 56–60                                 HLA Explorer software................................................... 543
Gen-Probe® LIFEMATCH Quick-Type™ ............... 59–60                               HLA Fusion™ (One Lambda).................................... 55–56
Germline................................. 392, 581, 583–585, 591, 593,               HLA Librarian™ software (Conexio Genomics,
           599–601, 606, 609, 613, 630                                                          Applecross, WA, Aust) .................................. 452
Gm allotypes ........................................................... 636–661     HLA microspheres ............................................................ 48
Gm-Am haplotypes................................................. 636, 638           Homozygosity
GMP. See Good manufacturing practice (GMP)                                              normalized deviate of homozygosity ................. 229–230
Good manufacturing practice (GMP) ..................... 495–497                      HPC. See Hematopoietic progenitor cell (HPC)
GPromo.R® ............................................................. 132, 137     HUGO. See Human Genome Organization (HUGO)
GPromo.S® ............................................................. 132, 137     Human Genome Organization (HUGO) ....................... 600
Graft versus host disease (GvHD) ...................183, 309, 310,                   Human genomes project.................................................. 178
           407–409, 469–471, 478, 493, 495, 504, 511, 512, 550                       Human leukocyte antigen (HLA)
Graft versus leukemia (GvL) ........................... 478, 511, 512                   compatibility ..............................................267–268, 270,
Granulocyte colony stimulating factor                                                           271, 472
           (G-CSF) ................................................ 471, 491            HLA-A ..............................2, 3, 9, 11, 61, 72, 75, 76, 78,
Granzymes ...................................................... 331, 392, 479                  143, 204, 268–271, 290, 303, 326, 393–394, 397,
GraphPad Prism® software ............................................. 248                      416, 472, 478, 480, 511, 512, 524, 525, 531, 533,
GvHD. See Graft versus host disease (GVHD)                                                      535, 536, 543, 545, 617
GvL. See Graft versus leukemia (GvL)                                                    HLA antibodies................. 267–276, 289–306, 359–360,
                                                                                                365–368, 371–372, 374, 379–381, 385, 386, 389
H                                                                                           clinical importance................................... 14–15, 302
HaploPrep™ kit (Qiagen, Valencia, CA, USA)............... 442                               detection and characterization ..................... 289–306
Haplo.stats software ........................................................ 224           donor specific (DSA) .................................. 272, 359,
Haplotypes                                                                                      366–368, 379, 380, 386, 389
   haploidentical ............................................................ 472      HLA-B.................................... 11, 29, 31–33, 35, 38, 44,
   haplo.stats package .................................................... 224                 72, 75, 76, 78, 119, 183, 238, 269, 326, 340, 397,
   rare estimated haplotypes .......................................... 225                     405, 416, 470, 472, 477–478, 480, 511, 524, 525,
Hardy Weinberg equilibrium (HWE)                                                                531, 539, 541, 543–545
   exact test for HWEP ......................................... 222, 223               HLA-C ........................70, 72, 75, 76, 78, 143, 204, 261,
   HWE proportions (HWEP) ............................. 220, 261                                262, 396, 397, 406, 416, 470, 472, 474, 475, 477,
HARPs. See Heterozygous ambiguity resolving primers                                             478, 480, 531, 534, 537, 539, 541, 543, 544
           (HARPs)                                                                      HLA-DP................................................... 531, 543–544
Helmberg sequence compilation and rearrangement                                         HLA-DQ ...............................................43–44, 299, 531
           evaluation (SCORE) ..................................... 206                 HLA-DR ................................... 9, 10, 28, 269–270, 531
Hemagglutination inhibition                                                             HLA-E ......................................................... 3, 143–157
           methodology .................................. 638, 670, 672                 HLA-G ..................................................... 123–140, 397
Hematopoietic progenitor cell                                                           isotypes
           (HPC)..................... 432, 471, 531–532, 535, 539                           IgA .............................................................. 271–272
Heterologous immunity................................................... 339                IgG ..............................................271–272, 326, 375,
Heterozygosity                                                                                  380, 385, 481
   HARPs ............................ 78, 94–95, 105, 107, 109, 112,                        IgM.............................................................. 271–272
           113, 115–118                                                              Human recombinant Interleukin-2 (IL-2) ...................... 314
                                                              IMMUNOGENETICS: METHODS AND APPLICATIONS IN CLINICAL PRACTICE
                                                                                                                    Index
                                                                                                                            685
HWE. See Hardy Weinberg equilibrium (HWE)                                              Internal positive control (IPC)........................ 296, 300, 399,
Hybridization .................................. 32, 41, 45, 58, 146, 151,                         418, 420, 423, 424, 426, 427, 564
          152, 426, 456, 457                                                           International HapMap Project ........................................ 178
Hydropathy index .................................................... 588, 625         Intracellular cytokine staining (ICS) ...................... 312, 313,
Hyperacute rejection........................................ 272–273, 379                          316–319, 326–328
                                                                                       Invitrogen™ .............................. 74, 130, 131, 137, 145, 314,
I                                                                                                  318, 334, 341, 352, 380, 384, 423, 435, 436, 442,
                                                                                                   443, 463, 472, 480, 514, 519, 521
ICS. See Intracellular cytokine staining (ICS)
                                                                                       IPC. See Internal positive control (IPC)
IgSF. See Immunoglobulin superfamily (IgSF)
                                                                                       IPD-KIR database........................... 402, 404, 441, 452, 461,
IMGT/HLA database ............................14, 88, 95, 143, 157
                                                                                                   462, 467
Immunogenes ..................................124, 197–212, 215–241,
                                                                                       Isoallotypes. ............................. 637–642, 644, 645, 647, 651,
            245–263, 510–511
                                                                                                   654, 656, 660, 667, 672
Immunogenetics information system®
                                                                                       Isoamyl alcohol ................................................ 442, 458, 463
    IMGT/Automat ........................................ 574, 587, 596
    IMGT/Colliers de Perles...........................574, 586, 602,                   J
            606, 610, 612, 624–630, 632, 642, 667
                                                                                       J-region...... .............................. 574–576, 578, 581, 585, 586,
    IMGT/DomainDisplay ..................................... 642, 667
                                                                                                    596, 598, 600, 607, 609, 612, 613, 619, 624, 630
    IMGT/DomainGapAlign .........................605–632, 636,
                                                                                       Junction analysis ......................................570, 572, 574–581,
            642, 643
                                                                                                    587–588, 590, 598, 601
    IMGT/3D structure-DB...................613, 615, 629, 636,
            644–648, 671, 672                                                          K
    IMGT/GENE-DB ...........................571, 600, 606, 636,
            642, 643, 667, 672                                                         Keratinocytes ................................................................... 511
    IMGT/HighV-QUEST .................................... 569–602                      Killer cell immunoglobulin like receptors (KIR)
    IMGT/JunctionAnalysis ...........................570, 572, 574,                        genes.... .......................202, 225, 226, 246, 261, 394–396,
            575, 577, 578, 580, 581, 587–588, 590, 598, 601                                         398–404, 407–408, 415–417, 425, 426, 431, 432,
    IMGT/Kaleidoscope ......................................... 570, 606                            434, 435, 443, 453, 462, 464, 466, 467, 470, 472,
    IMGT/LIGM-DB database .....................576, 579, 580,                                       474, 492
            582, 584, 586–589, 593                                                         ligands........................ 246, 261, 262, 394–397, 405–408,
    IMGT/PhyloGene .................................... 585, 594, 595                               415, 416, 470, 474, 492
    IMGT Repertoire Web resources .............................. 636                       polymorphism............................ 202, 216, 224, 397, 402,
    IMGT/Scientific chart ......................630, 642, 644, 651,                                 403, 416, 426, 435
            656, 659, 668, 671                                                             typing..........................207, 208, 212, 225, 261, 401, 416,
    IMGT/V-QUEST .................................... 569–602, 610                                  419, 422, 423, 425–429, 431–467, 472, 474, 497
    Immunogenomics data analysis working group                                         KIR. See Killer cell immunoglobulin like receptors (KIR)
            (IDAWG) .............................................. 204, 206            KIR haplotypes
Immunoglobulin                                                                             centromeric ......................... 261, 399–401, 406, 425, 492
    heavy chain ........................................................ 635, 641          gene content ...................................................... 398–401
    IgG........................................... . 271–272, 279, 280, 284,               telomeric .................................................... 401, 408, 492
            287, 292–294, 296, 315, 324–326, 341, 361, 369,                            Km allotypes ............................. 635, 637, 640, 667, 669, 670
            370, 372, 374–377, 379–382, 385, 386, 388, 472,
                                                                                       L
            473, 481, 536, 636–638, 642, 648, 650, 651, 660,
            670, 672                                                                   LabScan™100 Flow Analyser ..................................... 54, 59
    IgM.................................... 271–272, 296, 341, 359–361,                LABType® SSO .................................................... 48–56, 61
            366, 369–372, 375–377, 379                                                 Lasergene™ software programme (DNASTAR, Madison
    light chain .................................................. 635, 665, 667                 USA).............................................. 153, 155, 157
Immunoglobulin superfamily (IgSF) ....................... 605–632                      LCL. See Lymphoblastic cell lines (LCL)
Immunoinformatics ......................................................... 570        LD. See Linkage disequilibrium (LD)
Immunotherapy ........................................416, 510, 512, 513               LDA. See Limiting dilution assay (LDA)
Indels (insertions/deletions) ............................2, 4, 103–107,               Leukapheresis ...........................................471, 495, 497, 498
            111, 128, 129, 202, 206                                                    Lifecodes™............................................ ................30, 48, 50,
Influenza virus ......................................................... 310, 318               56–61, 64
Innate immunity .............................................................. 392     Life Technologies™ .............................................71, 74, 314,
Intergenic regions ................................................................5             316–318, 331
    IMMUNOGENETICS: METHODS AND APPLICATIONS IN CLINICAL PRACTICE
686 Index
Restriction enzyme ................... 433, 442–443, 457–458, 665                Side scatter (SSC) .................... 329, 383, 386, 426, 484, 485
Restriction fragment length polymorphism                                         Single nucleotide polymorphisms (SNPs) .................. 2, 4, 5,
           (RFLP) ............... 46–47, 144, 161, 637, 639, 665                              30–32, 42–45, 124–129, 134–136, 173–181, 202,
RFLP. See Restriction fragment length polymorphism                                            206, 207, 210, 224, 225, 253, 256, 257, 523,
           (RFLP)                                                                             550–552, 554, 559, 560, 562
RoboSep® automated magnetic cell separation                                      Single strand conformational polymorphism
           (stem cell technologies).................................. 361                     (SSCP)................................................... 184, 426
Rodgers (Rg) determinant ............................................... 161     Single transfected K562 cells (SALs) ...................... 339–349
Rosette sep® (stem cell technologies) ..................... 333, 480,            SLE. See Systemic lupus erythematosus (SLE)
           482, 487                                                              Smith-Waterman score............................................ 612, 631
ROX™ dye. ..................................................... 176, 178, 179    SNPs. See Single nucleotide polymorphisms (SNPs)
RPE. See Relative predispositional effects (RPE)                                 Sodium azide ............................................294, 381, 386, 481
R-phycoerythrin-conjugated streptavidin                                          Solid phase immunoassay .......................30, 41–42, 289–306
           (SAPE) ....................... .32, 40, 48–50, 53–55, 58,             Somatic hypermutations ...................570, 575, 581, 584–585
           59, 62, 64, 146, 152, 156, 157                                        Southern blotting ............................................................ 161
RPMI cell culture medium ......................314–317, 319, 320,                Spleen........ ...............................................351–357, 428, 491
           322, 333, 348, 352–354, 356, 361, 368, 380–382,                       SSP. See Sequence specific priming (SSP)
           480–482, 487                                                          STATA software package ................................................ 257
RR. See Relative risk (RR)                                                       Statistical analysis system (SAS®) ............197, 218, 248, 257
RSCA. See Reference strand conformational analysis (RSCA)                        Statistical package for social sciences (SPSS®) ........ 248, 257
RunModule Rapidseq™ (Applied Biosystems) ............... 461                     Streptavidin
RunOne™ electrophoresis unit (Embi Tee, San Diego,                                   Streptavidin-horseradish peroxidase (HRP) ................ 19
           CA,USA) ....................................................... 436   SYBR Safe™ (Invitrogen) ............................... 130, 134, 423
                                                                                 Systemic lupus erythematosus (SLE) ...................... 160, 161
S
                                                                                 T
Saponin (Sigma) .............................................. 317, 328, 334
SBT. See Sequence based typing (SBT)                                             TAMRA (6-carboxy-tetramethyl-rhodamine).. ............. 162,
SBTEngine® software for allele assignment (genome                                            174, 176, 177
           diagnostics) .................................... 520, 522, 523       Taqman® chemistry
Sephadex G-50 (GE healthcare) ..................... 514, 521, 522                   copy number assay ...................... 162, 164, 167, 168, 170
Sequence based typing (SBT)..........................14, 23, 28, 29,                copy number reference RNase P ................................ 167
           31–39, 45, 67, 70, 71, 74, 75, 81, 84, 87–121, 129,                      genotyping master mix ...................32, 42, 162, 164, 170
           144, 146–148, 153, 157, 183–195, 212, 452, 456,                       T cell receptor (TCR)...................... 309, 339, 340, 343, 348,
           462, 514, 526                                                                     396, 513, 540, 569–602, 605
Sequence electropherograms ............................... 84, 89–106            TCR. See T cell receptor (TCR)
Sequence specific oligonucleotide (SSO) assay                                    TDT. See Transmission disequilibrium test (TDT)
   biotinylated oligonucleotide probes. ................13, 16, 29,              TdT. See Terminal deoxynucleotidyl transferase (TdT)
           30, 44, 48, 130, 150, 325                                             Tepnel®
   DELFIA hybridization ............................. 30, 32, 41–42                 beads.... ...................................................................... 287
   hybridization buffer ..............................41, 146, 456, 457          Terasaki plates ................................................. 361, 363, 371
   microbead hybridization typing .................281–283, 388,                 Terminal deoxynucleotidyl transferase (TdT) ........ 259–260,
           496, 497, 499                                                                     570, 578
   reverse SSO ..................................................... 13–19, 48   Thermal sealing film (ISC BioExpress)........................... 185
   wash buffer .........................24, 32, 40, 41, 49, 52–54, 62,           Thermocycler............................... 9, 15–18, 21, 40, 144, 146,
           130, 146, 152, 156, 280, 292, 293, 386                                            148–152, 154, 155, 185, 187–190, 422
Sequence specific priming (SSP) .......................9–24, 28, 29,             Thermowell® sealing tape ....................................... 281–282
           31–34, 44, 45, 47–48, 84, 87, 144, 161, 184, 212,                     Thymoglobulin ................................................................ 297
           415–429, 432, 464, 514, 553                                           TOPO TA Cloning® kit (Invitrogen) ..................... 443, 459
Sequencher™ sequence software (Ann Arbor,                                        TrackItTM DNA ladder (Invitrogen) ................................... 74
           MI, USA) .............................................. 452, 463      Transfection .......290, 319, 323, 326, 327, 331, 332, 339–349
Sequencing primer. ........................ 36–38, 69, 70, 73, 78, 83,           Transmission disequilibrium test (TDT)......... 259–260, 551
           84, 89, 97, 105, 116, 118–119, 129, 131, 139, 147,                    Transplantation
           155, 157, 184, 186, 188, 189, 193, 195, 432, 443,                        haematopoietic stem cell (HSCT) ...............14, 407, 408,
           444, 460, 463, 464, 466, 521                                                      469, 471, 478, 483, 486
                                                             IMMUNOGENETICS: METHODS AND APPLICATIONS IN CLINICAL PRACTICE
                                                                                                                   Index
                                                                                                                           689
   heart................................................................... 273–276   Vic® dye....................................................162, 165, 174, 175
   kidney.......................... 184, 267–271, 273–276, 379–389                    Viral immunity ................................................................ 312
   liver........................................................... . 273–276, 407    Vogt-Koyanagi-Harda (VKH) disease .................... 406–407
   pretransplantation ...............................268, 272, 274, 275               V-region...................................................574–578, 580–588,
Transversions ............................ 583, 599, 607, 631, 650, 670                           591–593, 596, 598, 599, 601, 602, 609, 612, 613,
Treponema pallidum .......................................................... 503                 619, 621, 630
Trypan blue ..............................................314, 320, 481, 487
                                                                                      W
U
                                                                                      Whatman® 96 well plate ................................................. 281
Ultraviolet (UV) transilluminator..........................16, 22, 52,                World Health Organization International Non proprietary
            58, 130, 131, 134, 135, 138, 146, 427, 436, 555                                    name (INN) programme................................ 601
Untranslated region ......................................... 124, 441, 554           World Health Organization-International Union of
                                                                                               Immunological Societies
V                                                                                              (WHO-IUS) ......................................... 600, 641
Variable domain....................................................... 570, 643
                                                                                      Y
V-domain................................. 570, 572, 577, 601, 606–607,
           609–610, 612, 613, 619, 621, 624, 625, 630–632                             Yates’ correction ........................................... 27–28, 248–249