Citation: Seoyoung Yoon, Yong-Ku Kim. The role of immunity and neuroinflammation in genetic predisposition and pathogenesis of Alzheimer's disease[J]. AIMS Genetics, 2015, 2(3): 230-249. doi: 10.3934/genet.2015.3.230
[1] | Sraboni Chaudhury . Epigenetic regulation in Autism spectrum disorder. AIMS Genetics, 2016, 3(4): 292-299. doi: 10.3934/genet.2016.4.292 |
[2] | Joanna L. Richens, Jonathan P. Bramble, Hannah L. Spencer, Fiona Cantlay, Molly Butler, Paul O’Shea . Towards defining the Mechanisms of Alzheimer’s disease based on a contextual analysis of molecular pathways. AIMS Genetics, 2016, 3(1): 25-48. doi: 10.3934/genet.2016.1.25 |
[3] | Tú Nguyen-Dumont, Jenna Stewart, Ingrid Winship, Melissa C. Southey . Rare genetic variants: making the connection with breast cancer susceptibility. AIMS Genetics, 2015, 2(4): 281-292. doi: 10.3934/genet.2015.4.281 |
[4] | Sandra Rugonyi . Genetic and flow anomalies in congenital heart disease. AIMS Genetics, 2016, 3(3): 157-166. doi: 10.3934/genet.2016.3.157 |
[5] | Michael J. Murray . Drosophila models of metastasis. AIMS Genetics, 2015, 2(1): 25-53. doi: 10.3934/genet.2015.1.25 |
[6] | Alena V Malakhova, Olga I Rudko, Vladimir V Sobolev, Artemii V Tretiakov, Elena A Naumova, Zarema G Kokaeva, Julia E Azimova, Eugene A Klimov . PDE4B gene polymorphism in Russian patients with panic disorder. AIMS Genetics, 2019, 6(3): 55-63. doi: 10.3934/genet.2019.3.55 |
[7] | Agnès Collet, Julien Tarabeux, Elodie Girard, Catherine Dubois DEnghien, Lisa Golmard, Vivien Deshaies, Alban Lermine, Anthony Laugé, Virginie Moncoutier, Cédrick Lefol, Florence Copigny, Catherine Dehainault, Henrique Tenreiro, Christophe Guy, Khadija Abidallah, Catherine Barbaroux, Etienne Rouleau, Nicolas Servant, Antoine De Pauw, Dominique Stoppa-Lyonnet, Claude Houdayer . Pros and cons of HaloPlex enrichment in cancer predisposition genetic diagnosis. AIMS Genetics, 2015, 2(4): 263-280. doi: 10.3934/genet.2015.4.263 |
[8] | Marcos Morey, Ana Fernández-Marmiesse, Jose Angel Cocho, María L. Couce . Influence of technology in genetic epidemiology. AIMS Genetics, 2015, 2(3): 219-229. doi: 10.3934/genet.2015.3.219 |
[9] | Eun Jeong Kim, Yong-Ku Kim . Panic disorders: The role of genetics and epigenetics. AIMS Genetics, 2018, 5(3): 177-190. doi: 10.3934/genet.2018.3.177 |
[10] | Harem Othman Smail . The epigenetics of diabetes, obesity, overweight and cardiovascular disease. AIMS Genetics, 2019, 6(3): 36-45. doi: 10.3934/genet.2019.3.36 |
Alzheimer’s disease is the major form of dementia, and is considered as important global concern. Alzheimer’s disease together with other forms of dementia is the 4th leading cause of years lost due to disability (YLD) in high income countries at 3.7 million YLD, representing 5.4% of the total YLD, which is higher than that of chronic obstructive pulmonary disease, diabetes mellitus, and cancer [1]. As both life expectancy and the size of the elderly population continue to increase, the prevalence of dementia is also increasing. In 2001, 24 million people were estimated to have dementia worldwide, and the prevalence of dementia is expected to double every 20 years to 42 million by 2020 and 81 million by 2040 [2]. There were 35.6 million people with dementia as of 2010 [3], approximately 43% of which require a high level of care such as nursing home care [4]. Alzheimer’s disease has a significant effect on society as well as patients and their caregivers; however, there is currently no validated disease modifying pharmacotherapy. Thus, there continues to be significant investigations aimed at understanding the pathogenesis of Alzheimer’s disease and treating the condition.
The exact pathogenesis of Alzheimer’s disease is largely obscure. The most representative pathological hallmarks of Alzheimer’s disease are senile plaques and neurofibrillary tangles. Other pathological findings such as granulovacuolar degeneration and amyloid vascular degeneration are also present. Senile plaques, which are also known as amyloid plaques, consist of extracellular deposition of amyloid-β (Aβ) peptides, Aβ40, and Aβ42. Likewise, neurofibrillary tangles are intracellular lesions consisting of structures of paired helical filaments composed of deposits of hyperphosphorylated tau protein. The amyloid cascade hypothesis suggests that Aβ protein is neurotoxic and causes neurofibrillary tangles, cell loss, vascular damage and dementia directly [5]. Indeed, the amyloid cascade hypothesis has been considered the leading pathogenesis of Alzheimer’s dementia for decades. The amyloid cascade hypothesis also suggests that, in addition to direct neurotoxic effects of Aβ on synapses and neurites, microglial and astrocytic activation induced by senile plaques and neurofibrillarytangles causes neuroinflammation, altered neuronal ionic homeostasis, and oxidative injury, eventually followed by aggravation of neuronal dysfunction and cell death. Decreased effective clearance of Aβ fibrils by microglia is also a key factor in pathogenesis of Alzheimer’s disease, as well as increased generation of Aβ fibrils [6]. Thus, immunological mechanism and neuroinflammation play an important role in the progression of Alzheimer’s disease. However, the process of immune reaction and neuroinflammation in Alzheimer’s disease and its molecular participants remain largely unknown.
Epidemiologicalstudies have revealed risk factors of Alzheimer’s disease, largely distinguished into vascular (such as, smoking, obesity, dyslipidemia, diabetes, hypertension and asymptomatic cerebral infarction), psychosocial (such as, lower education, poor social engagement and physical activities) and genetic factors [7]. Genetic factors play important role in Alzheimer’s disease and positive family history is a strong risk factor. Indeed, the Multi-Institutional Research in Alzheimer Genetic Epidemiology (MIRAGE) project reported that the lifetime risk of Alzheimer’s disease in first degree relatives is more than doubled compared to the general population [8]. Likewise, several large studies on genetically identical twins have confirmed the high heritability for Alzheimer’s disease, ranging from 58% to 80% [9,10]. Alzheimer’s disease can be divided into two groups based on genetic aspects, namely, “early-onset familial Alzheimer’s disease” (familial EOAD) and “late-onset Alzheimer’s disease” (LOAD). Familial EOAD seems to be inherited following Mendelian genetic distributionin an autosomal dominant pattern, with an average age of onset mostly before 60. The loci of causative genetic mutations are the amyloid precursor protein gene (APP gene in chromosome 21) and two presenilin genes (PSEN1 and PSEN2 located on chromosomes 14 and 1, respectively), all of which have complete penetrance. Conversely, LOAD accounts for more than 90% of Alzheimer’s disease patients, and is considered sporadic without a well-defined mode of transmission. On the other hand, LOAD is associated with substantial genetic effects, having heredity as the important causal factor [11,12]. In this way, LOAD appears to be inherited following “complex” or “non-Mendelian” genetic distribution, rather than sporadic. The APOE (apolipoprotein E) gene, which encoding ApoE has been confirmed as a risk factor for LOAD [13,14]. Specifically, ApoE plays a role in mediating cell signaling, synaptic plasticity, and neuroinflammation as well as in cholesterol and lipid transport [15]. APOE-ε4 alleles significantly increase the risk of Alzheimer’s disease in a dose dependent manner in both Caucasians and Asians. Furthermore, individuals with two APOE-ε4 alleles have from 15 to 17 times higher risk of Alzheimer’s disease than those without the APOE-ε4 allele [16,17]. However, the APOE genotype explains only about 20% of Alzheimer’s disease risk, and thus does not sufficiently explain the genetic variance of LOAD [18]. In addition to APOE, more than 500 candidate genes were suggested to be involved in LOAD between 1996 and 2005. Subsequently, the Alz Gene database (www.alzgene.org) was created in 2007 and includes updated information on 1395 studies, 320 meta-analyses, 695 genes, with 2973 polymorphisms [19]. Among the genes in the Alz Gene database, sortilin-related receptor 1 (SORL1) is associated with Alzheimer’s disease according to multiple meta-analysis [20,21].
Genome-wide association studies (GWAS) performed since 2007 have identified many genes not previously suspected of being associated with complex diseases such as Alzheimer’s disease [22]. Indeed, more than 20 genes associated with LOAD have been successfully identified with relative risks ranging from 1.1-1.3. Specifically, bridging integrator 1 (BIN1), complement receptor 1 (CR1), clusterin (CLU), phosphatidylinositol-binding clathrin assembly protein (https://www.aimspress.com/aimspress-data/aimsg/2015/3/PICALM), CD33, EPH receptor A1 (EPHA1), membrane-spanning 4-domains subfamily A (MS4A/MS4A6), ATP-binding cassette, sub-family A member 7 (ABCA7), CD2-associated protein(CD2AP), SORL1, major histocompatibility complex, class II (HLA-DRB5/DRB1), protein tyrosine kinase 2 beta (PTK2B), solute carrier family 24 member 4 (SLC24A4-RIN3), inositol polyphosphate-5-phosphatase (INPP5D), myocyte enhancer factor 2C (MEF2C), NME/NM23 family member 8 (NME8), zinc finger, CW type with PWWP domain 1 (ZCWPW1), CUGBP Elav-like family member 1 (CELF1), fermitin family member 2 (FERMT2), Cas scaffolding protein family member 4 (CASS4), and thyroid hormone receptor interactor 4 (TRIP4) have been identified as near-novel AD genes[23].
Whole genome sequencing methods have also been employed to identify new rare genetic variants. One such study showed that the triggering receptor expressed on myeloid cells 2 (TREM2) gene has an effect on the risk of developing LOAD [24]. Likewise, another whole exome sequencing (WES) study identified a rare variant of phospholipase D3 (PLD3) that doubles the risk of Alzheimer’s disease [25]. The underlying biological mechanisms of these newly identified genes in the development of Alzheimer’s disease are not yet fully understood, although a thorough understanding of the functional implications of these genes may suggest significant and novel pathogeneses of Alzheimer’s disease, providing new biomarkers and treatment strategies. Among possible underlying causes of Alzheimer’s disease, neuroinflammation remains a promising pathogenic mechanism. In this article, we aimed to review the role of neuroinflammation in development of Alzheimer’s disease and study the influence of known and novel genes.
Microglia are resident macrophages in the central nervous system (CNS) and serve as key cellular elements of the innate immune system of the CNS. Microglia survey their local microenvironment and defend against invading pathogens via phagocytosis, production of cytokines, oxyradicals, and activation of the complement cascade [26].Microglia also play roles in neurodevelopment such as synaptic remodeling and inducing apoptosis in supernumerary Purkinje cells, and may also produce neural growth factor and influence vascular development. In this way, microglia help to maintaining homeostasis of other cells such as astrocytes and neurons in the CNS [27,28].
Clustered microglia and fibrous astrocytes around extracellular deposits of Aβ have been observed in multiple publications [28,29]. Increased Aβ proteins, especially aggregation-prone Aβ42 species, lead to formation of amyloid fibrils and Aβ oligomers in Alzheimer’s disease. These misfolded Aβ peptides produce chemokines and also act directly as microglial attractants. Chemokine receptors such as CCR2, CCR3, CCR5, and CX3CR1 are present both in the microglia and brain of Alzheimer’s disease patients, and CXCR2 and CXCR33 are expressed in the vicinity of neuritic plaques [30]. Microglia, which are attracted to amyloid plaques, recognize and bind to Aβ oligomers and Aβ fibrils via cell surface receptors, such as CD36 [31,32], integrin-associated protein/CD47, α6β1 integrin [32], scavenger receptor A-1 (SCARA1) [33], Toll-like receptors (TLRs) [34,35,36], and associated receptors (e.g., CD14) [37]. After receptor ligation with Aβ fibrils, microglia engulf Aβ fibrils by phagocytosis in vitro and degrade soluble Aβ species via extracellular proteases, neprilysin, and insulin-degrading enzyme (IDE) [38,39]. In turn, ligation of CD36 and TLRs 4 and 6 with Aβ activate microglia to stimulate production of proinflammatory cytokines and chemokines [40].
Microglia activation is classified into several phenotypes, namely, the proinflammatory classical (M1) phenotype, non-inflammatory alternative (M2) phenotype, and acquired deactivation [41,42]. M1 is induced by T-helper-type 1 (Th1)cell-derived cytokines and causes a release of high levels of proinflammatory cytokines such as tumor necrosis factor-α (TNF-α), STAT3, interleukin-1β (IL-1β), IL-6, IL-12, IL-18 IL-23, reactive oxygen, and nitrogen species and corresponding toxic intermediates followed by impaired phagocytic capacity [43]. Inflammatory cytokines, reactive oxygen and nitrogen species can cause cell damage. M1 can also cause deterioration of the extracellular matrix by releasing proteolytic enzymes and degrade cellular integrity that lead to cellular destruction [42]. M2 is induced by Th2 cell-derived cytokines and causes increased release of anti-inflammatory cytokines such as IL-4, IL-10, IL-13 and transforming growth factor-β (TGF-β). This activation is also characterized by increased phagocytic capacity without toxic nitric oxide(NO)production. M2 macrophages also play a role in immunomodulation, homeostasis, neurogenesis, repair, and tissue remodeling as well as inflammation resolution [42,44]. The heterogeneity of such microglial activation states likely impacts development and progression of neurodegenerative diseases. Indeed, the characteristic switch from M2 to M1 seems to align with disease progression and age [45,46]. However, recent articles suggest that simple categorization according to the M1/M2 paradigm is insufficient, and that mononuclear phagocytes interact with various environmental challenges, are more complex, and exhibit mixed phenotypes [47].
Activation of microglia both beneficially and adversely affects the pathogenesis of Alzheimer’s disease. As the primary role of microglia is to defend against recognized pathologic changes while minimizing tissue damage, microglia have a central role in clearing toxic senile plaques via phagocytosis [48]. Microglia also degrade soluble Aβ through internalization with a fluid phase macropinocytic mechanism and subsequent proteolytic degradation in endolysosomal compartments [49]. Sphingolipid-modulated exosomes seem to promote microglia-mediated clearance of Aβ [50]. The complement system, a major constituent of the innate immune system, includes anaphylatoxins and opsonins and is associated with Aβ clearance and genetic ablation of complement factor C3 in mice, resulting in augmentation of Aβ deposition and neurodegeneration [51]. Likewise, microglia and astrocytes are responsible for production of proteins of the complement system [52].Decreased clearance of Aβ by microglia is considered to contribute to the pathogenesis of Alzheimer’s disease, and senescence of microglia and failure to appropriately convert the microglial phenotype may influence the effectiveness of microglial amyloid clearance [45,53].
Chronic microglial activation is problematic in the pathogenesis of Alzheimer’s disease. Specifically, a positive feedback loop between inflammation and APP processing results in chronic, non-resolving inflammation [54,55,56,57,58,59]. As M2promotescessation of inflammation, conversion of microglia is important. Such conversion is mediated by modification of proinflammatory signaling pathways (e.g. NLPR3 inflammasome) and heterodimeric type II nuclear receptors (e.g., PPARγ/RXR, PPARδ/RXR and LXR/RXR) [60]. Chronic inflammation causes enormous and sustained exposure to proinflammatory cytokines, chemokines, and other inflammatory mediators resulting in neuronal damage. Studies on the progression of mild cognitive impairment to Alzheimer’s dementia have shown that increased levels of proinflammatory cytokines such as TNF-α and decreased levels of the anti-inflammatory cytokine TGF-β are associated with the conversion to dementia [61]. Microglial activation and inflammatory responses promote activation of mitogen-activated protein kinase (MAPK), resulting in exacerbated hyperphosphorylation of tau protein in mouse models [57,62,63]. Specifically, cytokine-directed influences on tau pathologies interfere with neuronal transport by damaging neurons and aggravating the detrimental effects of tau on intraneuronal transport [64]. Inducible nitric oxide synthase (iNOS) in microglia and astroglia is stimulated in M1 and releases high concentrations of NO and other reactive oxygen species (ROS). ROS and reactive nitrogen species (RNS) are highly reactive and are capable of destabilizing proteins, lipids, carbohydrates, DNA, and RNA, leading to various types of damage including cell death [65].Furthermore, oxidative stress may aggravate the Aβ aggregation process [66]. Lastly, iNOS is expressed in the brain of patients with Alzheimer’s disease, and genetic ablation of iNOS in mice has a protective effect on the development of Alzheimer’s disease [67].
The paradoxical actions of inflammation and microglial activation on progression of Alzheimer’s disease, having both detrimental and beneficial effects, make the development of anti-inflammatory therapies difficult [68]. In PS1-APP micestudy, an animalmodel of Alzheimer’s disease, as mice getting older they exhibited decreased expression of some Aβ receptors such as SCARA, CD36, receptor for advanced-glycosylation end-products (RAGE) and Aβ degrading enzymes such as insulysin, neprilysin, and matrix metalloproteinase 9 (MMP9), whereas increased expression of the proinflammatory cytokines IL-1β and TNF-α. Further, TNF-α downregulates SCARA and CD36 expression. This findings suggest that early microglial activation is neuroprotective due to its Aβ clearance function, but as the disease progresses, proinflammatory cytokines downregulate genes involved in Aβclearance, promoting Aβ accumulation [69].
Macrophages distinct from microglia may take part in the pathogenesis of Alzheimer’s disease. Specifically, infiltrated blood macrophages from periphery and perivascular macrophages continue to be of interest in Alzheimer’s disease [70]. Stimulation of perivascular macrophages reduces cerebral amyloid angiopathy and deposition of Aβ in cerebral blood vessels and leptomeninges in mouse model of Alzheimer’s disease [71]. In addition, CCR2 is needed for Aβ clearance by perivascular macrophages [72]. Several mouse studies have reported higher infiltration rates of peripheral macrophages in the CNS of Alzheimer’s disease model mice compared to control mice [73,74]. The function of infiltrating macrophages in the clearance of Aβ is somewhat inconclusive, appearing transient with non-phagocytic activity [73], but exhibiting co-localization with lysosomes indicative of phagocytosis [75]. The possibility of access and activity in physiological milieu of macrophage in CNS is also questionable due to the use of radiation in previous studies, which may affect the integrity of the blood brain barrier and inflammatory milieu [76,77].
The main kind of neuroinflammation evoked in pathogenesis of traditional neuroinflammatory diseases, such as multiple sclerosis and encephalitis, and that of neurodegenerative diseases, such as Alzheimer’s disease, have been known to be different. The former is driven primarily by adaptive immune system (blood derived T and B lymphocytes) but the latter by CNS-resident immune cells (microglia, perivascular macrophages and astrocytes) [78]. On the other hand, multiple studies suggested Alzheimer’s disease is related with increased CNS T lymphocyte infiltration [79,80]. The exact mechanism of entering T lymphocytes into brain through blood-brain barrier and their roles in pathogenesis of Alzheimer’s disease is largely unknown. But several studies provide possible explanations [81]. Aβfibrils may served as modified self-antigen and induce Aβspecific T lymphocytes. Blood vessels adjacent to Aβfibrils express increased level of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) and lead extravasation of activated Aβspecific T lymphocytes [82]. In Alzheimer’s disease model mice, infiltration of IL-17 producing Th17 cells into brain parenchyma was found, which promotes microglial activation, releases proinflammatory cytokines and increases neuroinflammation [83,84,85]. Some other studies reported beneficial effects of T lymphocytes in Alzheimer’s disease. Injection of Aβ specific Th1 in mouse model of Alzheimer’s disease showed increased Aβ uptake and facilitated neurogenesis [86].
Change in lymphocytepopulation was also detected in peripheral immune system. Decreased peripheral T and B lymphocyte numbers were observed in Alzheimer’s disease patients [87]. And especially in severe stage, decrement in naïve CD4+ T lymphocyte subpopulation levels, CD19+ B lymphocytes, naïve B cells (IgD+CD27−) with increased double negative (IgD−CD27−) memory B cells were detected [88,89]. Decrease in naïve lymphocytes may be result of exhaustion due to chronic inflammation in Alzheimer’s disease. The expression of chemokine receptors in lymphocytes also altered in Alzheimer’s disease. Expression of pro-inflammatory chemokine receptors, CCR6 and CCR7, on B lymphocytes was increased, leading to chronic inflammatory condition [89]. Number of CD8+CD28− suppressor T lymphocytes and IL-10 production were decreased in Alzheimer’s disease, suggesting diminished immunosuppressive capabilities [90].
Astrocytes are the most abundant glial cells in the CNS where they play important roles such as maintenance of brain homeostasis, neurotransmitter regulation, signal transmission, and blood flow regulation. There is evidence that impaired and modified astroglial function contributes to the pathogenesis of Alzheimer’s disease [91,92,93,94]. Astrocytes may also contribute to progression of Alzheimer’s disease by directly and indirectly contributing to neuroinflammation.Activatedmicroglia produce cytokines, chemokines, ROS and recruit astrocytes around plaquespromotingtheir proliferation [95]. In addition, Aβitself could directlyactivate astrocytes [96]. Activated astrocytes take part in Aβ clearance via internalization and degradation of Aβ, and ApoE seems to facilitate this function [97,98]. Activated astrocytes release proinflammatory cytokines, chemokines and NO and ROS, and thus may participate in chronic inflammation resulting in neuronal damage. The overexpression of β-site amyloid precursor protein cleaving enzyme (BACE) and α1-antichymotrypsin in activated astrocytes may also induce further amyloidogenesis and tau hyperphosphorylation resulting in inhibition of Aβ breakdown, respectively [99].
The amyloid cascade hypothesis is regarded as a major pathogenetic hypothesis of Alzheimer’s disease. As mentioned above, amyloid fibrils and Aβ oligomers both play a major role in neuroinflammation by activating microglia and astrocytes, which in turn release proinflammatory cytokines, proteolytic enzymes, and NO, and subsequently activate the complement system and induce oxidative stress. Processing of APP can be divided into amyloidogenic and non-amyloidogenic pathways. In the non-amyloidogenic pathway, APP is cleaved by α-secretase, which is followed by further cleavage by γ-secretase. Non-toxic P3 peptide, α-APP, and the intracellular domain of APP (AICD) are secreted by this pathway. Conversely, β-secretase and γ-secretase producing β-APP, AICD, and Aβ peptides, which are the major pathological findings in Alzheimer’s disease, are secreted via the amyloidogenic pathway. The exact mechanism is largely unknown, but mutations in the APP genes (Chromosome 21) and genetic mutation of presenilin, the catalytic core of the γ-secretase (PSEN1 and PSEN2 on Chromosomes 14 and 1, respectively) results in relatively increased generation of aggregation prone Aβ42 species, which are largely responsible for familial EOAD [100].
Recent studies have suggested that some novel susceptibility genes may also influence Aβ production in LOAD. Specifically, the A disintegrin and metalloproteinase domain (ADAM) protease family is the executer of APP cleavage by α-secretase, and two rare mutations in ADAM10 have been suggested to predispose individuals to LOAD [101]. β-secretase cleavage requires recycling of APP from the cell surface, which in turn requires clathrin-mediated endocytosis of APP and its targeting to specific subcellular compartments. https://www.aimspress.com/aimspress-data/aimsg/2015/3/PICALM and SORL1 are involved in this process, and variants of these genes are associated with LOAD [102,103,104,105]. Formation of oligomers seems to exert increased Aβ neurotoxicity. In addition, clusterin influences aggregation and disaggregation of Aβ, and variants of CLU are associated with LOAD [106,107].
ApoE is a high density lipoprotein that plays important roles in the CNS, including regulation of phospholipid and cholesterol distribution, synaptogenesis, and synaptoplasticity. There are three common alleles of APOE (ε2, ε3, and ε4), among which APOE-ε4 significantly increases the risk of Alzheimer’s disease in a dose dependent manner while APOE-ε2 has a protective effect [16,17]. ApoE also seems to regulate innate immunity in the brain, influencing neuroinflammation and pathogenesis of Alzheimer’s disease. In addition, ApoE promotes co-localization, degradation and clearance of Aβ by astrocytes, and astrocyte-dependent lipidation of ApoE facilitates Aβ clearance by microglia [98,108]. ApoE also enhances endolytic degradation of Aβ by microglial neprilysin and related enzymes [109]. Interestingly, different isoforms of ApoE seem to influence inflammatory receptor signaling differently. In animal studies, binding of lipopolysaccharide (LPS) to CD14/TLR4 co-receptor (which bind to Aβ in Alzheimer’s disease) leads to dual pathway activation, namely, NF-κB mediating signaling, which is associated with immune modulation, and p38/MAPK-dependant signaling, which is related to increased paracrine damage by neurons. Targeted replacement (TR) of ε4 in mice is also associated with microglial p38/MAPK-dependent cytokine secretion and neuronal damage. Indeed, TR of ε2 revealed an association with the NF-κB mediated pathway [110,111]. Macrophages expressing APO-ε2 also exhibit greater Aβ degradation via MMP9 compared with APO-ε3 or ε4 [112].
GWAS and meta-analysis of LOAD consortium data have suggested several novel loci associated with LOAD risk: BIN1, CR1, CLU, https://www.aimspress.com/aimspress-data/aimsg/2015/3/PICALM, CD33, EPHA1, MS4A/MS4A6, ABCA7, CD2AP, SORL1, HLA-DRB5/DRb1, PTK2B, SLC24A4-RIN3, INPP5D, MEF2C, NME8, ZCWPW1, CELF1, FERMT2, CASS4, and TRIP4 [22,113,114,115]. In addition, whole genome sequencing methods such as WES identified rare genetic variants of TREM2 and PLD3 that are associated with increased risk of LOAD [24,25]. These loci may help to identify novel mechanisms of the pathogenesis in Alzheimer’s disease, deepening our understanding of the disease, and raising the possibility of finding new therapeutic options based on its pathogenetic mechanisms. Importantly, these novel genes seem to be associated with multiple mechanisms such as cell migration, lipid transport and endocytosis, amyloidogenesis, tauopathy, synaptic, and cytoskeletal function, and may have a role in immune processes and neuroinflammation [116].
CD33, TREM2, and CR1 are expressed on and modulate microglia, which play a major role in Aβ clearance and neuroinflammation as described above. Genetic variations of CD33 are associated with Alzheimer’s disease [114,115,117,118]. CD33 is located on 19q13.3 and encodes a member of the sialic acid-binding Ig-like lectin family, which is expressed on myeloid/microglial cells. CD33 is an inhibitory lectin, and mediates cell-cell interactions and inhibit immune cell function. In the CNS, CD33 impairs microglia-mediated Aβ clearance, resulting in facilitation of Aβ pathology. The C allele of rs3865444 is associated with greater cell surface expression of CD33 and increased Alzheimer’s disease risk [119,120]. Consistently, microglia with higher expression of CD33 exhibit diminished Aβ internalization resulting in accumulation of Aβ pathology on brain imaging with increased numbers of microglial activation [120]. CD33 may also play a role in other neuroinflammatory pathways mediated by microglia and should be studied further [119].
TREM2 is located on chromosome 6q21.1, and its expression on microglia facilitates phagocytosis and down-regulation of cytokine production and inflammation [121]. Mutation of TREM2 is associated with some forms of dementia such as Nasu-Hakola disease, a rare autosomal recessive form of dementia that presents with bone cysts, and familial frontotemporal dementia-like syndrome [122,123]. Rare missense mutations and rare multiple coding variants in TREM2 increase the risk of LOAD. According to recent meta-analyses, the most common variant of TREM2, R47H(rs75932628), is associated with from 2.70 to 4.11 increased LOAD risk relative to non-carriers [124,125]. Other TREM2 variants rs104894002 and rs143332484 are also associated with higher risk of Alzheimer’s disease with ORs of 7.21 and 1.65 respectively, although these risks were established with a small sample size and limited ethnicity data [124]. Consistently, Alzheimer’s disease patients that carry a TREM2 mutation have more extensive brain atrophy than non-carriers [126].
CR1 is located on chromosome 1q32 and encodes CR1 protein, which functions in the complement response as a receptor for C3b and C4b. Activation of complement cascade leads to opsonization and lysis of pathogens. Aβ can activate the complement system in vitro [127]. Single nucleotide polymorphisms (SNPs)rs6656401 and rs3818361 of CR1 are associated with increased LOAD risk, especially the latter in the presence of the APOE-ε4 allele [107]. Measurement of gene expression level on brain tissue revealed that higher CR1 expression is associated with increased cognitive decline and Alzheimer’s disease [128]. Among the isoforms of CR1, CR1-B/S increases complement activity via extra binding site for C3b/C4b, which is associated with increased Alzheimer’s disease risk [129]. CR1 and C3b moderate the complement activity, and presence of CR1 on phagocytic cells make them ingest particles that activate the complement response. A previous study demonstrated that clearance of Aβ in the circulation is dependent upon binding of C3b to erythrocyte-associated CR1, and also suggested that the purpose of CR1 expression in the brain is to act as a receptor for Aβ [130]. Together, these findings suggest that genetic variance in CR1 can alter the rate of Aβ clearance by the immune response. The complement system may also exert detrimental inflammation. Presence of complement system element and membrane attack complex C5b-9 is correlated with a high level of synaptic loss and Alzheimer’s disease [131]. Insufficient clearance of Aβ and increased chronic inflammation by CR1 genetic variation seems to affect pathogenesis of Alzheimer’s disease, with a recent study suggesting that forms of CR1 with impaired function are associated with increased risk of Alzheimer's disease through decreased clearance of Aβ [132].
Other LOAD related genes such as CLU and ABCA7 are also associated with the complement system. CLU is located on chromosome 8p21.1 and encodes the stress activated chaperone protein clusterin. SNPs including rs11136000, rs9331888, rs2279590, rs7982, and rs7012010 have a protective effect on LOAD risk, while rs9331896 is associated with increased risk of LOAD [114,115,118,133]. Increased plasma concentrations of clusterin are associated with entorhinal cortex atrophy and increased Aβ burden in the medial temporal lobe[134]. Clusterin was also described above as being involved in Aβ aggregation, with functions in lipid transport, membrane protection, cell-cell interaction, apoptosis, and complement regulation. With respect to the complement system, clusterin modulates the membrane attack complex and inhibits complement activation associated inflammatory response [135]. Furthermore, a mouse study showed that binding of Aβ to clusterin increases the rate of Aβ clearance and efflux through the blood-brain barrier [136]. ABCA7 is located on chromosome 19p13.3 and several SNPs, namely rs3764650, rs4147929, are associated with LOAD [115,118,133]. ABCA7, a member of the ABC transporter superfamily, transports substrates across cell membranes. ABCA7 modulates the C1q complement pathway and affects microglial phagocytosis of apoptotic cells and Aβ [137,138,139]. ABCA7 may also affect cholesterol transfer to ApoE and clearing Aβ aggregates [140,141]. Despite these findings and ongoing research, the precise mechanism of how the complement system influences the pathogenesis of Alzheimer’s disease remains largely unknown.
Several other LOAD associated genes such as MS4A, EPHA, INPP5D, MEF2C, and HLA- DRB5-DRB1 are related with immunity and inflammation, although the functional implication of these genes in the pathogenesis of Alzheimer’s disease is largely unknown. The MS4A gene cluster, composed of 12 related genes, is located on chromosome 11q12 [142], of which the SNPs rs983392 and rs670139 are associated with LOAD [114,115,118]. Overall the MS4A gene cluster is poorly characterized; however, MS4A1 (CD20), MS4A2 (FcεRIβ), and MS4A3 (HTm4) are best characterized of the cluster. It is known that the MS4A family functions to regulate calcium homeostasis, and that increased intracellular calcium may be associated with Aβ accumulation, tau hyperphosphorylation, and neuronal death. The MS4A family also affects progression of Alzheimer’s disease by modulating neuroinflammation. Specifically, MS4A1 (CD20) is expressed mainly by B lymphocytes, regulatingcalcium influx after activating B lymphocyte antigen receptor and related with immunological diseases [143,144]. In mouse study, neuroinflammation with B lymphocytes infiltration after stroke was related with development of dementia and anti-CD20 antibody prevented cognitive deficits [145]. Likewise, overexpression of MS4A genes increases T lymphocyte activation, facilitating the trafficking of T lymphocyte through the blood-brain barrier and decreasing T lymphocyte apoptosis. Activated T lymphocytes may also interact with microglia to promote the release of proinflammatory cytokines leading to immunesystem dysfunction and ensuing neuronal damage [142,146].
EPHA1 is located on chromosome 7q34 and encodes the ephrin type-A receptor, a member of the ephrin receptor subfamily of tyrosine kinase family. The SNPs rs11767557 and rs11771145 are associated with decreased LOAD risk [114,115,118]. In addition, a recent study found that EPHA1 (rs11771145) prevents hippocampal atrophy in mild cognitive impairment over a 2-year follow up period and is associated with decreased atrophy and greater cerebral glucose metabolic rate in the lateral occipitotemporal and inferior temporal gyrus in Alzheimer’s disease, without association with Aβ and tauopathy [147]. Thus, EPHA1 seems to contribute to axonal guidance and synaptic plasticity, modulation of the MAPK pathway, and response at glutamatergic synapses [148,149,150]. EPHA1 also seems to modulate immune function and chronic inflammation. The relationship between Eph/Ephrin signaling and inflammatory disease such as atherosclerosis, rheumatoid arthritis, and inflammatory bowel disease has also been reported [151,152]. Furthermore, Eph receptors reduce monocyte chemotaxis via expression in CD4-positive T lymphocytes and monocytes [153]. Ultimately, the pathogenetic contribution of Eph receptors to Alzheimer’s disease remains unclear.
INPP5D, a member of the INPP5 family, is located on chromosome 2q37.1 and encodes Src homology 2 domain-containing inositol-5-phosphatase (SHIP1), of which the SNP rs35349669 is associated with increased LOAD risk [115]. SHIP plays a central role in multiple signaling pathways, such as phosphatidylinositol 3-kinase pathway. SHIP1 attenuates immunoreceptor signaling and hematopoietic progenitor cell proliferation [154]. SHIP1 knockout mice also exhibit immunologic dysfunction with increased myeloid-derived suppressor cells and regulatory T cells as well as altered natural killer cell development [155]. MEF2C is located on 5q14.3, of which the SNP rs190982 is associated with increased LOAD risk [115]. MEF2C participates in the development and maintenance of multiple organ systems, and is also an effector of neurogenesis. Indeed, mutations and deletions in MEF2C are associated with mental retardation, seizure, cerebral malformation, stereotypic movements, and atony [156]. MEF2C also appears to downregulate inflammation. Specifically, MEF2C knockout mice have increased expression of proinflammatory molecules and NF-κB, a proinflammatory transcription factor. Consistently, overexpression of MEF2C suppresses both TNF-α levels and induction of proinflammatory molecules such as those involved in leukocyte adhesion [157]. HLA-DRB5/DRB1 is located on chromosome 6 and encodes HLA class II, DRβ5, and DRβ1, and is also associated with Alzheimer’s disease [115]. These HLA class II beta chain paralogues participate in the immune system by presenting peptides derived from extracellular protein. HLA-DRs seem to affect glial activity differently according to subtype. One study reported decreased glial fibrillary acidic protein in the hippocampus of Alzheimer’s disease patients who are HLA-DR4 allele carriers compared with HLA-DR4 negative subjects [158]. HLA-DR also influences T lymphocyte responses to Aβ differently according to subtype, and this difference in Aβ immunogenicity is considered critical for evaluating therapeutic and/or pathogenic potential of Aβ vaccines [159]. Currently, however, the autoimmune response in Alzheimer’s disease and how HLA subtypes affect the pathogenesis of Alzheimer’s disease is not well understood [160].
Neuroinflammation and immune reactions affect the pathogenesis of psychiatric disorders such as dementia, depression, and schizophrenia [161,162,163]. The pathogenesis of Alzheimer’s disease is also affected by immunological mechanisms. Aβ induced activation of immune system, such as microglia, astrocytes, other macrophages and lymphocytes, causes chronic inflammation withincreased levels of proinflammatory cytokines, ROS, and complement system giving detrimental effects on CNS. To date, the significance of immunological mechanisms has likely been underestimatedas part of the amyloid cascade hypothesis and much of the hereditary aspects of Alzheimer’s disease have pointed to APP processing and generation of Aβ. Conversely, GWAS studies and relatively new whole exome and genome sequencing techniques have revealed multiple novel genes associated with LOAD risk, many of which are related to the immune system. These findings suggest that immunological mechanisms are more than just a passive reaction to Aβ, and that alterations and the complex heterogeneity of the immune system in different individuals may modify development of Alzheimer’s disease, including time of onset and progression.
Many studies have attempted to target immune reactions and neuroinflammation as therapeutic targets in Alzheimer’s disease, but the results of these efforts have not been successful to date. Indeed, the heterogeneity of immunity in different individuals, different stage specific roles of genes, and different microglia phenotypes that may be modulated by multiple factors make such approaches difficult to pursue. Nevertheless, the detailed mechanisms of neuroinflammation and immune reactions associated with Alzheimer’s disease, including the identity of involved molecules, should be investigated more deeply. Newly identified LOAD risk genes indicate the possibility of novel, more complicated, and multifaceted mechanisms of neuroinflammation. Thus, future studies should be performed to determine the exact and precise mechanisms of these novel genes on Alzheimer’s disease, and integrate these findings together with those of previous studies. Together, such efforts will help to suggest new biomarkers beneficial for diagnosing Alzheimer’s disease as well as developing novel, individualized treatment strategies with superior disease-modifying outcomes.
The authors declare that there are no conflicts of interest related to this paper.
[1] | World Health Organization, The global burden of disease: 2004 update. Geneva 27, Switzerland, World Health Organization, 2008. Available from: http: //www.who.int/healthinfo/global_burden_disease/2004_report_update/en/ |
[2] |
Ferri CP, Prince M, Brayne C, et al. (2005) Global prevalence of dementia: a Delphi consensus study. Lancet 366: 2112-2117. doi: 10.1016/S0140-6736(05)67889-0
![]() |
[3] |
Prince M, Bryce R, Albanese E, et al. (2013) The global prevalence of dementia: a systematic review and metaanalysis. Alzheimers Dement 9: 63-75.e62. doi: 10.1016/j.jalz.2012.11.007
![]() |
[4] |
Brookmeyer R, Johnson E, Ziegler-Graham K, et al. (2007) Forecasting the global burden of Alzheimer's disease. Alzheimers Dement 3: 186-191. doi: 10.1016/j.jalz.2007.04.381
![]() |
[5] |
Hardy JA, Higgins GA (1992) Alzheimer's disease: the amyloid cascade hypothesis. Science 256: 184-185. doi: 10.1126/science.1566067
![]() |
[6] |
Hardy J, Selkoe DJ (2002) The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science 297: 353-356. doi: 10.1126/science.1072994
![]() |
[7] |
Povova J, Ambroz P, Bar M, et al. (2012) Epidemiological of and risk factors for Alzheimer's disease: a review. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 156: 108-114. doi: 10.5507/bp.2012.055
![]() |
[8] |
Lautenschlager NT, Cupples LA, Rao VS, et al. (1996) Risk of dementia among relatives of Alzheimer's disease patients in the MIRAGE study: What is in store for the oldest old? Neurology 46: 641-650. doi: 10.1212/WNL.46.3.641
![]() |
[9] |
Raiha I, Kaprio J, Koskenvuo M, et al. (1996) Alzheimer's disease in Finnish twins. Lancet 347: 573-578. doi: 10.1016/S0140-6736(96)91272-6
![]() |
[10] |
Gatz M, Reynolds CA, Fratiglioni L, et al. (2006) Role of genes and environments for explaining Alzheimer disease. Arch Gen Psychiatry 63: 168-174. doi: 10.1001/archpsyc.63.2.168
![]() |
[11] | Bergem AL, Engedal K, Kringlen E (1997) The role of heredity in late-onset Alzheimer disease and vascular dementia. A twin study. Arch Gen Psychiatry 54: 264-270. |
[12] | Gatz M, Pedersen NL, Berg S, et al. (1997) Heritability for Alzheimer's disease: the study of dementia in Swedish twins. J Gerontol A Biol Sci Med Sci 52: M117-125. |
[13] |
Coon KD, Myers AJ, Craig DW, et al. (2007) A high-density whole-genome association study reveals that APOE is the major susceptibility gene for sporadic late-onset Alzheimer's disease. J Clin Psychiatry 68: 613-618. doi: 10.4088/JCP.v68n0419
![]() |
[14] | Couzin J (2008) Genetics. Once shunned, test for Alzheimer's risk headed to market. Science 319: 1022-1023. |
[15] | Holtzman DM, Herz J, Bu G (2012) Apolipoprotein E and apolipoprotein E receptors: normal biology and roles in Alzheimer disease. Cold Spring Harb Perspect Med 2: a006312. |
[16] | Farrer LA, Cupples LA, Haines JL, et al. (1997) Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis Consortium. JAMA 278: 1349-1356. |
[17] |
Kim KW, Jhoo JH, Lee KU, et al. (1999) Association between apolipoprotein E polymorphism and Alzheimer's disease in Koreans. Neurosci Lett 277: 145-148. doi: 10.1016/S0304-3940(99)00867-8
![]() |
[18] |
Lambert JC, Amouyel P (2011) Genetics of Alzheimer's disease: new evidences for an old hypothesis? Curr Opin Genet Dev 21: 295-301. doi: 10.1016/j.gde.2011.02.002
![]() |
[19] |
Bertram L, McQueen MB, Mullin K, et al. (2007) Systematic meta-analyses of Alzheimer disease genetic association studies: the AlzGene database. Nat Genet 39: 17-23. doi: 10.1038/ng1934
![]() |
[20] |
Rogaeva E, Meng Y, Lee JH, et al. (2007) The neuronal sortilin-related receptor SORL1 is genetically associated with Alzheimer disease. Nat Genet 39: 168-177. doi: 10.1038/ng1943
![]() |
[21] |
Reitz C, Cheng R, Rogaeva E, et al. (2011) Meta-analysis of the association between variants in SORL1 and Alzheimer disease. Arch Neurol 68: 99-106. doi: 10.1001/archneurol.2010.346
![]() |
[22] |
Wellcome Trust Case Control Consortium (2007) Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447: 661-678. doi: 10.1038/nature05911
![]() |
[23] |
Chouraki V, Seshadri S (2014) Genetics of Alzheimer's disease. Adv Genet 87: 245-294. doi: 10.1016/B978-0-12-800149-3.00005-6
![]() |
[24] |
Jonsson T, Stefansson H, Steinberg S, et al. (2013) Variant of TREM2 associated with the risk of Alzheimer's disease. N Engl J Med 368: 107-116. doi: 10.1056/NEJMoa1211103
![]() |
[25] | Cruchaga C, Karch CM, Jin SC, et al. (2014) Rare coding variants in the phospholipase D3 gene confer risk for Alzheimer's disease. Nature 505: 550-554. |
[26] |
Graeber MB, Streit WJ (2010) Microglia: biology and pathology. Acta Neuropathol 119: 89-105. doi: 10.1007/s00401-009-0622-0
![]() |
[27] |
Ransohoff RM, Perry VH (2009) Microglial physiology: unique stimuli, specialized responses. Annu Rev Immunol 27: 119-145. doi: 10.1146/annurev.immunol.021908.132528
![]() |
[28] |
Streit WJ (2002) Microglia as neuroprotective, immunocompetent cells of the CNS. Glia 40: 133-139. doi: 10.1002/glia.10154
![]() |
[29] | Dickson DW, Farlo J, Davies P, et al. (1988) Alzheimer's disease. A double-labeling immunohistochemical study of senile plaques. Am J Pathol 132: 86-101. |
[30] |
Guillot-Sestier MV, Town T (2013) Innate immunity in Alzheimer's disease: a complex affair. CNS Neurol Disord Drug Targets 12: 593-607. doi: 10.2174/1871527311312050008
![]() |
[31] |
Coraci IS, Husemann J, Berman JW, et al. (2002) CD36, a class B scavenger receptor, is expressed on microglia in Alzheimer's disease brains and can mediate production of reactive oxygen species in response to beta-amyloid fibrils. Am J Pathol 160: 101-112. doi: 10.1016/S0002-9440(10)64354-4
![]() |
[32] | Bamberger ME, Harris ME, McDonald DR, et al. (2003) A cell surface receptor complex for fibrillar beta-amyloid mediates microglial activation. J Neurosci 23: 2665-2674. |
[33] | Frenkel D, Wilkinson K, Zhao L, et al. (2013) Scara1 deficiency impairs clearance of soluble amyloid-beta by mononuclear phagocytes and accelerates Alzheimer's-like disease progression. Nat Commun 4: 2030. |
[34] |
Walter S, Letiembre M, Liu Y, et al. (2007) Role of the toll-like receptor 4 in neuroinflammation in Alzheimer's disease. Cell Physiol Biochem 20: 947-956. doi: 10.1159/000110455
![]() |
[35] | Scholtzova H, Chianchiano P, Pan J, et al. (2014) Amyloid beta and Tau Alzheimer's disease related pathology is reduced by Toll-like receptor 9 stimulation. Acta Neuropathol Commun 2: 101. |
[36] |
Wang LZ, Tian Y, Yu JT, et al. (2011) Association between late-onset Alzheimer's disease and microsatellite polymorphisms in intron II of the human toll-like receptor 2 gene. Neurosci Lett 489: 164-167. doi: 10.1016/j.neulet.2010.12.008
![]() |
[37] |
Liu Y, Walter S, Stagi M, et al. (2005) LPS receptor (CD14): a receptor for phagocytosis of Alzheimer's amyloid peptide. Brain 128: 1778-1789. doi: 10.1093/brain/awh531
![]() |
[38] |
Prokop S, Miller KR, Heppner FL (2013) Microglia actions in Alzheimer's disease. Acta Neuropathol 126: 461-477. doi: 10.1007/s00401-013-1182-x
![]() |
[39] |
Lee CY, Landreth GE (2010) The role of microglia in amyloid clearance from the AD brain. J Neural Transm 117: 949-960. doi: 10.1007/s00702-010-0433-4
![]() |
[40] |
Stewart CR, Stuart LM, Wilkinson K, et al. (2010) CD36 ligands promote sterile inflammation through assembly of a Toll-like receptor 4 and 6 heterodimer. Nat Immunol 11: 155-161. doi: 10.1038/ni.1836
![]() |
[41] |
Town T, Nikolic V, Tan J (2005) The microglial ""activation"" continuum: from innate to adaptive responses. J Neuroinflammation 2: 24. doi: 10.1186/1742-2094-2-24
![]() |
[42] |
Varnum MM, Ikezu T (2012) The classification of microglial activation phenotypes on neurodegeneration and regeneration in Alzheimer's disease brain. Arch Immunol Ther Exp (Warsz) 60: 251-266. doi: 10.1007/s00005-012-0181-2
![]() |
[43] |
Mantovani A, Sozzani S, Locati M, et al. (2002) Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol 23: 549-555. doi: 10.1016/S1471-4906(02)02302-5
![]() |
[44] |
Goerdt S, Orfanos CE (1999) Other functions, other genes: alternative activation of antigen-presenting cells. Immunity 10: 137-142. doi: 10.1016/S1074-7613(00)80014-X
![]() |
[45] |
Jimenez S, Baglietto-Vargas D, Caballero C, et al. (2008) Inflammatory response in the hippocampus of PS1M146L/APP751SL mouse model of Alzheimer's disease: age-dependent switch in the microglial phenotype from alternative to classic. J Neurosci 28: 11650-11661. doi: 10.1523/JNEUROSCI.3024-08.2008
![]() |
[46] |
Hoozemans JJ, Veerhuis R, Rozemuller JM, et al. (2006) Neuroinflammation and regeneration in the early stages of Alzheimer's disease pathology. Int J Dev Neurosci 24: 157-165. doi: 10.1016/j.ijdevneu.2005.11.001
![]() |
[47] | Martinez FO, Gordon S (2014) The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep 6: 13. |
[48] |
Simard AR, Soulet D, Gowing G, et al. (2006) Bone marrow-derived microglia play a critical role in restricting senile plaque formation in Alzheimer's disease. Neuron 49: 489-502. doi: 10.1016/j.neuron.2006.01.022
![]() |
[49] |
Mandrekar S, Jiang Q, Lee CY, et al. (2009) Microglia mediate the clearance of soluble Abeta through fluid phase macropinocytosis. J Neurosci 29: 4252-4262. doi: 10.1523/JNEUROSCI.5572-08.2009
![]() |
[50] |
Yuyama K, Sun H, Mitsutake S, et al. (2012) Sphingolipid-modulated exosome secretion promotes clearance of amyloid-beta by microglia. J Biol Chem 287: 10977-10989. doi: 10.1074/jbc.M111.324616
![]() |
[51] |
Maier M, Peng Y, Jiang L, et al. (2008) Complement C3 deficiency leads to accelerated amyloid beta plaque deposition and neurodegeneration and modulation of the microglia/macrophage phenotype in amyloid precursor protein transgenic mice. J Neurosci 28: 6333-6341. doi: 10.1523/JNEUROSCI.0829-08.2008
![]() |
[52] |
Webster S, Lue LF, Brachova L, et al. (1997) Molecular and cellular characterization of the membrane attack complex, C5b-9, in Alzheimer's disease. Neurobiol Aging 18: 415-421. doi: 10.1016/S0197-4580(97)00042-0
![]() |
[53] |
Flanary BE, Streit WJ (2004) Progressive telomere shortening occurs in cultured rat microglia, but not astrocytes. Glia 45: 75-88. doi: 10.1002/glia.10301
![]() |
[54] | Griffin WS, Sheng JG, Royston MC, et al. (1998) Glial-neuronal interactions in Alzheimer's disease: the potential role of a 'cytokine cycle' in disease progression. Brain Pathol 8: 65-72. |
[55] |
Griffin WS, Stanley LC, Ling C, et al. (1989) Brain interleukin 1 and S-100 immunoreactivity are elevated in Down syndrome and Alzheimer disease. Proc Natl Acad Sci U S A 86: 7611-7615. doi: 10.1073/pnas.86.19.7611
![]() |
[56] |
Barger SW, Harmon AD (1997) Microglial activation by Alzheimer amyloid precursor protein and modulation by apolipoprotein E. Nature 388: 878-881. doi: 10.1038/42257
![]() |
[57] | Li Y, Liu L, Barger SW, et al. (2003) Interleukin-1 mediates pathological effects of microglia on tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway. J Neurosci 23: 1605-1611. |
[58] | Li Y, Liu L, Kang J, et al. (2000) Neuronal-glial interactions mediated by interleukin-1 enhance neuronal acetylcholinesterase activity and mRNA expression. J Neurosci 20: 149-155. |
[59] |
Sheng JG, Ito K, Skinner RD, et al. (1996) In vivo and in vitro evidence supporting a role for the inflammatory cytokine interleukin-1 as a driving force in Alzheimer pathogenesis. Neurobiol Aging 17: 761-766. doi: 10.1016/0197-4580(96)00104-2
![]() |
[60] |
Yamanaka M, Ishikawa T, Griep A, et al. (2012) PPARgamma/RXRalpha-induced and CD36-mediated microglial amyloid-beta phagocytosis results in cognitive improvement in amyloid precursor protein/presenilin 1 mice. J Neurosci 32: 17321-17331. doi: 10.1523/JNEUROSCI.1569-12.2012
![]() |
[61] |
Tarkowski E, Andreasen N, Tarkowski A, et al. (2003) Intrathecal inflammation precedes development of Alzheimer's disease. J Neurol Neurosurg Psychiatry 74: 1200-1205. doi: 10.1136/jnnp.74.9.1200
![]() |
[62] |
Sheng JG, Jones RA, Zhou XQ, et al. (2001) Interleukin-1 promotion of MAPK-p38 overexpression in experimental animals and in Alzheimer's disease: potential significance for tau protein phosphorylation. Neurochem Int 39: 341-348. doi: 10.1016/S0197-0186(01)00041-9
![]() |
[63] |
Munoz L, Ralay Ranaivo H, Roy SM, et al. (2007) A novel p38 alpha MAPK inhibitor suppresses brain proinflammatory cytokine up-regulation and attenuates synaptic dysfunction and behavioral deficits in an Alzheimer's disease mouse model. J Neuroinflammation 4: 21. doi: 10.1186/1742-2094-4-21
![]() |
[64] |
Yoshiyama Y, Higuchi M, Zhang B, et al. (2007) Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron 53: 337-351. doi: 10.1016/j.neuron.2007.01.010
![]() |
[65] |
Butterfield DA, Swomley AM, Sultana R (2013) Amyloid beta-peptide (1-42)-induced oxidative stress in Alzheimer disease: importance in disease pathogenesis and progression. Antioxid Redox Signal 19: 823-835. doi: 10.1089/ars.2012.5027
![]() |
[66] |
Thiabaud G, Pizzocaro S, Garcia-Serres R, et al. (2013) Heme binding induces dimerization and nitration of truncated beta-amyloid peptide Abeta16 under oxidative stress. Angew Chem Int Ed Engl 52: 8041-8044. doi: 10.1002/anie.201302989
![]() |
[67] |
Nathan C, Calingasan N, Nezezon J, et al. (2005) Protection from Alzheimer's-like disease in the mouse by genetic ablation of inducible nitric oxide synthase. J Exp Med 202: 1163-1169. doi: 10.1084/jem.20051529
![]() |
[68] | Aboud O, Parcon PA, DeWall KM, et al. (2015) Aging, Alzheimer's, and APOE genotype influence the expression and neuronal distribution patterns of microtubule motor protein dynactin-P50. Front Cell Neurosci 9: 103. |
[69] |
Hickman SE, Allison EK, El Khoury J (2008) Microglial dysfunction and defective beta-amyloid clearance pathways in aging Alzheimer's disease mice. J Neurosci 28: 8354-8360. doi: 10.1523/JNEUROSCI.0616-08.2008
![]() |
[70] |
Lai AY, McLaurin J (2012) Clearance of amyloid-beta peptides by microglia and macrophages: the issue of what, when and where. Future Neurol 7: 165-176. doi: 10.2217/fnl.12.6
![]() |
[71] |
Hawkes CA, McLaurin J (2009) Selective targeting of perivascular macrophages for clearance of beta-amyloid in cerebral amyloid angiopathy. Proc Natl Acad Sci U S A 106: 1261-1266. doi: 10.1073/pnas.0805453106
![]() |
[72] |
Mildner A, Schlevogt B, Kierdorf K, et al. (2011) Distinct and non-redundant roles of microglia and myeloid subsets in mouse models of Alzheimer's disease. J Neurosci 31: 11159-11171. doi: 10.1523/JNEUROSCI.6209-10.2011
![]() |
[73] |
Stalder AK, Ermini F, Bondolfi L, et al. (2005) Invasion of hematopoietic cells into the brain of amyloid precursor protein transgenic mice. J Neurosci 25: 11125-11132. doi: 10.1523/JNEUROSCI.2545-05.2005
![]() |
[74] |
Malm TM, Koistinaho M, Parepalo M, et al. (2005) Bone-marrow-derived cells contribute to the recruitment of microglial cells in response to beta-amyloid deposition in APP/PS1 double transgenic Alzheimer mice. Neurobiol Dis 18: 134-142 doi: 10.1016/j.nbd.2004.09.009
![]() |
[75] |
Fiala M, Liu PT, Espinosa-Jeffrey A, et al. (2007) Innate immunity and transcription of MGAT-III and Toll-like receptors in Alzheimer's disease patients are improved by bisdemethoxycurcumin. Proc Natl Acad Sci U S A 104: 12849-12854. doi: 10.1073/pnas.0701267104
![]() |
[76] |
Mildner A, Schmidt H, Nitsche M, et al. (2007) Microglia in the adult brain arise from Ly-6ChiCCR2+ monocytes only under defined host conditions. Nat Neurosci 10: 1544-1553. doi: 10.1038/nn2015
![]() |
[77] |
Ajami B, Bennett JL, Krieger C, et al. (2007) Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nat Neurosci 10: 1538-1543. doi: 10.1038/nn2014
![]() |
[78] |
Heppner FL, Ransohoff RM, Becher B (2015) Immune attack: the role of inflammation in Alzheimer disease. Nat Rev Neurosci 16: 358-372. doi: 10.1038/nrn3880
![]() |
[79] |
Togo T, Akiyama H, Iseki E, et al. (2002) Occurrence of T cells in the brain of Alzheimer's disease and other neurological diseases. J Neuroimmunol 124: 83-92. doi: 10.1016/S0165-5728(01)00496-9
![]() |
[80] |
Town T, Tan J, Flavell RA, et al. (2005) T-cells in Alzheimer's disease. Neuromolecular Med 7: 255-264. doi: 10.1385/NMM:7:3:255
![]() |
[81] |
Hohsfield LA, Humpel C (2015) Migration of blood cells to beta-amyloid plaques in Alzheimer's disease. Exp Gerontol 65: 8-15. doi: 10.1016/j.exger.2015.03.002
![]() |
[82] | Fisher Y, Nemirovsky A, Baron R, et al. (2011) Dendritic cells regulate amyloid-beta-specific T-cell entry into the brain: the role of perivascular amyloid-beta. J Alzheimers Dis 27: 99-111. |
[83] |
Zhang J, Ke KF, Liu Z, et al. (2013) Th17 cell-mediated neuroinflammation is involved in neurodegeneration of abeta1-42-induced Alzheimer's disease model rats. PLoS One 8: e75786. doi: 10.1371/journal.pone.0075786
![]() |
[84] |
Gonzalez H, Pacheco R (2014) T-cell-mediated regulation of neuroinflammation involved in neurodegenerative diseases. J Neuroinflammation 11: 201. doi: 10.1186/s12974-014-0201-8
![]() |
[85] |
Browne TC, McQuillan K, McManus RM, et al. (2013) IFN-gamma Production by amyloid beta-specific Th1 cells promotes microglial activation and increases plaque burden in a mouse model of Alzheimer's disease. J Immunol 190: 2241-2251. doi: 10.4049/jimmunol.1200947
![]() |
[86] |
Fisher Y, Strominger I, Biton S, et al. (2014) Th1 polarization of T cells injected into the cerebrospinal fluid induces brain immunosurveillance. J Immunol 192: 92-102. doi: 10.4049/jimmunol.1301707
![]() |
[87] |
Richartz-Salzburger E, Batra A, Stransky E, et al. (2007) Altered lymphocyte distribution in Alzheimer's disease. J Psychiatr Res 41: 174-178. doi: 10.1016/j.jpsychires.2006.01.010
![]() |
[88] |
Bonotis K, Krikki E, Holeva V, et al. (2008) Systemic immune aberrations in Alzheimer's disease patients. J Neuroimmunol 193: 183-187. doi: 10.1016/j.jneuroim.2007.10.020
![]() |
[89] | Bulati M, Buffa S, Martorana A, et al. (2015) Double negative (IgG+IgD-CD27-) B cells are increased in a cohort of moderate-severe Alzheimer's disease patients and show a pro-inflammatory trafficking receptor phenotype. J Alzheimers Dis 44: 1241-1251. |
[90] |
Speciale L, Calabrese E, Saresella M, et al. (2007) Lymphocyte subset patterns and cytokine production in Alzheimer's disease patients. Neurobiol Aging 28: 1163-1169. doi: 10.1016/j.neurobiolaging.2006.05.020
![]() |
[91] |
Xiao M, Hu G (2014) Involvement of aquaporin 4 in astrocyte function and neuropsychiatric disorders. CNS Neurosci Ther 20: 385-390. doi: 10.1111/cns.12267
![]() |
[92] |
Grolla AA, Fakhfouri G, Balzaretti G, et al. (2013) Abeta leads to Ca(2)(+) signaling alterations and transcriptional changes in glial cells. Neurobiol Aging 34: 511-522. doi: 10.1016/j.neurobiolaging.2012.05.005
![]() |
[93] |
Kato S, Gondo T, Hoshii Y, et al. (1998) Confocal observation of senile plaques in Alzheimer's disease: senile plaque morphology and relationship between senile plaques and astrocytes. Pathol Int 48: 332-340. doi: 10.1111/j.1440-1827.1998.tb03915.x
![]() |
[94] |
Kulijewicz-Nawrot M, Verkhratsky A, Chvatal A, et al. (2012) Astrocytic cytoskeletal atrophy in the medial prefrontal cortex of a triple transgenic mouse model of Alzheimer's disease. J Anat 221: 252-262. doi: 10.1111/j.1469-7580.2012.01536.x
![]() |
[95] |
Heneka MT, O'Banion MK, Terwel D, et al. (2010) Neuroinflammatory processes in Alzheimer's disease. J Neural Transm 117: 919-947. doi: 10.1007/s00702-010-0438-z
![]() |
[96] |
Avila-Munoz E, Arias C (2014) When astrocytes become harmful: functional and inflammatory responses that contribute to Alzheimer's disease. Ageing Res Rev 18: 29-40. doi: 10.1016/j.arr.2014.07.004
![]() |
[97] |
Wyss-Coray T, Loike JD, Brionne TC, et al. (2003) Adult mouse astrocytes degrade amyloid-beta in vitro and in situ. Nat Med 9: 453-457. doi: 10.1038/nm838
![]() |
[98] |
Koistinaho M, Lin S, Wu X, et al. (2004) Apolipoprotein E promotes astrocyte colocalization and degradation of deposited amyloid-beta peptides. Nat Med 10: 719-726. doi: 10.1038/nm1058
![]() |
[99] | Dezsi L, Tuka B, Martos D, et al. (2015) Alzheimer's disease, astrocytes and kynurenines. Curr Alzheimer Res 12: 462-480. |
[100] |
Chavez-Gutierrez L, Bammens L, Benilova I, et al. (2012) The mechanism of gamma-Secretase dysfunction in familial Alzheimer disease. Embo j 31: 2261-2274. doi: 10.1038/emboj.2012.79
![]() |
[101] |
Suh J, Choi SH, Romano DM, et al. (2013) ADAM10 missense mutations potentiate beta-amyloid accumulation by impairing prodomain chaperone function. Neuron 80: 385-401. doi: 10.1016/j.neuron.2013.08.035
![]() |
[102] |
Jun G, Naj AC, Beecham GW, et al. (2010) Meta-analysis confirms CR1, CLU, and PICALM as alzheimer disease risk loci and reveals interactions with APOE genotypes. Arch Neurol 67: 1473-1484. doi: 10.1001/archneurol.2010.201
![]() |
[103] | Jin C, Liu X, Zhang F, et al. (2013) An updated meta-analysis of the association between SORL1 variants and the risk for sporadic Alzheimer's disease. J Alzheimers Dis 37: 429-437. |
[104] |
Li Y, Rowland C, Catanese J, et al. (2008) SORL1 variants and risk of late-onset Alzheimer's disease. Neurobiol Dis 29: 293-296. doi: 10.1016/j.nbd.2007.09.001
![]() |
[105] |
Xiao Q, Gil SC, Yan P, et al. (2012) Role of phosphatidylinositol clathrin assembly lymphoid-myeloid leukemia (PICALM) in intracellular amyloid precursor protein (APP) processing and amyloid plaque pathogenesis. J Biol Chem 287: 21279-21289. doi: 10.1074/jbc.M111.338376
![]() |
[106] | Narayan P, Orte A, Clarke RW, et al. (2012) The extracellular chaperone clusterin sequesters oligomeric forms of the amyloid-beta(1-40) peptide. Nat Struct Mol Biol 19: 79-83. |
[107] |
Lambert JC, Heath S, Even G, et al. (2009) Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer's disease. Nat Genet 41: 1094-1099. doi: 10.1038/ng.439
![]() |
[108] |
Terwel D, Steffensen KR, Verghese PB, et al. (2011) Critical role of astroglial apolipoprotein E and liver X receptor-alpha expression for microglial Abeta phagocytosis. J Neurosci 31: 7049-7059. doi: 10.1523/JNEUROSCI.6546-10.2011
![]() |
[109] |
Jiang Q, Lee CY, Mandrekar S, et al. (2008) ApoE promotes the proteolytic degradation of Abeta. Neuron 58: 681-693. doi: 10.1016/j.neuron.2008.04.010
![]() |
[110] |
Maezawa I, Maeda N, Montine TJ, et al. (2006) Apolipoprotein E-specific innate immune response in astrocytes from targeted replacement mice. J Neuroinflammation 3: 10. doi: 10.1186/1742-2094-3-10
![]() |
[111] | Maezawa I, Nivison M, Montine KS, et al. (2006) Neurotoxicity from innate immune response is greatest with targeted replacement of E4 allele of apolipoprotein E gene and is mediated by microglial p38MAPK. FASEB J 20: 797-799 |
[112] |
Zhao L, Lin S, Bales KR, et al. (2009) Macrophage-mediated degradation of beta-amyloid via an apolipoprotein E isoform-dependent mechanism. J Neurosci 29: 3603-3612. doi: 10.1523/JNEUROSCI.5302-08.2009
![]() |
[113] | Seshadri S, Fitzpatrick AL, Ikram MA, et al. (2010) Genome-wide analysis of genetic loci associated with Alzheimer disease. JAMA 303: 1832-1840 |
[114] |
Naj AC, Jun G, Beecham GW, et al. (2011) Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer's disease. Nat Genet 43: 436-441. doi: 10.1038/ng.801
![]() |
[115] |
Lambert JC, Ibrahim-Verbaas CA, Harold D, et al. (2013) Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer's disease. Nat Genet 45: 1452-1458. doi: 10.1038/ng.2802
![]() |
[116] |
Reitz C (2014) Genetic loci associated with Alzheimer's disease. Future Neurol 9: 119-122 doi: 10.2217/fnl.14.1
![]() |
[117] |
Malik M, Simpson JF, Parikh I, et al. (2013) CD33 Alzheimer's risk-altering polymorphism, CD33 expression, and exon 2 splicing. J Neurosci 33: 13320-13325. doi: 10.1523/JNEUROSCI.1224-13.2013
![]() |
[118] |
Hollingworth P, Harold D, Sims R, et al. (2011) Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer's disease. Nat Genet 43: 429-435. doi: 10.1038/ng.803
![]() |
[119] |
Jiang T, Yu JT, Hu N, et al. (2014) CD33 in Alzheimer's disease. Mol Neurobiol 49: 529-535. doi: 10.1007/s12035-013-8536-1
![]() |
[120] |
Bradshaw EM, Chibnik LB, Keenan BT, et al. (2013) CD33 Alzheimer's disease locus: altered monocyte function and amyloid biology. Nat Neurosci 16: 848-850. doi: 10.1038/nn.3435
![]() |
[121] | Rohn TT (2013) The triggering receptor expressed on myeloid cells 2: ""TREM-ming"" the inflammatory component associated with Alzheimer's disease. Oxid Med Cell Longev 2013: 860959. |
[122] | Le Ber I, De Septenville A, Guerreiro R, et al. (2014) Homozygous TREM2 mutation in a family with atypical frontotemporal dementia. Neurobiol Aging 35: 2419.e2423-2415. |
[123] | Sasaki A, Kakita A, Yoshida K, et al. (2015) Variable expression of microglial DAP12 and TREM2 genes in Nasu-Hakola disease. Neurogenetics. In press. |
[124] | Lu Y, Liu W, Wang X (2015) TREM2 variants and risk of Alzheimer's disease: a meta-analysis. Neurol Sci. In press. |
[125] | Ruiz A, Dols-Icardo O, Bullido MJ, et al. (2014) Assessing the role of the TREM2 p.R47H variant as a risk factor for Alzheimer's disease and frontotemporal dementia. Neurobiol Aging 35: 444.e441-444. |
[126] | Rajagopalan P, Hibar DP, Thompson PM (2013) TREM2 and neurodegenerative disease. N Engl J Med 369: 1565-1567. |
[127] |
Strohmeyer R, Ramirez M, Cole GJ, et al. (2002) Association of factor H of the alternative pathway of complement with agrin and complement receptor 3 in the Alzheimer's disease brain. J Neuroimmunol 131: 135-146. doi: 10.1016/S0165-5728(02)00272-2
![]() |
[128] |
Karch CM, Jeng AT, Nowotny P, et al. (2012) Expression of novel Alzheimer's disease risk genes in control and Alzheimer's disease brains. PLoS One 7: e50976. doi: 10.1371/journal.pone.0050976
![]() |
[129] |
Brouwers N, Van Cauwenberghe C, Engelborghs S, et al. (2012) Alzheimer risk associated with a copy number variation in the complement receptor 1 increasing C3b/C4b binding sites. Mol Psychiatry 17: 223-233. doi: 10.1038/mp.2011.24
![]() |
[130] |
Rogers J, Li R, Mastroeni D, et al. (2006) Peripheral clearance of amyloid beta peptide by complement C3-dependent adherence to erythrocytes. Neurobiol Aging 27: 1733-1739. doi: 10.1016/j.neurobiolaging.2005.09.043
![]() |
[131] |
Lue LF, Brachova L, Civin WH, et al. (1996) Inflammation, A beta deposition, and neurofibrillary tangle formation as correlates of Alzheimer's disease neurodegeneration. J Neuropathol Exp Neurol 55: 1083-1088. doi: 10.1097/00005072-199655100-00008
![]() |
[132] | Mahmoudi R, Kisserli A, Novella JL, et al. (2015) Alzheimer's disease is associated with low density of the long CR1 isoform. Neurobiol Aging 36: 1766.e1765-1712. |
[133] |
Harold D, Abraham R, Hollingworth P, et al. (2009) Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer's disease. Nat Genet 41: 1088-1093. doi: 10.1038/ng.440
![]() |
[134] |
Thambisetty M, Simmons A, Velayudhan L, et al. (2010) Association of plasma clusterin concentration with severity, pathology, and progression in Alzheimer disease. Arch Gen Psychiatry 67: 739-748. doi: 10.1001/archgenpsychiatry.2010.78
![]() |
[135] |
Jones SE, Jomary C (2002) Clusterin. Int J Biochem Cell Biol 34: 427-431. doi: 10.1016/S1357-2725(01)00155-8
![]() |
[136] | Bell RD, Sagare AP, Friedman AE, et al. (2007) Transport pathways for clearance of human Alzheimer's amyloid beta-peptide and apolipoproteins E and J in the mouse central nervous system. J Cereb Blood Flow Metab 27: 909-918. |
[137] |
Jehle AW, Gardai SJ, Li S, et al. (2006) ATP-binding cassette transporter A7 enhances phagocytosis of apoptotic cells and associated ERK signaling in macrophages. J Cell Biol 174: 547-556. doi: 10.1083/jcb.200601030
![]() |
[138] |
Tanaka N, Abe-Dohmae S, Iwamoto N, et al. (2011) HMG-CoA reductase inhibitors enhance phagocytosis by upregulating ATP-binding cassette transporter A7. Atherosclerosis 217: 407-414. doi: 10.1016/j.atherosclerosis.2011.06.031
![]() |
[139] |
Wildsmith KR, Holley M, Savage JC, et al. (2013) Evidence for impaired amyloid beta clearance in Alzheimer's disease. Alzheimers Res Ther 5: 33. doi: 10.1186/alzrt187
![]() |
[140] |
Chan SL, Kim WS, Kwok JB, et al. (2008) ATP-binding cassette transporter A7 regulates processing of amyloid precursor protein in vitro. J Neurochem 106: 793-804. doi: 10.1111/j.1471-4159.2008.05433.x
![]() |
[141] |
Kim WS, Li H, Ruberu K, et al. (2013) Deletion of Abca7 increases cerebral amyloid-beta accumulation in the J20 mouse model of Alzheimer's disease. J Neurosci 33: 4387-4394. doi: 10.1523/JNEUROSCI.4165-12.2013
![]() |
[142] |
Proitsi P, Lee SH, Lunnon K, et al. (2014) Alzheimer's disease susceptibility variants in the MS4A6A gene are associated with altered levels of MS4A6A expression in blood. Neurobiol Aging 35: 279-290. doi: 10.1016/j.neurobiolaging.2013.08.002
![]() |
[143] |
Zuccolo J, Bau J, Childs SJ, et al. (2010) Phylogenetic analysis of the MS4A and TMEM176 gene families. PLoS One 5: e9369. doi: 10.1371/journal.pone.0009369
![]() |
[144] |
Cruse G, Beaven MA, Music SC, et al. (2015) The CD20 homologue MS4A4 directs trafficking of KIT toward clathrin-independent endocytosis pathways and thus regulates receptor signaling and recycling. Mol Biol Cell 26: 1711-1727. doi: 10.1091/mbc.E14-07-1221
![]() |
[145] |
Doyle KP, Quach LN, Sole M (2015) B-lymphocyte-mediated delayed cognitive impairment following stroke. J Neurosci 35: 2133-2145. doi: 10.1523/JNEUROSCI.4098-14.2015
![]() |
[146] |
Ma J, Yu JT, Tan L (2015) MS4A Cluster in Alzheimer's Disease. Mol Neurobiol 51: 1240-1248. doi: 10.1007/s12035-014-8800-z
![]() |
[147] | Wang HF, Tan L, Hao XK, et al. (2015) Effect of EPHA1 genetic variation on cerebrospinal fluid and neuroimaging biomarkers in healthy, mild cognitive impairment and Alzheimer's disease cohorts. J Alzheimers Dis 44: 115-123. |
[148] |
Gerlai R (2001) Eph receptors and neural plasticity. Nat Rev Neurosci 2: 205-209. doi: 10.1038/35058582
![]() |
[149] | Gerlai R (2002) EphB and NMDA receptors: components of synaptic plasticity coming together. Trends Neurosci 25: 180-181. |
[150] |
Kullander K, Klein R (2002) Mechanisms and functions of Eph and ephrin signalling. Nat Rev Mol Cell Biol 3: 475-486. doi: 10.1038/nrm856
![]() |
[151] |
Coulthard MG, Morgan M, Woodruff TM, et al. (2012) Eph/Ephrin signaling in injury and inflammation. Am J Pathol 181: 1493-1503. doi: 10.1016/j.ajpath.2012.06.043
![]() |
[152] |
Ieguchi K (2015) Eph as a target in inflammation. Endocr Metab Immune Disord Drug Targets 15: 119-128. doi: 10.2174/1871530315666150316121302
![]() |
[153] |
Sakamoto A, Sugamoto Y, Tokunaga Y, et al. (2011) Expression profiling of the ephrin (EFN) and Eph receptor (EPH) family of genes in atherosclerosis-related human cells. J Int Med Res 39: 522-527. doi: 10.1177/147323001103900220
![]() |
[154] |
Viernes DR, Choi LB, Kerr WG, et al. (2014) Discovery and development of small molecule SHIP phosphatase modulators. Med Res Rev 34: 795-824. doi: 10.1002/med.21305
![]() |
[155] | Gold MJ, Hughes MR, Antignano F, et al. (2015) Lineage-specific regulation of allergic airway inflammation by the lipid phosphatase Src homology 2 domain-containing inositol 5-phosphatase (SHIP-1). J Allergy Clin Immunol. In press. |
[156] | Nowakowska BA, Obersztyn E, Szymanska K, et al. (2010) Severe mental retardation, seizures, and hypotonia due to deletions of MEF2C. Am J Med Genet B Neuropsychiatr Genet 153b: 1042-1051. |
[157] |
Xu Z, Yoshida T, Wu L, et al. (2015) Transcription factor MEF2C suppresses endothelial cell inflammation via regulation of NF-kappaB and KLF2. J Cell Physiol 230: 1310-1320. doi: 10.1002/jcp.24870
![]() |
[158] | Aisen PS, Luddy A, Durner M, et al. (1998) HLA-DR4 influences glial activity in Alzheimer's disease hippocampus. J Neurol Sci 161: 66-69. |
[159] |
Zota V, Nemirovsky A, Baron R, et al. (2009) HLA-DR alleles in amyloid beta-peptide autoimmunity: a highly immunogenic role for the DRB1*1501 allele. J Immunol 183: 3522-3530. doi: 10.4049/jimmunol.0900620
![]() |
[160] |
Mansouri L, Messalmani M, Klai S, et al. (2015) Association of HLA-DR/DQ polymorphism with Alzheimer's disease. Am J Med Sci 349: 334-337. doi: 10.1097/MAJ.0000000000000416
![]() |
[161] |
Kobrosly R, van Wijngaarden E (2010) Associations between immunologic, inflammatory, and oxidative stress markers with severity of depressive symptoms: an analysis of the 2005-2006 National Health and Nutrition Examination Survey. Neurotoxicology 31: 126-133. doi: 10.1016/j.neuro.2009.10.005
![]() |
[162] |
Na KS, Jung HY, Kim YK (2014) The role of pro-inflammatory cytokines in the neuroinflammation and neurogenesis of schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 48: 277-286. doi: 10.1016/j.pnpbp.2012.10.022
![]() |
[163] |
Heneka MT, Carson MJ, El Khoury J, et al. (2015) Neuroinflammation in Alzheimer's disease. Lancet Neurol 14: 388-405. doi: 10.1016/S1474-4422(15)70016-5
![]() |
1. | Shenglong Guo, Qi Lei, Hena Guo, Qian Yang, Yanli Xue, Ruili Chen, Edaravone Attenuates Aβ 1-42-Induced Inflammatory Damage and Ferroptosis in HT22 Cells, 2023, 48, 0364-3190, 570, 10.1007/s11064-022-03782-y | |
2. | Marta Sochocka, Breno Satler Diniz, Jerzy Leszek, Inflammatory Response in the CNS: Friend or Foe?, 2017, 54, 0893-7648, 8071, 10.1007/s12035-016-0297-1 |