Skip to main content

Noteworthy perspectives on microglia in neuropsychiatric disorders

Abstract

Microglia are so versatile that they not only provide immune surveillance for central nervous system, but participate in neural circuitry development, brain blood vessels formation, blood–brain barrier architecture, and intriguingly, the regulation of emotions and behaviors. Microglia have a profound impact on neuronal survival, brain wiring and synaptic plasticity. As professional phagocytic cells in the brain, they remove dead cell debris and neurotoxic agents via an elaborate mechanism. The functional profile of microglia varies considerately depending on age, gender, disease context and other internal or external environmental factors. Numerous studies have demonstrated a pivotal involvement of microglia in neuropsychiatric disorders, including negative affection, social deficit, compulsive behavior, fear memory, pain and other symptoms associated with major depression disorder, anxiety disorder, autism spectrum disorder and schizophrenia. In this review, we summarized the latest discoveries regarding microglial ontogeny, cell subtypes or state spectrum, biological functions and mechanistic underpinnings of emotional and behavioral disorders. Furthermore, we highlight the potential of microglia-targeted therapies of neuropsychiatric disorders, and propose outstanding questions to be addressed in future research of human microglia.

Background

Microglia, as the only resident cells of mononuclear–phagocyte lineage in the brain parenchyma, account for approximately 5–10% of the central nervous system (CNS) cells in mice and 0.5–16.6% in humans, with a higher density in white matter than grey matter [1, 2]. Since the advent of sequencing and imaging techniques, ontogeny of microglia has been deciphered from hematopoietic progenitors, macrophages, early microglia, pre-microglia to adult microglia, as well as their turnover with aging [2,3,4,5,6]. The gene profile of microglia in most mammals are relatively conservative, while human microglia exhibit extensive heterogeneity that predominantly enriched in host defense and immune response modulation [7,8,9,10]. Mature microglia within the parenchyma cannot be sustained by circulating blood cells; instead, it is solely dependent on their self-renewal coupled with proliferation and apoptosis, enabling a whole repopulation of microglia once about every 100 days [2, 11]. Specially, retinal microglia are repopulated not only from residual microglia but also from macrophages in the ciliary body and iris [12]. The development, maturation and maintenance of microglia are regulated by complex signals, such as CSF1R, RUNX1, CX3CR1, TGF-β, IL-34, etc. [13,14,15,16,17]. Neurons and astrocytes also collaborate to preserve microglial ramified morphology and homeostatic gene signature [18,19,20].

Throughout lifespan, microglia perform pleiotropic biological functions through intricate interactions with other cells, including neurons, astrocytes, oligodendrocytes endothelial cells, T cells, etc. by either chemical signals or direct contact. Microglial dysfunction in these processes can lead to persistent disturbance in neurogenesis, blood–brain barrier (BBB) integrity, immune homeostasis and synaptic plasticity [21]. Other emerging discoveries of microglia, such as innate immune memory [22], self-renewal [11] and durotaxis (mechanosensing) [23], have revealed the multifaced role of microglia in different (patho)physiological conditions. As immunosensors, microglia possess abundant receptor repertoire binding with threat-induced endocrine hormones (e.g., glucocorticoids), immune mediators (e.g., DAMPs, IL-1), neural transmitters (e.g., norepinephrine, 5-HT); in turn, they release various immunological factors to impinge neuro-immune microenvironment [24]. As neural modulators, microglial impairment in neurogenesis, neuronal activity and synaptic remodeling leads to neural maladaptation during stress exposure [25], all of which puts microglia in the unique position linking biological or social stress with emotional and behavioral consequences. Based on mastering the advances in microglial biology, we further systematically discuss the notion that microglia play a critical role in the regulation of emotion and behavior, and explain how fetal exposure, lifestyle and other internal or external environmental stress impact microglia functional states, leading to the vulnerability or progression of neuropsychiatric disorders.

Microglial diversity and state spectrum

Microglia are distributed around neuronal somas, axons, dendrites and blood vessels, exhibiting spatially and temporally restricted profiles determined by both genetic and environmental factors [26,27,28,29,30]. Since microglia classification has entered an exquisite phase with single-cell sequencing (scRNA-seq), the dichotomized M1/M2 polarization framework has been gradually discarded [10, 31,32,33,34]. Here, microglial “subtypes” are described by the relatively stable molecular and functional profiles, while a vast landscape of microglial “states” is distinguished corresponding to different pathologies, disease stages, or even culture conditions.

Keratan sulfate proteoglycan (KSPG) microglia

A subpopulation with abundant expression of KSPG has a preferential distribution in the hippocampus, brainstem, olfactory bulb (OB), spinal cord and retina, though relatively little is understood about their function [10].

Hox8b microglia

Hox8b microglia is much lesser than non-Hoxb8 microglia, but their loss cannot be compensated by the latter [35]. This subtype is associated with abnormal behaviors. Ablation of Hox8b microglia can induce obsessive–compulsive disorder (OCD)-like and anxiety-like behaviors in mice [36,37,38]. However, optogenetic stimulation of Hox8b microglia in specific brain areas results in enhanced anxiety, compulsive grooming, or both abnormalities [39].

TREM2 microglia

In the brain, TREM2 is exclusively expressed by microglia regulating their survival, proliferation, metabolism and phagocytosis [40]. Whereas, not all microglia express TREM2, as demonstrated by the region-specific distribution in murine brain [41]. TREM2 microglia has been intensively investigated in response to amyloid β (Aβ) and tau [40, 42, 43], while recent studies revealed distinct responses of cortical and subcortical TREM2 microglia to the Alzheimer’s disease (AD) pathology [44]. This subpopulation also participates in TDP-43-associated neurodegeneration [45] and axonal injury repair [46].

CD11c microglia

This subpopulation is the major source of insulin-like growth factor 1 (IGF-1) that is vital to myelinogenesis and neurodevelopment [47, 48]. Although CD11c+ microglia, proliferative region-associated microglia (PAM), and axon tract-associated microglia (ATM) are described by different terms, they may represent the same population referred as “developmental CD11c+ microglia” [47, 49, 50]. During neurodegenerative settings, microglia are transformed into DAM, activated microglia, or microglial neurodegenerative phenotype (MGnD) associated with the brain pathology, all of which exhibit an upregulation of CD11c [48, 51,52,53].

Arginase-1 (ARG1) microglia

In physiological condition, ARG1 microglia is enriched in the basal forebrain and ventral striatum during early development, displaying upregulation of Apoe, Clec7a, Igf1, Mgl2 and Lgals3 compared to ARG1 negative microglia. This subpopulation plays a pivotal role in shaping cholinergic forebrain–hippocampus circuits involved in cognitive function [54]. ARG1-expressing microglia induced by IL-4 appear to be neuroprotective by restoring hippocampal neurogenesis associated with stress resistance [55] and enhancing Aβ clearance in the context of neuroinflammation [56].

Microglia supporting neurogenesis

In active neurogenetic niches, including OB, subventricular zone and rostral migratory stream, microglia can be TREM2-negative and Iba1-negative, and they persist beyond the ontogenetic period [57, 58]. In the subgranular zone of dentate gyrus, microglia expressing Clec7a is correlated with adult neurogenesis [49].

Repopulating microglia

Repopulating microglia cells demonstrate enhanced cell-cycle, migration and survival-related genes to support their rapid replenishment [11]. However, substantial microgliosis is only observed in the gray matter following diphtheria toxin-induced ablation but not in the white matter, indicating the regional heterogeneity of microglia repopulation [59].

Lipid droplet accumulating microglia (LDAMs)

This notion emerges from the “foamy macrophages” in atherosclerosis. These glial cells containing massive lipid droplets were found in the brains of aging [60], AD [61, 62], and diabetes-associated cognitive impairment [63], displaying pro-inflammatory states and phagocytic defects.

Microglia in neurodegeneration

Various microglia states were identified under different neurodegenerative contexts. Disease-associated microglia (DAMs) were first described as a protective phagocytic microglia population in AD, generated from homeostatic microglia through a two-step TREM2-independent and -dependent mechanism [64, 65]. Yet, proinflammatory DAMs driven by receptor-interacting protein kinase 1 (RIPK1) also existed and were deleterious in the etiology of AD [66, 67]. Silvin et al. further identified embryonically derived TREM2-dependent DAM with neuroprotective signature and monocyte-derived TREM2-independent disease inflammatory macrophages (DIMs) accumulating in aging brain [68]. Activated response microglia (ARMs) were detected in Aβ (but not tau) models exhibiting upregulation of Apoe, Clec7a, MHC class II and putative tissue repair genes, which might be the converging point of AD risk factors [69, 70]. In hippocampal CA1 and temporal cortex, Aβ plaque-associated microglia were positive for all NLRP3 inflammasome components and cleaved gasdermin D, presenting pyroptosis activation in microglia responding to amyloidosis [71]. Interferon response microglia (IRMs) were defined from Aβ models by enhanced interferon type I pathways, including Ifit2, Ifit3, Ifitm3 and Irf7 [70, 72]. They displayed efficiency at restricting Aβ accumulation in AD mice [72], but also existed in young wild-type mice and increased over normal aging [70]. Pathology-specific AD1–microglia subgroups localized to Aβ and AD2–microglia subgroups associated with p-tau [73], as well as temporally specific early stage AD-associated microglia (EADAMs) and late-stage AD-associated microglia (LADAMs) [74], were identified by distinct genetic and functional signatures.

In addition to AD, MGnD was defined across AD, amyotrophic lateral sclerosis (ALS) and multiple sclerosis (MS) associated with neurodegenerative progression [75]. A subtype with a distinct ferroptosis-associated transcriptomic signature was enriched in the spinal cord of ALS and midbrain of Parkinson’s disease (PD) patients [76]. In the nigra of PD patients, microglia demonstrated a proinflammatory phenotype as a part of “pan-glial” activation network [77]. These findings revealed markable plasticity of microglial states, yet whether they are strictly correlated with certain diseases or more universal core properties, and whether they are conserved in humans, still need exploration [34, 43].

Senescent microglia

Microglial senescence was characterized by less ramified, reduced process motility and increased inflammatory responses to brain injury [78,79,80,81]. Age-associated changes in human microglia were enriched in axonal guidance, cell adhesion, actin (dis)assembly and surface receptor expression, and they can engulf the dead neurons in aged brain [8, 82].

Microglia in vitro

Mature microglia lose their signature (Tmem119, P2ry12, Sall1) and dedifferentiate within hours of isolation. Cultured microglia exhibit downregulated inflammation but upregulated cell-cycle and complement-related proteins [16]. Microglial profile in vitro cannot perfectly mimic the situation in vivo, for which stringent experimental conditions (e.g., astrocyte-conditioned medium) are required to maintain microglial homeostasis in vitro [83,84,85,86].

Biological functions of microglia

Microglia in brain development and neurogenesis

Distinct from prevailing ramified microglia in the gray matter, a proliferative region-associated CD11c+ microglia subpopulation displaying amoeboid morphology is discovered in the white matter which phagocytoses newly generated oligodendrocytes or myelin sheath and engages in the myelinogenesis [49, 50]. They are highly efficient at the clearance of apoptotic debris produced by dead cells during brain development [87, 88]. Importantly, trophic support from the CNS cellular milieu is required for neurogenesis. In the developing cortex, microglia-derived IGF-1 is essential for the survival of layer V neurons [89]. These activities also occur in young adults, as the unchallenged microglia in the subgranular zone niche dynamically remodel hippocampal circuitry by phagocytosing apoptotic newborn cells [90]. After traumatic brain injury, repopulating microglia can robustly promote adult neurogenesis in the hippocampus through IL-6/sIL-6R signaling [91].

Immune surveillance and homeostasis

In healthy brains, microglia provide continuous surveillance for CNS microenvironment by extending and retracting their processes. They are sensitive to pathogen infections, damage-associated molecular patterns (DAMPs) and peripherally produced neurotoxins to maintain CNS homeostasis. Neuronal injury initiates long-range Ca2+ transients and ATP release as guidance cues for the migration of P2Y12-expressing microglia [92,93,94,95]. However, constant microglial ramification and surveillance are independent of purinergic signaling. Instead, it requires the tonic activity of several channels, such as TWIK and TRPV1 [96,97,98]. Another aspect worth considering is that both immune surveillance and response are highly energy-demanding processes, which are coupled to metabolic adaptations, such as glycolytic flux and mitochondrial activity in microglia [99]. Microglia participate in antiviral immune by initiating CD4 + T cells responses and cross-presenting antigen to CD8 + T cells that remove the infected neurons by noncytolytic mechanism [100,101,102]. At the early stage of viral infection, phosphatidylserine (PtdSer) serves as a “eat-me” signaling recognized by microglial TAM receptors and triggers the phagocytosis of compromised cells [94, 95]. Candida infection in the parenchyma evokes focal microgliosis and astrocytosis surrounding the fungal cells to form fungal-induced glial granuloma (FIGG), in which microglia release cytokines, such as IL-1β, IL-6, CXCL1 and TNF to enhance fungal clearance [103, 104]. Several non-infectious CNS diseases are also characterized by neuroinflammation driven by microglial sensors or pathways, such as RIPK1, interferon-induced protein 35 (IFP35) and Toll-like receptors (TLRs) [105,106,107].

Microglia regulate vascular architecture and cerebral blood flow

Microglial regulation of angiogenesis was corroborated in the retinas of mice and zebrafish, as they promote vascular anastomosis during developmental neovascularization via an ambiguous TGF-β-dependent but VEGFA-independent mechanism [108,109,110]. In adults, microglia retain the capability to regulate endothelial function in the presence of chronic activation of TGF-β signaling. For example, obesity can elicit microgliosis and activation of microglial TGF-β-activated kinase 1 (Tak1), which further increases the secretion of IL-18 that directly inhibits the activity of endothelial nitric oxide synthase (eNOS) and hence eliminates NO production, leading to cerebrovascular dysfunction [111]. Low oxygen concentration is another driving force for neovascularization, which is partly related to the recruitment of microglia towards the sites of endothelial cell anastomosis induced by MAS receptor (encoded by Mas1 gene) signaling [112]. He et al. reported a proangiogenic receptor-interacting protein 3 positive (RIP3+) microglia subpopulation that can be activated by hypoxia to release fibroblast growth factor 2 (FGF2) through RIP3-mediated necroptosis and associated with retinopathy [113]. Capillary-associated microglia (CAMs) are the bona fide microglia residing in the perivascular space. They have direct purinergic interactions with endothelium and periarterial smooth muscle cells, contributing to the maintenance of optimal capillary radius, vascular reactivity and cerebral blood flow, which may be particularly crucial in pathological conditions, such as hypoperfusion and hypercapnia [114, 115].

Microglia affect BBB permeability

Being a component of the neurovascular unit (NVU) and glial–vascular unit (GVU), ongoing studies indicate a critical role of microglia in the disruption of perivascular niche and BBB integrity [116, 117]. Microglia activated by ischemic stroke can secrete TNF, adipocyte fatty acid-binding protein (A-FABP) and metalloproteinase 9 (MMP-9) to trigger endothelial necroptosis and BBB leakage [118,119,120]. Research from Haruwaka and colleagues described the time-dependent dual effects of microglia on BBB permeability. Microglia initially migrate towards the cerebral blood vessels driven by CCR5 and protect BBB integrity by producing tight-junction protein Claudin-5 (CLDN5) and physically interacting with the endothelium. However, persistent inflammation boosts these vessel-associated microglia to engulf astrocytic end-feet and wreck BBB function [121].

Microglia as sculptors of synaptic plasticity

Microglia interact with synaptic structures throughout the lifespan, particularly in the first two postnatal weeks. During early development, microglial TREM2 critically controls the refinement of excessive synapses [122]. P2Y12 expressed on microglia is required for synaptic remodeling in the developing visual cortex, indicating the contribution of ATP to this process [123]. Complement system (C1q, CR3, CR5) is well-known to mediate microglial pruning and engulfment of synapses [124,125,126]. Yet in the barrel cortex, synapse elimination is CR3-independent but regulated by CX3CR1 [127]. Cytokine IL-33, a novel regulator of microglial activation, can be secreted by developing astrocytes or adult hippocampal neurons and promote microglia-dependent synapse remodeling under physiological conditions [128, 129]. Serotonin (5-HT) is another important signaling directing microglial pruning during neonatal phase. Impaired 5-HT sensing reduced phagolysosomal compartments in microglia and synapse–microglia proximity, leading to impaired synaptic and axonal sculpting in the hippocampus, cortex and thalamus [130]. In contrast, SIRPα–CD47 functions as “don’t eat me” signal to prevent excessive microglial engulfment for the maintenance of synaptic structural and functional homeostasis [131]. Recent work by Zhong et al. found the formation of TREM2–C1q complexes in human AD brains; in murine AD models, microglial TREM2 can bind with C1q to rescue synaptic loss [132]. The question remains as how distinct pathways converge or antagonize their effects on lifelong synaptic modeling, as well as how microglia interact with other cells, such as astrocytes and oligodendrocytes to modulate these processes.

Microglial phagocytosis and efferocytosis

Microglia can phagocytose surviving or dead cells, a three-step process characterized by recognition, phagosome formation and ingestion. During the development of cerebellum, microglia promote the phagocytosis-mediated death of Purkinje fibers and clearance of apoptotic cells induced by phagocytosis-related genes epigenetically restricted by polycomb repressive complex 2 (PRC2) [27]; in the hippocampus, microglial CD11b and DAP12 are involved in this process [133].

The mechanism whereby microglia scavenge cell debris is related to numerous candidate molecules, such as P2Y12, BAI1, TIM-4, TAM receptors (Tyro3, Axl, MerTK), CD22, CD36, SYK, and TREM2. These receptors act at different levels, including recognition, target biding, and phagosome formation [29, 134,135,136,137,138,139,140,141]. Receptor recognition launches signaling to form the phagocytic cups, and then phagosomes are formed and gradually mature by fusion and fission with endosomes and lysosomes endowed with essential molecular markers, such as Lamp1, Rab5 and Rab7 [142,143,144]. With the assistance of Slc37a2, hybrid vesicles (phagolysosomes) are formed and fused into “gastrosome” containing membrane fragments and cellular debris, which facilitates the cargo degradation by digestive enzymes, such as Cathepsins [145,146,147]. Insufficient microglial clearance of cell debris and neurotoxic agents plays an important role in the development of diseases, including emotional disorders, stroke, aging and AD [134,135,136, 138, 141].

Interactions between microglia and other CNS or peripheral cells

Microglia constantly sense and respond to different signals to ensure neuronal activity under tight check [148]. In physiological conditions, direct microglia–neuron communication exist at the membrane-to-membrane junctions between microglial P2Y12-expressing processes and neuronal somatic Kv2.1-clustering regions, located in the vicinity of neuronal mitochondria and coupled with neuronal status switch [149]. Node of Ranvier is a major site of direct microglia–neuron interaction associated with myelin regeneration [150]. Local mediators, such as purinergic, serotonergic and complement signaling have been widely investigated in microglia–neuron crosstalk across physiological or disease-specific contexts [151,152,153]. Recent studies addressed microglia as the “brake” to dampen neuronal activity. In the striatum, microglia inhibit D1 neuronal activation via an ATP/AMP/ADO/A1R-dependent negative feedback, which is critical to prevent seizure vulnerability [154]. They also constitutively release platelet-derived growth factor B (PDGFB) that promotes the expression of Kv4.3 channels on neurons to increase neuronal potassium currents, while disruption of this pathway elevates basal sympathetic tonicity and predisposes hypertension [155]. In the thalamic reticular nucleus (TRN), microglia send circadian ceramide signaling projecting at neurons to modulate wake–sleep transition [156].

Glia–glia interactions represent a delicate balance affecting neural functions in health and diseases [157]. Inflammatory mediators and mitochondrial fragments from microglia can convert astrocytes to a neurotoxic A1 phenotype that occurs in many neurological diseases [158,159,160]. Conversely, microglia-derived TGF-α acts via ErbB1 in astrocytes to limit their neurotoxic activities. In demyelinating diseases, microglial extracellular vesicles release multimodal and multitarget signaling mediators acting on astrocytes and oligodendrocyte precursor cells (OPCs) around myelin lesions [161]. AD pathology drives microglia to express IL-3Rα that renders them responsive to IL-3 constitutively produced by astrocytes, which endows microglia with an acute immune response and recruitment towards Aβ plaque and neurofibrillary p-tau [162].

Cell-to-cell transfer is an interesting process, whereby microglia facilitate either the clearance or propagation of pathological factors. The F-actin-dependent membranous tubular connections between microglia enable them to share the α-synuclein (α-syn) burden for effective degradation, accompanied by the donation of intact mitochondria from naïve to affected microglia to attenuate inflammatory reactions [163, 164]. Burden transfer also occurs between microglia and neurons, yet it may be deleterious, since microglia act as the carriers of Aβ migrating towards unaffected neurons [165].

Microglia interactions with peripheral immune cells were studied in infectious and autoimmune encephalopathy. Upon recovery from neurotropic flaviviruses infection, IFN-γ derived from CD8+ T cell provokes microglial activation and excessive synapse elimination, contributing to post-infectious cognitive sequelae [102]. Conversely, CNS immune microenvironment re-established by microglia can also be the trigger for T cell-associated neuroinflammatory diseases [102, 166, 167]. In addition, regulatory T cells (Treg)–microglia interaction enhances the reparative activity of microglia, subsequently promoting oligodendrogenesis and white matter repair after stroke [168]. Lymphocyte–microglia–astrocyte crosstalk was described in chronic MS, in which C1q-activated microglia inflamed in MS (MIMS) and astrocytes inflamed in MS form a central hub to connect with other immune clusters [169]. In AD pathogenesis, meningeal lymphatic drainage is coupled with microglial activation and neurovascular response [170]. Together, interaction between microglia and other cells is ubiquitous at both physiological and pathological states; therefore, microglia dysfunction may have a domino effect on the entire CNS cellular network which demands further study.

Microglial priming and innate immune memory

Innate immune memory refers to that microglia exposed to a priming or desensitizing stimulus can react stronger (training) or weaker (tolerance) to the subsequent inflammatory insult [171, 172]. This has been observed in stress [173], aging [174, 175], optic nerve crush [176], MS [177], PD [178]and AD [179, 180]. Pregnant or neonatal mice that undergone inflammatory stimuli exhibit altered microglial responses upon a secondary disturbance during adulthood [181,182,183,184]. Early postnatal exposure to LPS primes microglial susceptibility to stress at adolescence, which enhances microglial engulfment of glutamatergic neuronal spines and depressive symptoms [185]. However, maternal immune activation (MIA) microglia in the offspring demonstrates a long-lasting decrease in immune reactivity (blunting) across the developmental trajectory [186]. In “tolerant” or “trained” microglia, the first stimulus endows them with epigenetic reprogramming that persists for at least 6 months [22, 187]. LPS-induced microglia priming is intimately linked to H3K4me3 and H3K27Ac peaks at promoters and enhancers, whereas the expression of tolerant genes correlated with ATAC and H3K27Ac peaks at enhancers [188]. Cerebral β-amyloidosis is exacerbated by immune training but alleviated by immune tolerance, which is modified by histone deacetylases-1/2 (HDAC1/2) [187].

Microglial neurotoxicity is usually aligned to primed microglia state due to their aggressive inflammatory state, while microglial tolerance provides sustained neuroprotection [189, 190]. However, the cytokine profile and phagocytotic activity of primed microglia should be taken into consideration, given that increased phagocytosis facilitates the clearance of cell debris, and the pleiotropic cytokines (e.g., IFN-β and IL-6) can be important for injury repair [174, 191]. This conclusion should not be made consistently but rather depend on the certain pathologies.

Microglial durotaxis

Mechanosensation of environmental cues, including extracellular protein burden and surface rigidity, can directly affect the level of immune response [192,193,194]. Microglia can sense the mechanical signals in their microenvironment and display a preference to migrate towards stiffer regions, termed as durotaxis [23]. The mechanical cues drive microglial adaptation of cytoskeletal composition and morphology to the surrounding stiffness, presenting spherical morphology with short lamellipodia on soft substrates, but more extended on stiff surface [192]. Multiple families of mechanosensors on microglia are identified [193, 195]. Piezo1 are highly expressed on microglia that can sense the stiffness of Aβ fibrils and engage in the compaction and engulfment of Aβ [196,197,198,199]. In retina, Kindlin3 is important for microglial to sense the stiffness of different layers [110] (see Fig. 1).

Fig. 1
figure 1

Multiple biological functions of microglia. A Microglia in brain development and neurogenesis. CD11c+ microglia are critical for neurodevelopment by providing pro-survival IGF-1 signaling for newborn neurons and oligodendrocytes. They also phagocytose apoptotic neurons, excessive myelin sheath and oligodendrocyte precursor cells to accomplish a structure–function equilibrium within neural circuits. B Immune surveillance and homeostasis. Microglia perform a dual-scale surveillance of CNS homeostasis by their processes sensitive to environmental perturbations. They are pivotal immune components in CNS infection and autoimmune diseases. C Microglia regulate angiogenesis and cerebral blood flow. Tissue stiffness and O2 concentration are two important cues for microglia to modulate angiogenesis throughout development. Capillary-associated microglia interact with brain vessels via purinergic signaling to maintain appropriate cerebral blood flow. D Dual-role of microglial in BBB integrity. Microglia can rescue BBB damage by secreting claudin 5 and phagocytosing infiltrated neutrophils to ameliorate further brain injury. On the other hand, they also secrete TNF-α to induce degradation of endothelial junctions, which aggravates BBB disruption. E Bi-directional role of microglia as sculptors of synaptic plasticity. Except for synaptic elimination via “eat me” signaling, microglia are also guardians of synaptic structures through “don’t eat me” signaling CD47–SIRPα. They engulf extracellular matrix deposition to support the formation and maturation of dendrite spines. F Microglial phagocytosis and efferocytosis. Microglia detect and phagocytose apoptotic neurons, myelin sheath and Aβ plaque by different receptors. Inside microglia, the phagosomes undergo hybrid phagolysosomes formation, gastrosomes fusion, and gastrosomes contraction to facilitate the scavenging of debris. G Interactions between microglia and other cells. Shared signaling pathways in microglia–other-cells crosstalk include purines, norepinephrine, serotonin, organelle component, complements and cytokines, which exerts profound effect on cellular survival, proliferation, metabolism, and functions. H Microglia durotaxis. Microglia react with the mechanical cues from neural tissue, Aβ plaque and foreign implants via mechanosensors, such as Piezos and integrins. The mechanism and significance of microglial durotaxis remain largely unknown

Microglia in behavioral and neuropsychiatric disorders

As discussed above, microglia perform a myriad of functions in brain development, synaptic plasticity, neuronal synchronization and neuroimmune homeostasis from embryo to adulthood. For neuropsychiatric disorders presenting emotional and behavioral abnormalities, microglia were initially suggested to reflect an immune response to relevant stress; however, recent genetic analyses revealed that a proportion of genetic risks exerts their impacts through myeloid cells, highlighting a causal role of microglia, the CNS resident myeloid cells, in the neuropsychiatric disorders [200, 201].

Depression and anxiety disorders

Depression is characterized by persistent sadness, anhedonia and diminished interest accompanied by daily function deficits [202]. Anxiety is arisen excessive fear and anticipation of real or imagined future threat, usually exhibiting avoidance behaviors and physical symptoms [203]. Depression and anxiety share genetic, neurobiological, and psychological risk mechanisms, including monoamine neurotransmission, hypothalamic–pituitary–adrenal axis changes, neuroinflammation, neuroplasticity, neurogenesis, etc., and they are often comorbid and difficult to distinguish, which causes more severe symptoms and treatment difficulties [202,203,204].

Inflammation is a common biological origin contributing to depression and anxiety. Innate and adaptive immune system dysregulation participate in symptom deterioration and compromise the curative effects of antidepressant and anti-anxiety treatments [205]. Microglia are the key initiators and regulators in CNS inflammatory cascade. Microglia activated by peripheral inflammation medium triggered an IL-6-mediated autocrine loop and the release of prostaglandin E2, which decreased the firing of striatal medium spiny neurons via delayed rectifying K+-channels, leading to negative affection [206]. In the model of post-stroke anxiety, HDAC3 was upregulated in the microglia of damaged cortex, which induced p65 deacetylation and the subsequent activation of NF-κB signaling involved in prostaglandin synthesis. Prostaglandin E2 released by cortical microglia could act on EP2 in the amygdala, potentially leading to the susceptibility to anxiety [207]. Discs large homolog 1 (Dlg1) is an adaptor protein regulating the activation of microglia, and Dlg1 knockout inhibited NF-κB and MAPK pathways in the microglia, leading to the alleviation of LPS-induced depression [208]. Adiponectin (APN), a circulatory hormone secreted by adipocytes, is associated with various neurological disorders. Studies demonstrated that APN deprivation exerted an antidepressant effect through regulating microglia NF-κB/Trk/BDNF signaling [209]. Glucagon-like peptide-1 receptor (GLP-1R) expressed on the microglia is another intriguing but understudied signaling with anti-inflammatory and neurotrophic properties [210]. Activation of GLP-1R ameliorated depression-like behaviors in diabetic db/db mice by promoting mitophagy in hippocampal microglia, which decreased ROS accumulation for final pyroptosis and pro-inflammatory cytokines release from microglia [211]. Interestingly, in animal model of neuropsychiatric systemic lupus erythematosus (NPSLE) that resembling symptoms, such as depression, anxiety and memory deficit, microglia were intrinsically activated in the presence of intact BBB during prenephritic stage, leading to increased IL-6 and IL-18 that elicited apoptosis of adult hippocampal neural stem cells and might gradually impair BBB integrity, indicating microglia as the spark of early neuropsychiatric diseases by altering neuroimmune microenvironment, which initiates BBB disruption and infiltration of peripheral immune components to provoke the disease [212].

An emerging understanding of “behavioral immune system” suggests that psychosocial stress can potentiate mental disorders through immunological and inflammatory pathways [213]. Wu et al. profiled the dorsolateral prefrontal cortex of non-human primates with depressive-like behaviors, showing that genetic alterations associated with depression-like behaviors predominantly enrich in microglia, and a subpopulation of pro-inflammatory microglia of depressive-like phenotypes (PIMID) was identified by upregulation of proinflammatory pathways and psychiatric risk genes, including BCL2 and SYNE1 [214]. In repeated social defeat stress (R-SDS) rodent models, IL-1α and TNF-α in mPFC microglia are upregulated in a TLR2/4-dependent manner, which gave rise to neuronal hypo-reactivity, dendritic atrophy and social avoidance [215]. Rather than TLRs, high mobility group box-1 protein (HMGB1) acts via receptor for advanced glycation endproducts (RAGE) on hippocampal microglia and causes anhedonia primed by chronic stress [216]. In the threat appraisal regions of the brain (prelimbic cortex, central amygdala, and hippocampus), R-SDS-induced microglia activation upregulates IL-1R1 on endothelial cells and hence facilitates monocyte recruitment, likely via chemokine gradient CCL2, to promote anxiety-like behaviors [217]. Along with stress accumulation, activation of NLRP3 inflammasome and its downstream inflammatory cytokines in hippocampal microglia contribute to the development of depressive-like behaviors. The mechanisms underlying NLRP3 inflammasome assembly could be associated with the activation of P2X7 receptors and increased serum glucocorticoid level, which activates glucocorticoid receptor (GR)/NF-κB/NLRP3 pathway [218,219,220]. During chronic mild stress, activation of microglial NLRP3 inflammasome triggers the release of A1 cocktail (TNF-α, IL-1α and C1q) to induce the production of A1-like astrocytes in hippocampus, which begins preceding to the onset of dendritic dysfunction and depression-like behaviors [221].

Microglia dysfunction in synaptic pruning and neuronal circuit modeling has been implicated in the development of depression and anxiety. Early life inflammation can prime microglia to perform enhanced CX3CR1-mediated engulfment of ACCGlu dendritic spines in response to later stress events, which causes the hypoactivity of ACCGlu and depressive vulnerability during adolescence [185]. At adulthood, neuron–microglia communication through CX3CR1–CX3CL1 also affects synaptic plasticity and neurogenesis in hippocampus associated with susceptibility to environmental stress and major depression [222, 223]. Chronic stress increases CSF1 secretion from neurons of mPFC to promote microglial phagocytosis of dendritic spines of pyramidal neurons, leading to depression-like and anxiety-like behaviors [224]. In contrast, early life adversity impairs microglial pruning of excitatory synapses onto stress-sensitive CRH-expressing neurons, provoking hormonal and behavioral stress responses in adulthood [184]. Under normal conditions, synaptic remodeling by microglia in the ventral hippocampal CA1 (vCA1) is active in the daytime, during which ROS and uncoupling protein 2 (UCP2) are produced. Knockout of UCP2 hinders the phased elimination of dendritic spines, resulting in disruption of hippocampal neural circuit and anxiety-like behaviors [225]. Fan et al. found that the exosomes secreted by microglia carrying mir-146a-5p are transported to the dentate gyrus, where they downregulates the expression of Krüppel-like factor 4 (KLF4) and cyclin-dependent kinase-like 5 (CDKL5), inhibits the proliferation and differentiation of neurons, and participates in the onset of depression [226].

Microglia could be the key mediator between negative affection and psychoactive substances, such as alcohol and tobacco. Adolescent ethanol consumption can drive the hyperactivation of microglia in raphe nuclei, leading to increased levels of TNF-α and IL-10, decreased serotonergic activity, and neuropsychiatric consequences of hyperalgesia and depression [227, 228]. Socodato et al. found that alcoholism activates microglia in the mPFC via Src/NF-κB/TNF pathway and selectively enhances the clearance of excitatory synapses, which inhibits nerve conduction and contributes to the anxiety-like behaviors [229]. Microglia depletion by CSF1R inhibitor reduces inhibitory GABAA and excitatory glutamate receptor-mediated transmission in the central nucleus of the amygdala (CeA) and improves alcohol-dependent anxiety [230]. During smoking cessation, increased expression of Nox2 and production of ROS from microglia in the nucleus accumbens potentiate the occurrence of nicotine withdrawal-related anxiety [231].

Although considerable researches have addressed the detrimental impact of microglial activation, the actual microglial role in human depression and anxiety remain largely unknown. Several translocator protein (TSPO) binding positron emission tomography (PET) studies demonstrated enhanced microglial activation in prefrontal cortex (PFC), ACC and insula of patients with major depressive disorder (MDD) [232,233,234]. However, evidence from post-mortem researches showed elevated homeostatic markers and unchanged or even inhibited inflammatory reactivity of microglia in MDD [37]. A distinct DAM phenotype, termed as DepDAM, was identified from the occipital cortex grey matter of MDD patients, displaying downregulated genes involved in immune response (C1QA/B/C, SPP1, MK167) and phagocytosis (CD14, CD163, FCGR1A/C, FCGR3A). Acquisition of this immune-suppressed status might be regulated by the increased “don’t eat me” CD200/CD47 signaling between neuron–microglia interaction [235]. A possible explanation is that reduced astrocytes, observed in post-mortem brains of MDD individuals, may cause disruption of BBB integrity and subsequently infiltration of monocytes into the CNS, which can also display increased TSPO in PET [236]; on the other hand, microglia activation may not persist but assume a role at specific stages of MDD [235].

Autism spectrum disorder (ASD)

ASD is a constellation of neurodevelopmental disorders characterized by early appearing social deficit, repetitive behaviors, restricted interests and sensory abnormality [237]. Hundreds of genes have been identified to decipher the genetic heterogeneity of ASD, pointing towards distinct biological processes, such as protein synthesis, chromatin remodeling, cell adhesion and synaptic function. Transcriptomic analysis provided corroborative evidence of microglial molecular perturbations preferentially in ASD patients [238]. Publication by Ma and colleagues revealed that disruption of microglial homeostasis impaired the supporting microenvironment of neural progenitor cells due to the loss of plasticity-related genes 3 (PRG3) triggering Wnt/β-catenin cascade, leading to abnormal neuronal plasticity during embryonic development and autistic behaviors at later stages [239].

Synaptic abnormality is a major hallmark of ASD, reflected as altered local protein synthesis, spine architecture and signaling transmission, and thus ASD is classified as a “synaptopathy” [240]. During brain development, synapses undergo substantial pruning and engulfment performed by microglia, which are essential for synaptic refinement and neural circuit maturation. It has been widely established that mutations lacking nuclear localization of phosphatase and tensin homolog deleted on chromosome ten (PTEN) are associated with ASD. Sarn et al. showed that cytoplasmic predominant–PTEN localization elicits excessive C1q-mediated synaptic pruning by microglia [241]. Engulfed synapses can be degraded by microglial autophagy, and depletion of autophagy-related gene 7 (Atg7) results in synaptic surplus and subsequent autistic behaviors due to impaired synaptosome degradation in microglia [242]. Patients aged in the developmental window coinciding with synaptic refinement (between 5 and 23 years) exhibits a significant decrease in TREM2 expression relative to healthy individuals. The absence of TREM2 attenuates microglia-mediated synapse elimination in CA1 accompanied by underconnectivity between prefrontal and hippocampal regions, contributing to the social defects and repetitive behavior in Trem2−/− mice. This study further supports the centrality of microglia in the nervous-immune crosstalk for correct brain development [122]. Among the neurotransmitter systems, serotonin (5-HT) is one of the longest standings in ASD etiology [243]. Microglia express 5-HT2B receptor throughout postnatal lifetime. During a critical developmental window from birth to P30, serotonergic signaling between synapses and microglia is pivotal for synaptic refinement and circuitry maturation involved in sociability and flexibility in novel environment [130]. The deficiency of glutaminase 1, an enzyme responsible for glutamate generation in the brain, can also impair microglial synapse pruning and results in ASD-like behaviors [244].

Fetal environmental exposure, especially MIA during pregnancy, is a long-term risk factor of ASD. Autistic behaviors such as social deficit, repetitiveness and anxiety have been observed in MIA-affected offspring [245,246,247]. Since prenatal BBB is not mature, maternal cytokines and pathogenic antibodies targeting fetal brain antigens can directly invade fetal CNS through placenta, or indirectly elicit the production of endogenous cytokines in fetus, which has profound impact on immune, neuronal and synaptic homeostasis of the offspring [247]. Human microglia progressively mature from gestational week 13, and by midgestation they have progressed towards homeostatic immune-sensors with the signature profiles of sensome, including Csf1R, Cx3cr1, Cxcl16, Ifngr1 and Ly86, rendering prenatal brain vulnerable to environmental disturbance during early development [248]. For example, contactin-associated protein-2 (CASPR2) is identified as a neuronal-surface target for anti-brain antibodies from the mothers of children with ASD [247]. Intrauterine exposure to CASPR2–IgGs leads to persistent microglial activation concurrent with abnormal location and synaptic loss of glutamatergic neurons in the somatosensory cortex, which contributes to the social deficits in adulthood [249]. Work from Rosin and colleagues demonstrated that embryotic hypothalamus is comprised of microglia that segregate into four subtypes distinct by unique gene signature, in which the Spp1+ population located along the third ventricle directly contact with nearby neural stem cells (NSCs). Maternal cold stress elevates the secretion of CCL3 and CCL4 from Spp1+ microglia, which alters the proliferation and differentiation of NSCs and decreases oxytocin neurons in paraventricular nucleus that associated with social behaviors [250]. In addition, patients with tuberous sclerosis (TSC), a genetic disorder with mutations in the Tsc1 or Tsc2, have a high prevalence of ASD. In Tsc2−/− mice undergoing early postnatal immune activation, social memory deficit correlates with a long-lasting upregulation of mTOR/IFN-β pathway in microglia, consistent with the observations in immune activated mouse model of microglia-restricted Tsc2 mutation, revealing the centrality of microglia in the synergistic interactions between early immune activation and genetic mutations associated with ASD [251].

Of note, the immune perturbations during pregnancy continuously impact microglia–synapse interaction in the offspring throughout lifetime. At both E17 and P60, MIA microglia exhibit upregulation of cellular protrusion/synaptogenic factors, such as catenin delta 2 (CTNND2), neuronal cell adhesion molecule 2 (NCAM2) and neurotrophic receptor tyrosine kinase 2 (NTRK2), suggesting that MIA drives lifelong disruption of microglial transcriptome towards an enhanced synaptogenic state. The aberrant microglial function coalesces with immature synaptic surplus of intrinsically bursting neurons in the mPFC and enhanced autistic-like behaviors in the MIA offspring, which can be reversed by microglia repopulation using CSF1R inhibitor [252]. In developing brain, deficient microglial elimination of hippocampal mossy fiber synapses is observed in MIA offspring with ASD phenotype. Moreover, voluntary exercise in adulthood ameliorates their synaptic and behavioral abnormalities by activating a portion of dentate granule cells that primes microglia to engulf weaker synapses [253]. Taken together, microglia sensing and responses to fetal stress put them in the unique position linking MIA with the susceptibility to ASD [247].

Dysregulation of protein synthesis is one of the recognized biochemical pathways underlying ASD pathophysiology. Mutations of several translation regulators, including eukaryotic initiation factor 4F (eIF4E), eIF4G and some upstream negative regulators of mTORC1 (PTEN, TSC1 and TSC2), have been reported associated with ASD [254,255,256]. The mechanistic underpinnings of these mutations remain largely unknown. Xu and colleagues elevated mRNA translation by overexpressing eIF4E and found that exaggerated protein synthesis in microglia, but not neurons, led to social deficit, repetitive behaviors, and cognitive defect in male mice. The authors further showed the decreased mobility of these microglia prevented them from clearing the synaptic surplus, which resulted in increased synapse density and excitation/inhibition (E/I) ratio in the cortex [257]. These findings indicate that microglia with abnormal protein metabolism could be the initiator indispensable to the onset of some ASD-related behaviors.

Schizophrenia

Schizophrenia is a psychiatric syndrome characterized by delusions, hallucinations, and negative symptoms, such as diminished motivation and expressiveness concomitant with cognitive deficits [258]. According to the unified gliocentric model of schizophrenia proposed by Dietz et al., microglia might be the key mediator between developmental risk factors (e.g., MIA and emotional maternal stress) and the vulnerability to schizophrenia. Early immune activation of microglia during brain development suppresses the proliferation and differentiation of glial progenitor cells, leading to delayed and deficient maturation of astrocytes and oligodendrocytes. Consequently, these glial impairments result in the disruption of white matter integrity and microenvironmental homeostasis in CNS, causing cerebral desynchronization and dysconnectivity [259].

MIA is a contributing factor underlying persistent high neuroimmune status in patients with schizophrenia [260]. Findings from in vitro cultures showed that co-culture with activated microglia compromised the metabolic pathways in developmental cortical interneuron (cIN) rather than the more expected immune disturbances, as indicated by impaired mitochondrial metabolism and synaptic formation. Moreover, after removal of activated microglia-conditioned medium, cINs from schizophrenia donors, but not healthy individuals, exhibited long-lasting metabolic deficits, suggesting an interaction between schizophrenia genetic backgrounds and inflammatory environment induced by microglial activation [261]. In addition, appropriately 25% of the schizophrenia patients have an incidence of catatonia, which might be triggered by activated microglia and low-grade inflammation in the white matter tracts [262].

Postmortem studies demonstrated that loss of dendritic spines in the PFC is a common neuropathological alteration in schizophrenia individuals. C4-mediated excessive synapse clearance is suggested to involve in the pathogenesis of schizophrenia. In individuals, the elevation of C4 level is significantly related to the increased TSPO in the brain [263]. Using schizophrenia–patient-derived neural cultures, Sellgreen and colleagues showed enhanced synapse uptake by microglia-like cells mediated by neuronal C4 deposition [264]. Likewise, C4a overexpression increased microglial pruning of synapses in the mPFC, rendering reduced synaptic density and altered behaviors in adult mice that resemble the negative symptoms experienced by schizophrenia individuals [265]. Given these findings, interventions targeting microglial immune response or synapse elimination merit further research for the prevention or treatment of schizophrenia.

Fear memory

Fear memory assumes a central role in the onset and development of trauma and stress-related neuropsychiatric disorders, such as post-traumatic stress disorder (PTSD) [266]. Transcriptomic analysis displayed the persistent gene expression programs in microglia supporting the consolidation of remote fear memory, specifically enriched in innate immunity (Il6r, Il1a, Stat6, Csf3r, CD86, etc.), cytoskeletal rearrangement and adhesion maintenance pathways (Cdc42, Vasp, Rhoa, Rhoh, Prkcd, etc.), implying that enhancement of inflammatory response and cell migration might be entangled in the maintenance of fear memory [267]. Nguyen and colleagues revealed that the synapse remodeling associated with memory encoding is governed by neuron–microglia signaling via IL-33. IL-33 is secreted by hippocampal neurons in an experience-dependent manner to prime microglial phagocytosis of extracellular matrix (ECM), and hence the loss of IL-33 results in accumulation of ECM surrounding synapses and decrease in newborn neurons, consequently leading to impaired synaptic plasticity and diminished remote fear memories [129]. In the dentate gyrus, microglia were shown to eliminate the dendritic spines of engram cells via C1q, promoting the disassociation of engrams and erasure of contextual fear memory [268].

Pain

The International Association for the Study of Pain defines pain as “an unpleasant emotional experience associated with, or resembling that associated with, actual or potential tissue damage”. Physical, mental or emotional pain have expanded the definition of pain and recently all of these have been included in personal emotional experience [269]. It is becoming clear that spinal cord microglia play an critical role in chronic neuropathic pain [270]. In human spinal cord undergoing peripheral nerve injury, there is a large subpopulation of microglia exhibiting a transcriptional profile (ApoE, Lpl, Spp1, CD63 and CTSB) that remarkably similar to the previously described injury-responsive microglia (IRM) and proliferating DAM/ATM. Particularly, the upregulation of ApoE might represent a central switcher controlling the transcriptional shift from immunoreactive microglia towards a metabolically altered state [271]. These activated microglia degrade the perineuronal nets (PNNs) that selectively enwrap the spinal parabrachial projection neurons responsible for nociceptive information integration and transduction in the dorsal horn, and thus they can augment the output of spinal nociceptive circuitry [272]. Persistent synaptic potentiation change also contributes to the chronic hyperalgesia, which is triggered by neuronal released neuromodulators (SP, ATP, BDNF, CASP6, or CSF1). Spinal long-term potentiation (LTP) at C-fiber synapses is hypothesized to underlie the chronic pain. High frequency stimulation (HFS) or sciatic nerve injury can induce LTP at the C-fiber and upregulate CSF1 only in calcitonin gene-related peptide (CGRP+) terminals and CSF1R on spinal microglia. Subsequently, this CSF1–microglial BDNF signaling increases the number of CGRP terminals in the spinal dorsal horn. CGRP+ C-terminals are critically involved in the initiation and maintenance of microglial activation in spinal dorsal horn. In turn, selective deletion of spinal microglia blocks the high-frequency induced LTP and chronic pain hypersensitivity. In this crosstalk, CSF1 and BDNF signaling in microglia are indispensable for spinal LTP and chronic pain [273]. CD11c+ microglia seem to exert pleiotropic effects on the neuropathic pain. In nerve-injured mice, CD11c+ microglial ablation hinders the recovery from pain hypersensitivity, whereas pain-recovered mice with depletion of CD11c+ microglia relapses in hyperalgesia. The authors further defined IGF1 signaling of CD11c+ microglia as the key regulator in the dynamic equilibrium between ongoing pain facilitation and repression, which might be the determinant for the remission and recurrence of pain [274]. Given the heterogeneity of microglia in neuropathic pain, further studies will apply microglial manipulation tools, including chemo- or optogenetic selective activation, to investigate their function in pain, aversion, and comorbidities during chronic pain diseases [275].

Compulsive or feeding disorders

Feeding and eating disorders include anorexia nervosa, bulimia nervosa, binge eating disorder, avoidant-restrictive food intake disorder, pica and rumination disorder, which are often companied with obsessive–compulsive or autism spectrum traits [276]. Within the CNS, hypothalamus controls energy metabolism but also directly senses and responds to peripheral hormones and glycolipid metabolites [277,278,279]. The interaction between neurons and microglia in hypothalamus has been shown to be engage with body weight homeostasis and diet-induced obesity (DIO) [280]. UCP2 is highly expressed in activated microglial cells in DIO accompanied by abnormal mitochondrial morphology. Microglial UCP2 deficiency prevents mitochondrial morphological alterations and dysfunction in the arcuate nucleus induced by high-fat diet (HFD), ultimately decreasing the susceptibility to obesity by remodeling the synaptic input organization and anorexigenic hypothalamic POMC neurons [281]. Besides, Cheng et al. reported that HFD consumption impedes threat-cue-induced suppression of compulsive sucrose-seeking behavior in mice. This compulsive behavior was attributed to enhanced cue-triggered neuronal activity in the anterior paraventricular thalamus (aPVT) due to HFD-induced microglia activation [282] (see Fig. 2).

Fig. 2
figure 2

Microglial contribution to neuropsychiatric disorders. Since embryonic period, biological and social stress hijack microglia to dysfunctional states that trigger neuroinflammation, synaptic abnormality, and diminished neurogenesis in various brain areas responsible for emotional and behavioral regulation, which consequently predispose individuals to the onset or deterioration of neuropsychiatric disorders. Further understanding of microglia will shed light on the microglia-targeted therapeutics for more efficient and personalized treatment of neuropsychiatric disorders

Future perspectives for the field

Sex bias: can microglia explain gender differences of neuropsychiatric disorders?

Sexual dimorphisms prevail in depression [202], anxiety [283], ASD [284] and schizophrenia [285]. Biological mechanisms including neuroinflammation [283], biochemical reactions [257], and more delicate transcriptional and proteomic divergence [286, 287] may underlie the pathophysiology differing between males and females; meanwhile, microglia may play a mediating role in these dimorphisms. Microglial differentiation and responses to environmental challenges diverge in males and females. They remain sensitive to sexual hormones throughout lifespan and modify sex-specific circuits in the preoptic area, cerebellum and amygdala [4, 288, 289]. The pro-inflammatory microglia of depressive-like phenotypes (PIMID), as introduced before, was found in female cynomolgus macaques that more vulnerable to social stress-associated depression than males [214]. In male developmental amygdala, microglia are more phagocytotic regulated by the androgen-induced endocannabinoid (eCB) tone and thereby shape the male social circuitry [290]. Elevating microglial protein synthesis only induced impaired synaptic refinement and autism-like behaviors in male but not female mice [257]. Microglia appear to be more vulnerable in male embryos and female adults, which may underly the higher incidence of ASD and schizophrenia in males at early stage, yet depression are more prevalent in females during adolescence or adulthood [4, 291,292,293].

Species diversity: how are human microglia involved in neuropsychiatric pathology?

Although considerable animal experiments have deciphered the involvement of microglia in various neuropsychiatric disorders, the confirmations in humans are still sparse. As discussed above, TSPO–PET and post-mortem studies of MDD patients remain controversial on whether microglia are activated as observed in most rodent models. The cellular repertoire in primate dorsolateral prefrontal cortex revealed the expression of neuropsychiatric risk gene FOXP2 in human-specific microglia, representing an evolutionary specialization of human microglia associated with psychiatric diseases [294]. Genome-wide association studies (GWASs) of human brain endorsed disease-specific alterations in microglia associated with ASD, schizophrenia and bipolar disorder, predominantly enriched in immune responses and phagocytosis [200, 201, 295]. Given the high inter-individual variations of neuropsychiatric disorders, large-scale clinical studies with more selective inclusion criteria are warranted to provide more compelling evidence. Of note, since the understanding of microglial diversity has been far beyond M1/M2 polarization, and their functional repertoire has immensely expanded over immunoreactivity and phagocytosis, whether microglia are involved in neuropsychiatric pathology should probably not be concluded by conventional inflammatory or phagocytotic markers. For this reason, confirming human microglia diversity and reliable markers at specific disease contexts could be an urgent problem.

Indeed, the gene expression profiles of human microglia are not identical to the data of rodents, showing differences in genes associated with immune regulation, proliferation and cell cycle [8]. Several researches revealed the unique spatiotemporal transcriptomics of human microglia obtained from individuals with aging, neuroinflammatory or neurodegenerative diseases [8, 26]. Furthermore, studies of live microglia behavior from fresh brain samples have extended the understanding of human microglial dynamics interacting with neurons, glial cells and multiple molecular signals within the pathological microenvironment. For example, work in human epileptic tissues exhibited distinct microglial live responses to purinergic stimulation in disease states, including membrane ruffling and process extension or retraction [296, 297]. The sophisticated combination of live imaging and human microglia models constructed by ex vivo acute brain slices, in vitro human-induced pluripotent cells or embryonic stem cells, brain organoids, and even in vivo human brain organoids using xenotransplantation [298,299,300,301], will allow neurologists towards better comprehension of human microglia pathology.

Bench to clinic: are microglia new targets for neuropsychiatric disorder treatment?

Although human microglia biology remains largely unknown, the prospect of targeting microglia for neuropsychiatric disorders treatment is fascinating. Current promising directions include:

Anti-inflammatory therapy

Microglia deletion (e.g., using CSF1R inhibitor) or inhibition (e.g., using minocycline) appears to mitigate neuroinflammation, emotional and behavioral disorders in animal models [219, 302,303,304,305]. In clinical trials, although minocycline seem not to improve treatment-resistant depression [306], schizophrenia [307, 308] and bipolar disorder [304], its add-on efficacy was demonstrated in MDD patients with low-grade peripheral inflammation (CRP ≥ 3 mg/L) [309]. More innovative microglia-targeted systems, such as a melatonin releasing system using cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) as a chimeric antigen receptor (CAR) to target CD86 on microglial surface (CAR-M) [310], and circDYM (mm9_circ_0007509) transfer system via extracellular vesicles (EVs) modified by acetylcholine receptor‐specific rabies virus glycoprotein (RVG-circDYM-EVs) [311], both of them can enter the brain across BBB, suppress microglial inflammatory activation, and alleviate depressive-like behaviors.

Induction of pro-neurogenesis microglia

Medications such as IL-4 [55] and (R)-ketamine [312] demonstrate antidepressant effects by reprograming microglia towards an ARG-1 expressing phenotype that promotes BDNF-dependent neurogenesis and rescues synaptic loss. Since activated microglia was found in the vicinity of new-born neurons to comprise their survival, microglia inhibition by minocycline may also have a pro-neurogenesis effect [305, 313].

Brain stimulation

Methods include transcranial magnetic stimulation, transcranial direct current stimulation, electroconvulsive stimulation (ECS) and deep brain stimulation [314]. Microglia appear to be necessary for the antidepressant effect of ECS, during which ECS can inhibit the immune checkpoint gene lymphocyte-activating gene-3 (LAG3) in microglia and enhance hippocampal neurogenesis [315]. Gamma entrainment has been investigated in treatment of various CNS diseases [316, 317]. In AD models, gamma entrainment enhanced microglial phagocytotic activity [318, 319]; in post-stroke models, gamma entrainment inhibited the pro-inflammatory activation of microglia and ameliorated anxiety susceptibility [207]. However, there is emerging controversy about the neuroprotective effect of gamma entrainment, and the gamma-band visual stimulation may be aversive for mice [320].

Cell replacement therapy

With the understanding of microglia repopulating properties, microglia replacement by pharmacologically induced repopulation, bone-marrow transplantation (mrBMT), peripheral blood (mrPB), or microglia transplantation (mrMT) have been explored as novel therapeutics [321,322,323]. The most recent work by Vieira et al. engrafted human glial progenitor cells (hGPCs) into mice chimerized with mutant Huntingtin (mHTT) expressing hGPCs, which archived effective repopulation with younger healthy glia that broadly replaced aged and diseased human glia, representing a markable progress of microglia replacement as a therapeutic strategy for CNS diseases [324].

In summary, high-throughput sequencing has unveiled the temporal–spatial heterogeneity of microglia about their ontogeny, phenotype diversity and biological functions. Pathophysiology of neuropsychiatric disorders is an intricate combination of genetic risk, biological stress, social threats, sex, age, comorbidity with other physical or psychiatric disorders, etc., and the versatile microglia may be involved in each catalyst. A deep comprehension of microglia biology will provide window into the microgliopathy of neuropsychiatric disorders, exemplified by the microglia supporting neurogenesis in depression, synaptic elimination in ASD and schizophrenia, microglia–neuron crosstalk in eating disorders and other to-be-explored mechanisms. Most importantly, these discoveries need to be confirmed in humans to break the bottleneck of neuropsychiatric disorders treatment. In the future, multiple advanced technologies and high-quality clinical researches will shed light on the shadow of microglia casting over human brain diseases.

Availability of data and materials

Not applicable.

Abbreviations

Aβ:

Amyloid β

AD:

Alzheimer’s disease

ALS:

Amyotrophic lateral sclerosis

ApoE:

Apolipoprotein E

ATM:

Axon tract-associated microglia

ATP:

Adenosine triphosphate

α-syn:

α-Synuclein

ACC:

Anterior cingulate cortex

ASD:

Autistic spectrum disorder

BBB:

Blood–brain barrier

BDNF:

Brain-derived neurotrophic factor

AMP:

Cyclic adenosine monophosphate

CARD9:

Domain-containing protein 9

CNS:

Central venous system

CSF1R:

Colony stimulating factor 1 receptor

CCR5:

C–C chemokine receptor type 5

CCRL2:

C–C motif chemokine receptor-like 2

CX3CR1:

C–X3–C motif chemokine receptor 1

CXCR4:

C–X–C chemokine receptor type 4

CXCL1:

C–X–C motif chemokine ligand 10

CAM:

Capillary-associated microglia

CLDN5:

Claudin-5

CASPR2:

Contactin-associated protein-2

cIN:

Cortical interneuron

CGRP:

Alcitonin gene-related peptide

DAM:

Disease-associated microglia

DIO:

Diet-induced obesity

ECM:

Extracellular matrix

eIF4E:

Eukaryotic initiation factor 4F

FGF2:

Fibroblast growth factor 2

GVU:

Glial–vascular unit

GLP-1R:

Glucagon-like peptide-1 receptor

HDAC:

Histone deacetylase

HFD:

High-fat diet

Iba-1:

Ionized calcium-binding adaptor molecule 1

iNOS:

Inducible nitric oxide synthase

IGF-1:

Insulin-like growth factor 1

IFN:

Interferon

IL:

Interleukin

IFP35:

Interferon-induced protein 35

IRM:

Injury-responsive microglia

LDAM:

Lipid droplet accumulating microglia

LPS:

Lipopolysaccharide

MGnD:

Microglial neurodegenerative phenotype

MS:

Multiple sclerosis

MMP-9:

Metalloproteinase 9

MIMS:

Microglia inflamed in MS

MIA:

Maternal immune activation

mrBMT:

Microglia replacement by bone-marrow transplantation

mrPB:

Microglia replacement by peripheral blood

mrMT:

Microglia replacement by microglia transplantation

mPFC:

Medial prefrontal cortex

MDD:

Major depressive disorder

NF-κB:

Nuclear factor kappa-B

NLRP3:

Nucleotide-binding oligomerization domain

NLRP3:

NOD-like receptor family pyrin domain-containing 3

NVU:

Neurovascular unit

OB:

Olfactory bulb

OCD:

Obsessive–compulsive disorder

OPCs:

Oligodendrocyte precursor cells

PAM:

Proliferative region-associated microglia

P2RY12:

Purinergic receptor P2Y12

PtdSer:

Phosphatidylserine

PDGFB:

Platelet-derived growth factor B

PTSD:

Post-traumatic stress disorder

PNNs:

Perineuronal nets

RIPK1:

Receptor interacting protein kinase 1

ROS:

Reactive oxygen species

RRIMs:

Regulated inflammatory microglia

R-SDS:

Repeated social defeat stress

Spp1:

Secreted phosphoprotein 1

SVZ:

Subventricular zone

SYK:

Spleen tyrosine kinase

ScRNA-seq:

Single-cell RNA-sequencing

SCI:

Spinal cord injury

TNF:

Tumor necrosis factor

TREM2:

Triggering receptor expressed on myeloid cells 2

TLR:

Toll-like receptors

TGF-β:

Transforming growth factor-β

THIK-1:

TWIK-related Halothane-inhibited K + channel

TREM2:

Triggering receptor expressed on myeloid cells 2

TRPV1:

Transient receptor potential vanilloid type

TSPO:

Translocator protein

TSC:

Tuberous sclerosis

UCP2:

Uncoupling protein 2

VEGFA:

Vascular endothelial growth factor A

References

  1. Aguzzi A, Barres BA, Bennett ML. Microglia: scapegoat, saboteur, or something else? Science. 2013;339(6116):156–61.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Askew K, Li K, Olmos-Alonso A, Garcia-Moreno F, Liang Y, Richardson P, et al. Coupled proliferation and apoptosis maintain the rapid turnover of microglia in the adult brain. Cell Rep. 2017;18(2):391–405.

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Zhang Y, Cui D. Evolving models and tools for microglial studies in the central nervous system. Neurosci Bull. 2021;37(8):1218–33.

    PubMed  PubMed Central  Google Scholar 

  4. Thion MS, Low D, Silvin A, Chen JM, Grisel P, Schulte-Schrepping J, et al. Microbiome influences prenatal and adult microglia in a sex-specific manner. Cell. 2018;172(3):500.

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Li Q, Barres BA. Microglia and macrophages in brain homeostasis and disease. Nat Rev Immunol. 2018;18(4):225–42.

    CAS  PubMed  Google Scholar 

  6. Füger P, Hefendehl JK, Veeraraghavalu K, Wendeln A-C, Schlosser C, Obermüller U, et al. Microglia turnover with aging and in an Alzheimer’s model via long-term in vivo single-cell imaging. Nat Neurosci. 2017;20(10):1371–6.

    PubMed  Google Scholar 

  7. Geirsdottir L, David E, Keren-Shaul H, Weiner A, Bohlen SC, Neuber J, et al. Cross-species single-cell analysis reveals divergence of the primate microglia program. Cell. 2019;179(7):1609-22.e16.

    CAS  PubMed  Google Scholar 

  8. Galatro TF, Holtman IR, Lerario AM, Vainchtein ID, Brouwer N, Sola PR, et al. Transcriptomic analysis of purified human cortical microglia reveals age-associated changes. Nat Neurosci. 2017;20(8):1162.

    CAS  PubMed  Google Scholar 

  9. Ryan EJ, Marshall AJ, Magaletti D, Floyd H, Draves KE, Olson NE, et al. Dendritic cell-associated lectin-1: a novel dendritic cell-associated, C-type lectin-like molecule enhances T cell secretion of IL-4. J Immunol. 2002;169(10):5638.

    CAS  PubMed  Google Scholar 

  10. Stratoulias V, Venero JL, Tremblay M-È, Joseph B. Microglial subtypes: diversity within the microglial community. EMBO J. 2019;38(17): e101997.

    PubMed  PubMed Central  Google Scholar 

  11. Huang Y, Xu Z, Xiong S, Sun F, Qin G, Hu G, et al. Repopulated microglia are solely derived from the proliferation of residual microglia after acute depletion. Nat Neurosci. 2018;21(4):530–40.

    CAS  PubMed  Google Scholar 

  12. Huang Y, Xu Z, Xiong S, Qin G, Sun F, Yang J, et al. Dual extra-retinal origins of microglia in the model of retinal microglia repopulation. Cell Discov. 2018;4(1):9.

    PubMed  PubMed Central  Google Scholar 

  13. Mossadegh-Keller N, Sarrazin S, Kandalla PK, Espinosa L, Stanley ER, Nutt SL, et al. M-CSF instructs myeloid lineage fate in single haematopoietic stem cells. Nature. 2013;497(7448):239–43.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Datta M, Staszewski O, Raschi E, Frosch M, Hagemeyer N, Tay TL, et al. Histone deacetylases 1 and 2 regulate microglia function during development, homeostasis, and neurodegeneration in a context-dependent manner. Immunity. 2018;48(3):514-29.e6.

    CAS  PubMed  Google Scholar 

  15. Gosselin D, Skola D, Coufal NG, Holtman IR, Schlachetzki JCM, Sajti E, et al. An environment-dependent transcriptional network specifies human microglia identity. Science. 2017;356(6344):eaa13222.

    Google Scholar 

  16. Bohlen CJ, Bennett FC, Tucker AF, Collins HY, Mulinyawe SB, Barres BA. Diverse requirements for microglial survival, specification, and function revealed by defined-medium cultures. Neuron. 2017;94(4):759-73.e8.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Utz SG, See P, Mildenberger W, Thion MS, Silvin A, Lutz M, et al. Early fate defines microglia and non-parenchymal brain macrophage development. Cell. 2020;181(3):557-73.e18.

    CAS  PubMed  Google Scholar 

  18. Baxter PS, Dando O, Emelianova K, He X, McKay S, Hardingham GE, et al. Microglial identity and inflammatory responses are controlled by the combined effects of neurons and astrocytes. Cell Rep. 2021;34(12):108882.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Chitu V, Gokhan Ş, Nandi S, Mehler MF, Stanley ER. Emerging roles for CSF-1 receptor and its ligands in the nervous system. Trends Neurosci. 2016;39(6):378–93.

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Kana V, Desland FA, Casanova-Acebes M, Ayata P, Badimon A, Nabel E, et al. CSF-1 controls cerebellar microglia and is required for motor function and social interaction. J Exp Med. 2019;216(10):2265–81.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Borst K, Dumas A, Prinz M. Microglia: Immune and non-immune functions. Immunity. 2021;54:2194–208.

    CAS  PubMed  Google Scholar 

  22. Martins-Ferreira R, Leal B, Costa PP, Ballestar E. Microglial innate memory and epigenetic reprogramming in neurological disorders. Prog Neurobiol. 2021;200:101971.

    CAS  PubMed  Google Scholar 

  23. Bollmann L, Koser D, Shahapure R, Gautier H, Holzapfel G, Scarcelli G, et al. Microglia mechanics: immune activation alters traction forces and durotaxis. Front Cell Neurosci. 2015;9:363.

    PubMed  PubMed Central  Google Scholar 

  24. Frank MG, Fonken LK, Watkins LR, Maier SF. Microglia: neuroimmune-sensors of stress. Semin Cell Dev Biol. 2019;94:176–85.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Zengeler K, Lukens J. Innate immunity at the crossroads of healthy brain maturation and neurodevelopmental disorders. Nat Rev Immunol. 2021;21:454.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Masuda T, Sankowski R, Staszewski O, Böttcher C, Amann L, Sagar, et al. Spatial and temporal heterogeneity of mouse and human microglia at single-cell resolution. Nature. 2019;566(7744):388–92.

    CAS  PubMed  Google Scholar 

  27. Ayata P, Badimon A, Strasburger HJ, Duff MK, Montgomery SE, Loh Y-HE, et al. Epigenetic regulation of brain region-specific microglia clearance activity. Nat Neurosci. 2018;21(8):1049–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Sato K. Effects of microglia on neurogenesis. Glia. 2015;63(8):1394–405.

    PubMed  PubMed Central  Google Scholar 

  29. Fourgeaud L, Través PG, Tufail Y, Leal-Bailey H, Lew ED, Burrola PG, et al. TAM receptors regulate multiple features of microglial physiology. Nature. 2016;532(7598):240–4.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Eyo UB, Mo M, Yi M-H, Murugan M, Liu J, Yarlagadda R, et al. P2Y12R-dependent translocation mechanisms gate the changing microglial landscape. Cell Rep. 2018;23(4):959–66.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Ransohoff RM. A polarizing question: do M1 and M2 microglia exist? Nat Neurosci. 2016;19(8):987–91.

    CAS  PubMed  Google Scholar 

  32. Kim CC, Nakamura MC, Hsieh CL. Brain trauma elicits non-canonical macrophage activation states. J Neuroinflammation. 2016;13(1):117.

    PubMed  PubMed Central  Google Scholar 

  33. Lloyd AF, Davies CL, Holloway RK, Labrak Y, Ireland G, Carradori D, et al. Central nervous system regeneration is driven by microglia necroptosis and repopulation. Nat Neurosci. 2019;22(7):1046–52.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Paolicelli RC, Sierra A, Stevens B, Tremblay M-E, Aguzzi A, Ajami B, et al. Microglia states and nomenclature: a field at its crossroads. Neuron. 2022;110(21):3458–83.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. De S, Van Deren D, Peden E, Hockin M, Boulet A, Titen S, et al. Two distinct ontogenies confer heterogeneity to mouse brain microglia. Development. 2018;145(13):dev152306.

    PubMed  PubMed Central  Google Scholar 

  36. Nagarajan N, Jones BW, West PJ, Marc RE, Capecchi MR. Corticostriatal circuit defects in Hoxb8 mutant mice. Mol Psychiatry. 2018;23(9):1868–77.

    CAS  PubMed  Google Scholar 

  37. Snijders GJLJ, Sneeboer MAM, Fernández-Andreu A, Udine E, Boks MP, Ormel PR, et al. Distinct non-inflammatory signature of microglia in post-mortem brain tissue of patients with major depressive disorder. Mol Psychiatry. 2021;26(7):3336–49.

    CAS  PubMed  Google Scholar 

  38. Chen S-K, Tvrdik P, Peden E, Cho S, Wu S, Spangrude G, et al. Hematopoietic origin of pathological grooming in Hoxb8 mutant mice. Cell. 2010;141:775–85.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Nagarajan N, Capecchi MR. Optogenetic stimulation of mouse Hoxb8 microglia in specific regions of the brain induces anxiety, grooming, or both. Mol Psychiatry. 2023. https://doi.org/10.1038/s41380-023-02019-w.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Yeh FL, Hansen DV, Sheng M. TREM2, Microglia, and neurodegenerative diseases. Trends Mol Med. 2017;23(6):512–33.

    CAS  PubMed  Google Scholar 

  41. Schmid CD, Sautkulis LN, Danielson PE, Cooper J, Hasel KW, Hilbush BS, et al. Heterogeneous expression of the triggering receptor expressed on myeloid cells-2 on adult murine microglia. J Neurochem. 2002;83(6):1309–20.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Ulland TK, Colonna M. TREM2—a key player in microglial biology and Alzheimer disease. Nat Rev Neurol. 2018;14(11):667–75.

    CAS  PubMed  Google Scholar 

  43. 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. Nat Med. 2020;26(1):131–42.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Winfree R, Seto M, Dumitrescu L, Menon V, Jager P, Wang Y, et al. TREM2 gene expression associations with Alzheimer’s disease neuropathology are region-specific: implications for cortical versus subcortical microglia. Acta Neuropathol. 2023;145:1–15.

    Google Scholar 

  45. Xie M, Liu YU, Zhao S, Zhang L, Bosco DB, Pang Y-P, et al. TREM2 interacts with TDP-43 and mediates microglial neuroprotection against TDP-43-related neurodegeneration. Nat Neurosci. 2022;25(1):26–38.

    CAS  PubMed  Google Scholar 

  46. Manich G, Gómez-López AR, Almolda B, Villacampa N, Recasens M, Shrivastava K, et al. Differential roles of TREM2+ microglia in anterograde and retrograde axonal injury models. Front Cell Neurosci. 2020;14:567404.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Benmamar-Badel A, Owens T, Wlodarczyk A. Protective microglial subset in development, aging, and disease: lessons from transcriptomic studies. Front Immunol. 2020;11:430.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Wlodarczyk A, Cedile O, Jensen KN, Jasson A, Mony JT, Khorooshi R, et al. Pathologic and protective roles for microglial subsets and bone marrow- and blood-derived myeloid cells in central nervous system inflammation. Front Immunol. 2015;6:1–11.

    CAS  Google Scholar 

  49. Li Q, Cheng Z, Zhou L, Darmanis S, Neff NF, Okamoto J, et al. Developmental heterogeneity of microglia and brain myeloid cells revealed by deep single-cell RNA sequencing. Neuron. 2019;101(2):207-23.e10.

    CAS  PubMed  Google Scholar 

  50. Hammond TR, Dufort C, Dissing-Olesen L, Giera S, Young A, Wysoker A, et al. Single-Cell RNA sequencing of microglia throughout the mouse lifespan and in the injured brain reveals complex cell-state changes. Immunity. 2019;50(1):253-71.e6.

    CAS  PubMed  Google Scholar 

  51. Böttcher C, Schlickeiser S, Sneeboer MAM, Kunkel D, Knop A, Paza E, et al. Human microglia regional heterogeneity and phenotypes determined by multiplexed single-cell mass cytometry. Nat Neurosci. 2019;22(1):78–90.

    PubMed  Google Scholar 

  52. Sato-Hashimoto M, Nozu T, Toriba R, Horikoshi A, Akaike M, Kawamoto K, et al. Microglial SIRPα regulates the emergence of CD11c+ microglia and demyelination damage in white matter. Elife. 2019;8: e42025.

    PubMed  PubMed Central  Google Scholar 

  53. Wlodarczyk A, Holtman IR, Krueger M, Yogev N, Bruttger J, Khorooshi R, et al. A novel microglial subset plays a key role in myelinogenesis in developing brain. EMBO J. 2017;36(22):3292–308.

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Stratoulias V, Ruiz R, Kanatani S, Osman A, Keane L, Armengol J, et al. ARG1-expressing microglia show a distinct molecular signature and modulate postnatal development and function of the mouse brain. Nat Neurosci. 2023;26:1–13.

    Google Scholar 

  55. Zhang J, Rong P, Zhang L, He H, Zhou T, Fan Y, et al. IL4-driven microglia modulate stress resilience through BDNF-dependent neurogenesis. Sci Adv. 2021;7(12):eabb9888.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Cherry J, Olschowka J, O’Banion M. Arginase 1+ microglia reduce Aβ plaque deposition during IL-1β-dependent neuroinflammation. J Neuroinflammation. 2015;12:1.

    Google Scholar 

  57. Xavier AL, Lima FR, Nedergaard M, Menezes JR. Ontogeny of CX3CR1-EGFP expressing cells unveil microglia as an integral component of the postnatal subventricular zone. Front Cell Neurosci. 2015;9:37.

    PubMed  PubMed Central  Google Scholar 

  58. Ribeiro Xavier AL, Kress BT, Goldman SA, Lacerda de Menezes JR, Nedergaard M. A distinct population of microglia supports adult neurogenesis in the subventricular zone. J Neurosci. 2015;35(34):11848.

    PubMed  PubMed Central  Google Scholar 

  59. Rubino SJ, Mayo L, Wimmer I, Siedler V, Brunner F, Hametner S, et al. Acute microglia ablation induces neurodegeneration in the somatosensory system. Nat Commun. 2018;9:4578.

    PubMed  PubMed Central  Google Scholar 

  60. Shimabukuro MK, Langhi LGP, Cordeiro I, Brito JM, Batista CMD, Mattson MP, et al. Lipid-laden cells differentially distributed in the aging brain are functionally active and correspond to distinct phenotypes. Sci Rep. 2016;6:23795.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. 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. Nat Neurosci. 2020;23(2):194.

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Claes C, Danhash EP, Hasselmann J, Chadarevian JP, Shabestari SK, England WE, et al. Plaque-associated human microglia accumulate lipid droplets in a chimeric model of Alzheimer’s disease. Mol Neurodegener. 2021;16(1):50.

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Li Q, Zhao Y, Guo H, Li Q, Yan C, Li Y, et al. Impaired lipophagy induced-microglial lipid droplets accumulation contributes to the buildup of TREM1 in diabetes-associated cognitive impairment. Autophagy. 2023. https://doi.org/10.1080/15548627.2023.2213984.

    Article  PubMed  PubMed Central  Google Scholar 

  64. 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.

    CAS  PubMed  Google Scholar 

  65. 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):1073–81.

    CAS  PubMed  Google Scholar 

  66. Ofengeim D, Mazzitelli S, Ito Y, DeWitt JP, Mifflin L, Zou CY, et al. RIPK1 mediates a disease-associated microglial response in Alzheimer’s disease. Proc Natl Acad Sci USA. 2017;114(41):E8788–97.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Rangaraju S, Dammer EB, Raza SA, Rathakrishnan P, Xiao HL, Gao TW, et al. Identification and therapeutic modulation of a pro-inflammatory subset of disease-associated-microglia in Alzheimer’s disease. Mol Neurodegener. 2018;13:1.

    Google Scholar 

  68. Silvin A, Uderhardt S, Piot C, Da Mesquita S, Yang K, Geirsdottir L, et al. Dual ontogeny of disease-associated microglia and disease inflammatory macrophages in aging and neurodegeneration. Immunity. 2022;55(8):1448-65.e6.

    CAS  PubMed  Google Scholar 

  69. Sierksma A, Lu A, Mancuso R, Fattorelli N, Thrupp N, Salta E, et al. Novel Alzheimer risk genes determine the microglia response to amyloid-β but not to TAU pathology. EMBO Mol Med. 2020;12(3): e10606.

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Sala Frigerio C, Wolfs L, Fattorelli N, Thrupp N, Voytyuk I, Schmidt I, et al. The major risk factors for Alzheimer’s disease: age, sex, and genes modulate the microglia response to Aβ plaques. Cell Rep. 2019;27(4):1293-306.e6.

    CAS  PubMed  Google Scholar 

  71. Moonen S, Koper MJ, Van Schoor E, Schaeverbeke JM, Vandenberghe R, von Arnim CAF, et al. Pyroptosis in Alzheimer’s disease: cell type-specific activation in microglia, astrocytes and neurons. Acta Neuropathol. 2023;145(2):175–95.

    CAS  PubMed  Google Scholar 

  72. Yin Z, Herron S, Silveira S, Kleemann K, Gauthier C, Mallah D, et al. Identification of a protective microglial state mediated by miR-155 and interferon-γ signaling in a mouse model of Alzheimer’s disease. Nat Neurosci. 2023;26(7):1196–207.

    CAS  PubMed  Google Scholar 

  73. Gerrits E, Brouwer N, Kooistra SM, Woodbury ME, Vermeiren Y, Lambourne M, et al. Distinct amyloid-β and tau-associated microglia profiles in Alzheimer’s disease. Acta Neuropathol. 2021;141(5):681–96.

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Kim DW, Tu KJ, Wei A, Lau AJ, Gonzalez-Gil A, Cao T, et al. Amyloid-beta and tau pathologies act synergistically to induce novel disease stage-specific microglia subtypes. Mol Neurodegener. 2022;17(1):83.

    CAS  PubMed  PubMed Central  Google Scholar 

  75. 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):566-81.e9.

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Ryan SK, Zelic M, Han Y, Teeple E, Chen L, Sadeghi M, et al. Microglia ferroptosis is regulated by SEC24B and contributes to neurodegeneration. Nat Neurosci. 2023;26(1):12–26.

    CAS  PubMed  Google Scholar 

  77. Smajić S, Prada C, Landoulsi Z, Dietrich C, Jarazo J, Henck J, et al. Single-cell sequencing of the human midbrain reveals glial activation and a neuronal state specific to Parkinson’s disease. Brain. 2022. https://doi.org/10.1093/brain/awab446.

    Article  PubMed  Google Scholar 

  78. Lopes KO, Sparks DL, Streit WJ. Microglial dystrophy in the aged and Alzheimer’s disease brain is associated with ferritin immunoreactivity. Glia. 2008;56(10):1048–60.

    PubMed  Google Scholar 

  79. Damani MR, Zhao LA, Fontainhas AM, Amaral J, Fariss RN, Wong WT. Age-related alterations in the dynamic behavior of microglia. Aging Cell. 2011;10(2):263–76.

    CAS  PubMed  Google Scholar 

  80. Koellhoffer EC, McCullough LD, Ritzel RM. Old maids: aging and its impact on microglia function. Int J Mol Sci. 2017;18(4):769.

    PubMed  PubMed Central  Google Scholar 

  81. Sierra A, Gottfried-Blackmore AC, McEwen BS, Bulloch K. Microglia derived from aging mice exhibit an altered inflammatory profile. Glia. 2007;55(4):412–24.

    PubMed  Google Scholar 

  82. Olah M, Patrick E, Villani A-C, Xu J, White C, Ryan K, et al. A transcriptomic atlas of aged human microglia. Nat Commun. 2018;9:539.

    PubMed  PubMed Central  Google Scholar 

  83. Haynes SE, Hollopeter G, Yang G, Kurpius D, Dailey ME, Gan WB, et al. The P2Y(12) receptor regulates microglial activation by extracellular nucleotides. Nat Neurosci. 2006;9(12):1512–9.

    CAS  PubMed  Google Scholar 

  84. Sievers J, Parwaresch R, Wottge HU. Blood monocytes and spleen macrophages differentiate into microglia-like cells on monolayers of astrocytes—morphology. Glia. 1994;12(4):245–58.

    CAS  PubMed  Google Scholar 

  85. Tanaka J, Maeda N. Microglial ramification requires nondiffusible factors derived from astrocytes. Exp Neurol. 1996;137(2):367–75.

    CAS  PubMed  Google Scholar 

  86. 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. Nat Neurosci. 2014;17(1):131–43.

    CAS  PubMed  Google Scholar 

  87. Hughes AN, Appel B. Microglia phagocytose myelin sheaths to modify developmental myelination. Nat Neurosci. 2020;23(9):1055–66.

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Hagemeyer N, Hanft KM, Akriditou MA, Unger N, Park ES, Stanley ER, et al. Microglia contribute to normal myelinogenesis and to oligodendrocyte progenitor maintenance during adulthood. Acta Neuropathol. 2017;134(3):441–58.

    PubMed  PubMed Central  Google Scholar 

  89. Ueno M, Fujita Y, Tanaka T, Nakamura Y, Kikuta J, Ishii M, et al. Layer V cortical neurons require microglial support for survival during postnatal development. Nat Neurosci. 2013;16(5):543–51.

    CAS  PubMed  Google Scholar 

  90. Sierra A, Encinas JM, Deudero JJP, Chancey JH, Enikolopov G, Overstreet-Wadiche LS, et al. Microglia shape adult hippocampal neurogenesis through apoptosis-coupled phagocytosis. Cell Stem Cell. 2010;7(4):483–95.

    CAS  PubMed  PubMed Central  Google Scholar 

  91. 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):833-46.e16.

    CAS  PubMed  Google Scholar 

  92. Bernier L-P, Bohlen CJ, York EM, Choi HB, Kamyabi A, Dissing-Olesen L, et al. Nanoscale surveillance of the brain by microglia via cAMP-regulated filopodia. Cell Rep. 2019;27(10):2895-908.e4.

    CAS  PubMed  Google Scholar 

  93. Sieger D, Moritz C, Ziegenhals T, Prykhozhij S, Peri F. Long-range Ca2+ waves transmit brain-damage signals to microglia. Dev Cell. 2012;22(6):1138–48.

    CAS  PubMed  Google Scholar 

  94. Tufail Y, Cook D, Fourgeaud L, Powers CJ, Merten K, Clark CL, et al. Phosphatidylserine exposure controls viral innate immune responses by microglia. Neuron. 2017;93(3):574-86.e8.

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Fekete R, Cserep C, Lenart N, Toth K, Orsolits B, Martinecz B, et al. Microglia control the spread of neurotropic virus infection via P2Y12 signalling and recruit monocytes through P2Y12-independent mechanisms. Acta Neuropathol. 2018;136(3):461–82.

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Madry C, Arancibia-Cárcamo IL, Kyrargyri V, Chan VTT, Hamilton NB, Attwell D. Effects of the ecto-ATPase apyrase on microglial ramification and surveillance reflect cell depolarization, not ATP depletion. Proc Natl Acad Sci. 2018;115(7):E1608–17.

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Madry C, Kyrargyri V, Arancibia-Cárcamo IL, Jolivet R, Kohsaka S, Bryan RM, et al. Microglial ramification, surveillance, and interleukin-1β release are regulated by the two-pore domain K+ channel THIK-1. Neuron. 2018;97(2):299-312.e6.

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Zhang Y, Hou B, Liang P, Lu X, Wu Y, Zhang X, et al. TRPV1 channel mediates NLRP3 inflammasome-dependent neuroinflammation in microglia. Cell Death Dis. 2021;12(12):1159.

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Hu Y, Cao K, Wang F, Wu W, Mai W, Qiu L, et al. Dual roles of hexokinase 2 in shaping microglial function by gating glycolytic flux and mitochondrial activity. Nat Metab. 2022;4(12):1756–74.

    CAS  PubMed  Google Scholar 

  100. Moseman EA, Blanchard AC, Nayak D, McGavern DB. T cell engagement of cross-presenting microglia protects the brain from a nasal virus infection. Sci Immunol. 2020;5(48):eabb1817.

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Wheeler DL, Sariol A, Meyerholz DK, Perlman S. Microglia are required for protection against lethal coronavirus encephalitis in mice. J Clin Investig. 2018;128(3):931–43.

    PubMed  PubMed Central  Google Scholar 

  102. Garber C, Soung A, Vollmer LL, Kanmogne M, Last A, Brown J, et al. T cells promote microglia-mediated synaptic elimination and cognitive dysfunction during recovery from neuropathogenic flaviviruses. Nat Neurosci. 2019;22(8):1276.

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Wu YF, Du SQ, Johnson JL, Tung HY, Landers CT, Liu YW, et al. Microglia and amyloid precursor protein coordinate control of transient Candida cerebritis with memory deficits. Nat Commun. 2019;10:58.

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Drummond RA, Swamydas M, Oikonomou V, Zhai B, Dambuza IM, Schaefer BC, et al. CARD9+ microglia promote antifungal immunity via IL-1β- and CXCL1-mediated neutrophil recruitment. Nat Immunol. 2019;20(5):559–70.

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Mifflin L, Hu Z, Dufort C, Hession CC, Walker AJ, Niu K, et al. A RIPK1-regulated inflammatory microglial state in amyotrophic lateral sclerosis. Proc Natl Acad Sci. 2021;118(13): e2025102118.

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Zelic M, Pontarelli F, Woodworth L, Zhu C, Mahan A, Ren Y, et al. RIPK1 activation mediates neuroinflammation and disease progression in multiple sclerosis. Cell Rep. 2021;35(6): 109112.

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Jing X, Yao Y, Wu D, Hong H, Feng X, Xu N, et al. IFP35 family proteins promote neuroinflammation and multiple sclerosis. Proc Natl Acad Sci. 2021;118(32): e2102642118.

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Fantin A, Vieira JM, Gestri G, Denti L, Schwarz Q, Prykhozhij S, et al. Tissue macrophages act as cellular chaperones for vascular anastomosis downstream of VEGF-mediated endothelial tip cell induction. Blood. 2010;116(5):829–40.

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Arnold T, Betsholtz C. The importance of microglia in the development of the vasculature in the central nervous system. Vasc cell. 2013;5(1):4.

    PubMed  PubMed Central  Google Scholar 

  110. Dudiki T, Meller J, Mahajan G, Liu H, Zhevlakova I, Stefl S, et al. Microglia control vascular architecture via a TGF beta 1 dependent paracrine mechanism linked to tissue mechanics. Nat Commun. 2020;11(1):986.

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Shen Q, Chen Z, Zhao F, Pan S, Zhang T, Cheng X, et al. Reversal of prolonged obesity-associated cerebrovascular dysfunction by inhibiting microglial Tak1. Nat Neurosci. 2020;23(7):832–41.

    CAS  PubMed  Google Scholar 

  112. Foulquier S, Caolo V, Swennen G, Milanova I, Reinhold S, Recarti C, et al. The role of receptor MAS in microglia-driven retinal vascular development. Angiogenesis. 2019;22(4):481–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  113. He C, Liu Y, Huang Z, Yang Z, Zhou T, Liu S, et al. A specific RIP3+ subpopulation of microglia promotes retinopathy through a hypoxia-triggered necroptotic mechanism. Proc Natl Acad Sci. 2021;118(11): e2023290118.

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Bisht K, Okojie KA, Sharma K, Lentferink DH, Sun Y-Y, Chen H-R, et al. Capillary-associated microglia regulate vascular structure and function through PANX1-P2RY12 coupling in mice. Nat Commun. 2021;12(1):5289.

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Csaszar E, Lenart N, Cserep C, Koernyei Z, Fekete R, Posfai B, et al. Microglia modulate blood flow, neurovascular coupling, and hypoperfusion via purinergic actions. J Exp Med. 2022;219(3): e20211071.

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Thurgur H, Pinteaux E. Microglia in the neurovascular unit: blood-brain barrier-microglia interactions after central nervous system disorders. Neuroscience. 2019;405:55–67.

    CAS  PubMed  Google Scholar 

  117. Yao D, Zhang R, Xie M, Ding F, Wang M, Wang W. Updated understanding of the glial-vascular unit in central nervous system disorders. Neurosci Bull. 2022. https://doi.org/10.1007/s12264-022-00977-9.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Chen A-Q, Fang Z, Chen XL, Yang S, Zhou Y-F, Mao L, et al. Microglia-derived TNF-α mediates endothelial necroptosis aggravating blood brain–barrier disruption after ischemic stroke. Cell Death Dis. 2019;10(7):487.

    PubMed  PubMed Central  Google Scholar 

  119. Liao BY, Geng LL, Zhang F, Shu LL, Wei L, Yeung PKK, et al. Adipocyte fatty acid-binding protein exacerbates cerebral ischaemia injury by disrupting the blood-brain barrier. Eur Heart J. 2020;41(33):3169.

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Otxoa-de-Amezaga A, Miro-Mur F, Pedragosa J, Gallizioli M, Justicia C, Gaja-Capdevila N, et al. Microglial cell loss after ischemic stroke favors brain neutrophil accumulation. Acta Neuropathol. 2019;137(2):321–41.

    CAS  PubMed  Google Scholar 

  121. Haruwaka K, Ikegami A, Tachibana Y, Ohno N, Konishi H, Hashimoto A, et al. Dual microglia effects on blood brain :barrier permeability induced by systemic inflammation. Nat Commun. 2019;10:5816.

    CAS  PubMed  PubMed Central  Google Scholar 

  122. 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):979-91.e8.

    CAS  PubMed  Google Scholar 

  123. Sipe GO, Lowery RL, Tremblay ME, Kelly EA, Lamantia CE, Majewska AK. Microglial P2Y12 is necessary for synaptic plasticity in mouse visual cortex. Nat Commun. 2016;7:10905.

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Chu YX, Jin XM, Parada I, Pesic A, Stevens B, Barres B, et al. Enhanced synaptic connectivity and epilepsy in C1q knockout mice. Proc Natl Acad Sci USA. 2010;107(17):7975–80.

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Stevens B, Allen N, Vazquez L, Howell G, Christopherson K, Nouri N, et al. The classical complement cascade mediates CNS synapse elimination. Cell. 2008;131:1164–78.

    Google Scholar 

  126. Schafer Dorothy P, Lehrman Emily K, Kautzman Amanda G, Koyama R, Mardinly Alan R, Yamasaki R, et al. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron. 2012;74(4):691–705.

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Gunner G, Cheadle L, Johnson KM, Ayata P, Badimon A, Mondo E, et al. Sensory lesioning induces microglial synapse elimination via ADAM10 and fractalkine signaling. Nat Neurosci. 2019;22(7):1075.

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Vainchtein ID, Molofsky AV. Astrocytes and microglia: in sickness and in health. Trends Neurosci. 2020;43(3):144–54.

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Nguyen PT, Dorman LC, Pan S, Vainchtein ID, Han RT, Nakao-Inoue H, et al. Microglial remodeling of the extracellular matrix promotes synapse plasticity. Cell. 2020;182(2):388-403.e15.

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Albertini G, D’Andrea I, Druart M, Béchade C, Nieves-Rivera N, Etienne F, et al. Serotonin sensing by microglia conditions the proper development of neuronal circuits and of social and adaptive skills. Mol Psychiatry. 2023. https://doi.org/10.1038/s41380-023-02048-5.

    Article  PubMed  Google Scholar 

  131. Lehrman EK, Wilton DK, Litvina EY, Welsh CA, Chang ST, Frouin A, et al. CD47 protects synapses from excess microglia-mediated pruning during development. Neuron. 2018;100(1):120.

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Zhong L, Sheng X, Wang W, Li Y, Zhuo R, Wang K, et al. TREM2 receptor protects against complement-mediated synaptic loss by binding to complement C1q during neurodegeneration. Immunity. 2023. https://doi.org/10.1016/j.immuni.2023.06.016.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Wakselman S, Béchade C, Roumier A, Bernard D, Triller A, Bessis A. Developmental neuronal death in hippocampus requires the microglial CD11b integrin and DAP12 immunoreceptor. J Neurosci. 2008;28(32):8138.

    CAS  PubMed  PubMed Central  Google Scholar 

  134. 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.

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Rawji K, Young A, Ghosh T, Michaels N, Mirzaei R, Kappen J, et al. Niacin-mediated rejuvenation of macrophage/microglia enhances remyelination of the aging central nervous system. Acta Neuropathol. 2020;139:893.

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Cai W, Dai X, Chen J, Zhao J, Xu M, Zhang L, et al. STAT6/Arg1 promotes microglia/macrophage efferocytosis and inflammation resolution in stroke mice. JCI Insight. 2019;4(20): e131355.

    PubMed  PubMed Central  Google Scholar 

  137. Ennerfelt H, Frost EL, Shapiro DA, Holliday C, Zengeler KE, Voithofer G, et al. SYK coordinates neuroprotective microglial responses in neurodegenerative disease. Cell. 2022;185(22):4135-52.e22.

    CAS  PubMed  PubMed Central  Google Scholar 

  138. 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.

    CAS  PubMed  Google Scholar 

  139. Wang S, Sudan R, Peng V, Zhou Y, Du S, Yuede CM, et al. TREM2 drives microglia response to amyloid-β via SYK-dependent and -independent pathways. Cell. 2022;185(22):4153-69.e19.

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Mazaheri F, Breus O, Durdu S, Haas P, Wittbrodt J, Gilmour D, et al. Distinct roles for BAI1 and TIM-4 in the engulfment of dying neurons by microglia. Nat Commun. 2014;5:4046.

    CAS  PubMed  Google Scholar 

  141. Henson PM. Cell removal: efferocytosis. Ann Rev Cell Dev Biol. 2017;33:127–44.

    CAS  Google Scholar 

  142. Pauwels AM, Trost M, Beyaert R, Hoffmann E. Patterns, receptors, and signals: regulation of phagosome maturation. Trends Immunol. 2017;38(6):407–22.

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Peri F, Nusslein-Volhard C. Live imaging of neuronal degradation by microglia reveals a role for v0-ATPase a1 in phagosomal fusion in vivo. Cell. 2008;133(5):916–27.

    CAS  PubMed  Google Scholar 

  144. Heckmann BL, Teubner BJW, Tummers B, Boada-Romero E, Harris L, Yang M, et al. LC3-associated endocytosis facilitates beta-amyloid clearance and mitigates neurodegeneration in murine Alzheimer’s disease. Cell. 2019;178(3):536.

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Villani A, Benjaminsen J, Moritz C, Henke K, Hartmann J, Norlin N, et al. Clearance by microglia depends on packaging of phagosomes into a unique cellular compartment. Dev Cell. 2019;49(1):77.

    CAS  PubMed  Google Scholar 

  146. Fairn GD, Grinstein S. How nascent phagosomes mature to become phagolysosomes. Trends Immunol. 2012;33(8):397–405.

    CAS  PubMed  Google Scholar 

  147. Levin R, Grinstein S, Canton J. The life cycle of phagosomes: formation, maturation, and resolution. Immunol Rev. 2016;273(1):156–79.

    CAS  PubMed  Google Scholar 

  148. Merlini M, Rafalski VA, Ma KR, Kim KY, Bushong EA, Coronado PER, et al. Microglial G(i)-dependent dynamics regulate brain network hyperexcitability. Nat Neurosci. 2021;24(1):19.

    CAS  PubMed  Google Scholar 

  149. Cserep C, Posfai B, Lenart N, Fekete R, Laszlo ZI, Lele Z, et al. Microglia monitor and protect neuronal function through specialized somatic purinergic junctions. Science. 2020;367(6477):528.

    CAS  PubMed  Google Scholar 

  150. Ronzano R, Roux T, Thetiot M, Aigrot MS, Richard L, Lejeune FX, et al. Microglia-neuron interaction at nodes of Ranvier depends on neuronal activity through potassium release and contributes to remyelination. Nat Commun. 2021;12(1):5219.

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Cserép C, Pósfai B, Dénes Á. Shaping neuronal fate: functional heterogeneity of direct microglia-neuron interactions. Neuron. 2021;109(2):222–40.

    PubMed  Google Scholar 

  152. Liu Y, Li M, Zhang Z, Ye Y, Zhou J. Role of microglia-neuron interactions in diabetic encephalopathy. Ageing Res Rev. 2018;42:28–39.

    CAS  PubMed  Google Scholar 

  153. Turkin A, Tuchina O, Klempin F. Microglia function on precursor cells in the adult hippocampus and their responsiveness to serotonin signaling. Front Cell Dev Biol. 2021;9:665739.

    PubMed  PubMed Central  Google Scholar 

  154. Badimon A, Strasburger HJ, Ayata P, Chen XH, Nair A, Ikegami A, et al. Negative feedback control of neuronal activity by microglia. Nature. 2020;586(7829):417.

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Bi Q, Wang C, Cheng G, Chen N, Wei B, Liu X, et al. Microglia-derived PDGFB promotes neuronal potassium currents to suppress basal sympathetic tonicity and limit hypertension. Immunity. 2022;55(8):1466-82.e9.

    CAS  PubMed  Google Scholar 

  156. Liu HX, Wang XX, Chen L, Chen L, Tsirka SE, Ge SY, et al. Microglia modulate stable wakefulness via the thalamic reticular nucleus in mice. Nat Commun. 2021;12(1):4646.

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Liddelow S, Marsh S, Stevens B. Microglia and astrocytes in disease: dynamic duo or partners in crime? Trends Immunol. 2020;41:820.

    CAS  PubMed  Google Scholar 

  158. Joshi AU, Minhas PS, Liddelow SA, Haileselassie B, Andreasson KI, Dorn GW, et al. Fragmented mitochondria released from microglia trigger A1 astrocytic response and propagate inflammatory neurodegeneration. Nat Neurosci. 2019;22(10):1635.

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Yun SP, Kam TI, Panicker N, Kim S, Oh Y, Park JS, et al. Block of A1 astrocyte conversion by microglia is neuroprotective in models of Parkinson’s disease. Nat Med. 2018;24(7):931.

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Borucki DM, Rothhammer VJ, Quintana FJ. Microglial control of astrocytes in response to microbial metabolites. Ann Neurol. 2018;84:S109–10.

    Google Scholar 

  161. Lombardi M, Parolisi R, Scaroni F, Bonfanti E, Gualerzi A, Gabrielli M, et al. Detrimental and protective action of microglial extracellular vesicles on myelin lesions: astrocyte involvement in remyelination failure. Acta Neuropathol. 2019;138(6):987–1012.

    CAS  PubMed  PubMed Central  Google Scholar 

  162. 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):701–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Scheiblich H, Dansokho C, Mercan D, Schmidt S, Bousset L, Wischhof L, et al. Microglia jointly degrade fibrillar alpha-synuclein cargo by distribution through tunneling nanotubes. Cell. 2021;184:5089.

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Wang Q, Xue X, Huang Z, Wang Y. Microglia share the burden. Neurosci Bull. 2022;38(6):695–8.

    PubMed  PubMed Central  Google Scholar 

  165. d’Errico P, Ziegler-Waldkirch S, Aires V, Hoffmann P, Mezö C, Erny D, et al. Microglia contribute to the propagation of Aβ into unaffected brain tissue. Nat Neurosci. 2022;25(1):20–5.

    PubMed  Google Scholar 

  166. Mohebiany AN, Ramphal NS, Karram K, Di Liberto G, Novkovic T, Klein M, et al. Microglial A20 protects the brain from CD8 T-cell-mediated immunopathology. Cell Rep. 2020;30(5):1585-97.e6.

    CAS  PubMed  Google Scholar 

  167. Shi Z, Yu P, Lin W-J, Chen S, Hu X, Chen S, et al. Microglia drive transient insult-induced brain injury by chemotactic recruitment of CD8+ T lymphocytes. Neuron. 2023. https://doi.org/10.1016/j.neuron.2022.12.009.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Shi LG, Sun ZY, Su W, Xu F, Xie D, Zhang QX, et al. Treg cell-derived osteopontin promotes microglia-mediated white matter repair after ischemic stroke. Immunity. 2021;54(7):1527.

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Absinta M, Maric D, Gharagozloo M, Garton T, Smith MD, Jin J, et al. A lymphocyte-microglia-astrocyte axis in chronic active multiple sclerosis. Nature. 2021;597(7878):709–14.

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Da Mesquita S, Papadopoulos Z, Dykstra T, Brase L, Farias FG, Wall M, et al. Meningeal lymphatics affect microglia responses and anti-Aβ immunotherapy. Nature. 2021;593(7858):255–60.

    PubMed  PubMed Central  Google Scholar 

  171. Cunningham C, Wilcockson DC, Campion S, Lunnon K, Perry VH. Central and systemic endotoxin challenges exacerbate the local inflammatory response and increase neuronal death during chronic neurodegeneration. J Neurosci. 2005;25(40):9275–84.

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Neher JJ, Cunningham C. Priming microglia for innate immune memory in the brain. Trends Immunol. 2019;40(4):358–74.

    CAS  PubMed  Google Scholar 

  173. Frank MG, Baratta MV, Sprunger DB, Watkins LR, Maier SF. Microglia serve as a neuroimmune substrate for stress-induced potentiation of CNS pro-inflammatory cytokine responses. Brain Behav Immun. 2007;21(1):47–59.

    CAS  PubMed  Google Scholar 

  174. Raj DDA, Jaarsma D, Holtman IR, Olah M, Ferreira FM, Schaafsma W, et al. Priming of microglia in a DNA-repair deficient model of accelerated aging. Neurobiol Aging. 2014;35(9):2147–60.

    CAS  PubMed  Google Scholar 

  175. Godbout JP, Chen J, Abraham J, Richwine AF, Berg BM, Kelley KW, et al. Exaggerated neuroinflammation and sickness behavior in aged mice after activation of the peripheral innate immune system. Faseb J. 2005;19(7):1329.

    CAS  PubMed  Google Scholar 

  176. Palin K, Cunningham C, Forse P, Perry VH, Platt N. Systemic inflammation switches the inflammatory cytokine profile in CNS Wallerian degeneration. Neurobiol Dis. 2008;30(1):19–29.

    CAS  PubMed  Google Scholar 

  177. Ramaglia V, Hughes TR, Donev RM, Ruseva MM, Wu X, Huitinga I, et al. C3-dependent mechanism of microglial priming relevant to multiple sclerosis. Proc Natl Acad Sci. 2012;109(3):965–70.

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Godoy MCP, Tarelli R, Ferrari CC, Sarchi MIS, Pitossi FJ. Central and systemic IL-1 exacerbates neurodegeneration and motor symptoms in a model of Parkinson’s disease. Brain. 2008;131:1880–94.

    PubMed Central  Google Scholar 

  179. Yin Z, Raj D, Saiepour N, Van Dam D, Brouwer N, Holtman IR, et al. Immune hyperreactivity of Aβ plaque-associated microglia in Alzheimer’s disease. Neurobiol Aging. 2017;55:115–22.

    CAS  PubMed  Google Scholar 

  180. Lopez-Rodriguez AB, Hennessy E, Murray C, Lewis A, de Barra N, Fagan S, et al. Microglial and astrocyte priming in the APP/PS1 model of Alzheimer’s disease: increased vulnerability to acute inflammation and cognitive deficits. Glia. 2019;67:E568.

    Google Scholar 

  181. Bilbo SD. Early-life infection is a vulnerability factor for aging-related glial alterations and cognitive decline. Neurobiol Learn Memory. 2010;94(1):57–64.

    Google Scholar 

  182. Hayes L, An K, Carloni E, Li F, Vincent E, Paranjpe M, et al. Prenatal immune stress induces a prolonged blunting of microglia activation that impacts striatal connectivity. bioRxiv. 2021. https://doi.org/10.1101/2021.12.27.473694.

    Article  PubMed  PubMed Central  Google Scholar 

  183. Mattei D, Ivanov A, Ferrai C, Jordan P, Guneykaya D, Buonfiglioli A, et al. Maternal immune activation results in complex microglial transcriptome signature in the adult offspring that is reversed by minocycline treatment. Transl Psychiatry. 2017;7: e1120.

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Bolton JL, Short AK, Othy S, Kooiker CL, Shao M, Gunn BG, et al. Early stress-induced impaired microglial pruning of excitatory synapses on immature CRH-expressing neurons provokes aberrant adult stress responses. Cell Rep. 2022;38(13): e1120.

    Google Scholar 

  185. Cao P, Chen C, Liu A, Shan Q, Zhu X, Jia C, et al. Early-life inflammation promotes depressive symptoms in adolescence via microglial engulfment of dendritic spines. Neuron. 2021;109(16):2573-89.e9.

    CAS  PubMed  Google Scholar 

  186. Hayes LN, An K, Carloni E, Li F, Vincent E, Trippaers C, et al. Prenatal immune stress blunts microglia reactivity, impairing neurocircuitry. Nature. 2022;610(7931):327–34.

    CAS  PubMed  Google Scholar 

  187. Wendeln AC, Degenhardt K, Kaurani L, Gertig M, Ulas T, Jain G, et al. Innate immune memory in the brain shapes neurological disease hallmarks. Nature. 2018;556(7701):332.

    CAS  PubMed  PubMed Central  Google Scholar 

  188. Zhang X, Kracht L, Lerario AM, Dubbelaar ML, Brouwer N, Wesseling EM, et al. Epigenetic regulation of innate immune memory in microglia. J Neuroinflammation. 2022;19(1):111.

    CAS  PubMed  PubMed Central  Google Scholar 

  189. Block ML, Zecca L, Hong J-S. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007;8(1):57–69.

    CAS  PubMed  Google Scholar 

  190. Haley MJ, Brough D, Quintin J, Allan SM. Microglial priming as trained immunity in the brain. Neuroscience. 2019;405:47–54.

    CAS  PubMed  Google Scholar 

  191. Longhi L, Gesuete R, Perego C, Ortolano F, Sacchi N, Villa P, et al. Long-lasting protection in brain trauma by endotoxin preconditioning. J Cereb Blood Flow Metab. 2011;31(9):1919–29.

    CAS  PubMed  PubMed Central  Google Scholar 

  192. Moshayedi P, Ng G, Kwok JCF, Yeo GSH, Bryant CE, Fawcett JW, et al. The relationship between glial cell mechanosensitivity and foreign body reactions in the central nervous system. Biomaterials. 2014;35(13):3919–25.

    CAS  PubMed  Google Scholar 

  193. Ayata P, Schaefer A. Innate sensing of mechanical properties of brain tissue by microglia. Curr Opin Immunol. 2020;62:123–30.

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Solis AG, Bielecki P, Steach HR, Sharma L, Harman CCD, Yun S, et al. Mechanosensation of cyclical force by PIEZO1 is essential for innate immunity. Nature. 2019;573(7772):69–74.

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Lim C-G, Jang J, Kim C. Cellular machinery for sensing mechanical force. BMB Rep. 2018;51:623.

    CAS  PubMed  PubMed Central  Google Scholar 

  196. Coste B, Mathur J, Schmidt M, Earley TJ, Ranade S, Petrus MJ, et al. Piezo1 and Piezo2 are essential components of distinct mechanically activated cation channels. Science. 2010;330(6000):55–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  197. Jin H, Zhu H, Yang Q, Shen H, Chai G, Zhang B, et al. Microglial Piezo1 senses Aβ fibrils stiffness to restrict Alzheimer’s disease. Neuron. 2022. https://doi.org/10.1016/j.neuron.2022.10.021.

    Article  PubMed  PubMed Central  Google Scholar 

  198. Ivkovic S, Major T, Mitic M, Loncarevic-Vasiljkovic N, Jovic M, Adzic M. Fatty acids as biomodulators of Piezo1 mediated glial mechanosensitivity in Alzheimer’s disease. Life Sci. 2022;297: 120470.

    CAS  PubMed  Google Scholar 

  199. Jäntti H, Sitnikova V, Ishchenko Y, Shakirzyanova A, Giudice L, Ugidos IF, et al. Microglial amyloid beta clearance is driven by PIEZO1 channels. J Neuroinflammation. 2022;19(1):147.

    PubMed  PubMed Central  Google Scholar 

  200. Lopes KdP, Snijders GJL, Humphrey J, Allan A, Sneeboer MAM, Navarro E, et al. Genetic analysis of the human microglial transcriptome across brain regions, aging and disease pathologies. Nat Genetics. 2022;54(1):4–17.

    CAS  PubMed  Google Scholar 

  201. Gandal MJ, Zhang P, Hadjimichael E, Walker RL, Chen C, Liu S, et al. Transcriptome-wide isoform-level dysregulation in ASD, schizophrenia, and bipolar disorder. Science. 2018;362(6420):8127.

    Google Scholar 

  202. Malhi GS, Mann JJ. Depression. Lancet. 2018;392(10161):2299–312.

    PubMed  Google Scholar 

  203. Penninx BWJH, Pine DS, Holmes EA, Reif A. Anxiety disorders. Lancet. 2021;397(10277):914–27.

    PubMed  PubMed Central  Google Scholar 

  204. McCarron RM, Shapiro B, Rawles J, Luo J. Depression. Ann Intern Med. 2021;174(5):65–80.

    Google Scholar 

  205. Beurel E, Toups M, Nemeroff CB. The bidirectional relationship of depression and inflammation: double trouble. Neuron. 2020;107(2):234–56.

    CAS  PubMed  PubMed Central  Google Scholar 

  206. Klawonn AM, Fritz M, Castany S, Pignatelli M, Canal C, Similä F, et al. Microglial activation elicits a negative affective state through prostaglandin-mediated modulation of striatal neurons. Immunity. 2021;54(2):225-34.e6.

    CAS  PubMed  Google Scholar 

  207. Hongrui Z, Guo Y, Huang A, Shen H, Chen Y, Song J, et al. HDAC3-regulated PGE2 production by microglia induces phobic anxiety susceptibility after stroke and pointedly exploiting a signal-targeted gamma visual stimulation new therapy. Front Immunol. 2022;13:845678.

    Google Scholar 

  208. Peng Z, Li X, Li J, Dong Y, Gao Y, Liao Y, et al. Dlg1 knockout inhibits microglial activation and alleviates lipopolysaccharide-induced depression-like behavior in mice. Neurosci Bull. 2021;37(12):1671–82.

    CAS  PubMed  PubMed Central  Google Scholar 

  209. Li W, Ali T, Zheng C, He K, Liu Z, Shah FA, et al. Anti-depressive-like behaviors of APN KO mice involve Trkb/BDNF signaling related neuroinflammatory changes. Mol Psychiatry. 2022;27(2):1047–58.

    CAS  PubMed  Google Scholar 

  210. Kopp KO, Glotfelty EJ, Li Y, Greig NH. Glucagon-like peptide-1 (GLP-1) receptor agonists and neuroinflammation: implications for neurodegenerative disease treatment. Pharmacol Res. 2022;186: 106550.

    CAS  PubMed  Google Scholar 

  211. Yang F, Wang X, Qi J, Zhang K, Jiang Y, Feng B, et al. Glucagon-like peptide 1 receptor activation inhibits microglial pyroptosis via promoting mitophagy to alleviate depression-like behaviors in diabetic mice. Nutrients. 2022;15:38.

    PubMed  PubMed Central  Google Scholar 

  212. Dionysis N, Theodora M, Alexia P, Anastasia F, George B, Yassemi K, et al. Microglia activation in the presence of intact blood–brain barrier and disruption of hippocampal neurogenesis via IL-6 and IL-18 mediate early diffuse neuropsychiatric lupus. Ann Rheum Dis. 2023;82(5):646.

    Google Scholar 

  213. Kopec AM, Smith CJ, Bilbo SD. Neuro-immune mechanisms regulating social behavior: dopamine as mediator? Trends Neurosci. 2019;42(5):337–48.

    CAS  PubMed  PubMed Central  Google Scholar 

  214. Wu J, Li Y, Huang Y, Liu L, Zhang H, Nagy C, et al. Integrating spatial and single-nucleus transcriptomic data elucidates microglial-specific responses in female cynomolgus macaques with depressive-like behaviors. Nat Neurosci. 2023. https://doi.org/10.1038/s41593-023-01379-4.

    Article  PubMed  PubMed Central  Google Scholar 

  215. Nie X, Kitaoka S, Tanaka K, Segi-Nishida E, Imoto Y, Ogawa A, et al. The innate immune receptors TLR2/4 mediate repeated social defeat stress-induced social avoidance through prefrontal microglial activation. Neuron. 2018;99(3):464-79.e7.

    CAS  PubMed  Google Scholar 

  216. Franklin TC, Wohleb ES, Zhang Y, Fogaça M, Hare B, Duman RS. Persistent increase in microglial RAGE contributes to chronic stress-induced priming of depressive-like behavior. Biol Psychiat. 2018;83(1):50–60.

    CAS  PubMed  Google Scholar 

  217. McKim DB, Weber MD, Niraula A, Sawicki CM, Liu X, Jarrett BL, et al. Microglial recruitment of IL-1β-producing monocytes to brain endothelium causes stress-induced anxiety. Mol Psychiatry. 2018;23(6):1421–31.

    CAS  PubMed  Google Scholar 

  218. Yue N, Huang H, Zhu X, Han Q, Wang Y, Li B, et al. Activation of P2X7 receptor and NLRP3 inflammasome assembly in hippocampal glial cells mediates chronic stress-induced depressive-like behaviors. J Neuroinflammation. 2017;14(1):102.

    PubMed  PubMed Central  Google Scholar 

  219. Wang Y-L, Han Q-Q, Gong W-Q, Pan D-H, Wang L-Z, Hu W, et al. Microglial activation mediates chronic mild stress-induced depressive- and anxiety-like behavior in adult rats. J Neuroinflammation. 2018;15(1):21.

    PubMed  PubMed Central  Google Scholar 

  220. Feng X, Zhao Y, Yang T, Song M, Wang C, Yao Y, et al. Glucocorticoid-driven NLRP3 inflammasome activation in hippocampal microglia mediates chronic stress-induced depressive-like behaviors. Front Mol Neurosci. 2019;12:210.

    CAS  PubMed  PubMed Central  Google Scholar 

  221. Li S, Fang Y, Zhang Y, Song M, Zhang X, Ding X, et al. Microglial NLRP3 inflammasome activates neurotoxic astrocytes in depression-like mice. Cell Rep. 2022;41: 111532.

    CAS  PubMed  Google Scholar 

  222. Milior G, Lecours C, Samson L, Bisht K, Poggini S, Pagani F, et al. Fractalkine receptor deficiency impairs microglial and neuronal responsiveness to chronic stress. Brain Behav Immun. 2016;55:114–25.

    CAS  PubMed  Google Scholar 

  223. Rimmerman N, Schottlender N, Reshef R, Dan-Goor N, Yirmiya R. The hippocampal transcriptomic signature of stress resilience in mice with microglial fractalkine receptor (CX3CR1) deficiency. Brain Behav Immun. 2017;61:184–96.

    CAS  PubMed  Google Scholar 

  224. Wohleb ES, Terwilliger R, Duman CH, Duman RS. Stress-induced neuronal colony stimulating factor 1 provokes microglia-mediated neuronal remodeling and depressive-like behavior. Biol Psychiat. 2018;83(1):38–49.

    CAS  PubMed  Google Scholar 

  225. Yasumoto Y, Stoiljkovic M, Kim Jd, Sestan Pesa M, Gao X-B, Diano S, et al. Ucp2-dependent microglia-neuronal coupling controls ventral hippocampal circuit function and anxiety-like behavior. Mol Psychiatry. 2021;26:1–13.

    Google Scholar 

  226. Fan C, Li Y, Lan T, Wang W, Long Y, Yu SY. Microglia secrete miR-146a-5p-containing exosomes to regulate neurogenesis in depression. Mol Ther. 2022;30(3):1300–14.

    CAS  PubMed  Google Scholar 

  227. Lee J-S, Lee S-B, Kim D-W, Shin N, Jeong S-J, Yang C-H, et al. Social isolation–related depression accelerates ethanol intake via microglia-derived neuroinflammation. Sci Adv. 2021;7(45):3400.

    Google Scholar 

  228. Khan KM, Bierlein-De La Rosa G, Biggerstaff N, Pushpavathi Selvakumar G, Wang R, Mason S, et al. Adolescent ethanol drinking promotes hyperalgesia, neuroinflammation and serotonergic deficits in mice that persist into adulthood. Brain, Behav Immunity. 2023;107:419–31.

    CAS  Google Scholar 

  229. Socodato R, Henriques JF, Portugal CC, Almeida TO, Tedim-Moreira J, Alves RL, et al. Daily alcohol intake triggers aberrant synaptic pruning leading to synapse loss and anxiety-like behavior. Sci Signal. 2020;13(650):5754.

    Google Scholar 

  230. Warden AS, Wolfe SA, Khom S, Varodayan FP, Patel RR, Steinman MQ, et al. Microglia control escalation of drinking in alcohol-dependent mice: genomic and synaptic drivers. Biol Psychiat. 2020;88(12):910–21.

    CAS  PubMed  Google Scholar 

  231. Adeluyi A, Guerin L, Fisher ML, Galloway A, Cole RD, Chan SSL, et al. Microglia morphology and proinflammatory signaling in the nucleus accumbens during nicotine withdrawal. Sci Adv. 2019;5(10):7031.

    Google Scholar 

  232. Setiawan E, Attwells S, Wilson AA, Mizrahi R, Rusjan PM, Miler L, et al. Association of translocator protein total distribution volume with duration of untreated major depressive disorder: a cross-sectional study. Lancet Psychiatry. 2018;5(4):339–47.

    PubMed  Google Scholar 

  233. Setiawan E, Wilson A, Mizrahi R, Rusjan P, Miler L, Rajkowska G, et al. Role of translocator protein density, a marker of neuroinflammation, in the brain during major depressive episodes. JAMA Psychiat. 2015;72:268.

    Google Scholar 

  234. Holmes SE, Hinz R, Conen S, Gregory CJ, Matthews JC, Anton-Rodriguez JM, et al. Elevated translocator protein in anterior cingulate in major depression and a role for inflammation in suicidal thinking: a positron emission tomography study. Biol Psychiat. 2018;83(1):61–9.

    CAS  PubMed  Google Scholar 

  235. Scheepstra KWF, Mizee MR, van Scheppingen J, Adelia A, Wever DD, Mason MRJ, et al. Microglia transcriptional profiling in major depressive disorder shows inhibition of cortical gray matter microglia. Biol Psychiatry. 2023. https://doi.org/10.1016/j.biopsych.2023.04.020.

    Article  PubMed  Google Scholar 

  236. Enache D, Pariante CM, Mondelli V. Markers of central inflammation in major depressive disorder: a systematic review and meta-analysis of studies examining cerebrospinal fluid, positron emission tomography and post-mortem brain tissue. Brain Behav Immun. 2019;81:24–40.

    PubMed  Google Scholar 

  237. Lord C, Elsabbagh M, Baird G, Veenstra-Vanderweele J. Autism spectrum disorder. Lancet. 2018;392(10146):508–20.

    PubMed  PubMed Central  Google Scholar 

  238. Velmeshev D, Schirmer L, Jung D, Haeussler M, Perez Y, Mayer S, et al. Single-cell genomics identifies cell type–specific molecular changes in autism. Science. 2019;364(6441):685–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  239. Su L, Zhang M, Ji F, Zhao J, Wang Y, Wang W, et al. Microglia homeostasis mediated by epigenetic ARID1A regulates neural progenitor cells response and leads to autism-like behaviors. Mol Psychiatry. 2022. https://doi.org/10.1038/s41380-022-01703-7.

    Article  PubMed  PubMed Central  Google Scholar 

  240. Bagni C, Zukin RS. A synaptic perspective of Fragile X syndrome and Autism spectrum disorders. Neuron. 2019;101(6):1070–88.

    CAS  PubMed  PubMed Central  Google Scholar 

  241. Sarn N, Jaini R, Thacker S, Lee H, Dutta R, Eng C. Cytoplasmic-predominant Pten increases microglial activation and synaptic pruning in a murine model with autism-like phenotype. Mol Psychiatry. 2021;26(5):1458–71.

    CAS  PubMed  Google Scholar 

  242. Kim HJ, Cho MH, Shim WH, Kim JK, Jeon EY, Kim DH, et al. Deficient autophagy in microglia impairs synaptic pruning and causes social behavioral defects. Mol Psychiatry. 2017;22(11):1576–84.

    CAS  PubMed  Google Scholar 

  243. Andersson M, Tangen Ä, Farde L, Bölte S, Halldin C, Borg J, et al. Serotonin transporter availability in adults with autism—a positron emission tomography study. Mol Psychiatry. 2021;26(5):1647–58.

    CAS  PubMed  Google Scholar 

  244. Ji C, Tang Y, Zhang Y, Huang X, Li C, Yang Y, et al. Glutaminase 1 deficiency confined in forebrain neurons causes autism spectrum disorder-like behaviors. Cell Rep. 2023;42: 112712.

    CAS  PubMed  Google Scholar 

  245. Hornig M, Bresnahan MA, Che X, Schultz AF, Ukaigwe JE, Eddy ML, et al. Prenatal fever and autism risk. Mol Psychiatry. 2018;23(3):759–66.

    CAS  PubMed  Google Scholar 

  246. Al-Haddad BJS, Jacobsson B, Chabra S, Modzelewska D, Olson EM, Bernier R, et al. Long-term risk of neuropsychiatric disease after exposure to infection in utero. JAMA Psychiat. 2019;76(6):594–602.

    Google Scholar 

  247. Han VX, Patel S, Jones HF, Dale RC. Maternal immune activation and neuroinflammation in human neurodevelopmental disorders. Nat Rev Neurol. 2021;17(9):564–79.

    PubMed  Google Scholar 

  248. Kracht L, Borggrewe M, Eskandar S, Brouwer N, de Sousa C, Lopes SM, Laman JD, et al. Human fetal microglia acquire homeostatic immune-sensing properties early in development. Science. 2020;369(6503):530–7.

    CAS  PubMed  Google Scholar 

  249. Coutinho E, Menassa DA, Jacobson L, West SJ, Domingos J, Moloney TC, et al. Persistent microglial activation and synaptic loss with behavioral abnormalities in mouse offspring exposed to CASPR2-antibodies in utero. Acta Neuropathol. 2017;134(4):567–83.

    CAS  PubMed  PubMed Central  Google Scholar 

  250. Rosin JM, Sinha S, Biernaskie J, Kurrasch DM. A subpopulation of embryonic microglia respond to maternal stress and influence nearby neural progenitors. Dev Cell. 2021;56(9):1326-45.e6.

    CAS  PubMed  Google Scholar 

  251. López-Aranda MF, Chattopadhyay I, Boxx GM, Fraley ER, Silva TK, Zhou M, et al. Postnatal immune activation causes social deficits in a mouse model of tuberous sclerosis: role of microglia and clinical implications. Sci Adv. 2021;7(38):eabf2073.

    PubMed  PubMed Central  Google Scholar 

  252. Ikezu S, Yeh H, Delpech J-C, Woodbury ME, Van Enoo AA, Ruan Z, et al. Inhibition of colony stimulating factor 1 receptor corrects maternal inflammation-induced microglial and synaptic dysfunction and behavioral abnormalities. Mol Psychiatry. 2021;26(6):1808–31.

    CAS  PubMed  Google Scholar 

  253. Andoh M, Shibata K, Okamoto K, Onodera J, Morishita K, Miura Y, et al. Exercise reverses behavioral and synaptic abnormalities after maternal inflammation. Cell Rep. 2019;27(10):2817-25.e5.

    CAS  PubMed  Google Scholar 

  254. Gkogkas CG, Khoutorsky A, Ran I, Rampakakis E, Nevarko T, Weatherill DB, et al. Autism-related deficits via dysregulated eIF4E-dependent translational control. Nature. 2013;493(7432):371–7.

    CAS  PubMed  Google Scholar 

  255. Gonatopoulos-Pournatzis T, Niibori R, Salter E, Weatheritt R, Tsang B, Farhangmehr S, et al. Autism-misregulated eIF4G microexons control synaptic translation and higher order cognitive functions. Mol Cell. 2020;77:1176.

    CAS  PubMed  Google Scholar 

  256. de la Torre-Ubieta L, Won H, Stein JL, Geschwind DH. Advancing the understanding of autism disease mechanisms through genetics. Nat Med. 2016;22(4):345–61.

    PubMed  PubMed Central  Google Scholar 

  257. Xu Z-X, Kim GH, Tan J-W, Riso AE, Sun Y, Xu EY, et al. Elevated protein synthesis in microglia causes autism-like synaptic and behavioral aberrations. Nat Commun. 2020;11(1):1797.

    CAS  PubMed  PubMed Central  Google Scholar 

  258. Marder SR, Cannon TD. Schizophrenia. N Engl J Med. 2019;381(18):1753–61.

    CAS  PubMed  Google Scholar 

  259. Dietz AG, Goldman SA, Nedergaard M. Glial cells in schizophrenia: a unified hypothesis. Lancet Psychiatry. 2020;7(3):272–81.

    PubMed  Google Scholar 

  260. Purves-Tyson TD, Weber-Stadlbauer U, Richetto J, Rothmond DA, Labouesse MA, Polesel M, et al. Increased levels of midbrain immune-related transcripts in schizophrenia and in murine offspring after maternal immune activation. Mol Psychiatry. 2021;26(3):849–63.

    PubMed  Google Scholar 

  261. Park G-H, Noh H, Shao Z, Ni P, Qin Y, Liu D, et al. Activated microglia cause metabolic disruptions in developmental cortical interneurons that persist in interneurons from individuals with schizophrenia. Nat Neurosci. 2020;23(11):1352–64.

    CAS  PubMed  PubMed Central  Google Scholar 

  262. Janova H, Arinrad S, Balmuth E, Mitjans M, Hertel J, Habes M, et al. Microglia ablation alleviates myelin-associated catatonic signs in mice. J Clin Investig. 2018;128(2):734–45.

    PubMed  Google Scholar 

  263. Da Silva T, Guma E, Hafizi S, Koppel A, Rusjan P, Kennedy JL, et al. Genetically predicted brain C4A expression is associated with TSPO and hippocampal morphology. Biol Psychiat. 2021;90(9):652–60.

    PubMed  Google Scholar 

  264. Sellgren C, Gracias J, Watmuff B, Biag J, Thanos J, Whittredge P, et al. Increased synapse elimination by microglia in schizophrenia patient-derived models of synaptic pruning. Nat Neurosci. 2019. https://doi.org/10.1038/s41593-018-0334-7.

    Article  PubMed  PubMed Central  Google Scholar 

  265. Yilmaz M, Yalcin E, Presumey J, Aw E, Ma M, Whelan CW, et al. Overexpression of schizophrenia susceptibility factor human complement C4A promotes excessive synaptic loss and behavioral changes in mice. Nat Neurosci. 2021;24(2):214–24.

    CAS  PubMed  Google Scholar 

  266. Shalev A, Liberzon I, Marmar C. Post-traumatic stress disorder. N Engl J Med. 2017;376(25):2459–69.

    PubMed  Google Scholar 

  267. Chen MB, Jiang X, Quake SR, Südhof TC. Persistent transcriptional programmes are associated with remote memory. Nature. 2020;587(7834):437–42.

    CAS  PubMed  PubMed Central  Google Scholar 

  268. 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):688–94.

    CAS  PubMed  Google Scholar 

  269. Raja SN, Carr DB, Cohen M, Finnerup NB, Flor H, Gibson S, et al. The revised International Association for the study of pain definition of pain: concepts, challenges, and compromises. Pain. 2020;161(9):1976.

    PubMed  PubMed Central  Google Scholar 

  270. Cao K, Hu Y, Gao Z. Sense to tune: engaging microglia with dynamic neuronal activity. Neurosci Bull. 2022. https://doi.org/10.1007/s12264-022-01010-9.

    Article  PubMed  PubMed Central  Google Scholar 

  271. Tansley S, Uttam S, Ureña Guzmán A, Yaqubi M, Pacis A, Parisien M, et al. Single-cell RNA sequencing reveals time- and sex-specific responses of mouse spinal cord microglia to peripheral nerve injury and links ApoE to chronic pain. Nat Commun. 2022;13(1):843.

    CAS  PubMed  PubMed Central  Google Scholar 

  272. Tansley S, Gu N, Guzmán AU, Cai W, Wong C, Lister KC, et al. Microglia-mediated degradation of perineuronal nets promotes pain. Science. 2022;377(6601):80–6.

    CAS  PubMed  Google Scholar 

  273. Zhou L-J, Peng J, Xu Y-N, Zeng W-J, Zhang J, Wei X, et al. Microglia are indispensable for synaptic plasticity in the spinal dorsal horn and chronic pain. Cell Rep. 2019;27(13):3844-59.e6.

    CAS  PubMed  PubMed Central  Google Scholar 

  274. Kohno K, Shirasaka R, Yoshihara K, Mikuriya S, Tanaka K, Takanami K, et al. A spinal microglia population involved in remitting and relapsing neuropathic pain. Science. 2022;376(6588):86–90.

    CAS  PubMed  Google Scholar 

  275. Parusel S, Yi M-H, Hunt CL, Wu L-J. Chemogenetic and optogenetic manipulations of microglia in chronic pain. Neurosci Bull. 2022. https://doi.org/10.1007/s12264-022-00937-3.

    Article  PubMed  PubMed Central  Google Scholar 

  276. Treasure J, Duarte T, Schmidt U. Eating disorders. Lancet. 2020;395:899–911.

    PubMed  Google Scholar 

  277. da Silva AA, Carmo do JM, Hall JE. CNS regulation of glucose homeostasis: role of the leptin-melanocortin system. Curr Diab Rep. 2020;20(7):29.

    PubMed  Google Scholar 

  278. Grayson BE, Seeley RJ, Sandoval DA. Wired on sugar: the role of the CNS in the regulation of glucose homeostasis. Nat Rev Neurosci. 2013;14(1):24–37.

    CAS  PubMed  Google Scholar 

  279. Schwartz MW, Seeley RJ, Tschöp MH, Woods SC, Morton GJ, Myers MG, et al. Cooperation between brain and islet in glucose homeostasis and diabetes. Nature. 2013;503(7474):59–66.

    CAS  PubMed  PubMed Central  Google Scholar 

  280. Thaler JP, Yi CX, Schur EA, Guyenet SJ, Hwang BH, Dietrich MO, et al. Obesity is associated with hypothalamic injury in rodents and humans. J Clin Invest. 2012;122(1):153–62.

    CAS  PubMed  Google Scholar 

  281. Kim JD, Yoon NA, Jin S, Diano S. Microglial UCP2 mediates inflammation and obesity induced by high-fat feeding. Cell Metab. 2019;30(5):952-62.e5.

    CAS  PubMed  PubMed Central  Google Scholar 

  282. Cheng J, Ma X, Li C, Ullah R, Wang X, Long J, et al. Diet-induced inflammation in the anterior paraventricular thalamus induces compulsive sucrose-seeking. Nat Neurosci. 2022;25:1009–13.

    CAS  PubMed  Google Scholar 

  283. Bekhbat M, Neigh GN. Sex differences in the neuro-immune consequences of stress: focus on depression and anxiety. Brain Behav Immun. 2018;67:1–12.

    PubMed  Google Scholar 

  284. Loomes R, Hull L, Mandy WPL. What is the male-to-female ratio in autism spectrum disorder? A systematic review and meta-analysis. J Am Acad Child Adolesc Psychiatry. 2017;56(6):466–74.

    PubMed  Google Scholar 

  285. Jauhar S, Johnstone M, McKenna P. Schizophrenia. The Lancet. 2022;399:473–86.

    CAS  Google Scholar 

  286. Tiihonen J, Koskuvi M, Storvik M, Hyötyläinen I, Gao Y, Puttonen KA, et al. Sex-specific transcriptional and proteomic signatures in schizophrenia. Nat Commun. 2019;10(1):3933.

    CAS  PubMed  PubMed Central  Google Scholar 

  287. Maitra M, Mitsuhashi H, Rahimian R, Chawla A, Yang J, Fiori LM, et al. Cell type specific transcriptomic differences in depression show similar patterns between males and females but implicate distinct cell types and genes. Nat Commun. 2023;14(1):2912.

    CAS  PubMed  PubMed Central  Google Scholar 

  288. Lenz KM, McCarthy MM. A starring role for microglia in brain sex differences. Neuroscientist. 2015;21(3):306–21.

    CAS  PubMed  Google Scholar 

  289. Nelson LH, Saulsbery AI, Lenz KM. Small cells with big implications: microglia and sex differences in brain development, plasticity and behavioral health. Prog Neurobiol. 2019;176:103–19.

    PubMed  Google Scholar 

  290. VanRyzin JW, Marquardt AE, Argue KJ, Vecchiarelli HA, Ashton SE, Arambula SE, et al. Microglial phagocytosis of newborn cells is induced by endocannabinoids and sculpts sex differences in juvenile rat social play. Neuron. 2019;102(2):435-49.e6.

    CAS  PubMed  PubMed Central  Google Scholar 

  291. Halladay AK, Bishop S, Constantino JN, Daniels AM, Koenig K, Palmer K, et al. Sex and gender differences in autism spectrum disorder: summarizing evidence gaps and identifying emerging areas of priority. Mol Autism. 2015;6:1.

    Google Scholar 

  292. Ochoa S, Usall J, Cobo J, Labad X, Kulkarni J. Gender differences in schizophrenia and first-episode psychosis: a comprehensive literature review. Schizophr Res Treat. 2012;2012:916198.

    Google Scholar 

  293. Ceasrine AM, Devlin BA, Bolton JL, Green LA, Jo YC, Huynh C, et al. Maternal diet disrupts the placenta–brain axis in a sex-specific manner. Nat Metab. 2022;4(12):1732–45.

    CAS  PubMed  PubMed Central  Google Scholar 

  294. Ma S, Skarica M, Li Q, Xu C, Risgaard RD, Tebbenkamp ATN, et al. Molecular and cellular evolution of the primate dorsolateral prefrontal cortex. Science. 2022. https://doi.org/10.1126/science.abo7257.

    Article  PubMed  PubMed Central  Google Scholar 

  295. Gandal MJ, Haney JR, Wamsley B, Yap CX, Parhami S, Emani PS, et al. Broad transcriptomic dysregulation occurs across the cerebral cortex in ASD. Nature. 2022;611(7936):532–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  296. Giampaolo M, Mélanie M-B, Farah C, Le Caroline D, Etienne S, Gilles H, et al. Distinct P2Y receptors mediate extension and retraction of microglial processes in epileptic and peritumoral human tissue. J Neurosci. 2020;40(7):1373.

    Google Scholar 

  297. Morin-Brureau M, Milior G, Royer J, Chali F, Le Duigou C, Savary E, et al. Microglial phenotypes in the human epileptic temporal lobe. Brain. 2018;141(12):3343–60.

    PubMed  Google Scholar 

  298. Dumas AA, Borst K, Prinz M. Current tools to interrogate microglial biology. Neuron. 2021;109(18):2805–19.

    CAS  PubMed  Google Scholar 

  299. Schafer S, Mansour AA, Schlachetzki J, Pena M, Ghassemzadeh S, Mitchell L, et al. An in vivo neuroimmune organoid model to study human microglia phenotypes. Cell. 2023;186:2111–26.

    CAS  PubMed  Google Scholar 

  300. Wang M, Zhang L, Gage FH. Modeling neuropsychiatric disorders using human induced pluripotent stem cells. Protein Cell. 2020;11(1):45–59.

    CAS  PubMed  Google Scholar 

  301. Andoh M, Koyama R. Assessing microglial dynamics by live imaging. Front Immunol. 2021;12:617564.

    CAS  PubMed  PubMed Central  Google Scholar 

  302. Li Y, Ritzel RM, Khan N, Cao T, He J, Lei Z, et al. Delayed microglial depletion after spinal cord injury reduces chronic inflammation and neurodegeneration in the brain and improves neurological recovery in male mice. Theranostics. 2020;10(25):11376–403.

    CAS  PubMed  PubMed Central  Google Scholar 

  303. Cheng D, Qin Z-S, Zheng Y, Xie J-Y, Liang S-S, Zhang J-L, et al. Minocycline, a classic antibiotic, exerts psychotropic effects by normalizing microglial neuroinflammation-evoked tryptophan-kynurenine pathway dysregulation in chronically stressed male mice. Brain Behav Immun. 2023;107:305–18.

    CAS  PubMed  Google Scholar 

  304. Tabassum S, Misrani A, Huo Q, Ahmed A, Long C, Yang L. Minocycline ameliorates chronic unpredictable mild stress-induced neuroinflammation and abnormal mPFC-HIPP oscillations in mice. Mol Neurobiol. 2022;59(11):6874–95.

    CAS  PubMed  Google Scholar 

  305. Bassett B, Subramaniyam S, Fan Y, Varney S, Pan H, Carneiro AMD, et al. Minocycline alleviates depression-like symptoms by rescuing decrease in neurogenesis in dorsal hippocampus via blocking microglia activation/phagocytosis. Brain Behav Immun. 2021;91:519–30.

    CAS  PubMed  Google Scholar 

  306. Hellmann-Regen J, Clemens V, Grözinger M, Kornhuber J, Reif A, Prvulovic D, et al. Effect of minocycline on depressive symptoms in patients with treatment-resistant depression: a randomized clinical trial. JAMA Netw Open. 2022;5(9):e2230367.

    PubMed  PubMed Central  Google Scholar 

  307. Deakin B, Suckling J, Barnes TRE, Byrne K, Chaudhry IB, Dazzan P, et al. The benefit of minocycline on negative symptoms of schizophrenia in patients with recent-onset psychosis (BeneMin): a randomised, double-blind, placebo-controlled trial. Lancet Psychiatry. 2018;5(11):885–94.

    PubMed  PubMed Central  Google Scholar 

  308. Krynicki CR, Dazzan P, Pariante CM, Barnes NM, Vincent RC, Roberts A, et al. Deconstructing depression and negative symptoms of schizophrenia; differential and longitudinal immune correlates, and response to minocycline treatment. Brain Behav Immun. 2021;91:498–504.

    CAS  PubMed  Google Scholar 

  309. Nettis MA, Lombardo G, Hastings C, Zajkowska Z, Mariani N, Nikkheslat N, et al. Augmentation therapy with minocycline in treatment-resistant depression patients with low-grade peripheral inflammation: results from a double-blind randomised clinical trial. Neuropsychopharmacology. 2021;46(5):939–48.

    CAS  PubMed  PubMed Central  Google Scholar 

  310. Liu Y, Hu P, Zheng Z, Zhong D, Xie W, Tang Z, et al. Photoresponsive vaccine-like CAR-M system with high-efficiency central immune regulation for inflammation-related depression. Adv Mater. 2022;34(11):2108525.

    CAS  Google Scholar 

  311. Yu X, Bai Y, Han B, Ju M, Tang T, Shen L, et al. Extracellular vesicle-mediated delivery of circDYM alleviates CUS-induced depressive-like behaviours. J Extracell Vesicles. 2022;11(1): e12185.

    CAS  PubMed  PubMed Central  Google Scholar 

  312. Yao W, Cao Q, Luo S, He L, Yang C, Chen J, et al. Microglial ERK-NRBP1-CREB-BDNF signaling in sustained antidepressant actions of (R)-ketamine. Mol Psychiatry. 2022;27(3):1618–29.

    CAS  PubMed  Google Scholar 

  313. Inta D, Guzman R, Gass P. Microglia activation and adult neurogenesis in the hippocampus: new clues about the antidepressant effect of minocycline. Brain Behav Immun. 2021;94:27–8.

    PubMed  Google Scholar 

  314. Marwaha S, Palmer E, Suppes T, Cons E, Young AH, Upthegrove R. Novel and emerging treatments for major depression. The Lancet. 2023;401(10371):141–53.

    CAS  Google Scholar 

  315. Rimmerman N, Verdiger H, Goldenberg H, Naggan L, Robinson E, Kozela E, et al. Microglia and their LAG3 checkpoint underlie the antidepressant and neurogenesis-enhancing effects of electroconvulsive stimulation. Mol Psychiatry. 2022;27(2):1120–35.

    CAS  PubMed  Google Scholar 

  316. Adaikkan C, Middleton SJ, Marco A, Pao P-C, Mathys H, Kim DN-W, et al. Gamma entrainment binds higher-order brain regions and offers neuroprotection. Neuron. 2019;102(5):929–43.

    CAS  PubMed  PubMed Central  Google Scholar 

  317. Guan A, Wang S, Huang A, Qiu C, Li Y, Li X, et al. The role of gamma oscillations in central nervous system diseases: mechanism and treatment. Front Cell Neurosci. 2022;16:962957.

    CAS  PubMed  PubMed Central  Google Scholar 

  318. Adaikkan C, Tsai L-H. Gamma entrainment: impact on neurocircuits, glia, and therapeutic opportunities. Trends Neurosci. 2020;43(1):24–41.

    CAS  PubMed  Google Scholar 

  319. Balbi M, Xiao D, Jativa Vega M, Hu H, Vanni MP, Bernier L-P, et al. Gamma frequency activation of inhibitory neurons in the acute phase after stroke attenuates vascular and behavioral dysfunction. Cell Rep. 2021;34(5): 108696.

    CAS  PubMed  Google Scholar 

  320. Soula M, Martín-Ávila A, Zhang Y, Dhingra A, Nitzan N, Sadowski MJ, et al. Forty-hertz light stimulation does not entrain native gamma oscillations in Alzheimer’s disease model mice. Nat Neurosci. 2023;26(4):570–8.

    CAS  PubMed  Google Scholar 

  321. Green KN, Crapser JD, Hohsfield LA. To kill a microglia: a case for CSF1R inhibitors. Trends Immunol. 2020;41(9):771–84.

    CAS  PubMed  PubMed Central  Google Scholar 

  322. Xu Z, Rao Y, Huang Y, Zhou T, Feng R, Xiong S, et al. Efficient strategies for microglia replacement in the central nervous system. Cell Rep. 2020;32(6): 108041.

    CAS  PubMed  Google Scholar 

  323. Shibuya Y, Kumar KK, Mader MM-D, Yoo Y, Ayala LA, Zhou M, et al. Treatment of a genetic brain disease by CNS-wide microglia replacement. Sci Transl Med. 2022;14(636): eabl9945.

    CAS  PubMed  PubMed Central  Google Scholar 

  324. Vieira R, Mariani JN, Huynh NPT, Stephensen HJT, Solly R, Tate A, et al. Young glial progenitor cells competitively replace aged and diseased human glia in the adult chimeric mouse brain. Nat Biotechnol. 2023. https://doi.org/10.1038/s41587-023-01798-5.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was funded by the National Natural Science Foundation of China (82272225, 81860249) and Wujieping Medical Foundation (320.6750. 2021-03-1).

Author information

Authors and Affiliations

Authors

Contributions

H-RZ conceptualized the review. H-RZ and AG were the primary authors writing and revising this manuscript. AG and J-YL provided the figure design. LP, ZZ and SW critically revised the manuscript. All authors read and approved the final version of the manuscript.

Corresponding authors

Correspondence to Hongrui Zhu, Zhi Zhang or Sheng Wang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhu, H., Guan, A., Liu, J. et al. Noteworthy perspectives on microglia in neuropsychiatric disorders. J Neuroinflammation 20, 223 (2023). https://doi.org/10.1186/s12974-023-02901-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12974-023-02901-y

Keywords