Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Aug 30.
Published before final editing as: J Physiol. 2025 Jun 18:10.1113/JP288573. doi: 10.1113/JP288573

Microglial Phagocytosis in Epilepsy: Mechanisms and Impact

Abhijeet S Barath 1,2,3, Long-Jun Wu 1,3
PMCID: PMC12396501  NIHMSID: NIHMS2105703  PMID: 40532092

Abstract

Microglia are resident immune cells critical in maintaining brain homeostasis via their surveillance and phagocytosis function. Under disease contexts like seizures and epilepsy, microglial phagocytic signaling is activated in response to both inflammatory and non-inflammatory cell death. This process involves a range of well-characterized ‘find me’ and ‘eat me’ signals, phagocytic receptors, and less well-characterized intracellular signaling pathways. In addition, epigenetic and transcriptional regulators orchestrate microglial responses to seizures, including the integration of phagocytic and inflammatory pathways. Interestingly, while inhibiting phagocytosis has been shown to improve neuronal survival and cognitive performance after seizures, it paradoxically increases the risk of developing spontaneous recurrent seizures (SRS). Reconciling these dual effects requires a deeper understanding the spatiotemporal dynamics of microglial phagocytosis. The objective of this review is to examine the mechanisms and impact of microglial phagocytosis in the context of epilepsy and to highlight unresolved questions that warrant further investigation in this emerging field.

Keywords: Epilepsy, seizures, microglia, phagocytosis, lysosomes, epileptogenesis, neuropathology

Graphical Abstract

graphic file with name nihms-2105703-f0004.jpg

Introduction

Epilepsy is a significant neurological disorder affecting approximately 50 million people worldwide, including 2.8 million in the United States (Beghi et al., 2019; Kobau et al., 2023). Among these individuals, 13-36% suffer from drug-resistant epilepsy, characterized by the inability to achieve sustained seizure freedom despite adequate trials of two or more anti-seizure medications (Sultana et al., 2021). One proposed explanation for this therapeutic failure is that current drugs may not adequately target key mechanisms, including those involving non-neuronal cells (Bazhanova et al., 2021). In this context, microglia have emerged as promising therapeutic targets for epilepsy. These cells are the resident immune cells of the central nervous system and function as professional phagocytes. Originating from the yolk sac during early embryonic development, microglia migrate to the CNS, where they are maintained through self-renewal (Saijo et al., 2011).

Microglia play a crucial role in regulating neuronal activity during acute seizures and orchestrating responses to tissue injury in the sub-acute and chronic phases (Bosco et al., 2018; Umpierre & Wu, 2021; Zhao et al., 2024). Neuronal damage following seizures activate microglia, which subsequently reshape neuronal organization through inflammatory and phagocytic signaling (Figure 1). These processes significantly impact cognition and future seizure risk, both of which are critical clinical outcomes (Wyatt-Johnson & Brewster, 2019; Novak et al., 2022; Yu et al., 2023). Aberrant microglial phagocytosis has also been implicated in epileptogenesis (Chu et al., 2010; Libbey et al., 2010; Ma et al., 2013). A deeper understanding of microglial phagocytosis in epilepsy may reveal novel therapeutic targets, particularly for patients with drug-resistant epilepsy. This review examines recent advances in understanding the mechanisms, regulatory elements, and effects of microglial phagocytic signaling in epilepsy. Additionally, we highlight major knowledge gaps, particularly regarding the spatiotemporal dynamics and regulatory mechanisms of microglial phagocytosis following seizures.

Figure 1: Seizure induced cellular injury activates microglia phagocytosis which in turn shapes tissue reorganization and behaviors.

Figure 1:

Seizure induced cellular injury is associated with release of ‘find me’ signals and pro-inflammatory cytokines, as well as display of ‘eat me’ signals by apoptotic and stressed neurons. The ‘find me’ and ‘eat me’ signals are recognized by microglial phagocytic receptors whose downstream signalling initiates cytoskeletal modifications necessary for approaching and engulfing the target. These targets include dead and injured neurons, synapses, and aberrant newborn neurons which in turn affects clinically significant behavioral outcomes.

A wide array of phagocytic receptors regulates microglial responses to seizures

Seizures lead to extensive neuronal damage and death through a variety of mechanisms (Dingledine et al., 2014). These include inflammatory modes such as necrosis, necroptosis, and pyroptosis and non-inflammatory modes like apoptosis, autophagy, and phagoptosis. Apoptotic cells are cleared by phagocytes including microglia through a process termed efferocytosis and is extensively reviewed elsewhere (Doran et al., 2019). However, microglia can also phagocytose stressed-but-viable neurons through phagoptosis where unlike in the case of apoptotic cell clearance, phagocytosis itself is the primary mode of cell death (Brown & Neher, 2014; Abiega et al., 2016; Marchi & Brewster, 2024). It occurs following reversible exposure of eat me signals like phosphatidylserine (PS) such that blocking any part of PS signaling cascade rescues a substantial proportion of neurons from cell death (Neher et al., 2011; Fricker et al., 2012; Bosco et al., 2025). Thus, microglia contribute to seizure-induced neuronal loss through both efferocytosis and phagoptosis.

The general mechanisms of microglia phagocytosis involve detection of apoptotic and damaged cells via a variety of find me signals, engulfment via eat me signals, intracellular signaling, phagolysosome formation and maturation (Figure 2). Engagement of phagocytic receptors lead to the activation of intracellular tyrosine kinases and cytoskeletal rearrangement. The specific mechanisms downstream of each receptor are not fully understood (Medina & Ravichandran, 2016). Additionally, phagocytic receptors also participate in signaling which influences microglia proliferation and the production or suppression of pro-inflammatory NFκB related cytokines (Wyatt-Johnson & Brewster, 2019). Below we discuss several of these phagocytic receptors, ligands, and their signaling mechanisms.

Figure 2: Ligand-receptor interactions regulating microglial phagocytosis in epilepsy.

Figure 2:

Following a seizure, a variety of ‘find me’ and ‘eat me’ signals promote microglia phagocytosis of dead and stressed neurons, while ‘don’t eat me’ signals protect healthy cells. This wide array of receptor-ligand interactions facilitates fine tuning of microglia response based on the substrate type and extracellular milieu, as well as in coordinating phagocytic response with other signaling pathways such as inflammatory cytokine release and proliferation. Purines (ATP, UDP, UTP), S1P, and sCX3CR1 are common find me signals which bind to a variety of GPCRs (G protein-coupled receptors). Similarly, phosphatidylserine (PS) and activated complement components are common ‘eat me’ signals. Receptors like BAI1, TIM4, MerTK, and TREM2 interact with PS either directly or via bridging molecules like ProtS, Gas6, tubby, and TULP.

Nucleotides and purinergic signaling

ATP, UTP, and UDP are amongst the best known find me signals in the nucleotide category (Figure 2) (Koizumi et al., 2007; Elliott et al., 2009). During apoptosis, ATP and UDP are released via a variety of mechanisms including through Pannexin 1 (PANX1) channels activated by action of caspases (Elliott et al., 2009; Chekeni et al., 2010), lysosomal or vesicular secretion (Martins et al., 2013), passive release from necrotic cells, or via other channels such as connexins (Dosch et al., 2018). P2Y6 and P2Y2 receptors respond to these nucleotide signals and facilitate removal of apoptotic cells (Hide et al., 2023; Umpierre et al., 2024). UDP-P2Y6 signaling has been studied in epilepsy and known to affect neuronal phagocytosis by modulating microglial calcium activity, promoting lysosome biogenesis (calcium dependent) and release of NFκB associated cytokines (calcium independent) (Koizumi et al., 2007; Umpierre et al., 2024). There is also some evidence supporting a role for the microglial P2Y12 receptor in phagocytosis (Eyo et al., 2014; Inouye, 2018; Diaz-Aparicio et al., 2020; Chidambaram et al., 2022). P2Y12 is a Gi coupled GPCR, and we recently showed that prolonged activation of Gi signaling via chemogenetic manipulation arrested microglia in a homeostatic state and reduced phagocytosis of neurons post seizures (Dheer et al., 2024).

Phosphatidylserine, MerTK, and TREM2 signaling

Phosphatidylserine (PS) is an asymmetrically distributed membrane phospholipid maintained in the inner leaflet of the membrane through the action of flippase. During apoptosis, caspases activate ‘scramblase’ which exposes PS on the outer membrane (Vorselen, 2022). This exposed PS is bound directly by microglial receptors like BAI4 and TIM-4 (Vorselen, 2022), providing an initial tether followed by binding of soluble bridging molecules with their receptors to complete engulfment (Fricker et al., 2012; Vorselen, 2022). Protein S, Gas6, tubby, and tubby-like protein 1 are such bridging molecules mediating interaction between PS and the TAM family of receptors (TYRO3, Axl, and Mer receptor tyrosine kinase or MerTK) on microglia (Figure 2) (Shafit-Zagardo et al., 2018).

MerTK and Axl activation leads to autophosphorylation and downstream activation of phospholipase C gamma (PLC-γ), phosphatidylinositol 3-kinase (PI3K), and subsequent activation of Rac GTPase (Yadav et al., 2025). This activates cytoskeletal elements leading to pseudopod extension and formation of phagocytic cup (Botelho & Grinstein, 2011). MerTK plays a role in both tethering and engulfment of apoptotic cells (Dransfield et al., 2015). Additionally, both MerTK and Axl signaling suppresses the pro-inflammatory NFκB signaling (Wu et al., 2018; Vago et al., 2021) and promotes resolution of inflammation (Cai et al., 2018). Microglial loss of TAM receptors was associated with deficient phagocytosis of excitatory synapses and newborn dentate gyrus neurons and an increased incidence of seizure-related mortality in mouse model of Alzheimer’s disease (Huang & Lemke, 2022).

TREM2 is another important PS receptor (Bosco et al., 2025). Mutations in TREM2 and its adaptor protein TYROBP have been identified as the causes of Nasu-Hakola disease, a neurodegenerative disease commonly presenting with seizures (Paloneva et al., 2000; Paloneva et al., 2001; Paloneva et al., 2002). PI3K, SYK, and PLC-γ mediate phagocytic signaling downstream of TREM2 (Yao et al., 2019). Like MerTK, TREM2 activation also suppresses pro-inflammatory NFκB signaling (Li et al., 2019; Yao et al., 2019). TREM2 deficiency was associated with decreased neuron loss and increased spontaneous seizures in mice (Bosco et al., 2025). Furthermore, TREM2 promotes microglial proliferation (Wang et al., 2020; Bosco et al., 2025) which is essential to meet the increased phagocytic demand in the aftermath of seizure induced neuronal damage.

Complement signaling

Levels of complement components C1q and C3 are increased in both rodent models of epilepsy as well as epileptic patients including those with drug resistant seizures (Wei et al., 2021; Gruber et al., 2022). C1q and its downstream component activated C3 (C3b and iC3b) opsonize immature CNS synapses during a discrete window of postnatal development (Stevens et al., 2021). These tagged synapses are eliminated through C3b mediated activation of microglial complement receptor 3 (CR3; Figure 2). Spleen tyrosine kinase (Syk) and Rho GTPase mediate the downstream signaling from the complement receptor 3 with subsequent engagement of cytoskeletal elements (Uribe-Querol & Rosales, 2020). Deficiency of complement components C1q and C3 is associated with increased seizures in both rodents and humans secondary to deficits in synaptic phagocytosis (Chu et al., 2010; Libbey et al., 2010; Schaarenburg et al., 2016).

Fractalkine (CX3CL1-CX3CR1) signaling

CX3CL1 is the only known member of the CX3C chemokine family (Zhao et al., 2023). It exists in two forms- membrane bound and soluble (sCX3CR1), the latter forming from cleavage of membrane bound form by metalloproteinases ADAM10 and ADAM17 (Figure 2) (Zhao et al., 2023). It signals via the CX3CR1 receptor that is expressed widely by immune cells including microglia (Sokolowski et al., 2014; Zhao et al., 2023). This signaling is multifaceted such that both membrane bound and soluble CX3CL1 can act as a chemoattractant, membrane bound CX3CL1 can additionally function as an adhesion molecule, and the activation of CX3CR1 receptor raises microglial calcium and promotes chemotaxis (Harrison et al., 1998; Zhao et al., 2023). Increased expression of CX3CL1 and its association with neuronal loss has been observed in the brains of epileptic patients and rodent models (Xu et al., 2012). Blocking either CX3CL1 or its microglial receptor CX3CR1 was found to reduce hippocampal neuronal loss after status epilepticus in rodent models (Yeo et al., 2011; Ali et al., 2015 Feb), highlighting the significance of fractalkine signaling in microglia-neuron interaction in epilepsy.

Sphingosine-1-phosphate (S1P) signaling

Apoptotic cells have been shown to upregulate sphingosine kinase 1 (SphK1) which in turn increases S1P secretion (Xiao et al., 2023). S1P acts as a potent chemoattractant for macrophages, monocytes, and microglia (Zahiri et al., 2021). While microglia can detect S1P via S1P receptors, a recent study identified that S1P can also influence phagocytosis by binding to and activating the microglial TREM2 (Figure 2) (Xue et al., 2021). S1P signaling plays important roles in many epilepsy related mechanisms including neuronal apoptosis, blood-brain barrier stability, and inflammation (Wang et al., 2024). While there is some evidence for a therapeutic role of S1P immunomodulators in reducing microglial activation and inflammatory responses in epilepsy (Leo et al., 2017), direct investigation of microglia specific S1P signaling is lacking.

Other find me and eat me signals

Lysophosphatidylcholine (LPC) and its receptor G2A are other important receptor/ ligand systems involved in ‘find me’ signaling (Lauber et al., 2003; Peter et al., 2008; Inose et al., 2015). Similarly, calreticulin is upregulated on the surface of apoptotic cells and recognized by the LDL-receptor related protein 1 (LRP1) receptor with their interaction modulated by the ‘don’t eat me’ signal CD47 (Gardai et al., 2005). Blocking LRP1 signaling in microglia has been shown to reduce neuroinflammation in neuropathic pain (Brifault et al., 2019). In the context of epilepsy, LRP1 expression has been found to be increased in epileptic foci in multiple cell types including microglia (Wang et al., 2021). However, there are currently no studies that have examined either LPC-G2A or calreticulin-LRP1 signaling in epilepsy is any detail.

Don’t eat me signals protect healthy cells from phagocytosis

Don’t eat me signals serve to protect healthy cells through negative regulation of phagocytosis. Currently known ‘don’t eat me signals’ include- CD47, programmed cell death ligand 1 (PDL-1), beta-2-microglobulin, and CD24 (Gardai et al., 2005; Barkal et al., 2019; Khalaji et al., 2023). Neuronal CD47 and its microglial receptor SIRPα have been shown to be involved in synaptic pruning (Lehrman et al., 2018). However, there is paucity of direct evidence for a role of ‘don’t eat me’ signals in epileptogenesis and post-seizure neuronal loss (Figure 2).

Lysosomal function and inflammatory cytokine release is closely linked with phagocytic signaling

Lysosomal function

Factors that affect microglial lysosomal function and their fusion with phagosomes also influence phagocytic and autophagic activity. For example, presenelin-1 (PSN1) gene mutations are the most common cause of autosomal dominant Alzheimer’s disease with 20% patients also presenting with epilepsy (Larner, 2011). Mechanistically, PSN1 gene is involved in regulating lysosomal acidification and its fusion with autophagosome (Quick et al., 2023). Similarly, progranulin is essential for maintaining lysosomal acidification (Tanaka et al., 2017) and its loss-of-function mutations are associated with neuronal ceroid lipofuscinoses type 11 which presents with neurodegeneration and refractory seizures in children (Nóbrega et al., 2025). Recently, P2Y6 signaling was also shown to regulate lysosomal biogenesis in the context of seizures (Umpierre et al., 2024). Lastly, microglia phagocytosis maybe an important mechanism in ATP6V1A encephalopathy where over 80% of patients present with seizures (Guerrini et al., 2022). Interestingly, a recent study showed that microglial glycoprotein non-metastatic melanoma B (GPNMB) is upregulated in seizures and interacts with ATP6V1A to facilitate phagocytosis. Deficiency of GPNMB was associated with phagocytic defects and worsened seizures (Liu et al., 2025). Thus, proteins regulating lysosomal function serve as important links between phagocytosis and genetically predisposed epilepsy development.

NFκB signaling

NFκB signaling complex controls multiple aspects of innate and adaptive immunity by regulating inflammatory responses (Liu et al., 2017b). Phagocytic receptors that respond to damage associated molecular patterns (DAMPS) or pathogen associated molecular patterns (PAMPS) upregulate NFκB mediated pro-inflammatory signaling (Figure 1). These include P2Y6, toll like receptors (TLRs), complement receptor 3 (CR3), and Fcγ receptors (Cao et al., 2010; Moretti & Blander, 2014; Matsuda et al., 2015; Reis et al., 2019; Umpierre et al., 2024). Conversely, the phagocytic receptors that respond to non-DAMP, non-PAMP signals and maintain homeostatic clearance functions of microglia downregulate NFκB associated pro-inflammatory cytokine release (Figure 1). These include MerTK, TREM2/ DAP12, and Axl receptors (Hasanbasic et al., 2004; Tibrewal et al., 2008; Yao et al., 2019). NFκB signaling is thus an important effector determining the pro- or non-inflammatory nature of phagocytic signaling. Downregulating NFκB signaling has been shown to be beneficial in epilepsy due to reduced pro-inflammatory responses (Cai & Lin, 2022). In macrophages, blocking NFκB has been shown to reduce phagocytic responses to pathogens (Zou & Shankar, 2015; Liu et al., 2017a). However, whether the same holds true in epilepsy remains scantly studied (Wu et al., 2022).

Epigenetic regulators and transcription factors provide high level regulation of phagocytic machinery

Epigenetic regulators: PRC2, Kdm6a, and Kdm6b

Regional heterogeneity in the expression of phagocytic machinery and phagocytic ability of microglia is well established (Biase et al., 2017; Ayata et al., 2018). This heterogeneity is maintained by epigenetic regulators such as the Polycomb repressive complex 2 (PRC2) and functions in matching the microglial phagocytic activity with the burden of apoptotic cell clearance under homeostatic conditions (Ayata et al., 2018). For example, cerebellar microglia have higher phagocytic activity than striatal microglia, in line with higher apoptotic cell burden in cerebellum (Ayata et al., 2018). PRC2 functions by silencing phagocytosis related genes by methylating histone H3 at lysine 27 (H3K27me3). Whereas in cerebellum, induction of histone demethylases Kdm6a and Kdm6b relieve this suppression (Ayata et al., 2018). Disruption of this spatially heterogenous phagocytic activity by suppression of PRC2 was associated with increased phagocytic activity of striatal microglia, altered morphology of striatal medium spiny neurons, behavioral abnormalities, and seizures (Figure 3) (Ayata et al., 2018).

Figure 3: Aberrant phagocytosis contributes to epileptogenesis.

Figure 3:

Phagocytic effectors, transcription factors, and epigenetic regulators maintain homeostatic control and regional heterogeneity of microglia phagocytosis. Both excessive as well as insufficient phagocytic activity can lead to epileptogenic remodeling of neuronal circuits. For example, elevated mTOR and PU.1 activity enhance microglial phagocytosis while deficiency of TREM2 and complements reduce phagocytosis, both results in increased seizures.

Transcription factors: PU.1, p53, mTOR, PPARs, and Nrf2

PU.1:

PU.1 (encoded by gene SPI1) is an essential transcription factor in microgliogenesis and defining microglia identity from erythromyeloid progenitors (Kierdorf et al., 2013). Silencing PU.1 downregulates phagocytic receptors and signaling partners like TREM2 and TYROBP/ DAP12, and reduces microglia phagocytosis, indicating its continued role in this process beyond development (Smith et al., 2013; Huang et al., 2017; Rustenhoven et al., 2018). Upregulation of SPI1/PU.1 is seen in tuberous sclerosis complex and focal cortical dysplasia which present with epilepsy (Figure 3) (Zimmer et al., 2021). There is some evidence to indicate that dysregulated phagocytosis may be involved in the pathogenesis of seizures in these cortical malformations (Zhao et al., 2018).

p53 tumor suppressor gene:

p53 is a tumor-suppressor gene and transcriptional regulator of cell cycle progression, DNA damage response, and apoptosis (Kruiswijk et al., 2015). Its activity is increased in microglia exposed to oxidative stress and DNA damage (Holtman et al., 2017). Knocking out or suppressing p53 in microglia has been shown to reduce pro-inflammatory cytokine release and increase phagocytic ability (Jayadev et al., 2011). Its effect on phagocytosis is mediated via the downstream regulation of Death Domain 1α (DD1α) receptor that engages in homophilic intermolecular interaction between apoptotic cell and macrophages (Yoon et al., 2015). Further, DD1α also promotes immune tolerance to apoptotic cell antigens through inhibitory co-stimulation of T cells (Yoon et al., 2015). It was observed that p53 general knockout mice experience more severe seizures with along with short but not long term neuroprotection in a kainate induced model of epilepsy (Engel et al., 2010). It is possible that the intricate balance between pro-inflammatory and phagocytic signaling, regulated by microglial p53, may influence these findings. Microglia-specific p53 deletion is needed to further elucidate its specific contributions.

mTOR:

mechanistic Target of Rapamycin (mTOR) signaling regulates fundamental cellular processes like growth, metabolism, and autophagy (Saxton & Sabatini, 2017). Selectively elevating mTOR signaling in microglia was associated with increased proliferation and phagocytic activity as well as development of spontaneous seizures, without a significant increase in pro-inflammatory cytokines (Figure 3) (Zhao et al., 2018). RNA-seq analysis revealed significant changes in many genes directly or indirectly involved in phagocytosis, thus highlighting that aberrant microglia phagocytosis may contribute to epileptogenesis (Zhao et al., 2018). However, microglia specific deletion of mTOR is also associated with worsened outcomes including increased neuronal loss and more severe spontaneous recurrent seizures, along with reduced proliferative responses to injury. (Zhao et al., 2020). These observations suggest that an optimal level of microglial mTOR activity is essential for a balanced response to seizure induced neuronal injury.

PPAR:

Peroxisome proliferator-activated receptors (PPARs) are a family of nuclear receptor transcription factors that function as lipid sensors and are involved in fatty acid metabolism (Tyagi et al., 2011). PPARγ agonists were found to increase amyloid-β phagocytosis through upregulation of phagocytic receptors Axl, MerTK, TREM2, and CD36 (Yamanaka et al., 2012; Savage et al., 2015). In the context of epilepsy, PPARγ agonists were found to rescue neuron loss, suppress the development of spontaneous recurrent seizures, reduce microglial activation, and inflammation by modulating the BDNF/TrkB signaling (Hong et al., 2013; Peng et al., 2019). However, the contribution of phagocytosis in these effects needs further evaluation.

Nrf2:

Nuclear factor erythroid 2-related factor 2 (Nrf2) is a master regulator of antioxidant response to oxidative stress and a proposed therapeutic target in epilepsy (Carmona-Aparicio et al., 2015). Activation of Nrf2 upregulates antioxidative defense and phagocytic ability of microglia, the latter via up-regulation of scavenger receptor CD36 (Zhao et al., 2015).

Phagocytosis shapes tissue reorganization after seizures

Post seizure pathology comprises widespread changes in tissue organization including loss of neurons and synapses, aberrant neurogenesis, and glial hypertrophy. Below we discuss the changes shaped by the phagocytic activity of microglia.

Neuronal loss

Epilepsy is associated with neuronal loss in multiple brain regions including hippocampus, amygdala, and entorhinal cortex . While some of the cell loss may be a direct consequence of seizure (Dingledine et al., 2014), microglial phagocytosis also plays an important role (Figure 1). This is evident from the neuroprotection observed when microglial phagocytic ability is compromised (Yeo et al., 2011; Umpierre et al., 2024; Bosco et al., 2025). For example, knocking out phagocytic receptors TREM2, P2Y6, C3R, or CX3CR1 as well as ‘eat me’ signals like complement 3 is associated with reduced neuronal loss after seizures (Yeo et al., 2011; Wei et al., 2021; Schartz et al., 2023; Umpierre et al., 2024; Bosco et al., 2025).

The mechanism of this neuroprotection likely involves a combination of reduced phagoptosis (Brown & Neher, 2014) and downregulation of pro-inflammatory NFκB associated cytokines downstream of phagocytic signaling (Umpierre et al., 2024). However, whether the stressed neurons that did not undergo phagoptosis function normally is unclear. In a kainate induced mice model of epilepsy, TREM2 knock out was associated with reduced phagocytic clearance of neurons as well as increased risk of spontaneous seizures (Bosco et al., 2025). The authors speculate that these ‘damaged neurons’ that survive may be pro-epileptogenic and contribute to the development of spontaneous recurrent seizures (Bosco et al., 2025). While others have suggested that neuronal death itself (Dudek et al., 2010) or the biochemical pathways causing programmed cell death may contribute to epileptogenesis (Dingledine et al., 2014).

Synapse reorganization

During brain development, microglia modulate synaptic connectivity in an activity dependent manner (Tremblay et al., 2010; Schafer et al., 2012; Nebeling et al., 2023). Although the mechanisms behind this, specially whether it involves a primarily microglia driven pruning of synapse has recently been debated (Pereira-Iglesias et al., 2024). Complement mediated signaling as described previously plays a key role in microglia-synapse interaction (Gasque, 2004; Stevens et al., 2007). Additionally, CX3CR1 (Paolicelli et al., 2011) and TREM2 (Filipello et al., 2018) signaling has also been shown to be important in microglial synaptic pruning. It has been observed that reduced microglial synaptic pruning during development such as in C1q knock out mice may predispose to recurrent seizures (Figure 3) (Chu et al., 2010; Ma et al., 2013). However, excessive synaptic pruning such as that observed with loss of Polycomb repressive complex 2 (PRC2), an epigenetic regulator of phagocytosis (Ayata et al., 2018) or with increased levels of C1q or C3 deposition in adult brain (Hong et al., 2016; Gomez-Arboledas et al., 2021) can also predispose to increased seizures (Figure 3). Conversely, a recent study showed that blocking C3 mediated inflammation and synaptic phagocytosis with captopril reduces risk of spontaneous seizures and cognitive deficits (Dong et al., 2022). Further, in the hours to days following seizures, microglia were shown to facilitate structural resolution of injured, beaded dendrites through direct physical interactions with their process pouches (Eyo et al., 2021). This may also play a role in synapse reorganization.

Aberrant neurogenesis

Aberrant neurogenesis is increased in the dentate gyrus following seizures (Victor & Tsirka, 2020; Lybrand et al., 2021). Non-inflammatory, homeostatic microglia keep excess neurogenesis in check through apoptosis coupled phagocytosis (Sierra et al., 2014). Further, following inflammatory challenge, microglia use various strategies to boost their phagocytic capacity to efficiently clear increased apoptotic cell load (Abiega et al., 2016). However, this apoptosis-phagocytosis coupling is lost in both patients as well as rodent model of mesial temporal lobe epilepsy (MTLE). These microglia show reduced surveillance and expression of apoptotic cell recognition receptors with resulting accumulation of apoptotic newborn neurons in the neurogenic niche (Abiega et al., 2016). However, microglial phagoptosis of viable newborn cells is increased around the same time, indicating a compensatory mechanism, specially keeping ectopic granule cells in check (Abiega et al., 2016; Luo et al., 2016; Eyo et al., 2017). It has been shown that aberrant hippocampal neurogenesis contributes to epileptogenesis and its associated cognitive decline (Cho et al., 2015; Lybrand et al., 2021). Interestingly, microglial ablation or P2Y12 knockout was found to be associated with reduced abberent neurogenesis and immature neuronal projections following seizures (Mo et al., 2019). Similarly, deficiency of either P2Y12 or MerTK/Axl phagocytic receptors was found to disrupt basal neurogenesis (Diaz-Aparicio et al., 2020). This could be due to disruption of the beneficial roles played by microglia in the proliferation and integration of neural progenitor cells, including through their phagocytic secretome (Matsuda et al., 2015; Sandvig et al., 2018; Diaz-Aparicio et al., 2020).

Microglia phagocytosis modulates epilepsy related behavioral outcomes

Cognitive function

Seizures are commonly associated with cognitive decline and behavioral abnormalities in both epilepsy patients and rodent models (Figure 1) (Elger et al., 2004; Novak et al., 2022). These abnormalities include deficits in memory and attention, low IQ, and poor scholastic performance (Lenck-Santini & Scott, 2015). The mechanisms of these deficits are multifactorial, involving a complex interplay between seizure etiology, the seizures themselves, interictal discharges, and anti-epileptic medications (Lenck-Santini & Scott, 2015). Neuronal loss in hippocampus is one such mechanism. Loss of hippocampal volume and neurons is seen in both patients as well as animal models of temporal lobe epilepsy and correlates with cognitive decline and psychiatric comorbidity (Cascino, 2003; Fuerst et al., 2003; Umpierre et al., 2024; Bosco et al., 2025). Reducing hippocampal neuronal loss by attenuating microglial phagocytosis and pro-inflammatory cytokine production is associated with improved cognitive performance (Tian et al., 2017; Schartz et al., 2023; Umpierre et al., 2024).

Thus, microglial phagocytic machinery maybe an attractive target for therapeutic interventions for improving cognition in epilepsy. However, a better understanding of the spatio-temporal dynamics of phagocytosis following acute seizures is required to selectively reduce detrimental effects and identify neuronal substrates underlying various cognitive and behavioral abnormalities. Also, the network effect of modulating phagocytosis, i.e. how it affects the interaction between different brain regions necessary for normal cognition has not been studied. Loss of homeostatic microglial function, such as regulating neuronal excitability (Badimon et al., 2020; Wu et al., 2020) following seizures may also be involved in cognitive deficits and need exploration.

Spontaneous recurrent seizures (SRS)

We have shown that microglia depletion increases the risk of SRS in mice (Wu et al., 2020). More recently we demonstrated that this increase in SRS is recapitulated in mice with TREM2 knockout associated phagocytic deficits (Bosco et al., 2025). The neuronal substrate that mediates the increased risk of seizures following status epilepticus and spared by microglial phagocytic defect is not well understood. However, aberrant neurogenesis in the dentate gyrus, ectopic granule cells, and immature neuronal projections after seizures are promising candidates.

While we have extensively discussed the effect of seizures on inducing phagocytosis, it is important to note that aberrant phagocytosis may itself be epileptogenic (Figure 3). C1q or C3 deficiency is associated with failure of synaptic pruning, enhanced excitatory synaptic connectivity and increased seizures in mice (Chu et al., 2010; Libbey et al., 2010; Ma et al., 2013). Seizures were also the most frequent neuropsychiatric symptoms reported in C1q deficient patients diagnosed with systemic lupus erythematosus (SLE) (Schaarenburg et al., 2016). Similarly, an abnormal increase in microglia phagocytosis can also increase seizure risk. Enhanced microglial mTOR signaling was associated with loss of cortical and hippocampal synapses, development of spontaneous recurrent seizures, and mortality (Zhao et al., 2018). Increased mTOR signaling is epileptogenic and observed in patients with tuberous sclerosis (Crino, 2016).

Lastly, it has been suggested that microglia may preferentially prune inhibitory synapses contributing to the excitation/ inhibition imbalance and epileptogenesis (Andoh et al., 2019; Fan et al., 2023). Interestingly, a recent study found that PS exposure may play a role in this. In mice with acute neuronal deletion of a flippase chaperone, a preferential exposure of PS in neuronal soma and a microglia MerTK dependent loss of inhibitory post-synapses were observed along with increased excitability and seizures (Park et al., 2021).

Concluding remarks and future directions

Over the past two decades, numerous groundbreaking discoveries have deepened our understanding of phagocytosis in epileptogenesis, post-seizure pathology, and the effects of seizures on phagocytic signaling (Wyatt-Johnson & Brewster, 2019). These advances include the identification of complementary ‘find me’ and ‘eat me’ signaling pathways, which facilitate the efficient clearance of damaged and stressed neurons (Medina & Ravichandran, 2016; Galloway et al., 2019; Wyatt-Johnson & Brewster, 2019). In parallel, a diverse array of phagocytic receptors (Figure 2) enables microglia to fine-tune their responses by recognizing DAMPS and apoptotic cells, leading to either the stimulation or suppression of pro-inflammatory cytokine release (Galloway et al., 2019; Wyatt-Johnson & Brewster, 2019). These responses are orchestrated by epigenetic regulators and transcription factors. However, significant gaps remain in our knowledge. The mechanisms underlying the dynamic switching between pro- and anti-inflammatory cytokine release downstream of phagocytic signaling are poorly understood (Paolicelli et al., 2022). Similarly, the role of ‘don’t eat me’ signals in epilepsy remains largely unexplored (Medina & Ravichandran, 2016). Furthermore, the interplay between phagocytic signaling and other pathways, such as those regulating proliferation and metabolism, which are concurrently altered in epilepsy, is yet to be fully elucidated.

Seizures exert a complex influence on microglial phagocytosis. Increased apoptotic and necrotic cell burden drives microglia to adopt compensatory mechanisms to meet heightened phagocytic demands (Abiega et al., 2016). However, in cases of recurrent or severe seizures, a prolonged pro-inflammatory environment can impair phagocytic capacity by downregulating phagocytic receptors (Cherry et al., 2014). This phenomenon underscores the growing interest in anti-inflammatory therapies for managing seizures and epilepsy, some of which target phagocytosis pathways (Dey et al., 2016; Dong et al., 2022).

Following status epilepticus, microglial phagocytosis has been implicated in cognitive deficits (Schartz et al., 2023; Umpierre et al., 2024). Current rodent studies predominantly focus on hippocampal-dependent memory assessments (Antunes & Biala, 2011). However, a more comprehensive evaluation is needed to characterize additional cognitive and psychological abnormalities, identify their underlying neuronal substrates, and investigate the role of microglial phagocytosis in shaping these behavioral outcomes.

Interestingly, microglial phagocytosis plays a pivotal role in modulating the risk of spontaneous recurrent seizures following status epilepticus. Emerging evidence suggests that aberrant neurogenesis in the dentate gyrus may serve as the primary substrate for this effect. Microglia exhibit a complex relationship with newborn neurons in the neurogenic niche: while they regulate aberrant neurogenesis by phagocytosing both apoptotic and viable newborn neurons, they also promote neurogenesis and support the growth of immature neuronal projections (Sierra et al., 2010; Abiega et al., 2016; Mo et al., 2019). The mechanisms through which microglia drive aberrant neurogenesis, however, remain poorly understood. Additionally, microglia contribute to epileptogenesis by mediating abnormal synaptic phagocytosis during synaptic development. Dysregulated microglial activity, whether excessive or insufficient, can have widespread effects on the central nervous system, as observed in conditions such as tuberous sclerosis or C1q deficiency-related seizures in systemic lupus erythematosus (Schaarenburg et al., 2016; Zhao et al., 2018). These insights underscore the potential of targeting microglial phagocytosis as a therapeutic strategy for such disorders.

Reconciling whether microglial phagocytosis is beneficial, or detrimental following status epilepticus may ultimately depend on contexts. Temporarily restricting phagocytosis could be advantageous, as it might enhance cognitive function without significantly increasing the risk of future seizures. This hypothesis could be tested in rodent models by attenuating microglial phagocytosis at various time points after an initial status epilepticus. However, prolonged attenuation of phagocytosis might elevate the risk of subsequent seizures, leading to cumulative cognitive decline over time. Another way to separate the beneficial from detrimental effect maybe by spatially restricting the attenuation of phagocytosis to the select brain regions. For instance, pharmacological agents that reduce phagocytosis could be delivered directly to the hippocampus following status epilepticus in rodent models. Subsequent assessments could evaluate both cognitive outcomes and the risk of developing spontaneous recurrent seizures (SRS). If these interventions show promise, future approaches could explore the use of small molecules targeting epigenetic regulators (Cheng et al., 2019; Singh et al., 2023) to modulate microglial phagocytosis. This strategy could enable precise, spatially restricted attenuation or enhancement of phagocytosis, optimizing therapeutic outcomes.

Acknowledgements

We thank Wu lab members at Mayo Clinic and UTHealth Houston for the insightful discussions.

Funding

This work was supported by the National Institutes of Health: R35NS132326 (L.-J.W.) and R01NS088627 (L.-J.W).

Footnotes

Competing interests

The authors declare no competing interests in relation to this work.

References

  1. Abiega O, Beccari S, Diaz-Aparicio I, Nadjar A, Layé S, Leyrolle Q, Gómez-Nicola D, Domercq M, Pérez-Samartín A, Sánchez-Zafra V, Paris I, Valero J, Savage JC, Hui C-W, Tremblay M-È, Deudero JJP, Brewster AL, Anderson AE, Zaldumbide L, Galbarriatu L, Marinas A, Vivanco Md, Matute C, Maletic-Savatic M, Encinas JM & Sierra A. (2016). Neuronal Hyperactivity Disturbs ATP Microgradients, Impairs Microglial Motility, and Reduces Phagocytic Receptor Expression Triggering Apoptosis/Microglial Phagocytosis Uncoupling. PLoS Biology 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Ali I, Chugh D & Ekdahl C. (2015. Feb). Role of fractalkine-CX3CR1 pathway in seizure-induced microglial activation, neurodegeneration, and neuroblast production in the adult rat brain. Neurobiology of disease 74. [DOI] [PubMed] [Google Scholar]
  3. Andoh M, Ikegaya Y & Koyama R. (2019). Synaptic Pruning by Microglia in Epilepsy. Journal of Clinical Medicine 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Antunes M & Biala G. (2011). The novel object recognition memory: neurobiology, test procedure, and its modifications. Cognitive Processing 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Ayata P, Badimon A, Strasburger HJ, Duff MK, Montgomery SE, Loh Y- HE, Ebert A, Pimenova AA, Ramirez BR, Chan AT, Sullivan JM, Purushothaman I, Scarpa JR, Goate AM, Busslinger M, Shen L, Losic B & Schaefer A. (2018). Epigenetic regulation of brain region-specific microglia clearance activity. Nature neuroscience 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Badimon A, Strasburger HJ, Ayata P, Chen X, Nair A, Ikegami A, Hwang P, Chan AT, Graves SM, Uweru JO, Ledderose C, Kutlu MG, Wheeler MA, Kahan A, Ishikawa M, Wang Y-C, Loh Y- HE, Jiang JX, Surmeier DJ, Robson SC, Junger WG, Sebra R, Calipari ES, Kenny PJ, Eyo UB, Colonna M, Quintana FJ, Wake H, Gradinaru V, Schaefer A, Badimon A, Strasburger HJ, Ayata P, Chen X, Nair A, Ikegami A, Hwang P, Chan AT, Graves SM, Uweru JO, Ledderose C, Kutlu MG, Wheeler MA, Kahan A, Ishikawa M, Wang Y-C, Loh Y- HE, Jiang JX, Surmeier DJ, Robson SC, Junger WG, Sebra R, Calipari ES, Kenny PJ, Eyo UB, Colonna M, Quintana FJ, Wake H, Gradinaru V & Schaefer A. (2020). Negative feedback control of neuronal activity by microglia. Nature 2020 586:7829 586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Barkal A, Brewer R, Markovic M, Kowarsky M, Barkal S, Zaro B, Krishnan V, Hatakeyama J, Dorigo O, Barkal L & Weissman I. (2019). CD24 signalling through macrophage Siglec-10 is a target for cancer immunotherapy - PubMed. Nature 572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bazhanova ED, Kozlov AA, Litovchenko AV, Bazhanova ED, Kozlov AA & Litovchenko AV. (2021). Mechanisms of Drug Resistance in the Pathogenesis of Epilepsy: Role of Neuroinflammation. A Literature Review. Brain Sciences 2021, Vol 11, Page 663 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Beghi E, Giussani G, Nichols E, Abd-Allah F, Abdela J, Abdelalim A, Abraha HN, Adib MG, Agrawal S, Alahdab F, Awasthi A, Ayele Y, Barboza MA, Belachew AB, Biadgo B, Bijani A, Bitew H, Carvalho F, Chaiah Y, Daryani A, Do HP, Dubey M, Endries AYY, Eskandarieh S, Faro A, Farzadfar F, Fereshtehnejad S-M, Fernandes E, Fijabi DO, Filip I, Fischer F, Gebre AK, Tsadik AG, Gebremichael TG, Gezae KE, Ghasemi-Kasman M, Weldegwergs KG, Degefa MG, Gnedovskaya EV, Hagos TB, Haj-Mirzaian A, Haj-Mirzaian A, Hassen HY, Hay SI, Jakovljevic M, Kasaeian A, Kassa TD, Khader YS, Khalil I, Khan EA, Khubchandani J, Kisa A, Krohn KJ, Kulkarni C, Nirayo YL, Mackay MT, Majdan M, Majeed A, Manhertz T, Mehndiratta MM, Mekonen T, Meles HG, Mengistu G, Mohammed S, Naghavi M, Mokdad AH, Mustafa G, Irvani SSN, Nguyen LH, Nixon MR, Ogbo FA, Olagunju AT, Olagunju TO, Owolabi MO, Phillips MR, Pinilla-Monsalve GD, Qorbani M, Radfar A, Rafay A, Rahimi-Movaghar V, Reinig N, Sachdev PS, Safari H, Safari S, Safiri S, Sahraian MA, Samy AM, Sarvi S, Sawhney M, Shaikh MA, Sharif M, Singh G, Smith M, Szoeke CEI, Tabarés-Seisdedos R, Temsah M-H, Temsah O, Tortajada-Girbés M, Tran BX, Tsegay AAT, Ullah I, Venketasubramanian N, Westerman R, Winkler AS, Yimer EM, Yonemoto N, Feigin VL, Vos T & Murray CJL. (2019). Global, regional, and national burden of epilepsy, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. The Lancet Neurology 18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Biase LMD, Schuebel KE, Fusfeld ZH, Jair K, Hawes IA, Cimbro R, Zhang H-Y, Liu Q-R, Shen H, Xi Z-X, Goldman D & Bonci A. (2017). Local Cues Establish and Maintain Region-Specific Phenotypes of Basal Ganglia Microglia. Neuron 95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Bosco DB, Kremen V, Haruwaka K, Zhao S, Wang L, Ebner BA, Zheng J, Xie M, Dheer A, Perry JF, Barath A, Nguyen AT, Worrell GA & Wu L-J. (2025). Microglial TREM2 promotes phagocytic clearance of damaged neurons after status epilepticus. Brain, behavior, and immunity 123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Bosco DB, Zheng J, Xu Z, Peng J, Eyo UB, Tang K, Yan C, Huang J, Feng L, Wu G, Richardson JR, Wang H & Wu L-J. (2018). RNAseq analysis of hippocampal microglia after kainic acid-induced seizures. Molecular Brain 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Botelho R & Grinstein S. (2011). Phagocytosis. Current biology 21. [DOI] [PubMed] [Google Scholar]
  14. Brifault C, Kwon H, Campana WM & Gonias SL. (2019). LRP1 deficiency in microglia blocks neuro-inflammation in the spinal dorsal horn and neuropathic pain processing. Glia 67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Brown GC & Neher JJ. (2014). Microglial phagocytosis of live neurons. Nat Rev Neurosci 15, 209–216. [DOI] [PubMed] [Google Scholar]
  16. Cai B, Kasikara C, Doran A, Ramakrishnan R, Birge R & Tabas I. (2018). MerTK signaling in macrophages promotes the synthesis of inflammation resolution mediators by suppressing CaMKII activity - PubMed. Science signaling 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Cai M & Lin W. (2022). The Function of NF-Kappa B During Epilepsy, a Potential Therapeutic Target. Frontiers in Neuroscience 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Cao S, Theodore S, Standaert DG, Cao S, Theodore S & Standaert DG. (2010). Fcγ receptors are required for NF-κB signaling, microglial activation and dopaminergic neurodegeneration in an AAV-synuclein mouse model of Parkinson’s disease. Molecular Neurodegeneration 2010 5:1 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Carmona-Aparicio L, Pérez-Cruz C, Zavala-Tecuapetla C, Granados-Rojas L, Rivera-Espinosa L, Montesinos-Correa H, Hernández-Damián J, Pedraza-Chaverri J, Sampieri AI, Coballase-Urrutia E & Cárdenas-Rodríguez N. (2015). Overview of Nrf2 as Therapeutic Target in Epilepsy. International Journal of Molecular Sciences 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Cascino GD. (2003). Progressive Damage in Epilepsy. Epilepsy Currents 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Chekeni F, Elliott M, Sandilos J, Walk S, Kinchen J, Lazarowski E, Armstrong A, Penuela S, Laird D, Salvesen G, Isakson B, Bayliss D & Ravichandran K. (2010). Pannexin 1 channels mediate ‘find-me’ signal release and membrane permeability during apoptosis - PubMed. Nature 467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Cheng Y, He C, Wang M, Ma X, Mo F, Yang S, Han J, Wei X, Cheng Y, He C, Wang M, Ma X, Mo F, Yang S, Han J & Wei X. (2019). Targeting epigenetic regulators for cancer therapy: mechanisms and advances in clinical trials. Signal Transduction and Targeted Therapy 2019 4:1 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Cherry JD, Olschowka JA, O’Banion MK, Cherry JD, Olschowka JA & O’Banion MK. (2014). Neuroinflammation and M2 microglia: the good, the bad, and the inflamed. Journal of Neuroinflammation 2014 11:1 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Chidambaram H, Das R & Chinnathambi S. (2022). G-protein coupled purinergic P2Y12 receptor interacts and internalizes TauRD-mediated by membrane-associated actin cytoskeleton remodeling in microglia - PubMed. European journal of cell biology 101. [DOI] [PubMed] [Google Scholar]
  25. Cho K, Lybrand Z, Ito N, Brulet R, Tafacory F, Zhang L, Good L, Ure K, Kernie S, Birnbaum S, Scharfman H, Eisch A & Hsieh J. (2015). Aberrant hippocampal neurogenesis contributes to epilepsy and associated cognitive decline. Nature communications 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Chu Y, Jin X, Parada I, Pesic A, Stevens B, Barres B & Prince DA. (2010). Enhanced synaptic connectivity and epilepsy in C1q knockout mice. Proceedings of the National Academy of Sciences of the United States of America 107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Crino P (2016). The mTOR signalling cascade: paving new roads to cure neurological disease. Nature reviews Neurology 12. [DOI] [PubMed] [Google Scholar]
  28. Dey A, Kang X, Qiu J, Du Y & Jiang J. (2016). Anti-inflammatory small molecules to treat seizures and epilepsy: from bench to bedside. Trends in pharmacological sciences 37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Dheer A, Bosco DB, Zheng J, Wang L, Zhao S, Haruwaka K, Yi M-H, Barath A, Tian D-S & Wu L-J. (2024). Chemogenetic approaches reveal dual functions of microglia in seizures. Brain, Behavior, and Immunity 115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Diaz-Aparicio I, Paris I, Sierra-Torre V, Plaza-Zabala A, Rodríguez-Iglesias N, Márquez-Ropero M, Beccari S, Huguet P, Abiega O, Alberdi E, Matute C, Bernales I, Schulz A, Otrokocsi L, Sperlagh B, Happonen KE, Lemke G, Maletic-Savatic M, Valero J & Sierra A. (2020). Microglia Actively Remodel Adult Hippocampal Neurogenesis through the Phagocytosis Secretome. Journal of Neuroscience 40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Dingledine R, Varvel NH & Dudek FE. (2014). When and How Do Seizures Kill Neurons, and Is Cell Death Relevant to Epileptogenesis? Advances in experimental medicine and biology 813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Dong X, Fan J, Lin D, Wang X, Kuang H, Gong L, Chen C, Jiang J, Xia N, He D, Shen W, Jiang P, Kuang R, Zeng L & Xie Y. (2022). Captopril alleviates epilepsy and cognitive impairment by attenuation of C3-mediated inflammation and synaptic phagocytosis - PubMed. Journal of neuroinflammation 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Doran AC, Yurdagul A, Tabas I, Doran AC, Yurdagul A & Tabas I. (2019). Efferocytosis in health and disease. Nature Reviews Immunology 2019 20:4 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Dosch M, Gerber J, Jebbawi F, Beldi G, Dosch M, Gerber J, Jebbawi F & Beldi G. (2018). Mechanisms of ATP Release by Inflammatory Cells. International Journal of Molecular Sciences 2018, Vol 19, Page 1222 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Dransfield I, Zagórska A, Lew E, Michail K & Lemke G. (2015). Mer receptor tyrosine kinase mediates both tethering and phagocytosis of apoptotic cells - PubMed. Cell death & disease 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Dudek FE, Ekstrand JJ & Staley KJ. (2010). Is Neuronal Death Necessary for Acquired Epileptogenesis in the Immature Brain? Epilepsy Currents 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Elger CE, Helmstaedter C & Kurthen M. (2004). Chronic epilepsy and cognition. The Lancet Neurology 3. [DOI] [PubMed] [Google Scholar]
  38. Elliott M, Chekeni F, Trampont P, Lazarowski E, Kadl A, Walk S, Park D, Woodson R, Ostankovich M, Sharma P, Lysiak J, Harden T, Leitinger N & Ravichandran K. (2009). Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance - PubMed. Nature 461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Engel T, Tanaka K, Jimenez-Mateos EM, Caballero-Caballero A, Prehn JHM, Henshall DC, Engel T, Tanaka K, Jimenez-Mateos EM, Caballero-Caballero A, Prehn JHM & Henshall DC. (2010). Loss of p53 results in protracted electrographic seizures and development of an aggravated epileptic phenotype following status epilepticus. Cell Death & Disease 2010 1:10 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Eyo UB, Haruwaka K, Mo M, Campos-Salazar AB, Wang L, Speros XS, Sabu S, Xu P & Wu L-J. (2021). Microglia provide structural resolution to injured dendrites after severe seizures. Cell Reports 35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Eyo UB, Murugan M & Wu L-J. (2017). Microglia–Neuron Communication in Epilepsy. Glia 65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Eyo UB, Peng J, Swiatkowski P, Mukherjee A, Bispo A & Wu L-J. (2014). Neuronal Hyperactivity Recruits Microglial Processes via Neuronal NMDA Receptors and Microglial P2Y12 Receptors after Status Epilepticus. Journal of Neuroscience 34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Fan J, Dong X, Tang Y, Wang X, Lin D, Gong L, Chen C, Jiang J, Shen W, Xu A, Zhang X, Xie Y, Huang X & Zeng L. (2023). Preferential pruning of inhibitory synapses by microglia contributes to alteration of the balance between excitatory and inhibitory synapses in the hippocampus in temporal lobe epilepsy - PubMed. CNS neuroscience & therapeutics 29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Filipello F, Morini R, Corradini I, Zerbi V, Canzi A, Michalski B, Erreni M, Markicevic M, Starvaggi-Cucuzza C, Otero K, Piccio L, Cignarella F, Perrucci F, Tamborini M, Genua M, Rajendran L, Menna E, Vetrano S, Fahnestock M, Paolicelli R & Matteoli M. (2018). The Microglial Innate Immune Receptor TREM2 Is Required for Synapse Elimination and Normal Brain Connectivity - PubMed. Immunity 48. [DOI] [PubMed] [Google Scholar]
  45. Fricker M, Neher JJ, Zhao J-W, Théry C, Tolkovsky AM & Brown GC. (2012). MFG-E8 Mediates Primary Phagocytosis of Viable Neurons during Neuroinflammation. Journal of Neuroscience 32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Fuerst D, Shah J, Shah A & Watson C. (2003). Hippocampal sclerosis is a progressive disorder: A longitudinal volumetric MRI study. Annals of Neurology 53. [DOI] [PubMed] [Google Scholar]
  47. Galloway DA, Phillips AEM, Owen DRJ & Moore CS. (2019). Phagocytosis in the Brain: Homeostasis and Disease. Frontiers in Immunology 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Gardai S, McPhillips K, Frasch S, Janssen W, Starefeldt A, Murphy-Ullrich J, Bratton D, Oldenborg P, Michalak M & Henson P. (2005). Cell-surface calreticulin initiates clearance of viable or apoptotic cells through trans-activation of LRP on the phagocyte - PubMed. Cell 123. [DOI] [PubMed] [Google Scholar]
  49. Gasque P (2004). Complement: a unique innate immune sensor for danger signals. Molecular Immunology 41. [DOI] [PubMed] [Google Scholar]
  50. Gomez-Arboledas A, Acharya MM & Tenner AJ. (2021). The Role of Complement in Synaptic Pruning and Neurodegeneration. ImmunoTargets and Therapy 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Gruber V-E, Luinenburg MJ, Colleselli K, Endmayr V, Anink JJ, Zimmer TS, Jansen F, Gosselaar P, Coras R, Scholl T, Blumcke I, Pimentel J, Hainfellner JA, Höftberger R, Rössler K, Feucht M, Scheppingen Jv, Aronica E & Mühlebner A. (2022). Increased expression of complement components in tuberous sclerosis complex and focal cortical dysplasia type 2B brain lesions. Epilepsia 63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Guerrini R, Mei D, Kerti-Szigeti K, Pepe S, Koenig MK, Von Allmen G, Cho MT, McDonald K, Baker J, Bhambhani V, Powis Z, Rodan L, Nabbout R, Barcia G, Rosenfeld JA, Bacino CA, Mignot C, Power LH, Harris CJ, Marjanovic D, Møller RS, Hammer TB, Study TD, Keski Filppula R, Vieira P, Hildebrandt C, Sacharow S, Network UD, Maragliano L, Benfenati F, Lachlan K, Benneche A, Petit F, de Sainte Agathe JM, Hallinan B, Si Y, Wentzensen IM, Zou F, Narayanan V, Matsumoto N, Boncristiano A, la Marca G, Kato M, Anderson K, Barba C, Sturiale L, Garozzo D, Bei R, collaborators AVA, Masuelli L, Conti V, Novarino G & Fassio A. (2022). Phenotypic and genetic spectrum of ATP6V1A encephalopathy: a disorder of lysosomal homeostasis. Brain 145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Harrison JK, Jiang Y, Chen S, Xia Y, Maciejewski D, McNamara RK, Streit WJ, Salafranca MN, Adhikari S, Thompson DA, Botti P, Bacon KB, Feng L, Harrison JK, Jiang Y, Chen S, Xia Y, Maciejewski D, McNamara RK, Streit WJ, Salafranca MN, Adhikari S, Thompson DA, Botti P, Bacon KB & Feng L. (1998). Role for neuronally derived fractalkine in mediating interactions between neurons and CX3CR1-expressing microglia. Proceedings of the National Academy of Sciences 95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Hasanbasic I, Cuerquis J, Varnum B & Blostein MD. (2004). Intracellular signaling pathways involved in Gas6-Axl-mediated survival of endothelial cells. American Journal of Physiology-Heart and Circulatory Physiology 287. [DOI] [PubMed] [Google Scholar]
  55. Hide I, Shiraki H, Masuda A, Maeda T, Kumagai M, Kunishige N, Yanase Y, Harada K, Tanaka S & Sakai N. (2023). P2Y2 receptor mediates dying cell removal via inflammatory activated microglia. Journal of Pharmacological Sciences 153. [DOI] [PubMed] [Google Scholar]
  56. Holtman IR, Skola D & Glass CK. (2017). Transcriptional control of microglia phenotypes in health and disease. The Journal of Clinical Investigation 127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Hong S, Beja-Glasser V, Nfonoyim B, Frouin A, Li S, Ramakrishnan S, Merry K, Shi Q, Rosenthal A, Barres B, Lemere C, Selkoe D & Stevens B. (2016). Complement and microglia mediate early synapse loss in Alzheimer mouse models - PubMed. Science (New York, NY) 352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Hong S, Xin Y, HaiQin W, GuiLian Z, Ru Z, ShuQin Z, HuQing W, Li Y, Ning B & YongNan L. (2013). The PPARγ agonist rosiglitazone prevents neuronal loss and attenuates development of spontaneous recurrent seizures through BDNF/TrkB signaling following pilocarpine-induced status epilepticus. Neurochemistry International 63. [DOI] [PubMed] [Google Scholar]
  59. Huang K-l, Marcora E, Pimenova AA, Narzo AFD, Kapoor M, Jin SC, Harari O, Bertelsen S, Fairfax BP, Czajkowski J, Chouraki V, Grenier-Boley B, Bellenguez C, Deming Y, McKenzie A, Raj T, Renton AE, Budde J, Smith A, Fitzpatrick A, Bis JC, DeStefano A, Adams HH, Ikram MA, Lee Svd, Del-Aguila JL, Fernandez MV, Ibañez L, Project TIGoAs, Initiative TAsDN, Sims R, Escott-Price V, Mayeux R, Haines JL, Farrer LA, Pericak-Vance MA, Lambert JC, Duijn Cv, Launer L, Seshadri S, Williams J, Amouyel P, Schellenberg GD, Zhang B, Borecki I, Kauwe JSK, Cruchaga C, Hao K & Goate AM. (2017). A common haplotype lowers PU.1 expression in myeloid cells and delays onset of Alzheimer’s disease. Nature neuroscience 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Huang Y & Lemke G. (2022). Early death in a mouse model of Alzheimer’s disease exacerbated by microglial loss of TAM receptor signaling. Proceedings of the National Academy of Sciences 119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Inose Y, Kato Y, Kitagawa K, Uchiyama S & Shibata N. (2015). Activated microglia in ischemic stroke penumbra upregulate MCP-1 and CCR2 expression in response to lysophosphatidylcholine derived from adjacent neurons and astrocytes - PubMed. Neuropathology : official journal of the Japanese Society of Neuropathology 35. [DOI] [PubMed] [Google Scholar]
  62. Inouye MO. (2018). Role of P2Y12 receptor-mediated ADP signalling at microglia in the phagocytosis of neurons. [Google Scholar]
  63. Jayadev S, Nesser NK, Hopkins S, Myers SJ, Case A, Lee RJ, Seaburg LA, Uo T, Murphy SP, Morrison RS & Garden GA. (2011). Transcription factor p53 influences microglial activation phenotype. Glia 59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Khalaji A, Yancheshmeh FB, Farham F, Khorram A, Sheshbolouki S, Zokaei M, Vatankhah F & Soleymani-Goloujeh M. (2023). Don’t eat me/eat me signals as a novel strategy in cancer immunotherapy. Heliyon 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Kierdorf K, Erny D, Goldmann T, Sander V, Schulz C, Perdiguero EG, Wieghofer P, Heinrich A, Riemke P, Hölscher C, Müller DN, Luckow B, Brocker T, Debowski K, Fritz G, Opdenakker G, Diefenbach A, Biber K, Heikenwalder M, Geissmann F, Rosenbauer F, Prinz M, Kierdorf K, Erny D, Goldmann T, Sander V, Schulz C, Perdiguero EG, Wieghofer P, Heinrich A, Riemke P, Hölscher C, Müller DN, Luckow B, Brocker T, Debowski K, Fritz G, Opdenakker G, Diefenbach A, Biber K, Heikenwalder M, Geissmann F, Rosenbauer F & Prinz M. (2013). Microglia emerge from erythromyeloid precursors via Pu.1- and Irf8-dependent pathways. Nature Neuroscience 2013 16:3 16. [DOI] [PubMed] [Google Scholar]
  66. Kobau R, Luncheon C & Greenlund K. (2023). Active epilepsy prevalence among U.S. adults is 1.1% and differs by educational level—National Health Interview Survey, United States, 2021. Epilepsy & behavior : E&B 142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Koizumi S, Shigemoto-Mogami Y, Nasu-Tada K, Shinozaki Y, Ohsawa K, Tsuda M, Joshi B, Jacobson K, Kohsaka S & Inoue K. (2007). UDP acting at P2Y6 receptors is a mediator of microglial phagocytosis - PubMed. Nature 446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Kruiswijk F, Labuschagne CF, Vousden KH, Kruiswijk F, Labuschagne CF & Vousden KH. (2015). p53 in survival, death and metabolic health: a lifeguard with a licence to kill. Nature Reviews Molecular Cell Biology 2015 16:7 16. [DOI] [PubMed] [Google Scholar]
  69. Larner A (2011). Presenilin-1 mutation Alzheimer’s disease: a genetic epilepsy syndrome? - PubMed. Epilepsy & behavior : E&B 21. [DOI] [PubMed] [Google Scholar]
  70. Lauber K, Bohn E, Kröber S, Xiao Y, Blumenthal S, Lindemann R, Marini P, Wiedig C, Zobywalski A, Baksh S, Xu Y, Autenrieth I, Schulze-Osthoff K, Belka C, Stuhler G & Wesselborg S. (2003). Apoptotic cells induce migration of phagocytes via caspase-3-mediated release of a lipid attraction signal - PubMed. Cell 113. [DOI] [PubMed] [Google Scholar]
  71. Lehrman EK, Wilton DK, Litvina EY, Welsh CA, Chang ST, Frouin A, Walker AJ, Heller MD, Umemori H, Chen C & Stevens B. (2018). CD47 protects synapses from excess microglia-mediated pruning during development. Neuron 100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Lenck-Santini P-P & Scott RC. (2015). Mechanisms Responsible for Cognitive Impairment in Epilepsy. Cold Spring Harbor Perspectives in Medicine 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Leo A, Citraro R, Marra R, Palma E, Paola E, Constanti A, De Sarro G & Russo E. (2017). The Sphingosine 1-Phosphate Signaling Pathway in Epilepsy: A Possible Role for the Immunomodulator Drug Fingolimod in Epilepsy Treatment. CNS & neurological disorders drug targets 16. [DOI] [PubMed] [Google Scholar]
  74. Li C, Zhao B, Lin C, Gong Z & An X. (2019). TREM2 inhibits inflammatory responses in mouse microglia by suppressing the PI3K/NF-κB signaling - PubMed. Cell biology international 43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Libbey J, Kirkman N, Wilcox K, White H & Fujinami R. (2010). Role for complement in the development of seizures following acute viral infection - PubMed. Journal of virology 84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Liu J, Xiang J, Li X, Blankson S, Zhao S, Cai J, Jiang Y, Redmond HP, Wang JH, Liu J, Xiang J, Li X, Blankson S, Zhao S, Cai J, Jiang Y, Redmond HP & Wang JH. (2017a). NF-κB activation is critical for bacterial lipoprotein tolerance-enhanced bactericidal activity in macrophages during microbial infection. Scientific Reports 2017 7:1 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Liu M, Zhu J, Zheng J, Han X, Jiang L, Tong X, Ke Y, Guo Z, Huang W, Cong J, Liu M, Lin S-Y, Zhu S, Mei L, Zhang X, Zhang W, Xin W-J, Zhang Z, Guo Y & Chen R. (2025). GPNMB and ATP6V1A interact to mediate microglia phagocytosis of multiple types of pathological particles. Cell Reports 44. [DOI] [PubMed] [Google Scholar]
  78. Liu T, Zhang L, Joo D, Sun S-C, Liu T, Zhang L, Joo D & Sun S-C. (2017b). NF-κB signaling in inflammation. Signal Transduction and Targeted Therapy 2017 2:1 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Luo C, Koyama R & Ikegaya Y. (2016). Microglia engulf viable newborn cells in the epileptic dentate gyrus - PubMed. Glia 64. [DOI] [PubMed] [Google Scholar]
  80. Lybrand Z, Goswami S, Zhu J, Jarzabek V, Merlock N, Aktar M, Smith C, Zhang L, Varma P, Cho K, Ge S & Hsieh J. (2021). A critical period of neuronal activity results in aberrant neurogenesis rewiring hippocampal circuitry in a mouse model of epilepsy - PubMed. Nature communications 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Ma Y, Ramachandran A, Ford N, Parada I & Prince DA. (2013). Remodeling of dendrites and spines in the C1q knockout model of genetic epilepsy. Epilepsia 54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Marchi N & Brewster AL. (2024). Pericytes and Microglia. Jasper’s Basic Mechanisms of the Epilepsies. [Google Scholar]
  83. Martins I, Wang Y, Michaud M, Ma Y, Sukkurwala AQ, Shen S, Kepp O, Métivier D, Galluzzi L, Perfettini J-L, Zitvogel L, Kroemer G, Martins I, Wang Y, Michaud M, Ma Y, Sukkurwala AQ, Shen S, Kepp O, Métivier D, Galluzzi L, Perfettini J-L, Zitvogel L & Kroemer G. (2013). Molecular mechanisms of ATP secretion during immunogenic cell death. Cell Death & Differentiation 2014 21:1 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Matsuda T, Murao N, Katano Y, Juliandi B, Kohyama J, Akira S, Kawai T & Nakashima K. (2015). TLR9 signalling in microglia attenuates seizure-induced aberrant neurogenesis in the adult hippocampus. Nature Communications 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Medina C & Ravichandran K. (2016). Do not let death do us part: ‘find-me’ signals in communication between dying cells and the phagocytes - PubMed. Cell death and differentiation 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Mo M, Eyo UB, Xie M, Peng J, Bosco DB, Umpierre AD, Zhu X, Tian D-S, Xu P & Wu L-J. (2019). Microglial P2Y12 Receptor Regulates Seizure-Induced Neurogenesis and Immature Neuronal Projections. The Journal of Neuroscience 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Moretti J & Blander JM. (2014). Insights into phagocytosis-coupled activation of pattern recognition receptors and inflammasomes. Current Opinion in Immunology 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Nebeling F, Poll S, Justus L, Steffen J, Keppler K, Mittag M & Fuhrmann M. (2023). Microglial motility is modulated by neuronal activity and correlates with dendritic spine plasticity in the hippocampus of awake mice - PubMed. eLife 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Neher JJ, Neniskyte U, Zhao J-W, Bal-Price A, Tolkovsky AM & Brown GC. (2011). Inhibition of Microglial Phagocytosis Is Sufficient To Prevent Inflammatory Neuronal Death. The Journal of Immunology 186, 4973–4983. [DOI] [PubMed] [Google Scholar]
  90. Nóbrega P, Paiva A, Amorim Junior A, Lima P, Cabral K, Barcelos I, Pessoa A, Souza-Lima C, Castro M, Freua F, Santos E, Rocha M, Maia R, Araújo R, Ramos J, Resende R, Carvalho G, Valença L, Lima de Carvalho J, Melo E, Pedroso J, Barsottini O, Houlden H, Kok F & Lynch D. (2025). Further description of the phenotypic spectrum of neuronal ceroid lipofuscinosis type 11 - PubMed. Genetics in medicine : official journal of the American College of Medical Genetics 27. [DOI] [PubMed] [Google Scholar]
  91. Novak A, Vizjak K & Rakusa M. (2022). Cognitive Impairment in People with Epilepsy. Journal of Clinical Medicine 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Paloneva J, Autti T, Raininko R, Partanen J, Salonen O, Puranen M, Hakola P & Haltia M. (2001). CNS manifestations of Nasu-Hakola disease: a frontal dementia with bone cysts - PubMed. Neurology 56. [DOI] [PubMed] [Google Scholar]
  93. Paloneva J, Kestilä M, Wu J, Salminen A, Böhling T, Ruotsalainen V, Hakola P, Bakker A, Phillips J, Pekkarinen P, Lanier L, Timonen T & Peltonen L. (2000). Loss-of-function mutations in TYROBP (DAP12) result in a presenile dementia with bone cysts - PubMed. Nature genetics 25. [DOI] [PubMed] [Google Scholar]
  94. Paloneva J, Manninen T, Christman G, Hovanes K, Mandelin J, Adolfsson R, Bianchin M, Bird T, Miranda R, Salmaggi A, Tranebjaerg L, Konttinen Y & Peltonen L. (2002). Mutations in two genes encoding different subunits of a receptor signaling complex result in an identical disease phenotype - PubMed. American journal of human genetics 71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Paolicelli R, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, Giustetto M, Ferreira T, Guiducci E, Dumas L, Ragozzino D & Gross C. (2011). Synaptic pruning by microglia is necessary for normal brain development - PubMed. Science (New York, NY) 333. [DOI] [PubMed] [Google Scholar]
  96. Paolicelli RC, Sierra A, Stevens B, Tremblay M-E, Aguzzi A, Ajami B, Amit I, Audinat E, Bechmann I, Bennett M, Bennett F, Bessis A, Biber K, Bilbo S, Blurton-Jones M, Boddeke E, Brites D, Brône B, Brown GC, Butovsky O & Wyss-Coray T. (2022). Microglia states and nomenclature: A field at its crossroads. Neuron 110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Park J, Choi Y, Jung E, Lee S, Sohn J & Chung W. (2021). Microglial MERTK eliminates phosphatidylserine-displaying inhibitory post-synapses - PubMed. The EMBO journal 40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Peng J, Wang K, Xiang W, Li Y, Hao Y & Guan Y. (2019). Rosiglitazone polarizes microglia and protects against pilocarpine-induced status epilepticus. CNS Neuroscience & Therapeutics 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Pereira-Iglesias M, Maldonado-Teixido J, Melero A, Piriz J, Galea E, Ransohoff RM, Sierra A, Pereira-Iglesias M, Maldonado-Teixido J, Melero A, Piriz J, Galea E, Ransohoff RM & Sierra A. (2024). Microglia as hunters or gatherers of brain synapses. Nature Neuroscience 2024 28:1 28. [DOI] [PubMed] [Google Scholar]
  100. Peter C, Waibel M, Radu C, Yang L, Witte O, Schulze-Osthoff K, Wesselborg S & Lauber K. (2008). Migration to apoptotic “find-me” signals is mediated via the phagocyte receptor G2A - PubMed. The Journal of biological chemistry 283. [DOI] [PubMed] [Google Scholar]
  101. Quick J, Silva C, Wong J, Lim K, Reynolds R, Barron A, Zeng J & Lo C. (2023). Lysosomal acidification dysfunction in microglia: an emerging pathogenic mechanism of neuroinflammation and neurodegeneration - PubMed. Journal of neuroinflammation 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Reis ES, Mastellos DC, Hajishengallis G, Lambris JD, Reis ES, Mastellos DC, Hajishengallis G & Lambris JD. (2019). New insights into the immune functions of complement. Nature Reviews Immunology 2019 19:8 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Rustenhoven J, Smith AM, Smyth LC, Jansson D, Scotter EL, Swanson MEV, Aalderink M, Coppieters N, Narayan P, Handley R, Overall C, Park TIH, Schweder P, Heppner P, Curtis MA, Faull RLM & Dragunow M. (2018). PU.1 regulates Alzheimer’s disease-associated genes in primary human microglia. Molecular Neurodegeneration 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Saijo K, Glass CK, Saijo K & Glass CK. (2011). Microglial cell origin and phenotypes in health and disease. Nature Reviews Immunology 2011 11:11 11. [DOI] [PubMed] [Google Scholar]
  105. Sandvig I, Augestad I, Håberg A & Sandvig A. (2018). Neuroplasticity in stroke recovery. The role of microglia in engaging and modifying synapses and networks - PubMed. The European journal of neuroscience 47. [DOI] [PubMed] [Google Scholar]
  106. Savage JC, Jay T, Goduni E, Quigley C, Mariani MM, Malm T, Ransohoff RM, Lamb BT & Landreth GE. (2015). Nuclear Receptors License Phagocytosis by Trem2+ Myeloid Cells in Mouse Models of Alzheimer’s Disease. Journal of Neuroscience 35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Saxton RA & Sabatini DM. (2017). mTOR Signaling in Growth, Metabolism, and Disease. Cell 168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Schaarenburg RAv, Magro-Checa C, Bakker JA, Teng YKO, Bajema IM, Huizinga TW, Steup-Beekman GM & Trouw LA. (2016). C1q Deficiency and Neuropsychiatric Systemic Lupus Erythematosus. Frontiers in Immunology 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Schafer DP, Lehrman EK, Kautzman AG, Koyama R, Mardinly AR, Yamasaki R, Ransohoff RM, Greenberg ME, Barres BA & Stevens B. (2012). Microglia Sculpt Postnatal Neural Circuits in an Activity and Complement-Dependent Manner. Neuron 74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Schartz ND, Aroor A, Li Y, Pinzón-Hoyos N & Brewster AL. (2023). Mice deficient in complement C3 are protected against recognition memory deficits and astrogliosis induced by status epilepticus. Frontiers in Molecular Neuroscience 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Shafit-Zagardo B, Gruber RC & DuBois JC. (2018). The role of TAM family receptors and ligands in the nervous system: From development to pathobiology. Pharmacology & Therapeutics 188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Sierra A, Beccari S, Diaz-Aparicio I, Encinas JM, Comeau S & Tremblay M-È. (2014). Surveillance, Phagocytosis, and Inflammation: How Never-Resting Microglia Influence Adult Hippocampal Neurogenesis. Neural Plasticity 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Sierra A, Encinas JM, Deudero JJ, Chancey JH, Enikolopov G, Overstreet-Wadiche LS, Tsirka SE & Maletic-Savatic M. (2010). Microglia shape adult hippocampal neurogenesis through apoptosis-coupled phagocytosis. Cell stem cell 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Singh S, Barik D, Arukha AP, Prasad S, Mohapatra I, Singh A, Singh G, Singh S, Barik D, Arukha AP, Prasad S, Mohapatra I, Singh A & Singh G. (2023). Small Molecule Targeting Immune Cells: A Novel Approach for Cancer Treatment. Biomedicines 2023, Vol 11, Page 2621 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Smith AM, Gibbons HM, Oldfield RL, Bergin PM, Mee EW, Faull RLM & Dragunow M. (2013). The transcription factor PU.1 is critical for viability and function of human brain microglia. Glia 61. [DOI] [PubMed] [Google Scholar]
  116. Sokolowski J, Chabanon-Hicks C, Han C, Heffron D & Mandell J. (2014). Fractalkine is a “find-me” signal released by neurons undergoing ethanol-induced apoptosis. Frontiers in cellular neuroscience 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS, Nouri N, Micheva KD, Mehalow AK, Huberman AD, Stafford B, Sher A, Litke Alan M, Lambris JD, Smith SJ, John SWM & Barres BA. (2007). The Classical Complement Cascade Mediates CNS Synapse Elimination. Cell 131. [DOI] [PubMed] [Google Scholar]
  118. Stevens B, Johnson MB, Stevens B & Johnson MB. (2021). The complement cascade repurposed in the brain. Nature Reviews Immunology 2021 21:10 21. [DOI] [PubMed] [Google Scholar]
  119. Sultana B, Panzini M, Veilleux Carpentier A, Comtois J, Rioux B, Gore G, Bauer P, Kwon C, Jetté N, Josephson C & Keezer M. (2021). Incidence and Prevalence of Drug-Resistant Epilepsy: A Systematic Review and Meta-analysis - PubMed. Neurology 96. [DOI] [PubMed] [Google Scholar]
  120. Tanaka Y, Suzuki G, Matsuwaki T, Hosokawa M, Serrano G, Beach T, Yamanouchi K, Hasegawa M & Nishihara M. (2017). Progranulin regulates lysosomal function and biogenesis through acidification of lysosomes - PubMed. Human molecular genetics 26. [DOI] [PubMed] [Google Scholar]
  121. Tian D, Peng J, Murugan M, Feng L, Liu J, Eyo U, Zhou L, Mogilevsky R, Wang W & Wu L. (2017). Chemokine CCL2-CCR2 Signaling Induces Neuronal Cell Death via STAT3 Activation and IL-1β Production after Status Epilepticus. The Journal of neuroscience : the official journal of the Society for Neuroscience 37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Tibrewal N, Wu Y, D’mello V, Akakura R, George T, Varnum B & Birge R. (2008). Autophosphorylation docking site Tyr-867 in Mer receptor tyrosine kinase allows for dissociation of multiple signaling pathways for phagocytosis of apoptotic cells and down-modulation of lipopolysaccharide-inducible NF-kappaB transcriptional activation. The Journal of biological chemistry 283. [DOI] [PubMed] [Google Scholar]
  123. Tremblay M-È, Lowery RL & Majewska AK. (2010). Microglial Interactions with Synapses Are Modulated by Visual Experience. PLoS Biology 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Tyagi S, Gupta P, Saini AS, Kaushal C & Sharma S. (2011). The peroxisome proliferator-activated receptor: A family of nuclear receptors role in various diseases. Journal of Advanced Pharmaceutical Technology & Research 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Umpierre A, Li B, Ayasoufi K, Simon W, Zhao S, Xie M, Thyen G, Hur B, Zheng J, Liang Y, Bosco D, Maynes M, Wu Z, Yu X, Sung J, Johnson A, Li Y & Wu L. (2024). Microglial P2Y6 calcium signaling promotes phagocytosis and shapes neuroimmune responses in epileptogenesis - PubMed. Neuron 112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Umpierre AD & Wu L-J. (2021). How microglia sense and regulate neuronal activity. Glia 69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Uribe-Querol E & Rosales C. (2020). Phagocytosis: Our Current Understanding of a Universal Biological Process. Frontiers in Immunology 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Vago JP, Amaral FA & Loo FAJvd. (2021). Resolving inflammation by TAM receptor activation. Pharmacology & Therapeutics 227. [DOI] [PubMed] [Google Scholar]
  129. Victor TR & Tsirka SE. (2020). Microglial contributions to aberrant neurogenesis and pathophysiology of epilepsy. Neuroimmunology and neuroinflammation 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Vorselen D (2022). Dynamics of phagocytosis mediated by phosphatidylserine. Biochemical Society Transactions 50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Wang C, Zhang J, Song S, Li Z, Yin S, Duan W, Wei Z, Qi M, Sun W, Zhang L, Chen L, Gao X, Mao Y, Wang H, Chen L & Li C. (2021). Imaging epileptic foci in mouse models via a low-density lipoprotein receptor-related protein-1 targeting strategy. EBioMedicine 63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Wang L, Kong Q, Leng X, Leung H & Li Y. (2024). The sphingosine-1-phosphate signaling pathway (sphingosine-1-phosphate and its receptor, sphingosine kinase) and epilepsy. Epilepsia Open. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Wang S, Mustafa M, Yuede C, Salazar S, Kong P, Long H, Ward M, Siddiqui O, Paul R, Gilfillan S, Ibrahim A, Rhinn H, Tassi I, Rosenthal A, Schwabe T & Colonna M. (2020). Anti-human TREM2 induces microglia proliferation and reduces pathology in an Alzheimer’s disease model - PubMed. The Journal of experimental medicine 217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Wei Y, Chen T, Bosco DB, Xie M, Zheng J, Dheer A, Ying Y, Wu Q, Lennon VA & Wu L-J. (2021). The complement C3-C3aR pathway mediates microglia-astrocyte interaction following status epilepticus. Glia 69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Wu G, McBride DW & Zhang JH. (2018). Axl activation attenuates neuroinflammation by inhibiting the TLR/TRAF/NF-κB pathway after MCAO in rats. Neurobiology of Disease 110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Wu Q, Wang H, Liu X, Zhao Y & Zhang J. (2022). The Role of the Negative Regulation of Microglia-Mediated Neuroinflammation in Improving Emotional Behavior After Epileptic Seizures. Frontiers in Neurology 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Wu W, Li Y, Wei Y, Bosco DB, Xie M, Zhao M-G, Richardson JR & Wu L-J. (2020). Microglial depletion aggravates the severity of acute and chronic seizures in mice. Brain, behavior, and immunity 89, 245–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Wyatt-Johnson SK & Brewster AL. (2019). Emerging Roles for Microglial Phagocytic Signaling in Epilepsy. Epilepsy Currents 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Xiao S, Peng K, Li C, Long Y, Yu Q, Xiao S, Peng K, Li C, Long Y & Yu Q. (2023). The role of sphingosine-1-phosphate in autophagy and related disorders. Cell Death Discovery 2023 9:1 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Xu Y, Zeng K, Han Y, Wang L, Chen D, Xi Z, Wang H, Wang X & Chen G. (2012). Altered expression of CX3CL1 in patients with epilepsy and in a rat model - PubMed. The American journal of pathology 180. [DOI] [PubMed] [Google Scholar]
  141. Xue T, Ji J, Sun Y, Huang X, Cai Z, Yang J, Guo W, Guo R, Cheng H & Sun X. (2021). Sphingosine-1-phosphate, a novel TREM2 ligand, promotes microglial phagocytosis to protect against ischemic brain injury. Acta Pharmaceutica Sinica B 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Yadav M, Sharma A, Patne K, Tabasum S, Suryavanshi J, Rawat L, Machaalani M, Eid M, Singh RP, Choueiri TK, Pal S & Sabarwal A. (2025). AXL signaling in cancer: from molecular insights to targeted therapies. Signal Transduction and Targeted Therapy 2025 10:1 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Yamanaka M, Ishikawa T, Griep A, Axt D, Kummer MP & Heneka MT. (2012). PPARγ/RXRα-Induced and CD36-Mediated Microglial Amyloid-β Phagocytosis Results in Cognitive Improvement in Amyloid Precursor Protein/Presenilin 1 Mice. Journal of Neuroscience 32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Yao H, Coppola K, Schweig JE, Crawford F, Mullan M & Paris D. (2019). Distinct Signaling Pathways Regulate TREM2 Phagocytic and NFκB Antagonistic Activities. Frontiers in Cellular Neuroscience 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Yeo S, Kim J, Ryu H, Seo C, Lee B, Choi I, Kim D & Kang T. (2011). The roles of fractalkine/CX3CR1 system in neuronal death following pilocarpine-induced status epilepticus - PubMed. Journal of neuroimmunology 234. [DOI] [PubMed] [Google Scholar]
  146. Yoon KW, Byun S, Kwon E, Hwang S-Y, Chu K, Hiraki M, Jo S-H, Weins A, Hakroush S, Cebulla A, Sykes DB, Greka A, Mundel P, Fisher DE, Mandinova A & Lee SW. (2015). Control of signaling-mediated clearance of apoptotic cells by the tumor suppressor p53. Science 349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Yu C, Deng X-j & Xu D. (2023). Microglia in epilepsy. Neurobiology of Disease 185. [DOI] [PubMed] [Google Scholar]
  148. Zahiri D, Burow P, Großmann C, Müller C, Klapperstück M & Markwardt F. (2021). Sphingosine-1-phosphate induces migration of microglial cells via activation of volume-sensitive anion channels, ATP secretion and activation of purinergic receptors. Biochimica et biophysica acta Molecular cell research 1868. [DOI] [PubMed] [Google Scholar]
  149. Zhao J, Li Q, Ouyang X, Wang F, Li Q, Xu Z, Ji D, Wu Q, Zhang J, Lu C, Ji S & Li S. (2023). The effect of CX3CL1/ CX3CR1 signal axis on microglia in central nervous system diseases. Journal of Neurorestoratology 11. [Google Scholar]
  150. Zhao S, Umpierre AD & Wu L-J. (2024). Tuning neural circuits and behaviors by microglia in the adult brain. Trends in Neurosciences 47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Zhao X-F, Liao Y, Alam MM, Mathur R, Feustel P, Mazurkiewicz JE, Adamo MA, Zhu XC & Huang Y. (2020). Microglial mTOR is Neuronal Protective and Antiepileptogenic in the Pilocarpine Model of Temporal Lobe Epilepsy. Journal of Neuroscience 40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Zhao X, Liao Y, Morgan S, Mathur R, Feustel P, Mazurkiewicz J, Qian J, Chang J, Mathern GW, Adamo MA, Ritaccio AL, Gruenthal M, Zhu X & Huang Y. (2018). Noninflammatory Changes of Microglia Are Sufficient to Cause Epilepsy. Cell Reports 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Zhao X, Sun G, Ting S-M, Song S, Zhang J, Edwards NJ & Aronowski J. (2015). Cleaning up after ICH: the role of Nrf2 in modulating microglia function and hematoma clearance. Journal of Neurochemistry 133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Zimmer T, Korotkov A, Zwakenberg S, Jansen F, Zwartkruis F, Rensing N, Wong M, Mühlebner A, van Vliet E, Aronica E & Mills J. (2021). Upregulation of the pathogenic transcription factor SPI1/PU.1 in tuberous sclerosis complex and focal cortical dysplasia by oxidative stress - PubMed. Brain pathology (Zurich, Switzerland) 31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Zou J & Shankar N. (2015). Roles of TLR/MyD88/MAPK/NF-κB Signaling Pathways in the Regulation of Phagocytosis and Proinflammatory Cytokine Expression in Response to E. faecalis Infection. PLOS ONE 10. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES