Next Article in Journal
HSP70 Expression Signature in Renal Cell Carcinoma: A Clinical and Bioinformatic Analysis Approach
Next Article in Special Issue
Whole Exome Sequencing Reveals a Novel Homozygous Variant in the Ganglioside Biosynthetic Enzyme, ST3GAL5 Gene in a Saudi Family Causing Salt and Pepper Syndrome
Previous Article in Journal
The Genetic Side of the Mood: A Scientometric Review of the Genetic Basis of Mood Disorders
Previous Article in Special Issue
Novel Variants in MPV17, PRX, GJB1, and SACS Cause Charcot–Marie–Tooth and Spastic Ataxia of Charlevoix–Saguenay Type Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Common Genetic Factors and Pathways in Alzheimer’s Disease and Ischemic Stroke: Evidences from GWAS

1
China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
2
Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
3
Department of Pharmacology, School of Medical Sciences, Faculty of Medicine & Health, UNSW, Sydney, NSW 2052, Australia
*
Author to whom correspondence should be addressed.
Genes 2023, 14(2), 353; https://doi.org/10.3390/genes14020353
Submission received: 13 December 2022 / Revised: 19 January 2023 / Accepted: 27 January 2023 / Published: 30 January 2023
(This article belongs to the Special Issue Genetics of Complex Human Disease)

Abstract

:
Alzheimer’s disease (AD) and ischemic stroke (IS) are common neurological disorders, and the comorbidity of these two brain diseases is often seen. Although AD and IS were regarded as two distinct disease entities, in terms of different etiologies and clinical presentation, recent genome-wide association studies (GWASs) revealed that there were common risk genes between AD and IS, indicating common molecular pathways and their common pathophysiology. In this review, we summarize AD and IS risk single nucleotide polymorphisms (SNPs) and their representative genes from the GWAS Catalog database, and find thirteen common risk genes, but no common risk SNPs. Furthermore, the common molecular pathways associated with these risk gene products are summarized from the GeneCards database and clustered into inflammation and immunity, G protein-coupled receptor, and signal transduction. At least seven of these thirteen genes can be regulated by 23 microRNAs identified from the TargetScan database. Taken together, the imbalance of these molecular pathways may give rise to these two common brain disorders. This review sheds light on the pathogenesis of comorbidity of AD and IS, and provides molecular targets for disease prevention, manipulation, and brain health maintenance.

1. Introduction

In the elderly, brain health is mainly jeopardized by dementia or stroke [1]. About 60–80% of dementia cases are Alzheimer’s disease (AD), which is a progressive and irreversible neurodegenerative disorder. Ischemic stroke (IS) accounts for almost 80% of total stroke cases [2], and it is a leading cause of disability and mortality in the elderly worldwide. Pathological studies have shown that the comorbidity of AD and IS is common, infarctions were present in 51.3% of patients with probable AD and 62.5% of patients with possible AD [3]. However, whether these two common brain disorders have the same underlying molecular mechanisms is controversial [4,5,6,7,8].
Clinically, AD is characterized by the inability to recall recent events, changes in personality, and difficulty in solving problems, at the disease’s early stage. AD patients may develop behavioral changes, confusion, disorientation, communication impairment, and diminished social skills at a later stage, and eventually, the patients may have difficulties in speaking, swallowing, and walking. Pathologically, AD is mainly characterized by hippocampal neuronal loss, amyloid β (Aβ) extracellular deposition forming senile plaques, and the formation of intracellular neurofibrillary tangles. Clinically, IS can present with a variety of physical and cognitive manifestations depending on the brain areas affected and associated with neuron lysis and death [9]. Multiple biological processes and molecular pathways are involved in both AD and IS. Vascular dysfunction (hypertension, hyperlipidemia, diabetes, coronary artery disease, etc.) was considered to have significant direct and indirect impacts on neurodegeneration in AD [10], and the presence of hyperlipidemia had a direct influence on the neuronal tau uptake in the entorhinal cortex [11]. The early appearance of vascular dysfunction in the preclinical stage of AD indicates that vascular events might be a part of the cause, rather than a passive contribution, of AD [12]. In addition, AD before IS was considered to be an independent predictor of death in patients under 80 years old [13]. The shared risk factors and protective factors between AD and IS were also identified. The risk factors include low education, sedentary lifestyle, heart diseases, advancing age [14,15], diabetes [16,17], and obesity [18,19], while the protective factors include high levels of psychological well-being, a rich social network, and active engagement in leisure activities [20].
Genetic risk factors play an important susceptible role in both AD and IS [6]. Genome-wide association studies (GWASs) with a large sample size, and their meta-analyses, have identified many AD and IS susceptible single nucleotide polymorphisms (SNPs) and genetic loci [21,22]. Through comparing the causal and susceptible genes of AD and IS by reviewing familial databases and GWAS databases, we aim to identify the common genetic contributors, summarize the common pathways, microRNAs (miRNAs) within the common mapped genes, and eventually provide molecular targets for the improvement of brain health.

2. Searching Methodology

The literature was reviewed by searching the GWAS Catalog database (https://www.ebi.ac.uk/gwas/, accessed on 14 September 2022 from the database’s inception to 14 September 2022. The GWAS Catalog data was extracted from the published GWASs, identified through a weekly PubMed search (https://pubmed.ncbi.nlm.nih.gov/, accessed on 14 September 2022). In the database, the search terms used were: “Alzheimer’s disease” and “ischemic stroke”. The information on risk SNPs and their mapped genes was extracted and aligned. If the SNP was located within a gene, that gene was listed, with multiple overlapping genes separated into several genes. If the gene was intergenic, the upstream and downstream genes were listed.
For familial genetic studies, the vascular neuropathologies of cases carrying familial causative AD genes were reviewed, and genetic variants of AD causative genes in relation to IS were investigated. Vice versa, the AD neuropathologies of cases carrying familial IS genes were reviewed, and genetic variants of IS causative genes in relation to AD were analyzed through a weekly Pubmed search.
Information on the pathways that the gene products were involved in was sourced from the GeneCards database (version 5.12, https://www.genecards.org/, accessed on 17 September 2022). The section “pathways” provides links to pathways according to information extracted from Cell Signaling Technology, R&D Systems, GeneGo (Thomson Reuters), Reactome, Sino Biological, Tocris Bioscience, PharmGKB, Qiagen, GeneTex, BosterBio, MedChemExpress, WikiPathways and PubChem, SuperPathways from PathCards, the protein-protein interaction network was derived based on the STRING database (https://cn.string-db.org/, accessed on 18 September 2022). We identified the miRNAs that regulate the target genes through the TargetScan database (https://www.targetscan.org/vert_80/, accessed on 18 September 2022). The molecular selection processes are shown in a flow chart (Figure 1).

3. The Roles of AD or IS Causative Genes in Its Counterpart

APP, PSEN1, and PSEN2 genes, are causal genes for AD, which encode proteins called amyloid precursor protein (APP), presenilin1 (PSEN1), and presenilin 2 (PSEN2), respectively. The Apolipoprotein E (ApoE) gene is a major player in sporadic AD. The major hereditary IS diseases are cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), cerebral autosomal recessive arteriopathy/arteriosclorosis with subcortical infarcts and leukoencephalopathy (CARASIL), and Fabry disease, which are caused by mutations in the notch receptor 3 (Notch3), htrA serine peptidase 1 (HTRA1), and galactosidase alpha (GLA) genes, respectively.

3.1. AD Causative Genes in IS

No study has found a direct effect of APP, PSEN1, or PSEN2 mutations on the risk of IS. However, they may influence the risk of IS through their effects on lipid metabolism [23]. When evaluating the protein oxidation and lipid peroxidation in the brain from knock-in mice expressing mutant human APP and PSEN1, it was observed that knock-in mice displayed increased oxidative stress, which is independent of dietary cholesterol [24]. Lipid peroxidation is closely related to endothelial dysfunction, and finally contributes to atherosclerosis and IS [25]. Statins are widely used drugs that elicit plaque stabilization and reduce inflammation in atherosclerotic plaque, and also reduce Aβ in human neurons through decreasing the generation of soluble APPβ and increasing the levels of full-length APP, indicating that AD and IS may have the same therapeutic target [26].
ApoE has been consistently considered as the major genetic risk for sporadic AD and cognitive decline post-IS [27]. The ApoE ε4 is associated with an increased number of Aβ plaques, but we did not find ApoE related to IS in the GWAS database, although ApoE has been shown to be associated with IS risk conditions, such as diabetes [28] and low density lipoprotein cholesterol (LDL-C) [29], which may finally lead to IS.

3.2. IS Causative Genes in AD

It has been suggested that Notch3 and HTRA1 are associated with AD [30,31]. Among 5617 participants with AD and 4594 controls, Notch3 rs149307620 allele missense mutation was observed in 10 participants with AD but not in the controls [30]. Notch3 was also an important hit in the gene-based analysis (combined effect of all Notch3 variants) of AD, suggesting its potential role as a modifier of AD [31]. It was found that the T allele of HTRA1 rs2293871 upregulated HTRA1 expression and was associated with an increased risk of AD and AD-by proxy [32]. The deficiency or increase of GLA induced by gene variations has not been shown to be associated with AD.

4. Common Susceptibility Genes of AD and IS

We found there were thirteen common mapped genes in AD and IS, although no common risk SNPs were found. The common genes are ALDH1A2, ANKRD22, ANTXR1, DIO2-AS1, HDAC9, JH3, KCNN3, LNC-LBCS, MMP3, MMP12, PCSK6, RBMS3, and RNU6-909P. Among them, ten genes are protein coding genes, including ALDH1A2, ANKRD22, ANTXR1, HDAC9, JPH3, KCNN3, MMP3, MMP12, PCSK6, and RBMS3. The shared genetic studies of AD and IS are summarized in Table 1. Among the thirteen genes, HDAC9, MMP3, and MMP12 have been relatively well studied in AD and IS.

4.1. HDAC9

Histone deacetylase 9 (HDAC9) is responsible for the lysine deacetylation residues on the N-terminal part of the core histones. Histone deacetylation is involved in epigenetic repression, and plays a significant role in transcriptional regulation, cell cycle progression, and some developmental events. In AD, the expression of HDAC9 was significantly reduced in the sampled prefrontal and visual cortices [46]. However, the level of blood HDAC9 mRNA expression was increased in AD twins compared to healthy twins [47]. The circRNA HDAC9 (circHDAC9) decreased miRNA (miR)-138 expression, which reversed excessive Aβ production, but circHDAC9 was reduced in AD patients [48]. Thus, in AD, whether the risk allele (A) of rs117756856 is associated with the increased or decreased expression of HDAC9 is not clear. An in vitro study showed that HDAC9 inhibition had neuroprotective effects on IS by inhibiting inflammation [49], which confirmed that depletion of HDAC9 could reduce cerebral injury in experimental stroke. In addition, HDAC9 contributed to brain micro-vessel endothelial cell dysfunction in IS, evidenced by reduced tight-junction proteins’ expression, endothelial cell permeability dysfunction, increased inflammatory responses, and cellular apoptosis [50]. It has also been demonstrated that a higher methylation level of HDAC9 is associated with a lower risk of IS [51]. The risk allele of rs2107595 increased the risk of atherosclerotic stroke through interaction with the HDAC9 promoter and increased the transcription capacity, which was related to higher HDAC9 mRNA levels [52]. Pathways associated with the risk alleles of rs2107595 are involved in cholesterol efflux, platelet aggregation, and IL-6 signaling [53]. Thus, the risk alleles of rs2107595, together with rs11984041, rs2023938, and rs71524263, may be associated with an increased expression of HDAC9 contributing to IS.

4.2. MMP3 and MMP12

Matrix metalloproteinase 3 (MMP3) and matrix metalloproteinase 12 (MMP12) are two members of the matrix metalloproteinases (MMPs) family, which are both involved in the breakdown of the extracellular matrix (ECM). Microglia surround the Aβ plaques, provoke an inflammatory response, and contribute to neuronal cell loss. Aβ1-42 induces the upregulation of MMP3 and MMP12 in microglia, which further enhances the inflammatory processes and accelerates the progression of AD [54]. The risk alleles of rs12808148 may be associated with the increase of MMP3 and MMP12 in AD. In carotid atherosclerosis plaques, MMP3 and MMP12 may play an important role in the plaque stability, and their low content in plaque is related to a higher risk of ipsilateral stroke [55]. A previous study indicated the causal link between lower serum MMP12 levels and a higher risk of IS [56], and MMP3 exerted a negative effect on the progression of IS [57]. The risk allele of rs72983521 may be associated with MMP3 and MMP12 levels in different sites, and thus is associated with IS.

4.3. Other Genes

ANTXR Cell Adhesion Molecule 1 (ANTXR1) is a type I transmembrane protein and a tumor-specific endothelial marker. The higher peptide from ANTXR1 showed a significant ability to discriminate AD patients from healthy controls, which suggested that the risk allele of rs7561207 may increase the risk of AD through enhancing the expression of ANTXR1 [58]. Ankyrin repeat domain 22 (ANKRD22) is a nuclear-encoded mitochondrial membrane protein. The expression of ANKRD22 in AD patients was significantly lower than those of normal controls. However, ANKRD22 stimulated the cytotoxic effect of Aβ and reduced hippocampal neuronal cell viability in an AD cell model [59]. The T allele of rs147285445 may influence the risk of AD differently in vivo and vitro. Proprotein convertase Subtilisin/Kexin type 6 (PCSK6) is a protease in the extracellular matrix and is expressed in many tissues, including the brain. The levels of PCSK6 were increased in the fibrous caps of symptomatic carotid plaques. PCSK6 is possibly involved in the inflammation, ECM remodeling, and dysregulation of smooth muscle cell proliferation in atherosclerosis, and thus could lead to IS [60]. The risk allele of rs528002287 may enhance the expression of PCSK6 and increase the risk of IS. The relationships of the aldehyde dehydrogenase 1 family member A2 (ALDH1A2), DIO2 antisense RNA 1 (DIO2-AS1), JH3, potassium calcium-activated channel subfamily N member 3 (KCNN3), long noncoding RNAs bladder and prostate cancer suppressor (LNC-LBCS), RNA binding motif single stranded interacting protein 3 (RBMS3) and RNA, U6 small nuclear 909, and pseudogene (RNU6-909P) genes, and AD, IS were only observed in populational studies and have not been confirmed in molecular studies yet.
To address the common AD and IS risk genes products in AD important molecules, a STRING analysis was performed. The thirteen common AD and IS risk genes in the AD important molecules network were identified. MMP3 and MMP12 links with ApoE, indicating that ApoE may associate with IS through connecting with MMP3 and MMP12 (Figure 2).

5. Common Molecular Pathways

We observed three common pathways with at least three gene products related together. The HDAC9, MMP3, and MMP12 gene products are involved in the G protein-coupled receptors (GPCR) pathway. Both HDAC9 and MMP3 gene products participate in the macrophage-migration inhibitory factor (MIF) mediated glucocorticoid regulation pathway, both MMP3 and MMP12 gene products are involved in the transendothelial migration of leukocytes pathway, and the two pathways are both immune related pathways. MMP3, ALDH1A2, HDAC9, and PCSK6 gene products participate in the signal transduction pathway; ALDH1A2 and MMP3 gene products are involved in the estrogen receptor (ESR)-mediated signaling and signaling by nuclear receptors pathways; ALDH1A2 and HDAC9 gene products are involved in the ethanol effects on histone modifications pathway; both MMP3 and MMP12 gene products participate in the urokinase-type plasminogen activator (uPA) and urokinase plasminogen activator receptor (uPAR)-mediated signaling and UPA-UPAR pathway, and these six pathways are signal transduction pathways (Supplementary Table S1).

5.1. Inflammation and Immunity

Immunity is the protection against a disease generated by immunization, previous infection, or other non-immunologic factors. Inflammation plays an important part in immunity. Inflammation is a process of removing damaged cells, infectious microorganisms, and starting to heal. When the trigger of the response is neutralized, immune cells change their activity towards a pro-resolution status via anti-inflammatory signaling. After a proper response, immune cells are recruited to the place where the attack occurs by pro-inflammatory signaling pathways. But when it becomes dysfunctional and chronic, systemic inflammation is an important factor in multiple diseases [61]. Increasing evidence has shown that peripheral and neuroinflammation are the main causes of various neuropsychiatric diseases, including AD and IS.
Neuritis aggravates brain injury, resulting in neuronal degeneration and synaptic dysfunction. Recent studies have shown that inflammation is one of the important factors in the pathogenesis of AD [62]. The type and severity of brain tissue damage is one of the important factors that determines the inflammatory mode of AD. Although the purpose of the initial inflammatory response is to protect the body from the effects of stress factors, if the duration or level of the stimulation is too high, it may cause damage [63]. After Aβ deposition, the pathological adaptations stimulate the release of pro-inflammatory cytokines [interleukin-6 (IL-6), interleukin-1β (IL-1β) or tumor necrosis factor-α (TNF-α)] and other pro-inflammatory molecules [macrophage inflammatory protein, monocyte chemoattractant protein, coagulation factor, reactive oxygen species (ROS), nitric oxide, protease, protease inhibitor, etc.], and some prostaglandin, thromboxane, leukotriene, and C-reactive protein (CRP) from glial cells [64]. Activated microglia respond to the Aβ, resulting in migration to the plaques as well as phagocytosis of the Aβ. However, when the microglia become enlarged, or after prolonged periods, they are no longer able to phagocytose the Aβ. Then peripheral macrophages may migrate to Aβ plaque deposition to clear the Aβ. However, peripheral macrophages recruitment into the brain is likely to exacerbate the effects of sustained inflammation and results in an exacerbation of AD pathology [65]. The deterioration of the environment will lead to additional Aβ accumulation and pro-inflammatory molecules [64], which will release reactive substances such as nitric oxide, proteolytic enzymes, excitatory amino acids and complementary factors, and cause damage to the adjacent neurons [66,67]. Although inflammation is mainly considered to be what happens after IS, it also has a close relationship with atherosclerosis, which is a key risk factor for IS. Numerous studies have shown that atherosclerosis is initiated by endothelial injury or LDLs accumulation within the arterial vascular wall, which generally involves in oxidization or modification. These modified or oxidized LDLs, and low-grade inflammation are caused by small endothelial injuries, activate innate and adaptive immune responses. These immune responses play important roles in the development of atherosclerosis [68]. Monocytes/macrophages, neutrophils, B lymphocytes, and T lymphocytes are the major cell subtypes in the components of atherosclerosis [69]. The increased levels of IL-6, CRP and lipoprotein-associated phospholipase A2 (Lp-PLA2) are related to an increased risk of IS [70].
MIF is a cytokine released from T-lymphocytes and macrophages stimulated by glucocorticoids. MIF counter-regulates the inhibitory effects of glucocorticoids on pro-inflammatory cytokines (IL-6, IL-8, IL-1β and TNF-α), and overcomes glucocorticoid’s inhibition of T-cell proliferation [71]. MIF may exacerbate the effects of sustained inflammation and leads to additional Aβ accumulation. In addition, T-lymphocytes are major components of atherosclerosis [72]. Increased levels of IL-6 are related to an increased risk of IS. Thus, MIF mediated glucocorticoid regulation is connected with an increased risk of both AD and IS. In the functional enrichment analysis of the 49 differently expressed genes among AD and control brain samples, transendothelial migration of leukocytes was enriched in the AD group [73]. Transendothelial leukocyte migration is a key step in the progression of vascular inflammation, the underlying molecular pathogenesis of atherosclerosis [74]. Moreover, the differently expressed long noncoding RNAs (lncRNAs) in IS are also mainly related to transendothelial leukocyte migration [75]. These indicate that transendothelial leukocyte migration is closely associated with both AD and IS.
The inflammatory responses are initiated locally under aberrant local conditions in both AD and IS, and elicited by the immune system (Figure 3). Therefore, AD and IS may have common biological pathways involved in the immune system.

5.2. GPCR

GPCRs are the largest membrane protein family of seven-transmembrane receptors in humans. They are involved in neuronal signal transduction in response to various extracellular signals such as hormones and neurotransmitters [76]. GPCRs may participate in AD pathology through three aspects: the amyloid hypothesis, the tau hypothesis, and the cholinergic hypothesis [77]. Microglia express several GPCRs to regulate microglial activation and its polarization status. Microglial GPCRs are involved in Aβ generation, degradation, clearance, and trigger multiple inflammatory pathways in response to Aβ [78]. Family C GPCRs also play important roles in Aβ. Family C of GPCRs contains the calcium-sensing receptor (CaSR), GABAB receptors (GABABRs), and other receptors. CaSRs are involved in the neurotransmitter system of human cortical astrocytes and neurons in vitro. The specific binding of Aβs to CaSRs hinders the release of soluble APP-α peptide and kills human cortical neurons [23]. GABABRs block the Aβ peptides’ synthesis and prevent neuronal hyperexcitability [79]. In AD, GPCRs are involved in tau phosphorylation via various downstream kinases including glycogen synthase kinase-3β (GSK-3β), cyclin-dependent kinase-5 (CDK-5), and extracellular signal-regulated kinases (ERKs) signaling cascade [80,81,82]. CX3C chemokine receptor 1 (CX3CR1) receptor is a microglia chemokine GPCR. Its binding and interaction with tau lead to the internalization of tau into microglia [83]. These GPCRs both promote and inhibit tau phosphorylation. In the cholinergic hypothesis, cholinergic dysfunction is characterized by reduced acetylcholine release and impaired coupling of muscarinic acetylcholine receptors (mAChRs) to heterotrimeric guanosine triphosphate (GTP)-binding proteins (G proteins). Cholinergic dysfunction is also associated with Aβ accumulation [84]. Atherosclerosis and type 2 diabetes are the risk factors for IS. GPCRs influence the risk of IS by affecting its risk factors. Lysophospholipids (LPLs) are second-generation bioactive lipid-derived signaling molecules. GPCRs mediate the biological effects of LPLs in the development of atherosclerosis [85]. β-cells and enteroendocrine cells are essential cells for insulin secretion modulation through expressing numerous GPCRs. GPCRs specific for free fatty acid ligands (lipid GPCRs) are the target for the treatment of type 2 diabetes because of their function in islet and gut hormone secretion [86]. Therefore, different GPCRs may have different influences on AD and IS (Figure 4).

5.3. Signal Transduction

The occurrence of both AD and IS are tightly regulated by a multitude of signal transduction pathways. Nuclear receptors generally act as ligand-activated transcription factors [87]. They regulate gene expression through binding to the specific ligand, and present as therapeutic targets in AD and IS. The receptor interacting protein-140 (RIP140) is known as a cofactor for some nuclear receptors. The overexpression of RIP140 was shown to reduce the generation of Aβ by decreasing the transcription of β-APP cleaving enzyme (BACE1) [88]. Elevated RIP140 also increases the risk of insulin resistance and atherosclerosis, which are risk factors of IS [89]. Retinoids binds to the specific nuclear receptor, such as retinoid X receptors (RXRs) and retinoic acid receptors, to regulate the expression of a variety of genes that code for enzymes, receptors, neuropeptide hormone, etc., which are responsible for slowing down the accumulation of amyloids, reducing neurodegeneration, and preventing pathogenesis of AD [90]. RXRs also negatively regulate the platelet functional responses and thrombus formation, which might delay the onset of IS [91]. Liver X receptor (LXR), also known as nuclear receptor subfamily 1 group H member 3 (NR1H3), is a member of the nuclear receptor superfamily of ligand-activated transcription factors, and plays a central role in the transcriptional control of cholesterol homeostasis. The C allele of rs7120118 of NR1H3 gene was shown to reduce the risk of AD, and the soluble Aβ42 levels were significantly reduced in the temporal cortex of patients with the CC genotype [92]. In an AD mice model, LXR activation restored microvascular morphology through decreasing tortuosity and increasing length, which were associated with decreased deposition of perivascular Aβ [93]. The TT genotype of LXRα rs2279238 is significantly associated with advanced carotid atherosclerosis, suggesting that this polymorphism may act as a genetic risk factor for atherosclerotic stroke [94]. Ex vivo LXR agonist treatment decreases early atherosclerosis in LDL receptor-deficient mice through inhibiting monocyte to endothelial adhesion [95]. In addition, the deficiency of LXR led to an increase in atherosclerosis, with enhanced inflammation in foam cells of atherosclerotic plaques [96]. Peroxisome proliferator-activated receptor γ (PPAR-γ) is a nuclear receptor that plays a crucial role in glucose and lipid homeostasis in the central nervous system. The PPAR-γ agonist pioglitazone significantly increased PPAR-γ expression, lowered amyloid-β levels, and improved the antioxidative capacity in the cortex of AD mice [97]. The PPAR-γ gene rs1801282 GG genotype may be associated with an increased risk of IS [98]. The PPAR-γ agonist improved the dyslipidemic profile and inflammatory status in atherosclerotic lesions in rats [99]. The nuclear receptor subfamily 4A 2 (NR4A2, also known as Nurr1) plays important roles in diverse brain functions and its overexpression alleviated AD pathology changes, including Aβ deposition and neuronal loss [100] in AD mice. The expression of nuclear receptor subfamily 4 group A member 1 (NR4A1, also known as Nur77) was significantly increased in the hippocampus of AD mice; the overexpression of NR4A1 promoted amyloidogenesis and accelerated tau hyperphosphorylation [101]. The absence of Nur77 in macrophages led to upregulated toll-like receptor signaling, and imbalanced macrophage polarization toward the proinflammatory M1 phenotype, indicating that Nur77 is an important target for modulating the inflammatory phenotype of macrophages and regulation of atherogenesis [102].
In AD hypothalamic medial mamillary (MMN), a higher nuclear ESRα intensity was significantly associated with larger nuclear and perikaryal sizes, indicating nuclear ESRα may mediate extra activation in MMN that acts as a unique brain area to prevent neurodegeneration [103]. In AD human brain tissue, ESRα co-localized with neurofibrillary pathology, and their interaction interrupted estrogen signaling, demonstrating that sequestration of ESRα by tau pathology decreased the neuroprotective role of estrogen in AD [104]. In both female aorta surgical sample and bilateral ovariectomized female ApoE -/- mice samples, assay results indicated that estrogen prevented atherosclerosis through upregulating ESRα expression to induce ESRα-mediated activation of autophagy and reduce inflammation and cell pyroptosis [105]. In addition, in an ovariectomized ApoE -/- mice model, it was demonstrated that ESRα inhibited the synthesis and secretion of proprotein convertase subtilisin/kexin type 9 (PCSK9) and subsequently lowered the accumulation of cholesterol and triglyceride to prevent post-menopausal atherosclerosis [106]. Hyperinsulinemia and insulin resistance are important causes of atherosclerosis. Insulin indirectly reduced the expression of ESRα, and thus interfered with estrogen regulation of vascular smooth muscle cells’ proliferation, leading to atherosclerosis [107].
Urokinase-type plasminogen gene rs2227564 C-positive genotype (CC+CT) has been reported to associate with a higher risk of developing AD [108]. Neuronal uPA protected the synapse from the deleterious effects of soluble Aβ. However, in the frontal cortex of an AD human brain and 5xFAD mice, uPA was decreased, leaving the deleterious effects of Aβ on the synapse unaffected [109]. UPAR was highly expressed in human symptomatic carotid endarterectomies, and mainly found in ruptured plaque segments, suggesting that UPAR may be connected with plaque rupture in the progression of symptomatic atherosclerotic lesions [110]. UPA stimulated the differentiation of monocytes into macrophages, prolonged the macrophage survival in the atherosclerotic lesion, increased lesion cellularity, and eventually accelerated lesion development [111]. The role of ethanol effects on histone modifications in AD and IS has not been reported.

6. Common miRNAs

At least nine of these thirteen genes can be regulated by 23 miRNAs identified from the TargetScan database. Among the 23 miRNAs, four miRNAs regulate eight genes, including hsa-miR-204-5p, hsa-miR-211-5p, hsa-miR-548c-3p, and hsa-miR-660-3p; and the other miRNAs regulate seven genes. Among them, we found that thirteen miRNAs were reported through a Pubmed search using the key words of each miRNA (Supplementary Table S2). However, only two miRNAs were reported to be related to AD and/or IS. Hsa-miR-204-5p is the product of the MIR204 gene, regulating ALDH1A2, ANKRD22, ANTXR1, HDAC9, JH3, KCNN3, MMP3, and RBMS3. Hsa-miR-664b-3p is the product of MIR664B, regulating ALDH1A2, ANKRD22, HDAC9, KCNN3, MMP3, MMP12, PCSK6, and RBMS3. Heavy metals like Pb acetate increased the expression of has-miR-204-5p, deteriorated the cognitive functions, and were associated with the overexpression of tau via pathways of neurodegeneration-multiple diseases [112]. Has-miR-204-5p also participated in mixed B vitamins’ better cognitive functions in AD, through linking to both mixed B vitamins and cognitive function related genes [113]. Hsa-miR-664b-3p was negatively associated with lead exposure and was decreased in AD human brain tissue compared with controls, and its target genes participated in potentially AD relevant pathways [114]. It was upregulated in human vascular smooth muscle cells (hVSMCs) during replicative senescence. Although hsa-miR-664b-3p was not functionally well annotated in hVSMCs to date, it may play a role in the adaptive immune system and toll-like receptor signaling 7/8/9 [115]. It may be associated with vascular aging and atherosclerosis, contributing to IS. Hsa-miR-664b-3p also regulates APP, PSEN1, MAPT, and HTRA1 (Supplementary Table S3).

7. Advantages and Limitations

Compared with other studies (Table 2), our study has summarized all up-to-date AD and IS GWAS studies to identify the common risk SNPs and related genes. In addition, we clustered the common molecular pathways indicated by the shared genes into three common potential pathogeneses for AD and IS. Finally, we searched for the miRNAs that regulated the commonly susceptible genes and identified 23 miRNAs that might regulate both AD and IS. There are some limitations in our study. First, our study was only based on GWAS datasets, so there might be other common susceptibility genes not covered in this review. Second, other types of stroke, such as intracerebral hemorrhage, small vessel disease, etc., might have their own specific genetic factors that are associated with AD, but they were not covered in this review.
In this study, we used genetic data from the GWAS Catalog database and PubMed to determine whether the same genetic loci contribute to AD and IS. We identified thirteen common risk genes, which are involved in three common molecular pathways: immunity, GPCR, and signaling transduction pathways. Furthermore, we identified 23 miRNAs that regulated at least seven of the common risk genes. In summary, there are some common genetic factors and pathways shared by AD and IS, which might be the molecular targets for maintaining good brain health jeopardized by AD or IS.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/genes14020353/s1, Table S1: Common molecular pathways related to the common susceptibility genes; Table S2: Summary of diseases/traits related to the 23 miRNAs in the literature [112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180]; Table S3: The common miRNAs that regulate the shared susceptibility genes of AD and IS.

Author Contributions

Conceptualization, W.D. and Y.H.; methodology, W.D and Y.H.; software, W.D.; validation, W.D. and Y.H.; writing—original draft preparation, W.D.; writing—review and editing, W.D. and Y.H.; supervision, Y.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China (No. 82071417, Y.H.) and neurogenetics for neurodegeneration PI Startup Fund, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (Y.H.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Release notice—Dementia and stroke comorbidity among Canadians aged 65 years and older: Highlights from the Canadian Chronic Disease Surveillance System. Health Promot. Chronic. Dis. Prev. Can. 2020, 40, 324. [CrossRef] [PubMed]
  2. GBD 2016 Stroke Collaborators. Global, regional, and national burden of stroke, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 439–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Schneider, J.A.; Arvanitakis, Z.; Bang, W.; Bennett, D.A. Mixed brain pathologies account for most dementia cases in community-dwelling older persons. Neurology 2007, 69, 2197–2204. [Google Scholar] [CrossRef] [PubMed]
  4. Traylor, M.; Adib-Samii, P.; Harold, D.; Alzheimer’s Disease Neuroimaging Initiative, The International Stroke Genetics Consortium (ISGC), UK Young Lacunar Stroke DNA resource; Dichgans, M.; Williams, J.; Lewis, C.M.; Markus, H.S.; Fornage, M.; Holliday, E.G.; et al. Shared genetic contribution to Ischaemic Stroke and Alzheimer’s Disease. Ann. Neurol. 2016, 79, 739–747. [Google Scholar] [CrossRef] [Green Version]
  5. Cui, P.; Ma, X.; Li, H.; Lang, W.; Hao, J. Shared Biological Pathways Between Alzheimer’s Disease and Ischemic Stroke. Front. Neurosci. 2018, 12, 605. [Google Scholar] [CrossRef]
  6. Wei, C.J.; Cui, P.; Li, H.; Lang, W.J.; Liu, G.Y.; Ma, X.F. Shared genes between Alzheimer’s disease and ischemic stroke. CNS Neurosci. Ther. 2019, 25, 855–864. [Google Scholar] [CrossRef] [Green Version]
  7. Rahman, M.R.; Islam, T.; Shahjaman, M.; Zaman, T.; Faruquee, H.M.; Jamal, M.; Huq, F.; Quinn, J.M.W.; Moni, M.A. Discovering biomarkers and pathways shared by Alzheimer’s disease and ischemic stroke to identify novel therapeutic targets. Medicina 2019, 55, 191. [Google Scholar] [CrossRef] [Green Version]
  8. Wang, T.; Ni, Q.B.; Wang, K.; Han, Z.; Sun, B.L. Stroke and Alzheimer’s disease: A mendelian randomization study. Front. Genet. 2020, 11, 581. [Google Scholar] [CrossRef]
  9. Feske, S.K. Ischemic stroke. Am. J. Med. 2021, 134, 1457–1464. [Google Scholar] [CrossRef]
  10. Jack, C.R., Jr.; Bennett, D.A.; Blennow, K.; Carrillo, M.C.; Dunn, B.; Haeberlein, S.B.; Holtzman, D.M.; Jagust, W.; Jessen, F.; Karlawish, J.; et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018, 14, 535–562. [Google Scholar] [CrossRef]
  11. Vemuri, P.; Lesnick, T.G.; Przybelski, S.A.; Knopman, D.S.; Lowe, V.J.; Graff-Radford, J.; Roberts, R.O.; Mielke, M.M.; Machulda, M.M.; Petersen, R.C.; et al. Age, vascular health, and Alzheimer disease biomarkers in an elderly sample. Ann. Neurol. 2017, 82, 706–718. [Google Scholar] [CrossRef] [PubMed]
  12. Govindpani, K.; McNamara, L.G.; Smith, N.R.; Vinnakota, C.; Waldvogel, H.J.; Faull, R.L.; Kwakowsky, A. Vascular dysfunction in Alzheimer’s iisease: A prelude to the pathological process or a consequence of it? J. Clin. Med. 2019, 8, 651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Zupanic, E.; von Euler, M.; Winblad, B.; Xu, H.; Secnik, J.; Kramberger, M.G.; Religa, D.; Norrving, B.; Garcia-Ptacek, S. Mortality after ischemic stroke in patients with Alzheimer’s disease dementia and other dementia disorders. J. Alzheimers Dis. 2021, 81, 1253–1261. [Google Scholar] [CrossRef]
  14. Hoyer, S. Age as risk factor for sporadic dementia of the Alzheimer type? Ann. N. Y. Acad. Sci. 1994, 719, 248–256. [Google Scholar] [CrossRef] [PubMed]
  15. Hunter, E.; Kelleher, J.D. Age specific models to capture the change in risk factor contribution by age to short term primary ischemic stroke risk. Front. Neurol. 2022, 13, 803749. [Google Scholar] [CrossRef] [PubMed]
  16. Cummings, J.; Ortiz, A.; Castellino, J.; Kinney, J. Diabetes: Risk factor and translational therapeutic implications for Alzheimer’s disease. Eur. J. Neurosci. 2022, 56, 5727–5757. [Google Scholar] [CrossRef]
  17. Khoury, J.C.; Kleindorfer, D.; Alwell, K.; Moomaw, C.J.; Woo, D.; Adeoye, O.; Flaherty, M.L.; Khatri, P.; Ferioli, S.; Broderick, J.P.; et al. Diabetes mellitus: A risk factor for ischemic stroke in a large biracial population. Stroke 2013, 44, 1500–1504. [Google Scholar] [CrossRef] [Green Version]
  18. Lloret, A.; Monllor, P.; Esteve, D.; Cervera-Ferri, A.; Lloret, M.A. Obesity as a risk factor for Alzheimer’s disease: Implication of leptin and glutamate. Front. Neurosci. 2019, 13, 508. [Google Scholar] [CrossRef]
  19. Cong, X.; Liu, S.; Wang, W.; Ma, J.; Li, J. Combined consideration of body mass index and waist circumference identifies obesity patterns associated with risk of stroke in a Chinese prospective cohort study. BMC Public Health 2022, 22, 347. [Google Scholar] [CrossRef]
  20. Wang, R.; Qiu, C.; Dintica, C.S.; Shang, Y.; Calderon Larranaga, A.; Wang, H.X.; Xu, W. Shared risk and protective factors between Alzheimer’s disease and ischemic stroke: A population-based longitudinal study. Alzheimers Dement. 2021, 17, 191–204. [Google Scholar] [CrossRef]
  21. Wightman, D.P.; Jansen, I.E.; Savage, J.E.; Shadrin, A.A.; Bahrami, S.; Holland, D.; Rongve, A.; Borte, S.; Winsvold, B.S.; Drange, O.K.; et al. A genome-wide association study with 1,126,563 individuals identifies new risk loci for Alzheimer’s disease. Nat. Genet. 2021, 53, 1276–1282. [Google Scholar] [CrossRef] [PubMed]
  22. Malik, R.; Chauhan, G.; Traylor, M.; Sargurupremraj, M.; Okada, Y.; Mishra, A.; Rutten-Jacobs, L.; Giese, A.K.; van der Laan, S.W.; Gretarsdottir, S.; et al. Multiancestry genome-wide association study of 520,000 subjects identifies 32 loci associated with stroke and stroke subtypes. Nat. Genet. 2018, 50, 524–537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Armato, U.; Chiarini, A.; Chakravarthy, B.; Chioffi, F.; Pacchiana, R.; Colarusso, E.; Whitfield, J.F.; Dal Pra, I. Calcium-sensing receptor antagonist (calcilytic) NPS 2143 specifically blocks the increased secretion of endogenous Abeta42 prompted by exogenous fibrillary or soluble Abeta25-35 in human cortical astrocytes and neurons-therapeutic relevance to Alzheimer’s disease. Biochim. Biophys. Acta 2013, 1832, 1634–1652. [Google Scholar] [CrossRef] [PubMed]
  24. Mohmmad Abdul, H.; Wenk, G.L.; Gramling, M.; Hauss-Wegrzyniak, B.; Butterfield, D.A. APP and PS-1 mutations induce brain oxidative stress independent of dietary cholesterol: Implications for Alzheimer’s disease. Neurosci. Lett. 2004, 368, 148–150. [Google Scholar] [CrossRef]
  25. Ito, F.; Sono, Y.; Ito, T. Measurement and clinical significance of lipid peroxidation as a biomarker of oxidative stress: Oxidative stress in diabetes, atherosclerosis, and chronic inflammation. Antioxidants 2019, 8, 72. [Google Scholar] [CrossRef] [Green Version]
  26. Langness, V.F.; van der Kant, R.; Das, U.; Wang, L.; Chaves, R.D.S.; Goldstein, L.S.B. Cholesterol-lowering drugs reduce APP processing to Abeta by inducing APP dimerization. Mol. Biol. Cell 2021, 32, 247–259. [Google Scholar] [CrossRef]
  27. Fekih-Mrissa, N.; Klai, S.; Mrad, M.; Mansour, M.; Zaouali, J.; Gritli, N.; Mrissa, R. Apolipoprotein E genotypes associated with Alzheimer disease and concomitant stroke. J. Stroke Cerebrovasc. Dis. 2014, 23, 681–685. [Google Scholar] [CrossRef]
  28. Mahajan, A.; Taliun, D.; Thurner, M.; Robertson, N.R.; Torres, J.M.; Rayner, N.W.; Payne, A.J.; Steinthorsdottir, V.; Scott, R.A.; Grarup, N.; et al. Fine-mapping type 2 diabetes loci to single-variant resolution using high-density imputation and islet-specific epigenome maps. Nat. Genet. 2018, 50, 1505–1513. [Google Scholar] [CrossRef] [Green Version]
  29. Chitnis, T.; Banwell, B.; Kappos, L.; Arnold, D.L.; Gucuyener, K.; Deiva, K.; Skripchenko, N.; Cui, L.Y.; Saubadu, S.; Hu, W.; et al. Safety and efficacy of teriflunomide in paediatric multiple sclerosis (TERIKIDS): A multicentre, double-blind, phase 3, randomised, placebo-controlled trial. Lancet Neurol. 2021, 20, 1001–1011. [Google Scholar] [CrossRef]
  30. Patel, D.; Mez, J.; Vardarajan, B.N.; Staley, L.; Chung, J.; Zhang, X.; Farrell, J.J.; Rynkiewicz, M.J.; Cannon-Albright, L.A.; Teerlink, C.C.; et al. Association of Rare Coding Mutations with Alzheimer Disease and Other Dementias Among Adults of European Ancestry. JAMA Netw. Open 2019, 2, e191350. [Google Scholar] [CrossRef]
  31. Sassi, C.; Nalls, M.A.; Ridge, P.G.; Gibbs, J.R.; Lupton, M.K.; Troakes, C.; Lunnon, K.; Al-Sarraj, S.; Brown, K.S.; Medway, C.; et al. Mendelian adult-onset leukodystrophy genes in Alzheimer’s disease: Critical influence of CSF1R and NOTCH3. Neurobiol. Aging 2018, 66, 179.e17–179.e29. [Google Scholar] [CrossRef] [PubMed]
  32. Liu, G.; Zhang, H.; Liu, B.; Ji, X. Rs2293871 regulates HTRA1 expression and affects cerebral small vessel stroke and Alzheimer’s disease. Brain 2019, 142, e61. [Google Scholar] [CrossRef] [PubMed]
  33. Schwartzentruber, J.; Cooper, S.; Liu, J.Z.; Barrio-Hernandez, I.; Bello, E.; Kumasaka, N.; Young, A.M.H.; Franklin, R.J.M.; Johnson, T.; Estrada, K.; et al. Genome-wide meta-analysis, fine-mapping and integrative prioritization implicate new Alzheimer’s disease risk genes. Nat. Genet. 2021, 53, 392–402. [Google Scholar] [CrossRef] [PubMed]
  34. Carty, C.L.; Keene, K.L.; Cheng, Y.C.; Meschia, J.F.; Chen, W.M.; Nalls, M.; Bis, J.C.; Kittner, S.J.; Rich, S.S.; Tajuddin, S.; et al. Meta-Analysis of Genome-Wide Association Studies Identifies Genetic Risk Factors for Stroke in African Americans. Stroke 2015, 46, 2063–2068. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Mez, J.; Chung, J.; Jun, G.; Kriegel, J.; Bourlas, A.P.; Sherva, R.; Logue, M.W.; Barnes, L.L.; Bennett, D.A.; Buxbaum, J.D.; et al. Two novel loci, COBL and SLC10A2, for Alzheimer’s disease in African Americans. Alzheimers Dement. 2017, 13, 119–129. [Google Scholar] [CrossRef] [Green Version]
  36. Kumar, A.; Chauhan, G.; Sharma, S.; Dabla, S.; Sylaja, P.N.; Chaudhary, N.; Gupta, S.; Agrawal, C.S.; Anand, K.S.; Srivastava, A.K.; et al. Association of SUMOylation Pathway Genes With Stroke in a Genome-Wide Association Study in India. Neurology 2021, 97, e345–e356. [Google Scholar] [CrossRef]
  37. Nazarian, A.; Yashin, A.I.; Kulminski, A.M. Genome-wide analysis of genetic predisposition to Alzheimer’s disease and related sex disparities. Alzheimers Res. Ther. 2019, 11, 5. [Google Scholar] [CrossRef] [Green Version]
  38. Chung, J.; Das, A.; Sun, X.; Sobreira, D.R.; Leung, Y.Y.; Igartua, C.; Mozaffari, S.; Chou, Y.F.; Thiagalingam, S.; Mez, J.; et al. Genome-wide association and multi-omics studies identify MGMT as a novel risk gene for Alzheimer’s disease among women. Alzheimers Dement. 2022. [Google Scholar] [CrossRef]
  39. Lee, T.H.; Ko, T.M.; Chen, C.H.; Chang, Y.J.; Lu, L.S.; Chang, C.H.; Huang, K.L.; Chang, T.Y.; Lee, J.D.; Chang, K.C.; et al. A genome-wide association study links small-vessel ischemic stroke to autophagy. Sci. Rep. 2017, 7, 15229. [Google Scholar] [CrossRef] [Green Version]
  40. Network, N.S.G.; International Stroke Genetics, C. Loci associated with ischaemic stroke and its subtypes (SiGN): A genome-wide association study. Lancet Neurol. 2016, 15, 174–184. [Google Scholar] [CrossRef]
  41. Malik, R.; Dau, T.; Gonik, M.; Sivakumar, A.; Deredge, D.J.; Edeleva, E.V.; Gotzfried, J.; van der Laan, S.W.; Pasterkamp, G.; Beaufort, N.; et al. Common coding variant in SERPINA1 increases the risk for large artery stroke. Proc. Natl. Acad. Sci. USA 2017, 114, 3613–3618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Traylor, M.; Malik, R.; Nalls, M.A.; Cotlarciuc, I.; Radmanesh, F.; Thorleifsson, G.; Hanscombe, K.B.; Langefeld, C.; Saleheen, D.; Rost, N.S.; et al. Genetic variation at 16q24.2 is associated with small vessel stroke. Ann. Neurol. 2017, 81, 383–394. [Google Scholar] [CrossRef] [Green Version]
  43. Traylor, M.; Persyn, E.; Tomppo, L.; Klasson, S.; Abedi, V.; Bakker, M.K.; Torres, N.; Li, L.; Bell, S.; Rutten-Jacobs, L.; et al. Genetic basis of lacunar stroke: A pooled analysis of individual patient data and genome-wide association studies. Lancet Neurol. 2021, 20, 351–361. [Google Scholar] [CrossRef]
  44. Jun, G.; Ibrahim-Verbaas, C.A.; Vronskaya, M.; Lambert, J.C.; Chung, J.; Naj, A.C.; Kunkle, B.W.; Wang, L.S.; Bis, J.C.; Bellenguez, C.; et al. A novel Alzheimer disease locus located near the gene encoding tau protein. Mol. Psychiatry 2016, 21, 108–117. [Google Scholar] [CrossRef] [Green Version]
  45. Kamboh, M.I.; Demirci, F.Y.; Wang, X.; Minster, R.L.; Carrasquillo, M.M.; Pankratz, V.S.; Younkin, S.G.; Saykin, A.J.; Alzheimer’s Disease Neuroimaging, I.; Jun, G.; et al. Genome-wide association study of Alzheimer’s disease. Transl. Psychiatry 2012, 2, e117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Chung, J.; Zhang, X.; Allen, M.; Wang, X.; Ma, Y.; Beecham, G.; Montine, T.J.; Younkin, S.G.; Dickson, D.W.; Golde, T.E.; et al. Genome-wide pleiotropy analysis of neuropathological traits related to Alzheimer’s disease. Alzheimers Res. Ther. 2018, 10, 22. [Google Scholar] [CrossRef] [PubMed]
  47. D’Addario, C.; Candia, S.B.; Arosio, B.; Di Bartolomeo, M.; Abbate, C.; Case, A.; Candeletti, S.; Romualdi, P.; Damanti, S.; Maccarrone, M.; et al. Transcriptional and epigenetic phenomena in peripheral blood cells of monozygotic twins discordant for alzheimer’s disease, a case report. J. Neurol. Sci. 2017, 372, 211–216. [Google Scholar] [CrossRef]
  48. Lu, Y.; Tan, L.; Wang, X. Circular HDAC9/microRNA-138/Sirtuin-1 pathway mediates synaptic and amyloid precursor protein processing deficits in Alzheimer’s disease. Neurosci. Bull. 2019, 35, 877–888. [Google Scholar] [CrossRef]
  49. Lu, S.; Li, H.; Li, K.; Fan, X.D. HDAC9 promotes brain ischemic injury by provoking IkappaBalpha/NF-kappaB and MAPKs signaling pathways. Biochem. Biophys. Res. Commun. 2018, 503, 1322–1329. [Google Scholar] [CrossRef]
  50. Shi, J.; Liu, H.; Wang, H.; Kong, X. MicroRNA expression signature in degenerative aortic stenosis. Biomed. Res. Int. 2016, 2016, 4682172. [Google Scholar] [CrossRef]
  51. Zhang, H.; Mo, X.; Wang, A.; Peng, H.; Guo, D.; Zhong, C.; Zhu, Z.; Xu, T.; Zhang, Y. Association of DNA methylation in blood pressure-related genes with ischemic stroke risk and prognosis. Front. Cardiovasc. Med. 2022, 9, 796245. [Google Scholar] [CrossRef] [PubMed]
  52. Prestel, M.; Prell-Schicker, C.; Webb, T.; Malik, R.; Lindner, B.; Ziesch, N.; Rex-Haffner, M.; Roh, S.; Viturawong, T.; Lehm, M.; et al. The atherosclerosis risk variant rs2107595 mediates allele-specific transcriptional regulation of HDAC9 via E2F3 and Rb1. Stroke 2019, 50, 2651–2660. [Google Scholar] [CrossRef] [PubMed]
  53. Shroff, N.; Ander, B.P.; Zhan, X.; Stamova, B.; Liu, D.; Hull, H.; Hamade, F.R.; Dykstra-Aiello, C.; Ng, K.; Sharp, F.R.; et al. HDAC9 Polymorphism Alters Blood Gene Expression in Patients with Large Vessel Atherosclerotic Stroke. Transl. Stroke Res. 2019, 10, 19–25. [Google Scholar] [CrossRef] [PubMed]
  54. Ito, S.; Kimura, K.; Haneda, M.; Ishida, Y.; Sawada, M.; Isobe, K. Induction of matrix metalloproteinases (MMP3, MMP12 and MMP13) expression in the microglia by amyloid-β stimulation via the PI3K/Akt pathway. Exp. Gerontol. 2007, 42, 532–537. [Google Scholar] [CrossRef]
  55. Asciutto, G.; Dias, N.V.; Edsfeldt, A.; Nitulescu, M.; Persson, A.; Nilsson, M.; Duner, P.; Nilsson, J.; Goncalves, I. Low elastin content of carotid plaques is associated with increased risk of ipsilateral stroke. PLoS ONE 2015, 10, e0121086. [Google Scholar] [CrossRef] [Green Version]
  56. Carcel-Marquez, J.; Cullell, N.; Muino, E.; Gallego-Fabrega, C.; Lledos, M.; Ibanez, L.; Krupinski, J.; Montaner, J.; Cruchaga, C.; Lee, J.M.; et al. Causal effect of MMP-1 (matrix metalloproteinase-1), MMP-8, and MMP-12 levels on ischemic stroke: A mendelian randomization study. Stroke 2021, 52, e316–e320. [Google Scholar] [CrossRef]
  57. Kuo, P.C.; Weng, W.T.; Scofield, B.A.; Furnas, D.; Paraiso, H.C.; Intriago, A.J.; Bosi, K.D.; Yu, I.C.; Yen, J.H. Interferon-β alleviates delayed tPA-induced adverse effects via modulation of MMP3/9 production in ischemic stroke. Blood Adv. 2020, 4, 4366–4381. [Google Scholar] [CrossRef]
  58. San Segundo-Acosta, P.; Montero-Calle, A.; Fuentes, M.; Rabano, A.; Villalba, M.; Barderas, R. Identification of Alzheimer’s Disease Autoantibodies and Their Target Biomarkers by Phage Microarrays. J. Proteome Res. 2019, 18, 2940–2953. [Google Scholar] [CrossRef]
  59. Han, J.; Feng, G.H.; Liu, H.W.; Yi, J.P.; Wu, J.B.; Yao, X.X. Classifying mild cognitive impairment and Alzheimer’s disease by constructing a 14-gene diagnostic model. Am. J. Transl. Res. 2022, 14, 4477–4492. [Google Scholar]
  60. Perisic, L.; Hedin, E.; Razuvaev, A.; Lengquist, M.; Osterholm, C.; Folkersen, L.; Gillgren, P.; Paulsson-Berne, G.; Ponten, F.; Odeberg, J.; et al. Profiling of atherosclerotic lesions by gene and tissue microarrays reveals PCSK6 as a novel protease in unstable carotid atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 2432–2443. [Google Scholar] [CrossRef]
  61. Alderton, G.; Scanlon, S.T. Inflammation. Science 2021, 374, 1068–1069. [Google Scholar] [CrossRef] [PubMed]
  62. Zhang, B.; Gaiteri, C.; Bodea, L.G.; Wang, Z.; McElwee, J.; Podtelezhnikov, A.A.; Zhang, C.; Xie, T.; Tran, L.; Dobrin, R.; et al. Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell 2013, 153, 707–720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Holmes, C.; Cunningham, C.; Zotova, E.; Woolford, J.; Dean, C.; Kerr, S.; Culliford, D.; Perry, V.H. Systemic inflammation and disease progression in Alzheimer disease. Neurology 2009, 73, 768–774. [Google Scholar] [CrossRef] [Green Version]
  64. Kinney, J.W.; Bemiller, S.M.; Murtishaw, A.S.; Leisgang, A.M.; Salazar, A.M.; Lamb, B.T. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement. 2018, 4, 575–590. [Google Scholar] [CrossRef] [PubMed]
  65. Baik, S.H.; Kang, S.; Son, S.M.; Mook-Jung, I. Microglia contributes to plaque growth by cell death due to uptake of amyloid β in the brain of Alzheimer’s disease mouse model. Glia 2016, 64, 2274–2290. [Google Scholar] [CrossRef] [PubMed]
  66. Shippy, D.C.; Watters, J.J.; Ulland, T.K. Transcriptional response of murine microglia in Alzheimer’s disease and inflammation. BMC Genom. 2022, 23, 183. [Google Scholar] [CrossRef]
  67. Bostanciklioglu, M. An update on the interactions between Alzheimer’s disease, autophagy and inflammation. Gene 2019, 705, 157–166. [Google Scholar] [CrossRef]
  68. Yao Mattisson, I.; Rattik, S.; Bjorkbacka, H.; Ljungcrantz, I.; Terrinoni, M.; Lebens, M.; Holmgren, J.; Fredrikson, G.N.; Gullstrand, B.; Bengtsson, A.A.; et al. Immune responses against oxidized LDL as possible targets for prevention of atherosclerosis in systemic lupus erythematosus. Vascul. Pharmacol. 2021, 140, 106863. [Google Scholar] [CrossRef]
  69. Xu, K.; Saaoud, F.; Yu, S.; Drummer, C.t.; Shao, Y.; Sun, Y.; Lu, Y.; Sun, J.; Yu, J.; Jiang, X.; et al. Monocyte adhesion assays for detecting endothelial cell activation in vascular inflammation and atherosclerosis. Methods Mol. Biol. 2022, 2419, 169–182. [Google Scholar] [CrossRef]
  70. Esenwa, C.C.; Elkind, M.S. Inflammatory risk factors, biomarkers and associated therapy in ischaemic stroke. Nat. Rev. Neurol. 2016, 12, 594–604. [Google Scholar] [CrossRef]
  71. Calandra, T.; Bucala, R. Macrophage Migration Inhibitory Factor (MIF): A Glucocorticoid Counter-Regulator within the Immune System. Crit. Rev. Immunol. 2017, 37, 359–370. [Google Scholar] [CrossRef] [PubMed]
  72. Wei, N.; Xu, Y.; Li, Y.; Shi, J.; Zhang, X.; You, Y.; Sun, Q.; Zhai, H.; Hu, Y. A bibliometric analysis of T cell and atherosclerosis. Front. Immunol. 2022, 13, 948314. [Google Scholar] [CrossRef] [PubMed]
  73. Tian, Y.; Lu, Y.; Cao, Y.; Dang, C.; Wang, N.; Tian, K.; Luo, Q.; Guo, E.; Luo, S.; Wang, L.; et al. Identification of diagnostic signatures associated with immune infiltration in Alzheimer’s disease by integrating bioinformatic analysis and machine-learning strategies. Front. Aging Neurosci. 2022, 14, 919614. [Google Scholar] [CrossRef]
  74. Matoba, K. Monitoring Leukocyte Migration During Atherosclerosis In Vivo. Methods Mol. Biol. 2022, 2419, 475–479. [Google Scholar] [CrossRef] [PubMed]
  75. Jiang, W.; Li, J.; Cai, Y.; Liu, W.; Chen, M.; Xu, X.; Deng, M.; Sun, J.; Zhou, L.; Huang, Y.; et al. The Novel lncRNA ENST00000530525 Affects ANO1, Contributing to Blood-Brain Barrier Injury in Cultured hCMEC/D3 Cells Under OGD/R Conditions. Front. Genet. 2022, 13, 873230. [Google Scholar] [CrossRef]
  76. Lefkowitz, R.J. A brief history of G-protein coupled receptors (Nobel Lecture). Angew. Chem. Int. Ed. Engl. 2013, 52, 6366–6378. [Google Scholar] [CrossRef]
  77. Zhao, J.; Deng, Y.; Jiang, Z.; Qing, H. G protein-coupled receptors (GPCRs) in Alzheimer’s disease: A focus on BACE1 related GPCRs. Front. Aging Neurosci. 2016, 8, 58. [Google Scholar] [CrossRef] [Green Version]
  78. Haque, M.E.; Kim, I.S.; Jakaria, M.; Akther, M.; Choi, D.K. Importance of GPCR-Mediated Microglial Activation in Alzheimer’s Disease. Front. Cell Neurosci. 2018, 12, 258. [Google Scholar] [CrossRef] [Green Version]
  79. Dal Prà, I.; Armato, U.; Chiarini, A. Family C G-protein-coupled receptors in Alzheimer’s disease and therapeutic implications. Front. Pharmacol. 2019, 10, 1282. [Google Scholar] [CrossRef]
  80. Liao, X.; Zhang, Y.; Wang, Y.; Wang, J. The effect of cdk-5 overexpression on tau phosphorylation and spatial memory of rat. Sci. China C Life Sci. 2004, 47, 251–257. [Google Scholar] [CrossRef]
  81. Siano, G.; Caiazza, M.C.; Olla, I.; Varisco, M.; Madaro, G.; Quercioli, V.; Calvello, M.; Cattaneo, A.; Di Primio, C. Identification of an ERK inhibitor as a therapeutic drug against tau aggregation in a new cell-based assay. Front. Cell Neurosci. 2019, 13, 386. [Google Scholar] [CrossRef] [PubMed]
  82. Reynolds, C.H.; Betts, J.C.; Blackstock, W.P.; Nebreda, A.R.; Anderton, B.H. Phosphorylation sites on tau identified by nanoelectrospray mass spectrometry: Differences in vitro between the mitogen-activated protein kinases ERK2, c-Jun N-terminal kinase and P38, and glycogen synthase kinase-3beta. J. Neurochem. 2000, 74, 1587–1595. [Google Scholar] [CrossRef] [PubMed]
  83. Bolos, M.; Llorens-Martin, M.; Perea, J.R.; Jurado-Arjona, J.; Rabano, A.; Hernandez, F.; Avila, J. Absence of CX3CR1 impairs the internalization of Tau by microglia. Mol. Neurodegener. 2017, 12, 59. [Google Scholar] [CrossRef] [Green Version]
  84. Thathiah, A.; De Strooper, B. G protein-coupled receptors, cholinergic dysfunction, and Abeta toxicity in Alzheimer’s disease. Sci. Signal. 2009, 2, re8. [Google Scholar] [CrossRef]
  85. Li, Y.F.; Li, R.S.; Samuel, S.B.; Cueto, R.; Li, X.Y.; Wang, H.; Yang, X.F. Lysophospholipids and their G protein-coupled receptors in atherosclerosis. Front. Biosci. 2016, 21, 70–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Ghislain, J.; Poitout, V. Targeting lipid GPCRs to treat type 2 diabetes mellitus—Progress and challenges. Nat. Rev. Endocrinol. 2021, 17, 162–175. [Google Scholar] [CrossRef] [PubMed]
  87. Jia, W.; Wang, C.; Zheng, J.; Li, Y.; Yang, C.; Wan, Q.L.; Shen, J. Pioglitazone Hydrochloride Extends the Lifespan of Caenorhabditis elegans by Activating DAF-16/FOXO- and SKN-1/NRF2-Related Signaling Pathways. Oxid. Med. Cell Longev. 2022, 2022, 8496063. [Google Scholar] [CrossRef] [PubMed]
  88. Blondrath, K.; Steel, J.H.; Katsouri, L.; Ries, M.; Parker, M.G.; Christian, M.; Sastre, M. The nuclear cofactor receptor interacting protein-140 (RIP140) regulates the expression of genes involved in Abeta generation. Neurobiol. Aging 2016, 47, 180–191. [Google Scholar] [CrossRef] [Green Version]
  89. Yi, Z.J.; Gong, J.P.; Zhang, W. Transcriptional co-regulator RIP140: An important mediator of the inflammatory response and its associated diseases (Review). Mol. Med. Rep. 2017, 16, 994–1000. [Google Scholar] [CrossRef] [Green Version]
  90. Das, B.C.; Dasgupta, S.; Ray, S.K. Potential therapeutic roles of retinoids for prevention of neuroinflammation and neurodegeneration in Alzheimer’s disease. Neural. Regen. Res. 2019, 14, 1880–1892. [Google Scholar] [CrossRef]
  91. Unsworth, A.J.; Flora, G.D.; Sasikumar, P.; Bye, A.P.; Sage, T.; Kriek, N.; Crescente, M.; Gibbins, J.M. RXR Ligands Negatively Regulate Thrombosis and Hemostasis. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 812–822. [Google Scholar] [CrossRef]
  92. Natunen, T.; Martiskainen, H.; Sarajarvi, T.; Helisalmi, S.; Pursiheimo, J.P.; Viswanathan, J.; Laitinen, M.; Makinen, P.; Kauppinen, T.; Rauramaa, T.; et al. Effects of NR1H3 genetic variation on the expression of liver X receptor α and the progression of Alzheimer’s disease. PLoS ONE 2013, 8, e80700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Sandoval-Hernandez, A.G.; Restrepo, A.; Cardona-Gomez, G.P.; Arboleda, G. LXR activation protects hippocampal microvasculature in very old triple transgenic mouse model of Alzheimer’s disease. Neurosci. Lett. 2016, 621, 15–21. [Google Scholar] [CrossRef]
  94. Grbic, E.; Gorkic, N.; Pleskovic, A.; Ljuca, F.; Gasparini, M.; Mrda, B.; Cilensek, I.; Fras, Z.; Petrovic, D. Association between the rs2279238 of the Liver X receptor α gene polymorphism and advanced carotid atherosclerosis in the Slovenian cohort. Gene 2022, 840, 146764. [Google Scholar] [CrossRef] [PubMed]
  95. Rasheed, A.; Shawky, S.A.; Tsai, R.; Jung, R.G.; Simard, T.; Saikali, M.F.; Hibbert, B.; Rayner, K.J.; Cummins, C.L. The secretome of liver X receptor agonist-treated early outgrowth cells decreases atherosclerosis in Ldlr-/- mice. Stem Cells Transl. Med. 2021, 10, 479–491. [Google Scholar] [CrossRef] [PubMed]
  96. Endo-Umeda, K.; Kim, E.; Thomas, D.G.; Liu, W.; Dou, H.; Yalcinkaya, M.; Abramowicz, S.; Xiao, T.; Antonson, P.; Gustafsson, J.A.; et al. Myeloid LXR (Liver X Receptor) Deficiency Induces Inflammatory Gene Expression in Foamy Macrophages and Accelerates Atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2022, 42, 719–731. [Google Scholar] [CrossRef]
  97. Chang, K.L.; Wong, L.R.; Pee, H.N.; Yang, S.; Ho, P.C. Reverting Metabolic Dysfunction in Cortex and Cerebellum of APP/PS1 Mice, a Model for Alzheimer’s Disease by Pioglitazone, a Peroxisome Proliferator-Activated Receptor γ (PPARgamma) Agonist. Mol. Neurobiol. 2019, 56, 7267–7283. [Google Scholar] [CrossRef]
  98. Cheng, F.; Si, X.M.; Yang, G.L.; Zhou, L. Relationship between PPAR-γ gene polymorphisms and ischemic stroke risk: A meta-analysis. Brain Behav. 2021, 11, e2434. [Google Scholar] [CrossRef]
  99. Sabry, M.M.; Dawood, A.F.; Rashed, L.A.; Sayed, S.M.; Hassan, S.; Younes, S.F. Relation between resistin, PPAR-γ, obesity and atherosclerosis in male albino rats. Arch. Physiol. Biochem. 2020, 126, 389–398. [Google Scholar] [CrossRef]
  100. Moon, M.; Jung, E.S.; Jeon, S.G.; Cha, M.Y.; Jang, Y.; Kim, W.; Lopes, C.; Mook-Jung, I.; Kim, K.S. Nurr1 (NR4A2) regulates Alzheimer’s disease-related pathogenesis and cognitive function in the 5XFAD mouse model. Aging Cell 2019, 18, e12866. [Google Scholar] [CrossRef] [Green Version]
  101. Zhao, L.G.; Tang, Y.; Tan, J.Z.; Wang, J.W.; Chen, G.J.; Zhu, B.L. The effect of NR4A1 on APP metabolism and tau phosphorylation. Genes Dis. 2018, 5, 342–348. [Google Scholar] [CrossRef] [PubMed]
  102. Hanna, R.N.; Shaked, I.; Hubbeling, H.G.; Punt, J.A.; Wu, R.; Herrley, E.; Zaugg, C.; Pei, H.; Geissmann, F.; Ley, K.; et al. NR4A1 (Nur77) deletion polarizes macrophages toward an inflammatory phenotype and increases atherosclerosis. Circ. Res. 2012, 110, 416–427. [Google Scholar] [CrossRef] [Green Version]
  103. Ishunina, T.A.; Bogolepova, I.N.; Swaab, D.F. Increased Neuronal Nuclear and Perikaryal Size in the Medial Mamillary Nucleus of Vascular Dementia and Alzheimer’s Disease Patients: Relation to Nuclear Estrogen Receptor α. Dement. Geriatr. Cogn. Disord. 2019, 47, 274–280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Wang, C.; Zhang, F.; Jiang, S.; Siedlak, S.L.; Shen, L.; Perry, G.; Wang, X.; Tang, B.; Zhu, X. Estrogen receptor-α is localized to neurofibrillary tangles in Alzheimer’s disease. Sci. Rep. 2016, 6, 20352. [Google Scholar] [CrossRef] [Green Version]
  105. Meng, Q.; Li, Y.; Ji, T.; Chao, Y.; Li, J.; Fu, Y.; Wang, S.; Chen, Q.; Chen, W.; Huang, F.; et al. Estrogen prevent atherosclerosis by attenuating endothelial cell pyroptosis via activation of estrogen receptor α-mediated autophagy. J. Adv. Res. 2021, 28, 149–164. [Google Scholar] [CrossRef] [PubMed]
  106. Chen, Q.; Chao, Y.; Zhang, W.; Zhang, Y.; Bi, Y.; Fu, Y.; Cai, D.; Meng, Q.; Li, Y.; Bian, H. Activation of estrogen receptor α (ERalpha) is required for Alisol B23-acetate to prevent post-menopausal atherosclerosis and reduced lipid accumulation. Life Sci. 2020, 258, 118030. [Google Scholar] [CrossRef]
  107. Min, J.; Weitian, Z.; Peng, C.; Yan, P.; Bo, Z.; Yan, W.; Yun, B.; Xukai, W. Correlation between insulin-induced estrogen receptor methylation and atherosclerosis. Cardiovasc. Diabetol. 2016, 15, 156. [Google Scholar] [CrossRef] [Green Version]
  108. Ji, X.; Jia, L.; Jia, J.; Qi, L. Genetic association of urokinase-type plasminogen activator gene rs2227564 site polymorphism with sporadic Alzheimer’s disease in the Han Chinese population. Neural. Regen. Res. 2012, 7, 2377–2383. [Google Scholar] [CrossRef]
  109. Diaz, A.; Merino, P.; Guo, J.D.; Yepes, M.A.; McCann, P.; Katta, T.; Tong, E.M.; Torre, E.; Rangaraju, S.; Yepes, M. Urokinase-Type Plasminogen Activator Protects Cerebral Cortical Neurons from Soluble Abeta-Induced Synaptic Damage. J. Neurosci. 2020, 40, 4251–4263. [Google Scholar] [CrossRef]
  110. Svensson, P.A.; Olson, F.J.; Hagg, D.A.; Ryndel, M.; Wiklund, O.; Karlstrom, L.; Hulthe, J.; Carlsson, L.M.; Fagerberg, B. Urokinase-type plasminogen activator receptor is associated with macrophages and plaque rupture in symptomatic carotid atherosclerosis. Int. J. Mol. Med. 2008, 22, 459–464. [Google Scholar]
  111. Paland, N.; Aharoni, S.; Fuhrman, B. Urokinase-type plasminogen activator (uPA) modulates monocyte-to-macrophage differentiation and prevents Ox-LDL-induced macrophage apoptosis. Atherosclerosis 2013, 231, 29–38. [Google Scholar] [CrossRef] [PubMed]
  112. Nguyen, H.D.; Kim, M.S. Exposure to a mixture of heavy metals induces cognitive impairment: Genes and microRNAs involved. Toxicology 2022, 471, 153164. [Google Scholar] [CrossRef] [PubMed]
  113. Nguyen, H.D.; Kim, M.S. The role of mixed B vitamin intakes on cognitive performance: Modeling, genes and miRNAs involved. J. Psychiatr. Res. 2022, 152, 38–56. [Google Scholar] [CrossRef]
  114. Wen, Q.; Verheijen, M.; Wittens, M.M.J.; Czurylo, J.; Engelborghs, S.; Hauser, D.; van Herwijnen, M.H.M.; Lundh, T.; Bergdahl, I.A.; Kyrtopoulos, S.A.; et al. Lead-exposure associated miRNAs in humans and Alzheimer’s disease as potential biomarkers of the disease and disease processes. Sci. Rep. 2022, 12, 15966. [Google Scholar] [CrossRef]
  115. Nguyen, D.D.N.; Zain, S.M.; Kamarulzaman, M.H.; Low, T.Y.; Chilian, W.M.; Pan, Y.; Ting, K.N.; Hamid, A.; Abdul Kadir, A.; Pung, Y.F. Intracellular and exosomal microRNAome profiling of human vascular smooth muscle cells during replicative senescence. Am. J. Physiol. Heart Circ. Physiol. 2021, 321, H770–H783. [Google Scholar] [CrossRef] [PubMed]
  116. Nguyen, H.D. Resveratrol, Endocrine Disrupting Chemicals, Neurodegenerative Diseases and Depression: Genes, Transcription Factors, microRNAs, and Sponges Involved. Neurochem. Res. 2022. [Google Scholar] [CrossRef] [PubMed]
  117. Shi, J.; Li, L. circKMT2E Protect Retina from Early Diabetic Retinopathy through SIRT1 Signaling Pathway via Sponging miR-204-5p. Comput. Math Methods Med. 2022, 2022, 7188193. [Google Scholar] [CrossRef]
  118. Liu, X.; Wu, J.; Nie, H.; Zhu, X.; Song, G.; Han, L.; Qin, W. Comprehensive Analysis of circRNAs, miRNAs, and mRNAs Expression Profiles and ceRNA Networks in Decidua of Unexplained Recurrent Spontaneous Abortion. Front. Genet. 2022, 13, 858641. [Google Scholar] [CrossRef]
  119. Tang, Y.F.; Wu, W.J.; Zhang, J.Y.; Zhang, J. Reconstruction and analysis of the aberrant lncRNA-miRNA-mRNA network based on competitive endogenous RNA in adenoid cystic carcinoma of the salivary gland. Transl. Cancer Res. 2021, 10, 5133–5149. [Google Scholar] [CrossRef]
  120. Yang, S.; Zheng, W.; Yang, C.; Zu, R.; Ran, S.; Wu, H.; Mu, M.; Sun, S.; Zhang, N.; Thorne, R.F.; et al. Integrated Analysis of Hub Genes and MicroRNAs in Human Placental Tissues from In Vitro Fertilization-Embryo Transfer. Front. Endocrinol. (Lausanne) 2021, 12, 774997. [Google Scholar] [CrossRef]
  121. Zhang, F.; Yu, X.; Lin, Z.; Wang, X.; Gao, T.; Teng, D.; Teng, W. Using Tumor-Infiltrating Immune Cells and a ceRNA Network Model to Construct a Prognostic Analysis Model of Thyroid Carcinoma. Front. Oncol. 2021, 11, 658165. [Google Scholar] [CrossRef] [PubMed]
  122. Liu, X.; Wang, P.; Teng, X.; Zhang, Z.; Song, S. Comprehensive Analysis of Expression Regulation for RNA m6A Regulators With Clinical Significance in Human Cancers. Front. Oncol. 2021, 11, 624395. [Google Scholar] [CrossRef] [PubMed]
  123. Hao, M.; Liu, W.; Ding, C.; Peng, X.; Zhang, Y.; Chen, H.; Dong, L.; Liu, X.; Zhao, Y.; Chen, X.; et al. Identification of hub genes and small molecule therapeutic drugs related to breast cancer with comprehensive bioinformatics analysis. PeerJ 2020, 8, e9946. [Google Scholar] [CrossRef] [PubMed]
  124. Zhai, M.; Zhu, Y.; Yang, M.; Mao, C. Human Mesenchymal Stem Cell Derived Exosomes Enhance Cell-Free Bone Regeneration by Altering Their miRNAs Profiles. Adv. Sci. (Weinh) 2020, 7, 2001334. [Google Scholar] [CrossRef] [PubMed]
  125. Zhou, H.; Chen, D.; Xie, G.; Li, J.; Tang, J.; Tang, L. LncRNA-mediated ceRNA network was identified as a crucial determinant of differential effects in periodontitis and periimplantitis by high-throughput sequencing. Clin. Implant Dent. Relat. Res. 2020, 22, 424–450. [Google Scholar] [CrossRef]
  126. Ma, T.; Liu, A.; Xu, D.; Zhang, T. Mechanisms underlying the promotion of osteosarcoma cell proliferation and invasion by lncRNA PBB12. Oncol. Rep. 2020, 43, 736–746. [Google Scholar] [CrossRef]
  127. Kondo, Y.; Kogure, T.; Ninomiya, M.; Fukuda, R.; Monma, N.; Ikeo, K.; Tanaka, Y. The reduction of miR146b-5p in monocytes and T cells could contribute to the immunopathogenesis of hepatitis C virus infection. Sci. Rep. 2019, 9, 13393. [Google Scholar] [CrossRef] [Green Version]
  128. Fan, C.; Liu, X.; Li, W.; Wang, H.; Teng, Y.; Ren, J.; Huang, Y. Circular RNA circ KMT2E is up-regulated in diabetic cataract lenses and is associated with miR-204-5p sponge function. Gene 2019, 710, 170–177. [Google Scholar] [CrossRef]
  129. Gumus, G.; Giray, D.; Bobusoglu, O.; Tamer, L.; Karpuz, D.; Hallioglu, O. MicroRNA values in children with rheumatic carditis: A preliminary study. Rheumatol. Int. 2018, 38, 1199–1205. [Google Scholar] [CrossRef]
  130. Su, L.N.; Song, X.Q.; Xue, Z.X.; Zheng, C.Q.; Yin, H.F.; Wei, H.P. Network analysis of microRNAs, transcription factors, and target genes involved in axon regeneration. J. Zhejiang Univ. Sci. B 2018, 19, 293–304. [Google Scholar] [CrossRef]
  131. Gao, W.; Wu, Y.; He, X.; Zhang, C.; Zhu, M.; Chen, B.; Liu, Q.; Qu, X.; Li, W.; Wen, S.; et al. MicroRNA-204-5p inhibits invasion and metastasis of laryngeal squamous cell carcinoma by suppressing forkhead box C1. J. Cancer 2017, 8, 2356–2368. [Google Scholar] [CrossRef] [PubMed]
  132. Zhang, T.; Liu, C.; Huang, S.; Ma, Y.; Fang, J.; Chen, Y. A Downmodulated MicroRNA Profiling in Patients with Gastric Cancer. Gastroenterol. Res. Pract. 2017, 2017, 1526981. [Google Scholar] [CrossRef] [PubMed]
  133. Zhu, J.; Ma, X.; Zhang, Y.; Ni, D.; Ai, Q.; Li, H.; Zhang, X. Establishment of a miRNA-mRNA regulatory network in metastatic renal cell carcinoma and screening of potential therapeutic targets. Tumour. Biol. 2016. [Google Scholar] [CrossRef] [PubMed]
  134. Hubens, W.H.G.; Krauskopf, J.; Beckers, H.J.M.; Kleinjans, J.C.S.; Webers, C.A.B.; Gorgels, T. Small RNA Sequencing of Aqueous Humor and Plasma in Patients With Primary Open-Angle Glaucoma. Investig. Ophthalmol. Vis. Sci. 2021, 62, 24. [Google Scholar] [CrossRef]
  135. Liu, S.; Wang, H.; Mu, J.; Wang, H.; Peng, Y.; Li, Q.; Mao, D.; Guo, L. MiRNA-211 triggers an autophagy-dependent apoptosis in cervical cancer cells: Regulation of Bcl-2. Naunyn. Schmiedebergs Arch. Pharmacol. 2020, 393, 359–370. [Google Scholar] [CrossRef]
  136. Zhong, Y.; Zhu, F.; Ding, Y. Differential microRNA expression profile in the plasma of preeclampsia and normal pregnancies. Exp. Ther. Med. 2019, 18, 826–832. [Google Scholar] [CrossRef]
  137. Shinawi, T.; Nasser, K.K.; Moradi, F.A.; Mujalli, A.; Albaqami, W.F.; Almukadi, H.S.; Elango, R.; Shaik, N.A.; Banaganapalli, B. A comparative mRNA- and miRNA transcriptomics reveals novel molecular signatures associated with metastatic prostate cancers. Front. Genet. 2022, 13, 1066118. [Google Scholar] [CrossRef]
  138. Dandare, A.; Rabia, G.; Khan, M.J. In silico analysis of non-coding RNAs and putative target genes implicated in metabolic syndrome. Comput. Biol. Med. 2021, 130, 104229. [Google Scholar] [CrossRef]
  139. Feng, C.; Li, Y.; Lin, Y.; Cao, X.; Li, D.; Zhang, H.; He, X. CircRNA-associated ceRNA network reveals ErbB and Hippo signaling pathways in hypopharyngeal cancer. Int. J. Mol. Med. 2019, 43, 127–142. [Google Scholar] [CrossRef] [Green Version]
  140. Jing, L.; Jin, C.; Lu, Y.; Huo, P.; Zhou, L.; Wang, Y.; Tian, Y. Investigation of microRNA expression profiles associated with human alcoholic cardiomyopathy. Cardiology 2015, 130, 223–233. [Google Scholar] [CrossRef]
  141. Li, S.; Song, F.; Lei, X.; Li, J.; Li, F.; Tan, H. hsa_circ_0004018 suppresses the progression of liver fibrosis through regulating the hsa-miR-660-3p/TEP1 axis. Aging (Albany N.Y.) 2020, 12, 11517–11529. [Google Scholar] [CrossRef] [PubMed]
  142. Liu, W.; Zhang, J.; Zou, C.; Xie, X.; Wang, Y.; Wang, B.; Zhao, Z.; Tu, J.; Wang, X.; Li, H.; et al. Microarray Expression Profile and Functional Analysis of Circular RNAs in Osteosarcoma. Cell Physiol. Biochem. 2017, 43, 969–985. [Google Scholar] [CrossRef] [PubMed]
  143. Zhao, X.; Dou, J.; Cao, J.; Wang, Y.; Gao, Q.; Zeng, Q.; Liu, W.; Liu, B.; Cui, Z.; Teng, L.; et al. Uncovering the potential differentially expressed miRNAs as diagnostic biomarkers for hepatocellular carcinoma based on machine learning in The Cancer Genome Atlas database. Oncol. Rep. 2020, 43, 1771–1784. [Google Scholar] [CrossRef] [Green Version]
  144. Yang, G.; Zhang, Y.; Yang, J. A Five-microRNA Signature as Prognostic Biomarker in Colorectal Cancer by Bioinformatics Analysis. Front. Oncol. 2019, 9, 1207. [Google Scholar] [CrossRef] [Green Version]
  145. Plata-Bello, J.; Farina-Jeronimo, H.; Betancor, I.; Salido, E. High Expression of FOXP2 Is Associated with Worse Prognosis in Glioblastoma. World Neurosurg. 2021, 150, e253–e278. [Google Scholar] [CrossRef]
  146. Wang, Y.; Li, Y.; Jia, D.; Zheng, J.; Wang, G. Correlation between single nucleotide polymorphisms in CXCR4 microRNA binding site and the susceptibility to knee osteoarthritis in Han Chinese population. J. Clin. Lab. Anal. 2021, 35, e23600. [Google Scholar] [CrossRef]
  147. Ekmekci, C.G.; Coskunpinar, E.; Avci, H.; Farooqi, A.A.; Orhan, K.S.; Akbas, F. Integrative analysis of mRNA and microRNA expression profiles in laryngeal squamous cell carcinoma. J. Cell Biochem. 2019, 120, 3415–3422. [Google Scholar] [CrossRef]
  148. Peng, Z.; Duan, Y.; Zhong, S.; Chen, J.; Li, J.; He, Z. RNA-seq analysis of extracellular vesicles from hyperphosphatemia-stimulated endothelial cells provides insight into the mechanism underlying vascular calcification. BMC Nephrol. 2022, 23, 192. [Google Scholar] [CrossRef]
  149. Chen, N.; Wan, G.; Zeng, X. Integrated Whole-Transcriptome Profiling and Bioinformatics Analysis of the Polypharmacological Effects of Ganoderic Acid Me in Colorectal Cancer Treatment. Front. Oncol. 2022, 12, 833375. [Google Scholar] [CrossRef] [PubMed]
  150. Baker, M.A.; Davis, S.J.; Liu, P.; Pan, X.; Williams, A.M.; Iczkowski, K.A.; Gallagher, S.T.; Bishop, K.; Regner, K.R.; Liu, Y.; et al. Tissue-Specific MicroRNA Expression Patterns in Four Types of Kidney Disease. J. Am. Soc. Nephrol. 2017, 28, 2985–2992. [Google Scholar] [CrossRef] [Green Version]
  151. Ju, L.; Han, M.; Li, X.; Zhao, C. MicroRNA Signature of Lung Adenocarcinoma with EGFR Exon 19 Deletion. J. Cancer 2017, 8, 1311–1318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Schoen, C.; Glennon, J.C.; Abghari, S.; Bloemen, M.; Aschrafi, A.; Carels, C.E.L.; Von den Hoff, J.W. Differential microRNA expression in cultured palatal fibroblasts from infants with cleft palate and controls. Eur. J. Orthod. 2018, 40, 90–96. [Google Scholar] [CrossRef] [PubMed]
  153. Pehserl, A.M.; Ress, A.L.; Stanzer, S.; Resel, M.; Karbiener, M.; Stadelmeyer, E.; Stiegelbauer, V.; Gerger, A.; Mayr, C.; Scheideler, M.; et al. Comprehensive Analysis of miRNome Alterations in Response to Sorafenib Treatment in Colorectal Cancer Cells. Int. J. Mol. Sci. 2016, 17, 2011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Wu, M.; Lu, L.; Dai, T.; Li, A.; Yu, Y.; Li, Y.; Xu, Z.; Chen, Y. Construction of a lncRNA-mediated ceRNA network and a genomic-clinicopathologic nomogram to predict survival for breast cancer patients. Cancer Biomark. 2022. [Google Scholar] [CrossRef] [PubMed]
  155. Hu, J.; Zhou, S.; Guo, W. Construction of the coexpression network involved in the pathogenesis of thyroid eye disease via bioinformatics analysis. Hum. Genom. 2022, 16, 38. [Google Scholar] [CrossRef]
  156. Li, S.; Zhang, M.; Xu, F.; Wang, Y.; Leng, D. Detection significance of miR-3662, miR-146a, and miR-1290 in serum exosomes of breast cancer patients. J. Cancer Res. Ther. 2021, 17, 749–755. [Google Scholar] [CrossRef]
  157. Liu, A.; Zhou, Y.; Zhao, T.; Tang, X.; Zhou, B.; Xu, J. MiRNA-3662 reverses the gemcitabine resistance in pancreatic cancer through regulating the tumor metabolism. Cancer Chemother. Pharmacol. 2021, 88, 343–357. [Google Scholar] [CrossRef]
  158. Liu, A.; Xu, J. Circ_03955 promotes pancreatic cancer tumorigenesis and Warburg effect by targeting the miR-3662/HIF-1alpha axis. Clin. Transl. Oncol. 2021, 23, 1905–1914. [Google Scholar] [CrossRef]
  159. Byun, Y.J.; Piao, X.M.; Jeong, P.; Kang, H.W.; Seo, S.P.; Moon, S.K.; Lee, J.Y.; Choi, Y.H.; Lee, H.Y.; Kim, W.T.; et al. Urinary microRNA-1913 to microRNA-3659 expression ratio as a non-invasive diagnostic biomarker for prostate cancer. Investig. Clin. Urol. 2021, 62, 340–348. [Google Scholar] [CrossRef]
  160. Ye, J.; Xiao, X.; Han, Y.; Fan, D.; Zhu, Y.; Yang, L. MiR-3662 suppresses cell growth, invasion and glucose metabolism by targeting HK2 in hepatocellular carcinoma cells. Neoplasma 2020, 67, 773–781. [Google Scholar] [CrossRef]
  161. Niveditha, D.; Jasoria, M.; Narayan, J.; Majumder, S.; Mukherjee, S.; Chowdhury, R.; Chowdhury, S. Common and Unique microRNAs in Multiple Carcinomas Regulate Similar Network of Pathways to Mediate Cancer Progression. Sci. Rep. 2020, 10, 2331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Chen, Z.; Zuo, X.; Zhang, Y.; Han, G.; Zhang, L.; Wu, J.; Wang, X. MiR-3662 suppresses hepatocellular carcinoma growth through inhibition of HIF-1alpha-mediated Warburg effect. Cell Death Dis. 2018, 9, 549. [Google Scholar] [CrossRef] [PubMed]
  163. Yasui, K.; Izumida, M.; Nakagawa, T.; Kubo, Y.; Hayashi, H.; Ito, T.; Ikeda, H.; Matsuyama, T. MicroRNA-3662 expression correlates with antiviral drug resistance in adult T-cell leukemia/lymphoma cells. Biochem. Biophys. Res. Commun. 2018, 501, 833–837. [Google Scholar] [CrossRef] [PubMed]
  164. Powrozek, T.; Kuznar-Kaminska, B.; Dziedzic, M.; Mlak, R.; Batura-Gabryel, H.; Sagan, D.; Krawczyk, P.; Milanowski, J.; Malecka-Massalska, T. The diagnostic role of plasma circulating precursors of miRNA-944 and miRNA-3662 for non-small cell lung cancer detection. Pathol. Res. Pract. 2017, 213, 1384–1387. [Google Scholar] [CrossRef]
  165. Powrozek, T.; Mlak, R.; Dziedzic, M.; Malecka-Massalska, T.; Sagan, D. Analysis of primary-miRNA-3662 and its mature form may improve detection of the lung adenocarcinoma. J. Cancer Res. Clin. Oncol. 2017, 143, 1941–1946. [Google Scholar] [CrossRef]
  166. Zhao, W.; Shen, W.W.; Cao, X.M.; Ding, W.Y.; Yan, L.P.; Gao, L.J.; Li, X.L.; Zhong, T.Y. Novel mechanism of miRNA-365-regulated trophoblast apoptosis in recurrent miscarriage. J. Cell Mol. Med. 2017, 21, 2412–2425. [Google Scholar] [CrossRef]
  167. Maharry, S.E.; Walker, C.J.; Liyanarachchi, S.; Mehta, S.; Patel, M.; Bainazar, M.A.; Huang, X.; Lankenau, M.A.; Hoag, K.W.; Ranganathan, P.; et al. Dissection of the Major Hematopoietic Quantitative Trait Locus in Chromosome 6q23.3 Identifies miR-3662 as a Player in Hematopoiesis and Acute Myeloid Leukemia. Cancer Discov. 2016, 6, 1036–1051. [Google Scholar] [CrossRef] [Green Version]
  168. Powrozek, T.; Krawczyk, P.; Kowalski, D.M.; Winiarczyk, K.; Olszyna-Serementa, M.; Milanowski, J. Plasma circulating microRNA-944 and microRNA-3662 as potential histologic type-specific early lung cancer biomarkers. Transl. Res. 2015, 166, 315–323. [Google Scholar] [CrossRef]
  169. Li, Z.; Sun, Y.; He, M.; Liu, J. Differentially-expressed mRNAs, microRNAs and long noncoding RNAs in intervertebral disc degeneration identified by RNA-sequencing. Bioengineered 2021, 12, 1026–1039. [Google Scholar] [CrossRef]
  170. Chen, W.; Tang, D.; Dai, Y.; Diao, H. Establishment of microRNA, transcript and protein regulatory networks in Alport syndrome induced pluripotent stem cells. Mol. Med. Rep. 2019, 19, 238–250. [Google Scholar] [CrossRef] [Green Version]
  171. Lan, X.; Han, J.; Wang, B.; Sun, M. Integrated analysis of transcriptome profiling of lncRNAs and mRNAs in livers of type 2 diabetes mellitus. Physiol. Genom. 2022, 54, 86–97. [Google Scholar] [CrossRef] [PubMed]
  172. Tarek, M.M.; Yahia, A.; El-Nakib, M.M.; Elhefnawi, M. Integrative assessment of CIP2A overexpression and mutational effects in human malignancies identifies possible deleterious variants. Comput. Biol. Med. 2021, 139, 104986. [Google Scholar] [CrossRef] [PubMed]
  173. Morenikeji, O.B.; Adegbaju, M.S.; Okoh, O.S.; Babalola, A.E.; Grytsay, A.; Braimah, O.A.; Akinyemi, M.O.; Thomas, B.N. Deciphering inhibitory mechanism of coronavirus replication through host miRNAs-RNA-dependent RNA polymerase interactome. Front. Genet. 2022, 13, 973252. [Google Scholar] [CrossRef] [PubMed]
  174. Barbier, R.H.; McCrea, E.M.; Lee, K.Y.; Strope, J.D.; Risdon, E.N.; Price, D.K.; Chau, C.H.; Figg, W.D. Abiraterone induces SLCO1B3 expression in prostate cancer via microRNA-579-3p. Sci. Rep. 2021, 11, 10765. [Google Scholar] [CrossRef]
  175. Hommers, L.G.; Richter, J.; Yang, Y.; Raab, A.; Baumann, C.; Lang, K.; Schiele, M.A.; Weber, H.; Wittmann, A.; Wolf, C.; et al. A functional genetic variation of SLC6A2 repressor hsa-miR-579-3p upregulates sympathetic noradrenergic processes of fear and anxiety. Transl. Psychiatry 2018, 8, 226. [Google Scholar] [CrossRef] [Green Version]
  176. Kasimanickam, V.; Kastelic, J. MicroRNA in sperm from Duroc, Landrace and Yorkshire boars. Sci. Rep. 2016, 6, 32954. [Google Scholar] [CrossRef] [Green Version]
  177. Yilmaz, S.G.; Geyik, S.; Neyal, A.M.; Soko, N.D.; Bozkurt, H.; Dandara, C. Hypothesis: Do miRNAs Targeting the Leucine-Rich Repeat Kinase 2 Gene (LRRK2) Influence Parkinson’s Disease Susceptibility? OMICS 2016, 20, 224–228. [Google Scholar] [CrossRef]
  178. Ulusan Bagci, O.; Caner, A. [miRNA Expression Profile in Ileocecal Adenocarcinoma Cells Infected with Cryptosporidium]. Mikrobiyol. Bul. 2022, 56, 449–465. [Google Scholar] [CrossRef]
  179. Tripathi, S.K.; Mathaiyan, J.; Kayal, S.; Nachiappa Ganesh, R. Identification of Differentially Expressed Mirna by Next Generation Sequencing in Locally Advanced Breast Cancer Patients of South Indian Origin. Asian Pac. J. Cancer Prev. 2022, 23, 2255–2261. [Google Scholar] [CrossRef]
  180. Jeong, S.; Kim, S.A.; Ahn, S.G. HOXC6-Mediated miR-188-5p Expression Induces Cell Migration through the Inhibition of the Tumor Suppressor FOXN2. Int. J. Mol. Sci. 2021, 23, 9. [Google Scholar] [CrossRef]
Figure 1. The molecular selection processes. Abbreviation: AD: Alzheimer’s disease; GWAS: genome-wide association study; IS: ischemic stroke; miRNAs: microRNAs; SNP: single nucleotide polymorphism.
Figure 1. The molecular selection processes. Abbreviation: AD: Alzheimer’s disease; GWAS: genome-wide association study; IS: ischemic stroke; miRNAs: microRNAs; SNP: single nucleotide polymorphism.
Genes 14 00353 g001
Figure 2. The protein-protein interaction network of APP, MAPT, ApoE, and thirteen common protein coding susceptibility genes products (confidence score ≥ 0.4). MMP3 and MMP12 link with ApoE. Abbreviation: ApoE: apolipoprotein E; APP: amyloid precursor protein; MAPT: microtubule associated protein tau; MMP12: matrix metalloproteinase 12; MMP3: matrix metalloproteinase 3.
Figure 2. The protein-protein interaction network of APP, MAPT, ApoE, and thirteen common protein coding susceptibility genes products (confidence score ≥ 0.4). MMP3 and MMP12 link with ApoE. Abbreviation: ApoE: apolipoprotein E; APP: amyloid precursor protein; MAPT: microtubule associated protein tau; MMP12: matrix metalloproteinase 12; MMP3: matrix metalloproteinase 3.
Genes 14 00353 g002
Figure 3. The involvements of inflammation and immunity in AD and IS. Abbreviation: Aβ: amyloid β; AD: Alzheimer’s disease; IL-1β: interleukin-1β; IL-6: interleukin-6; IS: ischemic stroke; LDL: low density lipoprotein; MIF: macrophage-migration inhibitory factor; TNF-α: tumor necrosis factor-α; +: promote the progression.
Figure 3. The involvements of inflammation and immunity in AD and IS. Abbreviation: Aβ: amyloid β; AD: Alzheimer’s disease; IL-1β: interleukin-1β; IL-6: interleukin-6; IS: ischemic stroke; LDL: low density lipoprotein; MIF: macrophage-migration inhibitory factor; TNF-α: tumor necrosis factor-α; +: promote the progression.
Genes 14 00353 g003
Figure 4. The involvements of GPCRs in AD and IS. Abbreviation: Aβ: amyloid β; AD: Alzheimer’s disease; CDK-5: cyclin-dependent kinase-5; CX3CR1: CX3C chemokine receptor 1; ERKs: extracellular signal-regulated kinases; GPCR: G protein-coupled receptor; GSK-3β: glycogen synthase kinase-3β; IS: ischemic stroke; LPL: lysophospholipids; mAchRs: muscarinic acetylcholine receptors.
Figure 4. The involvements of GPCRs in AD and IS. Abbreviation: Aβ: amyloid β; AD: Alzheimer’s disease; CDK-5: cyclin-dependent kinase-5; CX3CR1: CX3C chemokine receptor 1; ERKs: extracellular signal-regulated kinases; GPCR: G protein-coupled receptor; GSK-3β: glycogen synthase kinase-3β; IS: ischemic stroke; LPL: lysophospholipids; mAchRs: muscarinic acetylcholine receptors.
Genes 14 00353 g004
Table 1. Common susceptibility genes and their related SNPs in AD and IS.
Table 1. Common susceptibility genes and their related SNPs in AD and IS.
Mapped GeneRegionSNPPosition Relative to GeneRisk Allelep ValueRisk toReference
ALDH1A215q21.3rs4775044intron variantN.A4.0 × 10−6ADSchwartzentruber et al., 2021 [33]
rs4471613intron variantA5.0 × 10−7ISCarty et al., 2015 [34]
ANKRD2210q23.31rs147285445intron variantT4.0 × 10−6ADMez et al., 2017 [35]
rs11202867intron variantN.A1.0 × 10−5ISKumar et al., 2021 [36]
ANTXR12p13.3rs7561207intron variantN.A4.0 × 10−6ADNazarian et al., 2019 [37]
rs149587156intergenic variantN.A5.0 × 10−6ISKumar et al., 2021 [36]
DIO2-AS114q31.1rs7155666intron variantA8.0 × 10−10ADChung et al., 2022 [38]
rs11846182intron variantT9.0 × 10−6ISLee et al., 2017 [39]
HDAC97p21.1rs117756856intron variantA9.0 × 10−6ADMez et al., 2017 [35]
rs11984041intron variantT5.0 × 10−9ISNetwork NSG. 2016 [40]
rs20239383’prime UTR variantG8.0 × 10−7ISMalik et al., 2017 [41]
rs71524263intron variantN.A2.0 × 10−12ISTraylor et al., 2017 [42]
rs2107595regulatory region variantN.A4.0 × 10−15ISMalik et al., 2018 [22]
JPH316q24.2rs117760708non coding transcript exon variantT3.0 × 10−6ADMez et al., 2017 [35]
rs12445022intergenic variantA9.0 × 10−8ISTraylor et al., 2021 [43]
KCNN31q21.3rs16830122intron variantA2.0 × 10−6ADJun et al., 2016 [44]
rs114812453intergenic variantN.A3.0 × 10−7ISKumar et al., 2021 [36]
LNC-LBCS6p22.3rs62402815intron variantN.A2.0 × 10−6ADNazarian et al., 2019 [37]
rs9348394intron variantN.A5.0 × 10−6ISKumar et al., 2021 [36]
MMP1211q22.2rs12808148intergenic variantN.A1.0 × 10−6ADKamboh et al., 2012 [45]
rs72983521intergenic variantN.A3.0 × 10−8ISMalik et al., 2018 [22]
MMP311q22.2rs12808148intergenic variantN.A1.0 × 10−6ADKamboh et al., 2012 [45]
rs72983521intergenic variantN.A5.0 × 10−8ISMalik et al., 2018 [22]
PCSK615q26.3rs146322114intron variantA2.0 × 10−6ADMez et al., 2017 [35]
rs528002287intron variantN.A6.0 × 10−6ISKumar et al., 2021 [36]
RBMS33p24.1rs17022021intron variantT8.0 × 10−6ADMez et al., 2017 [35]
rs115182009intron variantN.A9.0 × 10−7ISKumar et al., 2021 [36]
RNU6-909P5p14.1rs150631144intron variantT1.0 × 10−7ADMez et al., 2017 [35]
rs13354619intron variantN.A2.0 × 10−6ISKumar et al., 2021 [36]
Abbreviation: AD: Alzheimer’s disease; IS: ischemic stroke; N.A: not available; SNP: single nucleotide polymorphisms.
Table 2. The studies about the common genetic factors, pathways and molecular mechanisms between AD and IS.
Table 2. The studies about the common genetic factors, pathways and molecular mechanisms between AD and IS.
No.ReferenceAD and ISCommon Molecular Pathways InvolvedOthers
NoSGNoSS
1Traylor et al. 2016 [4]00Phospholipid efflux, cholesterol efflux, reverse cholesterol transport, negative regulation of nuclear factor kappa B (NF-κB) transcription factor activity (AD and small vessel disease) One region (ATP5H/KCTD2/ICT1) associated with both AD and small vessel disease
2Cui et al. 2018 [5]//Glioma, toll-like receptor signaling pathway, non-small cell lung cancer, natural killer cell mediated cytotoxicity, phospholipase D signaling pathway, hepatitis B, cadherin signaling pathway, wnt signaling pathway, immunoregulatory interactions between a lymphoid and a non-lymphoid cell, synthesis of PIPs at the plasma membrane, cooperation of PDCL (PhLP1) and TRiC/CCT in G-protein beta folding, PI metabolism, signaling pathways in glioblastoma 56 biological processes, 95 cellular components, and 28 molecular functions
3Wei et al. 2019 [6]16/Immune system /
4Rahman MR et al. 2019 [7]22/Alcoholism, MAPK signaling, glycine metabolism, serine metabolism, threonine metabolism Transcriptional regulator: SPIB, SMAD3, and SOX2
5Wang T, et al.2020 [8]00Different types of stroke, including any stroke, any ischemic stroke, large artery stroke, and cardio-embolic stroke would not be causally associated with AD risk/
6This study130MIF mediated glucocorticoid regulation, transendothelial migration of leukocytes, GPCR pathway, signal transduction, signaling by nuclear receptors, ESR-mediated signaling, ethanol effects on histone modifications, urokinase-type plasminogen activator (uPA) and uPAR-mediated signaling, UPA-UPAR pathway23 miRNAs regulate more than seven common risk genes
Abbreviation: ATP5H: adenosine triphosphate (ATP) synthase, H+ transporting, mitochondrial F0; ESR: estrogen receptor; GPCR: G protein-coupled receptors; ICT1: Immature colon carcinoma transcript 1; KCTD2: Potassium channel tetramerization domain-containing protein 2; MAPK: Mitogen-activated protein kinase; MIF: macrophage-migration inhibitory factor; miRNA: microRNA; NoSG: Number of Shared Genes, NoSS: Number of Shared SNP, PDCL: Phosducin like; PhLP1: PH domain and leucine rich repeat protein phosphatase 1; PI : Phosphatidylinositol; SMAD3: SMAD family member 3; SOX2: SRY-Box transcription factor 2; SPIB: Spi-B Transcription factor; TRiC/CCT: TCP1 ring complex/chaperonin containing TCP1 complex; uPAR: urokinase plasminogen activator receptor; /: not reported.8. Summary.
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.

Share and Cite

MDPI and ACS Style

Dong, W.; Huang, Y. Common Genetic Factors and Pathways in Alzheimer’s Disease and Ischemic Stroke: Evidences from GWAS. Genes 2023, 14, 353. https://doi.org/10.3390/genes14020353

AMA Style

Dong W, Huang Y. Common Genetic Factors and Pathways in Alzheimer’s Disease and Ischemic Stroke: Evidences from GWAS. Genes. 2023; 14(2):353. https://doi.org/10.3390/genes14020353

Chicago/Turabian Style

Dong, Wei, and Yue Huang. 2023. "Common Genetic Factors and Pathways in Alzheimer’s Disease and Ischemic Stroke: Evidences from GWAS" Genes 14, no. 2: 353. https://doi.org/10.3390/genes14020353

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop