阿尔茨海默病综合概述:病因学见解和减量策略
阿尔茨海默病综合概述:病因学见解和减量策略
Molecular Sciences
Review
Comprehensive Overview of Alzheimer’s Disease: Etiological
Insights and Degradation Strategies
Manish Kumar Singh 1,2 , Yoonhwa Shin 1,2 , Songhyun Ju 1,2,3 , Sunhee Han 1,2,3 , Sung Soo Kim 1,2,3, *
and Insug Kang 1,2,3, *
1 Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University,
Seoul 02447, Republic of Korea; [email protected] (M.K.S.); [email protected] (Y.S.);
[email protected] (S.J.); [email protected] (S.H.)
2 Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
3 Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
* Correspondence: [email protected] (S.S.K.); [email protected] (I.K.);
Tel.: +82-2-961-0524 (S.S.K.); +82-2-961-0922 (I.K.)
Abstract: Alzheimer’s disease (AD) is the most prevalent neurodegenerative disorder and affects
millions of individuals globally. AD is associated with cognitive decline and memory loss that
worsens with aging. A statistical report using U.S. data on AD estimates that approximately 6.9 million
individuals suffer from AD, a number projected to surge to 13.8 million by 2060. Thus, there is a
critical imperative to pinpoint and address AD and its hallmark tau protein aggregation early to
prevent and manage its debilitating effects. Amyloid-β and tau proteins are primarily associated
with the formation of plaques and neurofibril tangles in the brain. Current research efforts focus on
degrading amyloid-β and tau or inhibiting their synthesis, particularly targeting APP processing and
tau hyperphosphorylation, aiming to develop effective clinical interventions. However, navigating
this intricate landscape requires ongoing studies and clinical trials to develop treatments that truly
make a difference. Genome-wide association studies (GWASs) across various cohorts identified
40 loci and over 300 genes associated with AD. Despite this wealth of genetic data, much remains to
be understood about the functions of these genes and their role in the disease process, prompting
continued investigation. By delving deeper into these genetic associations, novel targets such as
Citation: Singh, M.K.; Shin, Y.; Ju, S.;
kinases, proteases, cytokines, and degradation pathways, offer new directions for drug discovery and
Han, S.; Kim, S.S.; Kang, I.
therapeutic intervention in AD. This review delves into the intricate biological pathways disrupted in
Comprehensive Overview of
AD and identifies how genetic variations within these pathways could serve as potential targets for
Alzheimer’s Disease: Etiological
Insights and Degradation Strategies.
drug discovery and treatment strategies. Through a comprehensive understanding of the molecular
Int. J. Mol. Sci. 2024, 25, 6901. https:// underpinnings of AD, researchers aim to pave the way for more effective therapies that can alleviate
doi.org/10.3390/ijms25136901 the burden of this devastating disease.
the synaptic connections among them. Molecular and genetic studies in AD patients
have identified three primary genetic mutations associated with Early-Onset Alzheimer’s
Disease (EOAD), linked with genes encoding amyloid precursor protein (APP), presenilin
1 (PSEN1), and presenilin 2 (PSEN2) [8]. However, the APOEε4 allele is considered the
predominant genetic risk factor for AD, notably in sporadic AD cases referred to as Late-
Onset Alzheimer’s Disease (LOAD). Additionally, variations in CD33 splicing play a
significant role in altering and influencing this genetic predisposition [9,10]. Several tests
used for diagnosing AD clinically include brain imaging techniques such as positron
emission tomography (PET), cerebrospinal fluid (CSF)-based biomarkers like Aβ1–42,
p-Tau, and neurogranin-a synaptic protein, as well as blood test-based biomarkers such
as proinflammatory cytokines including IL-6, ICAM-1, and VCAM-1 [11]. Furthermore,
genome-wide association studies (GWASs) conducted across diverse populations have
implicated rare non-synonymous variants like TREM2, PLCG2, and ABI3 in LOAD [12].
Additionally, inflammatory pathways and various neuroinflammation-modulated signaling
pathways play significant roles in exacerbating Aβ toxicity and AD pathology. Thus,
understanding these genetic and molecular intricacies is vital for developing targeted
therapies aimed at mitigating the devastating effects of AD on cognitive function and
quality of life [13].
Existing evidence has revealed that various enzymes are dysregulated in AD, which is
involved in the cleavage of amyloid precursor protein (APP) and tau hyperphosphoryla-
tion, resulting in increased levels of amyloid-β and tau fibrils in neurons and, ultimately,
neurodegeneration. This process includes numerous endosomal–lysosomal proteases, such
as cathepsins, calpains, caspases, and matrix metalloproteinases (MMPs), which exhibit
a crucial role in tau cleavage and contribute to AD pathology [14]. Consequently, several
small molecule inhibitors targeting these proteases and kinases are currently under clinical
trials for treatment, aiming to inhibit the aggregation of amyloid-β and tau fibrils in AD
patients. Here, we comprehensively elucidate the important genetic factors and degra-
dation pathways implicated in the prevention of AD. Our object is to shed light on the
genetic risk factors, both high and low, and signaling pathways involved in Aβ cytotoxicity.
Additionally, we examine key degradation pathways, including the Receptor for Advance
Glycation End Product (RAGE), Endoplasmic Reticulum-Associated Degradation (ERAD),
and autophagy, which play crucial roles in the clearance of toxic Aβ plaques and NFTs
from the brain. We aim to highlight the genetic risk factors and neuronal markers that
warrant further investigation as potential early biomarkers and therapeutic targets that can
be utilized in the development of therapeutics and strategies to manage the progression of
AD effectively.
2. Methods
This review outlines potential strategies based on GWAS data in AD. We reviewed an
enormous amount of the literature on GWASs derived from transcriptomic, expression QTL
(eQTL), methylation QTL (mQTL), and methylome data. Identifying potential candidate
SNPs as risk factors for disease etiology was challenging because the significance threshold
(p-value < 0.05) was difficult to meet. Understanding the causal genetic variants and genes
influencing AD risk on susceptible loci remains limited, with only a few genes extensively
studied in AD patients. Despite a GWAS with a large population size identifying a locus
covering 3% of AD cases, attributing heritability to these loci is still ambiguous.
It was difficult to provide detailed information about each genetic locus, the af-
fected genes, and subsequent proteins associated with AD. This review encompassed
data from the last two and a half decades, sourced from the AD forum and the World
Health Organization (WHO). These data were collected through extensive searches of
online databases from January 2021 onwards such as https://www.alzforum.org, https:
//forum.alzheimers.org.uk, and https://www.who.int/data for gathering literature. For
research articles, Medline https://www.nlm.nih.gov/medline, https://clarivate.com/
webofscience-medline, PubMed https://pubmed.ncbi.nlm.nih.gov, and Google Scholar
Int. J. Mol. Sci. 2024, 25, 6901 3 of 28
https://scholar.google.com databases were used to find the most relevant and recent pub-
lications. The most important and recent research articles from each relevant author in
AD and tauopathy were included. The collected research allowed the inclusion of detailed
information related to GWASs across various populations, focusing on the most relevant
genes and data related to mechanisms affecting Aβ production and clearance of Aβ. Data
not directly linked with AD but rather other forms of dementia were excluded. This review
primarily focuses on the disease etiology based on GWASs and degradation pathways for
early biomarkers and therapeutic perspectives. A detailed discussion of each section with
significant mechanisms is provided, and references are cited for all encoded statements in
this review.
Figure 1. This image illustrates the processing of amyloid precursor protein (APP) and the subsequent
formation of amyloid plaques (A) as well as tau hyperphosphorylation and the formation of neurofib-
rillary tangles (B). (A). Processing of APP involves two pathways including the non-amyloidogenic
and amyloidogenic pathways. In the non-amyloidogenic pathway, α-secretase cleaves APP, while in
the amyloidogenic pathway, β-secretase and γ-secretase are involved in the cleavage process, leading
to the release of Aβ into the extracellular space. The initial cleavage of APP by proteases releases the
APP intracellular domain (AICD) into the intracellular space. (B). The tau protein normally binds to
microtubules, stabilizing their structure. However, hyperphosphorylation of tau leads to the release of
tau filaments and destabilization of the microtubule structure. This results in the formation of various
tau aggregates, including dimers, oligomers, paired helical filaments (PHFs), and neurofibrillary
tangles (NFTs). Adapted from Ref [17].
The APOE-ε4 gene significantly affects the splicing of CD33, while APOE2 gene
variants are associated with the regulation of HMOX-1 expression and possess antioxidant
properties relevant to AD [27]. APOE2 carriers show increased levels of SLAM family
member 8 (SLAMF8), a CD2 family member, implicated in modulating reactive oxygen
species and inflammation in the brain, as well as influencing macrophage function and
supporting the growth of neoplastic mast cells via SHP-2 [28]. Recent findings have
demonstrated that APOEε4 loss-of-function is associated with a high risk of AD. However
genetic analysis has revealed that ε4 drives AD risk via a gain in abnormal function rather
than a loss of function in AD pathogenesis. Supporting this hypothesis, studies have found
that APOEε4 increases tauopathy and neurodegeneration by promoting lipid accumulation
and impairing cholesterol metabolism in a tauopathy model [24,29]. The precise role of
APOE in AD pathogenesis remains elusive, and further studies are warranted to investigate
this potential mechanism in large populations carrying APOE mutations.
Tauopathies manifest as progressive cognitive and motor impairment, encompassing
conditions such as progressive supranuclear palsy, corticobasal degeneration, chronic trau-
matic encephalopathy, and certain forms of frontotemporal dementia. Genetic evidence
from model organisms like mice suggests that tau plays a pivotal role in age-related neu-
rodegeneration. However, fewer studies have demonstrated tau protein’s involvement in
progressive synaptic dysfunction and neuronal loss, likely stemming from various cellu-
lar derangements including oxidative and immune-mediated injury, altered proteostasis,
aberrant transcription, and post-translational modifications [18]. Tau, primarily localized
Int. J. Mol. Sci. 2024, 25, 6901 5 of 28
in the axons of healthy neurons, relocates to the soma and dendrites under pathological
conditions [30]. Axons play a crucial role in maintaining neuronal function by facilitating
anterograde and retrograde transport of cargo [31]. The mechanisms underlying tau seed-
ing and spreading remain elusive, but some in vivo studies suggest the involvement of
either exocytosis through vesicles or inflammation. The relevance of these mechanisms in
AD pathogenesis remains unclear. Studies in transgenic mice have linked the tau-mediated
reduction of kinesin-1 light chain to impaired anterograde axonal trafficking [32]. In AD,
hyperphosphorylation of tau increases, leading to the formation of neurofibrillary tangles
(NFTs) [11]. Tau-induced neuronal damage initiates in the entorhinal cortex and progres-
sively spreads to the hippocampus and other cortical regions. Later, tau oligomers spread
to other areas of the brain such as the corpus callosum and the mediolateral axis [33,34].
Additionally, extracellular oligomeric tau has been implicated in memory impairment
and cognitive dysfunction in mice [35]. Mutations in MAPT genes are reported in fa-
milial frontotemporal dementia (FTD), characterized by prominent neurofibrillary tangle
deposition [18].
Several studies have highlighted the cerebrospinal fluid (CSF) tau level as an early
indicator of Aβ pathogenesis. However, plasma Aβ also changes in AD patients compared
with controls, and the difference between amyloid-positive and -negative individuals is a
relatively small-fold change [36]. Recent research, utilizing positron emission tomography
(PET) analysis on CSF samples from Alzheimer’s disease (AD) patients, identified three
distinct phosphorylated tau (p-tau) epitopes, including p-tau 181, p-tau 217, and p-tau 231,
evident across clinical, preclinical, and pre-amyloid phases of AD [37,38]. Among these,
p-tau 231 exhibited a significant increase, accumulating notably in brain regions associated
with the default mode network, such as the precuneus, posterior cingulate cortex, and
orbitofrontal cortex [39]. As a result, cerebrospinal fluid (CSF) and brain regions affected
early in AD progression, such as the cortex and hippocampus, indicate that CSF p-tau
levels and Aβ PET are pivotal markers for the early detection of AD [40]. A parallel study
revealed elevated p-tau 217 and p-tau 181 levels in aggregated Aβ, detectable preceding the
onset of aggregated tau pathology [41]. This finding was supported by another study where
p-tau 181 was elevated in the CSF of patients with amyloid pathology and Braak NFT ≥ III
detected by Aβ and Tau PET screening in early AD stages, confirming an escalating trend
with disease progression [42,43]. Notably, 18F-fluorodeoxyglucose (FDG) PET has been
employed to differentiate between typical and atypical AD based on distinctive frontal and
parietal atrophy and hypometabolism [44]. Moreover, the levels of p-tau forms such as
p-tau 205 and total tau surged with the emergence of LOAD symptoms [45]. Notably, Phan
and Cho developed aptamer-mediated biosensors tailored to specifically detect p-tau at
threonine 231, a crucial early p-tau isotope frequently utilized in the diagnosis of AD [46].
Additionally, Gonzalez-Ortiz et al. demonstrated that plasma brain-derived tau (BD-Tau)
with p-tau alongside Aβ42/Aβ40 as a blood-based based-biomarker for diagnosis of AD.
This approach enhances agreement with autopsy results or those derived from CSF or
neuroimaging biomarkers [47,48]. Other blood biomarkers for AD including GFAP and β-
synuclein are significantly observed in AD [49,50]. Various studies have demonstrated that
combined biomarkers provide better diagnostic accuracy for AD than individual measures
alone [51]. For instance, the combination of three plasma biomarkers such as APP669–711
with Aβ and p-tau217, plasma Aβ42/Aβ40, and plasma NFL showed improved diagnostic
performance when the APOE genotype was included [52–54]. Further studies that analyze
combined biomarkers, including plasma Aβ42/Aβ40 and other measurements, may confer
even more accurate diagnoses from blood samples, representing a valuable avenue for
future investigation.
Additionally, modifiers of p-tau levels, such as the α7 subtype of nicotinic acetyl-
choline receptors (α7nAChRs), were found to induce tau phosphorylation at specific sites
(Ser 199, Ser 396, and Thr 205) in the hippocampus of 12-month-old α7 knockout mice.
Interestingly, these mice exhibited increased levels of APP and Aβ without the formation
of senile plaques, suggesting a potential role for α7nAChRs within the tripartite interplay
Int. J. Mol. Sci. 2024, 25, 6901 6 of 28
involving α7nAChRs, Aβ, and tau [55]. Increased oxidative stress leads to elevated level
of GSK-3β and protein kinase, and phosphatase PP2A activates tau phosphorylation at
ser396, ser404, and thr231. In the AD hippocampus, the decreased expression of peptidyl
prolyl cis-transferase (PIN1) and increased p-tau level act as an additive factor in AD
pathogenesis [56]. This observation is reinforced by subsequent experiments using cortical
neuronal cells with H2 O2 , which results in the activation of GSK-3β and PIN1. Thus, kinase
inhibitors could be beneficial in ameliorating p-tau in AD. Some kinase inhibitors including
Tideglusib and lithium, are utilized to modulate GSK-3β activity, although clinical trials
are ongoing in AD and other types of dementia [57]. Therefore, further investigations are
warranted to gain deeper insights into the pathological and non-pathological conditions of
p-tau and NFT production in AD and related tauopathies.
3.2. GWAS and Genetic Risk Factors Associated with AD and Synaptic Plasticity
High-throughput genomic analysis such as genome-wide association studies (GWASs)
identified several genes linked with disease progression and implicated in synaptic dysfunc-
tion and memory impairment [58]. GWAS screening primarily aims to identify susceptible
genes and investigate their impact on AD pathology and early detection. These studies
provide a novel approach to identifying genetic variations associated with AD, with a sig-
nificance threshold (p-value ≤ 0.05) for single nucleotide polymorphisms (SNPs), indicating
a potential risk factor for AD [59]. GWASs are implicated in early biomarkers for various
diseases, including cancers and age-related disorders [60,61]. A recent study utilizing the
U.K. Biobank for AD familial data, known as GWAS-by-proxy, uncovered 12 genomic loci
associated with LOAD [12]. However, our understanding of the causal genetic variants and
genes influencing AD risk at these loci remains limited, with only a few genes extensively
studied in AD patients.
Despite a large population size exceeding more than 1.1 million individuals and
the identification of 38 independent genome-wide significant loci, only 3% of AD cases
can be attributed to heritability [62,63]. Notably, novel loci such as rs5011804 at 12p12.1
have shown significant associations with the levels of CDRSB, FAQ, FS fusiform, and
ADAS13 in various AD cognitive and neuroimaging analyses, underscoring the necessity
for multiple measurements such as neuroimaging data (MRI), fluid biomarkers (blood
and CSF), cognitive impairment/dementia, and familial data to establish SNP association
as an AD risk factor [64,65]. New AD risk loci have been identified through higher-
density genotype imputation, shedding light on candidate causal variants at both new and
established risk loci. A GWAS meta-analysis also revealed early-age risk factors in AD
brain and lymph node samples such as SORL1, PTK2B, SLC24A4, and ZCWPW1 associated
with AD. It is necessary to conduct thorough sequencing studies and in-depth post-GWAS
analyses to fully understand the candidate genes and functional variants associated with
AD susceptibility. These investigations are crucial in order to determine the functional
significance of the identified loci in the pathophysiology of AD [12,66,67]. Furthermore, a
robust association was observed between the NYAP1 SNP and PILRA/PILRB protein in the
brain, with implications for regulating acute inflammatory reactions in the AD brain [28,68].
Numerous other candidate genes identified in GWASs as AD risk factors are local-
ized on different loci, such as rs6705798 encoding EPS15-homology domain-containing
protein1/2 (EHBP1), implicated in Glut4 transportation and expressed in various tissues
including the brain. Another SNP, rs73045836, encoding secreted extracellular calcium-
binding protein 2 (SMOC2) and its isoform SMOC1, has been identified as a novel biomarker
for AD in CSF and brain tissues [26]. In addition to APOE4, other genes associated with
AD risk, such as CLU, PICALM, CD33, MS4A4, MS4A6A, TREM2, ABCA7, CD2AP, and
EPHA1, have been identified in GWASs using meta-analysis methods involving both AD
patients and non-AD populations (Table 1). Among these AD risk genes, the myeloid cell
surface antigen CD33, rs3865444, and rs3836656 are linked to microglia-mediated clearance,
potentially promoting the accumulation of senile plaques [69,70]. Clusterin (CLU), also
known as apolipoprotein J (ApoJ), is a significant risk gene associated with AD that resides
Int. J. Mol. Sci. 2024, 25, 6901 7 of 28
in the central nervous system. It is synthesized and released by both astrocytes and neu-
rons in the brain, where it plays a role in regulating lipid metabolism. In the Caucasian
population, CLU variants, such as rs93331888 and rs11136000, have been identified [71].
However, in the Asian population no significant associations have been observed, indicat-
ing a notable variation in the impact of CLU on AD. Recently, additional candidate loci
including BIN1, TREM2, SORL1, MS4A, SPI1, and TOMM40 have also been identified
via GWAS meta-analysis (Table 1). These genes demand continued attention and further
exploration in the forthcoming studies [70].
Moreover, non-synonymous gene variants are significantly associated with trait as-
sociations, with most human trait-associated variants affecting gene expression rather
than altering protein-coding sequences. These variants likely mediate their effects via
altered gene expression, which may vary depending on cell type [2]. Functional map-
ping of variants to genes, utilizing positional data and expression quantitative trait loci
(eQTL) information from brain and immune tissues/cells, unveiled 989 genes linked to
38 genomic risk loci. These genes predominantly pertain to inflammatory signaling and
are associated with immune cells such as microglia, astrocytes, and oligodendrocytes in
LOAD patients [60,63]. GWASs identify intergenic regions, wherein all protein-coding
genes within 500 kilobases (kb) of the sentinel variant linkage disequilibrium (LD) region
(r2 > 0.5) are implicated as potential AD risk factors.
Transcriptomic analyses and whole-genome sequencing of brain samples from AD
patients have revealed alterations in the expression of genes involved in various signaling
pathways. Several mutations and loci associated with AD have been identified through
GWASs and collaborations such as the International Genomics of Alzheimer’s Project
(IGAP). Over the past two decades, at least 38 loci and more than 300 gene mutations
have been linked to AD. Some of the frequently mutated genes in AD cases include
CD2AP, APOEε4, PTK2B, CASS4, EPHA1, Zyxin, PACSIN, CD33, and CYP3A (Table 1) [60].
Recent analyses combining eQTL and expression transcriptome-wide association studies
(eTWASs) have revealed that the downregulation of EGFR is significantly associated with
reduced risk of AD [72]. Conversely, colocalization of eQTL-GWAS and methylation QTL
(mQTL) signals has identified TSPAN14, derived from lymphoblastoid cell lines, as being
correlated with AD risk [72]. Additionally, pathway enrichment analysis using STRING
has identified immune and tumor necrosis factor (TNF)-mediated signaling pathways,
involving genes such as SHARPIN, RBCK1, and LUBAC, regulated by OTULIN, as having
strong associations with AD risk [72,73].
A study on the Mexican American population with mild cognitive impairment (MCI)
revealed various methylation regions between control and MCI patients along with genes
implicated in neuronal death, metabolic dysfunction, and inflammatory processes [74].
Numerous environmental factors are associated with epigenetic modification and can be in-
herited. For instance, exposure to organophosphate pesticides has been shown to promote
tau hyperphosphorylation and microtubule dysfunction [75,76]. The methylation of genes,
such as ABCA7, BIN1, SORL1, and SLC24A4, is significantly associated with the patho-
logical processing of the tau protein and Aβ peptide in the dorsolateral prefrontal cortex.
Additionally, histone acetyltransferase and histone deacetylase inhibitors elevate histone
acetylation, thereby exerting various positive effects on AD including preventing memory
impairment, cognitive dysfunction, less deposition of the Aβ peptide, and reduced tau
phosphorylation and formation of NFTs [77]. Therefore, identifying polymorphisms associ-
ated with multiple environmental factors through GWASs could facilitate the development
of effective diagnostic and therapeutic strategies.
Furthermore, GWAS data serve as a valuable tool for understanding how AD progres-
sion is connected to the development of other diseases. Post-GWAS functional genomic
analysis is required to prioritize genes that modulate disease susceptibility and identify
candidate causal genes for further functional validation in AD animal models [78]. For
instance, Lee and colleagues used blood samples from AD patient and found associations
between the expression of GPBP1, also known as Vasculin, in both vascular wall and plasma,
Int. J. Mol. Sci. 2024, 25, 6901 8 of 28
Table 1. Cont.
Figure 2. This image depicts various biological processes affected by AD. It illustrates increased
amyloid-β plaques, tau hyperphosphorylation, elevated ROS levels, and impaired amyloid-β clear-
ance pathways, along with metabolic abnormalities. Reduced degradation, heightened lipid oxidation
and metabolite accumulation, mitochondrial dysfunction, and the aging process contribute to the
progression of AD. Vascular impairments and defects in the exosome pathway heighten the risk
of amyloid-β accumulation, while calcium release and metal dyshomeostasis exacerbate cognitive
impairment and neuronal cell death.
GWASs focus on investigating potential risk factors associated with AD using clinical
samples, leading to an increase in the number of loci and identification of new risk factors,
SNPs, and mutations significantly associated with AD. However, these GWAS-identified
Int. J. Mol. Sci. 2024, 25, 6901 11 of 28
Figure 3. This image represents different conditions of AD in normal (A), AD (B), and treatment
conditions (C). The image illustrates the imbalance in Aβ formation and clearance of Aβ aggregates
in human brains over the course of aging. The equilibrium state depicts a balance between Aβ
production and Aβ clearance. However, in the AD condition, there is an increase in Aβ production
and decreased Aβ clearance, which contribute to disease progression. AD treatment aims to restore
brain functions by reducing Aβ aggregate formation and inducing Aβ clearance via various treatment
strategies such as induced autophagy, ubiquitination, immunotherapy, small molecule therapy, and
degradation of Aβ and NFTs.
Figure 4. This image illustrates various factors associated with AD and highlights the impact of
several stressors on a healthy brain and the subsequent consequences that lead to disease progression.
GWASs and related genetic factors, as well as specific pathways, are shown to ameliorate disease
conditions. Targeting different pathways can slow disease progression and improve cognitive
functions. However, genetic manipulations and gene therapy for AD treatment still require extensive
in vivo testing. This figure also depicts strategies to restore brain functions by reducing Aβ aggregates
and promoting Aβ clearance through induced autophagy, UPS, RAGE, and ERAD. Additionally,
immunotherapy and small molecule therapies are illustrated as approaches to reduce inflammation
and maintain energy homeostasis, ultimately slowing AD progression. Red ‘?’ shows the effects are
remains exclusive.
Prominent genetic risk factors associated with Alzheimer’s disease (AD), along with
numerous molecular factors, play a crucial role in the early detection of AD risk genes.
Int. J. Mol. Sci. 2024, 25, 6901 18 of 28
Additionally, genes exhibiting common variants in AD and other types of dementia and
neurodegeneration may establish an earlier link with AD progression, warranting in-
depth investigation to determine the precise time point markers at which they intersect.
Autophagy and ERAD pathways emerge as potential therapeutic avenues for managing
neurodegenerative diseases, including AD. Understanding the specific proteolytic processes
involved in the processing of proteins like APP and tau is paramount for unraveling the
molecular mechanisms underlying AD.
In AD, metabolic dysregulation plays a critical role in disease progression and synaptic
dysfunction. T2D, which is characterized by hyperglycemia and IR, affects brain glucose
levels and promotes the accumulation of Aβ in the brain [216]. T2D is recognized as a risk
factor for the progression of AD. However, the mechanism by which IR influences glucose
metabolism, including its effects on memory and synapse plasticity, remains inadequately
explored. Impaired glucose metabolism and the resulting formation of Aβ plaques and
NFTs induce oxidative stress, leading to elevated intracellular free Ca2+ levels and trig-
gering a cascade of detrimental effects that culminate in neuronal death [217]. Therefore,
maintaining optimal brain glucose metabolism could mitigate oxidative damage, reduce
the level of reactive oxygen species (ROS) and reactive nitrogen species (RNS), and protect
the brain from adverse effects, thereby slowing disease progression. Continued research
into glucose metabolism, oxidative stress, and mitochondrial dysfunction is essential for a
more comprehensive understanding of AD pathogenesis and progression. These studies
will also enhance the ability to monitor the therapeutic efficacy of novel drugs and small
molecules targeting Aβ production and synaptic dysfunction, ultimately aiming to prevent
neuronal cell death [218].
Furthermore, defective lysosomal acidification plays a crucial role in AD pathogen-
esis and progression. Impaired functions of microglia and astrocytes contribute to the
accumulation and insufficient clearance of Aβ plaques and NFTs, leading to enhanced
neuroinflammation and ultimately neuronal cell death. Lysosomal acidification defects
and impaired glial functions occur early in the disease process, contributing to subsequent
neuronal dysfunction. Therefore, the early detection of lysosomal acidification dysfunction
and the development of therapeutic agents to restore lysosomal function are essential
for effective AD therapy. Non-invasive, real-time detection methods would significantly
advance this therapeutic approach [161,169]. Recent reports indicate that autolysosome
acidification declines in neurons well before extracellular amyloid deposition, characterized
by significantly reduced V-ATPase activity and the accumulation of Aβ/APP-βCTF within
enlarged deacidified autolysosomes. These profuse Aβ-positive autophagic vacuoles (AVs)
cluster into large membrane blebs, forming flower-like perikaryal rosettes known as PAN-
THOS, which are observed in AD brains. These AVs merge into perinuclear networks
of membrane tubules where fibrillar β-amyloid accumulates intraluminal. This process
leads to lysosomal membrane permeabilization, cathepsin release, and lysosomal cell
death, followed by microglial invasion and phagocytosis. Additionally, neurons exhibiting
PANTHOS are identified as the primary source of senile plaques in APP-associated AD
models. This hypothesis suggests that the early detection of these molecular markers could
serve as early diagnosis biomarkers for Alzheimer’s disease and aid in the development of
therapeutic agents [219].
Moreover, the role of autophagy and ERAD in neurodegenerative diseases remains
an active and evolving field of study. Optimizing pharmacological agents, such as small
molecules and nanomedicines, for clinical application is critical. This includes enhancing
their properties for better efficacy, bioavailability, and safety in therapeutic use [169].
Notably, these research strategies may provide deeper insights into the roles of specific
proteases within various cellular compartments, aiding in the development of targeted
therapies to inhibit plaque formation and prevent the development of NFTs. Future research
should prioritize obtaining more population-based GWAS data from diverse cohorts and
clinical samples. This approach will be crucial for identifying novel AD risk factors and
potential targets for innovative drug therapies and treatment strategies.
Int. J. Mol. Sci. 2024, 25, 6901 19 of 28
Author Contributions: Conceptualization, M.K.S., I.K. and S.S.K.; resources, M.K.S., S.J., S.H. and
Y.S.; writing—original draft preparation, M.K.S. and S.H.; figures, M.K.S. and S.J.; review and
editing, M.K.S., Y.S. and S.H; supervision, M.K.S., S.S.K. and I.K.; project administration, S.S.K.
and I.K.; funding acquisition, S.S.K. All authors have read and agreed to the published version of
the manuscript.
Funding: This study was supported by the National Research Foundation of Korea (NRF) grants
funded by the Korean government (MEST) (grant NRF-2018R1A6A1A03025124).
Conflicts of Interest: The authors declare no conflicts of interest.
Abbreviations
AD Alzheimer’s disease
APP amyloid precursor protein
APH1 anterior pharynx defective 1
BACE1 β-site APP-cleaving enzyme-1
BBB blood–brain barrier
CSF cerebrospinal fluid
EOAD Early-Onset Alzheimer’s Disease
ER endoplasmic reticulum
ERAD ER-associated protein degradation
eQTL expression quantitative trait loci
eTWASs expression transcriptome-wide association studies
FTD familial frontotemporal dementia
GWASs genome-wide association studies
IGAP International Genomics of Alzheimer’s Project
LOAD late-onset Alzheimer’s disease
LRP-1 LDL receptor-related protein-1
LD linkage disequilibrium
MMPs matrix metalloproteinases
MAPT microtubule-associated protein
mQTL methylation QTL
NFTs neurofibrillary tangles
PICALM phosphatidylinositol-binding clathrin assembly
RAGE Receptor for Advanced Glycation End Product
SRP14 Single Recognition Particle 14
α7nAChRs α7-Nicotinic Acetylcholine Receptors
SNPs single nucleotide polymorphisms
TNF tumor necrosis factor
TREM2 triggering receptor expressed on myeloid cells 2
UPR unfolded protein response
UPS ubiquitous proteasomal system
XBP1 X-box binding protein 1
References
1. Breijyeh, Z.; Karaman, R. Comprehensive review on alzheimer’s disease: Causes and treatment. Molecules 2020, 25, 5789.
[CrossRef] [PubMed]
2. Knopman, D.S.; Amieva, H.; Petersen, R.C.; Chételat, G.; Holtzman, D.M.; Hyman, B.T.; Nixon, R.A.; Jones, D.T. Alzheimer
disease. Nat. Rev. Dis. Primers 2021, 7, 1–21. [CrossRef] [PubMed]
3. Wang, Y.; Mandelkow, E. Tau in physiology and pathology. Nat. Rev. Neurosci. 2016, 17, 22–35. [CrossRef]
4. Zhou, L.; McInnes, J.; Wierda, K.; Holt, M.; Herrmann, A.G.; Jackson, R.J.; Wang, Y.-C.; Swerts, J.; Beyens, J.; Miskiewicz, K.; et al.
Tau association with synaptic vesicles causes presynaptic dysfunction. Nat. Commun. 2017, 8, 15295. [CrossRef]
5. Better, M.A. 2024 alzheimer’s disease facts and figures. Alzheimer’s Dement. 2024, 20, 3708–3821.
6. Hanger, D.P.; Byers, H.L.; Wray, S.; Leung, K.-Y.; Saxton, M.J.; Seereeram, A.; Reynolds, C.H.; Ward, M.A.; Anderton, B.H. Novel
phosphorylation sites in tau from alzheimer brain support a role for casein kinase 1 in disease pathogenesis. J. Biol. Chem. 2007,
282, 23645–23654. [CrossRef]
7. Ittner, A.; Chua, S.W.; Bertz, J.; Volkerling, A.; Van Der Hoven, J.; Gladbach, A.; Przybyla, M.; Bi, M.; Van Hummel, A.; Stevens,
C.H. Site-specific phosphorylation of tau inhibits amyloid-β toxicity in alzheimer’s mice. Science 2016, 354, 904–908. [CrossRef]
Int. J. Mol. Sci. 2024, 25, 6901 20 of 28
8. Andrade-Guerrero, J.; Santiago-Balmaseda, A.; Jeronimo-Aguilar, P.; Vargas-Rodríguez, I.; Cadena-Suárez, A.R.; Sánchez-Garibay,
C.; Pozo-Molina, G.; Méndez-Catalá, C.F.; Cardenas-Aguayo, M.-d.-C.; Diaz-Cintra, S. Alzheimer’s disease: An updated overview
of its genetics. Int. J. Mol. Sci. 2023, 24, 3754. [CrossRef]
9. Kim, J.; Basak, J.M.; Holtzman, D.M. The role of apolipoprotein e in alzheimer’s disease. Neuron 2009, 63, 287–303. [CrossRef]
10. Liu, C.-C.; Zhao, N.; Fu, Y.; Wang, N.; Linares, C.; Tsai, C.-W.; Bu, G. Apoe4 accelerates early seeding of amyloid pathology.
Neuron 2017, 96, 1024–1032.e3. [CrossRef]
11. Nystuen, K.L.; McNamee, S.M.; Akula, M.; Holton, K.M.; DeAngelis, M.M.; Haider, N.B. Alzheimer’s disease: Models and
molecular mechanisms informing disease and treatments. Bioengineering 2024, 11, 45. [CrossRef] [PubMed]
12. Jansen, I.E.; Savage, J.E.; Watanabe, K.; Bryois, J.; Williams, D.M.; Steinberg, S.; Sealock, J.; Karlsson, I.K.; Hägg, S.; Athanasiu, L.
Genome-wide meta-analysis identifies new loci and functional pathways influencing alzheimer’s disease risk. Nat. Genet. 2019,
51, 404–413. [CrossRef] [PubMed]
13. Takatori, S.; Wang, W.; Iguchi, A.; Tomita, T. Genetic risk factors for alzheimer disease: Emerging roles of microglia in disease
pathomechanisms. In Reviews on Biomarker Studies in Psychiatric and Neurodegenerative Disorders; Springer: Cham, Switzerland,
2019; pp. 83–116.
14. J Baranello, R.; L Bharani, K.; Padmaraju, V.; Chopra, N.; K Lahiri, D.; H Greig, N.; A Pappolla, M.; Sambamurti, K. Amyloid-beta
protein clearance and degradation (abcd) pathways and their role in alzheimer’s disease. Curr. Alzheimer Res. 2015, 12, 32–46.
[CrossRef] [PubMed]
15. Vassar, R.; Kovacs, D.M.; Yan, R.; Wong, P.C. The β-secretase enzyme bace in health and alzheimer’s disease: Regulation, cell
biology, function, and therapeutic potential. J. Neurosci. 2009, 29, 12787–12794. [CrossRef] [PubMed]
16. Kimberly, W.T.; LaVoie, M.J.; Ostaszewski, B.L.; Ye, W.; Wolfe, M.S.; Selkoe, D.J. Γ-secretase is a membrane protein complex
comprised of presenilin, nicastrin, aph-1, and pen-2. Proc. Natl. Acad. Sci. USA 2003, 100, 6382–6387. [CrossRef]
17. Hampel, H.; Hardy, J.; Blennow, K.; Chen, C.; Perry, G.; Kim, S.H.; Villemagne, V.L.; Aisen, P.; Vendruscolo, M.; Iwatsubo, T.; et al.
The amyloid-β pathway in alzheimer’s disease. Mol. Psychiatry 2021, 26, 5481–5503. [CrossRef] [PubMed]
18. Li, C.; Götz, J. Tau-based therapies in neurodegeneration: Opportunities and challenges. Nat. Rev. Drug Discov. 2017, 16, 863–883.
[CrossRef] [PubMed]
19. Nozal, V.; Martinez, A. Tau tubulin kinase 1 (ttbk1), a new player in the fight against neurodegenerative diseases. Eur. J. Med.
Chem. 2019, 161, 39–47. [CrossRef] [PubMed]
20. Monteiro, A.R.; Barbosa, D.J.; Remião, F.; Silva, R. Alzheimer’s disease: Insights and new prospects in disease pathophysiology,
biomarkers and disease-modifying drugs. Biochem. Pharmacol. 2023, 211, 115522. [CrossRef]
21. Li, Z.; Shue, F.; Zhao, N.; Shinohara, M.; Bu, G. Apoe2: Protective mechanism and therapeutic implications for alzheimer’s
disease. Mol. Neurodegener. 2020, 15, 63. [CrossRef]
22. Shinohara, M.; Kanekiyo, T.; Tachibana, M.; Kurti, A.; Shinohara, M.; Fu, Y.; Zhao, J.; Han, X.; Sullivan, P.M.; Rebeck, G.W. Apoe2
is associated with longevity independent of alzheimer’s disease. Elife 2020, 9, e62199. [CrossRef] [PubMed]
23. Arboleda-Velasquez, J.F.; Lopera, F.; O’Hare, M.; Delgado-Tirado, S.; Marino, C.; Chmielewska, N.; Saez-Torres, K.L.; Amarnani,
D.; Schultz, A.P.; Sperling, R.A. Resistance to autosomal dominant alzheimer’s disease in an apoe3 christchurch homozygote: A
case report. Nat. Med. 2019, 25, 1680–1683. [CrossRef] [PubMed]
24. Chemparathy, A.; Le Guen, Y.; Chen, S.; Lee, E.-G.; Leong, L.; Gorzynski, J.E.; Jensen, T.D.; Ferrasse, A.; Xu, G.; Xiang, H. Apoe
loss-of-function variants: Compatible with longevity and associated with resistance to alzheimer’s disease pathology. Neuron
2023, 112, 1110–1116.e5. [CrossRef] [PubMed]
25. Parhizkar, S.; Holtzman, D.M. Apoe mediated neuroinflammation and neurodegeneration in alzheimer’s disease. Semin. Immunol.
2022, 59, 101594. [CrossRef] [PubMed]
26. Homann, J.; Osburg, T.; Ohlei, O.; Dobricic, V.; Deecke, L.; Bos, I.; Vandenberghe, R.; Gabel, S.; Scheltens, P.; Teunissen, C.E.
Genome-wide association study of alzheimer’s disease brain imaging biomarkers and neuropsychological phenotypes in the
european medical information framework for alzheimer’s disease multimodal biomarker discovery dataset. Front. Aging Neurosci.
2022, 14, 840651. [CrossRef] [PubMed]
27. Barone, E.; Di Domenico, F.; Mancuso, C.; Butterfield, D.A. The janus face of the heme oxygenase/biliverdin reductase system in
alzheimer disease: It’s time for reconciliation. Neurobiol. Dis. 2014, 62, 144–159. [CrossRef]
28. Heath, L.; Earls, J.C.; Magis, A.T.; Kornilov, S.A.; Lovejoy, J.C.; Funk, C.C.; Rappaport, N.; Logsdon, B.A.; Mangravite, L.M.;
Kunkle, B.W. Manifestations of alzheimer’s disease genetic risk in the blood are evident in a multiomic analysis in healthy adults
aged 18 to 90. Sci. Rep. 2022, 12, 6117. [CrossRef] [PubMed]
29. Litvinchuk, A.; Suh, J.H.; Guo, J.L.; Lin, K.; Davis, S.S.; Bien-Ly, N.; Tycksen, E.; Tabor, G.T.; Serrano, J.R.; Manis, M. Amelioration
of tau and apoe4-linked glial lipid accumulation and neurodegeneration with an lxr agonist. Neuron 2024, 112, 384–403.e8.
[CrossRef]
30. Tracy, T.E.; Gan, L. Tau-mediated synaptic and neuronal dysfunction in neurodegenerative disease. Curr. Opin. Neurobiol. 2018,
51, 134–138. [CrossRef]
31. Maday, S.; Twelvetrees, A.E.; Moughamian, A.J.; Holzbaur, E.L. Axonal transport: Cargo-specific mechanisms of motility and
regulation. Neuron 2014, 84, 292–309. [CrossRef]
Int. J. Mol. Sci. 2024, 25, 6901 21 of 28
32. Sherman, M.A.; LaCroix, M.; Amar, F.; Larson, M.E.; Forster, C.; Aguzzi, A.; Bennett, D.A.; Ramsden, M.; Lesné, S.E. Soluble
conformers of aβ and tau alter selective proteins governing axonal transport. J. Neurosci. 2016, 36, 9647–9658. [CrossRef]
[PubMed]
33. Abyadeh, M.; Gupta, V.; Paulo, J.A.; Mahmoudabad, A.G.; Shadfar, S.; Mirshahvaladi, S.; Gupta, V.; Nguyen, C.T.; Finkelstein,
D.I.; You, Y. Amyloid-beta and tau protein beyond alzheimer’s disease. Neural Regen. Res. 2024, 19, 1262–1276. [CrossRef]
[PubMed]
34. Wang, J.; Gu, B.J.; Masters, C.L.; Wang, Y.-J. A systemic view of alzheimer disease—Insights from amyloid-β metabolism beyond
the brain. Nat. Rev. Neurol. 2017, 13, 612–623. [CrossRef] [PubMed]
35. Fá, M.; Puzzo, D.; Piacentini, R.; Staniszewski, A.; Zhang, H.; Baltrons, M.A.; Li Puma, D.D.; Chatterjee, I.; Li, J.; Saeed, F.
Extracellular tau oligomers produce an immediate impairment of ltp and memory. Sci. Rep. 2016, 6, 19393. [CrossRef] [PubMed]
36. Brand, A.L.; Lawler, P.E.; Bollinger, J.G.; Li, Y.; Schindler, S.E.; Li, M.; Lopez, S.; Ovod, V.; Nakamura, A.; Shaw, L.M. The
performance of plasma amyloid beta measurements in identifying amyloid plaques in alzheimer’s disease: A literature review.
Alzheimer’s Res. Ther. 2022, 14, 195. [CrossRef] [PubMed]
37. Leuzy, A.; Janelidze, S.; Mattsson-Carlgren, N.; Palmqvist, S.; Jacobs, D.; Cicognola, C.; Stomrud, E.; Vanmechelen, E.; Dage, J.L.;
Hansson, O. Comparing the clinical utility and diagnostic performance of csf p-tau181, p-tau217, and p-tau231 assays. Neurology
2021, 97, e1681–e1694. [CrossRef]
38. Suárez-Calvet, M.; Karikari, T.K.; Ashton, N.J.; Lantero Rodriguez, J.; Milà-Alomà, M.; Gispert, J.D.; Salvadó, G.; Minguillon,
C.; Fauria, K.; Shekari, M. Novel tau biomarkers phosphorylated at t181, t217 or t231 rise in the initial stages of the preclinical
alzheimer’s continuum when only subtle changes in aβ pathology are detected. EMBO Mol. Med. 2020, 12, e12921. [CrossRef]
[PubMed]
39. Ashton, N.J.; Benedet, A.L.; Pascoal, T.A.; Karikari, T.K.; Lantero-Rodriguez, J.; Brum, W.S.; Mathotaarachchi, S.; Therriault,
J.; Savard, M.; Chamoun, M. Cerebrospinal fluid p-tau231 as an early indicator of emerging pathology in alzheimer’s disease.
EBioMedicine 2022, 76, 103836. [CrossRef] [PubMed]
40. Dolphin, H.; Dyer, A.H.; Morrison, L.; Shenkin, S.D.; Welsh, T.; Kennelly, S.P. New horizons in the diagnosis and management of
alzheimer’s disease in older adults. Age Ageing 2024, 53, afae005. [CrossRef]
41. Thijssen, E.H.; La Joie, R.; Wolf, A.; Strom, A.; Wang, P.; Iaccarino, L.; Bourakova, V.; Cobigo, Y.; Heuer, H.; Spina, S. Diagnostic
value of plasma phosphorylated tau181 in alzheimer’s disease and frontotemporal lobar degeneration. Nat. Med. 2020, 26,
387–397. [CrossRef]
42. Janelidze, S.; Mattsson, N.; Palmqvist, S.; Smith, R.; Beach, T.G.; Serrano, G.E.; Chai, X.; Proctor, N.K.; Eichenlaub, U.; Zetterberg, H.
Plasma p-tau181 in alzheimer’s disease: Relationship to other biomarkers, differential diagnosis, neuropathology and longitudinal
progression to alzheimer’s dementia. Nat. Med. 2020, 26, 379–386. [CrossRef]
43. Kurihara, M.; Matsubara, T.; Morimoto, S.; Arakawa, A.; Ohse, K.; Kanemaru, K.; Iwata, A.; Murayama, S.; Saito, Y. Neuropatholog-
ical changes associated with aberrant cerebrospinal fluid p-tau181 and aβ42 in alzheimer’s disease and other neurodegenerative
diseases. Acta Neuropathol. Commun. 2024, 12, 48. [CrossRef] [PubMed]
44. Graff-Radford, J.; Yong, K.X.; Apostolova, L.G.; Bouwman, F.H.; Carrillo, M.; Dickerson, B.C.; Rabinovici, G.D.; Schott, J.M.; Jones,
D.T.; Murray, M.E. New insights into atypical alzheimer’s disease in the era of biomarkers. Lancet Neurol. 2021, 20, 222–234.
[CrossRef]
45. Barthélemy, N.R.; Li, Y.; Joseph-Mathurin, N.; Gordon, B.A.; Hassenstab, J.; Benzinger, T.; Buckles, V.; Fagan, A.M.; Perrin, R.J.;
Goate, A.M. A soluble phosphorylated tau signature links tau, amyloid and the evolution of stages of dominantly inherited
alzheimer’s disease. Nat. Med. 2020, 26, 398–407. [CrossRef]
46. Phan, L.M.T.; Cho, S. Fluorescent aptasensor and colorimetric aptablot for p-tau231 detection: Toward early diagnosis of
alzheimer’s disease. Biomedicines 2022, 10, 93. [CrossRef]
47. Gonzalez-Ortiz, F.; Turton, M.; Kac, P.R.; Smirnov, D.; Premi, E.; Ghidoni, R.; Benussi, L.; Cantoni, V.; Saraceno, C.; Rivolta, J.
Brain-derived tau: A novel blood-based biomarker for alzheimer’s disease-type neurodegeneration. Brain 2023, 146, 1152–1165.
[CrossRef] [PubMed]
48. Cardoso, S.; Carvalho, C.; Correia, S.C. Alzheimer’s disease—115 years after its discovery. Biomedicines 2024, 12, 478. [CrossRef]
49. Oeckl, P.; Halbgebauer, S.; Anderl-Straub, S.; von Arnim, C.A.; Diehl-Schmid, J.; Froelich, L.; Grimmer, T.; Hausner, L.; Denk, J.;
Jahn, H. Targeted mass spectrometry suggests beta-synuclein as synaptic blood marker in alzheimer’s disease. J. Proteome Res.
2020, 19, 1310–1318. [CrossRef] [PubMed]
50. Cicognola, C.; Janelidze, S.; Hertze, J.; Zetterberg, H.; Blennow, K.; Mattsson-Carlgren, N.; Hansson, O. Plasma glial fibrillary
acidic protein detects alzheimer pathology and predicts future conversion to alzheimer dementia in patients with mild cognitive
impairment. Alzheimer’s Res. Ther. 2021, 13, 68. [CrossRef]
51. Kaneko, N.; Nakamura, A.; Washimi, Y.; Kato, T.; Sakurai, T.; Arahata, Y.; Bundo, M.; Takeda, A.; Niida, S.; Ito, K. Novel plasma
biomarker surrogating cerebral amyloid deposition. Proc. Jpn. Acad. Ser. B 2014, 90, 353–364. [CrossRef]
52. Pérez-Grijalba, V.; Arbizu, J.; Romero, J.; Prieto, E.; Pesini, P.; Sarasa, L.; Guillen, F.; Monleón, I.; San-José, I.; Martínez-Lage, P.
Plasma aβ42/40 ratio alone or combined with fdg-pet can accurately predict amyloid-pet positivity: A cross-sectional analysis
from the ab255 study. Alzheimer’s Res. Ther. 2019, 11, 96. [CrossRef]
Int. J. Mol. Sci. 2024, 25, 6901 22 of 28
53. Palmqvist, S.; Janelidze, S.; Stomrud, E.; Zetterberg, H.; Karl, J.; Zink, K.; Bittner, T.; Mattsson, N.; Eichenlaub, U.; Blennow, K.
Performance of fully automated plasma assays as screening tests for alzheimer disease–related β-amyloid status. JAMA Neurol.
2019, 76, 1060–1069. [CrossRef]
54. Palmqvist, S.; Stomrud, E.; Cullen, N.; Janelidze, S.; Manuilova, E.; Jethwa, A.; Bittner, T.; Eichenlaub, U.; Suridjan, I.; Kollmorgen,
G. An accurate fully automated panel of plasma biomarkers for alzheimer’s disease. Alzheimer’s Dement. 2023, 19, 1204–1215.
[CrossRef]
55. Tropea, M.R.; Puma, D.D.L.; Melone, M.; Gulisano, W.; Arancio, O.; Grassi, C.; Conti, F.; Puzzo, D. Genetic deletion of α7
nicotinic acetylcholine receptors induces an age-dependent alzheimer’s disease-like pathology. Prog. Neurobiol. 2021, 206, 102154.
[CrossRef]
56. Perluigi, M.; Di Domenico, F.; Butterfield, D.A. Oxidative damage in neurodegeneration: Roles in the pathogenesis and
progression of alzheimer disease. Physiol. Rev. 2024, 104, 103–197. [CrossRef]
57. Paul, P.; Bhattacharjee, A.; Bordoloi, S.K.; Paul, U.K. The evolution of alzheimer’s disease therapies: A comprehensive review.
Ann. Med. Sci. Res. 2024, 3, 11–19. [CrossRef]
58. Fu, W.-Y.; Ip, N.Y. The role of genetic risk factors of alzheimer’s disease in synaptic dysfunction. Semin. Cell Dev. Biol. 2023, 139,
3–12. [CrossRef]
59. Morabito, S.; Miyoshi, E.; Michael, N.; Swarup, V. Integrative genomics approach identifies conserved transcriptomic networks in
alzheimer’s disease. Hum. Mol. Genet. 2020, 29, 2899–2919. [CrossRef]
60. Kunkle, B.W.; Grenier-Boley, B.; Sims, R.; Bis, J.C.; Damotte, V.; Naj, A.C.; Boland, A.; Vronskaya, M.; Van Der Lee, S.J.; Amlie-Wolf,
A. Genetic meta-analysis of diagnosed alzheimer’s disease identifies new risk loci and implicates aβ, tau, immunity and lipid
processing. Nat. Genet. 2019, 51, 414–430. [CrossRef]
61. Schwartzentruber, J.; Cooper, S.; Liu, J.Z.; Barrio-Hernandez, I.; Bello, E.; Kumasaka, N.; Young, A.M.; Franklin, R.J.; Johnson,
T.; Estrada, K. Genome-wide meta-analysis, fine-mapping and integrative prioritization implicate new alzheimer’s disease risk
genes. Nat. Genet. 2021, 53, 392–402. [CrossRef]
62. Escott-Price, V.; Hardy, J. Genome-wide association studies for alzheimer’s disease: Bigger is not always better. Brain Commun.
2022, 4, fcac125. [CrossRef]
63. Wightman, D.P.; Jansen, I.E.; Savage, J.E.; Shadrin, A.A.; Bahrami, S.; Holland, D.; Rongve, A.; Børte, S.; Winsvold, B.S.; Drange,
O.K. A genome-wide association study with 1,126,563 individuals identifies new risk loci for alzheimer’s disease. Nat. Genet.
2021, 53, 1276–1282. [CrossRef]
64. Deming, Y.; Li, Z.; Kapoor, M.; Harari, O.; Del-Aguila, J.L.; Black, K.; Carrell, D.; Cai, Y.; Fernandez, M.V.; Budde, J. Genome-wide
association study identifies four novel loci associated with alzheimer’s endophenotypes and disease modifiers. Acta Neuropathol.
2017, 133, 839–856. [CrossRef]
65. Lee, B.; Yao, X.; Shen, L. Genome-wide association study of quantitative biomarkers identifies a novel locus for alzheimer’s
disease at 12p12. 1. BMC Genom. 2022, 23, 85. [CrossRef]
66. Lambert, J.-C.; Ibrahim-Verbaas, C.A.; Harold, D.; Naj, A.C.; Sims, R.; Bellenguez, C.; DeStafano, A.; Bis, J.; Beecham, G.; Grenier-
Boley, B. European alzheimer’s disease initiative (eadi); genetic and environmental risk in alzheimer’s disease; alzheimer’s
disease genetic consortium; cohorts for heart and aging research in genomic epidemiology. Meta-analysis of 74,046 individuals
identifies 11 new susceptibility loci for alzheimer’s disease. Nat Genet 2013, 45, 1452–1458.
67. Marioni, R.E.; Harris, S.E.; Zhang, Q.; McRae, A.F.; Hagenaars, S.P.; Hill, W.D.; Davies, G.; Ritchie, C.W.; Gale, C.R.; Starr, J.M.
Gwas on family history of alzheimer’s disease. Transl. Psychiatry 2018, 8, 99. [CrossRef]
68. Karch, C.M.; Ezerskiy, L.A.; Bertelsen, S.; Consortium, A.s.D.G.; Goate, A.M. Alzheimer’s disease risk polymorphisms regulate
gene expression in the zcwpw1 and the celf1 loci. PLoS ONE 2016, 11, e0148717. [CrossRef]
69. Karch, C.M.; Goate, A.M. Alzheimer’s disease risk genes and mechanisms of disease pathogenesis. Biol. Psychiatry 2015, 77, 43–51.
[CrossRef]
70. Kong, F.; Wu, T.; Dai, J.; Cai, J.; Zhai, Z.; Zhu, Z.; Xu, Y.; Sun, T. Knowledge domains and emerging trends of genome-wide
association studies in alzheimer’s disease: A bibliometric analysis and visualization study from 2002 to 2022. PLoS ONE 2024,
19, e0295008. [CrossRef]
71. Zhu, B.; Wang, R.M.; Wang, J.T.; Chen, R.L.; Zheng, Y.F.; Zhang, L.; Zhao, Z.G. Correlation of rs9331888 polymorphism with
alzheimer’s disease among caucasian and chinese populations: A meta-analysis and systematic review. Metab. Brain Dis. 2017, 32,
981–989. [CrossRef]
72. Bellenguez, C.; Küçükali, F.; Jansen, I.E.; Kleineidam, L.; Moreno-Grau, S.; Amin, N.; Naj, A.C.; Campos-Martin, R.; Grenier-Boley,
B.; Andrade, V. New insights into the genetic etiology of alzheimer’s disease and related dementias. Nat. Genet. 2022, 54, 412–436.
[CrossRef]
73. Iwai, K. Lubac-mediated linear ubiquitination: A crucial regulator of immune signaling. Proc. Jpn. Acad. Ser. B 2021, 97, 120–133.
[CrossRef]
74. Pathak, G.A.; Silzer, T.K.; Sun, J.; Zhou, Z.; Daniel, A.A.; Johnson, L.; O’Bryant, S.; Phillips, N.R.; Barber, R.C. Genome-wide
methylation of mild cognitive impairment in mexican americans highlights genes involved in synaptic transport, alzheimer’s
disease-precursor phenotypes, and metabolic morbidities. J. Alzheimer’s Dis. 2019, 72, 733–749. [CrossRef]
75. Yong, W.-S.; Hsu, F.-M.; Chen, P.-Y. Profiling genome-wide DNA methylation. Epigenetics Chromatin 2016, 9, 26. [CrossRef]
Int. J. Mol. Sci. 2024, 25, 6901 23 of 28
76. Sabarwal, A.; Kumar, K.; Singh, R.P. Hazardous effects of chemical pesticides on human health–cancer and other associated
disorders. Environ. Toxicol. Pharmacol. 2018, 63, 103–114. [CrossRef]
77. Gao, X.; Chen, Q.; Yao, H.; Tan, J.; Liu, Z.; Zhou, Y.; Zou, Z. Epigenetics in alzheimer’s disease. Front. Aging Neurosci. 2022,
14, 911635. [CrossRef]
78. Andrews, S.J.; Renton, A.E.; Fulton-Howard, B.; Podlesny-Drabiniok, A.; Marcora, E.; Goate, A.M. The complex genetic
architecture of alzheimer’s disease: Novel insights and future directions. EBioMedicine 2023, 90, 104511. [CrossRef]
79. Lee, T.; Lee, H.; Initiative, A.S.D.N. Identification of disease-related genes that are common between alzheimer’s and cardiovascu-
lar disease using blood genome-wide transcriptome analysis. Biomedicines 2021, 9, 1525. [CrossRef]
80. Xu, M.; Zhang, D.-F.; Luo, R.; Wu, Y.; Zhou, H.; Kong, L.-L.; Bi, R.; Yao, Y.-G. A systematic integrated analysis of brain expression
profiles reveals yap1 and other prioritized hub genes as important upstream regulators in alzheimer’s disease. Alzheimer’s Dement.
2018, 14, 215–229. [CrossRef]
81. Dumanis, S.B.; DiBattista, A.M.; Miessau, M.; Moussa, C.E.; Rebeck, G.W. Apoe genotype affects the pre-synaptic compartment of
glutamatergic nerve terminals. J. Neurochem. 2013, 124, 4–14. [CrossRef]
82. Li, X.; Zhang, J.; Li, D.; He, C.; He, K.; Xue, T.; Wan, L.; Zhang, C.; Liu, Q. Astrocytic apoe reprograms neuronal cholesterol
metabolism and histone-acetylation-mediated memory. Neuron 2021, 109, 957–970.e8. [CrossRef] [PubMed]
83. Lin, Y.-T.; Seo, J.; Gao, F.; Feldman, H.M.; Wen, H.-L.; Penney, J.; Cam, H.P.; Gjoneska, E.; Raja, W.K.; Cheng, J. Apoe4 causes
widespread molecular and cellular alterations associated with alzheimer’s disease phenotypes in human ipsc-derived brain cell
types. Neuron 2018, 98, 1141–1154.e7. [CrossRef] [PubMed]
84. Litvinchuk, A.; Huynh, T.P.V.; Shi, Y.; Jackson, R.J.; Finn, M.B.; Manis, M.; Francis, C.M.; Tran, A.C.; Sullivan, P.M.; Ulrich, J.D.
Apolipoprotein e4 reduction with antisense oligonucleotides decreases neurodegeneration in a tauopathy model. Ann. Neurol.
2021, 89, 952–966. [CrossRef] [PubMed]
85. Marcello, E.; Saraceno, C.; Musardo, S.; Vara, H.; de La Fuente, A.G.; Pelucchi, S.; Di Marino, D.; Borroni, B.; Tramontano,
A.; Pérez-Otaño, I. Endocytosis of synaptic adam10 in neuronal plasticity and alzheimer’s disease. J. Clin. Investig. 2013, 123,
2523–2538. [CrossRef] [PubMed]
86. Kuhn, P.-H.; Colombo, A.V.; Schusser, B.; Dreymueller, D.; Wetzel, S.; Schepers, U.; Herber, J.; Ludwig, A.; Kremmer, E.; Montag,
D. Systematic substrate identification indicates a central role for the metalloprotease adam10 in axon targeting and synapse
function. Elife 2016, 5, e12748. [CrossRef]
87. De Roeck, A.; Van Broeckhoven, C.; Sleegers, K. The role of abca7 in alzheimer’s disease: Evidence from genomics, transcriptomics
and methylomics. Acta Neuropathol. 2019, 138, 201–220. [CrossRef] [PubMed]
88. Dib, S.; Pahnke, J.; Gosselet, F. Role of abca7 in human health and in alzheimer’s disease. Int. J. Mol. Sci. 2021, 22, 4603. [CrossRef]
89. Yi, B.; Jahangir, A.; Evans, A.K.; Briggs, D.; Ravina, K.; Ernest, J.; Farimani, A.B.; Sun, W.; Rajadas, J.; Green, M. Discovery of novel
brain permeable and g protein-biased beta-1 adrenergic receptor partial agonists for the treatment of neurocognitive disorders.
PLoS ONE 2017, 12, e0180319. [CrossRef] [PubMed]
90. Ubelmann, F.; Burrinha, T.; Salavessa, L.; Gomes, R.; Ferreira, C.; Moreno, N.; Guimas Almeida, C. Bin1 and cd 2 ap polarise the
endocytic generation of beta-amyloid. EMBO Rep. 2017, 18, 102–122. [CrossRef]
91. Calafate, S.; Flavin, W.; Verstreken, P.; Moechars, D. Loss of bin1 promotes the propagation of tau pathology. Cell Rep. 2016, 17,
931–940. [CrossRef]
92. Crotti, A.; Sait, H.R.; McAvoy, K.M.; Estrada, K.; Ergun, A.; Szak, S.; Marsh, G.; Jandreski, L.; Peterson, M.; Reynolds, T.L. Bin1
favors the spreading of tau via extracellular vesicles. Sci. Rep. 2019, 9, 9477. [CrossRef] [PubMed]
93. Yu, Y.; Niccoli, T.; Ren, Z.; Woodling, N.S.; Aleyakpo, B.; Szabadkai, G.; Partridge, L. Picalm rescues glutamatergic neurotransmis-
sion, behavioural function and survival in a drosophila model of aβ42 toxicity. Hum. Mol. Genet. 2020, 29, 2420–2434. [CrossRef]
[PubMed]
94. Crehan, H.; Holton, P.; Wray, S.; Pocock, J.; Guerreiro, R.; Hardy, J. Complement receptor 1 (cr1) and alzheimer’s disease.
Immunobiology 2012, 217, 244–250. [CrossRef] [PubMed]
95. Adams, C.J.; Kopp, M.C.; Larburu, N.; Nowak, P.R.; Ali, M.M. Structure and molecular mechanism of er stress signaling by the
unfolded protein response signal activator ire1. Front. Mol. Biosci. 2019, 6, 11. [CrossRef] [PubMed]
96. Duran-Aniotz, C.; Cornejo, V.H.; Espinoza, S.; Ardiles, Á.O.; Medinas, D.B.; Salazar, C.; Foley, A.; Gajardo, I.; Thielen, P.; Iwawaki,
T. Ire1 signaling exacerbates alzheimer’s disease pathogenesis. Acta Neuropathol. 2017, 134, 489–506. [CrossRef] [PubMed]
97. Wang, Y.; Fu, W.-Y.; Cheung, K.; Hung, K.-W.; Chen, C.; Geng, H.; Yung, W.-H.; Qu, J.Y.; Fu, A.K.; Ip, N.Y. Astrocyte-secreted il-33
mediates homeostatic synaptic plasticity in the adult hippocampus. Proc. Natl. Acad. Sci. USA 2021, 118, e2020810118. [CrossRef]
[PubMed]
98. Djurisic, M.; Brott, B.K.; Saw, N.L.; Shamloo, M.; Shatz, C.J. Activity-dependent modulation of hippocampal synaptic plasticity
via pirb and endocannabinoids. Mol. Psychiatry 2019, 24, 1206–1219. [CrossRef]
99. Barić, N. The decline of the expression of low density lipoprotein receptor-related protein 1 (lrp1) during normal ageing and in
alzheimer’s disease. Glycative Stress Res. 2022, 9, 42–54.
100. Belinson, H.; Lev, D.; Masliah, E.; Michaelson, D.M. Activation of the amyloid cascade in apolipoprotein e4 transgenic mice
induces lysosomal activation and neurodegeneration resulting in marked cognitive deficits. J. Neurosci. 2008, 28, 4690–4701.
[CrossRef]
Int. J. Mol. Sci. 2024, 25, 6901 24 of 28
101. Candela, P.; Saint-Pol, J.; Kuntz, M.; Boucau, M.-C.; Lamartiniere, Y.; Gosselet, F.; Fenart, L. In vitro discrimination of the role of
lrp1 at the bbb cellular level: Focus on brain capillary endothelial cells and brain pericytes. Brain Res. 2015, 1594, 15–26. [CrossRef]
102. Deane, R.; Wu, Z.; Sagare, A.; Davis, J.; Du Yan, S.; Hamm, K.; Xu, F.; Parisi, M.; LaRue, B.; Hu, H.W. Lrp/amyloid β-peptide
interaction mediates differential brain efflux of aβ isoforms. Neuron 2004, 43, 333–344. [CrossRef] [PubMed]
103. Shinohara, M.; Tachibana, M.; Kanekiyo, T.; Bu, G. Role of lrp1 in the pathogenesis of alzheimer’s disease: Evidence from
clinical and preclinical studies: Thematic review series: Apoe and lipid homeostasis in alzheimer’s disease. J. Lipid Res. 2017, 58,
1267–1281. [CrossRef] [PubMed]
104. Hsin, H.; Kim, M.J.; Wang, C.-F.; Sheng, M. Proline-rich tyrosine kinase 2 regulates hippocampal long-term depression. J. Neurosci.
2010, 30, 11983–11993. [CrossRef] [PubMed]
105. de Pins, B.; Mendes, T.; Giralt, A.; Girault, J.-A. The non-receptor tyrosine kinase pyk2 in brain function and neurological and
psychiatric diseases. Front. Synaptic Neurosci. 2021, 13, 749001. [CrossRef] [PubMed]
106. Nackenoff, A.G.; Hohman, T.J.; Neuner, S.M.; Akers, C.S.; Weitzel, N.C.; Shostak, A.; Ferguson, S.M.; Mobley, B.; Bennett, D.A.;
Schneider, J.A. Pld3 is a neuronal lysosomal phospholipase d associated with β-amyloid plaques and cognitive function in
alzheimer’s disease. PLoS Genet. 2021, 17, e1009406. [CrossRef] [PubMed]
107. Radford, H.; Moreno, J.A.; Verity, N.; Halliday, M.; Mallucci, G.R. Perk inhibition prevents tau-mediated neurodegeneration in a
mouse model of frontotemporal dementia. Acta Neuropathol. 2015, 130, 633–642. [CrossRef]
108. Yuan, S.H.; Hiramatsu, N.; Liu, Q.; Sun, X.V.; Lenh, D.; Chan, P.; Chiang, K.; Koo, E.H.; Kao, A.W.; Litvan, I. Tauopathy-associated
perk alleles are functional hypomorphs that increase neuronal vulnerability to er stress. Hum. Mol. Genet. 2018, 27, 3951–3963.
[CrossRef]
109. Schmidt, V.; Subkhangulova, A.; Willnow, T.E. Sorting receptor sorla: Cellular mechanisms and implications for disease. Cell. Mol.
Life Sci. 2017, 74, 1475–1483. [CrossRef]
110. Malik, A.R.; Willnow, T.E. Vps10p domain receptors: Sorting out brain health and disease. Trends Neurosci. 2020, 43, 870–885.
[CrossRef]
111. Ulland, T.K.; Colonna, M. Trem2—A key player in microglial biology and alzheimer disease. Nat. Rev. Neurol. 2018, 14, 667–675.
[CrossRef]
112. Deczkowska, A.; Weiner, A.; Amit, I. The physiology, pathology, and potential therapeutic applications of the trem2 signaling
pathway. Cell 2020, 181, 1207–1217. [CrossRef] [PubMed]
113. Di Paolo, G.; Kim, T.-W. Linking lipids to alzheimer’s disease: Cholesterol and beyond. Nat. Rev. Neurosci. 2011, 12, 284–296.
[CrossRef]
114. González, A.; Calfío, C.; Churruca, M.; Maccioni, R.B. Glucose metabolism and ad: Evidence for a potential diabetes type 3.
Alzheimer’s Res. Ther. 2022, 14, 56. [CrossRef] [PubMed]
115. Akhtar, A.; Sah, S.P. Insulin signaling pathway and related molecules: Role in neurodegeneration and alzheimer’s disease.
Neurochem. Int. 2020, 135, 104707. [CrossRef] [PubMed]
116. Perluigi, M.; Di Domenico, F.; Butterfield, D.A. Mtor signaling in aging and neurodegeneration: At the crossroad between
metabolism dysfunction and impairment of autophagy. Neurobiol. Dis. 2015, 84, 39–49. [CrossRef] [PubMed]
117. Urbanska, M.; Gozdz, A.; Swiech, L.J.; Jaworski, J. Mammalian target of rapamycin complex 1 (mtorc1) and 2 (mtorc2) control the
dendritic arbor morphology of hippocampal neurons. J. Biol. Chem. 2012, 287, 30240–30256. [CrossRef] [PubMed]
118. Banks, W.A.; Owen, J.B.; Erickson, M.A. Insulin in the brain: There and back again. Pharmacol. Ther. 2012, 136, 82–93. [CrossRef]
[PubMed]
119. Neth, B.J.; Craft, S. Insulin resistance and alzheimer’s disease: Bioenergetic linkages. Front. Aging Neurosci. 2017, 9, 345. [CrossRef]
120. Zhou, S.; Tu, L.; Chen, W.; Yan, G.; Guo, H.; Wang, X.; Hu, Q.; Liu, H.; Li, F. Alzheimer’s disease, a metabolic disorder: Clinical
advances and basic model studies. Exp. Ther. Med. 2024, 27, 63. [CrossRef]
121. Stanciu, G.D.; Rusu, R.N.; Bild, V.; Filipiuc, L.E.; Tamba, B.-I.; Ababei, D.C. Systemic actions of sglt2 inhibition on chronic mtor
activation as a shared pathogenic mechanism between alzheimer’s disease and diabetes. Biomedicines 2021, 9, 576. [CrossRef]
122. Rosa, E.; Fahnestock, M. Creb expression mediates amyloid β-induced basal bdnf downregulation. Neurobiol. Aging 2015, 36,
2406–2413. [CrossRef] [PubMed]
123. Cheng, Y.; Tian, D.-Y.; Wang, Y.-J. Peripheral clearance of brain-derived aβ in alzheimer’s disease: Pathophysiology and
therapeutic perspectives. Transl. Neurodegener. 2020, 9, 16. [CrossRef] [PubMed]
124. Fahnestock, M. Brain-derived neurotrophic factor: The link between amyloid-β and memory loss. Future Neurol. 2011, 6, 627–639.
[CrossRef]
125. Campos-Pena, V.; Toral-Rios, D.; Becerril-Pérez, F.; Sánchez-Torres, C.; Delgado-Namorado, Y.; Torres-Ossorio, E.; Franco-
Bocanegra, D.; Carvajal, K. Metabolic syndrome as a risk factor for alzheimer’s disease: Is aβ a crucial factor in both pathologies?
Antioxid. Redox Signal. 2017, 26, 542–560. [CrossRef] [PubMed]
126. Chen, Z.; Zhong, C. Oxidative stress in alzheimer’s disease. Neurosci. Bull. 2014, 30, 271–281. [CrossRef] [PubMed]
127. Ioannou, M.S.; Jackson, J.; Sheu, S.-H.; Chang, C.-L.; Weigel, A.V.; Liu, H.; Pasolli, H.A.; Xu, C.S.; Pang, S.; Matthies, D. Neuron-
astrocyte metabolic coupling protects against activity-induced fatty acid toxicity. Cell 2019, 177, 1522–1535.e14. [CrossRef]
[PubMed]
128. Fazzari, P.; Horre, K.; Arranz, A.M.; Frigerio, C.S.; Saito, T.; Saido, T.C.; De Strooper, B. Pld3 gene and processing of app. Nature
2017, 541, E1–E2. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2024, 25, 6901 25 of 28
129. Cai, R.; Wang, Y.; Huang, Z.; Zou, Q.; Pu, Y.; Yu, C.; Cai, Z. Role of rhoa/rock signaling in alzheimer’s disease. Behav. Brain Res.
2021, 414, 113481. [CrossRef] [PubMed]
130. Xiang, X.; Wang, X.; Jin, S.; Hu, J.; Wu, Y.; Li, Y.; Wu, X. Activation of gpr55 attenuates cognitive impairment and neurotoxicity in
a mouse model of alzheimer’s disease induced by aβ1–42 through inhibiting rhoa/rock2 pathway. Prog. Neuro-Psychopharmacol.
Biol. Psychiatry 2022, 112, 110423. [CrossRef]
131. Van Eldik, L.J.; Carrillo, M.C.; Cole, P.E.; Feuerbach, D.; Greenberg, B.D.; Hendrix, J.A.; Kennedy, M.; Kozauer, N.; Margolin, R.A.;
Molinuevo, J.L. The roles of inflammation and immune mechanisms in alzheimer’s disease. Alzheimer’s Dement. Transl. Res. Clin.
Interv. 2016, 2, 99–109. [CrossRef]
132. Shi, J.-Q.; Shen, W.; Chen, J.; Wang, B.-R.; Zhong, L.-L.; Zhu, Y.-W.; Zhu, H.-Q.; Zhang, Q.-Q.; Zhang, Y.-D.; Xu, J. Anti-tnf-α
reduces amyloid plaques and tau phosphorylation and induces cd11c-positive dendritic-like cell in the app/ps1 transgenic mouse
brains. Brain Res. 2011, 1368, 239–247. [CrossRef] [PubMed]
133. Nguyen, H.D.; Vu, H.H. Molecular mechanisms implicated in protein changes in the alzheimer’s disease human hippocampus.
Mech. Ageing Dev. 2024, 219, 111930. [CrossRef]
134. Wang, Y.; Li, L.; Wu, Y.; Zhang, S.; Ju, Q.; Yang, Y.; Jin, Y.; Shi, H.; Sun, C. Cd44 deficiency represses neuroinflammation and
rescues dopaminergic neurons in a mouse model of parkinson’s disease. Pharmacol. Res. 2022, 177, 106133. [CrossRef] [PubMed]
135. Smith, R.; Klein, P.; Koc-Schmitz, Y.; Waldvogel, H.J.; Faull, R.L.; Brundin, P.; Plomann, M.; Li, J.Y. Loss of snap-25 and rabphilin
3a in sensory-motor cortex in huntington’s disease. J. Neurochem. 2007, 103, 115–123. [CrossRef] [PubMed]
136. Wu, M.; Fang, K.; Wang, W.; Lin, W.; Guo, L.; Wang, J. Identification of key genes and pathways for alzheimer’s disease via
combined analysis of genome-wide expression profiling in the hippocampus. Biophys. Rep. 2019, 5, 98–109. [CrossRef]
137. Ali, M.; Falkenhain, K.; Njiru, B.N.; Murtaza-Ali, M.; Ruiz-Uribe, N.E.; Haft-Javaherian, M.; Catchers, S.; Nishimura, N.; Schaffer,
C.B.; Bracko, O. Vegf signalling causes stalls in brain capillaries and reduces cerebral blood flow in alzheimer’s mice. Brain 2022,
145, 1449–1463. [CrossRef] [PubMed]
138. García-Culebras, A.; Cuartero, M.I.; Peña-Martínez, C.; Moraga, A.; Vázquez-Reyes, S.; de Castro-Millán, F.J.; Cortes-Canteli,
M.; Lizasoain, I.; Moro, M.Á. Myeloid cells in vascular dementia and alzheimer’s disease: Possible therapeutic targets? Br. J.
Pharmacol. 2024, 181, 777–798. [CrossRef] [PubMed]
139. Kang, L.; Yu, H.; Yang, X.; Zhu, Y.; Bai, X.; Wang, R.; Cao, Y.; Xu, H.; Luo, H.; Lu, L. Neutrophil extracellular traps released by
neutrophils impair revascularization and vascular remodeling after stroke. Nat. Commun. 2020, 11, 2488. [CrossRef] [PubMed]
140. Cai, Z.; Wan, C.-Q.; Liu, Z. Astrocyte and alzheimer’s disease. J. Neurol. 2017, 264, 2068–2074. [CrossRef]
141. Fernandez, C.G.; Hamby, M.E.; McReynolds, M.L.; Ray, W.J. The role of apoe4 in disrupting the homeostatic functions of astrocytes
and microglia in aging and alzheimer’s disease. Front. Aging Neurosci. 2019, 11, 14. [CrossRef]
142. Hickman, S.E.; El Khoury, J. Trem2 and the neuroimmunology of alzheimer’s disease. Biochem. Pharmacol. 2014, 88, 495–498.
[CrossRef] [PubMed]
143. Kleinberger, G.; Yamanishi, Y.; Suárez-Calvet, M.; Czirr, E.; Lohmann, E.; Cuyvers, E.; Struyfs, H.; Pettkus, N.; Wenninger-
Weinzierl, A.; Mazaheri, F. Trem2 mutations implicated in neurodegeneration impair cell surface transport and phagocytosis. Sci.
Transl. Med. 2014, 6, 243ra86. [CrossRef] [PubMed]
144. Guerreiro, R.; Wojtas, A.; Bras, J.; Carrasquillo, M.; Rogaeva, E.; Majounie, E.; Cruchaga, C.; Sassi, C.; Kauwe, J.S.; Younkin, S.
Trem2 variants in alzheimer’s disease. N. Engl. J. Med. 2013, 368, 117–127. [CrossRef] [PubMed]
145. Steinberg, S.; Stefansson, H.; Jonsson, T.; Johannsdottir, H.; Ingason, A.; Helgason, H.; Sulem, P.; Magnusson, O.T.; Gudjonsson,
S.A.; Unnsteinsdottir, U. Loss-of-function variants in abca7 confer risk of alzheimer’s disease. Nat. Genet. 2015, 47, 445–447.
[CrossRef] [PubMed]
146. Lian, H.; Yang, L.; Cole, A.; Sun, L.; Chiang, A.C.-A.; Fowler, S.W.; Shim, D.J.; Rodriguez-Rivera, J.; Taglialatela, G.; Jankowsky, J.L.
Nfκb-activated astroglial release of complement c3 compromises neuronal morphology and function associated with alzheimer’s
disease. Neuron 2015, 85, 101–115. [CrossRef]
147. Jayaraman, A.; Htike, T.T.; James, R.; Picon, C.; Reynolds, R. Tnf-mediated neuroinflammation is linked to neuronal necroptosis
in alzheimer’s disease hippocampus. Acta Neuropathol. Commun. 2021, 9, 159. [CrossRef] [PubMed]
148. Picon, C.; Jayaraman, A.; James, R.; Beck, C.; Gallego, P.; Witte, M.E.; van Horssen, J.; Mazarakis, N.D.; Reynolds, R. Neuron-
specific activation of necroptosis signaling in multiple sclerosis cortical grey matter. Acta Neuropathol. 2021, 141, 585–604.
[CrossRef] [PubMed]
149. Asimakidou, E.; Reynolds, R.; Barron, A.M.; Lo, C.H. Autolysosomal acidification impairment as a mediator for tnfr1 induced
neuronal necroptosis in alzheimer’s disease. Neural Regen. Res. 2024, 19, 1690–1870. [CrossRef] [PubMed]
150. Song, L.; Pei, L.; Yao, S.; Wu, Y.; Shang, Y. Nlrp3 inflammasome in neurological diseases, from functions to therapies. Front. Cell.
Neurosci. 2017, 11, 63. [CrossRef]
151. Tripathi, P.; Rodriguez-Muela, N.; Klim, J.R.; de Boer, A.S.; Agrawal, S.; Sandoe, J.; Lopes, C.S.; Ogliari, K.S.; Williams, L.A.;
Shear, M. Reactive astrocytes promote als-like degeneration and intracellular protein aggregation in human motor neurons by
disrupting autophagy through tgf-β1. Stem Cell Rep. 2017, 9, 667–680. [CrossRef]
152. Feng, T.; Lacrampe, A.; Hu, F. Physiological and pathological functions of tmem106b: A gene associated with brain aging and
multiple brain disorders. Acta Neuropathol. 2021, 141, 327–339. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2024, 25, 6901 26 of 28
153. Chandrasekaran, A.; Dittlau, K.S.; Corsi, G.I.; Haukedal, H.; Doncheva, N.T.; Ramakrishna, S.; Ambardar, S.; Salcedo, C.; Schmidt,
S.I.; Zhang, Y. Astrocytic reactivity triggered by defective autophagy and metabolic failure causes neurotoxicity in frontotemporal
dementia type 3. Stem Cell Rep. 2021, 16, 2736–2751. [CrossRef] [PubMed]
154. Iyer, H.; Shen, K.; Meireles, A.M.; Talbot, W.S. A lysosomal regulatory circuit essential for the development and function of
microglia. Sci. Adv. 2022, 8, eabp8321. [CrossRef] [PubMed]
155. Jayaraman, A.; Reynolds, R. Diverse pathways to neuronal necroptosis in alzheimer’s disease. Eur. J. Neurosci. 2022, 56, 5428–5441.
[CrossRef] [PubMed]
156. Udeochu, J.C.; Shea, J.M.; Villeda, S.A. Microglia communication: Parallels between aging and alzheimer’s disease. Clin. Exp.
Neuroimmunol. 2016, 7, 114–125. [CrossRef] [PubMed]
157. Lee, J.; Chan, S.L.; Mattson, M.P. Adverse effect of a presenilin-1 mutation in microglia results in enhanced nitric oxide and
inflammatory cytokine responses to immune challenge in the brain. Neuromol. Med. 2002, 2, 29–45.
158. Fung, S.; Smith, C.L.; Prater, K.E.; Case, A.; Green, K.; Osnis, L.; Winston, C.; Kinoshita, Y.; Sopher, B.; Morrison, R.S. Early-onset
familial alzheimer disease variant psen2 n141i heterozygosity is associated with altered microglia phenotype. J. Alzheimer’s Dis.
2020, 77, 675–688. [CrossRef] [PubMed]
159. Deus, C.M.; Yambire, K.F.; Oliveira, P.J.; Raimundo, N. Mitochondria–lysosome crosstalk: From physiology to neurodegeneration.
Trends Mol. Med. 2020, 26, 71–88. [CrossRef] [PubMed]
160. Quick, J.D.; Silva, C.; Wong, J.H.; Lim, K.L.; Reynolds, R.; Barron, A.M.; Zeng, J.; Lo, C.H. Lysosomal acidification dysfunction in
microglia: An emerging pathogenic mechanism of neuroinflammation and neurodegeneration. J. Neuroinflamm. 2023, 20, 185.
[CrossRef]
161. Vest, R.T.; Chou, C.-C.; Zhang, H.; Haney, M.S.; Li, L.; Laqtom, N.N.; Chang, B.; Shuken, S.; Nguyen, A.; Yerra, L. Small molecule
c381 targets the lysosome to reduce inflammation and ameliorate disease in models of neurodegeneration. Proc. Natl. Acad. Sci.
USA 2022, 119, e2121609119. [CrossRef]
162. Chen, C.-C.; Keller, M.; Hess, M.; Schiffmann, R.; Urban, N.; Wolfgardt, A.; Schaefer, M.; Bracher, F.; Biel, M.; Wahl-Schott, C. A
small molecule restores function to trpml1 mutant isoforms responsible for mucolipidosis type iv. Nat. Commun. 2014, 5, 4681.
[CrossRef] [PubMed]
163. Tong, B.C.-K.; Huang, A.S.; Wu, A.J.; Iyaswamy, A.; Ho, O.K.-Y.; Kong, A.H.-Y.; Sreenivasmurthy, S.G.; Zhu, Z.; Su, C.; Liu, J.
Tetrandrine ameliorates cognitive deficits and mitigates tau aggregation in cell and animal models of tauopathies. J. Biomed. Sci.
2022, 29, 85. [CrossRef] [PubMed]
164. Yao, X.C.; Xue, X.; Zhang, H.T.; Zhu, M.M.; Yang, X.W.; Wu, C.F.; Yang, J.Y. Pseudoginsenoside-f11 alleviates oligomeric
β-amyloid-induced endosome-lysosome defects in microglia. Traffic 2019, 20, 61–70. [CrossRef] [PubMed]
165. Chin, M.Y.; Ang, K.-H.; Davies, J.; Alquezar, C.; Garda, V.G.; Rooney, B.; Leng, K.; Kampmann, M.; Arkin, M.R.; Kao, A.W.
Phenotypic screening using high-content imaging to identify lysosomal ph modulators in a neuronal cell model. ACS Chem.
Neurosci. 2022, 13, 1505–1516. [CrossRef] [PubMed]
166. Zeng, J.; Shirihai, O.S.; Grinstaff, M.W. Modulating lysosomal ph: A molecular and nanoscale materials design perspective. J. Life
Sci. 2020, 2, 25. [CrossRef] [PubMed]
167. Brouillard, M.; Barthélémy, P.; Dehay, B.; Crauste-Manciet, S.; Desvergnes, V. Nucleolipid acid-based nanocarriers restore neuronal
lysosomal acidification defects. Front. Chem. 2021, 9, 736554. [CrossRef]
168. Cunha, A.; Gaubert, A.; Latxague, L.; Dehay, B. Plga-based nanoparticles for neuroprotective drug delivery in neurodegenerative
diseases. Pharmaceutics 2021, 13, 1042. [CrossRef] [PubMed]
169. Lo, C.H.; Zeng, J. Defective lysosomal acidification: A new prognostic marker and therapeutic target for neurodegenerative
diseases. Transl. Neurodegener. 2023, 12, 29. [CrossRef]
170. Frost, G.R.; Li, Y.-M. The role of astrocytes in amyloid production and alzheimer’s disease. Open Biol. 2017, 7, 170228. [CrossRef]
171. Jablonski, A.M.; Warren, L.; Usenovic, M.; Zhou, H.; Sugam, J.; Parmentier-Batteur, S.; Voleti, B. Astrocytic expression of the
alzheimer’s disease risk allele, apoeε4, potentiates neuronal tau pathology in multiple preclinical models. Sci. Rep. 2021, 11, 3438.
[CrossRef]
172. Rojas-Gutierrez, E.; Muñoz-Arenas, G.; Treviño, S.; Espinosa, B.; Chavez, R.; Rojas, K.; Flores, G.; Díaz, A.; Guevara, J. Alzheimer’s
disease and metabolic syndrome: A link from oxidative stress and inflammation to neurodegeneration. Synapse 2017, 71, e21990.
[CrossRef]
173. Ni, J.; Xie, Z.; Quan, Z.; Meng, J.; Qing, H. How brain ‘cleaners’ fail: Mechanisms and therapeutic value of microglial phagocytosis
in alzheimer’s disease. Glia 2024, 72, 227–244. [CrossRef] [PubMed]
174. Mancuso, R.; Fryatt, G.; Cleal, M.; Obst, J.; Pipi, E.; Monzón-Sandoval, J.; Ribe, E.; Winchester, L.; Webber, C.; Nevado, A.; et al.
Csf1r inhibitor jnj-40346527 attenuates microglial proliferation and neurodegeneration in p301s mice. Brain 2019, 142, 3243–3264.
[CrossRef] [PubMed]
175. Chen, J.; Zhou, Y.; Mueller-Steiner, S.; Chen, L.-F.; Kwon, H.; Yi, S.; Mucke, L.; Gan, L. Sirt1 protects against microglia-dependent
amyloid-β toxicity through inhibiting nf-κb signaling. J. Biol. Chem. 2005, 280, 40364–40374. [CrossRef] [PubMed]
176. Wang, C.; Fan, L.; Khawaja, R.R.; Liu, B.; Zhan, L.; Kodama, L.; Chin, M.; Li, Y.; Le, D.; Zhou, Y. Microglial nf-κb drives tau
spreading and toxicity in a mouse model of tauopathy. Nat. Commun. 2022, 13, 1969. [CrossRef]
177. Yan, S.D.; Bierhaus, A.; Nawroth, P.P.; Stern, D.M. Rage and alzheimer’s disease: A progression factor for amyloid-β-induced
cellular perturbation? J. Alzheimer’s Dis. 2009, 16, 833–843. [CrossRef]
Int. J. Mol. Sci. 2024, 25, 6901 27 of 28
178. Farmer, D.G.; Ewart, M.-A.; Mair, K.M.; Kennedy, S. Soluble receptor for advanced glycation end products (srage) attenuates
haemodynamic changes to chronic hypoxia in the mouse. Pulm. Pharmacol. Ther. 2014, 29, 7–14. [CrossRef]
179. Heilmann, R.M.; Otoni, C.C.; Jergens, A.E.; Grützner, N.; Suchodolski, J.S.; Steiner, J.M. Systemic levels of the anti-inflammatory
decoy receptor soluble rage (receptor for advanced glycation end products) are decreased in dogs with inflammatory bowel
disease. Vet. Immunol. Immunopathol. 2014, 161, 184–192. [CrossRef]
180. Kook, S.-Y.; Seok Hong, H.; Moon, M.; Mook-Jung, I. Disruption of blood-brain barrier in alzheimer disease pathogenesis. Tissue
Barriers 2013, 1, 8845–8854. [CrossRef]
181. Wan, W.; Chen, H.; Li, Y. The potential mechanisms of aβ-receptor for advanced glycation end-products interaction disrupting
tight junctions of the blood-brain barrier in alzheimer’s disease. Int. J. Neurosci. 2014, 124, 75–81. [CrossRef]
182. Patel, M. Implications of the clearance methods for amyloid-beta plaques in alzheimer’s disease. Int. J. High Sch. Res. 2024, 6,
107–113.
183. Munoz, L.; Ranaivo, H.R.; Roy, S.M.; Hu, W.; Craft, J.M.; McNamara, L.K.; Chico, L.W.; Van Eldik, L.J.; Watterson, D.M. A
novel p38α mapk inhibitor suppresses brain proinflammatory cytokine up-regulation and attenuates synaptic dysfunction and
behavioral deficits in an alzheimer’s disease mouse model. J. Neuroinflamm. 2007, 4, 21. [CrossRef] [PubMed]
184. Puzzo, D.; Privitera, L.; Leznik, E.; Fa, M.; Staniszewski, A.; Palmeri, A.; Arancio, O. Picomolar amyloid-β positively modulates
synaptic plasticity and memory in hippocampus. J. Neurosci. 2008, 28, 14537–14545. [CrossRef] [PubMed]
185. Arancio, O.; Zhang, H.P.; Chen, X.; Lin, C.; Trinchese, F.; Puzzo, D.; Liu, S.; Hegde, A.; Yan, S.F.; Stern, A. Rage potentiates
aβ-induced perturbation of neuronal function in transgenic mice. EMBO J. 2004, 23, 4096–4105. [CrossRef]
186. Vincenz-Donnelly, L.; Hipp, M.S. The endoplasmic reticulum: A hub of protein quality control in health and disease. Free Radic.
Biol. Med. 2017, 108, 383–393. [CrossRef] [PubMed]
187. Fregno, I.; Molinari, M. Proteasomal and lysosomal clearance of faulty secretory proteins: Er-associated degradation (erad) and
er-to-lysosome-associated degradation (erlad) pathways. Crit. Rev. Biochem. Mol. Biol. 2019, 54, 153–163. [CrossRef] [PubMed]
188. Xu, S.; Liu, H.; Wang, C.; Deng, Y.; Xu, B.; Yang, T.; Liu, W. Dual roles of uprer and uprmt in neurodegenerative diseases. J. Mol.
Med. 2023, 101, 1499–1512. [CrossRef] [PubMed]
189. Montibeller, L.; De Belleroche, J. Amyotrophic lateral sclerosis (als) and alzheimer’s disease (ad) are characterised by differential
activation of er stress pathways: Focus on upr target genes. Cell Stress Chaperones 2018, 23, 897–912. [CrossRef] [PubMed]
190. Yu, Z.; Dou, F.; Wang, Y.; Hou, L.; Chen, H. Ca2+ -dependent endoplasmic reticulum stress correlation with astrogliosis involves
upregulation of kca3. 1 and inhibition of akt/mtor signaling. J. Neuroinflamm. 2018, 15, 316. [CrossRef]
191. Wan, Y.-W.; Al-Ouran, R.; Mangleburg, C.G.; Perumal, T.M.; Lee, T.V.; Allison, K.; Swarup, V.; Funk, C.C.; Gaiteri, C.; Allen, M.
Meta-analysis of the alzheimer’s disease human brain transcriptome and functional dissection in mouse models. Cell Rep. 2020,
32, 107908. [CrossRef]
192. Cissé, M.; Duplan, E.; Lorivel, T.; Dunys, J.; Bauer, C.; Meckler, X.; Gerakis, Y.; Lauritzen, I.; Checler, F. The transcription factor
xbp1s restores hippocampal synaptic plasticity and memory by control of the kalirin-7 pathway in alzheimer model. Mol.
Psychiatry 2017, 22, 1562–1575. [CrossRef] [PubMed]
193. Cozachenco, D.; Ribeiro, F.C.; Ferreira, S.T. Defective proteostasis in alzheimer’s disease. Ageing Res. Rev. 2023, 85, 101862.
[CrossRef]
194. Ahn, J.H.; Cho, H.; Kim, J.-H.; Kim, S.H.; Ham, J.-S.; Park, I.; Suh, S.H.; Hong, S.P.; Song, J.-H.; Hong, Y.-K. Meningeal lymphatic
vessels at the skull base drain cerebrospinal fluid. Nature 2019, 572, 62–66. [CrossRef] [PubMed]
195. Storck, S.E.; Meister, S.; Nahrath, J.; Meißner, J.N.; Schubert, N.; Di Spiezio, A.; Baches, S.; Vandenbroucke, R.E.; Bouter, Y.; Prikulis,
I. Endothelial lrp1 transports amyloid-β 1–42 across the blood-brain barrier. J. Clin. Investig. 2016, 126, 123–136. [CrossRef]
[PubMed]
196. Soto, I.; Grabowska, W.A.; Onos, K.D.; Graham, L.C.; Jackson, H.M.; Simeone, S.N.; Howell, G.R. Meox2 haploinsufficiency
increases neuronal cell loss in a mouse model of alzheimer’s disease. Neurobiol. Aging 2016, 42, 50–60. [CrossRef] [PubMed]
197. Nixon, R.A.; Yang, D.-S. Autophagy failure in alzheimer’s disease—Locating the primary defect. Neurobiol. Dis. 2011, 43, 38–45.
[CrossRef] [PubMed]
198. Nilsson, P.; Loganathan, K.; Sekiguchi, M.; Matsuba, Y.; Hui, K.; Tsubuki, S.; Tanaka, M.; Iwata, N.; Saito, T.; Saido, T.C. Aβ
secretion and plaque formation depend on autophagy. Cell Rep. 2013, 5, 61–69. [CrossRef] [PubMed]
199. Nilsson, P.; Sekiguchi, M.; Akagi, T.; Izumi, S.; Komori, T.; Hui, K.; Sörgjerd, K.; Tanaka, M.; Saito, T.; Iwata, N. Autophagy-related
protein 7 deficiency in amyloid β (aβ) precursor protein transgenic mice decreases aβ in the multivesicular bodies and induces
aβ accumulation in the golgi. Am. J. Pathol. 2015, 185, 305–313. [CrossRef] [PubMed]
200. Hara, T.; Nakamura, K.; Matsui, M.; Yamamoto, A.; Nakahara, Y.; Suzuki-Migishima, R.; Yokoyama, M.; Mishima, K.; Saito, I.;
Okano, H. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature 2006, 441, 885–889.
[CrossRef]
201. Komatsu, M.; Waguri, S.; Chiba, T.; Murata, S.; Iwata, J.-i.; Tanida, I.; Ueno, T.; Koike, M.; Uchiyama, Y.; Kominami, E. Loss of
autophagy in the central nervous system causes neurodegeneration in mice. Nature 2006, 441, 880–884. [CrossRef]
202. Harris, H.; Rubinsztein, D.C. Control of autophagy as a therapy for neurodegenerative disease. Nat. Rev. Neurol. 2012, 8, 108–117.
[CrossRef] [PubMed]
203. Liu, J.; Li, L. Targeting autophagy for the treatment of alzheimer’s disease: Challenges and opportunities. Front. Mol. Neurosci.
2019, 12, 203. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2024, 25, 6901 28 of 28
204. Du, Y.; Wooten, M.C.; Gearing, M.; Wooten, M.W. Age-associated oxidative damage to the p62 promoter: Implications for
alzheimer disease. Free Radic. Biol. Med. 2009, 46, 492–501. [CrossRef] [PubMed]
205. Rohn, T.T.; Wirawan, E.; Brown, R.J.; Harris, J.R.; Masliah, E.; Vandenabeele, P. Depletion of beclin-1 due to proteolytic cleavage
by caspases in the alzheimer’s disease brain. Neurobiol. Dis. 2011, 43, 68–78. [CrossRef] [PubMed]
206. Esteves, A.; Palma, A.; Gomes, R.; Santos, D.; Silva, D.; Cardoso, S. Acetylation as a major determinant to microtubule-dependent
autophagy: Relevance to alzheimer’s and parkinson disease pathology. Biochim. Et Biophys. Acta (BBA) Mol. Basis Dis. 2019, 1865,
2008–2023. [CrossRef] [PubMed]
207. Sola-Sevilla, N.; Puerta, E. Sirt2 as a potential new therapeutic target for alzheimer’s disease. Neural Regen. Res. 2024, 19, 124–131.
[CrossRef] [PubMed]
208. Alamro, H.; Thafar, M.A.; Albaradei, S.; Gojobori, T.; Essack, M.; Gao, X. Exploiting machine learning models to identify novel
alzheimer’s disease biomarkers and potential targets. Sci. Rep. 2023, 13, 4979. [CrossRef] [PubMed]
209. Long, J.M.; Holtzman, D.M. Alzheimer disease: An update on pathobiology and treatment strategies. Cell 2019, 179, 312–339.
[CrossRef] [PubMed]
210. Wojtunik-Kulesza, K.; Rudkowska, M.; Orzeł-Sajdłowska, A. Aducanumab—Hope or disappointment for alzheimer’s disease.
Int. J. Mol. Sci. 2023, 24, 4367. [CrossRef]
211. Van Dyck, C.H.; Swanson, C.J.; Aisen, P.; Bateman, R.J.; Chen, C.; Gee, M.; Kanekiyo, M.; Li, D.; Reyderman, L.; Cohen, S.
Lecanemab in early alzheimer’s disease. N. Engl. J. Med. 2023, 388, 9–21. [CrossRef]
212. Cummings, J.; Zhou, Y.; Lee, G.; Zhong, K.; Fonseca, J.; Cheng, F. Alzheimer’s disease drug development pipeline: 2023.
Alzheimer’s Dement. Transl. Res. Clin. Interv. 2023, 9, e12385. [CrossRef] [PubMed]
213. Jadhav, D.; Saraswat, N.; Vyawahare, N.; Shirode, D. Targeting the molecular web of alzheimer’s disease: Unveiling pathways for
effective pharmacotherapy. Egypt. J. Neurol. Psychiatry Neurosurg. 2024, 60, 7. [CrossRef]
214. Peng, Y.; Jin, H.; Xue, Y.-h.; Chen, Q.; Yao, S.-y.; Du, M.-q.; Liu, S. Current and future therapeutic strategies for alzheimer’s disease:
An overview of drug development bottlenecks. Front. Aging Neurosci. 2023, 15, 1206572. [CrossRef]
215. Jones, R.S.; Minogue, A.M.; Fitzpatrick, O.; Lynch, M.A. Inhibition of jak2 attenuates the increase in inflammatory markers in
microglia from app/ps1 mice. Neurobiol. Aging 2015, 36, 2716–2724. [CrossRef] [PubMed]
216. Arnold, S.E.; Arvanitakis, Z.; Macauley-Rambach, S.L.; Koenig, A.M.; Wang, H.-Y.; Ahima, R.S.; Craft, S.; Gandy, S.; Buettner, C.;
Stoeckel, L.E. Brain insulin resistance in type 2 diabetes and alzheimer disease: Concepts and conundrums. Nat. Rev. Neurol.
2018, 14, 168–181. [CrossRef] [PubMed]
217. Abolhassani, N.; Leon, J.; Sheng, Z.; Oka, S.; Hamasaki, H.; Iwaki, T.; Nakabeppu, Y. Molecular pathophysiology of impaired
glucose metabolism, mitochondrial dysfunction, and oxidative DNA damage in alzheimer’s disease brain. Mech. Ageing Dev.
2017, 161, 95–104. [CrossRef]
218. Butterfield, D.A.; Halliwell, B. Oxidative stress, dysfunctional glucose metabolism and alzheimer disease. Nat. Rev. Neurosci.
2019, 20, 148–160. [CrossRef]
219. Lee, J.-H.; Yang, D.-S.; Goulbourne, C.N.; Im, E.; Stavrides, P.; Pensalfini, A.; Chan, H.; Bouchet-Marquis, C.; Bleiwas, C.; Berg, M.J.
Faulty autolysosome acidification in alzheimer’s disease mouse models induces autophagic build-up of aβ in neurons, yielding
senile plaques. Nat. Neurosci. 2022, 25, 688–701. [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.