0% found this document useful (0 votes)
4 views14 pages

Lim 2014

Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
4 views14 pages

Lim 2014

Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 14

Arch Toxicol (2014) 88:1773–1786

DOI 10.1007/s00204-014-1338-z

Review Article

Antioxidative defense mechanisms controlled by Nrf2:


state‑of‑the‑art and clinical perspectives in neurodegenerative
diseases
Jamie L. Lim · Micha M. M. Wilhelmus ·
Helga E. de Vries · Benjamin Drukarch ·
Jeroen J. M. Hoozemans · Jack van Horssen

Received: 24 June 2014 / Accepted: 12 August 2014 / Published online: 28 August 2014
© Springer-Verlag Berlin Heidelberg 2014

Abstract Activation of microglial cells and impaired disorders, only a few Nrf2-activating compounds have
mitochondrial function are common pathological charac- been tested in a clinical setting. We here provide a com-
teristics of many neurological diseases and contribute to prehensive synopsis on the role of ROS in common neu-
increased generation of reactive oxygen species (ROS). rodegenerative disorders and discuss the therapeutic
It is nowadays accepted that oxidative damage and mito- potential of the Nrf2 pathway.
chondrial dysfunction are key hallmarks of classical neu-
roinflammatory and neurodegenerative diseases, such Keywords Reactive oxygen species · Oxidative stress ·
as multiple sclerosis, Alzheimer’s disease, Parkinson’s Neurodegenerative disorders · Nrf2 · Antioxidant enzymes ·
disease and Huntington’s disease. To counteract the det- Free radicals
rimental effects of ROS and restore the delicate redox
balance in the central nervous system (CNS), cells are Abbreviations
equipped with an endogenous antioxidant defense mecha- Aβ  β Amyloid
nism consisting of several antioxidant enzymes. The pro- AD Alzheimer’s disease
duction of many antioxidant enzymes is regulated at the ARE Antioxidant response elements
transcriptional level by the transcription factor nuclear BBB Blood–brain barrier
factor E2-related factor 2 (Nrf2). Although evidence is CNS Central nervous system
accumulating that activation of the Nrf2 pathway rep- CSF Cerebrospinal fluid
resents a promising therapeutic approach to restore the DMF Dimethyl fumarate
CNS redox balance by reducing ROS-mediated neuronal EAE Experimental autoimmune encephalomyelitis
damage in experimental models of neurodegenerative HD Huntington’s disease
HTT Huntingtin
iNOS Inducible nitric oxide synthase
J. L. Lim · H. E. de Vries · J. van Horssen (*)
Keap1 Kelch-like ECH-associated protein 1
Department of Molecular Cell Biology and Immunology,
Neuroscience Campus Amsterdam, VU University Medical MMF Monomethyl fumarate
Center Amsterdam, P.O. Box 7057, 1007 MB Amsterdam, MPTP 1-Methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine
The Netherlands MS Multiple sclerosis
e-mail: [email protected]
mtDNA Mitochondrial DNA
J. L. Lim NADPH Nicotinamide adenine dinucleotide phosphate
e-mail: [email protected]
NOX Nicotinamide adenine dinucleotide phosphate
M. M. M. Wilhelmus · B. Drukarch oxidase
Anatomy and Neurosciences, Neuroscience Campus Amsterdam, Nrf2 Nuclear factor E2-related factor 2
VU University Medical Center, Amsterdam, The Netherlands 6-OHDA 6-hydroxydopamine
OxPhos Oxidative phosphorylation
J. J. M. Hoozemans
Pathology, Neuroscience Campus Amsterdam, VU University PERK Protein kinase RNA-like endoplasmic
Medical Center, Amsterdam, The Netherlands reticulum kinase

13
1774 Arch Toxicol (2014) 88:1773–1786

PD Parkinson’s disease (Wagener et al. 2003). Heme oxygenases are localized to dif-
ROS Reactive oxygen species ferent cellular compartments, such as the endoplasmic reticu-
SN Substantia nigra lum, nucleus and plasma membrane (Ryter et al. 2006).
tBHQ Tert-butylhydroquinone ROS levels need to be tightly regulated in the central
nervous system (CNS) as it is well known that the brain is
particularly sensitive to oxidative stress. The CNS is char-
Introduction acterized by high oxygen utilization and relatively high
amounts of trace metals, including iron and copper, which
Reactive oxygen species (ROS) are small biological mol- together with hydrogen peroxide give rise to highly reac-
ecules that contain oxygen and one or more unpaired elec- tive hydroxyl radicals, one of the most potent oxidants in
trons. Aerobic organisms continuously produce ROS as a nature. In addition, the CNS is enriched in polyunsaturated
natural by-product of oxygen metabolism. ROS perform fatty acids, making it vulnerable to lipid peroxidation.
diverse physiological functions and play an important role in When ROS levels are dramatically increased in the CNS,
host defense, cell signaling, regulation of gene expression and e.g., under pathological conditions, the cellular antioxidant
cell differentiation (Bedard and Krause 2007). ROS-synthe- capacity is overwhelmed, resulting in oxidative stress and
sizing enzyme systems, including myeloperoxidase, xanthine significant oxidative damage to essential cellular structures.
oxidase and nicotinamide adenine dinucleotide phosphate ROS react rapidly with biological molecules, and various
(NADPH) oxidase (NOX), contribute to ROS production. oxidative markers have been described to study and quantify
In addition, mitochondria, the energy-producing organelles the occurrence of oxidative injury. Protein carbonyl groups and
of the cell, form ROS as a by-product of cellular respiration, 3-nitrotyrosine are biochemical markers used for the detection
particularly at complex I and III of the mitochondrial electron of oxidative modification of amino acid residues in proteins
transport chain. Superoxide, the most common ROS product (Beal 2002). Likewise, 8-hydroxydeoxyguanosine adducts are
of both mitochondrial respiration and NOX, is rapidly con- used as a footprint for oxidative damage to nucleotides (Evans
verted, either spontaneously or enzymatically, by superoxide et al. 2004; Kasai 2002). Mitochondrial DNA (mtDNA) is
dismutases. These metal-containing enzymes, including Cu, highly susceptible to ROS-induced damage since it is located
Zn-superoxide dismutase-1 (McCord and Fridovich 1969), in close vicinity to the initial production site of mitochondria-
Mn-superoxide dismutase-2 (McCord 1976) and extracellu- derived ROS, and mtDNA repair mechanisms are limited. Con-
lar Cu,Zn-superoxide dismutase-3 (Marklund 1982), provide sequently, mutations accrue in mtDNA, ultimately leading to a
the first line of defense against oxidative stress and catalyze decline in mitochondrial function and concomitant enhanced
the dismutation of superoxide anion to molecular oxygen and ROS production (Lin and Beal 2006). Reaction of free radi-
hydrogen peroxide (Johnson and Giulivi 2005). cals with unsaturated lipids may trigger the lipid peroxidation
Hydrogen peroxide itself is not a free radical, but it can eas- chain reaction resulting in the oxidative breakdown of cellular
ily diffuse across biological membranes and may cause severe membranes. Lipid peroxidation is detected using various bio-
oxidative damage to biological macromolecules. However, markers, such as malondialdehyde, 4-hydroxy-2-nonenal or F2
under normal conditions, hydrogen peroxide is effectively isoprostane (Mariani et al. 2005; Morrow et al. 1999). Markers
detoxified by endogenous antioxidant enzymes, like catalase, for oxidative damage can be measured in plasma, cerebrospinal
glutathione peroxidases and peroxiredoxins. Catalase is an fluid (CSF), urine or postmortem tissue by means of high-per-
intracellular antioxidant enzyme that catalyzes the conversion formance liquid chromatography and immunohistochemistry.
of hydrogen peroxide into water and molecular oxygen. It is As oxidative injury is a hallmark of several neurodegen-
located in the peroxisomes and is ubiquitously expressed in a erative diseases, antioxidant therapies have been proposed
wide variety of cells. The family of peroxiredoxins represents to prevent ROS-induced injury. Unfortunately, most studies
antioxidant proteins that are involved in the enzymatic degra- reported disappointing results upon investigating the thera-
dation of hydrogen peroxide, organic hydroperoxides and per- peutic potential of exogenous antioxidants (Moosmann and
oxynitrite (Rhee et al. 2005). Peroxiredoxins are abundantly Behl 2002), indicating the need for alternative strategies to
expressed in the cytosol; however, several isoforms are local- therapeutically counteract the damaging effects of ROS and
ized to mitochondria, peroxisomes, nuclei and membranes restore the cellular redox balance under neurodegenerative
(Hofmann et al. 2002). The glutathione system represents an conditions. To maintain a physiological redox balance, cells
important component of the non-enzymatic branch of ROS are endowed with a selection of endogenous antioxidant
detoxification. Glutathione is able to directly neutralize ROS enzymes. Transcription of these cytoprotective proteins is
or indirectly via glutathione peroxidase. Although heme oxy- under control of the nuclear transcription factor NF-E2-re-
genase-1 and oxygenase-2 are not able to detoxify free radicals lated factor 2 (Nrf2), which plays a central role in the regu-
directly, they protect cells from oxidative stress by converting lation of the cellular redox status (Itoh et al. 2003; Nguyen
toxic heme into the potent antioxidants biliverdin and bilirubin et al. 2003, 2004). Under homeostatic conditions, Nrf2

13
Arch Toxicol (2014) 88:1773–1786 1775

transcription is repressed by its negative regulator Kelch- lesions revealed a striking increase in genes and proteins
like ECH-associated protein 1 (Keap1) (Itoh et al. 2003), involved in ROS production. Particularly, NADPH oxidase
but upon exposure to ROS, Nrf2 dissociates from cytosolic subunits were profoundly upregulated in microglia and
Keap1 and translocates to the nucleus where it binds to anti- macrophages in active MS lesions, indicating the impor-
oxidant response elements (ARE) in the promoter region of tance of oxidative burst by activated microglia and infil-
hundreds of genes involved in antioxidant protection and trated macrophages. In addition, enhanced expression of
detoxification, including SODs (McCord and Edeas 2005), inducible nitric oxide synthase (Bo et al. 1994; Liu et al.
glutathione peroxidases (Brigelius-Flohe and Maiorino 2001; De Groot et al. 1997) and myeloperoxidase (Marik
2013), peroxiredoxins (Dringen et al. 2005; Kim et al. 2007), et al. 2007; Gray et al. 2008a, b; Chen et al. 2008) has
heme oxygenases (Ishii et al. 2000; Wagener et al. 2003) and been detected in the brains of patients with MS. Besides
NAD(P)H:quinone oxidoreductase-1 (Li and Jaiswal 1992; enhanced expression of enzymes involved in ROS produc-
Long and Jaiswal 2000; Iskander et al. 2006; Jaiswal 2000). tion, MS brain tissue is characterized by abundant mito-
Together, these free radical scavenging enzymes repre- chondrial defects, which represents another pathogenic
sent a powerful antioxidant defense mechanism. Nowadays, process contributing to oxidative stress. Oxidative damage
various natural and synthetic Nrf2-activating compounds to mtDNA in MS lesions was associated with decreased
have been identified, and the number of reports describ- activity of complex I of the oxidative phosphorylation
ing the beneficial effects of Nrf2 activation in experimental (OxPhos) chain in MS lesions (Lu et al. 2000), whereas
models of neurodegeneration is steadily increasing. Even complex IV activity was significantly increased (Mahad
more excitingly, some of these compounds have entered et al. 2009; Witte et al. 2009). Free radicals produced by
into clinical trials or are already on the market. Here, we activated microglia and macrophages through oxidative
will discuss the current literature on the role of ROS- burst likely contribute to intra-axonal mitochondrial injury
induced injury in neurodegenerative disorders, including by inhibiting OxPhos (Witte et al. 2010; Bolanos et al.
multiple sclerosis, Alzheimer’s disease, Parkinson’s disease 1997; Higgins et al. 2010). In animals with experimental
and Huntington’s disease. Moreover, we will provide an autoimmune encephalomyelitis (EAE), the most common
overview on protective effects of Nrf2-induced antioxidant MS animal model, antioxidant administration, attenuated
protection in experimental models of neurodegeneration. mitochondrial pathology and concomitant axonal degenera-
tion (Nikic et al. 2011). Nitration of mitochondrial proteins
and mitochondrial changes even preceded infiltration of
Reactive oxygen species in multiple sclerosis leukocytes in animals suffering from EAE indicating that
mitochondrial defects occur in the early stages of neuroin-
Multiple sclerosis (MS) is a chronic inflammatory and flammation (Qi et al. 2006). Mitochondrial defects are not
demyelinating disease of the CNS. Patients may suf- restricted to MS white matter lesions, but can be observed
fer from a wide variety of clinical symptoms, including throughout the gray matter. Accumulation of neuronal
visual, motor and cognitive disturbances. The course of mtDNA deletions and decreased neuronal levels of key
the disease is generally episodic, with frequent intervals mitochondrial proteins, including specific OxPhos proteins
of exacerbations followed by periods of remission, known and mitochondrial antioxidant enzymes, have been reported
as relapsing–remitting MS. In time, most patients gradu- (Campbell et al. 2011). Taken together, increased expres-
ally develop secondary progressive MS, a stage character- sion of ROS-generating enzymes by inflammatory cells and
ized by progressive permanent neurological deficits. The mitochondrial dysfunction results in enhanced ROS forma-
immune system is particularly involved in the initial stage tion during neuroinflammation.
of the disease as active demyelinating MS lesions are char- It is now well accepted that ROS play a key role in
acterized by massive microglial cell activation and leuko- pathogenic processes involved in MS lesion development
cyte infiltrates consisting of lymphocytes and macrophages. and progression. ROS induce blood–brain barrier (BBB)
Additional pathological hallmarks of MS plaques include permeability, thereby promoting leukocyte infiltration and
destruction of myelin sheaths, oligodendrocyte cell death, lesion formation. In the early phase of MS lesion forma-
neuro-axonal damage and glial scar formation. During an tion, locally produced ROS induce BBB breakdown,
inflammatory attack, macrophages and activated microglia thereby facilitating transendothelial leukocyte migration
produce a plethora of inflammatory mediators, including (Van der Goes et al. 2001; Schreibelt et al. 2006). Sec-
cytokines, chemokines, nitric oxide and ROS. ondly, ROS cause oxidative damage to CNS cells, resulting
To identify possible sources of ROS production in rela- in oligodendrocyte injury, demyelination and axonal dam-
tion to demyelination and neurodegeneration in MS, a age. Increased levels of indicators of oxidative stress and/or
genome-wide microarray analysis has been performed decreased levels of antioxidant enzymes and antioxidants
(Fischer et al. 2012). Analysis of active demyelinating MS have been observed in blood and CSF during the active

13
1776 Arch Toxicol (2014) 88:1773–1786

phases of MS (Karg et al. 1999; Greco et al. 1999; Cala- shown to be markedly increased in active demyelinating
brese et al. 1994; Ferretti et al. 2005; Koch et al. 2006). MS lesions, suggesting activation of the Nrf2 pathway dur-
Additionally, enhanced expression of markers of oxidative ing a neuroinflammatory attack (van Horssen et al. 2008,
damage in MS brain tissue has been reported, indicating the 2010). Immunohistochemical analysis revealed that Nrf2
occurrence of widespread oxidative injury in demyelinating and its downstream targets heme oxygenase-1 and NAD(P)
MS plaques. Immunohistochemical detection of nitrotyros- H:quinone oxidoreductase-1 were upregulated in inflamma-
ine, a footprint for peroxynitrite formation, demonstrated tory MS lesions, particularly in hypertrophic astrocytes and
extensive nitrotyrosine staining in early MS lesions. Addi- myelin-laden macrophages. Intriguingly, oligodendrocytes
tionally, markers for lipid peroxidation, such as malondial- and neurons, the primary cellular victims in MS, expressed
dehyde and 4-hydroxy-2-nonenal, and oxidative damage to relatively low levels of Nrf2 and antioxidant enzymes.
nucleotides are increased in MS lesions (van Horssen et al. Despite the increase in endogenous antioxidant enzymes in
2008; Haider et al. 2011). Oligodendrocytes, in particular active demyelinating MS lesions, oxidative stress and dam-
oligodendrocyte precursor cells, are highly susceptible to age are still prominent in inflammatory lesions. This sug-
oxidative damage as they contain relatively low levels of gests that the observed increase in antioxidant enzymes is
endogenous antioxidant enzymes compared to astrocytes not sufficient to counteract the increased ROS production
(Mitrovic et al. 1994; Thorburne and Juurlink 1996; Husain in MS brain tissue. Hence, additional activation of the Nrf2
and Juurlink 1995; Juurlink et al. 1998). Moreover, oli- pathway may be a potential therapeutic strategy to restore
godendrocytes contain large myelin sheets, which mainly the redox balance in MS.
consist of polyunsaturated fatty acids that can react with Johnson and co-workers elegantly showed the protec-
ROS, thereby triggering lipid peroxidation. It has been tive role of Nrf2 in the EAE animal model. They showed
demonstrated that free radicals are able to block matura- that induction of EAE in Nrf2-deficient mice resulted in
tion of oligodendrocyte precursor cells into myelin-forming more severe disease symptoms and an increase in infil-
cells by downregulation of genes involved in differentia- trated immune cells (Johnson et al. 2010). Sulforaphane
tion and upregulation of genes associated with inhibition of and derivatives of the natural triterpene oleanolic acid, both
maturation (French et al. 2009). well-known Nrf2 activators, suppressed disease in a murine
model of MS by inhibiting the immune system and induc-
ing antioxidant genes (Pareek et al. 2011; Li et al. 2013). In
Nrf2 in multiple sclerosis addition, there is ample evidence that fumaric acid esters
are able to boost the production of endogenous antioxidant
Based on the prominent role of ROS in the pathogenesis enzymes via activation of the Nrf2 pathway (Scannevin
of MS, it is conceivable that stimulating antioxidant pro- et al. 2012). Scannevin and colleagues demonstrated that
duction may be beneficial and can protect CNS cells from administration of dimethyl fumarate (DMF) or monomethyl
ROS-induced damage, thereby limiting disease progres- fumarate (MMF) prior to an oxidative challenge was able
sion. Administration of compounds with antioxidant prop- to prevent ROS-induced cell death of primary astrocytes
erties was shown to ameliorate clinical manifestations and and neurons in an Nrf2-dependent manner. Oral adminis-
inflammation in the EAE animal model (Mao et al. 2013; tration of fumarates attenuated disability progression in a
Hendriks et al. 2004). In contrast, other reports found no chronic EAE mouse model, and more recently, it was dem-
evident protective effect of exogenous antioxidants (Fie- onstrated that preventive DMF treatment reduced the clini-
biger et al. 2013; Spitsin et al. 2002). A possible explana- cal signs of disability in a chronic EAE mouse model and
tion for the lack of therapeutic efficacy is that most exog- that the beneficial effects were Nrf2-dependent, as DMF
enous antioxidants are hydrophilic and therefore not able administration did not affect the disease course in Nrf2-
to efficiently cross the BBB (Gilgun-Sherki et al. 2001; deficient animals. DMF treatment resulted in induction of
Moosmann and Behl 2002; Carlson and Rose 2006). Nrf2 expression in neurons, oligodendrocytes and astro-
Unfortunately, clinical data on successful antioxidant ther- cytes, preservation of myelin and axonal structures and a
apy in MS patients are very limited, emphasizing the need marked decrease in astrogliosis (Linker et al. 2011). Fuma-
for further epidemiological and clinical studies. Another rates have been used in a clinical setting for more than half
therapeutic option would be to activate Nrf2-driven antioxi- a century in the treatment of psoriasis, a chronic skin disor-
dant gene expression in the CNS, thereby limiting oxida- der. Based on the positive results in the treatment of psoria-
tive stress and oxidative cellular injury in MS. sis, Schimrigk et al. performed the first exploratory clinical
Nrf2-regulated proteins are upregulated in brain sam- trial with fumarates in a small cohort of relapsing–remit-
ples of EAE animals, particularly at the peak of disease ting MS patients (Schimrigk et al. 2006). Fumarate treat-
(van Horssen et al. 2011). Likewise, the expression of ment decreased the mean number of gadolinium-enhancing
Nrf2 as well as Nrf2-mediated antioxidant proteins was lesions, an indication of active inflammation, significantly.

13
Arch Toxicol (2014) 88:1773–1786 1777

More recently, the potential protective effects of Tecfidera®, 4-hydroxy-2-nonenal levels were shown to be significantly
enteric-coated microtablets containing DMF, have been elevated in the ventricular fluid of AD subjects compared
investigated in two large randomized, double-blind, pla- to control subjects, demonstrating increased lipid peroxida-
cebo-controlled Phase III studies. In these trials, relapsing– tion in AD brain (Lovell et al. 1997).
remitting MS patients were randomly assigned to receive Different potential sources of oxidative stress are pre-
oral Tecfidera® at a dose of 240 mg twice or three times sent in AD brain at already early stages of pathology. AD
daily or placebo (Gold et al. 2012; Fox et al. 2012). Both brains show increased presence of neurotoxic trace ele-
clinical studies clearly demonstrated the clinical efficacy ments, iron, aluminum, mercury and copper, capable of
of Tecfidera®. Taken together, there is now ample evidence enhancing free radical generation (Markesbery 1997;
from experimental animal studies and clinical trials that Shcherbatykh and Carpenter 2007). The abnormal interac-
fumarates represent a promising novel therapeutic in the tions of trace metals with Aβ and NFTs are considered to
treatment of MS. be a potential source of oxidative stress (Bush 2003; Huang
et al. 2004; Doraiswamy and Finefrock 2004). In addition,
neuroinflammation is a prominent feature in AD brain and
ROS in Alzheimer’s disease pathology provides a significant source of oxidative stress (Colton and
Gilbert 1987; Colton et al. 2000; Eckert et al. 2003). Astro-
Alzheimer’s disease (AD) is a progressive neurodegen- cytes and microglia are associated with amyloid plaques
erative disease and the most common type of adult onset and found to be increased in the surrounding tissue (Akiy-
dementia. AD is characterized by loss of neuronal integrity ama et al. 2000). Neuropathological studies show elevated
and consequent memory impairment and cognitive decline expression levels of inducible nitric oxide synthase (iNOS)
as a result of loss of neurons and synapses (Cummings and NOX subunits, particularly in amyloid-plaque-associ-
2004). The major pathological features of AD are amyloid- ated astrocytes and/or microglial cells (Lee et al. 1993; Shi-
containing plaques and neurofibrillary changes, which mohama et al. 2000). Induction of iNOS expression in glial
occur prominently in the neocortical areas. Plaques primar- cells may result in excess release of nitric oxide and nitra-
ily consist of amyloid β (Aβ), which is a cleavage product tive stress (Wallace et al. 1997; Smith et al. 1997), and acti-
of the amyloid precursor protein. As a result of a misbal- vation of the microglial respiratory burst may lead to the
ance between production and removal of Aβ from the brain, NOX catalyzed reduction of molecular oxygen to super-
Aβ builds up in extracellular deposits in the parenchyma oxide and the formation of hydrogen peroxide (Colton and
as well as in the brain blood vessels. Several types of neu- Gilbert 1987; Lambeth 2004). Although the respiratory
rofibrillary changes occur in the AD brain, neurofibrillary burst of microglia is likely to be modulated by numerous
tangles (NFTs), dystrophic neurites and neuropil threads factors, in vitro studies have lucidly shown that Aβ itself
(Braak and Braak 1991). These are composed of intracel- is a potent trigger of microglial respiratory burst activity
lular accumulation of paired helical filaments (PHF), which (Colton et al. 2000; Van Muiswinkel et al. 1996, 1999).
mainly consist of hyperphosphorylated microtubule-associ-
ated protein tau.
AD brains show increased levels of oxidative DNA Nrf2 in Alzheimer’s disease
damage, both nuclear DNA (nDNA) and to a much greater
extent mtDNA, and oxidative damage to RNA (Mecocci In vitro studies show that Nrf2 signaling is essential
et al. 1994; Wang et al. 2005; Ding et al. 2006; Nunomura for counteracting oxidative damage and neuronal death
et al. 2006). In addition, proteins derived from AD brains induced by Aβ (Karkkainen et al. 2014; Kanninen et al.
are more oxidized than those derived from age-matched 2008). Immunohistochemical studies indicate that in nor-
controls and are most pronounced in regions containing mal hippocampus, Nrf2 is predominantly expressed in neu-
severe neurodegeneration (Sultana et al. 2006). Oxidative rons where it is localized in the cytoplasm and the nucleus
damage to proteins often affects the function of proteins (Ramsey et al. 2007). In AD brain, Nrf2 localization in the
and makes them liable for aggregation. Several studies have nucleus is dramatically decreased while the cytoplasmic
shown increased lipid peroxidation in AD brain (Sultana localization of Nrf2 remains unchanged. Nrf2 in AD hip-
et al. 2006; Volkel et al. 2006). Lipid peroxidation produces pocampal neurons does not co-localize with the pathologi-
large amounts of aldehydes, particularly the 4-hydroxyalk- cal hallmarks of AD: Aβ and phosphorylated tau (Ramsey
enals, which are produced by the oxidation of polyunsatu- et al. 2007). Remarkably, neurons in AD brain positive for
rated fatty acids (Markesbery 1997). 4-hydroxy-2-nonenal, 7-hydroxynonenal, a marker of lipid peroxidation, do not
one of the major products of polyunsaturated fatty acid per- contain Nrf2 in the nucleus. So despite the presence of oxi-
oxidation, is highly reactive and responsible for cytotoxic dative stimuli or neurotoxic Aβ, Nrf2 is not translocated
effects found with oxidative stress (Esterbauer et al. 1991). to the nucleus, indicating dysfunctional Nrf2 signaling in

13
1778 Arch Toxicol (2014) 88:1773–1786

AD neurons. In APP/PS1 transgenic mice, a mouse model Nrf2-knockout mice show increased levels of phospho-
for AD, the Nrf2-ARE pathway is attenuated in neurons rylated and sarkosyl-insoluble tau in the brain concurrent
when Aβ plaques start to appear in these mice (Kanninen with decreased levels of NDP52, and NDP52 overexpres-
et al. 2008). These findings are in line with the observed sion promotes clearance of phosphorylated tau. These data
decreased neuronal levels of Nrf2 in AD brain (Ramsey suggest that Nrf2 signaling plays a key role in autophagy-
et al. 2007). Interestingly, intrahippocampal gene delivery mediated degradation of phosphorylated tau in vivo (Jo
of Nrf2 in APP/PS1 transgenic mice results in a reduction et al. 2014).
of spatial learning deficits in aged mice (Kanninen et al.
2009). Together with the observation that Nrf2 immuno-
reactivity is prominently present in neurons in human and ROS in Parkinson’s disease pathology
mouse brain, these data suggest that the Nrf2–ARE path-
way plays an important role in protecting neuronal function With a prevalence of approximately 160 per 100,000 in
and preventing cognitive dysfunction. the Western world, Parkinson’s disease is the second most
There have been several studies suggesting that phos- common neurodegenerative disease after AD (Dawson and
phorylation of Nrf2 may contribute to its nuclear exclusion Dawson 2003). PD is characterized by rest tremor, brad-
and degradation. Nrf2 contains many serine, threonine and ykinesia, rigidity and loss of postural reflexes (Jankovic
tyrosine residues, which may provide sites for phospho- 2008). In addition, other clinical features include second-
rylation by different kinases (Bryan et al. 2013). Glycogen ary motor symptoms (e.g., shuffling gait, dysarthria) and
synthase kinase-3β, which shows increased kinase activity non-motor symptoms, such as cognitive and sensory abnor-
in AD and has been proposed as the main kinase involved malities. At the cellular level, PD is characterized by the
in tau phosphorylation, is able to directly phosphorylate preferential loss of dopaminergic neurons in the substan-
Nrf2, thereby inducing its nuclear extraction (Salazar et al. tia nigra (SN) and the presence of α-synuclein-containing
2006). Antisense oligonucleotide against glycogen synthase inclusion bodies (known as Lewy bodies) in the cytoplasm
kinase-3β in an AD mouse model resulted in increased of neurons in various regions of the brain, including the
nuclear localization of Nrf2, a decrease in oxidative stress SN, olfactory bulb, and neocortex (Mosley et al. 2006).
and improved learning and memory (Farr et al. 2014). Although intracellular deposits of highly aggregated forms
Another kinase that is able to directly phosphorylate Nrf2 of α-synuclein are characteristic of PD, in a number of
is protein kinase RNA-like endoplasmic reticulum kinase other neurodegenerative disorders, such as multiple system
(PERK) (Cullinan et al. 2003). PERK is an endoplasmic atrophy and Lewy body dementia, these aggregated forms
reticulum stress sensor and controls the activation of one of of α-synuclein are also found. Therefore, these disorders
the three independent signaling pathways that together act are collectively known as the synucleinopathies (Jellinger
as the unfolded protein response (Walter and Ron 2011). 2008; Beyer and Ariza 2007; Uversky 2007; Springer and
The unfolded protein response is a stress response of the Kahle 2006). In spite of the tremendous efforts over the
endoplasmic reticulum that is activated when misfolded past years, the pathogenesis of idiopathic PD still remains
proteins start to accumulate in the endoplasmic reticulum, largely enigmatic. Nevertheless, in the past two decades,
for instance due to presence of ROS. Increased levels of evidence is mounting that oxidative stress and mitochon-
activated PERK can be observed in neurons already in the drial dysfunction play a major role in the degeneration of
early stages of AD pathology (Hoozemans et al. 2009). nigral dopaminergic neurons.
While Nrf2 is maintained in the cytoplasm in unstressed Increased stress resulting from ROS production is one of
cells via association with Keap1, activation of PERK leads the key mechanisms that results in death of dopaminergic
to phosphorylation and dissociation of Nrf2, enabling neurons in PD, and mitochondrial complex I is considered
nuclear import (Walter and Ron 2011). This mechanism to be the primary source of ROS. Postmortem analysis of
is important to restore protein homeostasis in the endo- PD brain demonstrated an early and profound loss of the
plasmic reticulum after oxidative damage to proteins. The antioxidant glutathione; a reduction in mitochondrial com-
PERK-dependent nuclear translocation of Nrf2 is a fast and plex I activity; increased lipid, protein, and DNA oxidation
transient response (within 2 hours). Prolonged activation and/or nitration; increased superoxide dismutase activ-
of the unfolded protein response results in re-localization ity; and elevated free iron levels in the SN of parkinso-
of Nrf2 back to cytoplasm (Walter and Ron 2011), a situ- nian patients (Van Muiswinkel et al. 2004; Yasuhara et al.
ation that may apply for neurons in AD brain. Recently, it 2007; Alam et al. 1997a, b; Dexter et al. 1989; Yoritaka
was shown that activation of the Nrf2 pathway reduces the et al. 1996; Sofic et al. 1992, 2006; Healy et al. 2004a).
levels of phosphorylated tau by induction of an autophagy In addition, purified mitochondria from frontal cortex of
adaptor protein, NDP52, in neurons. NDP52 has three ARE PD patients showed mitochondrial complex I deficiency
in its promoter region and is strongly induced by Nrf2. (Parker et al. 2008), and in CSF samples of PD patients,

13
Arch Toxicol (2014) 88:1773–1786 1779

oxidized coenzyme Q10 and 8-hydroxy-2′-deoxyguanosine Within this context, it is notable that proper DJ-1 function-
were significantly increased, indicating the occurrence of ing is essential for optimal induction of NAD(P)H:quinone
oxidative damage to mitochondria and DNA in PD patients oxidoreductase-1, as loss of DJ-1 was accompanied by
(Isobe et al. 2010). In addition, certain autosomal reces- deficits in NAD(P)H:quinone oxidoreductase-1 expression.
sively inherited forms of PD can be caused by loss of func- Furthermore, DJ-1 stabilizes Nrf2 by preventing associa-
tion mutations in genes coding for mitochondrial proteins tion with its inhibitor Keap1 and Nrf2’s subsequent ubiqui-
(i.e., PINK1 and DJ-1) involved in antioxidant protection, tination (Clements et al. 2006). When DJ-1 is not intact, the
as shown by several genetic studies (Healy et al. 2004a, Nrf2 protein is unstable and transcriptional responses will
b; Valente et al. 2004). These above-described findings be reduced both under basal conditions and after exposure
are supported by a wealth of data obtained from animal to inducing agents like ROS (Clements et al. 2006).
and in vitro models of PD and corroborated by epidemio- Recently, various in vitro and in vivo models provided
logical data that link exposure to mitochondrial toxins, like information on a possible protective role of the Nrf2–ARE
1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), pathway in PD. For instance, the highly neurotoxic dopa-
rotenone and paraquat, to PD (Beal 2001; Drechsel and mine analog 6-hydroxydopamine (6-OHDA) activates the
Patel 2008; Bove et al. 2005; Przedborski and Ischiropou- Nrf2–ARE system as part of a cellular defense mecha-
los 2005). In line with these data is the notion that the SN nism to protect against ROS-mediated damage (Jakel
is more vulnerable to complex I dysfunction compared to et al. 2005). On the other hand, the loss of Nrf2 increases
other brain areas due to the generation of ROS by nigral vulnerability to 6-OHDA both in vitro and in vivo, while
dopaminergic neurons during dopamine metabolism upregulation of Nrf2 using tert-butylhydroquinone (tBHQ)
(Chinta and Andersen 2008). However, although an impor- protected against 6-OHDA-induced cell death in an in vitro
tant role for ROS in PD pathophysiology seems firmly setting (Jakel et al. 2005). Transplantation of astroglial
established, it still remains unclear whether oxidative stress cells overexpressing Nrf2 into mouse brain demonstrated
is a primary trigger for PD or merely a by-product of cel- a significant reduction in susceptibility toward 6-OHDA-
lular malfunction. mediated neurotoxicity (Jakel et al. 2007). Likewise, astro-
cytic Nrf2 overexpression attenuated MPTP-mediated tox-
icity in mice indicating that astrocytic modulation of the
Nrf2 in Parkinson’s disease Nrf2-ARE pathway represents an attractive therapeutic
strategy aimed at reducing or preventing neuronal death
Information regarding the functional status of the Nrf2– in PD (Chen et al. 2009). Alternatively, deprenyl, a drug
ARE system in PD is limited, although activation of this applied in the treatment of PD, was shown to stimulate
system in postmortem PD brain tissue is suggested by stud- Nrf2 activity as part of its cytoprotective mechanism of
ies demonstrating strong upregulation of nigral immunore- action (Nakaso et al. 2006). In in vitro studies using cul-
activity for Nrf2-regulated proteins, like NAD(P)H:quinone tured rat neuronal PC12 and glial C6 cells, the flavonoid
oxidoreductase-1 (Van Muiswinkel et al. 2004) and heme luteolin provided protection against the mitochondrial toxin
oxygenase-1 (Schipper 2004; Riedl et al. 1999). However, MPP + (i.e., the active metabolite of MPTP, see above) by
the increased expression of heme oxygenase-1 and NAD(P) activation of the Nrf2 pathway (Wruck et al. 2007). In this
H:quinone oxidoreductase-1 seems largely restricted to latter model, the knockdown of Nrf2 nullified the effect,
(astro)glial cells and is virtually absent in dopaminergic demonstrating that luteolin-induced protection is mediated
neurons. In contrast to NAD(P)H:quinone oxidoreductase-1 solely through Nrf2. Similar effects in PC12 cells were
and heme oxygenase-1, the distribution of Nrf2 in the SN found upon use of the Nrf2 transcriptional activator tBHQ
of PD brain demonstrated both a cytoplasmic and a strong as the cells were protected against the toxicity of another
nuclear immunoreactivity in neurons (Ramsey et al. 2007). PD-associated neurotoxin, deltamethrin (Li et al. 2007).
Interestingly, neurons that survive in the SN of PD patients Together these data suggest that amelioration of the PD
revealed nuclear Nrf2 localization. These data suggest that phenotype in vivo by activation of the Nrf2–ARE pathway
Nrf2 is functional in these neurons, although it remains may be feasible. Finally, genetic variation in the Nrf2 gene
unclear whether the transcriptional machinery is also intact has been linked to both AD progression and PD progres-
(Ramsey et al. 2007). In line with the findings in PD brain sion (von Otter et al. 2010a, b).
tissue, olfactory neurosphere-derived cells from patients
with sporadic PD demonstrated lower glutathione levels,
which could be restored by the Nrf2-activating agent L-sul- ROS in Huntington’s disease pathology
foraphane (Cook et al. 2011). Thus, although the Nrf2–
ARE signaling route appears to be activated in PD, other Huntington’s disease (HD) is a progressive neurodegen-
factors may be hampering effective Nrf2 gene transcription. erative disease of genetic origin clinically characterized

13
1780 Arch Toxicol (2014) 88:1773–1786

by cognitive deficits, progressive motor dysfunction and suggesting that early superoxide dismutase upregulation
psychiatric disturbance (Walker 2007). HD is caused by represents a compensatory mechanism to protect cells, but
an expanded CAG repeat (≥35 repeats) in the hunting- that this endogenous defense system becomes less func-
tin (HTT) gene, located in the short arm of chromosome tional in time (Santamaria et al. 2001). In line with these
4. This polyglutamine expansion induces conformational findings, it was shown that cytosolic superoxide dismutase
changes in the HTT protein resulting in the generation of activity was slightly reduced in HD cortex and cerebellum
large nuclear and cytoplasmic aggregates (Scherzinger (Browne et al. 1997). Activities of superoxide dismutase-1
et al. 1997). The predominant pathological hallmark of and glutathione peroxidase are reduced in leukocytes of
HD is severe atrophy of the striatum, but as the disease HD patients when compared to controls (Chen et al. 2007).
progresses, the cerebral cortex and other subcortical struc- GPx activity was unaltered in the striatum of mice trans-
tures are affected as well. The mutation that causes HD was genic for the HD mutation compared with control animals
identified decades ago, yet the precise mechanism leading (Perez-Severiano et al. 2004). Recently, Mason and co-
to the degeneration of neurons in specific brain regions workers, using a combination of genetic and pharmacologi-
remains poorly understood. However, defective mitochon- cal approaches, demonstrated that boosting glutathione per-
drial bioenergetics, oxidative stress and associated impaired oxidase activity is neuroprotective in experimental models
neuronal energy metabolism are likely involved in the etiol- of HD (Mason et al. 2013). Heme oxygenase-1 immuno-
ogy of the disease. reactivity is markedly enhanced in HD brain tissue, pre-
Immunohistochemical analysis revealed increased lev- dominantly in advanced stages of the disease (Browne et al.
els of oxidized nucleotides and lipid peroxidation markers 1999).
in the brain tissue of HD animal models and HD patients Overexpression of superoxide dismutase-1 attenuated
(Browne et al. 1997; Perez-Severiano et al. 2013; Bog- proteasomal dysfunction, mutant HTT aggregation and cell
danov et al. 2001; Browne and Beal 2006). In addition, death in mutant HTT expressing cells, indicating the thera-
increased plasma and urine levels of oxidized nucleotides peutic potential of enhancing antioxidant enzyme levels
have been reported in both this experimental HD model (Goswami et al. 2006). Yet, only a few studies have focused
and HD patients (Hersch et al. 2006; Stoy et al. 2005; Bog- on the effects of the Nrf2-ARE pathway in mouse models
danov et al. 2001), as well as increased concentrations of of HD (Shih et al. 2005a; Calkins et al. 2005). Nrf2-defi-
8-hydroxydeoxyguanosine concentrations with HD dis- cient cells and Nrf2 knockout mice were shown to be sig-
ease progression (Long et al. 2012). Oxidative damage to nificantly more vulnerable to malonic acid and 3-nitropro-
mtDNA is evident in the parietal cortex of late-stage HD pionic acid, two mitochondrial toxins that inhibit complex
individuals and might contribute to subsequent mitochon- II and cause striatal damage reminiscent of HD (Shih et al.
drial dysfunction (Siddiqui et al. 2012). Overall, there is 2005a). Injection of Nrf2-overexpressing astrocytes in the
ample evidence for the occurrence of oxidative damage to striatum before treatment with malonic acid and 3-nitro-
essential biomolecules in experimental models of HD and propionic acid provided further protection against com-
HD patients (yala-Pena 2013). NOX activity was elevated plex II inhibition and had a strong neuroprotective effect
in HD cortex and striatum, particularly in pre-symptomatic (Calkins et al. 2005). Therapeutic strategies aimed at boost-
patients indicating that increased NOX activity is an early ing endogenous antioxidant levels in HD animal models
pathological event occurring before clinical deficits become show promising results. Administration of DMF resulted
apparent (Valencia et al. 2013). Pharmacological inhibition in enhanced Nrf2 immunostaining in a subset of neurons
of NOX by apocynin led to reduction in lipid peroxidation (not astrocytes), improved motor impairment, preserva-
and cellular injury in an experimental HD rat model (Mal- tion of striatal and motor cortex neurons and increased
donado et al. 2010). life span (Ellrichmann et al. 2011). Oral administration of
synthetic triterpenoids, potent inducers of the Nrf2 path-
way, increased levels of Nrf2-driven genes in the CNS,
Nrf2 in Huntington’s disease attenuated oxidative stress, ameliorated motor behavior and
increased longevity in a transgenic mouse model of HD
The involvement of free radicals in the pathology of HD (Stack et al. 2010).
is further highlighted by the observation that antioxi-
dant treatment strategies attenuate disease symptoms in
HD animal models (Klivenyi et al. 2003; Ferrante et al. Conclusion and future perspectives
2002). Relatively few studies focused on the role of endog-
enous antioxidant enzymes in HD animal models and Several research groups have highlighted the protective
patients. Superoxide dismutase activities were increased role of Nrf2 under different pathological conditions indicat-
in young transgenic HD mice, but decreased in older mice, ing that Nrf2 is critically involved in the protection against

13
Arch Toxicol (2014) 88:1773–1786 1781

oxidative stress. A large variety of natural and synthetic in Parkinson’s but not incidental Lewy body disease. J Neuro-
compounds that activate the Nrf2 pathway, also referred to chem 69:1326–1329
Alam ZI, Jenner A, Daniel SE, Lees AJ, Cairns N, Marsden CD, Jen-
as ‘Nrf2 activators,’ have been identified, including among ner P, Halliwell B (1997b) Oxidative DNA damage in the par-
others, tert-butyl hydroquinone, sulforaphane, curcumin kinsonian brain: an apparent selective increase in 8-hydroxy-
and dithiolethiones. Still, new small molecule Nrf2 acti- guanine levels in substantia nigra. J Neurochem 69:1196–1203
vators are being designed and synthesized. The Johnson Beal MF (2001) Experimental models of Parkinson’s disease. Nat Rev
Neurosci 2:325–334
lab used an elegant computational approach to discover Beal MF (2002) Oxidatively modified proteins in aging and disease.
novel compounds with the ability to induce Nrf2-mediated Free Radic Biol Med 32:797–803
gene expression (Williamson et al. 2012). The therapeutic Bedard K, Krause KH (2007) The NOX family of ROS-generating
potential of these novel identified compounds needs to be NADPH oxidases: physiology and pathophysiology. Physiol
Rev 87:245–313
explored in animal models of neurodegenerative diseases Beyer K, Ariza A (2007) Protein aggregation mechanisms in synucle-
in future. Importantly, preclinical data are emerging that inopathies: commonalities and differences. J Neuropathol Exp
activation of the Nrf2 pathway by these compounds results Neurol 66:965–974
in enhanced protection against oxidative damage and con- Bo L, Dawson TM, Wesselingh S, Mork S, Choi S, Kong PA, Han-
ley D, Trapp BD (1994) Induction of nitric oxide synthase in
comitant neurodegeneration (Shih et al. 2005a, b; Zhao demyelinating regions of multiple sclerosis brains. Ann Neurol
et al. 2006, 2007; Wang and Shuaib 2007). 36:778–786
Although there are ample preclinical data demonstrat- Bogdanov MB, Andreassen OA, Dedeoglu A, Ferrante RJ, Beal
ing the therapeutic efficacy of Nrf2 activators, only a few MF (2001) Increased oxidative damage to DNA in a trans-
genic mouse model of Huntington’s disease. J Neurochem
Nrf2-activating compounds have been tested in a clinical 79:1246–1249
setting. Biogen Idec developed an oral formulation of DMF Bolanos JP, Almeida A, Stewart V, Peuchen S, Land JM, Clark JB,
known as Tecfidera®, which is now approved for the treat- Heales SJ (1997) Nitric oxide-mediated mitochondrial damage
ment of relapsing–remitting MS. Future studies are needed in the brain: mechanisms and implications for neurodegenera-
tive diseases. J Neurochem 68:2227–2240
to reveal the efficacy of Tecfidera® in progressive MS Bove J, Prou D, Perier C, Przedborski S (2005) Toxin-induced models
patients. Intriguingly, fumaric acid esters are also effective of Parkinson’s disease. NeuroRx 2:484–494
in experimental animal models of HD (Ellrichmann et al. Braak H, Braak E (1991) Neuropathological stageing of Alzheimer-
2011), indicating that the clinical application of Tecfidera® related changes. Acta Neuropathol 82:239–259
Brigelius-Flohe R, Maiorino M (2013) Glutathione peroxidases. Bio-
may extend beyond MS. chim Biophys Acta 1830:3289–3303
Free radical formation and concomitant oxidative stress Browne SE, Beal MF (2006) Oxidative damage in Huntington’s dis-
are not restricted to the neurodegenerative diseases dis- ease pathogenesis. Antioxid Redox Signal 8:2061–2073
cussed in this review, but represent common pathological Browne SE, Bowling AC, MacGarvey U, Baik MJ, Berger SC, Muqit
MM, Bird ED, Beal MF (1997) Oxidative damage and meta-
features of many neurological disorders, including motor bolic dysfunction in Huntington’s disease: selective vulnerabil-
neuron disorders, cerebral hemorrhages and traumatic brain ity of the basal ganglia. Ann Neurol 41:646–653
injury. Future studies are warranted to further investigate Browne SE, Ferrante RJ, Beal MF (1999) Oxidative stress in Hun-
the therapeutic potential and clinical applicability of Nrf2 tington’s disease. Brain Pathol 9:147–163
Bryan HK, Olayanju A, Goldring CE, Park BK (2013) The Nrf2 cell
activators in experimental animal models for neurodegen- defence pathway: Keap1-dependent and -independent mecha-
eration and ultimately clinical trials. nisms of regulation. Biochem Pharmacol 85:705–717
Bush AI (2003) The metallobiology of Alzheimer’s disease. Trends
Acknowledgments This work was supported by grants from Neurosci 26:207–214
‘Stichting Vrienden MS Research,’ The Netherlands, project numbers Calabrese V, Raffaele R, Cosentino E, Rizza V (1994) Changes in
MS 05-358c (J. van Horssen). cerebrospinal fluid levels of malondialdehyde and glutathione
reductase activity in multiple sclerosis. Int J Clin Pharmacol
Res 14:119–123
Calkins MJ, Jakel RJ, Johnson DA, Chan K, Kan YW, Johnson JA
References (2005) Protection from mitochondrial complex II inhibition
in vitro and in vivo by Nrf2-mediated transcription. Proc Natl
Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM, Cooper Acad Sci USA 102:244–249
NR, Eikelenboom P, Emmerling M, Fiebich BL, Finch CE, Campbell GR, Ziabreva I, Reeve AK, Krishnan KJ, Reynolds R,
Frautschy S, Griffin WS, Hampel H, Hull M, Landreth G, Lue Howell O, Lassmann H, Turnbull DM, Mahad DJ (2011) Mito-
L, Mrak R, Mackenzie IR, McGeer PL, O’Banion MK, Pachter chondrial DNA deletions and neurodegeneration in multiple
J, Pasinetti G, Plata-Salaman C, Rogers J, Rydel R, Shen Y, sclerosis. Ann Neurol 69:481–492
Streit W, Strohmeyer R, Tooyoma I, Van Muiswinkel FL, Veer- Carlson NG, Rose JW (2006) Antioxidants in multiple sclerosis: do
huis R, Walker D, Webster S, Wegrzyniak B, Wenk G, Wyss- they have a role in therapy? CNS Drugs 20:433–441
Coray T (2000) Inflammation and Alzheimer’s disease. Neuro- Chen CM, Wu YR, Cheng ML, Liu JL, Lee YM, Lee PW, Soong BW,
biol Aging 21:383–421 Chiu DT (2007) Increased oxidative damage and mitochondrial
Alam ZI, Daniel SE, Lees AJ, Marsden DC, Jenner P, Halliwell B abnormalities in the peripheral blood of Huntington’s disease
(1997a) A generalised increase in protein carbonyls in the brain patients. Biochem Biophys Res Commun 359:335–340

13
1782 Arch Toxicol (2014) 88:1773–1786

Chen JW, Breckwoldt MO, Aikawa E, Chiang G, Weissleder R (2008) oligonucleotide against GSK-3beta in brain of SAMP8 mice
Myeloperoxidase-targeted imaging of active inflammatory improves learning and memory and decreases oxidative stress:
lesions in murine experimental autoimmune encephalomyelitis. involvement of transcription factor Nrf2 and implications for
Brain 131:1123–1133 Alzheimer disease. Free Radic Biol Med 67:387–395
Chen PC, Vargas MR, Pani AK, Smeyne RJ, Johnson DA, Kan YW, Ferrante RJ, Andreassen OA, Dedeoglu A, Ferrante KL, Jenkins BG,
Johnson JA (2009) Nrf2-mediated neuroprotection in the MPTP Hersch SM, Beal MF (2002) Therapeutic effects of coenzyme
mouse model of Parkinson’s disease: critical role for the astro- Q10 and remacemide in transgenic mouse models of Hunting-
cyte. Proc Natl Acad Sci USA 106:2933–2938 ton’s disease. J Neurosci 22:1592–1599
Chinta SJ, Andersen JK (2008) Redox imbalance in Parkinson’s dis- Ferretti G, Bacchetti T, Principi F, Di Ludovico F, Viti B, Angeleri
ease. Biochim Biophys Acta 1780:1362–1367 VA, Danni M, Provinciali L (2005) Increased levels of lipid
Clements CM, McNally RS, Conti BJ, Mak TW, Ting JP (2006) DJ-1, hydroperoxides in plasma of patients with multiple sclerosis: a
a cancer- and Parkinson’s disease-associated protein, stabilizes relationship with paraoxonase activity. Mult Scler 11:677–682
the antioxidant transcriptional master regulator Nrf2. Proc Natl Fiebiger SM, Bros H, Grobosch T, Janssen A, Chanvillard C, Paul F,
Acad Sci USA 103:15091–15096 Dorr J, Millward JM, Infante-Duarte C (2013) The antioxidant
Colton CA, Gilbert DL (1987) Production of superoxide anions by a idebenone fails to prevent or attenuate chronic experimental
CNS macrophage, the microglia. FEBS Lett 223:284–288 autoimmune encephalomyelitis in the mouse. J Neuroimmunol
Colton CA, Chernyshev ON, Gilbert DL, Vitek MP (2000) Microglial 262:66–71
contribution to oxidative stress in Alzheimer’s disease. Ann N Y Fischer MT, Sharma R, Lim JL, Haider L, Frischer JM, Drexhage
Acad Sci 899:292–307 J, Mahad D, Bradl M, van Horssen J, Lassmann H (2012)
Cook AL, Vitale AM, Ravishankar S, Matigian N, Sutherland NADPH oxidase expression in active multiple sclerosis lesions
GT, Shan J, Sutharsan R, Perry C, Silburn PA, Mellick GD, in relation to oxidative tissue damage and mitochondrial injury.
Whitelaw ML, Wells CA, kay-Sim A, Wood SA (2011) NRF2 Brain 135:886–899
activation restores disease related metabolic deficiencies in Fox RJ, Miller DH, Phillips JT, Hutchinson M, Havrdova E, Kita
olfactory neurosphere-derived cells from patients with sporadic M, Yang M, Raghupathi K, Novas M, Sweetser MT, Viglietta
Parkinson’s disease. PLoS one 6:e21907 V, Dawson KT (2012) Placebo-controlled phase 3 study of
Cullinan SB, Zhang D, Hannink M, Arvisais E, Kaufman RJ, Diehl oral BG-12 or glatiramer in multiple sclerosis. N Engl J Med
JA (2003) Nrf2 is a direct PERK substrate and effector of 367:1087–1097
PERK-dependent cell survival. Mol Cell Biol 23:7198–7209 French HM, Reid M, Mamontov P, Simmons RA, Grinspan JB (2009)
Cummings JL (2004) Alzheimer’s disease. N Engl J Med 351:56–67 Oxidative stress disrupts oligodendrocyte maturation. J Neuro-
Dawson TM, Dawson VL (2003) Molecular pathways of neurodegen- sci Res 87:3076–3087
eration in Parkinson’s disease. Science 302:819–822 Gilgun-Sherki Y, Melamed E, Offen D (2001) Oxidative stress induced-
De Groot CJ, Ruuls SR, Theeuwes JW, Dijkstra CD, Van der Valk P neurodegenerative diseases: the need for antioxidants that pen-
(1997) Immunocytochemical characterization of the expression etrate the blood brain barrier. Neuropharmacology 40:959–975
of inducible and constitutive isoforms of nitric oxide synthase Gold R, Kappos L, Arnold DL, Bar-Or A, Giovannoni G, Selmaj K,
in demyelinating multiple sclerosis lesions. J Neuropathol Exp Tornatore C, Sweetser MT, Yang M, Sheikh SI, Dawson KT
Neurol 56:10–20 (2012) Placebo-controlled phase 3 study of oral BG-12 for
Dexter DT, Carter CJ, Wells FR, Javoy-Agid F, Agid Y, Lees A, Jen- relapsing multiple sclerosis. N Engl J Med 367:1098–1107
ner P, Marsden CD (1989) Basal lipid peroxidation in sub- Goswami A, Dikshit P, Mishra A, Mulherkar S, Nukina N, Jana NR
stantia nigra is increased in Parkinson’s disease. J Neurochem (2006) Oxidative stress promotes mutant huntingtin aggrega-
52:381–389 tion and mutant huntingtin-dependent cell death by mimick-
Ding Q, Markesbery WR, Cecarini V, Keller JN (2006) Decreased ing proteasomal malfunction. Biochem Biophys Res Commun
RNA, and increased RNA oxidation, in ribosomes from early 342:184–190
Alzheimer’s disease. Neurochem Res 31:705–710 Gray E, Thomas TL, Betmouni S, Scolding N, Love S (2008a) Ele-
Doraiswamy PM, Finefrock AE (2004) Metals in our minds: thera- vated activity and microglial expression of myeloperoxidase in
peutic implications for neurodegenerative disorders. Lancet demyelinated cerebral cortex in multiple sclerosis. Brain Pathol
Neurol 3:431–434 18:86–95
Drechsel DA, Patel M (2008) Role of reactive oxygen species in the Gray E, Thomas TL, Betmouni S, Scolding N, Love S (2008b) Ele-
neurotoxicity of environmental agents implicated in Parkinson’s vated myeloperoxidase activity in white matter in multiple scle-
disease. Free Radic Biol Med 44:1873–1886 rosis. Neurosci Lett 444:195–198
Dringen R, Pawlowski PG, Hirrlinger J (2005) Peroxide detoxifica- Greco A, Minghetti L, Sette G, Fieschi C, Levi G (1999) Cerebro-
tion by brain cells. J Neurosci Res 79:157–165 spinal fluid isoprostane shows oxidative stress in patients with
Eckert A, Keil U, Marques CA, Bonert A, Frey C, Schussel K, Mul- multiple sclerosis. Neurology 53:1876–1879
ler WE (2003) Mitochondrial dysfunction, apoptotic cell death, Haider L, Fischer MT, Frischer JM, Bauer J, Hoftberger R, Bot-
and Alzheimer’s disease. Biochem Pharmacol 66:1627–1634 ond G, Esterbauer H, Binder CJ, Witztum JL, Lassmann H
Ellrichmann G, Petrasch-Parwez E, Lee DH, Reick C, Arning L, Saft (2011) Oxidative damage in multiple sclerosis lesions. Brain
C, Gold R, Linker RA (2011) Efficacy of fumaric acid esters 134:1914–1924
in the R6/2 and YAC128 models of Huntington’s disease. PLoS Healy DG, bou-Sleiman PM, Valente EM, Gilks WP, Bhatia K, Quinn
one 6:e16172 N, Lees AJ, Wood NW (2004a) DJ-1 mutations in Parkinson’s
Esterbauer H, Schaur RJ, Zollner H (1991) Chemistry and biochemis- disease. J Neurol Neurosurg Psychiatry 75:144–145
try of 4-hydroxynonenal, malonaldehyde and related aldehydes. Healy DG, bou-Sleiman PM, Wood NW (2004b) PINK, PANK, or
Free Radic Biol Med 11:81–128 PARK? A clinicians’ guide to familial parkinsonism. Lancet
Evans MD, Dizdaroglu M, Cooke MS (2004) Oxidative DNA dam- Neurol 3:652–662
age and disease: induction, repair and significance. Mutat Res Hendriks JJ, Alblas J, Van Der Pol SM, van Tol EA, Dijkstra CD, De
567:1–61 Vries HE (2004) Flavonoids influence monocytic GTPase activ-
Farr SA, Ripley JL, Sultana R, Zhang Z, Niehoff ML, Platt TL, Mur- ity and are protective in experimental allergic encephalitis. J
phy MP, Morley JE, Kumar V, Butterfield DA (2014) Antisense Exp Med 200:1667–1672

13
Arch Toxicol (2014) 88:1773–1786 1783

Hersch SM, Gevorkian S, Marder K, Moskowitz C, Feigin A, Cox M, Koistinaho J (2008) Nuclear factor erythroid 2-related factor 2
Como P, Zimmerman C, Lin M, Zhang L, Ulug AM, Beal MF, protects against beta amyloid. Mol Cell Neurosci 39:302–313
Matson W, Bogdanov M, Ebbel E, Zaleta A, Kaneko Y, Jen- Kanninen K, Heikkinen R, Malm T, Rolova T, Kuhmonen S,
kins B, Hevelone N, Zhang H, Yu H, Schoenfeld D, Ferrante R, Leinonen H, Yla-Herttuala S, Tanila H, Levonen AL, Koisti-
Rosas HD (2006) Creatine in Huntington disease is safe, toler- naho M, Koistinaho J (2009) Intrahippocampal injection of a
able, bioavailable in brain and reduces serum 8OH2′dG. Neu- lentiviral vector expressing Nrf2 improves spatial learning in a
rology 66:250–252 mouse model of Alzheimer’s disease. Proc Natl Acad Sci USA
Higgins GC, Beart PM, Shin YS, Chen MJ, Cheung NS, Nagley P 106:16505–16510
(2010) Oxidative stress: emerging mitochondrial and cellu- Karg E, Klivenyi P, Nemeth I, Bencsik K, Pinter S, Vecsei L (1999)
lar themes and variations in neuronal injury. J Alzheimers Dis Nonenzymatic antioxidants of blood in multiple sclerosis. J
20(Suppl 2):S453–S473 Neurol 246:533–539
Hofmann B, Hecht HJ, Flohe L (2002) Peroxiredoxins. Biol Chem Karkkainen V, Pomeshchik Y, Savchenko E, Dhungana H, Kurronen
383:347–364 A, Lehtonen S, Naumenko N, Tavi P, Levonen AL, Yamamoto
Hoozemans JJ, van Haastert ES, Nijholt DA, Rozemuller AJ, Eike- M, Malm T, Magga J, Kanninen KM, Koistinaho J (2014) Nrf2
lenboom P, Scheper W (2009) The unfolded protein response regulates neurogenesis and protects neural progenitor cells
is activated in pretangle neurons in Alzheimer’s disease hip- against Abeta toxicity. Stem Cells 32:1904–1916
pocampus. Am J Pathol 174:1241–1251 Kasai H (2002) Chemistry-based studies on oxidative DNA damage: for-
Huang X, Moir RD, Tanzi RE, Bush AI, Rogers JT (2004) Redox- mation, repair, and mutagenesis. Free Radic Biol Med 33:450–456
active metals, oxidative stress, and Alzheimer’s disease pathol- Kim YJ, Ahn JY, Liang P, Ip C, Zhang Y, Park YM (2007) Human prx1
ogy. Ann N Y Acad Sci 1012:153–163 gene is a target of Nrf2 and is up-regulated by hypoxia/reoxy-
Husain J, Juurlink BH (1995) Oligodendroglial precursor cell suscep- genation: implication to tumor biology. Cancer Res 67:546–554
tibility to hypoxia is related to poor ability to cope with reactive Klivenyi P, Ferrante RJ, Gardian G, Browne S, Chabrier PE, Beal
oxygen species. Brain Res 698:86–94 MF (2003) Increased survival and neuroprotective effects of
Ishii T, Itoh K, Takahashi S, Sato H, Yanagawa T, Katoh Y, Bannai BN82451 in a transgenic mouse model of Huntington’s disease.
S, Yamamoto M (2000) Transcription factor Nrf2 coordinately J Neurochem 86:267–272
regulates a group of oxidative stress-inducible genes in mac- Koch M, Ramsaransing GS, Arutjunyan AV, Stepanov M, Teelken A,
rophages. J Biol Chem 275:16023–16029 Heersema DJ, De Keyser J (2006) Oxidative stress in serum and
Iskander K, Li J, Han S, Zheng B, Jaiswal AK (2006) NQO1 and peripheral blood leukocytes in patients with different disease
NQO2 regulation of humoral immunity and autoimmunity. J courses of multiple sclerosis. J Neurol 253:483–487
Biol Chem 281:30917–30924 Lambeth JD (2004) NOX enzymes and the biology of reactive oxy-
Isobe C, Abe T, Terayama Y (2010) Levels of reduced and oxidized gen. Nat Rev Immunol 4:181–189
coenzyme Q-10 and 8-hydroxy-2′-deoxyguanosine in the cer- Lee SC, Dickson DW, Liu W, Brosnan CF (1993) Induction of nitric
ebrospinal fluid of patients with living Parkinson’s disease dem- oxide synthase activity in human astrocytes by interleukin-1
onstrate that mitochondrial oxidative damage and/or oxidative beta and interferon-gamma. J Neuroimmunol 46:19–24
DNA damage contributes to the neurodegenerative process. Li Y, Jaiswal AK (1992) Regulation of human NAD(P)H:quinone
Neurosci Lett 469:159–163 oxidoreductase gene. Role of AP1 binding site contained
Itoh K, Wakabayashi N, Katoh Y, Ishii T, O’Connor T, Yamamoto M within human antioxidant response element. J Biol Chem
(2003) Keap1 regulates both cytoplasmic-nuclear shuttling and 267:15097–15104
degradation of Nrf2 in response to electrophiles. Genes Cells Li HY, Zhong YF, Wu SY, Shi N (2007) NF-E2 related factor 2 acti-
8:379–391 vation and heme oxygenase-1 induction by tert-butylhydroqui-
Jaiswal AK (2000) Regulation of genes encoding NAD(P)H:quinone none protect against deltamethrin-mediated oxidative stress in
oxidoreductases. Free Radic Biol Med 29:254–262 PC12 cells. Chem Res Toxicol 20:1242–1251
Jakel RJ, Kern JT, Johnson DA, Johnson JA (2005) Induction of the Li B, Cui W, Liu J, Li R, Liu Q, Xie XH, Ge XL, Zhang J, Song XJ,
protective antioxidant response element pathway by 6-hydroxy- Wang Y, Guo L (2013) Sulforaphane ameliorates the develop-
dopamine in vivo and in vitro. Toxicol Sci 87:176–186 ment of experimental autoimmune encephalomyelitis by antag-
Jakel RJ, Townsend JA, Kraft AD, Johnson JA (2007) Nrf2-mediated onizing oxidative stress and Th17-related inflammation in mice.
protection against 6-hydroxydopamine. Brain Res 1144:192–201 Exp Neurol 250:239–249
Jankovic J (2008) Parkinson’s disease: clinical features and diagnosis. Lin MT, Beal MF (2006) Mitochondrial dysfunction and oxidative
J Neurol Neurosurg Psychiatry 79:368–376 stress in neurodegenerative diseases. Nature 443:787–795
Jellinger KA (2008) Neuropathological aspects of Alzheimer disease, Linker RA, Lee DH, Ryan S, van Dam AM, Conrad R, Bista P, Zeng
Parkinson disease and frontotemporal dementia. Neurodegener W, Hronowsky X, Buko A, Chollate S, Ellrichmann G, Bruck
Dis 5:118–121 W, Dawson K, Goelz S, Wiese S, Scannevin RH, Lukashev M,
Jo C, Gundemir S, Pritchard S, Jin YN, Rahman I, Johnson GV (2014) Gold R (2011) Fumaric acid esters exert neuroprotective effects
Nrf2 reduces levels of phosphorylated tau protein by inducing in neuroinflammation via activation of the Nrf2 antioxidant
autophagy adaptor protein NDP52. Nat Commun 5:3496 pathway. Brain 134:678–692
Johnson F, Giulivi C (2005) Superoxide dismutases and their impact Liu JS, Zhao ML, Brosnan CF, Lee SC (2001) Expression of induc-
upon human health. Mol Asp Med 26:340–352 ible nitric oxide synthase and nitrotyrosine in multiple sclerosis
Johnson DA, Amirahmadi S, Ward C, Fabry Z, Johnson JA (2010) lesions. Am J Pathol 158:2057–2066
The absence of the pro-antioxidant transcription factor Nrf2 Long DJ, Jaiswal AK (2000) NRH:quinone oxidoreductase2 (NQO2).
exacerbates experimental autoimmune encephalomyelitis. Toxi- Chem Biol Interact 129:99–112
col Sci 114:237–246 Long JD, Matson WR, Juhl AR, Leavitt BR, Paulsen JS (2012)
Juurlink BH, Thorburne SK, Hertz L (1998) Peroxide-scavenging def- 8OHdG as a marker for Huntington disease progression. Neu-
icit underlies oligodendrocyte susceptibility to oxidative stress. robiol Dis 46:625–634
Glia 22:371–378 Lovell MA, Ehmann WD, Mattson MP, Markesbery WR (1997) Ele-
Kanninen K, Malm TM, Jyrkkanen HK, Goldsteins G, Keksa-Gold- vated 4-hydroxynonenal in ventricular fluid in Alzheimer’s dis-
steine V, Tanila H, Yamamoto M, Yla-Herttuala S, Levonen AL, ease. Neurobiol Aging 18:457–461

13
1784 Arch Toxicol (2014) 88:1773–1786

Lu F, Selak M, O’Connor J, Croul S, Lorenzana C, Butunoi C, Kal- Nguyen T, Yang CS, Pickett CB (2004) The pathways and molecular
man B (2000) Oxidative damage to mitochondrial DNA and mechanisms regulating Nrf2 activation in response to chemical
activity of mitochondrial enzymes in chronic active lesions of stress. Free Radic Biol Med 37:433–441
multiple sclerosis. J Neurol Sci 177:95–103 Nikic I, Merkler D, Sorbara C, Brinkoetter M, Kreutzfeldt M, Bareyre
Mahad DJ, Ziabreva I, Campbell G, Lax N, White K, Hanson PS, FM, Bruck W, Bishop D, Misgeld T, Kerschensteiner M (2011)
Lassmann H, Turnbull DM (2009) Mitochondrial changes A reversible form of axon damage in experimental autoimmune
within axons in multiple sclerosis. Brain 132:1161–1174 encephalomyelitis and multiple sclerosis. Nat Med 17:495–499
Maldonado PD, Molina-Jijon E, Villeda-Hernandez J, Galvan-Arzate Nunomura A, Honda K, Takeda A, Hirai K, Zhu X, Smith MA, Perry
S, Santamaria A, Pedraza-Chaverri J (2010) NAD(P)H oxidase G (2006) Oxidative damage to RNA in neurodegenerative dis-
contributes to neurotoxicity in an excitotoxic/prooxidant model eases. J Biomed Biotechnol 2006:82323
of Huntington’s disease in rats: protective role of apocynin. J Pareek TK, Belkadi A, Kesavapany S, Zaremba A, Loh SL, Bai L,
Neurosci Res 88:620–629 Cohen ML, Meyer C, Liby KT, Miller RH, Sporn MB, Letterio
Mao P, Manczak M, Shirendeb UP, Reddy PH (2013) MitoQ, a mito- JJ (2011) Triterpenoid modulation of IL-17 and Nrf-2 expres-
chondria-targeted antioxidant, delays disease progression and sion ameliorates neuroinflammation and promotes remyelina-
alleviates pathogenesis in an experimental autoimmune enceph- tion in autoimmune encephalomyelitis. Sci Rep 1:201
alomyelitis mouse model of multiple sclerosis. Biochim Bio- Parker WD Jr, Parks JK, Swerdlow RH (2008) Complex I deficiency
phys Acta 1832:2322–2331 in Parkinson’s disease frontal cortex. Brain Res 1189:215–218
Mariani E, Polidori MC, Cherubini A, Mecocci P (2005) Oxidative stress Perez-Severiano F, Santamaria A, Pedraza-Chaverri J, Medina-Cam-
in brain aging, neurodegenerative and vascular diseases: an over- pos ON, Rios C, Segovia J (2004) Increased formation of reac-
view. J Chromatogr B Analyt Technol Biomed Life Sci 827:65–75 tive oxygen species, but no changes in glutathione peroxidase
Marik C, Felts PA, Bauer J, Lassmann H, Smith KJ (2007) Lesion activity, in striata of mice transgenic for the Huntington’s dis-
genesis in a subset of patients with multiple sclerosis: a role for ease mutation. Neurochem Res 29:729–733
innate immunity? Brain 130:2800–2815 Perez-Severiano F, Montes S, Geronimo-Olvera C, Segovia J
Markesbery WR (1997) Oxidative stress hypothesis in Alzheimer’s (2013) Study of oxidative damage and antioxidant systems in
disease. Free Radic Biol Med 23:134–147 two Huntington’s disease rodent models. Methods Mol Biol
Marklund SL (1982) Human copper-containing superoxide dis- 1010:177–200
mutase of high molecular weight. Proc Natl Acad Sci USA Przedborski S, Ischiropoulos H (2005) Reactive oxygen and nitrogen
79:7634–7638 species: weapons of neuronal destruction in models of Parkin-
Mason RP, Casu M, Butler N, Breda C, Campesan S, Clapp J, Green son’s disease. Antioxid Redox Signal 7:685–693
EW, Dhulkhed D, Kyriacou CP, Giorgini F (2013) Glutathione Qi X, Lewin AS, Sun L, Hauswirth WW, Guy J (2006) Mitochondrial
peroxidase activity is neuroprotective in models of Huntington’s protein nitration primes neurodegeneration in experimental
disease. Nat Genet 45:1249–1254 autoimmune encephalomyelitis. J Biol Chem 281:31950–31962
McCord JM (1976) Iron- and manganese-containing superoxide dis- Ramsey CP, Glass CA, Montgomery MB, Lindl KA, Ritson GP, Chia
mutases: structure, distribution, and evolutionary relationships. LA, Hamilton RL, Chu CT, Jordan-Sciutto KL (2007) Expres-
Adv Exp Med Biol 74:540–550 sion of Nrf2 in neurodegenerative diseases. J Neuropathol Exp
McCord JM, Edeas MA (2005) SOD, oxidative stress and human Neurol 66:75–85
pathologies: a brief history and a future vision. Biomed Phar- Rhee SG, Chae HZ, Kim K (2005) Peroxiredoxins: a historical
macother 59:139–142 overview and speculative preview of novel mechanisms and
McCord JM, Fridovich I (1969) Superoxide dismutase. An enzy- emerging concepts in cell signaling. Free Radic Biol Med
mic function for erythrocuprein (hemocuprein). J Biol Chem 38:1543–1552
244:6049–6055 Riedl AG, Watts PM, Brown CT, Jenner P (1999) P450 and heme
Mecocci P, MacGarvey U, Beal MF (1994) Oxidative damage to oxygenase enzymes in the basal ganglia and their roles in Par-
mitochondrial DNA is increased in Alzheimer’s disease. Ann kinson’s disease. Adv Neurol 80:271–286
Neurol 36:747–751 Ryter SW, Alam J, Choi AM (2006) Heme oxygenase-1/carbon mon-
Mitrovic B, Ignarro LJ, Montestruque S, Smoll A, Merrill JE (1994) oxide: from basic science to therapeutic applications. Physiol
Nitric oxide as a potential pathological mechanism in demy- Rev 86:583–650
elination: its differential effects on primary glial cells in vitro. Salazar M, Rojo AI, Velasco D, de Sagarra RM, Cuadrado A (2006)
Neuroscience 61:575–585 Glycogen synthase kinase-3beta inhibits the xenobiotic and
Moosmann B, Behl C (2002) Antioxidants as treatment for neurode- antioxidant cell response by direct phosphorylation and
generative disorders. Expert Opin Investig Drugs 11:1407–1435 nuclear exclusion of the transcription factor Nrf2. J Biol Chem
Morrow JD, Chen Y, Brame CJ, Yang J, Sanchez SC, Xu J, Zackert 281:14841–14851
WE, Awad JA, Roberts LJ (1999) The isoprostanes: unique Santamaria A, Perez-Severiano F, Rodriguez-Martinez E, Maldonado
prostaglandin-like products of free-radical-initiated lipid per- PD, Pedraza-Chaverri J, Rios C, Segovia J (2001) Comparative
oxidation. Drug Metab Rev 31:117–139 analysis of superoxide dismutase activity between acute phar-
Mosley RL, Benner EJ, Kadiu I, Thomas M, Boska MD, Hasan K, macological models and a transgenic mouse model of Hunting-
Laurie C, Gendelman HE (2006) Neuroinflammation, Oxidative ton’s disease. Neurochem Res 26:419–424
Stress and the pathogenesis of Parkinson’s disease. Clin Neuro- Scannevin RH, Chollate S, Jung MY, Shackett M, Patel H, Bista P,
sci Res 6:261–281 Zeng W, Ryan S, Yamamoto M, Lukashev M, Rhodes KJ (2012)
Nakaso K, Nakamura C, Sato H, Imamura K, Takeshima T, Fumarates promote cytoprotection of central nervous system
Nakashima K (2006) Novel cytoprotective mechanism of anti- cells against oxidative stress via the nuclear factor (erythroid-
parkinsonian drug deprenyl: PI3K and Nrf2-derived induc- derived 2)-like 2 pathway. J Pharmacol Exp Ther 341:274–284
tion of antioxidative proteins. Biochem Biophys Res Commun Scherzinger E, Lurz R, Turmaine M, Mangiarini L, Hollenbach B,
339:915–922 Hasenbank R, Bates GP, Davies SW, Lehrach H, Wanker EE
Nguyen T, Sherratt PJ, Pickett CB (2003) Regulatory mechanisms (1997) Huntingtin-encoded polyglutamine expansions form
controlling gene expression mediated by the antioxidant amyloid-like protein aggregates in vitro and in vivo. Cell
response element. Annu Rev Pharmacol Toxicol 43:233–260 90:549–558

13
Arch Toxicol (2014) 88:1773–1786 1785

Schimrigk S, Brune N, Hellwig K, Lukas C, Bellenberg B, Rieks M, Valencia A, Sapp E, Kimm JS, McClory H, Reeves PB, Alexander J,
Hoffmann V, Pohlau D, Przuntek H (2006) Oral fumaric acid Ansong KA, Masso N, Frosch MP, Kegel KB, Li X, DiFiglia
esters for the treatment of active multiple sclerosis: an open- M (2013) Elevated NADPH oxidase activity contributes to oxi-
label, baseline-controlled pilot study. Eur J Neurol 13:604–610 dative stress and cell death in Huntington’s disease. Hum Mol
Schipper HM (2004) Heme oxygenase expression in human central Genet 22:1112–1131
nervous system disorders. Free Radic Biol Med 37:1995–2011 Valente EM, bou-Sleiman PM, Caputo V, Muqit MM, Harvey K,
Schreibelt G, Musters RJ, Reijerkerk A, de Groot LR, Van Der Pol Gispert S, Ali Z, Del TD, Bentivoglio AR, Healy DG, Alba-
SM, Hendrikx EM, Dopp ED, Dijkstra CD, Drukarch B, De nese A, Nussbaum R, Gonzalez-Maldonado R, Deller T, Salvi
Vries HE (2006) Lipoic acid affects cellular migration into the S, Cortelli P, Gilks WP, Latchman DS, Harvey RJ, Dallapi-
central nervous system and stabilizes blood-brain barrier integ- ccola B, Auburger G, Wood NW (2004) Hereditary early-onset
rity. J Immunol 177:2630–2637 Parkinson’s disease caused by mutations in PINK1. Science
Shcherbatykh I, Carpenter DO (2007) The role of metals in the etiol- 304:1158–1160
ogy of Alzheimer’s disease. J Alzheimers Dis 11:191–205 Van der Goes A, Wouters D, Van Der Pol SM, Huizinga R, Ronken
Shih AY, Imbeault S, Barakauskas V, Erb H, Jiang L, Li P, Murphy E, Adamson P, Greenwood J, Dijkstra CD, De Vries HE (2001)
TH (2005a) Induction of the Nrf2-driven antioxidant response Reactive oxygen species enhance the migration of monocytes
confers neuroprotection during mitochondrial stress in vivo. J across the blood-brain barrier in vitro. FASEB J 15:1852–1854
Biol Chem 280:22925–22936 van Horssen J, Schreibelt G, Drexhage J, Hazes T, Dijkstra CD, Van
Shih AY, Li P, Murphy TH (2005b) A small-molecule-inducible Nrf2- der Valk P, De Vries HE (2008) Severe oxidative damage in
mediated antioxidant response provides effective prophylaxis multiple sclerosis lesions coincides with enhanced antioxidant
against cerebral ischemia in vivo. J Neurosci 25:10321–10335 enzyme expression. Free Radic Biol Med 45:1729–1737
Shimohama S, Tanino H, Kawakami N, Okamura N, Kodama H, van Horssen J, Drexhage JA, Flor T, Gerritsen W, Van der Valk P, De
Yamaguchi T, Hayakawa T, Nunomura A, Chiba S, Perry G, Vries HE (2010) Nrf2 and DJ1 are consistently upregulated in
Smith MA, Fujimoto S (2000) Activation of NADPH oxidase inflammatory multiple sclerosis lesions. Free Radic Biol Med
in Alzheimer’s disease brains. Biochem Biophys Res Commun 49:1283–1289
273:5–9 van Horssen J, Witte ME, Schreibelt G, De Vries HE (2011) Radical
Siddiqui A, Rivera-Sanchez S, Castro MR, cevedo-Torres K, Rane A, changes in multiple sclerosis pathogenesis. Biochim Biophys
Torres-Ramos CA, Nicholls DG, Andersen JK, yala-Torres S Acta 1812:141–150
(2012) Mitochondrial DNA damage is associated with reduced Van Muiswinkel FL, Veerhuis R, Eikelenboom P (1996) Amyloid
mitochondrial bioenergetics in Huntington’s disease. Free Radic beta protein primes cultured rat microglial cells for an enhanced
Biol Med 53:1478–1488 phorbol 12-myristate 13-acetate-induced respiratory burst activ-
Smith MA, Harris PLR, Sayre LM, Beckman JS, Perry G (1997) ity. J Neurochem 66:2468–2476
Widespread peroxynitrite-mediated damage in Alzheimer’s dis- Van Muiswinkel FL, Raupp SF, de Vos NM, Smits HA, Verhoef J,
ease. J Neurosci 17:2653–2657 Eikelenboom P, Nottet HS (1999) The amino-terminus of the
Sofic E, Lange KW, Jellinger K, Riederer P (1992) Reduced and oxi- amyloid-beta protein is critical for the cellular binding and
dized glutathione in the substantia nigra of patients with Parkin- consequent activation of the respiratory burst of human mac-
son’s disease. Neurosci Lett 142:128–130 rophages. J Neuroimmunol 96:121–130
Sofic E, Sapcanin A, Tahirovic I, Gavrankapetanovic I, Jellinger Van Muiswinkel FL, de Vos RA, Bol JG, Andringa G, Jansen Steur
K, Reynolds GP, Tatschner T, Riederer P (2006) Antioxidant EN, Ross D, Siegel D, Drukarch B (2004) Expression of
capacity in postmortem brain tissues of Parkinson’s and Alzhei- NAD(P)H:quinone oxidoreductase in the normal and Parkinso-
mer’s diseases. J Neural Transm Suppl 71:39-43 nian substantia nigra. Neurobiol Aging 25:1253–1262
Spitsin SV, Scott GS, Mikheeva T, Zborek A, Kean RB, Brimer CM, Volkel W, Sicilia T, Pahler A, Gsell W, Tatschner T, Jellinger K, Lebl-
Koprowski H, Hooper DC (2002) Comparison of uric acid and huber F, Riederer P, Lutz WK, Gotz ME (2006) Increased brain
ascorbic acid in protection against EAE. Free Radic Biol Med levels of 4-hydroxy-2-nonenal glutathione conjugates in severe
33:1363–1371 Alzheimer’s disease. Neurochem Int 48:679–686
Springer W, Kahle PJ (2006) Mechanisms and models of alpha-synu- von Otter M, Landgren S, Nilsson S, Celojevic D, Bergstrom P,
clein-related neurodegeneration. Curr Neurol Neurosci Rep Hakansson A, Nissbrandt H, Drozdzik M, Bialecka M, Kurzaw-
6:432–436 ski M, Blennow K, Nilsson M, Hammarsten O, Zetterberg H
Stack C, Ho D, Wille E, Calingasan NY, Williams C, Liby K, Sporn (2010a) Association of Nrf2-encoding NFE2L2 haplotypes with
M, Dumont M, Beal MF (2010) Triterpenoids CDDO-ethyl Parkinson’s disease. BMC Med Genet 11:36
amide and CDDO-trifluoroethyl amide improve the behavioral von Otter M, Landgren S, Nilsson S, Zetterberg M, Celojevic D,
phenotype and brain pathology in a transgenic mouse model of Bergstrom P, Minthon L, Bogdanovic N, Andreasen N, Gus-
Huntington’s disease. Free Radic Biol Med 49:147–158 tafson DR, Skoog I, Wallin A, Tasa G, Blennow K, Nilsson M,
Stoy N, Mackay GM, Forrest CM, Christofides J, Egerton M, Stone Hammarsten O, Zetterberg H (2010b) Nrf2-encoding NFE2L2
TW, Darlington LG (2005) Tryptophan metabolism and oxida- haplotypes influence disease progression but not risk in Alz-
tive stress in patients with Huntington’s disease. J Neurochem heimer’s disease and age-related cataract. Mech Ageing Dev
93:611–623 131:105–110
Sultana R, Perluigi M, Butterfield DA (2006) Protein oxidation and Wagener FA, Volk HD, Willis D, Abraham NG, Soares MP, Adema
lipid peroxidation in brain of subjects with Alzheimer’s disease: GJ, Figdor CG (2003) Different faces of the heme–heme oxyge-
insights into mechanism of neurodegeneration from redox prot- nase system in inflammation. Pharmacol Rev 55:551–571
eomics. Antioxid Redox Signal 8:2021–2037 Walker FO (2007) Huntington’s Disease. Semin Neurol 27:143–150
Thorburne SK, Juurlink BH (1996) Low glutathione and high iron Wallace MN, Geddes JG, Farquhar DA, Masson MR (1997) Nitric
govern the susceptibility of oligodendroglial precursors to oxi- oxide synthase in reactive astrocytes adjacent to beta-amyloid
dative stress. J Neurochem 67:1014–1022 plaques. Exp Neurol 144:266–272
Uversky VN (2007) Neuropathology, biochemistry, and biophysics of Walter P, Ron D (2011) The unfolded protein response: from stress
alpha-synuclein aggregation. J Neurochem 103:17–37 pathway to homeostatic regulation. Science 334:1081–1086

13
1786 Arch Toxicol (2014) 88:1773–1786

Wang CX, Shuaib A (2007) Neuroprotective effects of free radical via an ERK dependent Keap1-Nrf2-ARE pathway. J Neural
scavengers in stroke. Drugs Aging 24:537–546 Transm Suppl 72:57-67
Wang J, Xiong S, Xie C, Markesbery WR, Lovell MA (2005) yala-Pena S (2013) Role of oxidative DNA damage in mitochondrial
Increased oxidative damage in nuclear and mitochondrial DNA dysfunction and Huntington’s disease pathogenesis. Free Radic
in Alzheimer’s disease. J Neurochem 93:953–962 Biol Med 62:102–110
Williamson TP, Amirahmadi S, Joshi G, Kaludov NK, Martinov MN, Yasuhara T, Hara K, Sethi KD, Morgan JC, Borlongan CV (2007)
Johnson DA, Johnson JA (2012) Discovery of potent, novel Increased 8-OHdG levels in the urine, serum, and substantia
Nrf2 inducers via quantum modeling, virtual screening, and in nigra of hemiparkinsonian rats. Brain Res 1133:49–52
vitro experimental validation. Chem Biol Drug Des 80:810–820 Yoritaka A, Hattori N, Uchida K, Tanaka M, Stadtman ER, Mizuno
Witte ME, Bo L, Rodenburg RJ, Belien JA, Musters R, Hazes T, Win- Y (1996) Immunohistochemical detection of 4-hydroxynonenal
tjes LT, Smeitink JA, Geurts JJ, De Vries HE, Van der Valk J, protein adducts in Parkinson disease. Proc Natl Acad Sci USA
van Horssen J (2009) Enhanced number and activity of mito- 93:2696–2701
chondria in multiple sclerosis lesions. J Pathol 219:193–204 Zhao J, Kobori N, Aronowski J, Dash PK (2006) Sulforaphane
Witte ME, Geurts JJ, De Vries HE, Van der Valk J, van Horssen J reduces infarct volume following focal cerebral ischemia in
(2010) Mitochondrial dysfunction: a potential link between rodents. Neurosci Lett 393:108–112
neuroinflammation and neurodegeneration? Mitochondrion Zhao J, Moore AN, Redell JB, Dash PK (2007) Enhancing expression
10:411–418 of Nrf2-driven genes protects the blood brain barrier after brain
Wruck CJ, Claussen M, Fuhrmann G, Romer L, Schulz A, Pufe T, injury. J Neurosci 27:10240–10248
Waetzig V, Peipp M, Herdegen T, Gotz ME (2007) Luteolin
protects rat PC12 and C6 cells against MPP + induced toxicity

13

You might also like