Abstract
The amyloid hypothesis has dominated Alzheimer’s disease (AD) research for almost 30 years. This hypothesis hinges on the predominant clinical role of the amyloid beta (Aβ) peptide in propagating neurofibrillary tangles (NFTs) and eventual cognitive impairment in AD. Recent research in the AD field has identified the brain resident macrophages, known as microglia, and their receptors as integral regulators of both the initiation and propagation of inflammation, Aβ accumulation, neuronal-loss, and memory decline in AD. Emerging studies have also begun to reveal critical roles for distinct innate immune pathways in AD pathogenesis, which has led to great interest in harnessing the innate immune response as a therapeutic strategy to treat AD. In this review, we will highlight recent advancements in our understanding of innate immunity and inflammation in AD onset and progression. Additionally, there has been mounting evidence suggesting pivotal contributions of environmental factors and lifestyle choices in AD pathogenesis. Therefore, we will also discuss recent findings suggesting that many of these AD risk factors influence AD progression via modulation of microglia and immune responses.
Keywords: amyloid beta, Alzheimer’s disease, microglia, neurodegenerative disease, neuroimmunology, TREM2
INTRODUCTION
AD pathology:
Alzheimer’s disease (AD) is a neurodegenerative disease characterized by neuronal loss, neuroinflammation, and pronounced memory decline.1,2 Risk factors for developing this neurodegenerative disease include age, genetics, sex, brain injury, and various environmental factors.3 Regardless of origin, neuropathologies consistent among AD patients are amyloid beta (Aβ) and neurofibrillary tangle (NFT) deposition.4 Aβ can deposit in the brain decades prior to clinical symptom onset and stimulate other AD pathologies such as hyperphosphorylated tau (p-Tau) aggregation which triggers subsequent NFTs and neuronal death.5–9 These NFTs can mediate neuronal damage that propagates mild cognitive impairment (MCI) and dementia.10 Given that Aβ accumulation precedes NFT formation and memory decline by many years, the focus on Aβ production has dominated AD research.10 Aβ is a cleavage product of amyloid precursor protein (APP).11 APP cleavage can follow two pathways: amyloidogenic and nonamyloidogenic. The former has a cleavage event spurred by β-secretase and γ-secretase to produce neurotoxic Aβ, while the latter undergoes APP cleavage by α-secretase and γ-secretase to generate distinct products, some of which have important neuroprotective functions.11,12
The Aβ peptide can act as a building block to produce several distinct higher-order forms of Aβ, some of which are more neurotoxic than others.11 For instance, monomers of Aβ peptide can accumulate to form oligomers that can travel throughout the brain and constitute the soluble fraction of Aβ levels in the AD brain.11 Aβ oligomers are the most toxic form of Aβ and have been shown to incite synaptic dysfunction and neuronal cell death signaling.13 Aβ oligomers can seed Aβ fibrils to promote the formation of Aβ aggregates known as “plaques”.14 Monomers of the Aβ peptide can have varying lengths depending on their cleavage.11 However, Aβ42 and Aβ40 are the most prevalent in AD pathogenesis, with Aβ42 being more likely to incorporate into Aβ aggregates.15 AD brains are also marked by dystrophic neurites.16 Dystrophic neurites exhibit axonal transport defects and are often found surrounding Aβ plaques.4 Recent evidence suggests that dystrophic neurites can directly contribute to AD pathology propagation by promoting β-secretase-mediated cleavage of APP and subsequent Aβ peptide generation.17
Biomarkers:
The identification of biomarkers relating to AD risk and pathology prior to clinical onset has become an important area of focus in hopes of manipulating AD onset and progression. For decades, the diagnosis of AD relied on post-mortem brain histological assessment to identify Aβ and NFT pathologies. However, recent technological advancements in genetic testing, PET imaging, and sampling of the cerebrospinal fluid (CSF) and blood have opened doors to the possibility of earlier AD diagnosis. There is great hope that the identification of reliable AD biomarkers will move the field toward preventative medicine; however, continued progress on this front is needed to make this a reality.
There are two classifications of AD based on disease origin and onset: (1) early-onset AD (EOAD) (also known as familial AD or FAD) and (2) late-onset AD (LOAD). EOAD accounts for 5% of all AD cases and is primarily caused by mutations in APP, presenilin-1 (PSEN1), and presenilin-2 (PSEN2), all of which can lead to elevated levels of total Aβ and increased production of Aβ40 and Aβ42.18,19 EOAD often occurs in individuals younger than 65 years of age.20 In contrast, the origin of LOAD is linked to an interplay between genetic mutations, environment, and neuroinflammatory responses.21 Interestingly, many of the mutations linked to LOAD risk are known to affect innate immune signaling pathways (Table 1). The most common variant associated with LOAD risk is apolipoprotein E gene (APOE) ε4 allele.18 APOE has three isoforms which include ε2, ε3, and ε4, with ε4 being the allele exerting the greatest AD risk.22 In homeostasis, APOE is synthesized for lipid transport.23 However, during stress, APOE ε4 is markedly more susceptible to proteolytic cleavage to generate products that promote NFT formation and mitochondrial dysfunction, both of which can contribute to AD progression.23 In addition, APOE has been reported to promote Aβ aggregation and is often found in large quantities within Aβ plaques, further implying its role in AD pathogenesis.24,25 Patients with the APOE ε4 allele are at 3-times higher risk for developing AD.22 The combination of APOE ε4 and chronic inflammation further increases risk for these carriers, emphasizing an important link between genetics and inflammation in AD risk.26 LOAD is also associated with mutations in receptors expressed by microglia, the brain’s resident immune cells.27 Recent research shows that multiple microglial receptors are critically involved in Aβ clearance and that mutations in multiple microglial immune receptors dramatically increases the risk for LOAD.27
Table 1.
AD GWAS variants associated with innate immune signaling
Gene | Function | Variant function in AD | References |
---|---|---|---|
APOE | Lipid transport | Aβ aggregation APP transcription Tangle pathology Mitochondrial dysfunction |
23,24,301 |
TREM2 | Inflammatory response Phagocytosis Cellular metabolism |
Aβ accumulation Impaired microglial response |
65,302,303 |
CD33 | Cellular activity inhibition | Aβ accumulation | 79,82,304,305 |
CLU | Extracellular chaperone Lipid transport Immune modulation Complement |
Binding of Aβ | 306–308 |
ABCA7 | Lipid homeostasis Cell membrane transport |
APP processing regulation Aβ secretion Impaired phagocytosis |
79,309,310 |
BIN1 | Endocytosis Inflammation Calcium homeostasis |
Tau propagation Neuronal degeneration |
311,312 |
CR1 | Complement Microglial phagocytosis |
Aβ accumulation Inflammation |
306,313 |
SORL1 | Endocytosis Lipid metabolism |
APP processing Aβ accumulation |
314 |
IL-34 | Survival and differentiation of monocytes | Aβ neurotoxicity | 168,315,316 |
MS4A gene cluster | Calcium signaling Immunity |
Reduced sTREM2 production | 79,304,317,318 |
CD2AP | Cytoskeletal dynamics Endocytosis Synapse function |
Enhanced Aβ generation Tau-induced toxicity |
79,304,319 |
EPHA1 | Immunity Endocytosis |
Uncharacterized | 79,304,320 |
INPP5D | Immunity Myeloid cell regulation |
Inhibition of TREM2 signaling | 321,322 |
SHARPIN | Inflammation NF-κB activation |
Attenuated inflammatory response | 323 |
TREML2 | Microglial proliferation | Neuronal degeneration | 324 |
SPPL2A | TNF signaling | Immune cell regulation | 308,325 |
HLA-DR | Immunity Antigen presentation |
Aβ accumulation Inflammation |
308,326 |
Diagnosing AD in patients has become amenable with the use of positron emission tomography (PET) imaging of Aβ and tau in the brain.28 PET imaging is able to detect high levels of Aβ in the brains of patients with cognitive impairment, allowing for highly sensitive diagnosis of AD.29–31 In addition, the detection of tau using PET has also proven to aid in the tracking of AD progression.28,32 An early diagnosis of AD is also made possible with PET imaging of the initial inflammation of disease, which is often characterized by microglial activation.33 Biomarkers in the CSF and blood are also used to diagnose and detect AD progression.34 CSF markers include levels of Aβ and tau, with AD patients presenting with a decrease in Aβ42 and a significant increase in phosphorylated and total tau.35,36 Blood is an alternative medium to measure for AD biomarkers, and is especially attractive due to its low invasiveness and medical cost.37 Blood biomarkers for AD include decreased levels of Aβ42, a lower Aβ42:Aβ40 ratio, and increased tau.37 In addition, the level of neurofilament light chains (NfLs) is another blood marker for AD.38 Increased levels of NfLs in the blood is distinct in EOAD patients prior to symptom onset, and changes in NfL levels are often greatest when AD patients transition from the presymptomatic to symptomatic stages of AD.38 Therefore, NfLs can be used to predict AD onset and track its progression in a non-invasive manner.
Innate immune function of AD pathologies:
While numerous physiological functions have been assigned to Aβ and tau, there is still a general lack of consensus as to what roles these AD-related molecules play in homeostasis. Interestingly, it has recently been proposed that Aβ may help provide protection against pathogens by functioning as an antimicrobial peptide (AMP) (Fig. 1).39 Such studies demonstrate that Aβ oligomers are critically involved in limiting both fungal and bacterial infections in cell culture, C. Elegans, and mice.40 Additionally, bacterial infections have been shown to incite the seeding of Aβ in the 5xFAD AD mouse model.40 Aβ’s AMP function may explain why the peptide has been conserved across a breadth of species for hundreds of millions of years.41 Further implicating Aβ in the innate immune response is the finding that certain microorganisms’ cell membranes are covered with amyloid-like fibrillous structures.42 Bacteria with these amyloid structures, also known as curli fibrils, present a pattern recognized by receptors on immune cells to initiate signaling to remove these invaders.43,44 A similar response is elicited when Aβ itself is recognized by microglial pattern recognition receptors (PRRs) in the brain.45 Unfortunately, chronic innate immune activation augmented by sustained Aβ recognition provokes subsequent inflammation that can lead to further propagation of AD pathologies and neurodegeneration.2,46
Figure 1. The antimicrobial protection hypothesis of amyloid beta and Alzheimer’s disease.
The cleavage of the amyloid precursor protein (APP) can produce amyloid beta (Aβ) in its monomeric form. The oligomerization of Aβ is thought to help protect the brain against CNS-invading microbes. Aβ oligomers are believed to limit pathogen spread in the brain by promoting the production of reactive oxygen species (ROS) and also by physically encapsulating microbes to limit invasion of host cells. In line with this reasoning, many different microbes have been found to associate with Aβ upon examination of post-mortem AD brains. While the antimicrobial functions of Aβ can potentially play beneficial roles, this process must be tightly controlled as the formation of Aβ aggregates in response to infection can incite the spread of pathological Aβ plaques.
Although the explicit link between Aβ-seeding and tau accumulation has remained elusive, recent research has begun to shed light on this topic.5 For instance, it has been demonstrated that Aβ-induced dystrophic neurites accumulate endogenous tau that eventually form into neurofibrillary tangles that correlate with cognitive deficits.5 In homeostasis, endogenous tau participates in the assemblage and stabilization of microtubules in neurons, and, as a result, tau is believed to play functionally important roles in neuronal transport.47 However, the detachment of tau from microtubules and hyperphosphorylation of tau can incite aberrant microglial activation and proinflammatory cytokine production.48 This feedback drives further production of pathogenic tau to enter a cycle that perpetuates its overproduction and neurotoxicity.48 Interestingly, herpes simplex virus (HSV-1), which infects two-thirds of the global population, has been shown to phosphorylate tau.49,50 In accordance with this finding, antiviral medication has been reported to decrease phosphorylated tau accumulation and Aβ load.51 A clinical trial is currently investigating the efficacy of the antiviral drug valacyclovir for AD patients with HSV-1 or HSV-2.52 This further suggests a link between both neuropathologies of AD and the innate immune response that requires further exploration.
The role of microglia in homeostasis and AD:
As current AD research acknowledges the role of the innate immune response and neuroinflammation in driving neurodegenerative disease, microglia have taken center stage.53 Mounting research suggests that LOAD is, at least in part, mediated by the failure of microglia to properly clear Aβ.54 In fact, frequent variants identified with genome-wide association study (GWAS) in AD patients link mutations in microglia pattern recognition receptors (PRRs) with disease risk.55 Despite microglia being integral for proper phagocytosis and degradation of Aβ, their chronic activation can also provoke maladaptive neuroinflammatory responses with the potential to propagate Aβ production and neuronal distress.56 This has sparked controversy over the enigmatic roles of microglia in AD56–58, which will be discussed in greater detail throughout this review.
During development, microglia arise from progenitor cells originating in the yolk sac before populating the brain.59 In homeostasis, microglia have been shown to undergo self-renewal to maintain their numbers in the central nervous system (CNS).59 Microglia act as innate immune sentinels of the brain parenchyma, and perform classical macrophage functions such as coordinating phagocytic disposal of debris and inflammatory cytokine production.60 However, microglia have additional functions in neurogenesis, blood-brain barrier (BBB) permeability, vasculogenesis, myelination, synapse pruning, and neuronal connectivity.60 Microglial morphology transforms during activation, going from a highly branched or “ramified” state during homeostasis to an “ameboid” activated state in response to infection, injury, protein aggregate deposition, and other overt activators of innate immune signaling.61 Microglia express a repertoire of cell surface receptors known as PRRs that enable them to sense and respond to a diverse array of immunostimulatory triggers in the CNS.62 Microglia associated with disease are commonly found to be in an activated state and upregulate PRR expression.63 Accordingly, microglia are capable of sensing endogenous damage/danger-associated molecular patterns (DAMPs) as well as exogenous pathogen-associated molecular patterns (PAMPs).62 Microglia can also sense neurodegeneration-associated molecular patterns (NAMPs) that are generated in AD and other forms of neurodegenerative disease.64 In turn, the heterogeneity of microglia populations based on activation state and cell surface receptor expression has piqued interest in their distinct roles in health and disease.
Microglia receptors implicated in AD:
TREM2 -
One of the best-characterized microglia PRRs involved in AD is triggering receptor expressed on myeloid cells 2 (TREM2) (Fig. 2). TREM2 engages with DAP12 as an adaptor protein, and upon TREM2 activation, the phosphorylation of the DAP12 immunoreceptor tyrosine-based activation motif (ITAM) is stimulated, leading to the recruitment of spleen tyrosine kinase (SYK) to these sites.65 Engagement of the TREM2 receptor is believed to stimulate pathways responsible for cytoskeletal reorganization, myeloid cell survival, phagocytosis, and the production of pro-inflammatory cytokines.65 The rare Arginine-47-Histidine (R47H) mutation of TREM2 significantly increases LOAD risk.66,67 This R47H mutation is thought to contribute to AD risk by limiting proper TREM2-mediated sensing of lipids by microglia.68 TREM2 recognizes Aβ, lipids released from damaged neurons, and the Aβ chaperone APOE in AD.68–70 Activation of TREM/DAP12 signaling limits Aβ accumulation and other harmful forms of AD pathology, as well as modulates cytokine production.65 Therefore, disruption of TREM2/DAP12 signaling can markedly contribute to the progression of Aβ pathology in AD.65 An additional mechanism by which TREM2 limits Aβ pathology is by promoting plaque compaction.71 Plaque morphology is dynamically regulated by microglia, with less-compacted and fibrillous plaques being more neurotoxic than compact plaques.72 Microglia function as a physical barrier to protect neurons from the neurotoxicity of Aβ plaques and curtail further Aβ seeding.72,73 Such studies suggest that the loss of Trem2 function impairs microglial sensing of Aβ, leading to defective Aβ clearance, increased Aβ plaque toxicity, and neuronal dystrophy in 5xFAD mice.68,71
Figure 2. Microglial surface receptors that coordinate innate immune responses in AD.
TREM2 recognizes Aβ, lipids released from damaged neurons, and the Aβ chaperone APOE in AD. TREM2 activation critically contributes to Aβ clearance and also regulates downstream cytokine production. Aβ activation of scavenger and Fc receptors (FcRs) coordinates microglial-mediated Aβ clearance. In contrast, CD33 and CD22 bind sialic acid that decorates Aβ plaques, and inhibit downstream signaling events that are required for beneficial microglial responses and Aβ clearance. RAGE, Clec7a, and TLRs are also known to modulate microglial cytokine production in response to Aβ and DAMPs associated with AD.
Patients with the TREM2 R47H mutation and mice with Trem2 deficient microglia also present with enhanced autophagy stemming from the dysregulation of the mammalian target of rapamycin (mTOR) signaling.74 This amplification of autophagy impairs microglial response to Aβ and suggests that TREM2 also regulates AD pathology through the maintenance of proper microglial metabolism.74 Recent studies also show that genetic ablation of TREM2 in APPPS1 AD mice (APPPS1;Trem2−/−) and loss-of-function TREM2 mutations in humans both result in reduced association of APOE with Aβ plaques.75 As a result, these APPPS1;Trem2−/− mice have increased seeding of Aβ compared with APPPS1 controls.75 Microglia depletion was also found to significantly reduce APOE levels that were associated with Aβ plaques, which further implicates a TREM2-APOE pathway in microglia.75 Interestingly, crosstalk between TREM2 and APOE has also been reported to promote a transcriptional shift in microglia during AD, which can further propagate neuronal loss.76
Recent studies also suggest the proteolytic cleavage of TREM2 and the subsequent secretion of soluble TREM2 (sTREM2) can ameliorate AD-related disease progression in mouse models. sTREM2 is found at higher levels in the CSF of AD patients, and as such, has been proposed as a potential future biomarker for disease.77 The supplementation of sTREM2 into the brain of 5xFAD mice influences microglia signaling to improve Aβ clearance preventing further neuritic dystrophy while promoting microglia proliferation and migration.78 Continuation of sTREM2 supplementation, using an adeno-associated virus (AAV) mediated expression technique, was also found to rescue memory and long-term potentiation (LTP) deficits seen in the 5xFAD mice.78 Overall, these studies implicate beneficial roles for TREM2 in the context of proper Aβ sensing and clearance.
CD33 & CD22 -
Microglial sialic-acid binding immunoglobulin-like lectin (Siglec) receptors have also been shown to play important roles in AD and aging. For instance, CD33, also known as Siglec-3, is another microglial receptor implicated by GWAS in AD risk.79 CD33 binds the sialic acids found ubiquitously on cell membranes.80,81 In addition, sialic acids are also decorated on the glycoproteins and glycolipids found in Aβ plaques, and can, therefore, stimulate CD33 signaling in the AD brain.80 However, in contrast to TREM2 which signals utilizing ITAMs, CD33 has intracellular immunoreceptor tyrosine-based inhibitory motifs (ITIMs).80 The phosphorylation of ITIMs promotes the docking of Src homology 2 domain-containing phosphatases (SHPs) to inhibit downstream microglial signaling mediated by ITAMs that would have otherwise led to the removal of Aβ in the brain parenchyma.80 The CD33 rs3865444C AD risk allele leads to increased CD33 receptor expression which results in impaired Aβ42 phagocytosis.82 Increased expression of CD33 is significantly associated with a higher clinical score in AD patients and elevated AD pathology.83,84 Further implicating CD33 in AD risk, 5xFAD mice lacking Cd33 (5xFAD;Cd33−/−) have decreased Aβ burden and improved memory compared with Cd33 sufficient 5xFAD mice.85
More recent studies demonstrate that like CD33, CD22 (also known as Siglec-2), can also hinder microglial phagocytosis in aging and models of AD.86 For instance, Cd22 is found upregulated in aged microglia, and antibody-mediated blockade was shown to increase Aβ clearance.86 In addition, microglia in aged mice take on an age-related transcriptional profile that under continued inhibition of Cd22, is reverted to a more homeostatic state.86 The inhibition of Cd22 also ameliorates age-related cognitive decline.86 In summary, both CD33 and CD22 are negative regulators of microglial phagocytic functions in AD that have emerged as attractive therapeutic targets.
CLEC7A -
Recent studies have shown that the receptor CLEC7A, also known as Dectin-1, is dynamically upregulated by microglia in AD, particularly in microglia in intimate contact with Aβ plaques.76 CLEC7A has been best described in fungal infections where it has been shown to coordinate cytokine production and phagocytosis.87 Similar to TREM2, CLEC7A signals through its intracellular ITAM.87 Although the exact role of CLEC7A in AD still remains to be determined, emerging evidence indicates that changes in microglial Clec7a expression are associated with AD progression.76 In fact, increased microglial Clec7a expression is a hallmark of numerous neurodegenerative disorders and aging.63 Interestingly, multiple human studies have begun to link fungal infections with AD. This work has identified that fungi can be detected in the brains of AD patients.88–90 Levels of chitinase, the enzyme that breaks down chitin found in fungal cell walls, have also been reported to be elevated in the CSF of AD patients.91 This has led to growing speculation that during CNS fungal infections, neuroinflammatory responses and Aβ production are mounted to protect the brain from the fungal pathogens.92 However, the downside of this is that it may inadvertently set AD pathology in motion. The essential role that CLEC7A is known to play in numerous fungal infections coupled with the pronounced upregulation of microglial CLEC7A expression observed in AD progression, points toward a potentially important role for CLEC7A in AD. However, additional studies probing the specific involvement of CLEC7a in AD pathogenesis are needed.
RAGE –
The binding of Aβ to the microglial receptor of advanced glycosylation end products (RAGE) stimulates NF-kB activation resulting in increased production of pro-inflammatory cytokines.93 RAGE is a PRR found on many immune cells that can be upregulated in response to increased ligand production during injury, aging, and neurodegeneration.94,95 In addition to being a potent inducer of NF-kB signaling and downstream inflammation, RAGE has also been shown to promote microglial survival and proliferation in a MAPK-dependent fashion.95,96 In the context of neurodegenerative disease, overexpression of RAGE in the mAPP AD mouse model triggers IL-1β and TNF-α production.97 RAGE has also been reported to instigate Aβ accumulation in the brain by aiding in the transport of Aβ across the BBB and into the brain.98 Additionally, RAGE signaling in microglia can incite Aβ accumulation and amplify memory deficits.97 The exaggerated inflammation incited by this Aβ and RAGE interaction can compromise neuronal health and in turn, negatively affect memory in AD mouse models.93 Despite increased RAGE expression found in the brains of AD patients, clinical trials targeting RAGE have yielded underwhelming results.99,100
Complement associated receptors: CR3 and C1qR -
CR3 and C1qR are microglial receptors that can coordinate deleterious synapse pruning in AD.101 The complement receptors CR3 and C1qR are activated by the complement cascade proteins C3 and C1q, respectively.101 In homeostatic neurodevelopment, C3 and C1q contribute to proper synapse pruning and are required to sculpt healthy synaptic connections.102 In contrast, reactivation of C3 and C1q signaling during aging is believed to cause detrimental synapse loss in the hippocampus, the brain structure critical for learning and memory.103,104 In fact, aged C3-deficient mice exhibit decreased synapse loss, reduced neuronal loss, and improved memory compared with age-matched WT controls.103 In the context of AD, genetic ablation of C3 in APPPS1 AD mice results in reduced synapse loss, lower numbers of microglia interacting with Aβ plaques, and improved behavior despite increased Aβ load.105 However, CR3 deficiency in APP AD mice has been shown to limit Aβ levels by a mechanism involving increased expression of proteolytic enzymes that are known to breakdown Aβ in the extracellular compartment.106 In turn, the presence of C3 or CR3 in WT mice actually enhances Aβ42 fibrillar phagocytosis.107 These findings suggest enigmatic roles for CR3 in modulating the AD brain environment, which are likely impacted by the kinetics and model of disease.
Recent studies demonstrate that microglial-derived C1q is typically found to be elevated in the hippocampus of aged mouse brains and AD human brains.104,108,109 C1q tags synapses that are meant to be eliminated by microglia.110 However, C1q-mediated synaptic tagging and subsequent microglial clearance can result in synapse loss in the hippocampus, specifically to the engram cell synapses, which are known to be involved in memory storage.110 Interestingly, the deletion of microglia, impairments in microglial phagocytosis, and inhibition of the complement pathway have all been shown to block this synapse loss and preserve memory in mice.110 Collectively, this illustrates that complement signaling regulated by microglia is needed to ensure proper synapse formation in neurodevelopment. However, unchecked activation of this pathway can propagate aberrant synapse loss and contribute to subsequent aging-induced memory decline and AD-associated phenotypes in mouse models.
Microglial clearance of Aβ and tau:
Clearance of AD pathology by microglia is executed by several discrete processes. These include receptor-mediated phagocytosis and endocytosis, expression of degradative factors, extracellular chaperone-mediated clearance, and induction of autophagy.111 Each mechanism allows for homeostatic maintenance of the brain parenchyma, and can potentially be targeted to ameliorate Aβ and tau accumulation in disease.
Clearance by phagocytosis and endocytosis –
Microglial phagocytosis and endocytosis is a critical method of Aβ clearance, and as a result, there has been tremendous interest in targeting these processes to treat AD. Receptors that mediate this form of clearance include TREM2, toll-like receptors (TLRs), scavenger receptors, and Fc receptors (FcRs) (Fig. 2).111 In recent years, TREM2 has emerged as a major mediator of Aβ endocytosis. Details regarding how TREM2 mechanistically coordinates endocytic clearance of Aβ can be found earlier in the review. TLRs are most highly expressed by microglia in the brain. Microglial TLRs have been reported to bind oligomeric and fibrillar forms Aβ and this is thought to initiate a signaling cascade that promotes subsequent phagocytic clearance of Aβ.112 Consistent with this idea, genetic deletion of TLR2 and TLR4 in transgenic mouse models of AD has been shown to promote increased Aβ deposition and this is generally believed to result from deficits in phagocytosis.113,114 TLR co-receptors including the TLR4 coreceptor and LPS detector CD14 have also been implicated in AD.115 In a similar fashion to TLRs, CD14 is thought to orchestrate the phagocytosis of fibrillar Aβ.116 The endocytosis of Aβ in both fibrillar and oligomeric forms can also be stimulated by class A and class B scavenger receptors. For example, the scavenger receptors CD40 and CD36 have been described to initiate Aβ endocytosis.111
Lastly, Aβ plaques that have been bound by either host-derived or engineered anti-Aβ antibodies can be phagocytosed following engagement of FcRs on microglia.117 Engineered anti-Aβ antibodies and vaccine strategies that elicit robust anti-Aβ antibody production have been shown to promote impressive clearance of Aβ from the brains of Aβ-overexpressing AD mouse models.118 FcR-mediated phagocytosis of Aβ complexes coated with antibodies is thought to be a major mechanism promoting Aβ reduction following anti-Aβ antibody infusions and anti-Aβ vaccination.118 Unfortunately, recent clinical trials in AD patients testing antibody-mediated Aβ reducing strategies have not been promising.119 Current thinking is that anti-Aβ antibodies were introduced too late in the disease process to protect neurons that were either already lost or well on their way to neuronal demise. Despite this setback, hope still remains that earlier interventions with anti-Aβ antibodies may still hold clinical promise. However, for this to become a reality, improved AD diagnostic biomarkers to identify high-risk patients at earlier stages of disease are required.
Clearance by degradative factors –
In addition to phagocytic pathways, microglia are able to clear AD pathology through their production of degradative factors. These mechanisms include the use of insulin degrading enzyme (IDE), neprilysin, endothelin degrading enzyme (EDE), and the proteasomal degradation pathway.111 IDE, neprilysin, and ECE are all metalloendopeptidases known to degrade Aβ.111 IDE can be produced by microglia and secreted into the extracellular space to degrade monomeric Aβ.120 Interestingly, there is a link between taking statins, which lower cholesterol, and a decreased risk for AD.121 One mechanism by which statins are believed to deter AD development is via their promotion of microglial IDE production to degrade Aβ.121 Neprilysin is another important enzyme in the breakdown of Aβ.122 In fact, an age-related neprilysin decline in AD brains is thought to contribute to the increased levels of Aβ that occurs with aging.122 In line with this hypothesis, areas of the brain with high Aβ plaque load, such as the hippocampus, are known to have decreased neprilysin levels.123 In addition, genetic ablation of neprilysin in APP transgenic AD mice has been reported to cause increased Aβ deposition in the brain and more severe cognitive decline.124 Similar to IDE and neprilysin, ECE can also promote Aβ degradation and ECE-deficient mice have increased Aβ burden in the brain.125 Finally, the proteasomal degradation of AD pathology is regulated by the ubiquitin proteasome system (UPS).126 This system tags mis-folded proteins with ubiquitin to mark them for proteasomal degradation.36 In AD, this process is believed to be impaired, which in turn leads to increased retention of tau and Aβ aggregates in the brain.36
Extracellular chaperone-mediated clearance –
Extracellular chaperone-mediated clearance is another mechanism by which Aβ can be cleared from the brain. It has been shown that APOE binds to Aβ and acts as a chaperone to promote its clearance via endosomes.127 The chaperone function of APOE is thought to prompt microglial response to Aβ plaques, driving Aβ compaction and in turn, reducing neuronal damage.128 Interestingly, an APOE-TREM2 pathway has emerged in which TREM2 function is shown to be critical for the APOE association with Aβ plaques to promote plaque compaction.75 The binding of APOE and Aβ to microglial lipoprotein receptor-related protein 1(LRP-1) receptor is also thought to initiate signaling through JNK and NF-kB pathways to promote pro-inflammatory cytokine production.129 The AD risk variant, APOE ε4, is characterized to be less effective at these chaperone-mediated Aβ clearance duties in addition to inciting an increase in neuroinflammation.127,130
Autophagic clearance -
Autophagy acts as an additional mechanism for the clearance of Aβ. Autophagy is a lysosome-dependent, homeostatic process, whereby organelles and proteins such as Aβ are broken down and recycled.131 Interestingly, autophagy is linked with many aspects of the innate immune response, including the regulation of pro-inflammatory cytokine production.131 Fibrillar Aβ can be cleared by microglial autophagy, and the impairment of this process has been shown to propagate NLRP3 inflammasome activation.132 In addition, treatment with trehalose to enhance autophagy markedly decreases tau aggregation in cell culture settings and can reduce neuronal tau inclusions in a tauopathy mouse model.133,134 Dystrophic neurites are also known to accumulate autophagic vesicles, further supporting a potential role for autophagy in regulating neuronal health.135 In line with a beneficial role for autophagy in AD, boosting autophagic signaling has been shown to ameliorate memory deficits in AD mice.136 Therefore, modulating autophagy presents another potential therapeutic avenue for the treatment of AD that will surely be intensely pursued in future studies.
Disease associated microglia:
The dispute surrounding whether microglia exert beneficial or detrimental roles in AD remains a hotly debated topic. Single-cell RNA-seq recently identified a new subset of microglia known as “disease associated microglia” (DAM).63 In the past, microglia were largely thought of as a homogenous cell population. However, this diversified transcriptional profile of microglia in the AD and aging brain environment has prompted many new questions surrounding their role in disease onset and progression. DAMs are classified by two distinct stages: the initial TREM2-independent Stage 1 and TREM2-dependent Stage 2.63 Stage 1 and 2 DAMs are identified by their downregulation of characteristic homeostatic microglial genes (i.e. P2ry12, Cx3cr1, and Tmem119). Stage 1 DAMs upregulate phagocytic related genes such as Apoe and Dap12 while Stage 2 DAMs upregulate Clec7A and Trem2.63 As previously discussed in this review, APOE variants are a major risk factor for LOAD, while TREM2, DAP12, and CLEC7A are receptors previously described in phagocytic pathways.23,65,87 Initiation of this shift of microglia from a homeostatic phenotype to the DAM signature is believed to be dependent on sensing of NAMPs, engagement of TREM2 signaling, and extended interactions of microglia with Aβ plaques.64
The DAM subtype is accepted to serve in a beneficial capacity through their coordination of Aβ clearance.63 However, this function remains strictly regulated by the inhibitory signaling of CX3CR1, perhaps acting as a harness on DAM function.63 Therefore, future research efforts will likely focus on stimulating the DAM phenotype early on in disease to reduce AD progression. However, the possibility also exists that over-excessive and/or chronic activation of the DAM phenotype may trigger neurotoxic consequences and propagate AD pathology. This possibility will likely encourage a new AD-microglia field focused on determining how to revert DAM back to their homeostatic microglial state. The multi-faceted roles of microglia underscore the temporal complexities of their function, and will likely keep them in the AD research spotlight for years to come.
Loss of microglial homeostatic function in AD:
Homeostatic microglia possess a signature defined by the expression of receptors such as CX3 chemokine receptor 1 (CX3CR1) and purinergic receptor P2RY12.27 CX3CR1, which is activated in response to its ligand fractalkine, performs inhibitory duties to sustain a resting-state microglia population.137 Although fractalkine is found at higher levels in the blood of AD patients, Cx3cr1 is downregulated in DAMs.63,138 CX3CR1 is thought to have complex roles in AD. For example, deficiency in Cx3cr1 reduces neuronal loss in the 3xTgAD mouse model, which develop both Aβ plaques and NFTs.139 In contrast, the loss of Cx3cr1 in the Aβ-mediated hAPP-J20 AD mouse model spurs microglial toxicity and intensifies cognitive deficits.140 The presence of Aβ also downregulates fractalkine in the hAPP-J20 AD mouse model.140 These findings suggest that the role of CX3CR1 is variable depending on the different models of AD being used. Despite these inconsistencies, CX3CR1 appears to be a critical modulator of DAM biology. However, without this CXC3R1 check and balance, excessive microglia activation may contribute to neuronal loss and functional decline, while its overabundance may act to inhibit necessary DAM response to AD pathology. Purinergic receptor P2RY12, on the other hand, is important for microglial migration, and it is highly expressed in homeostatic microglia.141,142 However, the role of P2RY12 downregulation in plaque-associated DAMs and AD remains unclear and requires further investigation.
Impact of microglial elimination in AD:
The often-contradictory roles of microglia in varying states and stages of neurodegenerative disease have prompted researchers to explore the impact of microglia depletion in AD. In recent years, treatment with the colony stimulating factor 1 receptor (CSF-1R) inhibitor, Plexxikon, has emerged as a popular approach to study the functional role of microglia in vivo. CSF-1R signaling is required for the survival of microglia and other peripheral macrophages, and as a result, Plexxikon treatment results in rapid and robust depletion of microglia from the brain.143 Results from recent studies using Plexxikon indicate a complex role for microglia in mouse models of AD that is greatly influenced by the timing and efficacy of depletion as well as the AD mouse model studied. For example, when 3xTgAD mice were depleted of 30% of their microglia, and aged to have existing Aβ plaques and tau aggregation, the mice were shown to have improved cognition but this partial deletion of microglia had no effect on Aβ plaque load or tau aggregation.144 In comparison, a 90% microglial depletion in PS19 AD mice, aged to have tau aggregation, led to significant reductions in tau propagation.145 Meanwhile, partial depletion of microglia in the APPPS1 mouse model of AD was found to improve memory and ameliorate synaptic degeneration, while having no effect on Aβ load.146 5xFAD mice depleted of 80% of their microglia similarly showed improved memory and decreased neuronal damage without modulation of Aβ levels.147 However, 5xFAD mice depleted of greater than 97% of their microglia prior to Aβ plaque seeding-onset (i.e. Plexxikon treatment beginning at 1.5 months of age) exhibited reductions in Aβ load as well as a rescue of anxiety-related behaviors.148 However, the almost complete depletion of microglia in these studies was also found to promote increased numbers of dystrophic neurites, as microglia were no longer present to protect surrounding neurons from the neurotoxicity of the few remaining Aβ plaques.148 Taken together, current data in mice indicates that partial depletion of microglia does not affect Aβ plaque and tau load but can result in improved memory and less neuronal damage compared to controls.144,146,147 However, near-complete depletion of microglia amid or prior to AD pathology onset attenuates Aβ and tau load.145,148 While these studies all point to critical roles of microglia in AD, they also highlight that microglia-targeting therapeutics must carefully take into account the status of disease and desired microglia depletion efficacy to maximize beneficial clinical outcomes and safety.
AD immune cells: microglia or macrophages?
These intriguing findings from Plexxikon depletion studies have prompted speculation over whether microglia or peripheral monocytes repopulate this niche when the depletion drug is removed. As mentioned above, CSF-1R inhibition with Plexxikon treatment can deplete microglia, peripheral macrophages in other tissues, and potentially other cell types reliant on CSF-1R signaling for survival.143 This has led to speculation over the discrete roles of microglia versus other peripherally derived cell types in AD. Plexxikon microglial depletion treatment by itself does not compromise the BBB, and expansion of the few remaining microglia is thought to repopulate the brain following cessation of Plexxikon treatment.149,150 However, injury models, such as traumatic brain injury (TBI), can cause BBB breakdown and subsequent infiltration of immune cells into the brain.151,152 Likewise, AD pathology is also thought to propagate the loss of BBB integrity.153 Microglial depletion and repopulation after TBI has been reported to replace chronically activated microglia with resting-state myeloid cells, resulting in decreased neuroinflammation and neuronal loss.151 The neuroprotection conferred by the repopulated myeloid cells post-TBI relies on their induction of IL-6 trans-signaling to enhance learning and memory.154 Therefore, the replacement of activated microglia with more quiescent myeloid cells may underlie neuroprotection. However, while it is speculated that depleted microglia die and are cleared away following Plexxikon treatment, the identity of the repopulating cells in BBB-compromised settings remains poorly understood.149 Multiple myeloid cell types have been proposed to fill this niche including peripheral macrophages and microglia from the small Plexxikon-resistant subset. However, future studies are needed to better understand this process and how it affects neurological disease. This further illuminates the complexity and heterogeneity of CNS macrophages, and maintains speculation surrounding their capacity to be manipulated during neurodegenerative disease.
Initial studies exploring the ability of CNS-infiltrating monocytes to influence AD-related disease in the APPPS1 AD mouse model initially showed that chemokine receptor CCR2 deficiency prevents monocytes from entering the brain and that this results in fewer IBA-1+ myeloid cells in the brain and impaired Aβ clearance.155 These data suggest that peripheral monocytes are important for properly controlling AD pathology. In contrast, CCR2 deficiency in the same mouse model was later shown by a different lab to increase plaque-associated microglia numbers and have minimal impact on Aβ accumulation.156 What underlies the discrepancies between these two studies remains to be fully resolved. One possibility explaining the divergent outcomes may be related to the latter study’s use of irradiation, which can encourage the recruitment and retention of peripherally-derived monocytes in the brain.157 Notably, recent research utilizing an irradiation-independent model has established brain engrafting macrophages that maintain a distinct transcriptional profile from microglia and are able to repopulate the brain following partial microglia depletion.158 Other work also demonstrates that TREM2 can promote peripheral monocytes to enter the brain in APPPS1 mice.159 In contrast to the belief that peripheral myeloid cells are critical for proper AD control, parabiosis studies with WT and 5xFAD mice show minimal infiltration of peripheral monocytes from WT mice into 5xFAD brains.71 Likewise, these parabiosis findings have also been confirmed in the APP/PS1 mouse model.71 In addition, recent research using tdTomato lineage tracing in 5xFAD mice convincingly shows that CX3CR1+ microglia comprise virtually all of the myeloid cells surrounding the Aβ plaques, with virtually no CCR2+ peripheral monocytes found in the vicinity of Aβ in the brain.57 In terms of Aβ aggregation mouse models, which include 5xFAD and APPPS1 transgenic mice, microglia appear to be the key myeloid cell type responding to Aβ, with little assistance from infiltrating monocytes. However, this work cannot exclude the potential of peripheral monocyte assistance under all conditions associated with human neurodegenerative disease, especially those that are not fully recapitulated in mouse models. In particular, it will be important for future studies exploring the roles of discrete myeloid cell lineages in AD to assess how combined Aβ and tau pathology can influence outcomes.
Inflammatory mediation of AD:
Neuroinflammation centrally contributes to various aspects of AD pathologies and this has led to increasing interest in harnessing inflammatory cytokine signaling to treat AD.2,160 The role of inflammation in AD remains a hotly debated topic in the field. While on the one hand, chronic inflammation can negatively impact neuronal health, an ever-increasing body of literature also demonstrates that controlled activation of specific inflammatory pathways can help to mobilize beneficial immune functions to promote Aβ clearance and neurotrophic factor production.161,162 In this section, we will discuss the double-edged nature of inflammation in AD.
Cytokines in AD:
Multiple innate immune cytokines including type I interferon (IFN), TNF-α, IL-18, IL-1β, IL-33, IL-34, IL-12, IL-23, and IL-10 have been described to play important roles in various aspects of AD pathogenesis.163–170 Resting levels of pro-inflammatory cytokines are found to be higher in the blood and cerebrospinal fluid (CSF) of AD patients, suggesting a role for them in disease pathogenesis.171 The deposition of Aβ, and resultant neuronal damage, provide ligands to activate microglial phagocytosis of Aβ while also promoting microglia-mediated production of proinflammatory cytokines.58 Acutely, this process can lead to the proper clearance of Aβ allowing the brain parenchyma to restore homeostasis (Fig. 3).172 However, these inflammatory mediators can have complex effects in AD. For example, under chronic inflammation, this process can lose its effectiveness in clearing amyloid beta, while continuing its inflammatory response to further propagate neurotoxicity in a feed-forward mechanism (Fig. 3).173 Catalysts for this continued and detrimental response include factors such as age, genetics, injury, and peripheral inflammation.2 The production of specific pro-inflammatory cytokines, such as IFN-γ, also has the potential to encourage further production of Aβ as well as to recruit microglia activation to respond to neurotoxic Aβ species.174 Therefore, cytokine production can promote a cycle that is difficult to resolve.
Figure 3. The evolving functions of microglia in homeostasis and disease.
In homeostasis, acute exposure of microglia to Aβ initiates a temporary cytokine response that promotes Aβ clearance. Following proper disposal of Aβ, microglia typically revert back to a homeostatic resting state. However, this exposure can also prime microglia, and upon secondary or chronic exposure to stress, such as inflammation, this can lead the activated microglia to initiate neurotoxic mechanisms that provoke neuronal loss and excessive synaptic pruning. Upon aging or chronic exposure to Aβ, as microglia witness in AD, excessive levels of cytokines are produced which can impair proper microglial clearance of Aβ. Deposition of Aβ can promote NLRP3 inflammasome activation, which further perpetuates inflammatory cytokine production and subsequent deficits in Aβ clearance. In both aging and AD, microglial populations are known to take on a transcriptional signature known as “disease associated microglia” or DAM. This change is distinguished by microglial downregulation of homeostatic genes and an upregulation of genes associated with increased microglial activation and inflammatory responses. The role of DAMs in the AD brain are thought to be enigmatic in nature. For instance, DAMs are neurotoxic and lead to neuronal damage and synapse loss. However, DAMs are also able to promote Aβ clearance.
As was alluded to above, IFN cytokines play significant roles in shaping microglial responses in AD and aging.163,175 Several AD mouse models show a distinct IFN-stimulated gene (ISG) signature.176 IFN has been described to be expressed by immune cells sensing nucleic acids.177 Of note, fibrillar amyloid beta often contains nucleic acids that can elicit an inflammatory ISG signature in microglia.176 This neurotoxic type of microgliosis has been shown to promote complement-mediated synapse loss that can be curtailed with IFN-receptor blockage.176 Researchers have also shown that knocking out IFN-γ receptor type I in transgenic APP Swedish mutant AD mice leads to a reduction in Aβ plaque load and reactive microglia.174 In addition, in vitro studies indicate that IFN-γ promotes microglial release of TNF-α, and both of these cytokines promote β-secretase cleavage of APP to produce more Aβ.174 These results strongly suggest that IFN is associated with AD propagation.
The role of TNF-α in AD has remained enigmatic, as some research has focused on the therapeutic benefit of its inhibition, while others show beneficial or benign roles for the cytokine. In support of a detrimental role for TNF-α in AD, it has been shown that higher CSF levels of TNF-α correlate with impaired “functional connectivity,” a measure of the communication between different brain regions underlying various functions such as learning, memory, inhibition, etc.178 In line with pathologic functions of TNF-α in AD, APPPS1 mice deficient in TNF receptor 1 (TNFR1) have been described to have decreased levels of Aβ, lower expression of inflammatory factors, improved maintenance of choroid plexus tissue and CSF-blood barrier integrity, and preserved memory compared to TNFR1 sufficient APPPS1 mice.179 In contrast, an injection of murine TNF-α AAV into the hippocampus of APP transgenic mice has been reported to attenuate Aβ deposition and incite increased microglial responses without increasing APP levels.180 This suggests that under certain spatial and temporal contexts TNF-α may promote plaque clearance. Finally, the proposed causal relationship between TNF-α and AD has recently come into question, as Mendelian randomization modeling of GWAS studies show no causality between peripheral TNF-α expression levels and the risk of developing AD.181
IL-1 family cytokines, such as pro-inflammatory IL-18, IL-1β, and IL-33, have also been implicated in modulating AD pathogenesis.167,182,183 For example, IL-18 promotes β-secretase mediated cleavage of APP to produce more Aβ40.184 However, loss of IL-18 in the APPPS1 AD mouse model was found to incite seizure through a proposed mechanism that involves excessive excitatory synapse activity and impaired dendritic pruning.185 IL-1β has conflicting roles in AD pathologies. Increased peripheral levels of IL-1β and its propensity to incite neuroinflammation have led scientists to focus on neutralizing its functions in hopes of therapeutic benefit in AD.171,186 For instance, IL-1 receptor inhibition in 3xTgAD mice was reported to rescue cognitive deficits, decrease tau phosphorylation, and modestly modulate Aβ levels.186 Interestingly, conditional overexpression of IL-1β in 3xTgAD mice was conversely found to markedly decrease Aβ levels while concomitantly leading to increased tau phosphorylation, further highlighting the complex role of neuroinflammation in AD.187 Increased IL-1 production is also known to promote S100B protein release from astrocytes.188 The S100B protein is a neurotrophic cytokine that often acts as a pro-inflammatory cytokine in AD.189 Higher levels of S100B have been documented in AD patients and appears to be associated with increased Aβ plaque load and gliosis.190–192
IL-33, an alarmin in the IL-1 cytokine family, has also been implicated in AD. IL-33 is expressed by oligodendrocytes and its receptor, ST2, is often expressed by microglia.193 IL-33 expression is reduced in the brains of AD patients, identifying this cytokine as a potential modulator of disease.194 Accordingly, intraperitoneal injection of IL-33 was recently shown to restore LTP deficiency, improve contextual memory, and decrease Aβ levels in APPPS1 AD mice.167 IL-33 treatment accomplishes this rescue by increasing microglial Aβ phagocytosis and anti-inflammatory gene expression, while also decreasing microglial pro-inflammatory gene expression.167
IL-34 and CSF-1 activate the CSF-1 receptor (CSF-1R) to promote microglial population survival and maintenance.149 Interestingly, AD patients have been shown to have increased expression of IL-34 in the white matter compared with age-matched controls, highlighting its importance in regulating AD pathogenesis.195 More recent work, however, shows reduced expression of IL-34 in the inferior temporal gyrus (ITG) and an increase in expression of CSF-1 in the ITG and middle temporal gyrus (MTG) of AD patients.196 On one hand, research has found neuroprotective roles for IL-34 in AD. For instance, intracerebroventricular injection of IL-34 into APPPS1 mice promotes improved memory and lower levels of oligomeric Aβ through a mechanism that involves increased expression of insulin degrading enzyme and heme-oxygenase-1.168 In contrast, using microglial cultures from AD human brains, researchers show that IL-34 stimulation decreases expression of genes associated with phagocytosis such as CD68, which is a lysosomal marker commonly used to evaluate phagocytic potential in AD mouse models.196 Collectively, these findings highlight the potential temporal and anatomical complexity of IL-34 in the AD brain.
Increasing evidence also supports the role of IL-10, IL-12, and IL-23 in AD pathogenesis. For example, the shared subunit of IL-12 and IL-23 (i.e. p40) and IL-10 are found at elevated levels in the CSF of AD patients.197,198 A blood biomarker study also found IL-10 and p40 at increased levels in cognitively normal patients that exhibit abnormally high levels of Aβ deposition.199 In turn, the ablation of p40 can attenuate cognitive deficits and Aβ levels in APPPS1 mice.197 Peripheral treatment with anti-p40 antibody in APPPS1 mice is also able to decrease Aβ levels.197 Interestingly, the loss of IL-12 and IL-23 signaling through p40 genetic ablation has sex-specific effects in which only males exhibit a reduction in Aβ plaques.200 The silencing of p40 in aged senescence-accelerated mouse prone-8 (SAMP8) mice also mitigates cognitive decline, Aβ load, and neuronal death.169 Lastly, ablation of IL-10 in the context of AD is also considered beneficial. In fact, APPPS1 mice lacking IL-10 show improved synaptic health and cognition compared with APPPS1 controls.170 Therefore, the unchecked functioning of these cytokines during chronic inflammation of AD poses a serious threat to brain health and function.
Role of inflammasomes in AD:
Recent studies suggest a detrimental role for inflammasomes in AD.201–204 Inflammasomes are multimolecular complexes that form following receptor activation in response to various DAMPs, PAMPs, and NAMPs.205 The formation of these complexes initiates a signaling cascade in which active caspase-1 promotes the cleavage of inflammatory pro-IL-1β and pro-IL-18 cytokines into their active forms and also incites cell death.205 Inflammasome-mediated cell death is termed “pyroptosis” and is executed by cleaved gasdermin-D.206 Gasdermin-D proteins are cleaved by caspase-1,−4,−5, or −11, and this product comes together to form a pore on the cell membrane to promote cell lysis and the exit of pro-inflammatory cytokines from the cell.207 The NLRP3 inflammasome, in particular, has been identified as an important contributor to AD pathogenesis.185,201,202,208–210 NLRP3 is upregulated and activated in AD, with increased mRNA and protein expression in immune cells.211 The loss of NLRP3 inflammasome in APPPS1 mice attenuates spatial memory and LTP deficits, in addition to decreasing Aβ burden.201 NLRP3 was also recently shown to promote tau hyperphosphorylation and aggregation.212,213 Accordingly, inhibitors such as the JC-124 drug or fenamate class NSAIDs have been developed to successfully inhibit the NLRP3 inflammasome to mirror these improvements in a potentially therapeutic format.202,214 Recent research shows that apoptosis-associated speck-like protein containing CARD (ASC), an adaptor protein critical for the formation of the NLRP3 inflammasome, binds with Aβ.215 This ASC-Aβ interaction initiates NLRP3 inflammasome activation and prevents proper clearance of Aβ.216 In turn, the increased Aβ levels promote microglial pyroptosis leading to the extracellular release of more ASC that can bind Aβ to activate the cycle again, generating a chronic neuroinflammatory cascade.216
The AIM2 inflammasome, which is activated by double-stranded DNA, has also been implicated in AD.204 Aim2 deficient 5xFAD AD mice have decreased Aβ plaque load, however, this is not sufficient to rescue memory deficits. Additionally, 5xFAD;Aim2−/− mice have increased inflammation mediated by preserved IL-1 expression and an increase in the expression of pro-inflammatory cytokines IL-6 and IL-18 in the brain.204 This may suggest parallel inflammatory mechanisms to compensate for the loss of Aim2 in AD mouse models. Interestingly, our lab and others have shown the loss of Aim2 impacts neuronal morphology and negatively influences several behaviors such as anxiety and memory.217,218 Furthermore, our recent studies demonstrate that defects in the AIM2 inflammasome can alter neurodevelopment.217 Thus, it is feasible that changes in brain development existing before Aβ deposition may influence disease outcomes in AD mouse models. Moving forward, it will be interesting to revisit the role of the AIM2 inflammasome in AD using approaches that conditionally ablate AIM2 signaling following neurodevelopment.
Role of age, environmental factors, and sex in AD:
In recent years, there has been increasing appreciation for the instrumental roles that aging, environmental factors, and lifestyle choices play in AD pathogenesis (Fig. 4).219 Interestingly, emerging work has shown that many of these environmental and lifestyle factors influence AD progression by modulating aspects of inflammatory signaling.2 In this section, we will spotlight up-and-coming work describing how changes to inflammatory responses are now believed to underlie the ability of many environmental and lifestyle factors to influence AD.
Figure 4. Immune dysfunction underlies the ability of AD risk factors to impact neurodegenerative disease development.
Several genetic and peripheral factors are known to modulate AD risk. Among these factors, increased AD risk can arise from impaired microglial function amidst aging, genetic variation, sleep deficits, and in females. Additionally, obesity, gut dysbiosis, systemic inflammation, and head injury are known to cause disruptions in blood-brain barrier (BBB) integrity. BBB disruptions can contribute to a heightened risk of disease by means of altered microglial function and impaired removal of AD pathology.
Age is by far the greatest risk factor for developing AD.220 Interestingly, recent studies suggest that aging profoundly impacts microglial biology and that this can hinder their ability to properly handle and dispose of Aβ.86 Along with their propensity to take on the DAM transcriptional profile, aging also alters microglia morphology, enhances their activation status, limits their motility, and curbs their ability to phagocytose Aβ.63,221–223 Unfortunately, all of these age-related microglial changes have the potential to promote AD pathology. The ability of microglia to self-sustain their population is also speculated to make them particularly susceptible to telomeric shortening and cellular senescence, and it has been proposed that the acquisition of this senescence phenotype can cause microglial dysfunction.224 An additional mechanism by which aging contributes to AD is the loss of “youthful” blood-derived factors as one ages. Such studies demonstrate that parabiosis or plasma transfer from young mice to old mice improves learning and memory.225,226 Two factors shown to be elevated in young blood include thrombospondin-4 and SPARCL1, which support synaptic connectivity.227 It has also been shown that blood from old mice negatively impacts cognition in young mice.228 One factor suggested to contribute to this phenomenon is the chemokine CCL1, as it is elevated in aged-mice plasma levels and directly impairs cognition in recipient young mice.228
Obesity and exercise have also been implicated in AD prevalence and severity.166,229–232 Researchers have shown that obese individuals have high levels of the short chain fatty acid known as palmitate in the CSF and that this can lead to impaired neuronal insulin signaling.166 They further report that increased levels of palmitate in the CSF of aged mice provokes pro-inflammatory TNF-α cytokine production and that this can lead to memory deficits.166 Indeed, they showed that TNF-α receptor deficient mice treated with palmitate perform better on memory tests compared with palmitate treated wildtype controls.166 In addition, an intracerebroventricular injection of infliximab, a TNF-α neutralizing antibody, was also found to improve memory and ameliorate microgliosis in mice on a high fat diet.166
Routine exercise, on the other hand, has been shown to help limit AD development and progression233. One mechanism that may explain this phenomenon is the release of the myokine FNDC5/irisin from the muscle to the brain during exercise.230 FNDC5/irisin production following exercise increases synaptic plasticity, a critical aspect of memory formation and maintenance230,234. A mechanism by which memory formation is promoted by irisin is its ability to activate the memory-associated cAMP–PKA–CREB signaling pathway.235 Frequent exercise is also linked with decreased risk of viral and bacterial infections.236 Therefore, an increased susceptibility to infection as a result of insufficient exercise, which is shown to contribute to neurodegenerative pathology, may account for another mechanism by which exercise is linked with AD pathogenesis.237
Oral hygiene also appears to play a role in AD risk.238 More specifically, people with the gum disease known as periodontitis have persistent inflammation that is linked with AD.238,239 For example, it has been demonstrated that serum antibody levels for periodontitis-associated bacteria are increased in AD patients, suggesting a potential link between AD and this peripheral chronic inflammation.240 In addition, levels of the pro-inflammatory cytokine TNF-α are increased in the serum of patients with periodontal disease.241 Accordingly, TNF-α levels are also significantly increased in AD patients, suggesting a potential link between this specific inflammatory cytokine resulting from oral infection and the propagation of AD brain pathology.241–243
Exposure to pathogens in the environment has also been shown to influence AD risk. Mounting evidence has produced a consequential list of bacteria, viruses, and fungi found in the AD brain, with some even directly associating with Aβ plaques and influencing tau phosphorylation.90,244–247 Moreover, it has been reported that AD patients experiencing acute systemic inflammatory responses characterized by increased levels of TNF-α exhibit a two-fold increase in cognitive deficits.248 In fact, patients with sepsis present with long-term cognitive deficits, hippocampal atrophy, and are at a higher risk for developing dementia.249,250
TBI has emerged as a major non-genetic risk factor for developing AD and other neurodegenerative disorders later in life. TBI not only significantly increases one’s risk of developing AD, but has also been linked to earlier onset and more aggressive disease progression.251–254 More specifically, findings from a number of studies demonstrate that moderate to severe brain injury can increase the risk of developing AD by between 2.3- to 4.5-fold.255–257 These numbers increase exponentially with repetitive brain injuries and interactions with genetic risk factors.258 For instance, possessing one allele of APOE ε4 doubles the risk of developing AD, whereas this risk increases by 10 fold for individuals who carry one copy of the APOE ε4 allele and have a history of brain injury.259 One mechanism thought to underlie the link between TBI and AD is BBB breakdown. TBI-induced vascular shear stress has been shown to appreciably disrupt BBB integrity.260 Cerebrovascular injury induced by TBI and AD has been postulated to be another pathological loop in which BBB breakdown propagates AD pathology and the presence of AD pathology can also incite the loss of BBB integrity.260 For instance, 30% of brains from TBI patients have Aβ plaques, and TBI can induce tau phosphorylation and Aβ aggregation.251,261,262 The accumulation of Aβ and tau can then promote BBB breakdown to initiate the cycle once again.260 These findings suggest a link between this inflammatory brain injury and AD progression.
An additional mechanism by which brain injury and neurodegenerative disease interact involves the (re)discovery of the meningeal lymphatics in the CNS.263 In particular, the role of the meningeal lymphatics in neuroinflammation and Aβ accumulation may provide another compelling angle to the relationship between TBI and AD.264,265 For instance, TBI has recently been shown to compromise meningeal lymphatic function, namely its ability to drain inflammatory byproducts to lymph nodes following injury.266 Interestingly, meningeal lymphatic drainage defects are known to significantly increase meningeal Aβ load and to provoke memory deficits in 5xFAD AD mice.265 Therefore, meningeal lymphatic dysfunction may provide another explanation for the association between AD pathogenesis and brain injury, with promising clinical implications that require further investigation.
BBB permeability is also known to be influenced by gut microbiota composition.267 Indeed, changes in intestinal microbiota landscape have been shown to have prominent effects on the brain either directly via production of microbial products that shape BBB function or indirectly through the modulation of peripheral immune responses.267,268 Increased gut permeability allows for bacteria, bacterial toxins, and LPS to enter the bloodstream and has been coined “leaky gut.”269,270 This leakiness is instigated by chronic inflammation and gut dysbiosis, which can contribute to systemic inflammation and the decreased BBB integrity associated with aging and AD.229,269,271 BBB breakdown can ultimately lead to impaired removal of toxic species from the CNS and entry of pathogenic mediators from the periphery.269,271 Interestingly, compared to WT controls, APPPS1 mice have altered bacterial species in the gut, including higher levels of the bacteria B. thetaiotaomicron.229 In these studies, it was also shown that modulating the gut microbiota landscape with probiotics and exercise treatment can improve cognitive functions in APPPS1 mice.229
In addition to rescuing memory, the modulation of microbiota composition upon fecal transfer from WT mice to ADLPAPT AD mice has been shown to attenuate Aβ load, tau pathology, and microgliosis.272 Antibiotic cocktail treatment to alter microbiota species in the gut of APPPS1–21 AD mice has likewise been reported to reduce Aβ load and microglial activation in males specifically.273 Accordingly, cognitively impaired patients with high levels of Aβ also have altered gut microbiota populations. Interestingly, this skew in microbiota composition seen in cognitively impaired individuals is commonly characterized by the outgrowth of commensal microbes that have been linked with enhanced pro-inflammatory cytokine production.274 Dietary changes in patients with MCI also appears to alter the microbiota population and their association with Aβ and p-Tau levels in the CSF.275 These data convey how peripheral inflammation induced by dysbiosis in the gut can have broad clinical applications for mitigating AD risk and pathology in the future.
Sleep deficits have long been associated with cognitive decline.276 In fact, many AD patients present with shifts in their circadian rhythm.277 However, research also shows that problems with sleep can precede neurodegeneration and increase the risk of developing AD.277,278 Notably, Aβ and tau levels are known to be elevated in the CSF and brains of sleep-deprived individuals.279–281 In healthy patients, levels of Aβ shift, with an increase during the day and a marked decline during sleep.282 However, following Aβ aggregation, this alignment with the sleep-wake cycle dissipates.282 The half-life of Aβ more than doubles with age, likely allowing for improper protein folding and aggregate formation acting as a magnet for extracellular Aβ and preventing its transport into the CSF.283 Impaired microglial clearance of Aβ likely contributes to this enhancement in Aβ half-life. Deficits in sleep itself shift microglia to an activated state and aged phenotype, characterized by impaired Aβ clearance.284 Additionally, sleep disturbances increase the risk for inflammatory disease, which also has the potential to prime microglia to contribute to their impaired function with aging.224,285
The disparity between female and male prevalence of AD has also sparked research to distinguish the pathways underlying this sexual dimorphism. The increase in incidence of AD in females is thought to involve females’ dramatic decrease in estrogen levels during menopause, which is known to impact mitochondrial metabolism.286–288 In line with this hypothesis, estrogen replacement therapy (ERT) has been shown to significantly reduce the risk of developing AD.289 The decrease in testosterone in males occurs as a gradual decline, and therefore, it is believed to have a less detrimental effect on mitochondrial function.287 The decrease in sex hormone levels, characteristic during aging, affect mitochondrial metabolism by decreasing mitochondrial energy production and calcium efflux while increasing oxidative stress and reactive oxygen species (ROS) release.287,290 The surge of oxidative stress and ROS amplifies inflammatory responses.291 Accordingly, the release of ROS can trigger NLRP3 inflammasome activation and lead to downstream proinflammatory cytokine production, pyroptosis, and ASC speck seeding of Aβ spread.201,202,213,216,291,292 Some AD researchers are in favor of a “mitochondrial cascade hypothesis” in which mitochondrial dysfunction precedes AD pathogenesis by stimulating an increase in Aβ production and leading to the phosphorylation of tau.293 In a feed-forward mechanism, the AD pathologies themselves also propagate mitochondrial dysfunction and inflammatory ROS production which is consistently found at higher levels in AD patients.294,295 This has led to great interest in AD therapeutics that promote mitophagy to eliminate impaired mitochondria.296 In fact, boosting mitophagy has been shown to attenuate cognitive deficits and promote microglial phagocytosis of Aβ while decreasing neuroinflammation in APPPS1 AD mice.297
An additional sex-specific factor involved in AD is the sexual dimorphism of microglia. Female microglia display distinct morphology, decreased density, and altered function.27 It has also been demonstrated that male and female microglia in mice exhibit distinct transcriptional profiles that are retained when transplanted into the opposite sex.298 This suggests that the sex-specific microglia transcriptome does not rely on maintenance by sex-steroids. However, this does not exclude the possibility that the sex-steroids have a permanent effect on microglia during development. In fact, the development of microglia, as measured by gene expression profiles, occurs at disparate paces between the sexes.299 In turn, there is also a sex-specific divergence in the functionality of microglia in mice. For example, using a 5xFAD AD mouse model, it has been shown that female APOE ε4/5xFAD mice exhibit decreased microglia-mediated compaction of Aβ-plaques, decreased Trem2 expression, and increased plaque burden compared with male APOE ε4/5xFAD mice.300 Such studies suggest that sex-based differences in microglial response might help to explain the higher rates of AD in females.
CONCLUSIONS
The role of innate immunity in the modulation of AD pathology and progression poses exciting new avenues for neurodegenerative research. Mounting evidence suggests that microglia and the maintenance of a healthy neuronal environment appear to be inextricably linked. Decades of research have been dedicated to targeting the processing and production of Aβ, while disappointing clinical results have left researchers to look broader. The new focus on innate immunity, microglia, and neuroinflammation in AD has generated a burgeoning field, and exploring their roles will likely help to identify new and much-needed drug targets to combat this devastating neurodegenerative disease. Recent advancements in AD and neuroimmunology research have begun to uncover critical roles for the innate immune system in AD onset, pathology, and progression. Results from this relatively new area of study suggest that AD treatment is subject to variables such as immune response, environmental factors, and lifestyle choices that are pointing the field to a more individualistic style of therapeutics.
ACKNOWLEDGEMENTS
We apologize to authors whose work could not be referenced in this review due to space limitations. We thank members of the Lukens lab and the Center for Brain Immunology and Glia (BIG) for valuable discussions. Graphical illustrations were made using BioRender (https://biorender.com/). This work was supported by The National Institutes of Health/National Institute of Neurological Disorders and Stroke (R01NS106383; awarded to J.R.L.), The Alzheimer’s Association (AARG-18-566113; awarded to J.R.L.), and The Owens Family Foundation (Awarded to J.R.L.). H.E.E was supported by a Cell and Molecular Biology Training Grant (T32GM008136).
Abbreviations:
- AAV
adeno-associated virus
- Aβ
amyloid beta
- AD
Alzheimer’s disease
- AMP
antimicrobial peptide
- APOE
apolipoprotein E gene
- APP
amyloid precursor protein
- ASC
apoptosis-associated speck-like protein containing CARD
- BBB
blood-brain barrier
- CNS
central nervous system
- CSF
cerebrospinal fluid
- CSF-1R
colony stimulating factor-1 receptor
- CX3CR1
CX3 chemokine receptor
- DAM
disease associated microglia
- DAMP
damage associated molecular pattern
- EDE
endothelin degrading enzyme
- EOAD
early onset AD
- ERT
estrogen replacement therapy
- FAD
familial AD
- FcR
Fc receptor
- GWAS
genome-wide association study
- HSV
herpes simplex virus
- IDE
insulin degrading enzyme
- IFN
interferon
- IL
interleukin
- ISG
IFN-stimulated gene
- ITAM
immunoreceptor tyrosine-based activation motif
- ITG
inferior temporal gyrus
- ITIM
immunoreceptor tyrosine-based inhibitory motif
- LOAD
late onset AD
- LTP
long-term potentiation
- MCI
mild cognitive impairment
- MTG
middle temporal gyrus
- NAMP
neurodegeneration-associated molecular patterns
- NfL
neurofilament light chain
- NFT
neurofibrillary tangle
- NLRP3
NOD-like receptor family pyrin domain containing 3
- NOD
Nucleotide-binding oligomerization domain
- PAMP
pathogen-associated molecular pattern
- PET
positron emission tomography
- PRR
pattern recognition receptor
- PSEN1
presenilin-1
- PSEN2
presenilin-2
- p-Tau
hyperphosphorylated tau
- R47H
rare Arginine-47-Histidine
- RAGE
receptor of advanced glycosylation end products
- ROS
reactive oxygen species
- SAMP8
senescence-accelerated mouse prone-8
- SHP
Src homology 2 domain-containing phosphatase
- Siglec
sialic-acid binding immunoglobulin-like lectin
- sTREM2
soluble TREM2
- SYK
spleen tyrosine kinase
- TBI
traumatic brain injury
- TLR
toll-like receptor
- TNF
tumor necrosis factor
- TNFR1
TNF receptor 1
- TREM2
triggering receptor expressed on myeloid cells 2
- UPS
ubiquitin proteasome system
Footnotes
COMPETING INTERESTS
The authors declare no competing financial interests
REFERENCES
- 1.Nussbaum RL, Ellis CE. Alzheimer’s disease and Parkinson’s disease. N Engl J Med. 2003;348(14):1356–1364. [DOI] [PubMed] [Google Scholar]
- 2.Heneka MT, Carson MJ, Khoury JE, et al. Neuroinflammation in Alzheimer’s disease. The Lancet Neurology. 2015;14(4):388–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Aging NIo. What Causes Alzheimer’s Disease? 2020.
- 4.Perl DP. Neuropathology of Alzheimer’s disease. Mt Sinai J Med. 2010;77(1):32–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.He Z, Guo JL, McBride JD, et al. Amyloid-beta plaques enhance Alzheimer’s brain tau-seeded pathologies by facilitating neuritic plaque tau aggregation. Nat Med. 2018;24(1):29–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.LaFerla FM, Tinkle BT, Bieberich CJ, Haudenschild CC, Jay G. The Alzheimer’s A beta peptide induces neurodegeneration and apoptotic cell death in transgenic mice. Nat Genet. 1995;9(1):21–30. [DOI] [PubMed] [Google Scholar]
- 7.Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297(5580):353–356. [DOI] [PubMed] [Google Scholar]
- 8.Selkoe DJ, Hardy J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol Med. 2016;8(6):595–608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Musiek ES, Holtzman DM. Three dimensions of the amyloid hypothesis: time, space and ‘wingmen’. Nat Neurosci. 2015;18(6):800–806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Jack CR Jr., Knopman DS, Jagust WJ, et al. Hypothetical model of dynamic biomarkers of the Alzheimer’s pathological cascade. Lancet Neurol. 2010;9(1):119–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Chen GF, Xu TH, Yan Y, et al. Amyloid beta: structure, biology and structure-based therapeutic development. Acta Pharmacol Sin. 2017;38(9):1205–1235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Chasseigneaux S, Allinquant B. Functions of Abeta, sAPPalpha and sAPPbeta : similarities and differences. J Neurochem. 2012;120 Suppl 1:99–108. [DOI] [PubMed] [Google Scholar]
- 13.Kayed R, Lasagna-Reeves CA. Molecular mechanisms of amyloid oligomers toxicity. J Alzheimers Dis. 2013;33 Suppl 1:S67–78. [DOI] [PubMed] [Google Scholar]
- 14.Katzmarski N, Ziegler-Waldkirch S, Scheffler N, et al. Abeta oligomers trigger and accelerate Abeta seeding. Brain Pathol. 2020;30(1):36–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Klein AM, Kowall NW, Ferrante RJ. Neurotoxicity and oxidative damage of beta amyloid 1–42 versus beta amyloid 1–40 in the mouse cerebral cortex. Ann N Y Acad Sci. 1999;893:314–320. [DOI] [PubMed] [Google Scholar]
- 16.Serrano-Pozo A, Frosch MP, Masliah E, Hyman BT. Neuropathological alterations in Alzheimer disease. Cold Spring Harb Perspect Med. 2011;1(1):a006189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Sadleir KR, Kandalepas PC, Buggia-Prevot V, Nicholson DA, Thinakaran G, Vassar R. Presynaptic dystrophic neurites surrounding amyloid plaques are sites of microtubule disruption, BACE1 elevation, and increased Abeta generation in Alzheimer’s disease. Acta Neuropathol. 2016;132(2):235–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Tanzi RE. The genetics of Alzheimer disease. Cold Spring Harb Perspect Med. 2012;2(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.LarsBertram CML, Tanzi Rudolph E.. The Genetics of Alzheimer Disease: Back to the Future. 2010.
- 20.Panegyres PK, Chen HY. Differences between early and late onset Alzheimer’s disease. Am J Neurodegener Dis. 2013;2(4):300–306. [PMC free article] [PubMed] [Google Scholar]
- 21.Isik AT. Late onset Alzheimer’s disease in older people. Clin Interv Aging. 2010;5:307–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Safieh M, Korczyn AD, Michaelson DM. ApoE4: an emerging therapeutic target for Alzheimer’s disease. BMC Med. 2019;17(1):64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Huang Y, Mahley RW. Apolipoprotein E: structure and function in lipid metabolism, neurobiology, and Alzheimer’s diseases. Neurobiol Dis. 2014;72 Pt A:3–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ma J, Yee A, Brewer HB Jr., Das S, Potter H. Amyloid-associated proteins alpha 1-antichymotrypsin and apolipoprotein E promote assembly of Alzheimer beta-protein into filaments. Nature. 1994;372(6501):92–94. [DOI] [PubMed] [Google Scholar]
- 25.Namba Y, Tomonaga M, Kawasaki H, Otomo E, Ikeda K. Apolipoprotein E immunoreactivity in cerebral amyloid deposits and neurofibrillary tangles in Alzheimer’s disease and kuru plaque amyloid in Creutzfeldt-Jakob disease. Brain Res. 1991;541(1):163–166. [DOI] [PubMed] [Google Scholar]
- 26.Tao Q, Ang TFA, DeCarli C, et al. Association of Chronic Low-grade Inflammation With Risk of Alzheimer Disease in ApoE4 Carriers. JAMA Netw Open. 2018;1(6):e183597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hemonnot AL, Hua J, Ulmann L, Hirbec H. Microglia in Alzheimer Disease: Well-Known Targets and New Opportunities. Front Aging Neurosci. 2019;11:233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Brier MR, Gordon B, Friedrichsen K, et al. Tau and Abeta imaging, CSF measures, and cognition in Alzheimer’s disease. Sci Transl Med. 2016;8(338):338ra366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Nordberg A PET imaging of amyloid in Alzheimer’s disease. Lancet Neurol. 2004;3(9):519–527. [DOI] [PubMed] [Google Scholar]
- 30.Sabri O, Sabbagh MN, Seibyl J, et al. Florbetaben PET imaging to detect amyloid beta plaques in Alzheimer’s disease: phase 3 study. Alzheimers Dement. 2015;11(8):964–974. [DOI] [PubMed] [Google Scholar]
- 31.Landau SM, Thomas BA, Thurfjell L, et al. Amyloid PET imaging in Alzheimer’s disease: a comparison of three radiotracers. Eur J Nucl Med Mol Imaging. 2014;41(7):1398–1407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Okamura N, Harada R, Furumoto S, Arai H, Yanai K, Kudo Y. Tau PET imaging in Alzheimer’s disease. Curr Neurol Neurosci Rep. 2014;14(11):500. [DOI] [PubMed] [Google Scholar]
- 33.Hamelin L, Lagarde J, Dorothee G, et al. Early and protective microglial activation in Alzheimer’s disease: a prospective study using 18F-DPA-714 PET imaging. Brain. 2016;139(Pt 4):1252–1264. [DOI] [PubMed] [Google Scholar]
- 34.Mila-Aloma M, Suarez-Calvet M, Molinuevo JL. Latest advances in cerebrospinal fluid and blood biomarkers of Alzheimer’s disease. Ther Adv Neurol Disord. 2019;12:1756286419888819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Blennow K, Zetterberg H. Cerebrospinal fluid biomarkers for Alzheimer’s disease. J Alzheimers Dis. 2009;18(2):413–417. [DOI] [PubMed] [Google Scholar]
- 36.Blennow K Cerebrospinal fluid protein biomarkers for Alzheimer’s disease. NeuroRx. 2004;1(2):213–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Hampel H, O’Bryant SE, Molinuevo JL, et al. Blood-based biomarkers for Alzheimer disease: mapping the road to the clinic. Nat Rev Neurol. 2018;14(11):639–652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Preische O, Schultz SA, Apel A, et al. Serum neurofilament dynamics predicts neurodegeneration and clinical progression in presymptomatic Alzheimer’s disease. Nat Med. 2019;25(2):277–283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Soscia SJ, Kirby JE, Washicosky KJ, et al. The Alzheimer’s disease-associated amyloid beta-protein is an antimicrobial peptide. PLoS One. 2010;5(3):e9505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kumar DK, Choi SH, Washicosky KJ, et al. Amyloid-beta peptide protects against microbial infection in mouse and worm models of Alzheimer’s disease. Sci Transl Med. 2016;8(340):340ra372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Moir RD, Lathe R, Tanzi RE. The antimicrobial protection hypothesis of Alzheimer’s disease. Alzheimers Dement. 2018;14(12):1602–1614. [DOI] [PubMed] [Google Scholar]
- 42.Fowler DM, Koulov AV, Balch WE, Kelly JW. Functional amyloid--from bacteria to humans. Trends Biochem Sci. 2007;32(5):217–224. [DOI] [PubMed] [Google Scholar]
- 43.Rapsinski GJ, Wynosky-Dolfi MA, Oppong GO, et al. Toll-like receptor 2 and NLRP3 cooperate to recognize a functional bacterial amyloid, curli. Infect Immun. 2015;83(2):693–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Tukel C, Wilson RP, Nishimori JH, Pezeshki M, Chromy BA, Baumler AJ. Responses to amyloids of microbial and host origin are mediated through toll-like receptor 2. Cell Host Microbe. 2009;6(1):45–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Salminen A, Ojala J, Kauppinen A, Kaarniranta K, Suuronen T. Inflammation in Alzheimer’s disease: amyloid-beta oligomers trigger innate immunity defence via pattern recognition receptors. Prog Neurobiol. 2009;87(3):181–194. [DOI] [PubMed] [Google Scholar]
- 46.Chen GY, Nunez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol. 2010;10(12):826–837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Guo T, Noble W, Hanger DP. Roles of tau protein in health and disease. Acta Neuropathol. 2017;133(5):665–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Laurent C, Buee L, Blum D. Tau and neuroinflammation: What impact for Alzheimer’s Disease and Tauopathies? Biomed J. 2018;41(1):21–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Wozniak MA, Frost AL, Itzhaki RF. Alzheimer’s disease-specific tau phosphorylation is induced by herpes simplex virus type 1. J Alzheimers Dis. 2009;16(2):341–350. [DOI] [PubMed] [Google Scholar]
- 50.Organization WH. Globally, an estimated two-thirds of the population under 50 are infected with herpes simplex virus type 1. 2015.
- 51.Wozniak MA, Frost AL, Preston CM, Itzhaki RF. Antivirals reduce the formation of key Alzheimer’s disease molecules in cell cultures acutely infected with herpes simplex virus type 1. PLoS One. 2011;6(10):e25152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Aging NIo. Valacyclovir for Mild Alzheimer’s Disease. 2018.
- 53.Lenz KM, Nelson LH. Microglia and Beyond: Innate Immune Cells As Regulators of Brain Development and Behavioral Function. Front Immunol. 2018;9:698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Wildsmith KR, Holley M, Savage JC, Skerrett R, Landreth GE. Evidence for impaired amyloid beta clearance in Alzheimer’s disease. Alzheimers Res Ther. 2013;5(4):33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Mhatre SD, Tsai CA, Rubin AJ, James ML, Andreasson KI. Microglial malfunction: the third rail in the development of Alzheimer’s disease. Trends Neurosci. 2015;38(10):621–636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Sarlus H, Heneka MT. Microglia in Alzheimer’s disease. J Clin Invest. 2017;127(9):3240–3249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Reed-Geaghan EG, Croxford AL, Becher B, Landreth GE. Plaque-associated myeloid cells derive from resident microglia in an Alzheimer’s disease model. J Exp Med. 2020;217(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Darío Tejera MTH. Microglia in Neurodegenerative Disorders. 2019. [DOI] [PubMed]
- 59.Ginhoux F, Lim S, Hoeffel G, Low D, Huber T. Origin and differentiation of microglia. Front Cell Neurosci. 2013;7:45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Sierra A, Paolicelli RC, Kettenmann H. Cien Anos de Microglia: Milestones in a Century of Microglial Research. Trends Neurosci. 2019;42(11):778–792. [DOI] [PubMed] [Google Scholar]
- 61.Parakalan R, Jiang B, Nimmi B, et al. Transcriptome analysis of amoeboid and ramified microglia isolated from the corpus callosum of rat brain. BMC Neurosci. 2012;13:64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Kigerl KA, de Rivero Vaccari JP, Dietrich WD, Popovich PG, Keane RW. Pattern recognition receptors and central nervous system repair. Exp Neurol. 2014;258:5–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Keren-Shaul H, Spinrad A, Weiner A, et al. A Unique Microglia Type Associated with Restricting Development of Alzheimer’s Disease. Cell. 2017;169(7):1276–1290 e1217. [DOI] [PubMed] [Google Scholar]
- 64.Deczkowska A, Keren-Shaul H, Weiner A, Colonna M, Schwartz M, Amit I. Disease-Associated Microglia: A Universal Immune Sensor of Neurodegeneration. Cell. 2018;173(5):1073–1081. [DOI] [PubMed] [Google Scholar]
- 65.Ulland TK, Colonna M. TREM2 - a key player in microglial biology and Alzheimer disease. Nat Rev Neurol. 2018;14(11):667–675. [DOI] [PubMed] [Google Scholar]
- 66.Guerreiro R, Wojtas A, Bras J, et al. TREM2 variants in Alzheimer’s disease. N Engl J Med. 2013;368(2):117–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Jonsson T, Stefansson H, Steinberg S, et al. Variant of TREM2 associated with the risk of Alzheimer’s disease. N Engl J Med. 2013;368(2):107–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wang Y, Cella M, Mallinson K, et al. TREM2 lipid sensing sustains the microglial response in an Alzheimer’s disease model. Cell. 2015;160(6):1061–1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Zhao Y, Wu X, Li X, et al. TREM2 Is a Receptor for beta-Amyloid that Mediates Microglial Function. Neuron. 2018;97(5):1023–1031 e1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Atagi Y, Liu CC, Painter MM, et al. Apolipoprotein E Is a Ligand for Triggering Receptor Expressed on Myeloid Cells 2 (TREM2). J Biol Chem. 2015;290(43):26043–26050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Wang Y, Ulland TK, Ulrich JD, et al. TREM2-mediated early microglial response limits diffusion and toxicity of amyloid plaques. J Exp Med. 2016;213(5):667–675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Condello C, Yuan P, Grutzendler J. Microglia-Mediated Neuroprotection, TREM2, and Alzheimer’s Disease: Evidence From Optical Imaging. Biol Psychiatry. 2018;83(4):377–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Condello C, Yuan P, Schain A, Grutzendler J. Microglia constitute a barrier that prevents neurotoxic protofibrillar Abeta42 hotspots around plaques. Nat Commun. 2015;6:6176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Ulland TK, Song WM, Huang SC, et al. TREM2 Maintains Microglial Metabolic Fitness in Alzheimer’s Disease. Cell. 2017;170(4):649–663 e613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Parhizkar S, Arzberger T, Brendel M, et al. Loss of TREM2 function increases amyloid seeding but reduces plaque-associated ApoE. Nat Neurosci. 2019;22(2):191–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Krasemann S, Madore C, Cialic R, et al. The TREM2-APOE Pathway Drives the Transcriptional Phenotype of Dysfunctional Microglia in Neurodegenerative Diseases. Immunity. 2017;47(3):566–581 e569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Piccio L, Deming Y, Del-Aguila JL, et al. Cerebrospinal fluid soluble TREM2 is higher in Alzheimer disease and associated with mutation status. Acta Neuropathol. 2016;131(6):925–933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Zhong L, Xu Y, Zhuo R, et al. Soluble TREM2 ameliorates pathological phenotypes by modulating microglial functions in an Alzheimer’s disease model. Nat Commun. 2019;10(1):1365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Hollingworth P, Harold D, Sims R, et al. Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer’s disease. Nat Genet. 2011;43(5):429–435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Zhao L. CD33 in Alzheimer’s Disease - Biology, Pathogenesis, and Therapeutics: A Mini-Review. Gerontology. 2019;65(4):323–331. [DOI] [PubMed] [Google Scholar]
- 81.Varki A. Sialic acids in human health and disease. Trends Mol Med. 2008;14(8):351–360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Bradshaw EM, Chibnik LB, Keenan BT, et al. CD33 Alzheimer’s disease locus: altered monocyte function and amyloid biology. Nat Neurosci. 2013;16(7):848–850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Walker DG, Whetzel AM, Serrano G, Sue LI, Beach TG, Lue LF. Association of CD33 polymorphism rs3865444 with Alzheimer’s disease pathology and CD33 expression in human cerebral cortex. Neurobiol Aging. 2015;36(2):571–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Karch CM, Jeng AT, Nowotny P, Cady J, Cruchaga C, Goate AM. Expression of novel Alzheimer’s disease risk genes in control and Alzheimer’s disease brains. PLoS One. 2012;7(11):e50976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Griciuc A, Patel S, Federico AN, et al. TREM2 Acts Downstream of CD33 in Modulating Microglial Pathology in Alzheimer’s Disease. Neuron. 2019;103(5):820–835 e827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Pluvinage JV, Haney MS, Smith BAH, et al. CD22 blockade restores homeostatic microglial phagocytosis in ageing brains. Nature. 2019;568(7751):187–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Brown GD. Dectin-1: a signalling non-TLR pattern-recognition receptor. Nat Rev Immunol. 2006;6(1):33–43. [DOI] [PubMed] [Google Scholar]
- 88.Alonso R, Pisa D, Marina AI, Morato E, Rabano A, Carrasco L. Fungal infection in patients with Alzheimer’s disease. J Alzheimers Dis. 2014;41(1):301–311. [DOI] [PubMed] [Google Scholar]
- 89.Pisa D, Alonso R, Juarranz A, Rabano A, Carrasco L. Direct visualization of fungal infection in brains from patients with Alzheimer’s disease. J Alzheimers Dis. 2015;43(2):613–624. [DOI] [PubMed] [Google Scholar]
- 90.Pisa D, Alonso R, Rabano A, Rodal I, Carrasco L. Different Brain Regions are Infected with Fungi in Alzheimer’s Disease. Sci Rep. 2015;5:15015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Watabe-Rudolph M, Song Z, Lausser L, et al. Chitinase enzyme activity in CSF is a powerful biomarker of Alzheimer disease. Neurology. 2012;78(8):569–577. [DOI] [PubMed] [Google Scholar]
- 92.Parady B Innate Immune and Fungal Model of Alzheimer’s Disease. J Alzheimers Dis Rep. 2018;2(1):139–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Doens D, Fernandez PL. Microglia receptors and their implications in the response to amyloid beta for Alzheimer’s disease pathogenesis. J Neuroinflammation. 2014;11:48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Chavakis T, Bierhaus A, Nawroth PP. RAGE (receptor for advanced glycation end products): a central player in the inflammatory response. Microbes Infect. 2004;6(13):1219–1225. [DOI] [PubMed] [Google Scholar]
- 95.Lee EJ, Park JH. Receptor for Advanced Glycation Endproducts (RAGE), Its Ligands, and Soluble RAGE: Potential Biomarkers for Diagnosis and Therapeutic Targets for Human Renal Diseases. Genomics Inform. 2013;11(4):224–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Sparvero LJ, Asafu-Adjei D, Kang R, et al. RAGE (Receptor for Advanced Glycation Endproducts), RAGE ligands, and their role in cancer and inflammation. J Transl Med. 2009;7:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Fang F, Lue LF, Yan S, et al. RAGE-dependent signaling in microglia contributes to neuroinflammation, Abeta accumulation, and impaired learning/memory in a mouse model of Alzheimer’s disease. FASEB J. 2010;24(4):1043–1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Deane R, Du Yan S, Submamaryan RK, et al. RAGE mediates amyloid-beta peptide transport across the blood-brain barrier and accumulation in brain. Nat Med. 2003;9(7):907–913. [DOI] [PubMed] [Google Scholar]
- 99.Yan SD, Chen X, Fu J, et al. RAGE and amyloid-beta peptide neurotoxicity in Alzheimer’s disease. Nature. 1996;382(6593):685–691. [DOI] [PubMed] [Google Scholar]
- 100.Walker D, Lue LF, Paul G, Patel A, Sabbagh MN. Receptor for advanced glycation endproduct modulators: a new therapeutic target in Alzheimer’s disease. Expert Opin Investig Drugs. 2015;24(3):393–399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Hong S, Beja-Glasser VF, Nfonoyim BM, et al. Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science. 2016;352(6286):712–716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Schafer DP, Lehrman EK, Kautzman AG, et al. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron. 2012;74(4):691–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Shi Q, Colodner KJ, Matousek SB, et al. Complement C3-Deficient Mice Fail to Display Age-Related Hippocampal Decline. J Neurosci. 2015;35(38):13029–13042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Stephan AH, Madison DV, Mateos JM, et al. A dramatic increase of C1q protein in the CNS during normal aging. J Neurosci. 2013;33(33):13460–13474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Shi Q, Chowdhury S, Ma R, et al. Complement C3 deficiency protects against neurodegeneration in aged plaque-rich APP/PS1 mice. Sci Transl Med. 2017;9(392). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Czirr E, Castello NA, Mosher KI, et al. Microglial complement receptor 3 regulates brain Abeta levels through secreted proteolytic activity. J Exp Med. 2017;214(4):1081–1092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Fu H, Liu B, Frost JL, et al. Complement component C3 and complement receptor type 3 contribute to the phagocytosis and clearance of fibrillar Abeta by microglia. Glia. 2012;60(6):993–1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Fonseca MI, Chu SH, Hernandez MX, et al. Cell-specific deletion of C1qa identifies microglia as the dominant source of C1q in mouse brain. J Neuroinflammation. 2017;14(1):48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Morgan BP. Complement in the pathogenesis of Alzheimer’s disease. Semin Immunopathol. 2018;40(1):113–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Wang C, Yue H, Hu Z, et al. Microglia mediate forgetting via complement-dependent synaptic elimination. Science. 2020;367(6478):688–694. [DOI] [PubMed] [Google Scholar]
- 111.Ries M, Sastre M. Mechanisms of Abeta Clearance and Degradation by Glial Cells. Front Aging Neurosci. 2016;8:160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Landreth GE, Reed-Geaghan EG. Toll-like receptors in Alzheimer’s disease. Curr Top Microbiol Immunol. 2009;336:137–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Richard KL, Filali M, Prefontaine P, Rivest S. Toll-like receptor 2 acts as a natural innate immune receptor to clear amyloid beta 1–42 and delay the cognitive decline in a mouse model of Alzheimer’s disease. J Neurosci. 2008;28(22):5784–5793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Song M, Jin J, Lim JE, et al. TLR4 mutation reduces microglial activation, increases Abeta deposits and exacerbates cognitive deficits in a mouse model of Alzheimer’s disease. J Neuroinflammation. 2011;8:92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Arroyo-Espliguero R, Avanzas P, Jeffery S, Kaski JC. CD14 and toll-like receptor 4: a link between infection and acute coronary events? Heart. 2004;90(9):983–988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Liu Y, Walter S, Stagi M, et al. LPS receptor (CD14): a receptor for phagocytosis of Alzheimer’s amyloid peptide. Brain. 2005;128(Pt 8):1778–1789. [DOI] [PubMed] [Google Scholar]
- 117.Bard F, Cannon C, Barbour R, et al. Peripherally administered antibodies against amyloid beta-peptide enter the central nervous system and reduce pathology in a mouse model of Alzheimer disease. Nat Med. 2000;6(8):916–919. [DOI] [PubMed] [Google Scholar]
- 118.van Dyck CH. Anti-Amyloid-beta Monoclonal Antibodies for Alzheimer’s Disease: Pitfalls and Promise. Biol Psychiatry. 2018;83(4):311–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Mullard A Anti-amyloid failures stack up as Alzheimer antibody flops. Nat Rev Drug Discov. 2019. [DOI] [PubMed] [Google Scholar]
- 120.Kurochkin IV, Guarnera E, Berezovsky IN. Insulin-Degrading Enzyme in the Fight against Alzheimer’s Disease. Trends Pharmacol Sci. 2018;39(1):49–58. [DOI] [PubMed] [Google Scholar]
- 121.Tamboli IY, Barth E, Christian L, et al. Statins promote the degradation of extracellular amyloid {beta}-peptide by microglia via stimulation of exosome-associated insulin-degrading enzyme (IDE) secretion. J Biol Chem. 2010;285(48):37405–37414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Hellstrom-Lindahl E, Ravid R, Nordberg A. Age-dependent decline of neprilysin in Alzheimer’s disease and normal brain: inverse correlation with A beta levels. Neurobiol Aging. 2008;29(2):210–221. [DOI] [PubMed] [Google Scholar]
- 123.Yasojima K, Akiyama H, McGeer EG, McGeer PL. Reduced neprilysin in high plaque areas of Alzheimer brain: a possible relationship to deficient degradation of beta-amyloid peptide. Neurosci Lett. 2001;297(2):97–100. [DOI] [PubMed] [Google Scholar]
- 124.Huang SM, Mouri A, Kokubo H, et al. Neprilysin-sensitive synapse-associated amyloid-beta peptide oligomers impair neuronal plasticity and cognitive function. J Biol Chem. 2006;281(26):17941–17951. [DOI] [PubMed] [Google Scholar]
- 125.Eckman EA, Watson M, Marlow L, Sambamurti K, Eckman CB. Alzheimer’s disease beta-amyloid peptide is increased in mice deficient in endothelin-converting enzyme. J Biol Chem. 2003;278(4):2081–2084. [DOI] [PubMed] [Google Scholar]
- 126.Oddo S The ubiquitin-proteasome system in Alzheimer’s disease. J Cell Mol Med. 2008;12(2):363–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Kanekiyo T, Xu H, Bu G. ApoE and Abeta in Alzheimer’s disease: accidental encounters or partners? Neuron. 2014;81(4):740–754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Ulrich JD, Ulland TK, Mahan TE, et al. ApoE facilitates the microglial response to amyloid plaque pathology. J Exp Med. 2018;215(4):1047–1058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Yang L, Liu CC, Zheng H, et al. LRP1 modulates the microglial immune response via regulation of JNK and NF-kappaB signaling pathways. J Neuroinflammation. 2016;13(1):304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Friedberg JS, Aytan N, Cherry JD, et al. Associations between brain inflammatory profiles and human neuropathology are altered based on apolipoprotein E epsilon4 genotype. Sci Rep. 2020;10(1):2924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Levine B, Mizushima N, Virgin HW. Autophagy in immunity and inflammation. Nature. 2011;469(7330):323–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Cho MH, Cho K, Kang HJ, et al. Autophagy in microglia degrades extracellular beta-amyloid fibrils and regulates the NLRP3 inflammasome. Autophagy. 2014;10(10):1761–1775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Kruger U, Wang Y, Kumar S, Mandelkow EM. Autophagic degradation of tau in primary neurons and its enhancement by trehalose. Neurobiol Aging. 2012;33(10):2291–2305. [DOI] [PubMed] [Google Scholar]
- 134.Schaeffer V, Lavenir I, Ozcelik S, Tolnay M, Winkler DT, Goedert M. Stimulation of autophagy reduces neurodegeneration in a mouse model of human tauopathy. Brain. 2012;135(Pt 7):2169–2177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Sanchez-Varo R, Trujillo-Estrada L, Sanchez-Mejias E, et al. Abnormal accumulation of autophagic vesicles correlates with axonal and synaptic pathology in young Alzheimer’s mice hippocampus. Acta Neuropathol. 2012;123(1):53–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Yang DS, Stavrides P, Mohan PS, et al. Reversal of autophagy dysfunction in the TgCRND8 mouse model of Alzheimer’s disease ameliorates amyloid pathologies and memory deficits. Brain. 2011;134(Pt 1):258–277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Butovsky O, Weiner HL. Microglial signatures and their role in health and disease. Nat Rev Neurosci. 2018;19(10):622–635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Kim TS, Lim HK, Lee JY, et al. Changes in the levels of plasma soluble fractalkine in patients with mild cognitive impairment and Alzheimer’s disease. Neurosci Lett. 2008;436(2):196–200. [DOI] [PubMed] [Google Scholar]
- 139.Fuhrmann M, Bittner T, Jung CK, et al. Microglial Cx3cr1 knockout prevents neuron loss in a mouse model of Alzheimer’s disease. Nat Neurosci. 2010;13(4):411–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Cho SH, Sun B, Zhou Y, et al. CX3CR1 protein signaling modulates microglial activation and protects against plaque-independent cognitive deficits in a mouse model of Alzheimer disease. J Biol Chem. 2011;286(37):32713–32722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Lou N, Takano T, Pei Y, Xavier AL, Goldman SA, Nedergaard M. Purinergic receptor P2RY12-dependent microglial closure of the injured blood-brain barrier. Proc Natl Acad Sci U S A. 2016;113(4):1074–1079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Haynes SE, Hollopeter G, Yang G, et al. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat Neurosci. 2006;9(12):1512–1519. [DOI] [PubMed] [Google Scholar]
- 143.Han J, Harris RA, Zhang XM. An updated assessment of microglia depletion: current concepts and future directions. Mol Brain. 2017;10(1):25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Dagher NN, Najafi AR, Kayala KM, et al. Colony-stimulating factor 1 receptor inhibition prevents microglial plaque association and improves cognition in 3xTg-AD mice. J Neuroinflammation. 2015;12:139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Asai H, Ikezu S, Tsunoda S, et al. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat Neurosci. 2015;18(11):1584–1593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Olmos-Alonso A, Schetters ST, Sri S, et al. Pharmacological targeting of CSF1R inhibits microglial proliferation and prevents the progression of Alzheimer’s-like pathology. Brain. 2016;139(Pt 3):891–907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Spangenberg EE, Lee RJ, Najafi AR, et al. Eliminating microglia in Alzheimer’s mice prevents neuronal loss without modulating amyloid-beta pathology. Brain. 2016;139(Pt 4):1265–1281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Spangenberg E, Severson PL, Hohsfield LA, et al. Sustained microglial depletion with CSF1R inhibitor impairs parenchymal plaque development in an Alzheimer’s disease model. Nat Commun. 2019;10(1):3758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Elmore MR, Najafi AR, Koike MA, et al. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron. 2014;82(2):380–397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Bruttger J, Karram K, Wortge S, et al. Genetic Cell Ablation Reveals Clusters of Local Self-Renewing Microglia in the Mammalian Central Nervous System. Immunity. 2015;43(1):92–106. [DOI] [PubMed] [Google Scholar]
- 151.Henry RJ, Ritzel RM, Barrett JP, et al. Microglial Depletion with CSF1R Inhibitor During Chronic Phase of Experimental Traumatic Brain Injury Reduces Neurodegeneration and Neurological Deficits. J Neurosci. 2020;40(14):2960–2974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Chodobski A, Zink BJ, Szmydynger-Chodobska J. Blood-brain barrier pathophysiology in traumatic brain injury. Transl Stroke Res. 2011;2(4):492–516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Sweeney MD, Sagare AP, Zlokovic BV. Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat Rev Neurol. 2018;14(3):133–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Willis EF, MacDonald KPA, Nguyen QH, et al. Repopulating Microglia Promote Brain Repair in an IL-6-Dependent Manner. Cell. 2020;180(5):833–846 e816. [DOI] [PubMed] [Google Scholar]
- 155.El Khoury J, Toft M, Hickman SE, et al. Ccr2 deficiency impairs microglial accumulation and accelerates progression of Alzheimer-like disease. Nat Med. 2007;13(4):432–438. [DOI] [PubMed] [Google Scholar]
- 156.Naert G, Rivest S. Hematopoietic CC-chemokine receptor 2 (CCR2) competent cells are protective for the cognitive impairments and amyloid pathology in a transgenic mouse model of Alzheimer’s disease. Mol Med. 2012;18:297–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Moravan MJ, Olschowka JA, Williams JP, O’Banion MK. Brain radiation injury leads to a dose- and time-dependent recruitment of peripheral myeloid cells that depends on CCR2 signaling. J Neuroinflammation. 2016;13:30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Cronk JC, Filiano AJ, Louveau A, et al. Peripherally derived macrophages can engraft the brain independent of irradiation and maintain an identity distinct from microglia. J Exp Med. 2018;215(6):1627–1647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Jay TR, Miller CM, Cheng PJ, et al. TREM2 deficiency eliminates TREM2+ inflammatory macrophages and ameliorates pathology in Alzheimer’s disease mouse models. J Exp Med. 2015;212(3):287–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Kinney JW. Inflammation as a central mechanism in Alzheimer’s disease. 2018. [DOI] [PMC free article] [PubMed]
- 161.Akiyama H, Barger S, Barnum S, et al. Inflammation and Alzheimer’s disease. Neurobiol Aging. 2000;21(3):383–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Kinney JW, Bemiller SM, Murtishaw AS, Leisgang AM, Salazar AM, Lamb BT. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement (N Y). 2018;4:575–590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Taylor JM, Moore Z, Minter MR, Crack PJ. Type-I interferon pathway in neuroinflammation and neurodegeneration: focus on Alzheimer’s disease. J Neural Transm (Vienna). 2018;125(5):797–807. [DOI] [PubMed] [Google Scholar]
- 164.Thawkar BS, Kaur G. Inhibitors of NF-kappaB and P2X7/NLRP3/Caspase 1 pathway in microglia: Novel therapeutic opportunities in neuroinflammation induced early-stage Alzheimer’s disease. J Neuroimmunol. 2019;326:62–74. [DOI] [PubMed] [Google Scholar]
- 165.Bhaskar K, Maphis N, Xu G, et al. Microglial derived tumor necrosis factor-alpha drives Alzheimer’s disease-related neuronal cell cycle events. Neurobiol Dis. 2014;62:273–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Melo HM, Seixas da Silva GdS, Sant’Ana MR, et al. Palmitate Is Increased in the Cerebrospinal Fluid of Humans with Obesity and Induces Memory Impairment in Mice via Pro-inflammatory TNF-α. Cell Reports. 2020;30(7):2180–2194.e2188. [DOI] [PubMed] [Google Scholar]
- 167.Fu AK, Hung KW, Yuen MY, et al. IL-33 ameliorates Alzheimer’s disease-like pathology and cognitive decline. Proc Natl Acad Sci U S A. 2016;113(19):E2705–2713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Mizuno T, Doi Y, Mizoguchi H, et al. Interleukin-34 selectively enhances the neuroprotective effects of microglia to attenuate oligomeric amyloid-beta neurotoxicity. Am J Pathol. 2011;179(4):2016–2027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Tan MS, Yu JT, Jiang T, Zhu XC, Guan HS, Tan L. IL12/23 p40 inhibition ameliorates Alzheimer’s disease-associated neuropathology and spatial memory in SAMP8 mice. J Alzheimers Dis. 2014;38(3):633–646. [DOI] [PubMed] [Google Scholar]
- 170.Guillot-Sestier MV, Doty KR, Gate D, et al. Il10 deficiency rebalances innate immunity to mitigate Alzheimer-like pathology. Neuron. 2015;85(3):534–548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Swardfager W, Lanctot K, Rothenburg L, Wong A, Cappell J, Herrmann N. A meta-analysis of cytokines in Alzheimer’s disease. Biol Psychiatry. 2010;68(10):930–941. [DOI] [PubMed] [Google Scholar]
- 172.Polazzi E, Monti B. Microglia and neuroprotection: from in vitro studies to therapeutic applications. Prog Neurobiol. 2010;92(3):293–315. [DOI] [PubMed] [Google Scholar]
- 173.Clayton KA, Van Enoo AA, Ikezu T. Alzheimer’s Disease: The Role of Microglia in Brain Homeostasis and Proteopathy. Front Neurosci. 2017;11:680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Yamamoto M, Kiyota T, Horiba M, et al. Interferon-gamma and tumor necrosis factor-alpha regulate amyloid-beta plaque deposition and beta-secretase expression in Swedish mutant APP transgenic mice. Am J Pathol. 2007;170(2):680–692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Gorle N, Vandenbroucke RE. Interferons: A molecular switch between damage and repair in ageing and Alzheimer’s disease. Mech Ageing Dev. 2019;183:111148. [DOI] [PubMed] [Google Scholar]
- 176.Roy ER, Wang B, Wan YW, et al. Type I interferon response drives neuroinflammation and synapse loss in Alzheimer disease. J Clin Invest. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Di Domizio J, Dorta-Estremera S, Gagea M, et al. Nucleic acid-containing amyloid fibrils potently induce type I interferon and stimulate systemic autoimmunity. Proc Natl Acad Sci U S A. 2012;109(36):14550–14555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Contreras JA, Aslanyan V, Sweeney MD, et al. Functional connectivity among brain regions affected in Alzheimer’s disease is associated with CSF TNF-alpha in APOE4 carriers. Neurobiol Aging. 2020;86:112–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Steeland S, Gorle N, Vandendriessche C, et al. Counteracting the effects of TNF receptor-1 has therapeutic potential in Alzheimer’s disease. EMBO Mol Med. 2018;10(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Chakrabarty P, Herring A, Ceballos-Diaz C, Das P, Golde TE. Hippocampal expression of murine TNFalpha results in attenuation of amyloid deposition in vivo. Mol Neurodegener. 2011;6:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Andrews SJ, Goate A. Mendelian randomization indicates that TNF is not causally associated with Alzheimer’s disease. Neurobiol Aging. 2019;84:241 e241–241 e243. [DOI] [PubMed] [Google Scholar]
- 182.Bossu P, Ciaramella A, Salani F, et al. Interleukin-18, from neuroinflammation to Alzheimer’s disease. Curr Pharm Des. 2010;16(38):4213–4224. [DOI] [PubMed] [Google Scholar]
- 183.Su F, Bai F, Zhang Z. Inflammatory Cytokines and Alzheimer’s Disease: A Review from the Perspective of Genetic Polymorphisms. Neurosci Bull. 2016;32(5):469–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Sutinen EM, Pirttila T, Anderson G, Salminen A, Ojala JO. Pro-inflammatory interleukin-18 increases Alzheimer’s disease-associated amyloid-beta production in human neuron-like cells. J Neuroinflammation. 2012;9:199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Tzeng TC, Hasegawa Y, Iguchi R, et al. Inflammasome-derived cytokine IL18 suppresses amyloid-induced seizures in Alzheimer-prone mice. Proc Natl Acad Sci U S A. 2018;115(36):9002–9007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Kitazawa M, Cheng D, Tsukamoto MR, et al. Blocking IL-1 signaling rescues cognition, attenuates tau pathology, and restores neuronal beta-catenin pathway function in an Alzheimer’s disease model. J Immunol. 2011;187(12):6539–6549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Ghosh S, Wu MD, Shaftel SS, et al. Sustained interleukin-1beta overexpression exacerbates tau pathology despite reduced amyloid burden in an Alzheimer’s mouse model. J Neurosci. 2013;33(11):5053–5064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.de Souza DF, Leite MC, Quincozes-Santos A, et al. S100B secretion is stimulated by IL-1beta in glial cultures and hippocampal slices of rats: Likely involvement of MAPK pathway. J Neuroimmunol. 2009;206(1–2):52–57. [DOI] [PubMed] [Google Scholar]
- 189.Cristovao JS, Gomes CM. S100 Proteins in Alzheimer’s Disease. Front Neurosci. 2019;13:463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Chaves ML, Camozzato AL, Ferreira ED, et al. Serum levels of S100B and NSE proteins in Alzheimer’s disease patients. J Neuroinflammation. 2010;7:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Peskind. Cerebrospinal fluid S100B is elevated in the earlier stages of Alzheimer’s disease. 2001. [DOI] [PubMed]
- 192.Mori T, Koyama N, Arendash GW, Horikoshi-Sakuraba Y, Tan J, Town T. Overexpression of human S100B exacerbates cerebral amyloidosis and gliosis in the Tg2576 mouse model of Alzheimer’s disease. Glia. 2010;58(3):300–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Liew FY, Girard JP, Turnquist HR. Interleukin-33 in health and disease. Nat Rev Immunol. 2016;16(11):676–689. [DOI] [PubMed] [Google Scholar]
- 194.Chapuis J, Hot D, Hansmannel F, et al. Transcriptomic and genetic studies identify IL-33 as a candidate gene for Alzheimer’s disease. Mol Psychiatry. 2009;14(11):1004–1016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Gomez-Nicola D, Fransen NL, Suzzi S, Perry VH. Regulation of microglial proliferation during chronic neurodegeneration. J Neurosci. 2013;33(6):2481–2493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Walker DG, Tang TM, Lue LF. Studies on Colony Stimulating Factor Receptor-1 and Ligands Colony Stimulating Factor-1 and Interleukin-34 in Alzheimer’s Disease Brains and Human Microglia. Front Aging Neurosci. 2017;9:244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Vom Berg J, Prokop S, Miller KR, et al. Inhibition of IL-12/IL-23 signaling reduces Alzheimer’s disease-like pathology and cognitive decline. Nat Med. 2012;18(12):1812–1819. [DOI] [PubMed] [Google Scholar]
- 198.Taipa R, das Neves SP, Sousa AL, et al. Proinflammatory and anti-inflammatory cytokines in the CSF of patients with Alzheimer’s disease and their correlation with cognitive decline. Neurobiol Aging. 2019;76:125–132. [DOI] [PubMed] [Google Scholar]
- 199.Pedrini S, Gupta VB, Hone E, et al. A blood-based biomarker panel indicates IL-10 and IL-12/23p40 are jointly associated as predictors of beta-amyloid load in an AD cohort. Sci Rep. 2017;7(1):14057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Eede P, Obst J, Benke E, et al. Interleukin-12/23 deficiency differentially affects pathology in male and female Alzheimer’s disease-like mice. EMBO Rep. 2020:e48530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Heneka MT, Kummer MP, Stutz A, et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature. 2013;493(7434):674–678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Yin J, Zhao F, Chojnacki JE, et al. NLRP3 Inflammasome Inhibitor Ameliorates Amyloid Pathology in a Mouse Model of Alzheimer’s Disease. Mol Neurobiol. 2018;55(3):1977–1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Voet S, Mc Guire C, Hagemeyer N, et al. A20 critically controls microglia activation and inhibits inflammasome-dependent neuroinflammation. Nat Commun. 2018;9(1):2036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Wu PJ, Hung YF, Liu HY, Hsueh YP. Deletion of the Inflammasome Sensor Aim2 Mitigates Abeta Deposition and Microglial Activation but Increases Inflammatory Cytokine Expression in an Alzheimer Disease Mouse Model. Neuroimmunomodulation. 2017;24(1):29–39. [DOI] [PubMed] [Google Scholar]
- 205.Rathinam VA, Vanaja SK, Fitzgerald KA. Regulation of inflammasome signaling. Nat Immunol. 2012;13(4):333–342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Man SM, Kanneganti TD. Gasdermin D: the long-awaited executioner of pyroptosis. Cell Res. 2015;25(11):1183–1184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Liu X, Zhang Z, Ruan J, et al. Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature. 2016;535(7610):153–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Mitroulis I, Skendros P, Ritis K. Targeting IL-1beta in disease; the expanding role of NLRP3 inflammasome. Eur J Intern Med. 2010;21(3):157–163. [DOI] [PubMed] [Google Scholar]
- 209.Pennisi M, Crupi R, Di Paola R, et al. Inflammasomes, hormesis, and antioxidants in neuroinflammation: Role of NRLP3 in Alzheimer disease. J Neurosci Res. 2017;95(7):1360–1372. [DOI] [PubMed] [Google Scholar]
- 210.Goldmann T, Tay TL, Prinz M. Love and death: microglia, NLRP3 and the Alzheimer’s brain. Cell Res. 2013;23(5):595–596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Saresella M, La Rosa F, Piancone F, et al. The NLRP3 and NLRP1 inflammasomes are activated in Alzheimer’s disease. Mol Neurodegener. 2016;11:23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Stancu IC, Cremers N, Vanrusselt H, et al. Aggregated Tau activates NLRP3-ASC inflammasome exacerbating exogenously seeded and non-exogenously seeded Tau pathology in vivo. Acta Neuropathol. 2019;137(4):599–617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Ising C, Venegas C, Zhang S, et al. NLRP3 inflammasome activation drives tau pathology. Nature. 2019;575(7784):669–673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Daniels MJ, Rivers-Auty J, Schilling T, et al. Fenamate NSAIDs inhibit the NLRP3 inflammasome and protect against Alzheimer’s disease in rodent models. Nat Commun. 2016;7:12504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Venegas C, Kumar S, Franklin BS, et al. Microglia-derived ASC specks cross-seed amyloid-beta in Alzheimer’s disease. Nature. 2017;552(7685):355–361. [DOI] [PubMed] [Google Scholar]
- 216.Friker LL, Scheiblich H, Hochheiser IV, et al. beta-Amyloid Clustering around ASC Fibrils Boosts Its Toxicity in Microglia. Cell Rep. 2020;30(11):3743–3754 e3746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Lammert CR, Frost EL, Bellinger CE, et al. AIM2 inflammasome surveillance of DNA damage shapes neurodevelopment. Nature. 2020;580(7805):647–652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Wu PJ, Liu HY, Huang TN, Hsueh YP. AIM 2 inflammasomes regulate neuronal morphology and influence anxiety and memory in mice. Sci Rep. 2016;6:32405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Heneka MT, Golenbock DT, Latz E. Innate immunity in Alzheimer’s disease. Nat Immunol. 2015;16(3):229–236. [DOI] [PubMed] [Google Scholar]
- 220.Guerreiro R, Bras J. The age factor in Alzheimer’s disease. Genome Med. 2015;7:106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Davies DS, Ma J, Jegathees T, Goldsbury C. Microglia show altered morphology and reduced arborization in human brain during aging and Alzheimer’s disease. Brain Pathol. 2017;27(6):795–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Schuitemaker A, van der Doef TF, Boellaard R, et al. Microglial activation in healthy aging. Neurobiol Aging. 2012;33(6):1067–1072. [DOI] [PubMed] [Google Scholar]
- 223.Koellhoffer EC, McCullough LD, Ritzel RM. Old Maids: Aging and Its Impact on Microglia Function. Int J Mol Sci. 2017;18(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Spittau B Aging Microglia-Phenotypes, Functions and Implications for Age-Related Neurodegenerative Diseases. Front Aging Neurosci. 2017;9:194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Villeda SA, Plambeck KE, Middeldorp J, et al. Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice. Nat Med. 2014;20(6):659–663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Rebo J, Mehdipour M, Gathwala R, et al. A single heterochronic blood exchange reveals rapid inhibition of multiple tissues by old blood. Nat Commun. 2016;7:13363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Gan KJ, Sudhof TC. Specific factors in blood from young but not old mice directly promote synapse formation and NMDA-receptor recruitment. Proc Natl Acad Sci U S A. 2019;116(25):12524–12533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Villeda SA, Luo J, Mosher KI, et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature. 2011;477(7362):90–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Abraham D, Feher J, Scuderi GL, et al. Exercise and probiotics attenuate the development of Alzheimer’s disease in transgenic mice: Role of microbiome. Exp Gerontol. 2019;115:122–131. [DOI] [PubMed] [Google Scholar]
- 230.Lourenco MV, Frozza RL, de Freitas GB, et al. Exercise-linked FNDC5/irisin rescues synaptic plasticity and memory defects in Alzheimer’s models. Nat Med. 2019;25(1):165–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Vukovic J, Colditz MJ, Blackmore DG, Ruitenberg MJ, Bartlett PF. Microglia modulate hippocampal neural precursor activity in response to exercise and aging. J Neurosci. 2012;32(19):6435–6443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Forny-Germano L, De Felice FG, Vieira M. The Role of Leptin and Adiponectin in Obesity-Associated Cognitive Decline and Alzheimer’s Disease. Front Neurosci. 2018;12:1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Maliszewska-Cyna E, Lynch M, Oore JJ, Nagy PM, Aubert I. The Benefits of Exercise and Metabolic Interventions for the Prevention and Early Treatment of Alzheimer’s Disease. Curr Alzheimer Res. 2017;14(1):47–60. [DOI] [PubMed] [Google Scholar]
- 234.Neves G, Cooke SF, Bliss TV. Synaptic plasticity, memory and the hippocampus: a neural network approach to causality. Nat Rev Neurosci. 2008;9(1):65–75. [DOI] [PubMed] [Google Scholar]
- 235.Kandel ER. The molecular biology of memory: cAMP, PKA, CRE, CREB-1, CREB-2, and CPEB. Mol Brain. 2012;5:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Campbell JP, Turner JE. Debunking the Myth of Exercise-Induced Immune Suppression: Redefining the Impact of Exercise on Immunological Health Across the Lifespan. Front Immunol. 2018;9:648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Honjo K, van Reekum R, Verhoeff NP. Alzheimer’s disease and infection: do infectious agents contribute to progression of Alzheimer’s disease? Alzheimers Dement. 2009;5(4):348–360. [DOI] [PubMed] [Google Scholar]
- 238.Ide M, Harris M, Stevens A, et al. Periodontitis and Cognitive Decline in Alzheimer’s Disease. PLoS One. 2016;11(3):e0151081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Holmer J, Eriksdotter M, Schultzberg M, Pussinen PJ, Buhlin K. Association between periodontitis and risk of Alzheimer’s disease, mild cognitive impairment and subjective cognitive decline: A case-control study. J Clin Periodontol. 2018;45(11):1287–1298. [DOI] [PubMed] [Google Scholar]
- 240.Sparks Stein P, Steffen MJ, Smith C, et al. Serum antibodies to periodontal pathogens are a risk factor for Alzheimer’s disease. Alzheimers Dement. 2012;8(3):196–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Kamer AR, Craig RG, Pirraglia E, et al. TNF-alpha and antibodies to periodontal bacteria discriminate between Alzheimer’s disease patients and normal subjects. J Neuroimmunol. 2009;216(1–2):92–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Kamer AR, Craig RG, Dasanayake AP, Brys M, Glodzik-Sobanska L, de Leon MJ. Inflammation and Alzheimer’s disease: possible role of periodontal diseases. Alzheimers Dement. 2008;4(4):242–250. [DOI] [PubMed] [Google Scholar]
- 243.Teixeira FB, Saito MT, Matheus FC, et al. Periodontitis and Alzheimer’s Disease: A Possible Comorbidity between Oral Chronic Inflammatory Condition and Neuroinflammation. Front Aging Neurosci. 2017;9:327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Balin BJ, Gerard HC, Arking EJ, et al. Identification and localization of Chlamydia pneumoniae in the Alzheimer’s brain. Med Microbiol Immunol. 1998;187(1):23–42. [DOI] [PubMed] [Google Scholar]
- 245.Zhan X, Stamova B, Jin LW, DeCarli C, Phinney B, Sharp FR. Gram-negative bacterial molecules associate with Alzheimer disease pathology. Neurology. 2016;87(22):2324–2332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Emery DC, Shoemark DK, Batstone TE, et al. 16S rRNA Next Generation Sequencing Analysis Shows Bacteria in Alzheimer’s Post-Mortem Brain. Front Aging Neurosci. 2017;9:195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Itzhaki RF, Lin WR, Shang D, Wilcock GK, Faragher B, Jamieson GA. Herpes simplex virus type 1 in brain and risk of Alzheimer’s disease. Lancet. 1997;349(9047):241–244. [DOI] [PubMed] [Google Scholar]
- 248.Holmes C, Cunningham C, Zotova E, et al. Systemic inflammation and disease progression in Alzheimer disease. Neurology. 2009;73(10):768–774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Semmler A, Widmann CN, Okulla T, et al. Persistent cognitive impairment, hippocampal atrophy and EEG changes in sepsis survivors. J Neurol Neurosurg Psychiatry. 2013;84(1):62–69. [DOI] [PubMed] [Google Scholar]
- 250.Chou CH, Lee JT, Lin CC, et al. Septicemia is associated with increased risk for dementia: a population-based longitudinal study. Oncotarget. 2017;8(48):84300–84308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Johnson VE, Stewart W, Smith DH. Traumatic brain injury and amyloid-beta pathology: a link to Alzheimer’s disease? Nat Rev Neurosci. 2010;11(5):361–370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Tran HT, LaFerla FM, Holtzman DM, Brody DL. Controlled cortical impact traumatic brain injury in 3xTg-AD mice causes acute intra-axonal amyloid-beta accumulation and independently accelerates the development of tau abnormalities. J Neurosci. 2011;31(26):9513–9525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Tajiri N, Kellogg SL, Shimizu T, Arendash GW, Borlongan CV. Traumatic brain injury precipitates cognitive impairment and extracellular Abeta aggregation in Alzheimer’s disease transgenic mice. PLoS One. 2013;8(11):e78851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Smith DH, Nakamura M, McIntosh TK, et al. Brain trauma induces massive hippocampal neuron death linked to a surge in beta-amyloid levels in mice overexpressing mutant amyloid precursor protein. Am J Pathol. 1998;153(3):1005–1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Plassman BL, Havlik RJ, Steffens DC, et al. Documented head injury in early adulthood and risk of Alzheimer’s disease and other dementias. Neurology. 2000;55(8):1158–1166. [DOI] [PubMed] [Google Scholar]
- 256.Fleminger S, Oliver DL, Lovestone S, Rabe-Hesketh S, Giora A. Head injury as a risk factor for Alzheimer’s disease: the evidence 10 years on; a partial replication. J Neurol Neurosurg Psychiatry. 2003;74(7):857–862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Lobo A, Launer LJ, Fratiglioni L, et al. Prevalence of dementia and major subtypes in Europe: A collaborative study of population-based cohorts. Neurologic Diseases in the Elderly Research Group. Neurology. 2000;54(11 Suppl 5):S4–9. [PubMed] [Google Scholar]
- 258.Smith DH, Johnson VE, Stewart W. Chronic neuropathologies of single and repetitive TBI: substrates of dementia? Nat Rev Neurol. 2013;9(4):211–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Mayeux R, Ottman R, Maestre G, et al. Synergistic effects of traumatic head injury and apolipoprotein-epsilon 4 in patients with Alzheimer’s disease. Neurology. 1995;45(3 Pt 1):555–557. [DOI] [PubMed] [Google Scholar]
- 260.Ramos-Cejudo J, Wisniewski T, Marmar C, et al. Traumatic Brain Injury and Alzheimer’s Disease: The Cerebrovascular Link. EBioMedicine. 2018;28:21–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Edwards G 3rd, Zhao J, Dash PK, Soto C, Moreno-Gonzalez I. Traumatic Brain Injury Induces Tau Aggregation and Spreading. J Neurotrauma. 2020;37(1):80–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Washington PM, Morffy N, Parsadanian M, Zapple DN, Burns MP. Experimental traumatic brain injury induces rapid aggregation and oligomerization of amyloid-beta in an Alzheimer’s disease mouse model. J Neurotrauma. 2014;31(1):125–134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Louveau A, Smirnov I, Keyes TJ, et al. Structural and functional features of central nervous system lymphatic vessels. Nature. 2015;523(7560):337–341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Louveau A, Herz J, Alme MN, et al. CNS lymphatic drainage and neuroinflammation are regulated by meningeal lymphatic vasculature. Nat Neurosci. 2018;21(10):1380–1391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Da Mesquita S, Louveau A, Vaccari A, et al. Functional aspects of meningeal lymphatics in ageing and Alzheimer’s disease. Nature. 2018;560(7717):185–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Bolte Ashley C. H ME, Smirnov Igor, Kovacs Michael A., McKee Celia A., Natale Nick, Ennerfelt Hannah E., Frost Elizabeth L., Lammert Catherine E., Kipnis Jonathan, Lukens John R.. Meningeal lymphatic dysfunction exacerbates traumatic brain injury pathogenesis. 2019. [DOI] [PMC free article] [PubMed]
- 267.Braniste V, Al-Asmakh M, Kowal C, et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med. 2014;6(263):263ra158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Carabotti M, Scirocco A, Maselli MA, Severi C. The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems. Ann Gastroenterol. 2015;28(2):203–209. [PMC free article] [PubMed] [Google Scholar]
- 269.Obrenovich MEM. Leaky Gut, Leaky Brain? Microorganisms. 2018;6(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Kowalski K, Mulak A. Brain-Gut-Microbiota Axis in Alzheimer’s Disease. J Neurogastroenterol Motil. 2019;25(1):48–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Jiang C, Li G, Huang P, Liu Z, Zhao B. The Gut Microbiota and Alzheimer’s Disease. J Alzheimers Dis. 2017;58(1):1–15. [DOI] [PubMed] [Google Scholar]
- 272.Kim MS, Kim Y, Choi H, et al. Transfer of a healthy microbiota reduces amyloid and tau pathology in an Alzheimer’s disease animal model. Gut. 2020;69(2):283–294. [DOI] [PubMed] [Google Scholar]
- 273.Dodiya HB, Kuntz T, Shaik SM, et al. Sex-specific effects of microbiome perturbations on cerebral Abeta amyloidosis and microglia phenotypes. J Exp Med. 2019;216(7):1542–1560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.Cattaneo A, Cattane N, Galluzzi S, et al. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol Aging. 2017;49:60–68. [DOI] [PubMed] [Google Scholar]
- 275.Nagpal R, Neth BJ, Wang S, Craft S, Yadav H. Modified Mediterranean-ketogenic diet modulates gut microbiome and short-chain fatty acids in association with Alzheimer’s disease markers in subjects with mild cognitive impairment. EBioMedicine. 2019;47:529–542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276.Mander BA, Winer JR, Walker MP. Sleep and Human Aging. Neuron. 2017;94(1):19–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Volicer L, Harper DG, Manning BC, Goldstein R, Satlin A. Sundowning and circadian rhythms in Alzheimer’s disease. Am J Psychiatry. 2001;158(5):704–711. [DOI] [PubMed] [Google Scholar]
- 278.Bubu OM, Brannick M, Mortimer J, et al. Sleep, Cognitive impairment, and Alzheimer’s disease: A Systematic Review and Meta-Analysis. Sleep. 2017;40(1). [DOI] [PubMed] [Google Scholar]
- 279.Shokri-Kojori E, Wang GJ, Wiers CE, et al. beta-Amyloid accumulation in the human brain after one night of sleep deprivation. Proc Natl Acad Sci U S A. 2018;115(17):4483–4488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280.Lucey BP, Hicks TJ, McLeland JS, et al. Effect of sleep on overnight cerebrospinal fluid amyloid beta kinetics. Ann Neurol. 2018;83(1):197–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Holth JK, Fritschi SK, Wang C, et al. The sleep-wake cycle regulates brain interstitial fluid tau in mice and CSF tau in humans. Science. 2019;363(6429):880–884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282.Holth J, Patel T, Holtzman DM. Sleep in Alzheimer’s Disease - Beyond Amyloid. Neurobiol Sleep Circadian Rhythms. 2017;2:4–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Patterson BW, Elbert DL, Mawuenyega KG, et al. Age and amyloid effects on human central nervous system amyloid-beta kinetics. Ann Neurol. 2015;78(3):439–453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284.Kaneshwaran K, Olah M, Tasaki S, et al. Sleep fragmentation, microglial aging, and cognitive impairment in adults with and without Alzheimer’s dementia. Sci Adv. 2019;5(12):eaax7331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285.Irwin MR, Opp MR. Sleep Health: Reciprocal Regulation of Sleep and Innate Immunity. Neuropsychopharmacology. 2017;42(1):129–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286.Pike CJ. Sex and the development of Alzheimer’s disease. J Neurosci Res. 2017;95(1–2):671–680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287.Gaignard P, Liere P, Therond P, Schumacher M, Slama A, Guennoun R. Role of Sex Hormones on Brain Mitochondrial Function, with Special Reference to Aging and Neurodegenerative Diseases. Front Aging Neurosci. 2017;9:406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288.Vina J, Lloret A. Why women have more Alzheimer’s disease than men: gender and mitochondrial toxicity of amyloid-beta peptide. J Alzheimers Dis. 2010;20 Suppl 2:S527–533. [DOI] [PubMed] [Google Scholar]
- 289.Song YJ, Li SR, Li XW, et al. The Effect of Estrogen Replacement Therapy on Alzheimer’s Disease and Parkinson’s Disease in Postmenopausal Women: A Meta-Analysis. Front Neurosci. 2020;14:157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290.Jadiya P, Kolmetzky DW, Tomar D, et al. Impaired mitochondrial calcium efflux contributes to disease progression in models of Alzheimer’s disease. Nat Commun. 2019;10(1):3885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291.Chen Y, Zhou Z, Min W. Mitochondria, Oxidative Stress and Innate Immunity. Front Physiol. 2018;9:1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292.Aminzadeh M, Roghani M, Sarfallah A, Riazi GH. TRPM2 dependence of ROS-induced NLRP3 activation in Alzheimer’s disease. Int Immunopharmacol. 2018;54:78–85. [DOI] [PubMed] [Google Scholar]
- 293.Swerdlow RH, Burns JM, Khan SM. The Alzheimer’s disease mitochondrial cascade hypothesis: progress and perspectives. Biochim Biophys Acta. 2014;1842(8):1219–1231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Kerr JS, Adriaanse BA, Greig NH, et al. Mitophagy and Alzheimer’s Disease: Cellular and Molecular Mechanisms. Trends Neurosci. 2017;40(3):151–166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295.Wang X, Wang W, Li L, Perry G, Lee HG, Zhu X. Oxidative stress and mitochondrial dysfunction in Alzheimer’s disease. Biochim Biophys Acta. 2014;1842(8):1240–1247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296.Lautrup S, Lou G, Aman Y, Nilsen H, Tao J, Fang EF. Microglial mitophagy mitigates neuroinflammation in Alzheimer’s disease. Neurochem Int. 2019;129:104469. [DOI] [PubMed] [Google Scholar]
- 297.Fang EF, Hou Y, Palikaras K, et al. Mitophagy inhibits amyloid-beta and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease. Nat Neurosci. 2019;22(3):401–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298.Villa A, Gelosa P, Castiglioni L, et al. Sex-Specific Features of Microglia from Adult Mice. Cell Rep. 2018;23(12):3501–3511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299.Hanamsagar R, Alter MD, Block CS, Sullivan H, Bolton JL, Bilbo SD. Generation of a microglial developmental index in mice and in humans reveals a sex difference in maturation and immune reactivity. Glia. 2017;65(9):1504–1520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300.Stephen TL, Cacciottolo M, Balu D, et al. APOE genotype and sex affect microglial interactions with plaques in Alzheimer’s disease mice. Acta Neuropathol Commun. 2019;7(1):82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301.Lee LC, Goh MQL, Koo EH. Transcriptional regulation of APP by apoE: To boldly go where no isoform has gone before: ApoE, APP transcription and AD: Hypothesised mechanisms and existing knowledge gaps. Bioessays. 2017;39(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302.Konishi H, Kiyama H. Microglial TREM2/DAP12 Signaling: A Double-Edged Sword in Neural Diseases. Front Cell Neurosci. 2018;12:206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303.Zheng H, Cheng B, Li Y, Li X, Chen X, Zhang YW. TREM2 in Alzheimer’s Disease: Microglial Survival and Energy Metabolism. Front Aging Neurosci. 2018;10:395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304.Naj AC, Jun G, Beecham GW, et al. Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer’s disease. Nat Genet. 2011;43(5):436–441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 305.Griciuc A, Serrano-Pozo A, Parrado AR, et al. Alzheimer’s disease risk gene CD33 inhibits microglial uptake of amyloid beta. Neuron. 2013;78(4):631–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306.Lambert JC, Heath S, Even G, et al. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer’s disease. Nat Genet. 2009;41(10):1094–1099. [DOI] [PubMed] [Google Scholar]
- 307.Foster EM, Dangla-Valls A, Lovestone S, Ribe EM, Buckley NJ. Clusterin in Alzheimer’s Disease: Mechanisms, Genetics, and Lessons From Other Pathologies. Front Neurosci. 2019;13:164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 308.Sims R, Hill M, Williams J. The multiplex model of the genetics of Alzheimer’s disease. Nat Neurosci. 2020;23(3):311–322. [DOI] [PubMed] [Google Scholar]
- 309.Kim WS, Weickert CS, Garner B. Role of ATP-binding cassette transporters in brain lipid transport and neurological disease. J Neurochem. 2008;104(5):1145–1166. [DOI] [PubMed] [Google Scholar]
- 310.Jehle AW, Gardai SJ, Li S, et al. ATP-binding cassette transporter A7 enhances phagocytosis of apoptotic cells and associated ERK signaling in macrophages. J Cell Biol. 2006;174(4):547–556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 311.Tan MS, Yu JT, Tan L. Bridging integrator 1 (BIN1): form, function, and Alzheimer’s disease. Trends Mol Med. 2013;19(10):594–603. [DOI] [PubMed] [Google Scholar]
- 312.Wang HF, Wan Y, Hao XK, et al. Bridging Integrator 1 (BIN1) Genotypes Mediate Alzheimer’s Disease Risk by Altering Neuronal Degeneration. J Alzheimers Dis. 2016;52(1):179–190. [DOI] [PubMed] [Google Scholar]
- 313.Zhu XC, Yu JT, Jiang T, Wang P, Cao L, Tan L. CR1 in Alzheimer’s disease. Mol Neurobiol. 2015;51(2):753–765. [DOI] [PubMed] [Google Scholar]
- 314.Yin RH, Yu JT, Tan L. The Role of SORL1 in Alzheimer’s Disease. Mol Neurobiol. 2015;51(3):909–918. [DOI] [PubMed] [Google Scholar]
- 315.Marioni RE, Harris SE, Zhang Q, et al. Correction: GWAS on family history of Alzheimer’s disease. Transl Psychiatry. 2019;9(1):161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 316.Guillonneau C, Bezie S, Anegon I. Immunoregulatory properties of the cytokine IL-34. Cell Mol Life Sci. 2017;74(14):2569–2586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317.Ma J, Yu JT, Tan L. MS4A Cluster in Alzheimer’s Disease. Mol Neurobiol. 2015;51(3):1240–1248. [DOI] [PubMed] [Google Scholar]
- 318.Deming Y, Filipello F, Cignarella F, et al. The MS4A gene cluster is a key modulator of soluble TREM2 and Alzheimer’s disease risk. Sci Transl Med. 2019;11(505). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 319.Tao QQ, Chen YC, Wu ZY. The role of CD2AP in the Pathogenesis of Alzheimer’s Disease. Aging Dis. 2019;10(4):901–907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 320.Karch CM, Goate AM. Alzheimer’s disease risk genes and mechanisms of disease pathogenesis. Biol Psychiatry. 2015;77(1):43–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 321.Efthymiou AG, Goate AM. Late onset Alzheimer’s disease genetics implicates microglial pathways in disease risk. Mol Neurodegener. 2017;12(1):43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 322.Pauls SD, Marshall AJ. Regulation of immune cell signaling by SHIP1: A phosphatase, scaffold protein, and potential therapeutic target. Eur J Immunol. 2017;47(6):932–945. [DOI] [PubMed] [Google Scholar]
- 323.Asanomi Y, Shigemizu D, Miyashita A, et al. A rare functional variant of SHARPIN attenuates the inflammatory response and associates with increased risk of late-onset Alzheimer’s disease. Mol Med. 2019;25(1):20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 324.Song YN, Li JQ, Tan CC, et al. TREML2 Mutation Mediate Alzheimer’s Disease Risk by Altering Neuronal Degeneration. Front Neurosci. 2019;13:455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 325.Miguel Tábuas-Pereira IS, Guerreiro Rita, Brás José. Alzheimer’s Disease Genetics: Review of Novel Loci Associated with Disease. Current Genetic Medicine Reports. 2020. [Google Scholar]
- 326.Steele NZ, Carr JS, Bonham LW, et al. Fine-mapping of the human leukocyte antigen locus as a risk factor for Alzheimer disease: A case-control study. PLoS Med. 2017;14(3):e1002272. [DOI] [PMC free article] [PubMed] [Google Scholar]