IMR Press / JIN / Volume 22 / Issue 6 / DOI: 10.31083/j.jin2206150
Open Access Review
The Pathogenesis in Alzheimer's Disease: TREM2 as a Potential Target
Show Less
1 Department of Physical Education, Henan Normal University, 453007 Xinxiang, Henan, China
2 College of Chemistry, Beijing Normal University, 100875 Beijing, China
*Correspondence: fanyongzhao@cupes.edu.cn (Yongzhao Fan); jiwenliang@bnu.edu.cn (Wenliang Ji)
J. Integr. Neurosci. 2023, 22(6), 150; https://doi.org/10.31083/j.jin2206150
Submitted: 11 May 2023 | Revised: 11 July 2023 | Accepted: 13 July 2023 | Published: 30 October 2023
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Alzheimer’s disease (AD) is ranked as the third-most expensive illness and sixth leading cause of mortality. It is associated with the deposition of extracellular amyloid-β (Aβ) in neural plaques (NPs), as well as intracellular hyperphosphorylated tau proteins that form neurofibrillary tangles (NFTs). As a new target in regulating neuroinflammation in AD, triggering receptor expressed on myeloid cells 2 (TREM2) is highly and exclusively expressed on the microglial surface. TREM2 interacts with adaptor protein DAP12 to initiate signal pathways that mainly dominant microglia phenotype and phagocytosis mobility. Furthermore, TREM2 gene mutations confer increased AD risk, and TREM2 deficiency exhibits more dendritic spine loss around neural plaques. Mechanisms for regulating TREM2 to alleviate AD has evolved as an area of AD research in recent years. Current medications targeting Aβ or tau proteins are unable to reverse AD progression. Emerging evidence implicating neuroinflammation may provide novel insights, as early microglia-related inflammation can be induced decades prior to the commencement of AD-related cognitive damage. Physical exercise can exert a neuroprotective effect over the course of AD progression. This review aims to (1) summarize the pathogenesis of AD and recent updates in the field, (2) assess the concept that AD cognitive impairment is closely correlated with microglia-related inflammation, and (3) review TREM2 functions and its role between exercise and AD, which is likely to be an ideal candidate target.

Keywords
AD
pathogenesis
neuroinflammation
TREM2
1. Introduction

Alzheimer’s disease (AD) is an irreversible and progressive neurological disorder [1, 2]. Various pathological factors are found in the postmortem brains of AD patients, such as abnormal amyloid-β (Aβ) plaques, hyperphosphorylated tau protein, and damaged neurons and synapses [3, 4]. Such lesions affect brain areas important for learning and memory, including the cerebral cortex, striatum, and hippocampus [5]. According to Global Alzheimer’s Information, AD affected 46.8 million individuals globally and is projected to affect 82 million by 2030 [6]. Without effective intervention, this figure will likely triple by 2050 with the aging of the population. Accumulating evidence from clinical investigations demonstrates the absence of effective treatments for curing AD or restricting its progress. In vitro and in vivo studies have shown that Aβ plaques are key drivers in AD pathogenesis [7]. However, findings from unsuccessful partial clinical studies directly targeting the Aβ protein indicate that reducing the Aβ load does not ameliorate cognitive impairment [8, 9]. The challenges in disease diagnosis have undermined the improvements made in the prevention and treatment of AD. The prevention and treatment of AD are impeded by the similarities between AD and other forms of dementia [10]. Many AD experiments have shown that microglia play a key role in regulating inflammation in the central nervous system [11]. Neuroinflammation aggravated by microglia contributes to AD pathogenesis and promotes protein aggregation, implying that microglia may function as a novel target in AD [12].

Triggering receptor expressed on myeloid cells 2 (TREM2) is expressed in myeloid progenitors. It is an innate immune receptor that is located on the microglial membrane. Findings from genome-wide association experiments have revealed rare TREM2 (R47H) mutations that are linked to an increased possibility of developing AD, as TREM2 is one of the most important receptors because of it’s effects on neuronal/microglia health [13]. The type I transmembrane glycoprotein TREM2 communicates with an immune adaptor DNAX activation protein of 12 kDa (DAP12) transmembrane area to facilitate signaling via the immune-receptor tyrosine-based activation motif ( ITAM) domain of DAP12 in migration, phagocytosis, cell survival, and inflammatory cytokine release [14, 15]. TREM2 is also implicated in the etiology of AD as it regulates microglial function and innate immunity [16]. The function of TREM2 in neuroinflammation and its relationship with AD have not been fully elucidated. This review focuses on current understanding of the pathogenesis of AD and the shift from the initial focus on abnormal Aβ plaques and hyperphosphorylated tau proteins to the current concept that AD cognitive impairment is closely correlated with microglia-induced neuroinflammation, suggesting it may be an ideal candidate target. Here, we summarize our current understanding of TREM2 function and its role in AD.

2. AD Pathology

AD is the most widespread age-related neurodegenerative disorder, with no effective cure that can help alleviate the financial and emotional burden of the disease [17, 18]. Studies on the pathogenesis and development of therapies are vital for preventing or alleviating AD symptoms. A number of hypotheses explain AD pathophysiology, including the hypotheses of the Aβ cascade, Aβ-tau cooperative action, and cholinergic and γ-aminobutyric acid (GABA) functions.

2.1 Aβ Cascade Hypothesis

Aβ is found in various assembly forms, such as fibrils, protofibrils, and oligomers [19]. Aβ is non-neurotoxic in its monomeric form. In contrast, protofibrillars and oligomers are regarded as effective blockers of long-term potentiation and strength. Fiber formation is strongly linked to protein misfolding [20].

Aβ induces fatality in neuronal cells, which is the primary cause of AD [21]. Significant evidence has shown that the production of Aβ oligomers (AbOs) in cortical neurons initiates AD [22, 23, 24]. AbOs acting on entorhinal neurons assist in tau oligomer development with progression to associated neurons. This occurs in the hippocampus and subsequently in the subiculum and associated neocortex [25]. The toxicity of tau oligomers can be damaging to neurons, where they are formed and subsequently released into neurons and synapses. Tau proteins are propagated and disseminated from neurons. AbOs may interact with tau oligomers during this phase to enhance neuronal and synaptic dysfunction [26]. Data further demonstrates that Aβ is not a bystander in AD [27]. Recent advances have revealed that extracellular Aβ deposits can be released from the brain via the blood-brain barrier (BBB) and other systems [28]. The structure and function of the BBB are damaged in AD [29]. The destruction of the BBB allows neurotoxic blood-derived debris and microbes to enter the brain, which is closely linked to neuroinflammation and immune responses that can trigger numerous neurodegenerative pathways [30, 31].

2.2 Aβ-tau Cooperated Hypothesis

Tau pathology is a hallmark of AD progression [32]. Tau present in the medial temporal lobe binds and stabilizes the microtubule architecture, where it is normally concentrated. However, hyperphosphorylated tau protein produced in AD dissociates from microtubules and accumulates inside neurons [33]. Once it spreads from the medial temporal lobe to the surrounding neocortex, hyperphosphorylated tau protein can cause cognitive impairment.

Findings from animal models have shown that Aβ spreads via neuronal connections from cell to cell [34, 35]. An increasing body of evidence demonstrates that the synergistic effects of Aβ on tau plays a vital role in AD pathogenesis [36, 37]. Busche et al. [38] recently showed that Aβ and tau interaction promotes neural circuit damage: in vivo multiphoton imaging showed that the combined existence of tau and Aβ pathology in the neocortex is linked to repressed neuronal activity and increase in spine loss and microglia injury. The repression of tau or Aβ pathology alone is not conducive to salvaging functional damage. In addition, the impact of tau and Aβ co-expression on neuronal activity was analyzed in Busche and Angulo’s studies. Their findings [39] showed that extracellular expression of Aβ caused hyper-excitability, whereas tau expression led to activity suppression. The co-expression of Aβ and tau also repressed activity, where the tau phenotype appeared to play a dominant role.

2.3 Cholinergic Hypothesis

Cholinergic neurons have been demonstrated to modulate neuronal circuits in the hippocampus and cortical regions and play an important role in hippocampus-dependent memory [40, 41]. Normally, endogenous nerve growth factor (NGF) is produced by hippocampal and post-synaptic cortical neurons and is expressed through related receptors present on presynaptic cholinergic terminals [42, 43]. Stimulated NGF is then transported from the target regions (hippocampus and cortex) to cholinergic nuclei, consequently initiating cholinergic signaling in these brain areas [44].

Findings from various animal models and human studies have shown that cholinergic neuron degeneration in the basal forebrain is linked to cognitive impairments in AD [45, 46]. The cholinergic hypothesis primarily focuses on the gradual damage of limbic and neocortical cholinergic innervations, which is linked to neurofibrillary degeneration and inefficient axonal transport and signaling [47]. The crosstalk between postsynaptic cortical/hippocampal neurons and presynaptic cholinergic terminals is altered during AD. These variations are related to the accessibility of mature NGF (mNGF) to basal forebrain cholinergic neurons, which subsequently results in cholinergic degeneration in hippocampal and cortical areas [48, 49].

The cholinergic hypothesis has transformed different areas of AD research, from the field of neuropathology to the modern concept of synaptic neurotransmission [50]. It was developed based on three parameters: (1) diminished presynaptic cholinergic indicators in the cerebral cortex [51, 52], (2) the nucleus basalis of Meynert (NBM) in the basal forebrain is a source of cortical cholinergic innervation that undergoes intense neurodegeneration in AD [53, 54] (3) cholinergic antagonists impair memory whereas agonists have a contrasting effect on memory [55].

2.4 GABAergic Hypothesis

GABAergic activity is important for brain development and plasticity. Under normal conditions, GABA is produced in the presynaptic membrane and helps synthesize glutamic acid decarboxylase (GAD) from glutamate. GABA can bind to GABA receptors present on the postsynaptic membrane through the synaptic cleft, which inhibits the post-synaptic neuron [56, 57]. However, in AD, imbalances in GABAergic neurons is associated with several disorders. In the AD brain, Aβ is toxic to GABAergic synapses and promotes the degeneration of axons and synaptic loss. At first, Aβ enhances neural activity, which supports Aβ release. The cycle between neural activity and Aβ can involve Aβ-induced GABAergic terminal loss, which results in impaired cognition and repressed long-term potentiation (LTP). In addition, β-site APP cleaving enzyme 1 (BACE1), Apolipoprotein E4 (APOEϵ4), and TREM2 can induce GABAergic dysfunction [58].

These outcomes have shown that GABAergic dysfunction is related to the E (excitation)/I (inhibition) imbalance in AD [59]. As mentioned Govindpani et al. [60], because of the narrow scope of existing treatments for disease modification, better insights into GABAergic remodeling in AD may help introduce innovative and unique therapeutic options.

3. The Relationship between Microglial Activation and Inflammation in AD

AD pathology involves the clustering of Aβ proteins and hyperphosphorylation of tau protein. However, therapies targeting tau and Aβ have failed in clinic trial. Several researchers have emphasized the significant contribution of neuroinflammation in initial and late AD stages, which is strongly associated with microglia activation [61, 62]. Inflammation during AD is predominantly associated with continuous microglia activation in response to cell death and Aβ plaque deposition [63]. Classical microglial activation appears to contribute to neuronal impairment in neurodegenerative disorders; however, the advantageous characteristics of microglial alternative activation were also observed [64, 65]. In this section, we report findings from an advanced report to highlight the link between microglial activation and inflammation in AD and the critical function of TREM2 in AD progression.

3.1 Neuroinflammation in AD

In recent decades, AD investigations have prioritized the mechanisms related to extracellular neural plaques (NPs) and intracellular neurofibrillary tangles (NFTs). Data from genetic, preclinical, and clinical findings have focused on the neuroinflammatory mediation of the innate immune system [66]. Many neurodegenerative diseases, particularly AD, have been linked to inflammation [67, 68]. Increasing evidence suggests that systemic inflammation plays a key role in AD [69]. Tau-protein tangles and Aβ plaques in the brain trigger cells, such as microglia and astrocytes, to release anti-inflammatory and pro-inflammatory mediators, including reactive oxygen species (ROS), neurotransmitters, chemokines, and cytokines [70, 71]. The release of mediators causes lymphocytes and monocytes to pass through the BBB, which boosts the release of inflammatory components.These findings indicate that inflammation promotes AD progression and accelerates disease progression [72, 73]. Trovato et al. [74] eported that an increase in oxidative stress and altered antioxidant systems cause NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome activation, resulting in cell damage and cognitive impairment.

3.2 Overactivation of Microglia is a Key Contributor to Neuroinflammation in AD Models

Microglia are the primary macrophages and provide basic immunological protection in disorders and injuries of the central nervous system [75, 76]. Plastic cells have dual functions in neuronal damage and healing and can adopt multiple phenotypes [77, 78]. Microglia have a ramified phenotype under normal physiological conditions, characterized by a small cell body and several processes, such as in “resting microglia”. Even in the resting state, microglial processes are dynamic and constantly scan to preserve central nervous system (CNS) cell types with neurons [79]. Evidently, they scan the brain environment continuously and contact synapses by using their fine branches to detect infections and damage in their environment [80].

Microglia, which represent the immune cells of the brain, engage in diverse functional programming, known as polarization, for responding to external stimulating factors [81]. In the activated state, microglial phenotypes are categorized by two major states: classical activation (pro-inflammatory microglia) and alternative activation (anti-inflammatory microglia) [82, 83]. Pro-inflammatory microglia primarily release proinflammatory substances, whereas anti-inflammatory microglia release anti-inflammatory cytokines. When triggered by lipopolysaccharide (LPS), the pro-inflammatory phenotype is acquired by microglia, which leads to neurotoxicity via secreted pro-inflammatory factors such as tumor necrosis factor-α (TNF-α) and Interleukin 1 (IL-1) [83]. In contrast, microglia acquire an anti-inflammatory state via the release of anti-inflammatory cytokines (e.g., Interleukin 4.), which aids neuroprotection and promotes tissue repair, both of which play crucial roles in maintaining the physiological environment [84, 85].

3.3 Microglial Activation Modulation is a Potential Target in AD-Induced Neuroinflammation

In neurodegenerative conditions, especially in AD models, persistently activated microglia may restrict CD3+/CD8+ T-cell entry into the brain. Local macrophages constitute a link between innate and adaptive immunity [86]. In response to Aβ aggregation, including disease progression, the production of proinflammatory cytokines downregulates the expression of Aβ clearance components and promotes Aβ-mediated neurodegeneration and Aβ aggregation [87]. The early activation of microglia during AD progression provides neuroprotection by supporting Aβ clearance. However, pro-inflammatory cytokines promote Aβ aggregation during disease progression. Evidence from multiple animal studies provides strong support for the production of Aβ, which increases AD deficits by upregulating microglial activation. Aβ-excess neurons, as primary proinflammatory factors, implicate the intraneuronal aggregation of Aβ as an important immunological element in AD pathogenesis [88].

Microglia show different phenotypic states in AD modesl, especially under chronic inflammatory conditions [89]. Some receptors (receptor for advanced glycation endproducts [RAGE] and NLRP3) of the proinflammatory microglial phenotype secrete proinflammatory cytokines, such as TNF-α and IL-1β, by triggering a signaling cascade to induce neuronal cell death. Meanwhile, other receptors (the class a macrophage scavenger receptor type I [SR-AI], TREM2) of the M2-like microglia participate in clearing Aβ by stimulating Aβ fibril internalization and synthesizing anti-inflammatory cytokines such as TGF-β, IL-10, and IL-4 [90, 91]. Activated microglia play a key role in enhancing the spreading of tau protein in the presence of Aβ. Aβ in its soluble form, along with additional elements, can activate microglia via microglial surface receptors. Tau is taken up by activated microglia and released into further bioactive types. The released tau can be taken up by neurons and subsequently released into the neuropil, in an activation-dependent manner [92]. Therefore, finding a method to transform pro-inflammatory microglia into anti-inflammatory microglia has a prospective advantage in treating AD.

4. The Function and Role of TREM2 in AD

Receptors located on microglial membranes are composed of soluble and membrane proteins that can receive various stimuli and trigger a series of responses to maintain microglial homeostasis [93]. The receptor TREM2 is primarily expressed on the microglial surface. Substantial evidence has shown that TREM2 has bioactive potential and the ability to connect with ligands, stimulate microglia, and regulate the immune system in AD progression, including proliferation, survival, and phagocytosis. Furthermore, a lack of TREM2 expression has been shown to increase the accumulation of Aβ and induce neuronal death in various AD animal models [94].

4.1 TREM2’s Structure and Signal

TREM2 is an innate immunological receptor found only on the surface of myeloid cells in the brain, such as microglia, macrophages, and monocytes [95, 96]. Mature TREM2 protein weights approximately 40 kDa. In situ hybridization and immunohistochemical labeling have been used to detect the protein [97]. TREM2 has a long ectodomain that interacts with the extracellular setting to control microglial function [98].

Current research indicates that minor changes in soluble TREM2 (sTREM2) levels (approximately 7%–10%), are sufficient for modulating AD risk [99]. According to Piccio et al. [100, 101], AD and other inflammatory central nervous system diseases have considerably higher CSF sTREM2 levels than normal controls. Deming et al. [102] discovered for the first time that, as a vital contributor in TREM2’s biological processes, the membrane-spanning 4-domains subfamily A (MS4A) gene cluster plays a significant role in regulating soluble TREM2 expression as well as AD pathology.

4.2 TREM2’s Function

TREM2 is crucial for cell maturation, proliferation, and survival during development under homeostatic conditions [103, 104]. Multiple TREM2 functions have been identified in the last decade, including the regulation of phagocytosis and modulation of inflammatory signaling [105, 106].

4.2.1 Regulation of Phagocytosis

The CNS, with its greater phagocytic potential, has myeloid cell subsets expressing TREM2 [107, 108]. Knockout animal studies have shown reduced phagocytosis in apoptotic neurons [109]. The overexpression or activation of TREM2 has been shown to increase substrate uptake [110, 111]. According to Yao et al. [112], phagocytosis depending on TREM2 requires the activation of the SYK/PI3K/AKT/PLC pathways. A novel TREM2 agonistic antibody treatment accelerated the clearing of myelin debris from the CNS demyelination model [113].

4.2.2 Modulation of Microglial Activation and Inflammatory Responses

TREM2 is traditionally classified as an anti-inflammatory receptor [114, 115, 116]. Its silencing stimulates an early pro-inflammatory response through the PI3K/NF-κB pathway, which downregulates CD163 expression, leading to virus repression [117]. TREM2 is vital for regulating the in vivo activation of microglia in response to damaged tissue [118]. TREM2 increases the survival of microglia by stimulating the Wnt/-catenin pathway, and Wnt/-catenin signaling can be restored when TREM2 activity is interrupted [119]. More recently, studies have shown that the TREM2-APOE pathway is the primary regulator of microglial phenotypic changes in neurodegenerative disorders and may serve as a target to restore homeostatic microglia [120].

4.3 TREM2 in AD

Genetic variations in TREM2 have been linked to several neurodegenerative diseases [121, 122]. Recent research has shown that microglial TREM2 expression is associated with AD pathology. The most common AD-associated TREM2 variant is rs75932628, a single-nucleotide polymorphism encoding an arginine-to-histidine missense substitution at amino acid 47 (R47H), which significantly increased sporadic AD incidence and impaired phospholipid ligand binding [123, 124]. Recent studies have proposed that R62H and R47H are partial loss-of-function variants and predominantly lead to minimized affinity for TREM2 ligands [125]. In contrast, upregulated TREM2 signaling can lead to several possibilities for immune-linked AD treatments [126, 127]. The regulation of TREM2 in AD represent a novel therapeutic approach that is currently under investigation in clinical trials [128, 129]. Therefore, microglial TREM2 is a potential therapeutic target for ameliorating neurodegeneration disease. However, caution should be exercised when targeting TREM2 as a therapeutic entry point for AD until its involvement in tau aggregation and propagation is better understood [130].

4.3.1 Interaction with Aβ and Tau Proteins

The accumulation of neurotoxic forms of tau and Aβ is a pathological characteristic of AD. TREM2 is an Aβ receptor that transduces AD-linked pathological and physiological effects. TREM2 is specifically upregulated in plaque-associated microglia [131]. Karanfilian et al. [132] and Singh et al. [133] reported that TREM2 is involved in modulating amyloid plaque deposition and removing amyloid plaques in AD. Findings from numerous studies have shown that plaque-associated microglia may regulate the pathogenesis of Aβ via plaque phagocytosis [134, 135]. TREM2-dependent microglia activity has been demonstrated to reduce amyloid plaque development by increasing Aβ phagocytosis [136]. Kim et al. [137] demonstrated that TREM2 enhances the phagocytosis of Aβ via the upregulation of C/EBPα depending on expression of CD36 in the microglia, which is necessary for protecting against memory loss and improving learning in cases of AD.

Recent evidence demonstrates that TREM2 insufficiency has varying effects on AD progression and initially suppresses amyloid pathology. However, the pathology is exacerbated in later disease stages [138]. Furthermore, deficiency of microglial TREM2 leads to heightened tau pathology coupled with widespread increases in activated neuronal stress kinases [139]. Tau is a neuronal intracellular protein. TREM2 insufficiency enhances the hyperphosphorylation of tau, even in the preliminary stages of AD and other neurodegenerative diseases [140]. Jiang T et al. [141] described that suppressing brain TREM2 expression increased tau pathology, a phenomenon that can be linked to neuroinflammation-induced tau kinase hyperactivation. The authors also pointed out that TREM2 suppresses kinase activity through tau by limiting neuroinflammation and therefore protects against tau pathology.

4.3.2 Modulation of Inflammatory Responses and Microglia Activation

An increase in inflammation and the classical activation of microglia are associated with AD [142, 143]. Human and in vivo studies have revealed that the NLRP3 inflammasome is triggered in AD and ASC specks are indicated in the amyloid plaque core [144]. Some studies have shown that TREM2 has anti-inflammatory functions. In cell lines, TREM2 insufficiency enhanced levels of pro-inflammatory mediators, such as TNF-α, IL-6, and IL-1 [145]. TREM2 anti-inflammatory activities can also be facilitated by the C-terminal fragment of TREM2 [146].

Furthermore, a critical function of TREM2 is to modulate microglial function in the CNS [147, 148]. A decrease in microglia surrounding plaque coverage was shown in TREM2-deficient mice, especially in plaques with higher volume [149]. In an AD model, TREM2-mediated early microglial responses reduced amyloid plaque transport and toxicity [150]. A genomic transgene-guided enhancement in TREM2 expression was shown to reprogram the responsivity of microglia and improve behavioral and neuropathological symptoms in AD rodent experients [151]. Recent reports suggest that TREM2 is an Aβ receptor that mediates function in microglia [152]. Zhao et al. [153] further demonstrated that TREM2/Aβ interaction mediates the downstream TREM2/DAP12 signaling pathway, thereby inducing the degradation of Aβ protein around microglia and microglial activation. Recent data have corroborated these findings by revealing that TREM2 promotes microglial survival and maintains microglial responses to Aβ [154]. In addition, TREM2 controls microglial function by modulating cellular energy and biosynthetic metabolism in an AD model [155].

4.3.3 Participation in the Formation of Dendritic Spines

Dendritic spines are important for forming neuronal connection and signal transmission in the nervous system and can alter the motility, density, and morphology of neurons in relatively shorter periods [156]. The characteristics of dendritic spines have attracted attention in investigations on neurobiological behavioral platforms. For instance, the structural aspects of dendritic spines are linked to variations in learning and memory, synaptic efficiency, and other cognitive functions [157]. Reportedly, microglia reshape synapses through presynaptic nourishment and spinal head filamentous foot induction [158].

TREM2 is crucial for microglia-assisted synaptic refinement during the preliminary stages of brain development. A lack of TREM2 expression causes ineffective synapse pruning, increases excitatory neurotransmission, and minimizes long-range functional connectivity [159]. Maturation studies have shown that variations in the structure and activity of synapses and dendrites are preliminary and critical occurrences in the pathogenesis of AD neurodegenerative procedures [160]. Aβ-induced changes in dendritic spine pathology has also been observed in the preliminary stages of AD [161]. In vivo dendritic spine analysis revealed that neuroinflammation alters the structural plasticity of dendritic spines, which can be ameliorated by anti-inflammatory drugs [162]. TREM2 deletion aggravated the loss of dendritic spines and axons in a transgenic AD mouse model. This indicates that serious neuron damage is more commonly a result of TREM2 insufficiency than of amyloid plaque load.

4.4 The Role of TREM2 in the Connection between Exercise and AD

There is plenty of literature demonstrating that regular physical exercise may slow disease progression or ameliorate symptoms in AD by increasing cerebrospinal biomarkers [163] and cardiovascular fitness [164], as well as decreasing AD-related biomarkers [165]. In addition, various animal models have demonstrated that exercise exerts neuroprotective effects on cognition in AD, such as object recognition memory [166], spatial learning and memory [167], and anxiety behavior [168]. Dao AT et al. [169] demonstrated that AD-impaired basal synaptic transmission and suppression of early-phase long-term potentiation in the dentate gyrus was prevented by prior moderate treadmill exercise. Using a transgenic model, Mu L et al. [170] demonstrated that strengthening structural synaptic plasticity may represent a potential mechanism by which treadmill exercise prevents decline in spatial learning and memory and synapse loss in 3 × Tg-AD mice. Liu Y et al. [171] reported that short-term resistance exercise inhibits neuroinflammation and attenuates neuropathological changes in AD mice. Lu Y et al. [172] demonstrated that treadmill exercise is neuroprotective and regulates microglial polarization and oxidative stress in AD model. Furthermore, pharmacological mimetics of exercise, such as by enhancing adult hippocampal neurogenesis (AHN) and elevating brain derived neurotrophic factor (BDNF) levels, may improve cognition in AD. Furthermore, when applied at early stages of AD, these mimetics may protect against subsequent neuronal cell death [173].

The interaction between microglia and neurons, mediated by TREM2, has been shown to contribute to AD pathogenesis. Therefore, the regulation of TREM2 to alleviate AD has garnered major interest in AD research. Recently, findings from an animal study showed that upregulation of TREM2 ameliorated inflammatory responses, neuronal injury, and cognitive deficits [174, 175]. More importantly, exercise can be viewed as a safe and economic option for improving cognitive performance in both normal and diseased states, including AD. However, few studies have revealed the critical role of TREM2 in exercise and AD.

Currently, the mechanism underlying the exercise-induced neuroprotective effect on cognitive function associated with TREM2 in an AD model can be illustrated from three major findings. First, exercise has been shown to regulate microglial function through TREM2 regulation in the brain. Improvement in recognition memory in an AD rat model is linked to the upregulation of the hippocampal TREM2/DAP12 pathway, which can inhibit excessive microglial activation as well as neuroinflammation [176]. Recent research using the APP/PS1 mouse model demonstrated that long-term running inhibits TREM2 shedding to maintain the levels of TREM2 protein as well as microglial metabolic activity [177]. Second, exercise can increase dendritic spine density by modulating TREM2 expression. TREM2 deficiency was shown to exacerbate dendritic spine loss in an AD mouse model [162]. In contrast, Mu et al. [170] showed that exercise protects memory function by enhancing dendritic spine density in the brain in a 3 × Tg-AD mouse model. Thus, it was widely speculated that exercise modulates TREM2 expression to protect dendritic spines in AD models. However, the in vivo/in vitro evidence should be validated further to confirm this hypothesis. Third, the enhancing effect of physical exercise onsTREM2 was measured in the cerebrospinal fluid of patients with AD. The experiments performed by Jensen et al. [178] indicated that the levels of sTREM2 in the cerebrospinal fluid of patients with AD increased with physical exercise intervention.

5. Conclusions

The pathology of AD is well-studied. The demand for AD prevention or treatment methods has increased owing to more than 15 years of failure in clinical studies. Fortunately, research on AD is currently underway. Emerging evidence implicating TREM2 can provide new insights into AD diagnostics and treatments because early TREM2-participated microglial inflammation may be present decades before the onset of AD-related cognitive impairments. TREM2 deficiency is closely associated with AD and other neurodegenerative disorders. In contrast, TREM2 upregulation in response to exercise can ameliorate AD-related neuropathological changes. Further studies are required to elucidate the protective mechanism of TREM2 in AD.

Author Contributions

GTL, LLZ and WLJ designed the research study. YZF conducted a literature review and revised the manuscript. YZF and WLJ supervised the study. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This research received no external funding.

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Shi Z, Zhang K, Zhou H, Jiang L, Xie B, Wang R, et al. Increased miR-34c mediates synaptic deficits by targeting synaptotagmin 1 through ROS-JNK-p53 pathway in Alzheimer’s Disease. Aging Cell. 2020; 19: e13125.
[2]
Weiner MW, Veitch DP, Aisen PS, Beckett LA, Cairns NJ, Green RC, et al. The Alzheimer’s Disease Neuroimaging Initiative: a review of papers published since its inception. Alzheimer’s & Dementia: the Journal of the Alzheimer’s Association. 2013; 9: e111–94.
[3]
Small SA, Duff K. Linking Abeta and tau in late-onset Alzheimer’s disease: a dual pathway hypothesis. Neuron. 2008; 60: 534–542.
[4]
Polanco JC, Li C, Bodea LG, Martinez-Marmol R, Meunier FA, Götz J. Amyloid-β and tau complexity - towards improved biomarkers and targeted therapies. Nature Reviews. Neurology. 2018; 14: 22–39.
[5]
Weiner MW, Veitch DP, Aisen PS, Beckett LA, Cairns NJ, Green CR, et al. Recent publications from the Alzheimer’s Disease Neuroimaging Initiative: Reviewing progress toward improved AD clinical trials. Alzheimers Dementia. 2017; 13: e1-e85.
[6]
Guzman-Martinez L, Maccioni RB, Farías GA, Fuentes P, Navarrete LP. Biomarkers for Alzheimer’s Disease. Current Alzheimer Research. 2019; 16: 518–528.
[7]
Zhang F, Gannon M, Chen Y, Yan S, Zhang S, Feng W, et al. β-amyloid redirects norepinephrine signaling to activate the pathogenic GSK3β/tau cascade. Science Translational Medicine. 2020; 12: eaay6931.
[8]
Hammond TC, Xing X, Wang C, Ma D, Nho K, Crane PK, et al. β-amyloid and tau drive early Alzheimer’s disease decline while glucose hypometabolism drives late decline. Communications Biology. 2020; 3: 352.
[9]
Hemonnot AL, Hua J, Ulmann L, Hirbec H. Microglia in Alzheimer Disease: Well-Known Targets and New Opportunities. Frontiers in Aging Neuroscience. 2019; 11: 233.
[10]
Livingston G, Huntley J, Sommerlad A, Ames D, Ballard C, Banerjee S, et al. Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. Lancet (London, England). 2020; 396: 413–446.
[11]
Iqubal A, Ahmed M, Iqubal MK, Pottoo FH, Haque SE. Polyphenols as Potential Therapeutics for Pain and Inflammation in Spinal Cord Injury. Current Molecular Pharmacology. 2021; 14: 714–730.
[12]
Guo T, Zhang D, Zeng Y, Huang TY, Xu H, Zhao Y. Molecular and cellular mechanisms underlying the pathogenesis of Alzheimer’s disease. Molecular Neurodegeneration. 2020; 15: 40.
[13]
Jonsson T, Stefansson H, Steinberg S, Jonsdottir I, Jonsson PV, Snaedal J, et al. Variant of TREM2 associated with the risk of Alzheimer’s disease. The New England Journal of Medicine. 2013; 368: 107–116.
[14]
Lessard CB, Malnik SL, Zhou Y, Ladd TB, Cruz PE, Ran Y, et al. High-affinity interactions and signal transduction between Aβ oligomers and TREM2. EMBO Molecular Medicine. 2018; 10: e9027.
[15]
Peng Q, Malhotra S, Torchia JA, Kerr WG, Coggeshall KM, Humphrey MB. TREM2- and DAP12-dependent activation of PI3K requires DAP10 and is inhibited by SHIP1. Science Signaling. 2010; 3: ra38.
[16]
Yeh FL, Hansen DV, Sheng M. TREM2, Microglia, and Neurodegenerative Diseases. Trends in Molecular Medicine. 2017; 23: 512–533.
[17]
Chen XQ, Mobley WC. Exploring the Pathogenesis of Alzheimer Disease in Basal Forebrain Cholinergic Neurons: Converging Insights From Alternative Hypotheses. Frontiers in Neuroscience. 2019; 13: 446.
[18]
Zholos AV, Moroz OF, Storozhuk MV. Curcuminoids and Novel Opportunities for the Treatment of Alzheimer’s Disease: Which Molecules are Actually Effective? Current Molecular Pharmacology. 2019; 12: 12–26.
[19]
LaFerla FM, Green KN, Oddo S. Intracellular amyloid-beta in Alzheimer’s disease. Nature Reviews. Neuroscience. 2007; 8: 499–509.
[20]
Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer’s disease. The Lancet. Neurology. 2015; 14: 388–405.
[21]
Li M, Chen L, Lee DHS, Yu LC, Zhang Y. The role of intracellular amyloid beta in Alzheimer’s disease. Progress in Neurobiology. 2007; 83: 131–139.
[22]
Piccialli I, Tedeschi V, Caputo L, Amato G, De Martino L, De Feo V, et al. The Antioxidant Activity of Limonene Counteracts Neurotoxicity Triggered byAβ1-42 Oligomers in Primary Cortical Neurons. Antioxidants (Basel, Switzerland). 2021; 10: 937.
[23]
Kar S. Role of amyloid β peptides in the regulation of central cholinergic function and its relevance to Alzheimer’s disease pathology. Drug Development Research. 2002; 56: 248–263.
[24]
Ferreiro E, Ferreira I, Cardoso J, Oliveira C, Pereira C, Rego A. P3‐200: Mitochondria control NMDA receptor‐dependent calcium rise evoked by amyloid‐β 1‐42 oligomers in rat cortical neurons. Alzheimer’s & Dementia. 2011; 7: S580.
[25]
Welikovitch LA, Do Carmo S, Maglóczky Z, Malcolm JC, Lőke J, Klein WL, et al. Early intraneuronal amyloid triggers neuron-derived inflammatory signaling in APP transgenic rats and human brain. Proceedings of the National Academy of Sciences of the United States of America. 2020; 117: 6844–6854.
[26]
Chen WT, Lu A, Craessaerts K, Pavie B, Sala Frigerio C, Corthout N, et al. Spatial Transcriptomics and In Situ Sequencing to Study Alzheimer’s Disease. Cell. 2020; 182: 976–991.e19.
[27]
Masters CL, Bateman R, Blennow K, Rowe CC, Sperling RA, Cummings JL. Alzheimer’s disease. Nature Reviews. Disease Primers. 2015; 1: 15056.
[28]
Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV. Blood-Brain Barrier: From Physiology to Disease and Back. Physiological Reviews. 2019; 99: 21–78.
[29]
Tarasoff-Conway JM, Carare RO, Osorio RS, Glodzik L, Butler T, Fieremans E, et al. Clearance systems in the brain–implications for Alzheimer diseaser. Nature Reviews. Neurology. 2016; 12: 248.
[30]
Sweeney MD, Sagare AP, Zlokovic BV. Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nature Reviews. Neurology. 2018; 14: 133–150.
[31]
Chen XQ, Mobley WC. Alzheimer Disease Pathogenesis: Insights From Molecular and Cellular Biology Studies of Oligomeric Aβ and Tau Species. Frontiers in Neuroscience. 2019; 13: 659.
[32]
He Z, Guo JL, McBride JD, Narasimhan S, Kim H, Changolkar L, et al. Amyloid-β plaques enhance Alzheimer’s brain tau-seeded pathologies by facilitating neuritic plaque tau aggregation. Nature Medicine. 2018; 24: 29–38.
[33]
Shahpasand K, Uemura I, Saito T, Asano T, Hata K, Shibata K, et al. Regulation of mitochondrial transport and inter-microtubule spacing by tau phosphorylation at the sites hyperphosphorylated in Alzheimer’s disease. The Journal of Neuroscience: the Official Journal of the Society for Neuroscience. 2012; 32: 2430–2441.
[34]
Vogel JW, Iturria-Medina Y, Strandberg OT, Smith R, Levitis E, Evans AC, et al. Spread of pathological tau proteins through communicating neurons in human Alzheimer’s disease. Nature Communications. 2020; 11: 2612.
[35]
Kametani F, Hasegawa M. Reconsideration of Amyloid Hypothesis and Tau Hypothesis in Alzheimer’s Disease. Frontiers in Neuroscience. 2018; 12: 25.
[36]
Bennett RE, DeVos SL, Dujardin S, Corjuc B, Gor R, Gonzalez J, et al. Enhanced Tau Aggregation in the Presence of Amyloid β. The American Journal of Pathology. 2017; 187: 1601–1612.
[37]
Busche MA, Wegmann S, Dujardin S, Commins C, Schiantarelli J, Klickstein N, et al. Tau impairs neural circuits, dominating amyloid-β effects, in Alzheimer models in vivo. Nature Neuroscience. 2019; 22: 57–64.
[38]
Busche MA, Hyman BT. Synergy between amyloid-β and tau in Alzheimer’s disease. Nature Neuroscience. 2020; 23: 1183–1193.
[39]
Angulo SL, Orman R, Neymotin SA, Liu L, Buitrago L, Cepeda-Prado E, et al. Tau and amyloid-related pathologies in the entorhinal cortex have divergent effects in the hippocampal circuit. Neurobiology of Disease. 2017; 108: 261–276.
[40]
Liu X, Carter AG. Ventral Hippocampal Inputs Preferentially Drive Corticocortical Neurons in the Infralimbic Prefrontal Cortex. The Journal of Neuroscience: the Official Journal of the Society for Neuroscience. 2018; 38: 7351–7363.
[41]
Haam J, Zhou J, Cui G, Yakel JL. Septal cholinergic neurons gate hippocampal output to entorhinal cortex via oriens lacunosum moleculare interneurons. Proceedings of the National Academy of Sciences of the United States of America. 2018; 115: E1886–E1895.
[42]
Eyjolfsdottir H, Eriksdotter M, Linderoth B, Lind G, Juliusson B, Kusk P, et al. Targeted delivery of nerve growth factor to the cholinergic basal forebrain of Alzheimer’s disease patients: application of a second-generation encapsulated cell biodelivery device. Alzheimer’s Research & Therapy. 2016; 8: 30.
[43]
Mitra S, Behbahani H, Eriksdotter M. Innovative Therapy for Alzheimer’s Disease-With Focus on Biodelivery of NGF. Frontiers in Neuroscience. 2019; 13: 38.
[44]
Eriksdotter-Jönhagen M, Linderoth B, Lind G, Aladellie L, Almkvist O, Andreasen N, et al. Encapsulated cell biodelivery of nerve growth factor to the Basal forebrain in patients with Alzheimer’s disease. Dementia and Geriatric Cognitive Disorders. 2012; 33: 18–28.
[45]
Fernández-Cabello S, Kronbichler M, Van Dijk KRA, Goodman JA, Spreng RN, Schmitz TW, et al. Basal forebrain volume reliably predicts the cortical spread of Alzheimer’s degeneration. Brain: a Journal of Neurology. 2020; 143: 993–1009.
[46]
Schmitz TW, Mur M, Aghourian M, Bedard MA, Spreng RN, Alzheimer’s Disease Neuroimaging Initiative. Longitudinal Alzheimer’s Degeneration Reflects the Spatial Topography of Cholinergic Basal Forebrain Projections. Cell Reports. 2018; 24: 38–46.
[47]
Hampel H, Mesulam MM, Cuello AC, Farlow MR, Giacobini E, Grossberg GT, et al. The cholinergic system in the pathophysiology and treatment of Alzheimer’s disease. Brain: a Journal of Neurology. 2018; 141: 1917–1933.
[48]
Baker-Nigh A, Vahedi S, Davis EG, Weintraub S, Bigio EH, Klein WL, et al. Neuronal amyloid-β accumulation within cholinergic basal forebrain in ageing and Alzheimer’s disease. Brain: a Journal of Neurology. 2015; 138: 1722–1737.
[49]
Mesulam MM, Lalehzari N, Rahmani F, Ohm D, Shahidehpour R, Kim G, et al. Cortical cholinergic denervation in primary progressive aphasia with Alzheimer pathology. Neurology. 2019; 92: e1580–e1588.
[50]
Ashford JW. Treatment of Alzheimer’s disease: the legacy of the cholinergic hypothesis, neuroplasticity, and future directions. Journal of Alzheimer’s Disease. 2015; 47: 149–156.
[51]
Bowen DM, Smith CB, White P, Davison AN. Neurotransmitter-related enzymes and indices of hypoxia in senile dementia and other abiotrophies. Brain: a Journal of Neurology. 1976; 99: 459–496.
[52]
Davies P, Maloney AJ. Selective loss of central cholinergic neurons in Alzheimer’s disease. Lancet (London, England). 1976; 2: 1403.
[53]
Mesulam M. A horseradish peroxidase method for the identification of the efferents of acetyl cholinesterase-containing neurons. The Journal of Histochemistry and Cytochemistry: Official Journal of the Histochemistry Society. 1976; 24: 1281–1285.
[54]
Whitehouse PJ, Price DL, Clark AW, Coyle JT, DeLong MR. Alzheimer disease: evidence for selective loss of cholinergic neurons in the nucleus basalis. Annals of Neurology. 1981; 10: 122–126.
[55]
Drachman DA, Leavitt J. Human memory and the cholinergic system. A relationship to aging? Archives of Neurology. 1974; 30: 113–121.
[56]
Root DH, Zhang S, Barker DJ, Miranda-Barrientos J, Liu B, Wang HL, et al. Selective Brain Distribution and Distinctive Synaptic Architecture of Dual Glutamatergic-GABAergic Neurons. Cell Reports. 2018; 23: 3465–3479.
[57]
Tremblay R, Lee S, Rudy B. GABAergic Interneurons in the Neocortex: From Cellular Properties to Circuits. Neuron. 2016; 91: 260–292.
[58]
Bi D, Wen L, Wu Z, Shen Y. GABAergic dysfunction in excitatory and inhibitory (E/I) imbalance drives the pathogenesis of Alzheimer’s disease. Alzheimer’s & Dementia: the Journal of the Alzheimer’s Association. 2020; 16: 1312–1329.
[59]
Maestú F, Haan WD, Busche MA, DeFelipe J. Neuronal excitation/inhibition imbalance: core element of a translational perspective on Alzheimer pathophysiology. Ageing Research Reviews. 2021; 69: 101372.
[60]
Govindpani K, Calvo-Flores Guzmán B, Vinnakota C, Waldvogel HJ, Faull RL, Kwakowsky A. Towards a Better Understanding of GABAergic Remodeling in Alzheimer’s Disease. International Journal of Molecular Sciences. 2017; 18: 1813.
[61]
Hopperton KE, Mohammad D, Trépanier MO, Giuliano V, Bazinet RP. Markers of microglia in post-mortem brain samples from patients with Alzheimer’s disease: a systematic review. Molecular Psychiatry. 2018; 23: 177–198.
[62]
Spangenberg EE, Green KN. Inflammation in Alzheimer’s disease: Lessons learned from microglia-depletion models. Brain, Behavior, and Immunity. 2017; 61: 1–11.
[63]
Madore C, Yin Z, Leibowitz J, Butovsky O. Microglia, Lifestyle Stress, and Neurodegeneration. Immunity. 2020; 52: 222–240.
[64]
Baufeld C, O’Loughlin E, Calcagno N, Madore C, Butovsky O. Differential contribution of microglia and monocytes in neurodegenerative diseases. Journal of Neural Transmission (Vienna, Austria: 1996). 2018; 125: 809–826.
[65]
Mendsaikhan A, Tooyama I, Walker DG. Microglial Progranulin: Involvement in Alzheimer’s Disease and Neurodegenerative Diseases. Cells. 2019; 8: 230.
[66]
Ardura-Fabregat A, Boddeke EWGM, Boza-Serrano A, Brioschi S, Castro-Gomez S, Ceyzériat K, et al. Targeting Neuroinflammation to Treat Alzheimer’s Disease. CNS Drugs. 2017; 31: 1057–1082.
[67]
Irwin MR, Vitiello MV. Implications of sleep disturbance and inflammation for Alzheimer’s disease dementia. The Lancet. Neurology. 2019; 18: 296–306.
[68]
Gammon K. Inflammation: A complex problem. Nature. 2013; 502: S86–S87.
[69]
Senior K. Inflammation speeds up cognitive decline in AD. Nature Reviews Neurology. 2009; 5: 577–577.
[70]
Holmes C. Review: systemic inflammation and Alzheimer’s disease. Neuropathology and Applied Neurobiology. 2013; 39: 51–68.
[71]
Meraz-Ríos MA, Toral-Rios D, Franco-Bocanegra D, Villeda-Hernández J, Campos-Peña V. Inflammatory process in Alzheimer’s Disease. Frontiers in Integrative Neuroscience. 2013; 7: 59.
[72]
McGeer EG, McGeer PL. Chronic inflammation in Alzheimer’s disease offers therapeutic opportunities. Expert Review of Neurotherapeutics. 2001; 1: 53–60.
[73]
Zimmer ER, Leuzy A, Benedet AL, Breitner J, Gauthier S, Rosa-Neto P. Tracking neuroinflammation in Alzheimer’s disease: the role of positron emission tomography imaging. Journal of Neuroinflammation. 2014; 11: 120.
[74]
Trovato Salinaro A, Pennisi M, Di Paola R, Scuto M, Crupi R, Cambria MT, et al. Neuroinflammation and neurohormesis in the pathogenesis of Alzheimer’s disease and Alzheimer-linked pathologies: modulation by nutritional mushrooms. Immunity & Ageing: i & a. 2018; 15: 8.
[75]
Moriyama M, Nishimura Y, Kurebayashi R, Minamihata T, Kawabe K, Takano K, et al. Acetate Suppresses Lipopolysaccharide-stimulated Nitric Oxide Production in Primary Rat Microglia but not in BV-2 Microglia Cells. Current Molecular Pharmacology. 2021; 14: 253–260.
[76]
Nayak D, Roth TL, McGavern DB. Microglia development and function. Annual Review of Immunology. 2014; 32: 367–402.
[77]
Kettenmann H. Neuroscience: the brain’s garbage men. Nature. 2007; 446: 987–989.
[78]
Brown GC, Neher JJ. Microglial phagocytosis of live neurons. Nature Reviews. Neuroscience. 2014; 15: 209–216.
[79]
Michell-Robinson MA, Touil H, Healy LM, Owen DR, Durafourt BA, Bar-Or A, et al. Roles of microglia in brain development, tissue maintenance and repair. Brain: a Journal of Neurology. 2015; 138: 1138–1159.
[80]
Kettenmann H, Kirchhoff F, Verkhratsky A. Microglia: new roles for the synaptic stripper. Neuron. 2013; 77: 10–18.
[81]
Zeng F, Wu Y, Li X, Ge X, Guo Q, Lou X, et al. Custom-Made Ceria Nanoparticles Show a Neuroprotective Effect by Modulating Phenotypic Polarization of the Microglia. Angewandte Chemie (International Ed. in English). 2018; 57: 5808–5812.
[82]
De Biase LM, Schuebel KE, Fusfeld ZH, Jair K, Hawes IA, Cimbro R, et al. Local Cues Establish and Maintain Region-Specific Phenotypes of Basal Ganglia Microglia. Neuron. 2017; 95: 341–356.e6.
[83]
Howes OD, McCutcheon R. Inflammation and the neural diathesis-stress hypothesis of schizophrenia: a reconceptualization. Translational Psychiatry. 2017; 7: e1024.
[84]
Subramaniam SR, Federoff HJ. Targeting Microglial Activation States as a Therapeutic Avenue in Parkinson’s Disease. Frontiers in Aging Neuroscience. 2017; 9: 176.
[85]
Zhang L, Zhang J, You Z. Switching of the Microglial Activation Phenotype Is a Possible Treatment for Depression Disorder. Frontiers in Cellular Neuroscience. 2018; 12: 306.
[86]
Unger MS, Schernthaner P, Marschallinger J, Mrowetz H, Aigner L. Microglia prevent peripheral immune cell invasion and promote an anti-inflammatory environment in the brain of APP-PS1 transgenic mice. Journal of Neuroinflammation. 2018; 15: 274.
[87]
Hickman SE, Allison EK, El Khoury J. Microglial dysfunction and defective beta-amyloid clearance pathways in aging Alzheimer’s disease mice. The Journal of Neuroscience: the Official Journal of the Society for Neuroscience. 2008; 28: 8354–8360.
[88]
Van Zeller M, Dias D, Sebastião AM, Valente CA. NLRP3 Inflammasome: A Starring Role in Amyloid-β- and Tau-Driven Pathological Events in Alzheimer’s Disease. Journal of Alzheimers Disease 2021; 83: 939–961.
[89]
Serrano-Pozo A, Gómez-Isla T, Growdon JH, Frosch MP, Hyman BT. A phenotypic change but not proliferation underlies glial responses in Alzheimer disease. The American Journal of Pathology. 2013; 182: 2332–2344.
[90]
Dong Y, Li X, Cheng J, Hou L. Drug Development for Alzheimer’s Disease: Microglia Induced Neuroinflammation as a Target? International Journal of Molecular Sciences. 2019; 20: 558.
[91]
Doens D, Fernández PL. Microglia receptors and their implications in the response to amyloid β for Alzheimer’s disease pathogenesis. Journal of Neuroinflammation. 2014; 11: 48.
[92]
Wang Y, Cella M, Mallinson K, Ulrich JD, Young KL, Robinette ML, et al. TREM2 lipid sensing sustains the microglial response in an Alzheimer’s disease model. Cell. 2015; 160: 1061–1071.
[93]
Hu X, Liou AKF, Leak RK, Xu M, An C, Suenaga J, et al. Neurobiology of microglial action in CNS injuries: receptor-mediated signaling mechanisms and functional roles. Progress in Neurobiology. 2014; 119–120: 60–84.
[94]
Li Y, Jiang J, Tang Q, Tian H, Wang S, Wang Z, et al. Microglia TREM2: A Potential Role in the Mechanism of Action of Electroacupuncture in an Alzheimer’s Disease Animal Model. Neural Plasticity. 2020; 2020: 8867547.
[95]
Gratuze M, Leyns CEG, Holtzman DM. New insights into the role of TREM2 in Alzheimer’s disease. Molecular Neurodegeneration. 2018; 13: 66.
[96]
Tanzi RE. TREM2 and Risk of Alzheimer’s Disease–Friend or Foe? The New England Journal of Medicine. 2015; 372: 2564–2565.
[97]
Hickman S, Izzy S, Sen P, Morsett L, El Khoury J. Microglia in neurodegeneration. Nature Neuroscience. 2018; 21: 1359–1369.
[98]
Yang J, Fu Z, Zhang X, Xiong M, Meng L, Zhang Z. TREM2 ectodomain and its soluble form in Alzheimer’s disease. Journal of Neuroinflammation. 2020; 17: 204.
[99]
Del-Aguila JL, Benitez BA, Li Z, Dube U, Mihindukulasuriya KA, Budde JP, et al. TREM2 brain transcript-specific studies in AD and TREM2 mutation carriers. Molecular Neurodegeneration. 2019; 14: 18.
[100]
Piccio L, Deming Y, Del-Águila JL, Ghezzi L, Holtzman DM, Fagan AM, et al. Cerebrospinal fluid soluble TREM2 is higher in Alzheimer disease and associated with mutation status. Acta Neuropathologica. 2016; 131: 925–933.
[101]
Piccio L, Buonsanti C, Cella M, Tassi I, Schmidt RE, Fenoglio C, et al. Identification of soluble TREM-2 in the cerebrospinal fluid and its association with multiple sclerosis and CNS inflammation. Brain: a Journal of Neurology. 2008; 131: 3081–3091.
[102]
Deming Y, Filipello F, Cignarella F, Cantoni C, Hsu S, Mikesell R, et al. The ms4a gene cluster is a key modulator of soluble trem2 and alzheimer disease risk. Alzhmer’s and Dementia. 2019; 15: 922.
[103]
Colonna M, Wang Y. TREM2 variants: new keys to decipher Alzheimer disease pathogenesis. Nature Reviews. Neuroscience. 2016; 17: 201–207.
[104]
Schlepckow K, Monroe KM, Kleinberger G, Cantuti-Castelvetri L, Parhizkar S, Xia D, et al. Enhancing protective microglial activities with a dual function TREM2 antibody to the stalk region. EMBO Molecular Medicine. 2020; 12: e11227.
[105]
Jay TR, von Saucken VE, Landreth GE. TREM2 in Neurodegenerative Diseases. Molecular Neurodegeneration. 2017; 12: 56.
[106]
Zhao N, Qiao W, Li F, Ren Y, Zheng J, Martens YA, et al. Elevating microglia TREM2 reduces amyloid seeding and suppresses disease-associated microglia. Journal of Experimental Medicine. 2022; 219: e20212479.
[107]
Schlepckow K, Kleinberger G, Fukumori A, Feederle R, Lichtenthaler SF, Steiner H, et al. An Alzheimer-associated TREM2 variant occurs at the ADAM cleavage site and affects shedding and phagocytic function. EMBO Molecular Medicine. 2017; 9: 1356–1365.
[108]
Spiteri AG, Wishart CL, Pamphlett R, Locatelli G, King NJC. Microglia and monocytes in inflammatory CNS disease: integrating phenotype and function. Acta Neuropathologica. 2022; 14: 179–224.
[109]
Hsieh CL, Koike M, Spusta SC, Niemi EC, Yenari M, Nakamura MC, et al. A role for TREM2 ligands in the phagocytosis of apoptotic neuronal cells by microglia. Journal of Neurochemistry. 2009; 109: 1144–1156.
[110]
Takahashi K, Prinz M, Stagi M, Chechneva O, Neumann H. TREM2-transduced myeloid precursors mediate nervous tissue debris clearance and facilitate recovery in an animal model of multiple sclerosis. PLoS Medicine. 2007; 4: e124.
[111]
Jiang T, Tan L, Zhu XC, Zhang QQ, Cao L, Tan MS, et al. Upregulation of TREM2 ameliorates neuropathology and rescues spatial cognitive impairment in a transgenic mouse model of Alzheimer’s disease. Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology. 2014; 39: 2949–2962.
[112]
Yao H, Coppola K, Schweig JE, Crawford F, Mullan M, Paris D. Distinct Signaling Pathways Regulate TREM2 Phagocytic and NFκB Antagonistic Activities. Frontiers in Cellular Neuroscience. 2019; 13: 457.
[113]
Cignarella F, Filipello F, Bollman B, Cantoni C, Locca A, Mikesell R, et al. TREM2 activation on microglia promotes myelin debris clearance and remyelination in a model of multiple sclerosis. Acta Neuropathologica. 2020; 140: 513–534.
[114]
Griciuc A, Patel S, Federico AN, Choi SH, Innes BJ, Oram MK, et al. TREM2 Acts Downstream of CD33 in Modulating Microglial Pathology in Alzheimer’s Disease. Neuron. 2019; 103: 820–835.e7.
[115]
Zhong L, Chen XF, Zhang ZL, Wang Z, Shi XZ, Xu K, et al. DAP12 Stabilizes the C-terminal Fragment of the Triggering Receptor Expressed on Myeloid Cells-2 (TREM2) and Protects against LPS-induced Pro-inflammatory Response. The Journal of Biological Chemistry. 2015; 290: 15866–15877.
[116]
Guo Y, Wei X, Yan H, Qin Y, Yan S, Liu J, et al. TREM2 deficiency aggravates α-synuclein-induced neurodegeneration and neuroinflammation in Parkinson’s disease models. FASEB Journal: Official Publication of the Federation of American Societies for Experimental Biology. 2019; 33: 12164–12174.
[117]
Zhu Z, Zhang X, Dong W, Wang X, He S, Zhang H, et al. TREM2 suppresses the proinflammatory response to facilitate PRRSV infection via PI3K/NF-κB signaling. PLoS Pathogens. 2020; 16: e1008543.
[118]
Cantoni C, Bollman B, Licastro D, Xie M, Mikesell R, Schmidt R, et al. TREM2 regulates microglial cell activation in response to demyelination in vivo. Acta Neuropathologica. 2015; 129: 429–447.
[119]
Zheng H, Jia L, Liu CC, Rong Z, Zhong L, Yang L, et al. TREM2 Promotes Microglial Survival by Activating Wnt/β-Catenin Pathway. The Journal of Neuroscience: the Official Journal of the Society for Neuroscience. 2017; 37: 1772–1784.
[120]
Krasemann S, Madore C, Cialic R, Baufeld C, Calcagno N, El Fatimy R, et al. The TREM2-APOE Pathway Drives the Transcriptional Phenotype of Dysfunctional Microglia in Neurodegenerative Diseases. Immunity. 2017; 47: 566–581.e9.
[121]
Kober DL, Brett TJ. TREM2-Ligand Interactions in Health and Disease. Journal of Molecular Biology. 2017; 429: 1607–1629.
[122]
Niemitz E. TREM2 and Alzheimer’s disease. Nature Genetics. 2013; 45: 11–11.
[123]
Gratuze M, Leyns CE, Sauerbeck AD, St-Pierre MK, Xiong M, Kim N, et al. Impact of TREM2R47H variant on tau pathology-induced gliosis and neurodegeneration. The Journal of Clinical Investigation. 2020; 130: 4954–4968.
[124]
Cheng-Hathaway PJ, Reed-Geaghan EG, Jay TR, Casali BT, Bemiller SM, Puntambekar SS, et al. The Trem2 R47H variant confers loss-of-function-like phenotypes in Alzheimer’s disease. Molecular Neurodegeneration. 2018; 13: 29.
[125]
Kober DL, Alexander-Brett JM, Karch CM, Cruchaga C, Colonna M, Holtzman MJ, et al. Neurodegenerative disease mutations in TREM2 reveal a functional surface and distinct loss-of-function mechanisms. ELife. 2016; 5: e20391.
[126]
Hammond TR, Marsh SE, Stevens B. Immune Signaling in Neurodegeneration. Immunity. 2019; 50: 955–974.
[127]
Li JT, Zhang Y. TREM2 regulates innate immunity in Alzheimer’s disease. Journal of Neuroinflammation. 2018; 15: 107.
[128]
Molgora M, Esaulova E, Vermi W, Hou J, Chen Y, Luo J, et al. TREM2 Modulation Remodels the Tumor Myeloid Landscape Enhancing Anti-PD-1 Immunotherapy. Cell. 2020; 182: 886–900.e17.
[129]
Deczkowska A, Weiner A, Amit I. The Physiology, Pathology, and Potential Therapeutic Applications of the TREM2 Signaling Pathway. Cell. 2020; 181: 1207–1217.
[130]
Lee-Gosselin A, Jury-Garfe N, You Y, Dabin L, Soni D, Dutta S, et al. TREM2-Deficient Microglia Attenuate Tau Spreading In Vivo. Cells. 2023; 12: 1597.
[131]
Frank S, Burbach GJ, Bonin M, Walter M, Streit W, Bechmann I, Bechmann I, et al. TREM2 is upregulated in amyloid plaque-associated microglia in aged APP23 transgenic mice. Glia. 2008; 56: 1438–1447.
[132]
Karanfilian L, Tosto MG, Malki K. The role of TREM2 in Alzheimer’s disease; evidence from transgenic mouse models. Neurobiology of Aging. 2020; 86: 39–53.
[133]
Singh AK, Mishra G, Maurya A, Awasthi R, Kumari K, Thakur A, et al. Role of TREM2 in Alzheimer’s Disease and its Consequences on β- Amyloid, Tau and Neurofibrillary Tangles. Current Alzheimer Research. 2019; 16: 1216–1229.
[134]
Keren-Shaul H, Spinrad A, Weiner A, Matcovitch-Natan O, Dvir-Szternfeld R, Ulland TK, et al. A Unique Microglia Type Associated with Restricting Development of Alzheimer’s Disease. Cell. 2017; 169: 1276–1290.e17.
[135]
Rubio-Araiz A, Finucane OM, Keogh S, Lynch MA. Anti-TLR2 antibody triggers oxidative phosphorylation in microglia and increases phagocytosis of β-amyloid. Journal of Neuroinflammation. 2018; 15: 247.
[136]
Parhizkar S, Arzberger T, Brendel M, Kleinberger G, Deussing M, Focke C, et al. Loss of TREM2 function increases amyloid seeding but reduces plaque-associated ApoE. Nature Neuroscience. 2019; 22: 191–204.
[137]
Kim SM, Mun BR, Lee SJ, Joh Y, Lee HY, Ji KY, et al. TREM2 promotes Aβ phagocytosis by upregulating C/EBPα-dependent CD36 expression in microglia. Scientific Reports. 2017; 7: 11118.
[138]
Jay TR, Hirsch AM, Broihier ML, Miller CM, Neilson LE, Ransohoff RM, et al. Disease Progression-Dependent Effects of TREM2 Deficiency in a Mouse Model of Alzheimer’s Disease. The Journal of Neuroscience: the Official Journal of the Society for Neuroscience. 2017; 37: 637–647.
[139]
Bemiller SM, McCray TJ, Allan K, Formica SV, Xu G, Wilson G, et al. TREM2 deficiency exacerbates tau pathology through dysregulated kinase signaling in a mouse model of tauopathy. Molecular Neurodegeneration. 2017; 12: 74.
[140]
Shi Y, Holtzman DM. Interplay between innate immunity and Alzheimer disease: APOE and TREM2 in the spotlight. Nature Reviews. Immunology. 2018; 18: 759–772.
[141]
Jiang T, Tan L, Zhu XC, Zhou JS, Cao L, Tan MS, et al. Silencing of TREM2 exacerbates tau pathology, neurodegenerative changes, and spatial learning deficits in P301S tau transgenic mice. Neurobiology of Aging. 2015; 36: 3176–3186.
[142]
Hur JY, Frost GR, Wu X, Crump C, Pan SJ, Wong E, et al. The innate immunity protein IFITM3 modulates γ-secretase in Alzheimer’s disease. Nature. 2020; 586: 735–740.
[143]
Cai Y, Liu J, Wang B, Sun M, Yang H. Microglia in the Neuroinflammatory Pathogenesis of Alzheimer’s Disease and Related Therapeutic Targets. Frontiers in Immunology. 2022; 13: 856376.
[144]
Venegas C, Kumar S, Franklin BS, Dierkes T, Brinkschulte R, Tejera D, et al. Microglia-derived ASC specks cross-seed amyloid-β in Alzheimer’s disease. Nature. 2017; 552: 355–361.
[145]
Yin J, Liu X, He Q, Zhou L, Yuan Z, Zhao S. Vps35-dependent recycling of Trem2 regulates microglial function. Traffic (Copenhagen, Denmark). 2016; 17: 1286–1296.
[146]
Painter MM, Atagi Y, Liu CC, Rademakers R, Xu H, Fryer JD, et al. TREM2 in CNS homeostasis and neurodegenerative disease. Molecular Neurodegeneration. 2015; 10: 43.
[147]
Hou J, Chen Y, Grajales-Reyes G, Colonna M. TREM2 dependent and independent functions of microglia in Alzheimer’s disease. Molecular Neurodegeneration. 2022; 17: 84.
[148]
Deming Y, Li Z, Benitez BA, Cruchaga C. Triggering receptor expressed on myeloid cells 2 (TREM2): a potential therapeutic target for Alzheimer disease? Expert Opinion on Therapeutic Targets. 2018; 22: 587–598.
[149]
Delizannis AT, Nonneman A, Tsering W, Bondt AD, Van den Wyngaert I, Zhang B, et al. Effects of microglial depletion and TREM2 deficiency on Aβ plaque burden and neuritic plaque tau pathology in 5XFAD mice. Acta Neuropathologica Communications. 2021; 9: 150.
[150]
Wang Y, Ulland TK, Ulrich JD, Song W, Tzaferis JA, Hole JT, et al. TREM2-mediated early microglial response limits diffusion and toxicity of amyloid plaques. The Journal of Experimental Medicine. 2016; 213: 667–675.
[151]
Lee CYD, Daggett A, Gu X, Jiang LL, Langfelder P, Li X, et al. Elevated TREM2 Gene Dosage Reprograms Microglia Responsivity and Ameliorates Pathological Phenotypes in Alzheimer’s Disease Models. Neuron. 2018; 97: 1032–1048.e5.
[152]
Ulrich JD, Finn MB, Wang Y, Shen A, Mahan TE, Jiang H, et al. Altered microglial response to Aβ plaques in APPPS1-21 mice heterozygous for TREM2. Molecular Neurodegeneration. 2014; 9: 20.
[153]
Zhao Y, Wu X, Li X, Jiang L, Gui X, Liu Y, et al. TREM2 Is a Receptor for β-Amyloid that Mediates Microglial Function[J]. Neuron, 2018; 97: 1023–1031.
[154]
Ulland TK, Song WM, Huang SCC, Ulrich JD, Sergushichev A, Beatty WL, et al. TREM2 Maintains Microglial Metabolic Fitness in Alzheimer’s Disease. Cell. 2017; 170: 649–663.e13.
[155]
Attardo A, Fitzgerald JE, Schnitzer MJ. Impermanence of dendritic spines in live adult CA1 hippocampus. Nature. 2015; 523: 592–596.
[156]
Stanika RI, Flucher BE, Obermair GJ. Regulation of Postsynaptic Stability by the L-type Calcium Channel CaV1.3 and its Interaction with PDZ Proteins. Current Molecular Pharmacology. 2015; 8: 95–101.
[157]
Weinhard L, di Bartolomei G, Bolasco G, Machado P, Schieber NL, Neniskyte U, et al. Microglia remodel synapses by presynaptic trogocytosis and spine head filopodia induction. Nature Communications. 2018; 9: 1228.
[158]
Filipello F, Morini R, Corradini I, Zerbi V, Canzi A, Michalski B, et al. The Microglial Innate Immune Receptor TREM2 Is Required for Synapse Elimination and Normal Brain Connectivity. Immunity. 2018; 48: 979–991.e8.
[159]
Mattson MP. Pathways towards and away from Alzheimer’s disease. Nature. 2004; 430: 631–639.
[160]
Yu W, Lu B. Synapses and dendritic spines as pathogenic targets in Alzheimer’s disease. Neural Plasticity. 2012; 2012: 247150.
[161]
Zou C, Shi Y, Ohli J, Schüller U, Dorostkar MM, Herms J. Neuroinflammation impairs adaptive structural plasticity of dendritic spines in a preclinical model of Alzheimer’s disease. Acta Neuropathologica. 2016; 131: 235–246.
[162]
Meilandt WJ, Ngu H, Gogineni A, Lalehzadeh G, Lee SH, Srinivasan K, et al. Trem2 Deletion Reduces Late-Stage Amyloid Plaque Accumulation, Elevates the Aβ42:Aβ40 Ratio, and Exacerbates Axonal Dystrophy and Dendritic Spine Loss in the PS2APP Alzheimer’s Mouse Model. The Journal of Neuroscience: the Official Journal of the Society for Neuroscience. 2020; 40: 1956–1974.
[163]
Devanand DP, Masurkar AV, Wisniewski T. Vigorous, regular physical exercise may slow disease progression in Alzheimer’s disease. Alzheimer’s & Dementia: the Journal of the Alzheimer’s Association. 2023; 19: 1592–1597.
[164]
Scarmeas N, Luchsinger JA, Schupf N, Brickman AM, Cosentino S, Tang MX, et al. Physical activity, diet, and risk of Alzheimer disease. JAMA. 2009; 302: 627–637.
[165]
Liang KY, Mintun MA, Fagan AM, Goate AM, Bugg JM, Holtzman DM, et al. Exercise and Alzheimer’s disease biomarkers in cognitively normal older adults. Annals of Neurology. 2010; 68: 311–318.
[166]
Lourenco MV, Frozza RL, de Freitas GB, Zhang H, Kincheski GC, Ribeiro FC, et al. Exercise-linked FNDC5/irisin rescues synaptic plasticity and memory defects in Alzheimer’s models. Nature Medicine. 2019; 25: 165–175.
[167]
Lu Y, Dong Y, Tucker D, Wang R, Ahmed ME, Brann D, et al. Treadmill Exercise Exerts Neuroprotection and Regulates Microglial Polarization and Oxidative Stress in a Streptozotocin-Induced Rat Model of Sporadic Alzheimer’s Disease. Journal of Alzheimer’s Disease: JAD. 2017; 56: 1469–1484.
[168]
Bernardo TC, Beleza J, Rizo-Roca D, Santos-Alves E, Leal C, Martins MJ, et al. Physical exercise mitigates behavioral impairments in a rat model of sporadic Alzheimer’s disease. Behavioural Brain Research. 2020; 379: 112358.
[169]
Dao AT, Zagaar MA, Alkadhi KA. Moderate Treadmill Exercise Protects Synaptic Plasticity of the Dentate Gyrus and Related Signaling Cascade in a Rat Model of Alzheimer’s Disease. Molecular Neurobiology. 2015; 52: 1067–1076.
[170]
Mu L, Cai J, Gu B, Yu L, Li C, Liu QS, et al. Treadmill Exercise Prevents Decline in Spatial Learning and Memory in 3×Tg-AD Mice through Enhancement of Structural Synaptic Plasticity of the Hippocampus and Prefrontal Cortex. Cells. 2022; 11: 244.
[171]
Liu Y, Chu JMT, Yan T, Zhang Y, Chen Y, Chang RCC, et al. Short-term resistance exercise inhibits neuroinflammation and attenuates neuropathological changes in 3×Tg Alzheimer’s disease mice. Journal of Neuroinflammation. 2020; 17: 4.
[172]
Lu Y, Dong Y, Tucker D, Wang R, Ahmed ME, Brann D, et al. Treadmill exercise exerts neuroprotection and regulates microglial polarization and oxidative stress in a streptozotocin-induced rat model of sporadic Alzheimer’s disease. Journal of Alzheimer’s Disease. 2017; 56: 1469–1484.
[173]
Choi SH, Bylykbashi E, Chatila ZK, Lee SW, Pulli B, Clemenson GD, et al. Combined adult neurogenesis and BDNF mimic exercise effects on cognition in an Alzheimer’s mouse model. Science (New York, N.Y.). 2018; 361: eaan8821.
[174]
Wang Y, Lin Y, Wang L, Zhan H, Luo X, Zeng Y, et al. TREM2 ameliorates neuroinflammatory response and cognitive impairment via PI3K/AKT/FoxO3a signaling pathway in Alzheimer’s disease mice. Aging. 2020; 12: 20862–20879.
[175]
Han X, Cheng X, Xu J, Liu Y, Zhou J, Jiang L, et al. Activation of TREM2 attenuates neuroinflammation via PI3K/Akt signaling pathway to improve postoperative cognitive dysfunction in mice. Neuropharmacology. 2022; 15: 219:109231.
[176]
Zhang L, Liu Y, Wang X, Wang D, Wu H, Chen H, et al. Treadmill exercise improve recognition memory by TREM2 pathway to inhibit hippocampal microglial activation and neuroinflammation in Alzheimer’s disease model. Physiology & Behavior. 2022; 251: 113820.
[177]
Zhang SS, Zhu L, Peng Y, Zhang L, Chao FL, Jiang L, et al. Long-term running exercise improves cognitive function and promotes microglial glucose metabolism and morphological plasticity in the hippocampus of APP/PS1 mice. Journal of Neuroinflammation. 2022; 19: 34.
[178]
Jensen CS, Bahl JM, Østergaard LB, Høgh P, Wermuth L, Heslegrave A, et al. Exercise as a potential modulator of inflammation in patients with Alzheimer’s disease measured in cerebrospinal fluid and plasma. Experimental Gerontology. 2019; 121: 91–98.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top