Open access peer-reviewed chapter

Microglia, TREM2, and Therapeutic Methods of Alzheimer’s Disease

Written By

Siwei Xu, Yaya Ji, Tianle Sha and Haoming Li

Submitted: 05 August 2021 Reviewed: 30 August 2021 Published: 20 September 2021

DOI: 10.5772/intechopen.100203

From the Edited Volume

Hippocampus - Cytoarchitecture and Diseases

Edited by Xinhua Zhang

Chapter metrics overview

400 Chapter Downloads

View Full Metrics

Abstract

Alzheimer’s disease (AD) is one of the most common causes of dementia all around the world. It is characterized by the deposition of amyloid-β protein (Aβ) and the formation of neurofibrillary tangles (NFTs), which contribute to neuronal loss and cognitive decline. Microglia, as innate immune cells in brain, plays dual roles in the pathological process of AD. Expression in different subtypes of microglia is diverse in AD genes. Triggering receptor expressed on myeloid cells 2 (TREM2) is a transmembrane glycoprotein mainly expressed on microglia in the central nervous system (CNS). Soluble TREM2 (sTREM2), a proteolytic product of TREM2, which is abundant in the cerebrospinal fluid, shows a dynamic change in different stages and ameliorates the pathological process of AD. The interplay between the different subtypes of apolipoprotein and TREM2 is closely related to the mechanism of AD and serves as important regulatory sites. Moreover, several therapeutic strategies targeting TREM2 have shown positive outcomes during clinical trials and some novel therapies at different points are in progress. In this review, we mainly talk about the interrelationships among microglia, TREM2, and AD, and hope to give an overview of the strategies of AD.

Keywords

  • Alzheimer’s disease (AD)
  • microglia
  • TREM2
  • sTREM2
  • APOE

1. Introduction

Nowadays, Alzheimer’s disease (AD) is one of the most common causes of dementia in the United States [1]. Alois Alzheimer discovered AD in 1907 and characterized AD as amyloid plaques, brain atrophy, neurofibrillary tangles, loss of neurons and synapses, and dystrophic neurites in histopathology [2].

Microglia are the resident immune cells in the CNS. They derive from erythromyeloid progenitor cells and then migrate to the brain [3]. Developing and adult microglia demonstrate distinct morphological features as ramified or ameboid [4], which was proved by recent comprehensive transcriptomic analyses [5]. Relative analyses also demonstrate the heterogeneity, abundance, steady state in embryonic, postnatal, juvenile, and adult mouse models [6, 7]. They are also featured as self-renewing, which requires several factors such as colony-stimulating factor-1 receptor (CSF1R) and transforming growth factor β (TGF-β) [8, 9, 10]. Moreover, the murine signature of microglia in AD was present in human microglial subtypes, especially clusters 4, 5, 7, and 8. Among which, cluster 7 stands out in the consequence of its high expression of AD gene decrease in the tissue sections in both AD dementia and pathological AD [11]. This can be a diagnostic standard for AD when the frequency of cluster 7 was diminished.

Hippocampus is an elongated structure that is part of the cerebral cortex [12]. It is one of the most severely affected structures in neurodegenerative diseases like AD [13]. Hippocampus, along with its accessory structure, was suggested to be related to space [14, 15], time [16, 17], and the creation of declarative memories (memories that can evoke conscious awareness and be verbalized) [18].

Hippocampus is vulnerable to the harmfulness of diseases such as epilepsy, hypoxia, ischemia, or encephalitis [18]. The entorhinal cortex is usually the first region that demonstrates tau pathology in AD patients [18]. Somatostatin-positive interneurons are also found lost in the hippocampus of AD patients [19]. In AD patients, degenerative cholinergic neurons in the basal forebrain were proved to lead to dysfunctional cholinergic neurotransmission in regions like hippocampus [20].

Advertisement

2. Harmful and beneficial effects of microglia

Microglia play the role of phagocytes in the CNS, thus, maintaining the homeostasis of the brain [21]. In aging brains, microglia will cause synaptic clearance leading to forgetting via complement pathway [22]. In AD pathology, microglia also prove to be phagocytose synapses [23, 24]. Nevertheless, with CSF1R blockade to remove microglia in Aβ models, increased Aβ is detected [25]. Despite negative outcomes of microglia, synapse loss and behavior deficits can be avoided [26, 27]. The production of neurotoxic inflammatory cytokines and reactive oxygen species are found to be related to chronic activation of microglia [28]. However, it still remains unclear whether microglia play positive or negative roles in the process of neurodegenerative diseases.

In recent research, microglia in patients with AD show specific characteristics such as aging and upregulation of apolipoprotein E (APOE) [29]. The fat droplets appearing in microglia of aged mice suggest that the main manifestations of aging are the accumulation of fat droplets and excessive secretion of pro-inflammatory factors [30], which may be a new biological hallmark of AD. Additionally, it is not difficult to find that the branching of microglia has been reduced in aged brains, thus cutting the size of microglia’s area for surveillance and leading to the harm of homeostatic functions [31, 32, 33, 34]. One important function of microglia in AD is the phagocytosis of Aβ amyloid. For instance, microglia can mediate clearance of Aβ via receptors including β1 integrin in neurodegenerative diseases [35]. The acute inflammatory response can also promote phagocytosis of impaired neurons and neuronal toxic accumulation [36]. Despite the protection of microglia, prolonged inflammatory reaction will exacerbate neuronal degeneration [37]. The TAM receptor tyrosine kinases (RTKs) are a distinct family of three protein tyrosine kinases, namely Tyro3, Axl, and Mer3, which play an important role in phagocytosis and phagocytic clearance of apoptotic cells and cell membranes in the adult tissues [38, 39]. Axl and Mer play pivotal roles in macrophages like phagocytosis of apoptotic cells and negative feedback inhibition of toll-like receptor and cytokine receptor signaling. In AD mice with double knockout of Axl or Mer, the ability of microglia to phagocytize the plaque is weakened, suggesting the inhibition of TAM signal promotes plaque formation [40]. A cluster of differentiation-22 (CD22), a canonical B-cell receptor and a negative regulator of phagocytosis, is found highly expressed in microglia of aged brains, and rarely in young brains [41]. The finding suggests that the inhibition of CD22 can delay aging-related dysfunction and neurodegenerative diseases. The pellino-1 (peli1) is a ubiquitin E3 ligase, expressed in many kinds of nerve cells in the mouse brain, and with the highest expression level of microglia [42]. Similarly, Peli1 negatively regulates the ability of phagocytosis of microglia to Aβ, resulting in the inability of clearance of deposition, leading to the deterioration of AD [43].

Perineuronal nets (PNNs), with their structure remaining unknown in detail, surround the cell bodies and dendrites, and spare free space for synaptic contact [44]. In the AD mice model and human cortical tissue, PNNs are largely lost in proportion to plaque burden and depletion of microglia. Loss is prevented regardless of plaque persistence and suggests that microglia can enhance the loss of PNNs in the AD brain [45]. Besides, CD163-positive amyloid-responsive microglia are depleted in TREM2 and APOE variants in AD like TREM2 R47H and APOE4 [46].

Microglia may be detrimental to neurons in the pathological process. Recently, interleukin 3 (IL-3) from astrocytes was found to re-encode microglia, thus improving the situation of Aβ pathology [47]. Injection of IL-3 enables microglia to focus on clearing amyloid deposition and neurofibrillary tangles instead of causing extensive neuroinflammation [47]. This signaling pathway is expected to provide ideas for new drug research and development in the future and bring new drugs for the treatment of AD. A study suggested that some damaging characteristics of microglia behavior may be reversible by short-term treatment with CSF1R inhibitors [48, 49, 50]. In the mice model, removal of microglia did not improve the cognitive ability in a traumatic brain injury (TBI) [51]. Interestingly, repopulating microglia can reverse the decrease of nerve regeneration caused by brain injury and improve cognitive dysfunction in mice in an IL-6-dependant manner [51]. This study opens up a new understanding of the role of microglia in the brain injury. Remarkably, the ubiquitin ligase COP1 (also called RFWD2) is shown to dampen the neuroinflammation through inhibiting the expression of the transcription factor CCAAT/enhancer-binding protein beta (c/EBPβ), which regulates the pro-inflammatory gene of microglia [52], marking a new target for suppressing neuroinflammation in AD patients.

Disease-associated microglia (DAM), which was identified in AD patients by single-cell RNA sequencing (RNA-seq) [53], has recently become a hot topic, characterized by molecules including Iba1, Cst3, and Hexb, typically expressed in microglia. DAM also experiences downregulation of physiologically expressed genes such as P2ry12, P2ry13, Cx3cr1, CD33, and Tmem119 [9]. It is remarkable to find that DAM is identified in areas that are affected by diseases such as cortical tissue [53] and postmortem human AD brain [54]. The evidence suggests that DAM is specifically expressed in CNS pathological process, serving as an important pathological diagnostic standard. However, in the late set of neurodegenerative diseases, its role still remains unclear, which needs further investigation.

Advertisement

3. Physiological function on microglia of TREM2

Recent years have witnessed the central role of TREM2 as a hub in diverse pathology. TREM2 is a receptor that interacts with a variety of ligands, many of which are markers of tissue damage. TREM2 is a single-pass transmembrane protein known to regulate immune responses in peripheral macrophages through lipopolysaccharide binding and bacterial phagocytosis [55, 56, 57]. RNA-seq data were analyzed across human tissues to investigate TREM2 expression, and it has been confirmed that TREM2 is expressed physiologically in a small group of macrophages that are tissue specific [58]. In CNS, TREM2 is mostly expressed on microglia. In addition to the expression on microglia, the analysis also showed its expression in macrophages from the adrenal gland, placenta, and adipose tissue [59]. TREM2 was thought to bind a wide range of molecules [60], and the interaction with different ligands can regulate the signal intensity and direction of TREM2 in turn [61]. Downstream signals mainly consist of those arrangements; for example, DAP10 is the key to activate extracellular signal regulated-kinase (ERK) and serine/threonine protein kinase (AKT1), while in murine macrophages, DAP12 is necessary for calcium mobilization [61, 62, 63]. Functional loss of TREM2 is related to polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy (PLOSL) [64].

Mice lacking TREM2 had defects in survival and differentiation of myeloid cells [65], as well as osteopenia and loss of microglia in CNS [62, 66]. TREM2-lacking cells may undergo a similar differential process as normal cells do despite a reduced life cycle [67]. Microglia deprived of TREM2 or expressing T66M variant demonstrated the impaired process of brain glucose metabolism and cerebral perfusion [68]. Mice TREM2 was involved in synaptic pruning through a microglia-dependent way to shape neuronal circuitry [69]. In rodents, TREM2-positive macrophages are found to be important regulators related to hair follicle stem cells [70]. Additionally, in TREM2-deficient microglia, increased autophagic vesicles can be found with defective activation of mTOR pathways [63], which partially regulate autophagy [71].

Deletion or impairment of TREM2 was proved to be detrimental to phagocytosis of lipoproteins, cellular debris, bacteria, and Aβ [68, 72, 73]. Moreover, overexpression of TREM2 in cells that are not functionally phagocytic like Chinese hamster ovary (CHO) cells showed induced phagocytosis of apoptotic cells and bacteria [57, 72].

TREM2 was also found to ameliorate neuroinflammation and neuronal apoptosis via PI3K/AKT signaling pathway in 5xFAD mice [74, 75]. TREM2 overexpression can also rescue cognitive barriers by reducing neuroinflammation via JAK/STAT/SOCS pathway [76] and the suppression of TREM2 demonstrated a defective ability to regulate the PI3K/Akt and NF-κB signaling pathways [77].

Recently, genome-wide association studies (GWAS) demonstrated a link between single-nucleotide polymorphisms (SNPs) and inflammation-related genes to increased AD risk, such as the R47H variant in TREM2 [78], which is one of the strongest genetic risk factors for AD [79]. TREM2 variant R47H, whose foundation was dysregulated peroxisome proliferator-activated receptor-γ (PPARγ)/p38MAPK signaling [80], was shown to decrease the expression of TREM2, thus deteriorating myeloid cell responses to Aβ pathology [81]. Furthermore, the R47H variants and R62H variants of TREM2 demonstrate a defective microglial transcriptional activation, implicating fully functional TREM2 seems to be the key for development of the human DAM [82].

Advertisement

4. TREM2 and AD

TREM2 gives protection against neurodegenerative disease. Depletion of TREM2 can induce impaired phagocytosis of the critical substrates such as APOE [83] and exacerbates tau pathology in AD [84].

4.1 TREM2 gets involved in AD pathogenesis via microglia

TREM2 is found to reduce tau seeding in neuritic plaques [85], which is essential for synapse clearance in the early stage of brain development, and TREM2-KO mice demonstrate altered sociability [69]. Moreover, TREM2 can induce microglia to gather around Aβ and restrict plaque expansion found in murine models of AD [86]. Similar conditions can also lead to exacerbation of axonal dystrophy and dendritic spine loss [87]. Another research shows that a dosage of TREM2 can reprogram the microglial response in downregulating the expression of DAM genes and ameliorating the pathological phenotype in AD mice [88]. In the absence of functional TREM2, amyloid plaque seeding increased, and microglial aggregation decreased [88]. A similar study shows that in human pluripotent stem cell (PSC), monocytes and transdifferentiated microglia-like cells, TREM2 R47H variant and loss of TREM2 on heterozygous or homozygous, display a significant decreased in phagocytosis [89]. On a recent finding, IL-4 and IL-10 enhance the phagocytosis of microglia via upregulation of TREM2 [90]. These findings support the hypothesis that reactive microglia and TREM2 are functionally necessary to alleviate neuronal damage. However, other studies give opposite outcomes that loss of TREM2 may be protective in AD mice [91].

Genetically, the immune cell-specific phospholipase C isoform γ2 (PLCG2), a rare coding variant, is identified [92]. Recent research has demonstrated that TREM2 can mediate phagocytosis, cell survival, lipid metabolism, and process neuronal debris through PLCG2 of microglia derived from human-induced pluripotent stem cell (iPSC) [93]. PLCG2 P552R variant has protective functions including weak-enhancing enzyme functions [94] and promoting survival functions of microglia in Plcγ2-P522R knock in mice [95]. These studies highlighted the critical role of the TREM2 pathway in AD and provided genetic evidence for the increase of TREM2 in the pathologic process of AD.

In recent years, different TREM2 ligands have been found and proposed, such as β-amyloid peptide [96] and APOE [97]. APOE-dependent molecular signature in microglia is identified in AD patients, mediating a switch from homeostatic to neurodegenerative status [98]. This can be a target in treating AD patients through restoring the homeostatic microglia.

4.2 TREM2 regulates APOE mediating AD risk

Although there is no difference in the quantity of activated microglia and reactive astrocytes between APOE4 carriers and noncarriers in the postmortem neocortex [99], relative transcriptomic studies have shown the connection between APOE and glia. Human APOE is expressed in three allelic variants, APOE2, APOE3, and APOE4, which exhibit different receptor binding properties [100]. APOE upregulation has been proved to be TREM2-dependent [101]. To some extent, TREM2 and APOE may have some special links [102], and the lack of TREM2 leads to a decrease of APOE4, while APOE3 remains unchanged [103]. Microglial plaque coverage and TREM2 are the highest in APOE3 male mice while significantly low in both APOE4 genotype and female sex [104], implicating a possible mechanism of AD between sex and APOE genotype. A reduction in plaque-associated APOE is also found in the brains of AD patients [105]. In another research, APOE3 is shown to promote the proliferation of microglia to injected Aβ, contribute to the uptake of Aβ, and improve cognition related to Aβ in preclinical models of AD [106]. Moreover, APOE was proven to stimulate different signal transduction cascades, ApoE4 > ApoE3 > ApoE2, in proportion to their AD risk [107]. This suggests that neuronal pathways may be related to the pathogenesis of AD. Human TREM2 (hTREM2) was bind to APOJ and APOE that are ligands of TREM2 under normal circumstances [73]. However, this binding is reduced in diseases or TREM2 KO mice microglia, leading to the impaired uptake of Aβ [73]. TREM2 is also an attractive target for drug regulation, but needs to be cautious because it is an important upstream mediator of microglia activation and phenotypic changes [53, 98]. In addition, single-cell transcriptomic studies pointing at microglia have shown a fascinating TREM2 ligand gal-3 that is related to neurodegenerative diseases [108]. Increased gal-3 is found in AD patients and 5xFAD mice, while decreased gal-3 shows improved cognitive ability and attenuates immune responses related to the TREM2-DAP pathway [109]. Therefore, suppressing gal-3 in the AD process may be a potential target in treatment.

Advertisement

5. sTREM2

sTREM2, a soluble form of TREM2, is derived from the non-proteolytic-mediated secretion of some TREM2 isoforms or due to extracellular domain of TREM2 being cleaved by different sheddases [110]. Years before the onset of dementia symptoms, sTREM2 increased in cerebrospinal fluid (CSF) of people with AD biomarker characteristics [111, 112, 113, 114]. Recently, it is found that in preclinical AD, CSF sTREM2 changes are dynamic. In the absence of tau deposition and neurodegeneration, sTREM2 is decreased with Aβ pathology [115]. Different mutants of sTREM2 showed differences in concentration in CSF [114]. sTREM2 has a protective effect on Aβ and AD, such as reducing amyloid plaque load and restoring spatial memory [116]. Similarly, in the absence of TREM2 [105], sTREM2 enhances microglial proliferation, migration, clustering around Aβ, and contributing to the uptake and degradation of Aβ [116]. sTREM2 administration can also stimulate the expression of inflammatory cytokines and induce morphological changes of microglia such as decreased cell process and increased cell body size, thus enhancing microglial survival [117]. In TREM2 KO mice, administration of sTREM2 also showed positive feedback, like rescuing apoptosis upon colony-stimulating factor (GM-CSF) withdrawal, inducing the proliferation and cell viability of the primary microglia [118, 119], compared with WT mice [117]. These results indicate the tremendous therapeutic potential of sTREM2, but warn that pro-inflammatory activation in the brain may lead to negative functional outcomes. Under stress, sTREM2 can promote myeloid cell survival too in a manner dependent on PI3K/AKT [117, 120]. Consequently, sTREM2 can be a target for AD therapy. But it is shown less potent for sTREM2-R47H and sTREM2-R62H variants to suppress apoptosis in AD context [117]. Among the three polymorphic forms (APOE2, APOE3, and APOE4), APOE4 proved to be more related to AD [121] and high levels of sTREM2 are associated with the decrease of APOE4 [122] and slower rates of Aβ accumulation [123]. In conclusion, the changes of sTREM2 can also be a biological hallmark for AD.

Advertisement

6. The prospect of treatment of AD

For a long time, it is considered that Aβ accumulation is the central and initial event in the pathological process of AD. The famous amyloid cascade hypothesis thinks that the increase of Aβ levels leads to the pathological events of AD [124, 125]. Extensive clinical medicine trials of Aβ finally come to an end, and results showed that reduced Aβ load does not affect the cognitive ability of patients with AD [126, 127]. So, finding a new target rather than Aβ may be our priority. Microglia play pivotal roles in the pathological process, and interfering with their detrimental process in AD can become our next focus.

Microglia are shown to maintain the function of neurons by clearing toxic damage in the early stage of AD [128]. Consequently, interfering with the activation of microglia to lengthen the period of anti-inflammatory seems to be a therapy for AD [129]. Other anti-inflammatory cytokines such as IL-2, IL-4, and IL-33 have the potential to ameliorate AD pathology by regulating microglial activation [128], despite its results are not decisive [130].

Moreover, TREM2 is shown to be a positive target for treating AD. Recently, AL002c, an anti-human TREM2 agonistic monoclonal antibody (mAb), gives a positive outcome in 5xFAD mice expressing both the R47H variant and the common variant (CV). Prolonged administration of AL002c ameliorates filamentous plaques, causes neurodystrophy, and regulates microglial inflammation. AL002 is a derivative product of AL002c, which is modified for clinical use. AL002 is proven safe and well-tolerated in a first-in-human phase I clinical trial [131]. Overexpression of TREM2 can attenuate the pro-inflammatory effect caused by LPS, which can contribute to the increase of NO, LDH, TNF-α, IL-1b, and the activation of AKT [132]. Thus, relative experiments can be conducted in CNS.

Another way to increase TREM2 expression in microglia is by preventing ADAM10/17 family proteases from shedding extracellular domain [133, 134]. To stabilize TREM2 on the cell surface and enhance its activity, a specific mAb against TREM2 called 4D9 was screened to selectively compete for α-secretase-mediated shedding [133]. Shedding is considered to end cell-autonomous TREM2 signaling, and data show an increased phagocytic capacity of cells that express TREM2 by inhibiting ADAM proteases [135]. Combined with another research, Aβ clearance is TREM2-dependant [136], and future treatments can combine anti-Aβ antibodies with microglia-stimulating antibodies (4D9). This view opens a new door to the treatment of AD. Another study evaluated aducanumab as another antibody that may treat AD, but clinical trial results are still unsatisfactory [137]. In addition to cross-linking and activating the TREM2-DAP12 signal, 4D9 also inhibits the shedding of TREM2, resulting in the decrease of soluble TREM2 in vitro and the increase of total TREM2 in the brain [133]. This research may consider the role of sTREM2 in AD [91]. Since TREM2 is expressed in peripheral myeloid cells, any effects of treatment for TREM2 should be evaluated for peripheral adipose tissue in liver, lung, bone, and spleen. However, this has not been thoroughly investigated [91].

A novel property, cyclocreatine, the creatine analog, which can generate a supply chain for ATP demand regardless of the TREM2-mTOR pathway [138], is found to ameliorate autophagy, induce microglia around Aβ, and decrease neuronal dystrophy during dietary administration in 5xFAD mice [63]. Based on metabolism, this is a new era for treating AD.

Another research provides a creative angle in treating AD. It is known that meningeal lymphatic vessels drain macromolecular substances from the brain into the deep cervical lymph nodes [139], in which meningeal lymphatic serves as a channel to transport substances such as an antibody. But ablation of meningeal lymphatic vessels in 5xFAD mice can lead to a switch of microglia from homeostasis to DAM [140] and inhibit the transportation of antibodies to specific locations, thus exacerbating the cognitive ability of AD patients. It may bring unexpected clinical effects to patients with AD, if the treatment is placed in the early stage, thus enhancing the meningeal lymphatic function and combined with immunotherapy, to better play the role of meningeal lymphatic vessels.

Recently, tau pathology is the study focus. Tau hyperphosphorylation causes abnormal aggregation and neurodegeneration in AD brains [141], and protein phosphatase 2A (PP2A) has the most robust dephosphorylation activity to tau protein in vitro and in vivo [142]. A novel DEPho-sphorylation Targeting Chimaera (DEPTAC) was designed to enhance the combination of tau and PP2A-Bα, which shows high efficiency in preventing tau accumulation in vitro and in vivo [143]. Further studies showed that DEPAC significantly improved the microtubule assembly, neurite plasticity, and hippocampus-dependent learning and memory in transgenic mice [143].

Advertisement

7. Conclusion

Microglia play important roles in the pathological process of AD. The dual role it plays (positive or negative outcomes), its distinctive phenotype, DAM, which is specifically expressed in certain regions in AD, still needs further investigation. In most findings, TREM2 exhibits positive feedback in inhibiting detrimental factors. sTREM2, a soluble form of TREM2 in CSF, and its soluble form in CSF and sTREM2 can be biological hallmarks for diagnosis. Moreover, a close relationship between the TREM2-APOE pathway and AD demonstrates an important pathological feature. A new therapeutic method based on TREM2 to manipulate the function of microglia is currently being tested. Although there are still numerous obstacles ahead to treating AD, it is expected that this field will move closer to understanding the influence of microglia regulation in AD, which is a breakthrough result for patients. Most therapeutic treatments targeting Aβ do not get expected feedback. Thus, genetic evidence and metabolic mechanism related to AD should be more explored in future studies.

References

  1. 1. James BD, Leurgans SE, Hebert LE, Scherr PA, Yaffe K, Bennett DA. Contribution of Alzheimer disease to mortality in the United States. Neurology. 2014;82(12):1045-1050. DOI: 10.1212/WNL.0000000000000240
  2. 2. Alzheimer, AJAzp. Uber eine eigenartige Erkrankung der Hirnrinde. Allgemeine Zeitschrife Psychiatrie.1907; 64: 146-148
  3. 3. Ginhoux F, Lim S, Hoeffel G, Low D, Huber T. Origin and differentiation of microglia. Frontiers in Cellular Neuroscience. 2013;7:45. DOI: 10.3389/fncel.2013.00045
  4. 4. Lawson LJ, Perry VH, Gordon S. Turnover of resident microglia in the normal adult mouse brain. Neuroscience. 1992;48(2):405-415. DOI: 10.1016/0306-4522(92)90500-2
  5. 5. Matcovitch-Natan O, Winter DR, Giladi A, Vargas Aguilar S, Spinrad A, Sarrazin S, et al. Microglia development follows a stepwise program to regulate brain homeostasis. Science. 2016;353(6301):aad8670. DOI: 10.1126/science.aad8670
  6. 6. Varol D, Mildner A, Blank T, Shemer A, Barashi N, Yona S, et al. Dicer deficiency differentially impacts microglia of the developing and adult brain. Immunity. 2017;46(6):1030-1044 e8. DOI: 10.1016/j.immuni.2017.05.003
  7. 7. Masuda T, Sankowski R, Staszewski O, Bottcher C, Amann L, Sagar, et al. Spatial and temporal heterogeneity of mouse and human microglia at single-cell resolution. Nature. 2019;566(7744):388-392. DOI: 10.1038/s41586-019-0924-x
  8. 8. Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. 2010;330(6005):841-845. DOI: 10.1126/science.1194637
  9. 9. Butovsky O, Jedrychowski MP, Moore CS, Cialic R, Lanser AJ, Gabriely G, et al. Identification of a unique TGF-beta-dependent molecular and functional signature in microglia. Nature Neuroscience. 2014;17(1):131-143. DOI: 10.1038/nn.3599
  10. 10. Elmore MR, Najafi AR, Koike MA, Dagher NN, Spangenberg EE, Rice RA, 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: 10.1016/j.neuron.2014.02.040
  11. 11. Olah M, Menon V, Habib N, Taga MF, Ma Y, Yung CJ, et al. Single cell RNA sequencing of human microglia uncovers a subset associated with Alzheimer's disease. Nature Communications. 2020;11(1):6129. DOI: 10.1038/s41467-020-19737-2
  12. 12. Gilbert PE, Brushfield AM. The role of the CA3 hippocampal subregion in spatial memory: A process oriented behavioral assessment. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2009;33(5):774-781. DOI: 10.1016/j.pnpbp.2009.03.037
  13. 13. Frisoni GB, Fox NC, Jack CR Jr, Scheltens P, Thompson PM. The clinical use of structural MRI in Alzheimer disease. Nature Reviews. Neurology. 2010;6(2):67-77. DOI: 10.1038/nrneurol.2009.215
  14. 14. Eckardt, Michael. The Hippocampus as a Cognitive Map. The Journal of Nervous and Mental Disease.1980; 168: 191-192.DOI: 10.1097/00005053-198003000-00018
  15. 15. Moser EI, Roudi Y, Witter MP, Kentros C, Bonhoeffer T, Moser MB. Grid cells and cortical representation. Nature Reviews. Neuroscience. 2014;15(7):466-481. DOI: 10.1038/nrn3766
  16. 16. Eichenbaum H. Time cells in the hippocampus: A new dimension for mapping memories. Nature Reviews. Neuroscience. 2014;15(11):732-744. DOI: 10.1038/nrn3827
  17. 17. Eichenbaum H. On the integration of space, time, and memory. Neuron. 2017;95(5):1007-1018. DOI: 10.1016/j.neuron.2017.06.036
  18. 18. Knierim JJ. The hippocampus. Current Biology. 2015;25(23):R1116-R1121. DOI: 10.1016/j.cub.2015.10.049
  19. 19. Chan-Palay V. Somatostatin immunoreactive neurons in the human hippocampus and cortex shown by immunogold/silver intensification on vibratome sections: Coexistence with neuropeptide Y neurons, and effects in Alzheimer-type dementia. The Journal of Comparative Neurology. 1987;260(2):201-223. DOI: 10.1002/cne.902600205
  20. 20. Perry E, Walker M, Grace J, Perry R. Acetylcholine in mind: A neurotransmitter correlate of consciousness? Trends in Neurosciences. 1999;22(6):273-280. DOI: 10.1016/s0166-2236(98)01361-7
  21. 21. Nayak D, Roth TL, McGavern DB. Microglia development and function. Annual Review of Immunology. 2014;32:367-402. DOI: 10.1146/annurev-immunol-032713-120240
  22. 22. Wang C, Yue H, Hu Z, Shen Y, Ma J, Li J, et al. Microglia mediate forgetting via complement-dependent synaptic elimination. Science. 2020;367(6478):688-694. DOI: 10.1126/science.aaz2288
  23. 23. Shi Q, Chowdhury S, Ma R, Le KX, Hong S, Caldarone BJ, et al. Complement C3 deficiency protects against neurodegeneration in aged plaque-rich APP/PS1 mice. Science Translational Medicine. 2017;9(392):eaaf6295. DOI: 10.1126/scitranslmed.aaf6295
  24. 24. Hong S, Beja-Glasser VF, Nfonoyim BM, Frouin A, Li S, Ramakrishnan S, et al. Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science. 2016;352(6286):712-716. DOI: 10.1126/science.aad8373
  25. 25. Dagher NN, Najafi AR, Kayala KM, Elmore MR, White TE, Medeiros R, et al. Colony-stimulating factor 1 receptor inhibition prevents microglial plaque association and improves cognition in 3xTg-AD mice. Journal of Neuroinflammation. 2015;12:139. DOI: 10.1186/s12974-015-0366-9
  26. 26. Olmos-Alonso A, Schetters ST, Sri S, Askew K, Mancuso R, Vargas-Caballero M, 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: 10.1093/brain/awv379
  27. 27. Spangenberg EE, Lee RJ, Najafi AR, Rice RA, Elmore MR, Blurton-Jones M, et al. Eliminating microglia in Alzheimer’s mice prevents neuronal loss without modulating amyloid-beta pathology. Brain. 2016;139(Pt 4):1265-1281. DOI: 10.1093/brain/aww016
  28. 28. Block ML, Zecca L, Hong JS. Microglia-mediated neurotoxicity: Uncovering the molecular mechanisms. Nature Reviews. Neuroscience. 2007;8(1):57-69. DOI: 10.1038/nrn2038
  29. 29. Srinivasan K, Friedman BA, Etxeberria A, Huntley MA, van der Brug MP, Foreman O, et al. Alzheimer’s patient microglia exhibit enhanced aging and unique transcriptional activation. Cell Reports. 2020;31(13):107843. DOI: 10.1016/j.celrep.2020.107843
  30. 30. Marschallinger J, Iram T, Zardeneta M, Lee SE, Lehallier B, Haney MS, et al. Lipid-droplet-accumulating microglia represent a dysfunctional and proinflammatory state in the aging brain. Nature Neuroscience. 2020;23(2):194-208. DOI: 10.1038/s41593-019-0566-1
  31. 31. Spittau B. Aging microglia-phenotypes, functions and implications for age-related neurodegenerative diseases. Frontiers in Aging Neuroscience. 2017;9:194. DOI: 10.3389/fnagi.2017.00194
  32. 32. 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 Pathology. 2017;27(6):795-808. DOI: 10.1111/bpa.12456
  33. 33. Rawji KS, Mishra MK, Michaels NJ, Rivest S, Stys PK, Yong VW. Immunosenescence of microglia and macrophages: Impact on the ageing central nervous system. Brain. 2016;139(Pt 3):653-661. DOI: 10.1093/brain/awv395
  34. 34. Bisht K, Sharma KP, Lecours C, Sanchez MG, El Hajj H, Milior G, et al. Dark microglia: A new phenotype predominantly associated with pathological states. Glia. 2016;64(5):826-839. DOI: 10.1002/glia.22966
  35. 35. Koenigsknecht J, Landreth G. Microglial phagocytosis of fibrillar beta-amyloid through a beta1 integrin-dependent mechanism. The Journal of Neuroscience. 2004;24(44):9838-9846. DOI: 10.1523/JNEUROSCI.2557-04.2004
  36. 36. Neher JJ, Neniskyte U, Zhao JW, Bal-Price A, Tolkovsky AM, Brown GC. Inhibition of microglial phagocytosis is sufficient to prevent inflammatory neuronal death. Journal of Immunology. 2011;186(8):4973-4983. DOI: 10.4049/jimmunol.1003600
  37. 37. Shabab T, Khanabdali R, Moghadamtousi SZ, Kadir HA, Mohan G. Neuroinflammation pathways: A general review. The International Journal of Neuroscience. 2017;127(7):624-633. DOI: 10.1080/00207454.2016.1212854
  38. 38. Lemke G, Rothlin CV. Immunobiology of the TAM receptors. Nature Reviews. Immunology. 2008;8(5):327-336. DOI: 10.1038/nri2303
  39. 39. Lemke G, Burstyn-Cohen T. TAM receptors and the clearance of apoptotic cells. Annals of the New York Academy of Sciences. 2010;1209:23-29. DOI: 10.1111/j.1749-6632.2010.05744.x
  40. 40. Huang Y, Happonen KE, Burrola PG, O’Connor C, Hah N, Huang L, et al. Microglia use TAM receptors to detect and engulf amyloid beta plaques. Nature Immunology. 2021;22(5):586-594. DOI: 10.1038/s41590-021-00913-5
  41. 41. Pluvinage JV, Haney MS, Smith BAH, Sun J, Iram T, Bonanno L, et al. CD22 blockade restores homeostatic microglial phagocytosis in ageing brains. Nature. 2019;568(7751):187-192. DOI: 10.1038/s41586-019-1088-4
  42. 42. Xiao Y, Jin J, Chang M, Chang JH, Hu H, Zhou X, et al. Peli1 promotes microglia-mediated CNS inflammation by regulating Traf3 degradation. Nature Medicine. 2013;19(5):595-602. DOI: 10.1038/nm.3111
  43. 43. Xu J, Yu T, Pietronigro EC, Yuan J, Arioli J, Pei Y, et al. Peli1 impairs microglial Abeta phagocytosis through promoting C/EBPbeta degradation. PLoS Biology. 2020;18(10):e3000837. DOI: 10.1371/journal.pbio.3000837
  44. 44. Matthews RT, Kelly GM, Zerillo CA, Gray G, Tiemeyer M, Hockfield S. Aggrecan glycoforms contribute to the molecular heterogeneity of perineuronal nets. The Journal of Neuroscience. 2002;22(17):7536-7547
  45. 45. Crapser JD, Spangenberg EE, Barahona RA, Arreola MA, Hohsfield LA, Green KN. Microglia facilitate loss of perineuronal nets in the Alzheimer’s disease brain. eBioMedicine. 2020;58:102919. DOI: 10.1016/j.ebiom.2020.102919
  46. 46. Nguyen AT, Wang K, Hu G, Wang X, Miao Z, Azevedo JA, et al. APOE and TREM2 regulate amyloid-responsive microglia in Alzheimer’s disease. Acta Neuropathologica. 2020;140(4):477-493. DOI: 10.1007/s00401-020-02200-3
  47. 47. McAlpine CS, Park J, Griciuc A, Kim E, Choi SH, Iwamoto Y, et al. Astrocytic interleukin-3 programs microglia and limits Alzheimer's disease. Nature. 2021;595(7869):701-706. DOI: 10.1038/s41586-021-03734-6
  48. 48. Rice RA, Spangenberg EE, Yamate-Morgan H, Lee RJ, Arora RP, Hernandez MX, et al. Elimination of microglia improves functional outcomes following extensive neuronal loss in the hippocampus. The Journal of Neuroscience. 2015;35(27):9977-9989. DOI: 10.1523/JNEUROSCI.0336-15.2015
  49. 49. Rice RA, Pham J, Lee RJ, Najafi AR, West BL, Green KN. Microglial repopulation resolves inflammation and promotes brain recovery after injury. Glia. 2017;65(6):931-944. DOI: 10.1002/glia.23135
  50. 50. Weber MD, McKim DB, Niraula A, Witcher KG, Yin W, Sobol CG, et al. The influence of microglial elimination and repopulation on stress sensitization induced by repeated social defeat. Biological Psychiatry. 2019;85(8):667-678. DOI: 10.1016/j.biopsych.2018.10.009
  51. 51. Willis EF, MacDonald KPA, Nguyen QH, Garrido AL, Gillespie ER, Harley SBR, et al. Repopulating microglia promote brain repair in an IL-6-dependent manner. Cell. 2020;180(5):833-846 e16. DOI: 10.1016/j.cell.2020.02.013
  52. 52. Ndoja A, Reja R, Lee SH, Webster JD, Ngu H, Rose CM, et al. Ubiquitin ligase COP1 suppresses neuroinflammation by degrading c/EBPbeta in microglia. Cell. 2020;182(5):1156-1169 e12. DOI: 10.1016/j.cell.2020.07.011
  53. 53. 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(7):1276-1290 e17. DOI: 10.1016/j.cell.2017.05.018
  54. 54. Friedman BA, Srinivasan K, Ayalon G, Meilandt WJ, Lin H, Huntley MA, et al. Diverse brain myeloid expression profiles reveal distinct microglial activation states and aspects of Alzheimer’s disease not evident in mouse models. Cell Reports. 2018;22(3):832-847. DOI: 10.1016/j.celrep.2017.12.066
  55. 55. Turnbull IR, Gilfillan S, Cella M, Aoshi T, Miller M, Piccio L, et al. Cutting edge: TREM-2 attenuates macrophage activation. Journal of Immunology. 2006;177(6):3520-3524. DOI: 10.4049/jimmunol.177.6.3520
  56. 56. Hamerman JA, Jarjoura JR, Humphrey MB, Nakamura MC, Seaman WE, Lanier LL. Cutting edge: Inhibition of TLR and FcR responses in macrophages by triggering receptor expressed on myeloid cells (TREM)-2 and DAP12. Journal of Immunology. 2006;177(4):2051-2055. DOI: 10.4049/jimmunol.177.4.2051
  57. 57. N'Diaye EN, Branda CS, Branda SS, Nevarez L, Colonna M, Lowell C, et al. TREM-2 (triggering receptor expressed on myeloid cells 2) is a phagocytic receptor for bacteria. The Journal of Cell Biology. 2009;184(2):215-223. DOI: 10.1083/jcb.200808080
  58. 58. Han X, Zhou Z, Fei L, Sun H, Wang R, Chen Y, et al. Construction of a human cell landscape at single-cell level. Nature. 2020;581(7808):303-309. DOI: 10.1038/s41586-020-2157-4
  59. 59. Jaitin DA, Adlung L, Thaiss CA, Weiner A, Li B, Descamps H, et al. Lipid-associated macrophages control metabolic homeostasis in a Trem2-dependent manner. Cell. 2019;178(3):686-698 e14. DOI: 10.1016/j.cell.2019.05.054
  60. 60. Kober DL, Brett TJ. TREM2-ligand interactions in health and disease. Journal of Molecular Biology. 2017;429(11):1607-1629. DOI: 10.1016/j.jmb.2017.04.004
  61. 61. 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(122):ra38. DOI: 10.1126/scisignal.2000500
  62. 62. Otero K, Shinohara M, Zhao H, Cella M, Gilfillan S, Colucci A, et al. TREM2 and beta-catenin regulate bone homeostasis by controlling the rate of osteoclastogenesis. Journal of Immunology. 2012;188(6):2612-2621. DOI: 10.4049/jimmunol.1102836
  63. 63. Ulland TK, Song WM, Huang SC, Ulrich JD, Sergushichev A, Beatty WL, et al. TREM2 maintains microglial metabolic fitness in Alzheimer’s disease. Cell. 2017;170(4):649-663 e13. DOI: 10.1016/j.cell.2017.07.023
  64. 64. Klunemann HH, Ridha BH, Magy L, Wherrett JR, Hemelsoet DM, Keen RW, et al. The genetic causes of basal ganglia calcification, dementia, and bone cysts: DAP12 and TREM2. Neurology. 2005;64(9):1502-1507. DOI: 10.1212/01.WNL.0000160304.00003.CA
  65. 65. Otero K, Turnbull IR, Poliani PL, Vermi W, Cerutti E, Aoshi T, et al. Macrophage colony-stimulating factor induces the proliferation and survival of macrophages via a pathway involving DAP12 and beta-catenin. Nature Immunology. 2009;10(7):734-743. DOI: 10.1038/ni.1744
  66. 66. Poliani PL, Wang Y, Fontana E, Robinette ML, Yamanishi Y, Gilfillan S, et al. TREM2 sustains microglial expansion during aging and response to demyelination. The Journal of Clinical Investigation. 2015;125(5):2161-2170. DOI: 10.1172/JCI77983
  67. 67. 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: 10.1016/j.cell.2018.05.003
  68. 68. Kleinberger G, Brendel M, Mracsko E, Wefers B, Groeneweg L, Xiang X, et al. The FTD-like syndrome causing TREM2 T66M mutation impairs microglia function, brain perfusion, and glucose metabolism. EMBO Journal. 2017;36(13):1837-1853. DOI: 10.15252/embj.201796516
  69. 69. 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(5):979-991 e8. DOI: 10.1016/j.immuni.2018.04.016
  70. 70. Wang ECE, Dai Z, Ferrante AW, Drake CG, Christiano AM. A subset of TREM2(+) dermal macrophages secretes oncostatin M to maintain hair follicle stem cell quiescence and inhibit hair growth. Cell Stem Cell. 2019;24(4):654-669 e6. DOI: 10.1016/j.stem.2019.01.011
  71. 71. Saxton RA, Sabatini DM. mTOR signaling in growth, metabolism, and disease. Cell. 2017;168(6):960-976. DOI: 10.1016/j.cell.2017.02.004
  72. 72. Takahashi K, Rochford CD, Neumann H. Clearance of apoptotic neurons without inflammation by microglial triggering receptor expressed on myeloid cells-2. The Journal of Experimental Medicine. 2005;201(4):647-657. DOI: 10.1084/jem.20041611
  73. 73. Yeh FL, Wang Y, Tom I, Gonzalez LC, Sheng M. TREM2 binds to apolipoproteins, including APOE and CLU/APOJ, and thereby facilitates uptake of amyloid-beta by microglia. Neuron. 2016;91(2):328-340. DOI: 10.1016/j.neuron.2016.06.015
  74. 74. 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 (Albany NY). 2020;12(20):20862, 10.18632/aging.104104-20879
  75. 75. Chen S, Peng J, Sherchan P, Ma Y, Xiang S, Yan F, et al. TREM2 activation attenuates neuroinflammation and neuronal apoptosis via PI3K/Akt pathway after intracerebral hemorrhage in mice. Journal of Neuroinflammation. 2020;17(1):168. DOI: 10.1186/s12974-020-01853-x
  76. 76. Ruganzu JB, Zheng Q, Wu X, He Y, Peng X, Jin H, et al. TREM2 overexpression rescues cognitive deficits in APP/PS1 transgenic mice by reducing neuroinflammation via the JAK/STAT/SOCS signaling pathway. Experimental Neurology. 2021;336:113506. DOI: 10.1016/j.expneurol.2020.113506
  77. 77. Xu H, Mu S, Qin W. Microglia TREM2 is required for electroacupuncture to attenuate neuroinflammation in focal cerebral ischemia/reperfusion rats. Biochemical and Biophysical Research Communications. 2018;503(4):3225-3234. DOI: 10.1016/j.bbrc.2018.08.130
  78. 78. Karch CM, Goate AM. Alzheimer's disease risk genes and mechanisms of disease pathogenesis. Biological Psychiatry. 2015;77(1):43-51. DOI: 10.1016/j.biopsych.2014.05.006
  79. 79. Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, Majounie E, et al. TREM2 variants in Alzheimer's disease. The New England Journal of Medicine. 2013;368(2):117-127. DOI: 10.1056/NEJMoa1211851
  80. 80. Piers TM, Cosker K, Mallach A, Johnson GT, Guerreiro R, Hardy J, et al. A locked immunometabolic switch underlies TREM2 R47H loss of function in human iPSC-derived microglia. The FASEB Journal. 2020;34(2):2436-2450. DOI: 10.1096/fj.201902447R
  81. 81. 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(1):29. DOI: 10.1186/s13024-018-0262-8
  82. 82. Zhou Y, Song WM, Andhey PS, Swain A, Levy T, Miller KR, et al. Human and mouse single-nucleus transcriptomics reveal TREM2-dependent and TREM2-independent cellular responses in Alzheimer's disease. Nature Medicine. 2020;26(1):131-142. DOI: 10.1038/s41591-019-0695-9
  83. 83. McQuade A, Kang YJ, Hasselmann J, Jairaman A, Sotelo A, Coburn M, et al. Gene expression and functional deficits underlie TREM2-knockout microglia responses in human models of Alzheimer’s disease. Nature Communications. 2020;11(1):5370. DOI: 10.1038/s41467-020-19227-5
  84. 84. 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(1):74. DOI: 10.1186/s13024-017-0216-6
  85. 85. Leyns CEG, Gratuze M, Narasimhan S, Jain N, Koscal LJ, Jiang H, et al. TREM2 function impedes tau seeding in neuritic plaques. Nature Neuroscience. 2019;22(8):1217-1222. DOI: 10.1038/s41593-019-0433-0
  86. 86. 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(5):667-675. DOI: 10.1084/jem.20151948
  87. 87. Meilandt WJ, Ngu H, Gogineni A, Lalehzadeh G, Lee SH, Srinivasan K, et al. Trem2 deletion reduces late-stage amyloid plaque accumulation, elevates the Abeta42:Abeta40 ratio, and exacerbates axonal dystrophy and dendritic spine loss in the PS2APP Alzheimer's mouse model. The Journal of Neuroscience. 2020;40(9):1956-1974. DOI: 10.1523/JNEUROSCI.1871-19.2019
  88. 88. 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(5):1032-1048 e5. DOI: 10.1016/j.neuron.2018.02.002
  89. 89. Claes C, Van Den Daele J, Boon R, Schouteden S, Colombo A, Monasor LS, et al. Human stem cell-derived monocytes and microglia-like cells reveal impaired amyloid plaque clearance upon heterozygous or homozygous loss of TREM2. Alzheimer's & Dementia. 2019;15(3):453-464. DOI: 10.1016/j.jalz.2018.09.006
  90. 90. Yi S, Jiang X, Tang X, Li Y, Xiao C, Zhang J, et al. IL-4 and IL-10 promotes phagocytic activity of microglia by up-regulation of TREM2. Cytotechnology. 2020;72(4):589-602. DOI: 10.1007/s10616-020-00409-4
  91. 91. Lewcock JW, Schlepckow K, Di Paolo G, Tahirovic S, Monroe KM, Haass C. Emerging microglia biology defines novel therapeutic approaches for Alzheimer's disease. Neuron. 2020;108(5):801-821. DOI: 10.1016/j.neuron.2020.09.029
  92. 92. Sims R, van der Lee SJ, Naj AC, Bellenguez C, Badarinarayan N, Jakobsdottir J, et al. Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer's disease. Nature Genetics. 2017;49(9):1373-1384. DOI: 10.1038/ng.3916
  93. 93. Andreone BJ, Przybyla L, Llapashtica C, Rana A, Davis SS, van Lengerich B, et al. Alzheimer’s-associated PLCgamma2 is a signaling node required for both TREM2 function and the inflammatory response in human microglia. Nature Neuroscience. 2020;23(8):927-938. DOI: 10.1038/s41593-020-0650-6
  94. 94. Magno L, Lessard CB, Martins M, Lang V, Cruz P, Asi Y, et al. Alzheimer's disease phospholipase C-gamma-2 (PLCG2) protective variant is a functional hypermorph. Alzheimer’s Research & Therapy. 2019;11(1):16. DOI: 10.1186/s13195-019-0469-0
  95. 95. Takalo M, Wittrahm R, Wefers B, Parhizkar S, Jokivarsi K, Kuulasmaa T, et al. The Alzheimer's disease-associated protective Plcgamma2-P522R variant promotes immune functions. Molecular Neurodegeneration. 2020;15(1):52. DOI: 10.1186/s13024-020-00402-7
  96. 96. Zhao Y, Wu X, Li X, Jiang LL, Gui X, Liu Y, et al. TREM2 Is a receptor for beta-amyloid that mediates microglial function. Neuron. 2018;97(5):1023-1031 e7. DOI: 10.1016/j.neuron.2018.01.031
  97. 97. Atagi Y, Liu CC, Painter MM, Chen XF, Verbeeck C, Zheng H, et al. Apolipoprotein E is a ligand for triggering receptor expressed on myeloid cells 2 (TREM2). The Journal of Biological Chemistry. 2015;290(43):26043-26050. DOI: 10.1074/jbc.M115.679043
  98. 98. 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(3):566-581 e9. DOI: 10.1016/j.immuni.2017.08.008
  99. 99. Serrano-Pozo A, Betensky RA, Frosch MP, Hyman BT. Plaque-associated local toxicity increases over the clinical course of Alzheimer disease. The American Journal of Pathology. 2016;186(2):375-384. DOI: 10.1016/j.ajpath.2015.10.010
  100. 100. Calandra S, Tarugi P, Speedy HE, Dean AF, Bertolini S, Shoulders CC. Mechanisms and genetic determinants regulating sterol absorption, circulating LDL levels, and sterol elimination: Implications for classification and disease risk. Journal of Lipid Research. 2011;52(11):1885-1926. DOI: 10.1194/jlr.R017855
  101. 101. Nugent AA, Lin K, van Lengerich B, Lianoglou S, Przybyla L, Davis SS, et al. TREM2 regulates microglial cholesterol metabolism upon chronic phagocytic challenge. Neuron. 2020;105(5):837-854 e9. DOI: 10.1016/j.neuron.2019.12.007
  102. 102. Damisah EC, Rai A, Grutzendler J. TREM2: Modulator of lipid metabolism in microglia. Neuron. 2020;105(5):759-761. DOI: 10.1016/j.neuron.2020.02.008
  103. 103. Fitz NF, Wolfe CM, Playso BE, Biedrzycki RJ, Lu Y, Nam KN, et al. Trem2 deficiency differentially affects phenotype and transcriptome of human APOE3 and APOE4 mice. Molecular Neurodegeneration. 2020;15(1):41. DOI: 10.1186/s13024-020-00394-4
  104. 104. Stephen TL, Cacciottolo M, Balu D, Morgan TE, LaDu MJ, Finch CE, et al. APOE genotype and sex affect microglial interactions with plaques in Alzheimer's disease mice. Acta Neuropathologica Communications. 2019;7(1):82. DOI: 10.1186/s40478-019-0729-z
  105. 105. 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(2):191-204. DOI: 10.1038/s41593-018-0296-9
  106. 106. Fitz NF, Nam KN, Wolfe CM, Letronne F, Playso BE, Iordanova BE, et al. Phospholipids of APOE lipoproteins activate microglia in an isoform-specific manner in preclinical models of Alzheimer’s disease. Nature Communications. 2021;12(1):3416. DOI: 10.1038/s41467-021-23762-0
  107. 107. Huang YA, Zhou B, Nabet AM, Wernig M, Sudhof TC. Differential signaling mediated by ApoE2, ApoE3, and ApoE4 in human neurons parallels Alzheimer's disease risk. The Journal of Neuroscience. 2019;39(37):7408-7427. DOI: 10.1523/JNEUROSCI.2994-18.2019
  108. 108. Butovsky O, Weiner HL. Microglial signatures and their role in health and disease. Nature Reviews. Neuroscience. 2018;19(10):622-635. DOI: 10.1038/s41583-018-0057-5
  109. 109. Boza-Serrano A, Ruiz R, Sanchez-Varo R, Garcia-Revilla J, Yang Y, Jimenez-Ferrer I, et al. Galectin-3, a novel endogenous TREM2 ligand, detrimentally regulates inflammatory response in Alzheimer's disease. Acta Neuropathologica. 2019;138(2):251-273. DOI: 10.1007/s00401-019-02013-z
  110. 110. 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(1):204. DOI: 10.1186/s12974-020-01878-2
  111. 111. Heslegrave A, Heywood W, Paterson R, Magdalinou N, Svensson J, Johansson P, et al. Increased cerebrospinal fluid soluble TREM2 concentration in Alzheimer's disease. Molecular Neurodegeneration. 2016;11:3. DOI: 10.1186/s13024-016-0071-x
  112. 112. Piccio L, Deming Y, Del-Aguila 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(6):925-933. DOI: 10.1007/s00401-016-1533-5
  113. 113. Suarez-Calvet M, Araque Caballero MA, Kleinberger G, Bateman RJ, Fagan AM, Morris JC, et al. Early changes in CSF sTREM2 in dominantly inherited Alzheimer's disease occur after amyloid deposition and neuronal injury. Science Translational Medicine. 2016;8(369):369ra178. DOI: 10.1126/scitranslmed.aag1767
  114. 114. Suarez-Calvet M, Morenas-Rodriguez E, Kleinberger G, Schlepckow K, Araque Caballero MA, Franzmeier N, et al. Early increase of CSF sTREM2 in Alzheimer's disease is associated with tau related-neurodegeneration but not with amyloid-beta pathology. Molecular Neurodegeneration. 2019;14(1):1. DOI: 10.1186/s13024-018-0301-5
  115. 115. Ma LZ, Tan L, Bi YL, Shen XN, Xu W, Ma YH, et al. Dynamic changes of CSF sTREM2 in preclinical Alzheimer's disease: The CABLE study. Molecular Neurodegeneration. 2020;15(1):25. DOI: 10.1186/s13024-020-00374-8
  116. 116. Zhong L, Xu Y, Zhuo R, Wang T, Wang K, Huang R, et al. Author correction: Soluble TREM2 ameliorates pathological phenotypes by modulating microglial functions in an Alzheimer's disease model. Nature Communications. 2019;10(1):2923. DOI: 10.1038/s41467-019-10950-2
  117. 117. Zhong L, Chen XF, Wang T, Wang Z, Liao C, Wang Z, et al. Soluble TREM2 induces inflammatory responses and enhances microglial survival. The Journal of Experimental Medicine. 2017;214(3):597-607. DOI: 10.1084/jem.20160844
  118. 118. Lee SC, Liu W, Brosnan CF, Dickson DW. GM-CSF promotes proliferation of human fetal and adult microglia in primary cultures. Glia. 1994;12(4):309-318. DOI: 10.1002/glia.440120407
  119. 119. Tomozawa Y, Inoue T, Takahashi M, Adachi M, Satoh M. Apoptosis of cultured microglia by the deprivation of macrophage colony-stimulating factor. Neuroscience Research. 1996;25(1):7-15. DOI: 10.1016/0168-0102(96)01021-8
  120. 120. Wu K, Byers DE, Jin X, Agapov E, Alexander-Brett J, Patel AC, et al. TREM-2 promotes macrophage survival and lung disease after respiratory viral infection. The Journal of Experimental Medicine. 2015;212(5):681-697. DOI: 10.1084/jem.20141732
  121. 121. Strittmatter WJ, Roses AD. Apolipoprotein E and Alzheimer’s disease. Annual Review of Neuroscience. 1996;19:53-77. DOI: 10.1146/annurev.ne.19.030196.000413
  122. 122. Franzmeier N, Suarez-Calvet M, Frontzkowski L, Moore A, Hohman TJ, Morenas-Rodriguez E, et al. Higher CSF sTREM2 attenuates ApoE4-related risk for cognitive decline and neurodegeneration. Molecular Neurodegeneration. 2020;15(1):57. DOI: 10.1186/s13024-020-00407-2
  123. 123. Ewers M, Biechele G, Suarez-Calvet M, Sacher C, Blume T, Morenas-Rodriguez E, et al. Higher CSF sTREM2 and microglia activation are associated with slower rates of beta-amyloid accumulation. EMBO Molecular Medicine. 2020;12(9):e12308. DOI: 10.15252/emmm.202012308
  124. 124. Hardy J, Allsop D. Amyloid deposition as the central event in the aetiology of Alzheimer's disease. Trends in Pharmacological Sciences. 1991;12(10):383-388. DOI: 10.1016/0165-6147(91)90609-v
  125. 125. Hardy JA, Higgins GA. Alzheimer's disease: The amyloid cascade hypothesis. Science. 1992;256(5054):184-185. DOI: 10.1126/science.1566067
  126. 126. Lannfelt L, Relkin NR, Siemers ER. Amyloid-ss-directed immunotherapy for Alzheimer's disease. Journal of Internal Medicine. 2014;275(3):284-295. DOI: 10.1111/joim.12168
  127. 127. Small SA, Duff K. Linking Abeta and tau in late-onset Alzheimer's disease: A dual pathway hypothesis. Neuron. 2008;60(4):534-542. DOI: 10.1016/j.neuron.2008.11.007
  128. 128. Leng F, Edison P. Neuroinflammation and microglial activation in Alzheimer disease: Where do we go from here? Nature Reviews. Neurology. 2021;17(3):157-172. DOI: 10.1038/s41582-020-00435-y
  129. 129. Grimaldi LM, Zappala G, Iemolo F, Castellano AE, Ruggieri S, Bruno G, et al. A pilot study on the use of interferon beta-1a in early Alzheimer's disease subjects. Journal of Neuroinflammation. 2014;11:30. DOI: 10.1186/1742-2094-11-30
  130. 130. Zheng C, Zhou XW, Wang JZ. The dual roles of cytokines in Alzheimer's disease: Update on interleukins, TNF-alpha, TGF-beta and IFN-gamma. Translational Neurodegeneration. 2016;5:7. DOI: 10.1186/s40035-016-0054-4
  131. 131. Wang S, Mustafa M, Yuede CM, Salazar SV, Kong P, Long H, et al. Anti-human TREM2 induces microglia proliferation and reduces pathology in an Alzheimer's disease model. Journal of Experimental Medicine. 2020;217(9):e20200785. DOI: 10.1084/jem.20200785
  132. 132. Li C, Zhao B, Lin C, Gong Z, An X. TREM2 inhibits inflammatory responses in mouse microglia by suppressing the PI3K/NF-kappaB signaling. Cell Biology International. 2019;43(4):360-372. DOI: 10.1002/cbin.10975
  133. 133. 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(4):e11227. DOI: 10.15252/emmm.201911227
  134. 134. Wunderlich P, Glebov K, Kemmerling N, Tien NT, Neumann H, Walter J. Sequential proteolytic processing of the triggering receptor expressed on myeloid cells-2 (TREM2) protein by ectodomain shedding and gamma-secretase-dependent intramembranous cleavage. The Journal of Biological Chemistry. 2013;288(46):33027-33036. DOI: 10.1074/jbc.M113.517540
  135. 135. Kleinberger G, Yamanishi Y, Suarez-Calvet M, Czirr E, Lohmann E, Cuyvers E, et al. TREM2 mutations implicated in neurodegeneration impair cell surface transport and phagocytosis. Science Translational Medicine. 2014;6(243):243ra86. DOI: 10.1126/scitranslmed.3009093
  136. 136. Xiang X, Werner G, Bohrmann B, Liesz A, Mazaheri F, Capell A, et al. TREM2 deficiency reduces the efficacy of immunotherapeutic amyloid clearance. EMBO Molecular Medicine. 2016;8(9):992-1004. DOI: 10.15252/emmm.201606370
  137. 137. Howard R, Liu KY. Questions EMERGE as biogen claims aducanumab turnaround. Nature Reviews. Neurology. 2020;16(2):63-64. DOI: 10.1038/s41582-019-0295-9
  138. 138. Kurosawa Y, Degrauw TJ, Lindquist DM, Blanco VM, Pyne-Geithman GJ, Daikoku T, et al. Cyclocreatine treatment improves cognition in mice with creatine transporter deficiency. The Journal of Clinical Investigation. 2012;122(8):2837-2846. DOI: 10.1172/JCI59373
  139. 139. Da Mesquita S, Louveau A, Vaccari A, Smirnov I, Cornelison RC, Kingsmore KM, et al. Functional aspects of meningeal lymphatics in ageing and Alzheimer's disease. Nature. 2018;560(7717):185-191. DOI: 10.1038/s41586-018-0368-8
  140. 140. Da Mesquita S, Papadopoulos Z, Dykstra T, Brase L, Farias FG, Wall M, et al. Meningeal lymphatics affect microglia responses and anti-Abeta immunotherapy. Nature. 2021;593(7858):255-260. DOI: 10.1038/s41586-021-03489-0
  141. 141. Mazanetz MP, Fischer PM. Untangling tau hyperphosphorylation in drug design for neurodegenerative diseases. Nature Reviews. Drug Discovery. 2007;6(6):464-479. DOI: 10.1038/nrd2111
  142. 142. Wang JZ, Gong CX, Zaidi T, Grundke-Iqbal I, Iqbal K. Dephosphorylation of Alzheimer paired helical filaments by protein phosphatase-2A and -2B. The Journal of Biological Chemistry. 1995;270(9):4854-4860. DOI: 10.1074/jbc.270.9.4854
  143. 143. Zheng J, Tian N, Liu F, Zhang Y, Su J, Gao Y, et al. A novel dephosphorylation targeting chimera selectively promoting tau removal in tauopathies. Signal Transduction and Targeted Therapy. 2021;6(1):269. DOI: 10.1038/s41392-021-00669-2

Written By

Siwei Xu, Yaya Ji, Tianle Sha and Haoming Li

Submitted: 05 August 2021 Reviewed: 30 August 2021 Published: 20 September 2021