This is an open access publication, available online and distributed under the terms of a Creative Commons Attribution-Non Commercial-No Derivatives 4.0 International licence (CC BY-NC-ND 4.0), a copy of which is available at https://creativecommons.org/licenses/by-nc-nd/4.0/. Subject to this license, all rights are reserved.
NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.
Noebels JL, Avoli M, Rogawski MA, et al., editors. Jasper's Basic Mechanisms of the Epilepsies. 5th edition. New York: Oxford University Press; 2024. doi: 10.1093/med/9780197549469.003.0029
Abstract
In seizure conditions, neuro-glio-vascular immune communications, blood–brain barrier (BBB) damage, and synaptic circuitry functional remodeling are dynamic processes that unfold over time and by brain region. These processes are associated with the generation and maintenance of epileptic networks. Accumulating evidence suggests that pericytes and microglial cells play critical roles in contributing to epileptogenic events, with a specific and renewed attention to pro- and anti-inflammatory equilibriums that strictly depend on the disease stage. Implications for pericytes include BBB dysfunction during and after seizures, while microglia is involved in cerebrovascular and neural network remodeling through altered surveillance, phagocytic, and inflammatory responses. These cellular modifications can span from the epileptogenic foci to seizure-propagating regions or networks. Understanding how pericyte and microglial cells interact with neuronal and cerebrovascular structures will facilitate the development of multitarget pharmacological strategies for epilepsy. Within this framework, the timing of pharmacological intervention could dictate therapeutic success, in the light of the varying and contrasting inflammatory and neurovascular trajectories that are cell-specific and unfold after a seizure or during epilepsy.
Introduction: Neuro-Glio-Vascular Regulatory Cells in Seizure Networks
Over the last decade, numerous studies introduced novel roles for pericytes and microglial cells in the development and maintenance of a healthy brain and the pathophysiology of neurological disorders, including epilepsy (Giannoni et al., 2018; Klement et al., 2018, 2019; Milesi et al., 2014; Cheng et al., 2018; Eyo et al., 2016, 2021; Eyo and Wu, 2019; Wyatt-Johnson and Brewster, 2020). Pericytes and microglia can work independently as well as together, both to ensure a tight homeostatic control within networks for proper neuronal communication and to safeguard blood-brain barrier (BBB) tightness and physiological functions at the neurovascular unit (NVU) (Obermeieret al., 2013). Briefly, the BBB consists of a network of brain capillaries with a monolayer endothelium sealed by tight junctions, interfacing and segregating the brain parenchyma from the peripheral blood. Exact communications between the endothelial cells and the adjacent pericyte, astrocyte end-feet, and microglial cells continuously control BBB properties and NVU functions (Figs. 29–1 and 29–2). In a healthy system, the BBB is mostly impermeable to blood-borne molecules, and it finely tunes the brain homeostasis through a bidirectional control of peripheral-brain gradients of cells, molecules, and ions for correct synaptic transmission (Sweeney et al., 2019; Ghosh et al., 2011; Janigro et al., 2020). Considerable evidence supports cerebrovascular damage as a pathophysiological player in experimental and clinical epilepsy (Gorter et al., 2015; Librizzi et al., 2018; Marchi et al., 2011, 2012; Noe and Marchi, 2019; Swissa et al., 2019). In the healthy brain, pericytes participate in capillary stability, angiogenesis, and tightness of the endothelium (Attwell et al., 2016; Giannoni et al., 2018; Obermeier et al., 2013; Rustenhoven et al., 2017; Sweeney et al., 2019), while microglial cells surveil the perivascular space (Giannoni et al., 2018; Klement et al., 2018, 2019) and, in turn, promote neuronal survival and maintain synaptic circuitry (Kierdorf and Prinz, 2017). Evidence from humans and experimental models of epilepsy suggests that pericytes and microglia regulate immune and inflammatory responses that may have beneficial and detrimental consequences in the pathology of acute seizures and chronic epilepsy (Klement et al., 2019; Arango-Lievano et al., 2018).

Figure 29–1.
The endothelial-pericyte-glia interface in health and seizure conditions. A. At the neurovascular unit, endothelial cells (pink), pericytes (or smooth muscle cells depending on vessel caliber) (blue), astrocytes (green), neighboring microglial cells (more...)

Figure 29–2.
Examples of glio-pericytes structures at the cerebrovasculature. A. Two-photon acquisition of cortical brain arteries and NG2DsRed mural cells. B. Distribution of GFAP astrocytes and NG2DsRED pericytes in the cortex. NG2DsRED pericytes processes outline (more...)
Both pericytes and microglia react to bursts of neuronal hyperexcitability that subsequently change their morphology and functions. During and following seizures, pericytes detach from their mural positioning at the BBB endothelium and undergo damage and proliferation (i.e., pericytosis), thereby allowing instability and permeability of the BBB to occur. At the same time, microglial cells rapidly activate in response to seizures and migrate toward parenchymal neurons or to the perivascular space where their time-dependent detrimental or healing roles are studied (Figs. 29–1 and 29–3). Here, we provide basic definitions of pericyte and microglial cells along with their recently described roles in the regulation of neuronal activity and the NVU through the modulation of immune, pro- and anti-inflammatory, responses (Rustenhoven et al., 2017). The implication of pericyte damage in seizure conditions integrates the notion of BBB dysfunction in promoting abnormal neuronal firing (Arango-Lievano et al., 2018; Garbelli et al., 2015; Giannoni et al., 2018; Klement et al., 2018, 2019; Librizzi et al., 2018; Marchi and Lerner-Natoli, 2013; Milesi et al., 2014). Alongside, contemporary studies have disclosed novel roles for microglia, including potential control of neuronal activity when seizures occur in otherwise healthy systems, and impaired or improper phagocytosis of synaptic structures and/or neurons, all of which may be guided by a myriad of immune molecules such as cytokines and complement proteins (reviewed in Eyo et al., 2016; Eyo and Wu, 2019; Wyatt-Johnson and Brewster, 2020). Important from a cellular mechanism standpoint, understanding the spatiotemporal trajectories of pericytes and microglia modifications, and their contribution to epileptogenic processes, could lead to the development of novel diagnostic or pharmacological strategies (Giannoni et al., 2018, 2020).

Figure 29–3.
Microglial-neuronal interactions in health and seizure conditions. A. Under physiological conditions, microglia (red) occupy nonoverlapping territories in the brain parenchyma where they interact with neurons (green). B. During acute seizures, microglial (more...)
What Is a Pericyte?
Pericytes are mural cells, presenting a distinctive rotund soma with slim and long ramifications lining the abluminal side of the BBB endothelium (Attwell et al., 2016; Sweeney et al., 2016; Figs. 29–1 and 29–2). These cells were first described in 1873 and were referred to as Rouget cells. Later on, their name was changed to pericytes to reflect their close association to the endothelial cells. Pericytes form a tight sleeve, embedded in basement membranes, that provides essential structural and molecular support to regulate BBB stability, (re)growth, and hemodynamics (Attwell et al., 2016; Sweeney et al., 2019).
A heterogeneous population of pericytes with varying morphologies and protein expression profiles has been reported (Attwell et al., 2016; Sweeney et al., 2019). Pericytes localize along and around the walls of capillaries which they can either completely ensheath (i.e., circumferential processes) or partially cover (i.e., mesh pericytes; see Attwell et al., 2016, for review; Fig. 29–1). The molecular and functional properties associated with morphological heterogeneity of capillary pericytes remain to be deciphered (Attwell et al., 2016). Pericytes can be identified using immunostaining with antibodies against the glycoprotein CD13 and the platelet-derived growth factor receptor beta (PDGFRβ) or with transgenic mice (i.e., NG2DsRed). At the molecular level, the dynamic and physiological interplay between pericytes and endothelial cells is mediated by molecules that include PDGFRβ, tumor growth factor-β (TGF-β), and Notch Receptor 3, among others (Fig. 29–1D). Signaling through PDGFRβ, a tyrosine kinase receptor, promotes cerebrovascular integrity. TGF-β secreted by endothelial cells and pericytes binds to the TGF-β receptor 2 (TGFβR2) to activate the downstream Smad signaling cascade. This TGF-β/Smad signaling cascade inhibits pericyte proliferation and migration, promotes pericyte differentiation and attachment, and triggers the expression of extracellular matrix proteins. Lastly, the activated Notch3 receptor cleaves its intracellular domain to promote pericyte survival and cooperates with TGF-β/TGFβR2 to augment BBB stability (see Sweeney et al., 2016, 2019, for a comprehensive review). The latter signaling is also implicated in perivascular astrocyte dysfunction and cell inflammatory phenotype (Ivens et al., 2007).
Because of their unique perivascular localization at the BBB, the contribution of pericytes to inflammation, from either brain or blood-borne origin, is emerging (Giannoni et al., 2018; Rustenhoven et al., 2017). Pericytes mediate blood leukocytes’ extravasation into the brain parenchyma, allowing for the bidirectional propagation of peripheral and brain pro-inflammatory events to occur (Rustenhoven et al., 2017). Importantly, during inflammatory states, pericytes express an array of cytokines, chemokines, adhesion molecules (ICAM, VCAM), reactive oxygen and nitrogen species (ROS/RNS), and metalloproteinases (MMP2-9), all of which are associated with mechanisms underlying epileptogenic processes (Rustenhoven et al., 2017; Marchi et al., 2014; Vezzani et al., 2011, 2015; Fig. 29–1D). Pericytes also express elements of damage-associated molecular patterns (DAMPS) and pathogen-associated molecular patterns (PAMPS) which enable the propagation of immune responses between the periphery and the brain. Their inflammatory machinery encompasses typical elements, including nuclear factor kappa light chain enhancer of activated B-cells (NF-κB), production of interferon-gamma (IFNγ) and tumor necrosis factor-alpha (TNFα), and the expression of major histocompatibility complex class II (MHCII) (see Rustenhoven et al., 2017, for review). In addition, pericytes have been shown to have phagocytic properties which may facilitate the uptake of damaged cells and waste products for interstitial clearance, to secrete mediators polarizing or activating microglia, and to internalize antigens for T-cell presentation (Rustenhoven et al., 2017).
Perivascular Inflammatory Cell Reactivity during Seizures: Focus on Pericytes
Pericyte damage, remodeling, and reactivity are implicated in the progression or severity of neurological diseases, including Alzheimer disease (AD), traumatic brain injury (TBI), ischemic stroke, and epilepsy (Giannoni et al., 2018; Sweeney et al., 2016, 2019). Here, we focus on the emerging roles of pericytes in experimental seizure conditions. Structural deconstruction and reactivity of pericytes and glial cells around the capillary endothelium (or perivascular) directly impact BBB permeability and it participates in neuroinflammation (Sweeney et al., 2019; Rustenhoven et al., 2017). Whether pericytes reactivity precedes or follows the occurrence of a seizure, BBB dysfunction, and increased permeability is unknown. Elucidation of these aspects will contribute to understanding pericyte involvement in the disease (Bar-Klein et al., 2017; Kastanauskaite et al., 2009; Marchi et al., 2007; Ivens et al., 2007; Pavlovsky et al., 2005; Seiffert et al., 2004). BBB damage can be prompted by pro-inflammatory factors such as IL-1β and IL-6 (Vezzani et al., 2011, 2019) and by local increases of extracellular glutamate concentrations (Vazana et al., 2016), which are events typically seen during seizure activity. A multicellular modification, or adaptation, occurs at the outer capillary wall after experimental status epilepticus (SE) and during seizure progression (Arango-Lievano et al., 2018; Giannoni et al., 2018; Klement et al., 2018, 2019; Milesi et al., 2014). By using NG2DsRed transgenic mice and FITC-Dextran micro-angiography, pericyte detachment from the BBB capillary in the dorsal hippocampus and overlying cortices was reported shortly after generalized SE induced by intra-peritoneal kainic acid (KA) injection (Milesi et al., 2014). Extending this initial evidence, during epileptogenesis and at spontaneous chronic seizures in a murine model of temporal lobe epilepsy (TLE, unilateral intra-hippocampal KA), NG2DsRed pericytes undergo a time-dependent detachment from the capillaries and a Ki67 + proliferation in the CA1 and CA3 hippocampal regions of the epileptic foci (Klement et al., 2018). During this process, pericytes lose their typical bump-on-a-log morphology, their minute perivascular ramifications, and they become hypertrophic (Klement et al., 2019; Librizzi et al., 2018; Boux et al., 2021). The latter resembled pericyte pathology (i.e., pericytosis) also reported after experimental traumatic brain injury (Zehendner et al., 2015). Precise and time-dependent dynamics of vessel-specific removal and addition of mural cells (smooth muscles and pericytes) were outlined using longitudinal 2-photon microscopy after a generalized SE in the somatosensory cortex (Arango-Lievano et al., 2018). Importantly, loss of pericytes negatively impacted cortical vasoreactivity in response to glutamate or endothelin-1 applications (Arango-Lievano et al., 2018). From a functional standpoint, the continuous remodeling of mural cells provoked by SE impacted in vivo vasoreactivity and blood flow (Arango-Lievano et al., 2018). Furthermore, BBB opening was preceded by capillary constriction with indications of pericyte injury-mediated loss of capillary integrity. Interestingly, reduced regional cerebrovascular reactivity was linked to mitochondrial depolarization in pericytes (Prager et al., 2019).
Importantly, pericytes were proposed to have immune-like properties and to participate in inflammatory responses, either peripheral or brain-borne (Smyth et al., 2018; Rustenhoven et al., 2017). This is important because overexpression of pro-inflammatory cytokines and chemokines occurs early in response to seizures, and players such as IL-1β can promote pericyte damage and microglia-pericyte assemblies at the cerebrovasculature (Klement et al., 2018). However, it remains to be understood whether these perivascular microglia-pericyte clusters are a component of restorative or detrimental processes.
What Are Microglial Cells?
Microglia are the tissue-specific macrophages and professional phagocytes of the central nervous system. These cells were first described by Spanish researcher Pío del Río Hortega in 1919. A century after, numerous scientific discoveries support that these cells are critically important for the development, maturation, and maintenance of neural circuitries (Prinz et al., 2019), and they contribute to the pathology of many neurological disorders (Hammond et al., 2018), including epilepsy (Wyatt-Johnson and Brewster, 2020; Eyo et al., 2016). Microglia are highly active cells that occupy and survey nonoverlapping territories throughout the brain (Fig. 29–3A), which in turn facilitates rapid responses to immune and homeostatic disturbances (Fig. 29–3B–D) (Eyo and Wu, 2019; Wyatt-Johnson and Brewster, 2020). These stimuli can trigger changes in microglial morphology from cells with highly ramified profiles to cells with hypertrophied and amoeboid shapes (Fig. 29–3D), which are associated with alterations in their functional status (i.e., surveillance, phagocytic, inflammatory).
The functions that microglia undertake under physiological and pathological conditions are tightly correlated to the expression of receptors specialized to sense and orchestrate appropriate responses to “find-me,” “eat-me,” and “don’t-eat-me” signals in their microenvironment (Sierra et al., 2013). Under physiological conditions, ramified microglia are constantly patrolling/surveying the brain by following ADT/ATP gradients (“find-me” signals) through purinergic receptors such as P2Y12 (Eyo and Wu, 2019). Phagocytic receptors, such as the triggering receptor expressed in myeloid cells 2 (Trem2), receptor Mer Tyrosine Kinase (MerTK), Vitronectin receptors (VNR), and complement receptors 1 and 3 (CR1, CR3), can promote the rearrangement of the cytoskeleton and help induce phagocytosis (Sierra et al., 2013; Brown and Neher, 2014; Konishi and Kiyama, 2018). Some “eat-me” signals that activate these receptors are lipids and DNA for Trem2, phosphatidylserine (PS) exposed on the surface of stressed or apoptotic cells bound to the opsonins Protein S (ProS) or Milk fat globule epidermal growth factor 8 (MFG-E8), which are ligands for MerTK and VNR, respectively, complement C1q for CR1, and complement C3b for CR3. These phagocytotic signals can be balanced by “don’t-eat-me” signals such as the integrin-associated protein CD47 that is sensed by the regulatory protein α (SIRP-α) receptor to inhibit phagocytosis. Receptors such as the colony-stimulating factor 1 receptor (CSF1R) are activated by the colony-stimulating factor 1 (CSF1) or interleukin 34 (IL-34) to regulate various functions involving inflammatory and phagocytic responses, as well as microglial survival and proliferation (Ulland et al., 2015; Oosterhof et al., 2018; Elmore et al., 2014). Activation of the complement system leads to downstream activation of C3a/b and C5a/b signaling that can regulate microglial phagocytic and inflammatory responses (Barnum, 2017; Schartz and Tenner, 2020), while Toll-like receptors (TLR) signal for the production of cytokines (van Vliet et al., 2018). Microglia can produce pro- and anti-inflammatory cytokines, including IL-1β and TNFα, which promote inflammation, and IL-4, IL-10, and IL-13, which help produce anti-inflammatory signals (Hiragi et al., 2018; Ravizza and Vezzani, 2018). Note that all these different signaling cascades can activate downstream pathways that can crosstalk with each other and thereby regulate or control the extent of both phagocytic and inflammatory responses (Sierra et al., 2013). New evidence over the last decade support that dysregulation of microglial surveillance, phagocytic, and inflammatory properties participate in the neuropathology and pathophysiology of epilepsy in humans and preclinical models. In this section, we summarize and discuss some of these findings.
Microglia Surveillance and Neuronal Interactions
Microglial cells are constantly surveying their microenvironment and making contacts with nearby neurons in an activity-dependent manner (Eyo and Wu, 2019; Eyo et al., 2016). This process is important for a rapid immune response and necessary for the proper development and maintenance of synaptic circuitries (Hammond et al., 2018). The glia-neuron interactions are mediated, in part, by microglial P2Y12R responses to microgradients of ATP (Eyo and Wu, 2019). Seizures promote an increase in the levels of ATP (Beamer et al., 2019), and along with this, microglial processes interact more frequently with neuronal elements (Hasegawa et al., 2007; Brewster et al., 2013; Eyo et al., 2014, 2015, 2016, 2017, 2021). Recent studies showed that seizure-induced increases in microglial-neuronal contacts were dependent on P2Y12R (Eyo et al., 2014), fractalkine signaling (CX3CL1-CX3CR1) that was coupled with increases in IL-1β (Eyo et al., 2017), neuronal NMDA receptor activation (Eyo et al., 2014), and extracellular calcium (Eyo et al., 2014, 2015). Interestingly, these studies also showed that mice deficient in P2Y12R or CX3CR1 displayed higher seizure severity following KA or pilocarpine-induced seizures (Eyo et al., 2014, 2016, 2017), suggesting that microglial surveillance functions are important to reduce neuronal hyperexcitability when a seizure is induced in an otherwise healthy system. This is further supported by a recent study showing that decreasing microglia ability to survey their microenvironment by inhibiting the Gi pathway, needed for the downstream activation of signaling pathways dependent on G protein–coupled receptors (GPCRs), results in unprovoked seizures and network hypersynchrony in mice (Merlini et al., 2021). This evidence suggests that microglial surveillance properties are altered in response to seizures and may work to dampen the sudden neuronal hyperexcitability that promotes more seizures. However, questions remain on how exactly microglia achieve this effect.
Microglia can shape the connectivity of synaptic networks in an activity- or experience-dependent manner (Schafer et al., 2012; Stevens et al., 2007; Prinz et al., 2019; Tay et al., 2017; Tremblay et al., 2010). The activity-driven increase in glial-neuronal contacts may result in modifications to synaptic structures because microglia have been shown to displace, eliminate, or promote the growth of synaptic elements (Tremblay et al., 2010; Weinhard et al., 2018; Miyamoto et al., 2016; Wake et al., 2009; Wan et al., 2020; Chen et al., 2014; Hammond et al., 2018). For example, microglial-mediated displacement of GABAergic synapses has been shown to occur in developing brains exposed to complex febrile seizures (FS) (Wan et al., 2020) and in adult brains subjected to lipopolysaccharide (LPS)-induced microglial activation (Chen et al., 2014). Following FS induced in young mice, an increased association of microglial processes with the soma of cortical neurons was coupled to an absence in GABAergic terminals, reduced GABAergic postsynaptic currents, and decreased frequency of evoked action potentials compared to neurons with less or without microglial associations (Wan et al., 2020). These effects were abolished with pharmacogenetic-induced reductions of P2Y12R, thereby suggesting that seizure-induced P2Y12R-dependent microglial-neuronal contacts are neuroprotective in the immature brain. Similarly, LPS-induced microglial activation in adult mice or rats resulted in increased microglial enwrapping of the soma of cortical neurons, reduced GABAergic neurotransmission, and increased synchronized neuronal firing (Chen et al., 2014). Even though activation of microglia had a similar effect in enwrapping and displacing GABAergic synapses in the immature and adult brain, the impact on network hyperexcitability was the opposite, likely due to the excitatory role for GABA early in life (Ben-Ari et al., 2012). The activated microglial-mediated displacement of GABAergic synapses in an adult system may potentiate the neuronal hyperactivity that results in seizures, suggesting age-specific roles for microglia in the pathogenic sequelae of prolonged seizures.
Complement proteins such as C1q and C3 act as guides in microglia-mediated synaptic stripping during normal development and in neurodegenerative disorders (Schafer et al., 2012, 2013; Stephan et al., 2012; Stevens et al., 2007; Carpanini et al., 2019; Prinz et al., 2019). In the normal developing retinogeniculate system, C1q and C3b promote the elimination of weak or uncoupled synaptic elements suggesting that the complement pathway contributes to the stability of the newly formed circuits (Schafer et al., 2012, 2013; Stephan et al., 2012; Stevens et al., 2007). Complement C3 is upregulated in aging, neurodegenerative disorders (Carpanini et al., 2019; Prinz et al., 2019), and epilepsy (Wyatt-Johnson and Brewster, 2020), suggesting that complement-mediated synapse elimination may be a mechanism associated with the microglia-neuronal contact-mediated regulation of neuronal activity discussed above. Clinical studies revealed higher levels of circulating C3 (Başaran et al., 1994), terminal complement proteins, and Factor H (Kopczynska et al., 2018), as well as increases in C1q and C3 mRNA and C3b protein levels in the brain (Aronica et al., 2007; Wyatt-Johnson and Brewster, 2020; Wyatt et al., 2017). In preclinical models, complement mRNA and proteins are upregulated in the brain both acutely after prolonged seizures and in the chronic epilepsy stage (Aronica et al., 2007; Schartz et al., 2016, 2018). In epileptic rats, increased C3 levels are significantly correlated with the frequency of spontaneous seizures and parallel deficits in hippocampal-dependent memory deficits (Kharatishvili et al., 2014; Schartz et al., 2018, 2019). In addition, significant correlations between increased levels of C3 protein and decreased levels of the synaptic markers PSD95, VGlut, and VGat were found in hippocampi of rats subjected to SE (Schartz et al., 2019). Studies done blocking downstream complement components such as C3 and C5 (and their receptors) reduced seizure frequency or severity (Libbey et al., 2010; Buckingham et al., 2014; Benson et al., 2015b), though suppressing C1q signaling upstream of C3 did not improve memory defects induced by SE (Schartz et al., 2019), and C1q KO mice developed epilepsy (DeVos et al., 2013; Chu et al., 2010). Taken together, this evidence suggests that microglia-neuronal contacts that occur following acute seizures or in chronic epilepsy may result in synaptic remodeling. Potential protective or detrimental outcomes would depend on the type of synapse targeted, brain region affected, and age (Andoh et al., 2019). Further investigation is required to fully understand how microglia-neuronal interactions modulate the epileptic brain and what immune molecules may be underlying the changes in synaptic activity.
Microglial Pro- and Anti-inflammatory Molecular Equilibriums in Experimental Epilepsy
Microglia with reactive properties are widely found in models of genetic and acquired epilepsies (Wyatt-Johnson and Brewster, 2020; Brewster, 2019; Eyo et al., 2016; Sabilallah et al., 2016). It is well established that microglia develop an inflammatory phenotype during and after seizures (van Vliet et al., 2018; Devinsky et al., 2013), and more recent studies support that this is coupled with dysfunction in microglial phagocytic activity (Abiega et al., 2016; Zhao et al., 2018; Sierra-Torre et al., 2020; Luo et al., 2016; Koizumi et al., 2007). In a rat model of acquired epilepsy triggered by pilocarpine-induced SE, drastic alterations in microglial morphologies and densities in the hippocampus that varied in a spatiotemporal-dependent manner were found during the period of epileptogenesis between 4 h and 5 weeks after SE (Wyatt-Johnson et al., 2017; Schartz et al., 2016). Early after SE (4 h), microglia developed a hypertrophied morphology that paralleled significant increases in the levels of TNFα and IL-6 measured in hippocampal homogenates. However, at 2 weeks post-SE, when microgliosis peaked in the hippocampus, or at 5 weeks when the animals reached chronic epilepsy, no significant changes were found in at least 24 different cytokines (Schartz et al., 2016), suggesting that the functions of microglia evolve during epileptogenesis. In agreement with these findings, studies reported increases in the pro-inflammatory cytokines IL-1β, TNFα, IL-6, and IL-12, as well as the anti-inflammatory molecules Arg1, Ym1, IL-10, and IL-4 in isolated microglia at 3 days after pilocarpine-induced seizures, all not modified in the chronic epilepsy phase (Benson et al., 2015a). In comparative analyses using the KA model of mesial (m)TLE, the pro-inflammatory cytokines at 3 days after SE did not last into the chronic phase (Benson et al., 2015a). The presence of reactive microglia with inflammatory signatures has been extensively reported in different types of epilepsies (see van Vliet et al., 2018), including TLE (Broekaart et al., 2018; Schartz et al., 2016, 2019), focal cortical dysplasia (FCD) (Nguyen et al., 2015), tuberous sclerosis complex (TSC) (Nguyen et al., 2019; Zhang et al., 2018; Zimmer et al., 2020), models of encephalitis (Pitsch et al., 2021; Bell et al., 2020), and developmental seizures (Patterson et al., 2015). While causal mechanisms underlying microglial inflammatory responses are not definitively known, existing evidence suggests that enhanced neuronal firing, accompanied by excitotoxicity, neuronal loss, and interplays of peripheral immune cells (i.e., CD8 + T-cells and monocytes) at the neurovascular unit can trigger the activation of microglial cells (Pitsch et al., 2021; Varvel et al., 2016). This microglia-mediated inflammation may have beneficial (i.e., tissue repair, neuroprotection) or detrimental (i.e., neurodegeneration) consequences to the surrounding neural networks depending on whether inflammation is short- or long-lasting (DiSabato et al., 2016) and the brain regions affected (i.e., epileptogenic foci and seizure-propagating).
In parallel to inflammatory changes, several studies demonstrated alterations in microglia phagocytic activity that may be pro-epileptogenic in genetic and acquired models of epilepsy (see Wyatt-Johnson and Brewster, 2020, for review). A recent study showed that deletion of the TSC1 gene specifically in microglial cells (TSC1CX3CR1CKO) caused enhanced activation of the mechanistic target of rapamycin (mTOR) in microglial cells and recurrent unprovoked seizures (Zhao et al., 2018). The microglial population in the TSC1CX3CR1CKO mice displayed amoeboid and bushy morphologies, decreased levels of IL-1β, IL-6, TNFα, and iNOS, among other inflammatory molecules, and increased phagocytic activity (Zhao et al., 2018), thereby suggesting that noninflammatory phagocytic microglia may underlie epilepsy in this TSC model. This study also showed that microglial mTOR signaling is necessary for proper microglial phagocytic functions as mTOR-deficient microglia showed a phagocytosis impairment, and the microglial mTOR-deficient mice developed severe spontaneous seizures (Zhao et al., 2020). Thus, it is possible that disrupting the fine balance of inflammatory/phagocytic properties of microglia can result in hyperactive neuronal networks. However, it is noted that a different study utilizing similar TSC1CX3CR1-CreCKO mice showed that enhanced mTOR signaling increased microglial cell size, although spontaneous seizures did not develop (Zhang et al., 2018; Wong, 2019). Yet a recent study interrogating epigenetic regulation of microglial clearance activity showed that mice lacking the polycomb repressive complex 2 (PRC2) molecule, which controls the expression of microglial-specific clearance genes, developed epilepsy (Ayata et al., 2018). This evidence further suggests that increased microglial phagocytosis activity may contribute to the generation of spontaneous recurrent seizures.
Abnormal microglia-mediated phagocytosis and its role in epileptogenesis and chronic epilepsy were also reported in animal models of acquired epilepsy (Wyatt-Johnson and Brewster, 2020). Following KA-induced seizures in the rat, microglial phagocytic activity was increased in the hippocampal CA3 region, and this was mediated by UDP/P2Y6-dependent signaling (Koizumi et al., 2007). In contrast, a microglial phagocytic deficiency was found in the hippocampal dentate gyrus (DG) following intrahippocampal administration of KA in mice. In this study, the molecular signatures of fluorescence-activated cell sorting (FACS)-isolated microglia included decreased mRNA levels of the phagocytosis receptors Trem2, MerTK, CR3, and GPR34, and increased levels of the purinergic receptors P2Y12, P2Y6, and P2X4, along with cytokines such as IL-1β, IL-6, and TNFα (Abiega et al., 2016). These molecular changes occurred in association with a decrease in both microglial phagocytic activity and surveillance/motility within the DG area. As a result, newborn apoptotic cells were not phagocytosed and accumulated, possibly potentiating detrimental and pro-epileptogenic inflammatory responses (Abiega et al., 2016). A microglial phagocytosis impairment in the hippocampal DG was also observed in a mouse model of progressive myoclonus epilepsy type 1 (Sierra-Torre et al., 2020). The differences in microglial phagocytic responses in the hippocampal CA3 versus DG areas are likely due to the extent of regional vulnerability to neuronal death, which can trigger increases in microglial P2Y6 receptors, thereby enhancing phagocytic activity of these cells (Koizumi et al., 2007). Interestingly, it has also been shown that microglial cells can engulf and phagocytose “healthy” nonapoptotic cells in the hippocampal DG area during epileptogenesis (Abiega et al., 2016; Luo et al., 2016). This process is known as phagoptosis (Brown and Neher, 2014) and may contribute to exacerbated neuronal loss in epilepsy. These observations suggest that seizures per se can cause long-lasting disruptions in the microglial phagocytic activity that are region-dependent. These findings, along with evidence from human drug-resistant epilepsies, support that dysregulation of the equilibrium of microglial phagocytic and inflammatory functions may be epileptogenic.
Microglia-Pericytes Perivascular Assembly and Reactivity during Seizures: Experimental and Clinical Evidence
The reported redistribution of mural cells (smooth muscle cells and pericyte) at the perivascular compartment is intertwined with dynamic and time-dependent modifications of astrocytes and microglial cells. Evidence exists for CNS diseases, in particular for experimental autoimmune encephalopathy (EAE) (Merlini et al., 2019; Davalos et al., 2012) or AD (Ryu and McLarnon, 2009; Halder and Milner, 2019), where the perivascular accumulation of microglial cells topographically overlapped with blood fibrinogen entry into the brain parenchyma, promoting neuronal and dendritic spine damage or elimination. In an experimental TLE model, GFAP-labeled astrocytes, IBA1-positive microglia, and NG2DsRed pericytes spatially converge to form a complex, multicellular and dynamic assembly at the outer wall of the BBB (Garbelli et al., 2015; Giannoni et al., 2018; Klement et al., 2018). In the hippocampal epileptic foci (ih KA), activated microglial cells accrue the outer BBB wall, possibly exerting a phagocytic action towards damaged pericytes (Fig. 29–1; Klement et al., 2018).
In these regions, increased PDGFRβ expression indicated a fibrotic-like mesh developing around the capillaries (Klement et al., 2019). Partaking in this multicellular perivascular reorganization, abnormal collagen deposits could confer stiffness to specific vascular portions, and exacerbate neuroinflammation (Klement et al., 2019). Evidence of collagen accumulation also derives from human TLE (Klement et al., 2019; Kastanauskaite et al., 2009). These data suggest the presence of fibrotic elements, indicating a possible perivascular scar at sites of BBB leakages, the latter identified by using intravenous fluorescein during epileptogenesis and at spontaneous seizures (Klement et al., 2019). Furthermore, a pathological matrix MMPs production was reported during epileptogenesis, and the use of an MMP2/9 inhibitor (IPR-179) reduced seizures in experimental models, also curbing seizure-associated cognitive defects (Broekaart et al., 2021). Further indicating the presence of pro-fibrotic events is an increase of parenchymal PDGFRβ reactivity during seizures in the hippocampal epileptic focus (Dias et al., 2018). The exact origin of PDGFRβ cells during disease conditions is debated, possibly originating from the activated or damaged pericytes detaching from the vascular wall (Dias et al., 2018; Goritz et al., 2011; Cheng et al., 2018) or, otherwise, proliferating from an existing pool of fibroblasts (Soderblom et al., 2013; Riew et al., 2018). In any case, this perivascular multicellular activation could promote localized rigid scars, with negative implications to the control of vascular tone (Arango-Lievano et al., 2018) and the regulation of neurovascular coupling during seizures (Prager et al., 2019b). The presence of perivascular microglial cells and pericyte damage was also reported in human TLE and FCD type IIb brain tissues (Garbelli et al., 2015; Klement et al., 2018; Milesi et al., 2014). In TLE with neuronal loss and gliosis in the hippocampus (hippocampal sclerosis; HS), microglial cells (IBA1 and activated HLA) were found around the capillaries, as compared to nonlesional TLE (Klement et al., 2018). Alongside, PDGFRβ+ immunoreactivity was evident at the capillary level in TLE-HS. In FCD IIb brain specimens, perivascular pericytes and microglia (IBA1, HLA) were reported in the core of the dysplastic regions, as compared to the adjacent normal-appearing areas (Garbelli et al., 2015; Klement et al., 2018).
Less obvious is whether pathological glio-vascular variations exist in seizure networks and in the absence of a clear-cut inflammatory phenotype or tissue lesion. The latter notion is relevant as varying physiological neuronal activities can finely tune glio-vascular gene expression patterns (Pulido et al., 2020), while nonlesional seizure-propagating brain regions can display distinct imaging signatures (Boux et al., 2021). Remodeling of capillary pericytes was shown to occur in the seizure propagating hippocampi of a TLE mouse model, in areas characterized by the absence of tissue sclerosis or apparent histological signs of inflammation (Boux et al., 2021). An increase in the MRI parameters apparent diffusion coefficient, blood volume fraction, and BBB permeability occurring in the contralateral hippocampus (of the unilateral ih KA model) correlated with an augmented CD13 pericytes length, an indication of cell hypertrophy or a previous angiogenesis (Boux et al., 2021). Taken together, these data indicate the presence of discrete pathological signatures generated by seizure propagation routes, independent from the existence of histological, tissue-level, lesions.
Microglial Profiles in Human Drug-Resistant Epilepsies
The heterogeneity of microglial morphological and immune signatures is evident in brain tissues surgically resected from patients with drug-resistant epilepsies, including FCD, TSC, Rasmussen encephalitis (RE), and mTLE-HS. Histological analyses of these tissues support a microglial pathology in epilepsy that is first seen through morphological alterations. For example, microglia with hypertrophied and amoeboid shapes are present in cortices and hippocampi of FCD, RE, TSC, and mTLE patients, suggesting a reactive phenotype (Wyatt et al., 2017; Wirenfeldt et al., 2009; Boer et al., 2008a, 2008b; Broekaart et al., 2018; Xu et al., 2018; Altmann et al., 2021). Microglia with these “reactive” shapes are more abundant in areas closer to the seizure focus as well as within microlesions characterized by high spiking activity (Dachet et al., 2015; Morin-Brureau et al., 2018). In human mTLE, amoeboid microglia were found in hippocampal areas CA1 and CA3 associated with high neuronal death when compared to the DG or subiculum where the neuronal loss was less prominent (Morin-Brureau et al., 2018). In addition, a recent study utilizing a systems-level analysis of image-based cortical structural profiles along with gene expression data from human epilepsy cases also showed a high density of microglial cells in areas characterized by reduced cortical thickness (Altmann et al., 2021). Even though the presence of reactive microglia in human epileptic brains correlates with areas of decreased neuronal densities and increased vascularity (Dachet et al., 2015; Morin-Brureau et al., 2018; Altmann et al., 2021), and microglia have been found in close association with injured dendrites (Wyatt et al., 2017; Wirenfeldt et al., 2009), dysmorphic neurons, cortical tubers (Boer et al., 2008a, 2008b), and apoptotic cells (Abiega et al., 2016), their role in the pathology of epilepsy is not definitively known.
To understand exactly how microglia may contribute to hyperexcitable circuits, it is necessary to obtain information on their molecular profiles. In human epileptic tissues, immunohistological studies showed that amoeboid microglia were abundant in MHCII, phagocytic-associated molecules CD68 and CD16/32a (Morin-Brureau et al., 2018), complement proteins (Aronica et al., 2007; Wyatt et al., 2017), and inflammatory molecules (Morin-Brureau et al., 2018; Hiragi et al., 2018; van Vliet et al., 2018), suggesting a reactive phenotype. In mTLE, both amoeboid and ramified microglia expressed the purinergic receptor P2Y12 and retracted their processes similarly in response to ADP exposure in culture (Morin-Brureau et al., 2018). This interesting finding contrasts the ADP/ATP-triggered extension of microglial processes seen under physiological conditions in preclinical models (Eyo and Wu, 2019). Furthermore, transcriptome analyses from whole-tissue biopsy homogenates showed differentially expressed microglial-specific genes that support altered microglial-related molecular signatures in areas of high and low spiking activity or neuronal death (Dachet et al., 2015; Morin-Brureau et al., 2018). High expression of markers for microglial M1 and M2 polarization (with higher M1/M2 ratio) (Dachet et al., 2015) and upregulations in levels of cytokines such as IL-10, IL-1β, CXCL8, CXCR4, and CCR5 were found in the epileptic tissues (Morin-Brureau et al., 2018). Other changes of microglial-related proteins recently described in drug-resistant epilepsies include increases in C1q, C3b, and MerTK (Wyatt et al., 2017) and decreases in Protein S, Trem2, CD47, and SIRP-α (Wyatt et al., 2017; Sun et al., 2016).
To specifically study microglia in surgically resected samples (biopsies) from epileptic patients, recent studies utilized high-throughput techniques based on combinations of single-cell RNA sequencing (scRNA-seq), multiplexed single-cell mass cytometry (CyTOF), FACS, and bioinformatics. The results support that the biological profiles of microglia vary in a spatial- and age-dependent manner in the human brain (Gosselin et al., 2017; Galatro et al., 2017; Sankowski et al., 2019; Masuda et al., 2019; Bottcher et al., 2019; Brewster, 2019). These studies used perilesional tissues from epilepsy surgeries along with human brain autopsy samples, among others, and while not specifically investigating the pathology of epilepsy, their findings can help elucidate specific microglial functions that are particularly relevant to the human epileptic brain. RNA-seq of FACS-isolated microglial cells demonstrated a robust expression of P2Y12, Trem2, and the CX3C chemokine receptor 1 (CX3CR1) along with the complement proteins C3 and C1q in these cells (Gosselin et al., 2017). Microglial cells extracted from epilepsy cases and processed through CyTOF showed a robust expression of the MHCII surface receptor human leukocyte antigen-DR isotype (HLA-DR), Trem2, P2Y12, interferon regulatory factor 8 (IRF8), and CD68 (Bottcher et al., 2019). In parallel, this study showed diverse microglial immune signatures across five different brain regions, with areas such as the subventricular zone containing microglia rich in CD68 along with the cyclins A and B1, and the nuclear protein Ki-67, which participate in cell growth and proliferation (Bottcher et al., 2019). Other regional changes include higher expression of CD68 and HLA-DR in microglia localized to the white matter compared to those in the grey matter (Sankowski et al., 2019). Comparative analyses of microglial immune profiles between human epilepsy biopsies and mice showed higher biological heterogeneity with more activated phenotypes in humans (Masuda et al., 2019), which could be related to not only intrinsic species differences but also to a history of drug-resistant seizures. Taken together these data support that microglial cells in the human epileptic brain have diverse biological features which may result in altered surveillance, inflammatory, and phagocytic roles that vary by brain region, as well as by the extent of neuronal density and seizure activity.
Additional evidence supporting a role for microglia in epilepsy comes from a study describing two cases in which homozygous mutations in the CSF1R gene, a signaling molecule that regulates microglial survival (Elmore et al., 2014), caused brain malformations, leukoencepatholpathy, and epilepsy (Oosterhof et al., 2019). Loss-of-function mutations in CSF1R are typically associated with reduced numbers of microglia in individuals diagnosed with adult-onset leukoencepatholpathy with axonal spheroids and pigmented glia (ALSP), which develop epilepsy in adulthood (Oosterhof et al., 2019). A study describes two pediatric cases in which CSFR1 mutations were associated with both the development of intractable epilepsy in an 8-month-old infant that died prematurely at 10 months, and with childhood epilepsy and developmental regression that occurred at age 12 years in the second individual. Interestingly, histological analysis showed a lack of microglial cells in the brain of the infant, suggesting that microglia are critically important for postnatal brain development and to modulate/control neural activity. These findings in human are also supported by pharmacogenetic manipulations of CSF1R signaling in preclinical models (discussed below).
Pharmacological Entry Points: Focus on Pericytes and Microglia
To study how microglia may contribute to the neuropathology and the pathophysiology of epilepsy, recent studies focused on pharmacogenetic manipulations of the CSF1R signaling cascade. CSF1R is mainly found in microglia and functions to control the survival of these cells (Ulland et al., 2015; Oosterhof et al., 2018; Elmore et al., 2014; Waisman et al., 2015). A systems-level framework analysis for target drug discovery using mRNA samples extracted from hippocampi of pilocarpine-treated mice identified CSF1R as a potential target for epilepsy treatment (Srivastava et al., 2018). In this study, CSF1R signaling was suppressed with the drug Plexxikon (PLX) 3397, which was administered for 2 weeks to chronically epileptic mice. PLX3397 caused a significant decrease in the microglial population in both hippocampus and cortex, and a reduction in seizure frequency and duration (Srivastava et al., 2018), suggesting that microglia contribute to the maintenance of an epileptic brain. In contrast, other studies have shown that depletion of microglia can worsen the seizure pathology. For example, microglial suppression starting at least 3 weeks after SE aggravated the seizure severity in chronically epileptic mice (Wu et al., 2020), and microglial depletion before the administration of chemoconvulsants/stimulants (KA, picrotoxin, or D1 agonist) or a Theiler’s murine encephalomyelitis virus resulted in more severe seizures (Wu et al., 2020; Badimon et al., 2020; Sanchez et al., 2019). Recent studies support that the timing of microglial suppression can impact the outcome of seizure burden and associated cognitive comorbidities in rodent models of SE and acquired TLE (Altmann et al., 2021; Di Nunzio et al., 2021; Wyatt-Johnson et al., 2021). PLX3397 treatment given immediately after SE and for 3 consecutive weeks during the period of epileptogenesis significantly decreased hippocampal microgliosis without altering hippocampal-dependent spatial learning and memory defects in rats (Wyatt-Johnson et al., 2021). Similarly, others showed that treatment with the CSF1R suppressor GW2580 during SE-induced epileptogenesis did not alter either the seizure or memory pathology (Di Nunzio et al., 2021). However, when GW2580 was given to mice with fully established epilepsy, seizure severity decreased and cognitive performance improved, suggesting that microglia may have different functions in developing and established epileptic networks (Di Nunzio et al., 2021). However, existing studies do not account for the heterogeneous populations of microglia that exist throughout the brain, with different molecular signatures and biological functions that may serve beneficial or detrimental effects in the epileptic brain. Therefore, additional research is required to differentiate the impact that alterations in different microglial functions (surveillance, inflammatory, and phagocytic) have on epilepsy pathophysiology. Once these are identified, more efficacious treatments that specifically target the detrimental signaling can be developed.
When studying the functional impact of pericytes in seizure conditions, PDGFRβ was targeted to modify the pathological neurovascular sequelae of SE in vivo or during epileptiform conditions in vitro (Klement et al., 2019; Arango-Lievano et al., 2018). A restorative or a protective effect was reported when the PDGFRβ agonist ligand PDGF-BB was administered early post-SE in mice (Arango-Lievano et al., 2018). However, during epileptogenesis leading to spontaneous seizures, the PDGFRβ increase was associated with a scarring process, advocating for the use of a receptor blocker, such as imatinib, to curb inflammation (Klement et al., 2019). From this evidence we understand that the outcome of PDGFRβ modulation may be contingent on the disease stage, that is, early post brain insults (SE, head trauma, ischemic stroke) pericytes necessitate support via PDGFRβ activation, whereas a pro-inflammatory involvement of PDGFRβ may emerge during chronic stages, perhaps necessitating inhibition. Because of the rapidly evolving pro- and anti-inflammatory equilibriums and dynamics during epileptogenesis, targeting a specific player may be oversimplistic, considering the multiple roles that neuro-glio-vascular cells have as a function of time and region.
Conclusion: Refining Timing and Targets for Pharmacological Interventions
Experimental and clinical epilepsies can be considered neuro-glio-vascular pathological conditions. This notion is important because the next antiseizure solutions could include the combination of neuronal and glio-vascular molecules, targeting multiple and interindependent culprits of seizures. Adding a level of complexity, time for pharmacological intervention needs to be carefully planned, because of the varying, even contrasting, functions that a specific cell type can acquire during disease progression. Based on contemporary evidence, we identify several research opportunities and questions: (1) what are the temporal dynamics governing the pericyte and microglial mediated anti- and pro-inflammatory cellular (dys) equilibrium unfolding during seizure progression? Recent evidence identified insufficient engagement of endogenous anti-inflammatory pathways (i.e., glucocorticoid receptors/annexin A1, n-3 docosapentaenoic acid) during experimental seizures, enabling inflammation (Frigerio et al., 2018; Zub et al., 2019). Promoting resolution of inflammation by boosting endogenous mechanisms could represent a new therapeutic frontier (Fullerton and Gilroy, 2016); (2) what are the roles of microglia activation and differential redistribution toward the capillaries and neurons? Recent evidence demonstrated that microglial cells can accumulate at the perivascular compartment in response to seizures, in models of autoimmune encephalitis, and AD (Bauer et al., 2008; Davalos et al., 2012; Giannoni et al., 2018; Halder and Milner, 2019; Klement et al., 2018, 2019; Merlini et al., 2019; Ryu and McLarnon, 2009), overlapping with regions of BBB permeability, as well as in areas of high neuronal spiking and cell death. At the same time, neuronal hyperexcitability and seizures trigger increases in the physical interactions between microglial processes and different neuronal structures (i.e., soma, dendrites), whose exact roles, pro- or anti-epileptogenic, are yet to be determined. Similarly, whether alterations in levels of phagocytosis-related molecules and microglial phagocytic actions (enhanced or impaired) as well as if the associated release of pro- or anti-inflammatory molecules are a cause or a result of seizures is not known. To determine how pericytes and microglial cells contribute to epilepsy, future studies must perform comprehensive spatiotemporal mapping of biological profiles of these cell types as well as the interactions with neurons and blood vessels both during and after acute seizures and in the chronic epilepsy stage. Based on recent evidence described throughout this review, populations of pericytes and microglia with different profiles that may be beneficial or detrimental can coexist in the epileptic brain. This suggests that time-targeted, regional, and cell-specific interventions may represent a strategy that can reduce the harmful signals or enhance the beneficial actions of antiepileptic drugs (Ghosh et al., 2011).
Acknowledgments
This work was supported by NIH 1R01NS096234 (ALB); ANR Epicyte, ANR EpiCatcher, ANR HepatoBrain, and ANR Neu-Vasc (NM).
Disclosure Statement
The authors declare no relevant conflicts.
References
- Abiega O, Beccari S, Diaz-Aparicio I, Nadjar A, Laye S, Leyrolle Q, Gomez-Nicola D, Domercq M, Perez-Samartin A, Sanchez-Zafra V, Paris I, Valero J, Savage JC, Hui CW, Tremblay ME, Deudero JJ, Brewster AL, Anderson AE, Zaldumbide L, Galbarriatu L, Marinas A, Vivanco M, Matute C, Maletic-Savatic M, Encinas JM, Sierra A. Neuronal hyperactivity disturbs atp microgradients, impairs microglial motility, and reduces phagocytic receptor expression triggering apoptosis/microglial phagocytosis uncoupling. PLoS Biol. 2016;14(5):e1002466. doi: 10.1371/journal.pbio.1002466. PubMed PMID: 27228556. [PMC free article: PMC4881984] [PubMed: 27228556]
- Altmann A, Ryten M, Di Nunzio M, Ravizza T, Tolomeo D, Reynolds RH, Somani A, Bacigaluppi M, Iori V, Micotti E, Di Sapia R, Cerovic M, Palma E, Ruffolo G, Botia JA, Absil J, Alhusaini S, Alvim MKM, Auvinen P, Bargallo N, Bartolini E, Bender B, Bergo FPG, Bernardes T, Bernasconi A, Bernasconi N, Bernhardt BC, Blackmon K, Braga B, Caligiuri ME, Calvo A, Carlson C, Carr SJA, Cavalleri GL, Cendes F, Chen J, Chen S, Cherubini A, Concha L, David P, Delanty N, Depondt C, Devinsky O, Doherty CP, Domin M, Focke NK, Foley S, Franca W, Gambardella A, Guerrini R, Hamandi K, Hibar DP, Isaev D, Jackson GD, Jahanshad N, Kalviainen R, Keller SS, Kochunov P, Kotikalapudi R, Kowalczyk MA, Kuzniecky R, Kwan P, Labate A, Langner S, Lenge M, Liu M, Martin P, Mascalchi M, Meletti S, Morita-Sherman ME, O’Brien TJ, Pariente JC, Richardson MP, Rodriguez-Cruces R, Rummel C, Saavalainen T, Semmelroch MK, Severino M, Striano P, Thesen T, Thomas RH, Tondelli M, Tortora D, Vaudano AE, Vivash L, von Podewils F, Wagner J, Weber B, Wiest R, Yasuda CL, Zhang G, Zhang J, Group EN-EW, Leu C, Avbersek A, Epi PGXC, Thom M, Whelan CD, Thompson P, McDonald CR, Vezzani A, Sisodiya SM. A systems-level analysis highlights microglial activation as a modifying factor in common epilepsies. Neuropathol Appl Neurobiol. 2022 Feb;48(1):e12758. Epub 2021/08/14. doi: 10.1111/nan.12758. PubMed PMID: 34388852. [PMC free article: PMC8983060] [PubMed: 34388852]
- Andoh M, Ikegaya Y, Koyama R. Synaptic pruning by microglia in epilepsy. J Clin Med. 2019 Dec 9;8(12):2170. doi: 10.3390/jcm8122170. PubMed PMID: 31818018. [PMC free article: PMC6947403] [PubMed: 31818018]
- Arango-Lievano M, Boussadia B, De Terdonck LDT, Gault C, Fontanaud P, Lafont C, Mollard P, Marchi N, Jeanneteau F. Topographic reorganization of cerebrovascular mural cells under seizure conditions. Cell Rep. 2018;23(4):1045–59. doi: 10.1016/j.celrep.2018.03.110. PubMed PMID: 29694884. [PubMed: 29694884]
- Aronica E, Boer K, van Vliet EA, Redeker S, Baayen JC, Spliet WG, van Rijen PC, Troost D, da Silva FH, Wadman WJ, Gorter JA. Complement activation in experimental and human temporal lobe epilepsy. Neurobiol Dis. 2007;26(3):497–511. doi: 10.1016/j.nbd.2007.01.015. PubMed PMID: 17412602. [PubMed: 17412602]
- Attwell D, Mishra A, Hall CN, O’Farrell FM, Dalkara T. What is a pericyte? J Cereb Blood Flow Metab. 2016;36(2):451–5. doi: 10.1177/0271678X15610340. PubMed PMID: 26661200. [PMC free article: PMC4759679] [PubMed: 26661200]
- Ayata P, Badimon A, Strasburger HJ, Duff MK, Montgomery SE, Loh YE, Ebert A, Pimenova AA, Ramirez BR, Chan AT, Sullivan JM, Purushothaman I, Scarpa JR, Goate AM, Busslinger M, Shen L, Losic B, Schaefer A. Epigenetic regulation of brain region-specific microglia clearance activity. Nat Neurosci. 2018;21(8):1049–60. doi: 10.1038/s41593-018-0192-3. PubMed PMID: 30038282. [PMC free article: PMC6090564] [PubMed: 30038282]
- 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 YC, Loh YE, 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. Negative feedback control of neuronal activity by microglia. Nature. 2020;586(7829):417–23. doi: 10.1038/s41586-020-2777-8. PubMed PMID: 32999463. [PMC free article: PMC7577179] [PubMed: 32999463]
- Bar-Klein G, Lublinsky S, Kamintsky L, Noyman I, Veksler R, Dalipaj H, Senatorov VV, Jr., Swissa E, Rosenbach D, Elazary N, Milikovsky DZ, Milk N, Kassirer M, Rosman Y, Serlin Y, Eisenkraft A, Chassidim Y, Parmet Y, Kaufer D, Friedman A. Imaging blood-brain barrier dysfunction as a biomarker for epileptogenesis. Brain. 2017;140(6):1692–705. Epub 2017/04/27. doi: 10.1093/brain/awx073. PubMed PMID: 28444141. [PubMed: 28444141]
- Barnum SR. Complement: A primer for the coming therapeutic revolution. Pharmacol Ther. 2017;172:63–72. doi: 10.1016/j.pharmthera.2016.11.014. PubMed PMID: 27914981. [PubMed: 27914981]
- Başaran N, Hincal F, Kansu E, Cǧer A. Humoral and cellular immune parameters in untreated and phenytoin- or carbamazepine-treated epileptic patients. International Journal of Immunopharmacology. 1994;16(12):1071–7. doi: https://doi
.org/10.1016 /0192-0561(94)90087-6. [PubMed: 7705969] - Bauer S, Koller M, Cepok S, Todorova-Rudolph A, Nowak M, Nockher WA, Lorenz R, Tackenberg B, Oertel WH, Rosenow F, Hemmer B, Hamer HM. Nk and cd4+ t cell changes in blood after seizures in temporal lobe epilepsy. Exp Neurol. 2008;211(2):370–7. Epub 2008/04/05. doi: 10.1016/j.expneurol.2008.01.017. PubMed PMID: 18387608. [PubMed: 18387608]
- Beamer E, Conte G, Engel T. Atp release during seizures—a critical evaluation of the evidence. Brain Res Bull. 2019;151:65–73. doi: 10.1016/j.brainresbull.2018.12.021. PubMed PMID: 30660718. [PubMed: 30660718]
- Bell LA, Wallis GJ, Wilcox KS. Reactivity and increased proliferation of ng2 cells following central nervous system infection with theiler’s murine encephalomyelitis virus. J Neuroinflamm. 2020;17(1):369. doi: ARTN 36910.1186/s12974-020-02043-5. PubMed PMID: WOS:000595994700001. [PMC free article: PMC7713670] [PubMed: 33272299]
- Ben-Ari Y, Khalilov I, Kahle KT, Cherubini E. The gaba excitatory/inhibitory shift in brain maturation and neurological disorders. Neuroscientist. 2012;18(5):467–86. doi: 10.1177/1073858412438697. PubMed PMID: 22547529. [PubMed: 22547529]
- Benson MJ, Manzanero S, Borges K. Complex alterations in microglial m1/m2 markers during the development of epilepsy in two mouse models. Epilepsia. 2015a;56(6):895–905. doi: 10.1111/epi.12960. PubMed PMID: 25847097. [PubMed: 25847097]
- Benson MJ, Thomas NK, Talwar S, Hodson MP, Lynch JW, Woodruff TM, Borges K. A novel anticonvulsant mechanism via inhibition of complement receptor c5ar1 in murine epilepsy models. Neurobiol Dis. 2015b;76:87–97. doi: 10.1016/j.nbd.2015.02.004. PubMed PMID: 25681535. [PubMed: 25681535]
- Boer K, Jansen F, Nellist M, Redeker S, van den Ouweland AM, Spliet WG, van Nieuwenhuizen O, Troost D, Crino PB, Aronica E. Inflammatory processes in cortical tubers and subependymal giant cell tumors of tuberous sclerosis complex. Epilepsy Res. 2008a;78(1):7–21. doi: 10.1016/j.eplepsyres.2007.10.002. PubMed PMID: 18023148. [PubMed: 18023148]
- Boer K, Troost D, Jansen F, Nellist M, van den Ouweland AM, Geurts JJ, Spliet WG, Crino P, Aronica E. Clinicopathological and immunohistochemical findings in an autopsy case of tuberous sclerosis complex. Neuropathology. 2008b;28(6):577–90. doi: 10.1111/j.1440-1789.2008.00920.x. PubMed PMID: 18410267. [PubMed: 18410267]
- Bottcher C, Schlickeiser S, Sneeboer MAM, Kunkel D, Knop A, Paza E, Fidzinski P, Kraus L, Snijders GJL, Kahn RS, Schulz AR, Mei HE, Psy NBB, Hol EM, Siegmund B, Glauben R, Spruth EJ, de Witte LD, Priller J. Human microglia regional heterogeneity and phenotypes determined by multiplexed single-cell mass cytometry. Nat Neurosci. 2019;22(1):78–90. doi: 10.1038/s41593-018-0290-2. PubMed PMID: 30559476. [PubMed: 30559476]
- Boux F, Forbes F, Collomb N, Zub E, Maziere L, de Bock F, Blaquiere M, Stupar V, Depaulis A, Marchi N, Barbier EL. Neurovascular multiparametric mri defines epileptogenic and seizure propagation regions in experimental mesiotemporal lobe epilepsy. Epilepsia. 2021;62(5):1244–55. Epub 2021/04/06. doi: 10.1111/epi.16886. PubMed PMID: 33818790. [PubMed: 33818790]
- Brewster AL. Human microglia seize the chance to be different. Epilepsy Curr. 2019;19(3):190–2. doi: 10.1177/1535759719843299. PubMed PMID: 31032668. [PMC free article: PMC6610380] [PubMed: 31032668]
- Brewster AL, Lugo JN, Patil VV, Lee WL, Qian Y, Vanegas F, Anderson AE. Rapamycin reverses status epilepticus-induced memory deficits and dendritic damage. PLoS One. 2013;8(3):e57808. Epub 2013/03/29. doi: 10.1371/journal.pone.0057808. PONE-D-12-26653 [pii]. PubMed PMID: 23536771. [PMC free article: PMC3594232] [PubMed: 23536771]
- Broekaart DW, Bertran A, Jia S, Korotkov A, Senkov O, Bongaarts A, Mills JD, Anink JJ, Seco J, Baayen JC, Idema S, Chabrol E, Becker AJ, Wadman WJ, Tarrago T, Gorter JA, Aronica E, Prades R, Dityatev A, van Vliet EA. The matrix metalloproteinase inhibitor ipr-179 has antiseizure and antiepileptogenic effects. J Clin Invest. 2021;131(1):e138332. doi: 10.1172/JCI138332. PubMed PMID: 33141761. [PMC free article: PMC7773344] [PubMed: 33141761]
- Broekaart DWM, Anink JJ, Baayen JC, Idema S, de Vries HE, Aronica E, Gorter JA, van Vliet EA. Activation of the innate immune system is evident throughout epileptogenesis and is associated with blood-brain barrier dysfunction and seizure progression. Epilepsia. 2018;59(10):1931–44. doi: 10.1111/epi.14550. PubMed PMID: 30194729. [PubMed: 30194729]
- Brown GC, Neher JJ. Microglial phagocytosis of live neurons. Nat Rev Neurosci. 2014;15(4):209–16. doi: 10.1038/nrn3710. PubMed PMID: 24646669. [PubMed: 24646669]
- Buckingham SC, Ramos TN, Barnum SR. Complement c5-deficient mice are protected from seizures in experimental cerebral malaria. Epilepsia. 2014;55(12):e139–e42. doi: 10.1111/epi.12858. [PMC free article: PMC4285666] [PubMed: 25385326]
- Carpanini SM, Torvell M, Morgan BP. Therapeutic inhibition of the complement system in diseases of the central nervous system. Front Immunol. 2019;10:362. doi: 10.3389/fimmu.2019.00362. PubMed PMID: 30886620. [PMC free article: PMC6409326] [PubMed: 30886620]
- Chen Z, Jalabi W, Hu W, Park HJ, Gale JT, Kidd GJ, Bernatowicz R, Gossman ZC, Chen JT, Dutta R, Trapp BD. Microglial displacement of inhibitory synapses provides neuroprotection in the adult brain. Nat Commun. 2014;5:4486. doi: 10.1038/ncomms5486. PubMed PMID: 25047355. [PMC free article: PMC4109015] [PubMed: 25047355]
- Cheng J, Korte N, Nortley R, Sethi H, Tang Y, Attwell D. Targeting pericytes for therapeutic approaches to neurological disorders. Acta Neuropathol. 2018;136(4):507–23. Epub 2018/08/12. doi: 10.1007/s00401-018-1893-0. PubMed PMID: 30097696. [PMC free article: PMC6132947] [PubMed: 30097696]
- Chu Y, Jin X, Parada I, Pesic A, Stevens B, Barres B, Prince DA. Enhanced synaptic connectivity and epilepsy in c1q knockout mice. Proc Natl Acad Sci USA. 2010;107(17):7975–80. doi: 10.1073/pnas.0913449107. PubMed PMID: 20375278. [PMC free article: PMC2867906] [PubMed: 20375278]
- Dachet F, Bagla S, Keren-Aviram G, Morton A, Balan K, Saadat L, Valyi-Nagy T, Kupsky W, Song F, Dratz E, Loeb JA. Predicting novel histopathological microlesions in human epileptic brain through transcriptional clustering. Brain. 2015;138(Pt 2):356–70. doi: 10.1093/brain/awu350. PubMed PMID: 25516101. [PMC free article: PMC4306820] [PubMed: 25516101]
- Davalos D, Ryu JK, Merlini M, Baeten KM, Le Moan N, Petersen MA, Deerinck TJ, Smirnoff DS, Bedard C, Hakozaki H, Gonias Murray S, Ling JB, Lassmann H, Degen JL, Ellisman MH, Akassoglou K. Fibrinogen-induced perivascular microglial clustering is required for the development of axonal damage in neuroinflammation. Nat Commun. 2012;3:1227. doi: 10.1038/ncomms2230. PubMed PMID: 23187627. [PMC free article: PMC3514498] [PubMed: 23187627]
- Devinsky O, Vezzani A, Najjar S, De Lanerolle NC, Rogawski MA. Glia and epilepsy: Excitability and inflammation. Trends Neurosci. 2013;36(3):174–84. doi: 10.1016/j.tins.2012.11.008. PubMed PMID: 23298414. [PubMed: 23298414]
- DeVos SL, Goncharoff DK, Chen G, Kebodeaux CS, Yamada K, Stewart FR, Schuler DR, Maloney SE, Wozniak DF, Rigo F, Bennett CF, Cirrito JR, Holtzman DM, Miller TM. Antisense reduction of tau in adult mice protects against seizures. J Neurosci. 2013;33(31):12887–97. Epub 2013/08/02. doi: 10.1523/JNEUROSCI.2107-13.2013. PubMed PMID: 23904623. [PMC free article: PMC3728694] [PubMed: 23904623]
- Di Nunzio M, Di Sapia R, Sorrentino D, Kebede V, Cerovic M, Gullotta GS, Bacigaluppi M, Audinat E, Marchi N, Ravizza T, Vezzani A. Microglia proliferation plays distinct roles in acquired epilepsy depending on disease stages. Epilepsia. 2021;62(8):1931–45. Epub 2021/06/16. doi: 10.1111/epi.16956. PubMed PMID: 34128226. [PubMed: 34128226]
- Dias DO, Kim H, Holl D, Werne Solnestam B, Lundeberg J, Carlen M, Goritz C, Frisen J. Reducing pericyte-derived scarring promotes recovery after spinal cord injury. Cell. 2018;173(1):153–65 e22. Epub 2018/03/06. doi: 10.1016/j.cell.2018.02.004. PubMed PMID: 29502968. [PMC free article: PMC5871719] [PubMed: 29502968]
- DiSabato DJ, Quan N, Godbout JP. Neuroinflammation: The devil is in the details. J Neurochem. 2016;139 Suppl 2:136–53. doi: 10.1111/jnc.13607. PubMed PMID: 26990767. [PMC free article: PMC5025335] [PubMed: 26990767]
- Elmore MR, Najafi AR, Koike MA, Dagher NN, Spangenberg EE, Rice RA, Kitazawa M, Matusow B, Nguyen H, West BL, Green KN. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron. 2014;82(2):380–97. doi: 10.1016/j.neuron.2014.02.040. PubMed PMID: 24742461. [PMC free article: PMC4161285] [PubMed: 24742461]
- Eyo UB, Gu N, De S, Dong H, Richardson JR, Wu LJ. Modulation of microglial process convergence toward neuronal dendrites by extracellular calcium. J Neurosci. 2015;35(6):2417–22. doi: 10.1523/JNEUROSCI.3279-14.2015. PubMed PMID: 25673836. [PMC free article: PMC4323526] [PubMed: 25673836]
- Eyo UB, Haruwaka K, Mo M, Campos-Salazar AB, Wang L, Speros XSt, Sabu S, Xu P, Wu LJ. Microglia provide structural resolution to injured dendrites after severe seizures. Cell Rep. 2021;35(5):109080. doi: 10.1016/j.celrep.2021.109080. PubMed PMID: 33951432. [PMC free article: PMC8164475] [PubMed: 33951432]
- Eyo UB, Murugan M, Wu LJ. Microglia-neuron communication in epilepsy. Glia. 2016;65(1):5–18. doi: 10.1002/glia.23006. PubMed PMID: 27189853. [PMC free article: PMC5116010] [PubMed: 27189853]
- Eyo UB, Peng J, Murugan M, Mo M, Lalani A, Xie P, Xu P, Margolis DJ, Wu LJ. Regulation of physical microglia-neuron interactions by fractalkine signaling after status epilepticus. eNeuro. 2016 Nov-Dec;3(6):ENEURO.0209-16.2016. doi: 10.1523/ENEURO.0209-16.2016. PubMed PMID: 28101527. [PMC free article: PMC5237828] [PubMed: 28101527]
- Eyo UB, Peng J, Swiatkowski P, Mukherjee A, Bispo A, Wu LJ. Neuronal hyperactivity recruits microglial processes via neuronal nmda receptors and microglial p2y12 receptors after status epilepticus. J Neurosci. 2014;34(32):10528–40. doi: 10.1523/JNEUROSCI.0416-14.2014. PubMed PMID: 25100587. [PMC free article: PMC4200107] [PubMed: 25100587]
- Eyo UB, Wu LJ. Microglia: Lifelong patrolling immune cells of the brain. Prog Neurobiol. 2019;179:101614. doi: 10.1016/j.pneurobio.2019.04.003. PubMed PMID: 31075285. [PMC free article: PMC6599472] [PubMed: 31075285]
- Frigerio F, Pasqualini G, Craparotta I, Marchini S, van Vliet EA, Foerch P, Vandenplas C, Leclercq K, Aronica E, Porcu L, Pistorius K, Colas RA, Hansen TV, Perretti M, Kaminski RM, Dalli J, Vezzani A. N-3 docosapentaenoic acid-derived protectin d1 promotes resolution of neuroinflammation and arrests epileptogenesis. Brain. 2018;141(11):3130–43. Epub 2018/10/12. doi: 10.1093/brain/awy247. PubMed PMID: 30307467. [PMC free article: PMC6202571] [PubMed: 30307467]
- Fullerton JN, Gilroy DW. Resolution of inflammation: A new therapeutic frontier. Nat Rev Drug Discov. 2016;15(8):551–67. Epub 2016/03/30. doi: 10.1038/nrd.2016.39. PubMed PMID: 27020098. [PubMed: 27020098]
- Galatro TF, Holtman IR, Lerario AM, Vainchtein ID, Brouwer N, Sola PR, Veras MM, Pereira TF, Leite REP, Moller T, Wes PD, Sogayar MC, Laman JD, den Dunnen W, Pasqualucci CA, Oba-Shinjo SM, Boddeke E, Marie SKN, Eggen BJL. Transcriptomic analysis of purified human cortical microglia reveals age-associated changes. Nat Neurosci. 2017;20(8):1162–71. doi: 10.1038/nn.4597. PubMed PMID: 28671693. [PubMed: 28671693]
- Garbelli R, de Bock F, Medici V, Rousset MC, Villani F, Boussadia B, Arango-Lievano M, Jeanneteau F, Daneman R, Bartolomei F, Marchi N. Pdgfrbeta(+) cells in human and experimental neuro-vascular dysplasia and seizures. Neuroscience. 2015;306:18–27. Epub 2015/08/19. doi: 10.1016/j.neuroscience.2015.07.090. PubMed PMID: 26283024. [PubMed: 26283024]
- Ghosh C, Puvenna V, Gonzalez-Martinez J, Janigro D, Marchi N. Blood-brain barrier p450 enzymes and multidrug transporters in drug resistance: A synergistic role in neurological diseases. Curr Drug Metab. 2011;12(8):742–9. Epub 2011/05/17. doi: 10.2174/138920011798357051. PubMed PMID: 21568937. [PMC free article: PMC4874186] [PubMed: 21568937]
- Giannoni P, Badaut J, Dargazanli C, De Maudave AF, Klement W, Costalat V, Marchi N. The pericyte-glia interface at the blood-brain barrier. Clin Sci (Lond). 2018;132(3):361–74. doi: 10.1042/CS20171634. PubMed PMID: 29439117. [PubMed: 29439117]
- Giannoni P, Claeysen S, Noe F, Marchi N. Peripheral routes to neurodegeneration: Passing through the blood-brain barrier. Front Aging Neurosci. 2020;12:3. Epub 2020/03/03. doi: 10.3389/fnagi.2020.00003. PubMed PMID: 32116645. [PMC free article: PMC7010934] [PubMed: 32116645]
- Goritz C, Dias DO, Tomilin N, Barbacid M, Shupliakov O, Frisen J. A pericyte origin of spinal cord scar tissue. Science. 2011;333(6039):238–42. doi: 10.1126/science.1203165. PubMed PMID: 21737741. [PubMed: 21737741]
- Gorter JA, van Vliet EA, Aronica E. Status epilepticus, blood-brain barrier disruption, inflammation, and epileptogenesis. Epilepsy Behav. 2015;49:13–6. Epub 2015/05/11. doi: 10.1016/j.yebeh.2015.04.047. PubMed PMID: 25958228. [PubMed: 25958228]
- Gosselin D, Skola D, Coufal NG, Holtman IR, Schlachetzki JCM, Sajti E, Jaeger BN, O’Connor C, Fitzpatrick C, Pasillas MP, Pena M, Adair A, Gonda DD, Levy ML, Ransohoff RM, Gage FH, Glass CK. An environment-dependent transcriptional network specifies human microglia identity. Science. 2017;356(6344):eaal3222. doi: 10.1126/science.aal3222. PubMed PMID: 28546318. [PMC free article: PMC5858585] [PubMed: 28546318]
- Halder SK, Milner R. A critical role for microglia in maintaining vascular integrity in the hypoxic spinal cord. Proc Natl Acad Sci USA. 2019;116(51):26029–37. doi: 10.1073/pnas.1912178116. PubMed PMID: 31772011. [PMC free article: PMC6925983] [PubMed: 31772011]
- Hammond TR, Robinton D, Stevens B. Microglia and the brain: Complementary partners in development and disease. Annu Rev Cell Dev Biol. 2018;34:523–44. doi: 10.1146/annurev-cellbio-100616-060509. PubMed PMID: 30089221. [PubMed: 30089221]
- Hasegawa S, Yamaguchi M, Nagao H, Mishina M, Mori K. Enhanced cell-to-cell contacts between activated microglia and pyramidal cell dendrites following kainic acid-induced neurotoxicity in the hippocampus. J Neuroimmunol. 2007;186(1–2):75–85. doi: 10.1016/j.jneuroim.2007.03.005. PubMed PMID: 17428546. [PubMed: 17428546]
- Hiragi T, Ikegaya Y, Koyama R. Microglia after seizures and in epilepsy. Cells. 2018;7(4):26. doi: 10.3390/cells7040026. PubMed PMID: 29597334. [PMC free article: PMC5946103] [PubMed: 29597334]
- Ivens S, Kaufer D, Flores LP, Bechmann I, Zumsteg D, Tomkins O, Seiffert E, Heinemann U, Friedman A. Tgf-beta receptor-mediated albumin uptake into astrocytes is involved in neocortical epileptogenesis. Brain. 2007;130(Pt 2):535–47. Epub 2006/11/24. doi: 10.1093/brain/awl317. PubMed PMID: 17121744. [PubMed: 17121744]
- Janigro D, Bailey DM, Lehmann S, Badaut J, O’Flynn R, Hirtz C, Marchi N. Peripheral blood and salivary biomarkers of blood-brain barrier permeability and neuronal damage: Clinical and applied concepts. Front Neurol. 2020;11:577312. Epub 2021/02/23. doi: 10.3389/fneur.2020.577312. PubMed PMID: 33613412. [PMC free article: PMC7890078] [PubMed: 33613412]
- Kastanauskaite A, Alonso-Nanclares L, Blazquez-Llorca L, Pastor J, Sola RG, DeFelipe J. Alterations of the microvascular network in sclerotic hippocampi from patients with epilepsy. J Neuropathol Exp Neurol. 2009;68(8):939–50. Epub 2009/07/17. doi: 10.1097/NEN.0b013e3181b08622. PubMed PMID: 19606060. [PubMed: 19606060]
- Kharatishvili I, Shan ZY, She DT, Foong S, Kurniawan ND, Reutens DC. Mri changes and complement activation correlate with epileptogenicity in a mouse model of temporal lobe epilepsy. Brain Struct Funct. 2014;219(2):683–706. doi: 10.1007/s00429-013-0528-4. PubMed PMID: 23474541. [PubMed: 23474541]
- Kierdorf K, Prinz M. Microglia in steady state. J Clin Invest. 2017;127(9):3201–9. doi: 10.1172/JCI90602. PubMed PMID: 28714861. [PMC free article: PMC5669563] [PubMed: 28714861]
- Klement W, Blaquiere M, Zub E, deBock F, Boux F, Barbier E, Audinat E, Lerner-Natoli M, Marchi N. A pericyte-glia scarring develops at the leaky capillaries in the hippocampus during seizure activity. Epilepsia. 2019;60(7):1399–411. doi: 10.1111/epi.16019. PubMed PMID: 31135065. [PubMed: 31135065]
- Klement W, Garbelli R, Zub E, Rossini L, Tassi L, Girard B, Blaquiere M, Bertaso F, Perroy J, de Bock F, Marchi N. Seizure progression and inflammatory mediators promote pericytosis and pericyte-microglia clustering at the cerebrovasculature. Neurobiol Dis. 2018;113:70–81. Epub 2018/02/13. doi: 10.1016/j.nbd.2018.02.002. PubMed PMID: 29432809. [PubMed: 29432809]
- Koizumi S, Shigemoto-Mogami Y, Nasu-Tada K, Shinozaki Y, Ohsawa K, Tsuda M, Joshi BV, Jacobson KA, Kohsaka S, Inoue K. Udp acting at p2y6 receptors is a mediator of microglial phagocytosis. Nature. 2007;446(7139):1091–5. doi: 10.1038/nature05704. PubMed PMID: 17410128. [PMC free article: PMC3464483] [PubMed: 17410128]
- Konishi H, Kiyama H. Microglial trem2/dap12 signaling: A double-edged sword in neural diseases. Front Cell Neurosci. 2018;12:206. doi: 10.3389/fncel.2018.00206. PubMed PMID: 30127720. [PMC free article: PMC6087757] [PubMed: 30127720]
- Kopczynska M, Zelek WM, Vespa S, Touchard S, Wardle M, Loveless S, Thomas RH, Hamandi K, Morgan BP. Complement system biomarkers in epilepsy. Seizure. 2018;60:1–7. Epub 2018/06/02. doi: 10.1016/j.seizure.2018.05.016. PubMed PMID: 29857269. [PubMed: 29857269]
- Libbey JE, Kirkman NJ, Wilcox KS, White HS, Fujinami RS. Role for complement in the development of seizures following acute viral infection. J Virol. 2010;84(13):6452–60. doi: 10.1128/JVI.00422-10. PubMed PMID: 20427530. [PMC free article: PMC2903264] [PubMed: 20427530]
- Librizzi L, de Cutis M, Janigro D, Runtz L, de Bock F, Barbier EL, Marchi N. Cerebrovascular heterogeneity and neuronal excitability. Neurosci Lett. 2018;667:75–83. Epub 2017/01/15. doi: 10.1016/j.neulet.2017.01.013. PubMed PMID: 28087439. [PubMed: 28087439]
- Luo C, Koyama R, Ikegaya Y. Microglia engulf viable newborn cells in the epileptic dentate gyrus. Glia. 2016;64(9):1508–17. doi: 10.1002/glia.23018. PubMed PMID: 27301702. [PubMed: 27301702]
- Marchi N, Angelov L, Masaryk T, Fazio V, Granata T, Hernandez N, Hallene K, Diglaw T, Franic L, Najm I, Janigro D. Seizure-promoting effect of blood-brain barrier disruption. Epilepsia. 2007;48(4):732–42. Epub 2007/02/27. doi: 10.1111/j.1528-1167.2007.00988.x. PubMed PMID: 17319915. [PMC free article: PMC4135474] [PubMed: 17319915]
- Marchi N, Granata T, Ghosh C, Janigro D. Blood-brain barrier dysfunction and epilepsy: Pathophysiologic role and therapeutic approaches. Epilepsia. 2012;53(11):1877–86. doi: 10.1111/j.1528-1167.2012.03637.x. PubMed PMID: 22905812. [PMC free article: PMC4842020] [PubMed: 22905812]
- Marchi N, Granata T, Janigro D. Inflammatory pathways of seizure disorders. Trends Neurosci. 2014;37(2):55–65. doi: 10.1016/j.tins.2013.11.002. PubMed PMID: 24355813. [PMC free article: PMC3977596] [PubMed: 24355813]
- Marchi N, Johnson AJ, Puvenna V, Johnson HL, Tierney W, Ghosh C, Cucullo L, Fabene PF, Janigro D. Modulation of peripheral cytotoxic cells and ictogenesis in a model of seizures. Epilepsia. 2011;52(9):1627–34. Epub 2011/06/02. doi: 10.1111/j.1528-1167.2011.03080.x. PubMed PMID: 21627645. [PMC free article: PMC3728674] [PubMed: 21627645]
- Marchi N, Lerner-Natoli M. Cerebrovascular remodeling and epilepsy. Neuroscientist. 2013;19(3):304–12. doi: 10.1177/1073858412462747. PubMed PMID: 23072899. [PMC free article: PMC3701121] [PubMed: 23072899]
- Masuda T, Sankowski R, Staszewski O, Bottcher C, Amann L, Sagar, Scheiwe C, Nessler S, Kunz P, van Loo G, Coenen VA, Reinacher PC, Michel A, Sure U, Gold R, Grun D, Priller J, Stadelmann C, Prinz M. Spatial and temporal heterogeneity of mouse and human microglia at single-cell resolution. Nature. 2019;566(7744):388–92. doi: 10.1038/s41586-019-0924-x. PubMed PMID: 30760929. [PubMed: 30760929]
- Merlini M, Rafalski VA, Ma K, Kim KY, Bushong EA, Rios Coronado PE, Yan Z, Mendiola AS, Sozmen EG, Ryu JK, Haberl MG, Madany M, Sampson DN, Petersen MA, Bardehle S, Tognatta R, Dean T, Jr., Acevedo RM, Cabriga B, Thomas R, Coughlin SR, Ellisman MH, Palop JJ, Akassoglou K. Microglial gi-dependent dynamics regulate brain network hyperexcitability. Nat Neurosci. 2021;24(1):19–23. doi: 10.1038/s41593-020-00756-7. PubMed PMID: 33318667. [PMC free article: PMC8118167] [PubMed: 33318667]
- Merlini M, Rafalski VA, Rios Coronado PE, Gill TM, Ellisman M, Muthukumar G, Subramanian KS, Ryu JK, Syme CA, Davalos D, Seeley WW, Mucke L, Nelson RB, Akassoglou K. Fibrinogen induces microglia-mediated spine elimination and cognitive impairment in an alzheimer’s disease model. Neuron. 2019;101(6):1099–108 e6. doi: 10.1016/j.neuron.2019.01.014. PubMed PMID: 30737131. [PMC free article: PMC6602536] [PubMed: 30737131]
- Milesi S, Boussadia B, Plaud C, Catteau M, Rousset MC, De Bock F, Schaeffer M, Lerner-Natoli M, Rigau V, Marchi N. Redistribution of pdgfrbeta cells and ng2dsred pericytes at the cerebrovasculature after status epilepticus. Neurobiol Dis. 2014;71:151–8. Epub 2014/08/05. doi: 10.1016/j.nbd.2014.07.010. PubMed PMID: 25088711. [PMC free article: PMC4179992] [PubMed: 25088711]
- Miyamoto A, Wake H, Ishikawa AW, Eto K, Shibata K, Murakoshi H, Koizumi S, Moorhouse AJ, Yoshimura Y, Nabekura J. Microglia contact induces synapse formation in developing somatosensory cortex. Nat Commun. 2016;7:12540. doi: 10.1038/ncomms12540. PubMed PMID: 27558646. [PMC free article: PMC5007295] [PubMed: 27558646]
- Morin-Brureau M, Milior G, Royer J, Chali F, LeDuigou C, Savary E, Blugeon C, Jourdren L, Akbar D, Dupont S, Navarro V, Baulac M, Bielle F, Mathon B, Clemenceau S, Miles R. Microglial phenotypes in the human epileptic temporal lobe. Brain. 2018;141(12):3343–60. doi: 10.1093/brain/awy276. PubMed PMID: 30462183. [PMC free article: PMC6277010] [PubMed: 30462183]
- Nguyen LH, Brewster AL, Clark ME, Regnier-Golanov A, Sunnen CN, Patil VV, D’Arcangelo G, Anderson AE. Mtor inhibition suppresses established epilepsy in a mouse model of cortical dysplasia. Epilepsia. 2015;56(4):636–46. doi: 10.1111/epi.12946. PubMed PMID: 25752454. [PMC free article: PMC4459784] [PubMed: 25752454]
- Nguyen LH, Mahadeo T, Bordey A. Mtor hyperactivity levels influence the severity of epilepsy and associated neuropathology in an experimental model of tuberous sclerosis complex and focal cortical dysplasia. J Neurosci. 2019;39(14):2762–73. doi: 10.1523/JNEUROSCI.2260-18.2019. PubMed PMID: 30700531. [PMC free article: PMC6445990] [PubMed: 30700531]
- Noe FM, Marchi N. Central nervous system lymphatic unit, immunity, and epilepsy: Is there a link? Epilepsia Open. 2019;4(1):30–9. Epub 2019/03/15. doi: 10.1002/epi4.12302. PubMed PMID: 30868113. [PMC free article: PMC6398113] [PubMed: 30868113]
- Obermeier B, Daneman R, Ransohoff RM. Development, maintenance and disruption of the blood-brain barrier. Nat Med. 2013;19(12):1584–96. doi: 10.1038/nm.3407. PubMed PMID: 24309662. [PMC free article: PMC4080800] [PubMed: 24309662]
- Oosterhof N, Chang IJ, Karimiani EG, Kuil LE, Jensen DM, Daza R, Young E, Astle L, van der Linde HC, Shivaram GM, Demmers J, Latimer CS, Keene CD, Loter E, Maroofian R, van Ham TJ, Hevner RF, Bennett JT. Homozygous mutations in csf1r cause a pediatric-onset leukoencephalopathy and can result in congenital absence of microglia. Am J Hum Genet. 2019;104(5):936–47. doi: 10.1016/j.ajhg.2019.03.010. PubMed PMID: 30982608. [PMC free article: PMC6506793] [PubMed: 30982608]
- Oosterhof N, Kuil LE, van der Linde HC, Burm SM, Berdowski W, van Ijcken WFJ, van Swieten JC, Hol EM, Verheijen MHG, van Ham TJ. Colony-stimulating factor 1 receptor (csf1r) regulates microglia density and distribution, but not microglia differentiation in vivo. Cell Rep. 2018;24(5):1203–17 e6. doi: 10.1016/j.celrep.2018.06.113. PubMed PMID: 30067976. [PubMed: 30067976]
- Patterson KP, Brennan GP, Curran M, Kinney-Lang E, Dube C, Rashid F, Ly C, Obenaus A, Baram TZ. Rapid, coordinate inflammatory responses after experimental febrile status epilepticus: Implications for epileptogenesis. eNeuro. 2015;2(5):ENEURO.0034-15.2015. doi: 10.1523/ENEURO.0034-15.2015. PubMed PMID: 26730400. [PMC free article: PMC4699830] [PubMed: 26730400]
- Pavlovsky L, Seiffert E, Heinemann U, Korn A, Golan H, Friedman A. Persistent bbb disruption may underlie alpha interferon-induced seizures. J Neurol. 2005;252(1):42–6. Epub 2005/01/27. doi: 10.1007/s00415-005-0596-3. PubMed PMID: 15672209. [PubMed: 15672209]
- Pitsch J, van Loo KMJ, Gallus M, Dik A, Kamalizade D, Baumgart AK, Gnatkovsky V, Muller JA, Opitz T, Hicking G, Naik VN, Wachsmuth L, Faber C, Surges R, Kurts C, Schoch S, Melzer N, Becker AJ. Cd8(+) t-lymphocyte-driven limbic encephalitis results in temporal lobe epilepsy. Ann Neurol. 2021;89(4):666–85. doi: 10.1002/ana.26000. PubMed PMID: 33368582. [PubMed: 33368582]
- Prager O, Kamintsky L, Hasam-Henderson LA, Schoknecht K, Wuntke V, Papageorgiou I, Swolinsky J, Muoio V, Bar-Klein G, Vazana U, Heinemann U, Friedman A, Kovacs R. Seizure-induced microvascular injury is associated with impaired neurovascular coupling and blood-brain barrier dysfunction. Epilepsia. 2019a;60(2):322–36. doi: 10.1111/epi.14631. PubMed PMID: 30609012. [PubMed: 30609012]
- Prinz M, Jung S, Priller J. Microglia biology: One century of evolving concepts. Cell. 2019;179(2):292–311. doi: 10.1016/j.cell.2019.08.053. PubMed PMID: 31585077. [PubMed: 31585077]
- Pulido RS, Munji RN, Chan TC, Quirk CR, Weiner GA, Weger BD, Rossi MJ, Elmsaouri S, Malfavon M, Deng A, Profaci CP, Blanchette M, Qian T, Foreman KL, Shusta EV, Gorman MR, Gachon F, Leutgeb S, Daneman R. Neuronal activity regulates blood-brain barrier efflux transport through endothelial circadian genes. Neuron. 2020;108(5):937–52 e7. Epub 2020/09/27. doi: 10.1016/j.neuron.2020.09.002. PubMed PMID: 32979312. [PMC free article: PMC7736535] [PubMed: 32979312]
- Ravizza T, Vezzani A. Pharmacological targeting of brain inflammation in epilepsy: Therapeutic perspectives from experimental and clinical studies. Epilepsia Open. 2018;3(Suppl Suppl 2):133–42. doi: 10.1002/epi4.12242. PubMed PMID: 30564772. [PMC free article: PMC6293065] [PubMed: 30564772]
- Riew TR, Choi JH, Kim HL, Jin X, Lee MY. Pdgfr-beta-positive perivascular adventitial cells expressing nestin contribute to fibrotic scar formation in the striatum of 3-np intoxicated rats. Front Mol Neurosci. 2018;11:402. Epub 2018/11/21. doi: 10.3389/fnmol.2018.00402. PubMed PMID: 30455628. [PMC free article: PMC6230557] [PubMed: 30455628]
- Rustenhoven J, Jansson D, Smyth LC, Dragunow M. Brain pericytes as mediators of neuroinflammation. Trends Pharmacol Sci. 2017;38(3):291–304. doi: 10.1016/j.tips.2016.12.001. PubMed PMID: 28017362. [PubMed: 28017362]
- Ryu JK, McLarnon JG. A leaky blood-brain barrier, fibrinogen infiltration and microglial reactivity in inflamed alzheimer’s disease brain. J Cell Mol Med. 2009;13(9A):2911–25. doi: 10.1111/j.1582-4934.2008.00434.x. PubMed PMID: 18657226. [PMC free article: PMC4498946] [PubMed: 18657226]
- Sabilallah M, Fontanaud P, Linck N, Boussadia B, Peyroutou R, Lasgouzes T, Rassendren FA, Marchi N, Hirbec HE. Evidence for status epilepticus and pro-inflammatory changes after intranasal kainic acid administration in mice. PLoS One. 2016;11(3):e0150793. Epub 2016/03/11. doi: 10.1371/journal.pone.0150793. PubMed PMID: 26963100. [PMC free article: PMC4786335] [PubMed: 26963100]
- Sanchez JMS, DePaula-Silva AB, Doty DJ, Truong A, Libbey JE, Fujinami RS. Microglial cell depletion is fatal with low level picornavirus infection of the central nervous system. J Neurovirol. 2019;25(3):415–21. doi: 10.1007/s13365-019-00740-3. PubMed PMID: 30859497. [PMC free article: PMC6635090] [PubMed: 30859497]
- Sankowski R, Bottcher C, Masuda T, Geirsdottir L, Sagar, Sindram E, Seredenina T, Muhs A, Scheiwe C, Shah MJ, Heiland DH, Schnell O, Grun D, Priller J, Prinz M. Mapping microglia states in the human brain through the integration of high-dimensional techniques. Nat Neurosci. 2019;22(12):2098–110. doi: 10.1038/s41593-019-0532-y. PubMed PMID: 31740814. [PubMed: 31740814]
- Schafer DP, Lehrman EK, Kautzman AG, Koyama R, Mardinly AR, Yamasaki R, Ransohoff RM, Greenberg ME, Barres BA, Stevens B. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron. 2012;74(4):691–705. doi: 10.1016/j.neuron.2012.03.026. PubMed PMID: 22632727. [PMC free article: PMC3528177] [PubMed: 22632727]
- Schafer DP, Lehrman EK, Stevens B. The “quad-partite” synapse: Microglia-synapse interactions in the developing and mature cns. Glia. 2013;61(1):24–36. doi: 10.1002/glia.22389. PubMed PMID: 22829357. [PMC free article: PMC4082974] [PubMed: 22829357]
- Schartz ND, Herr SA, Madsen L, Butts SJ, Torres C, Mendez LB, Brewster AL. Spatiotemporal profile of map2 and microglial changes in the hippocampal ca1 region following pilocarpine-induced status epilepticus. Sci Rep. 2016;6:24988. doi: 10.1038/srep24988. PubMed PMID: 27143585. [PMC free article: PMC4855223] [PubMed: 27143585]
- Schartz ND, Sommer AL, Colin SA, Mendez LB, Brewster AL. Early treatment with c1 esterase inhibitor improves weight but not memory deficits in a rat model of status epilepticus. Physiol Behav. 2019;212:112705. doi: 10.1016/j.physbeh.2019.112705. PubMed PMID: 31628931. [PMC free article: PMC6879103] [PubMed: 31628931]
- Schartz ND, Tenner AJ. The good, the bad, and the opportunities of the complement system in neurodegenerative disease. J Neuroinflammation. 2020;17(1):354. doi: 10.1186/s12974-020-02024-8. PubMed PMID: 33239010. [PMC free article: PMC7690210] [PubMed: 33239010]
- Schartz ND, Wyatt-Johnson SK, Price LR, Colin SA, Brewster AL. Status epilepticus triggers long-lasting activation of complement c1q-c3 signaling in the hippocampus that correlates with seizure frequency in experimental epilepsy. Neurobiol Dis. 2018;109(Pt A):163–73. doi: 10.1016/j.nbd.2017.10.012. PubMed PMID: 29074125. [PubMed: 29074125]
- Seiffert E, Dreier JP, Ivens S, Bechmann I, Tomkins O, Heinemann U, Friedman A. Lasting blood-brain barrier disruption induces epileptic focus in the rat somatosensory cortex. J Neurosci. 2004;24(36):7829–36. Epub 2004/09/10. doi: 10.1523/JNEUROSCI.1751-04.2004. PubMed PMID: 15356194. [PMC free article: PMC6729929] [PubMed: 15356194]
- Sierra-Torre V, Plaza-Zabala A, Bonifazi P, Abiega O, Diaz-Aparicio I, Tegelberg S, Lehesjoki AE, Valero J, Sierra A. Microglial phagocytosis dysfunction in the dentate gyrus is related to local neuronal activity in a genetic model of epilepsy. Epilepsia. 2020;61(11):2593–608. doi: 10.1111/epi.16692. PubMed PMID: 32940364. [PMC free article: PMC7756777] [PubMed: 32940364]
- Sierra A, Abiega O, Shahraz A, Neumann H. Janus-faced microglia: Beneficial and detrimental consequences of microglial phagocytosis. Front Cell Neurosci. 2013;7:6. doi: 10.3389/fncel.2013.00006. PubMed PMID: 23386811. [PMC free article: PMC3558702] [PubMed: 23386811]
- Smyth LCD, Rustenhoven J, Park TI, Schweder P, Jansson D, Heppner PA, O’Carroll SJ, Mee EW, Faull RLM, Curtis M, Dragunow M. Unique and shared inflammatory profiles of human brain endothelia and pericytes. J Neuroinflammation. 2018;15(1):138. doi: 10.1186/s12974-018-1167-8. PubMed PMID: 29751771. [PMC free article: PMC5948925] [PubMed: 29751771]
- Soderblom C, Luo X, Blumenthal E, Bray E, Lyapichev K, Ramos J, Krishnan V, Lai-Hsu C, Park KK, Tsoulfas P, Lee JK. Perivascular fibroblasts form the fibrotic scar after contusive spinal cord injury. J Neurosci. 2013;33(34):13882–7. doi: 10.1523/JNEUROSCI.2524-13.2013. PubMed PMID: 23966707. [PMC free article: PMC3755723] [PubMed: 23966707]
- Srivastava PK, van Eyll J, Godard P, Mazzuferi M, Delahaye-Duriez A, Steenwinckel JV, Gressens P, Danis B, Vandenplas C, Foerch P, Leclercq K, Mairet-Coello G, Cardenas A, Vanclef F, Laaniste L, Niespodziany I, Keaney J, Gasser J, Gillet G, Shkura K, Chong SA, Behmoaras J, Kadiu I, Petretto E, Kaminski RM, Johnson MR. A systems-level framework for drug discovery identifies csf1r as an anti-epileptic drug target. Nat Commun. 2018;9(1):3561. doi: 10.1038/s41467-018-06008-4. PubMed PMID: 30177815. [PMC free article: PMC6120885] [PubMed: 30177815]
- Stephan AH, Barres BA, Stevens B. The complement system: An unexpected role in synaptic pruning during development and disease. Annu Rev Neurosci. 2012;35:369–89. doi: 10.1146/annurev-neuro-061010-113810. PubMed PMID: 22715882. [PubMed: 22715882]
- Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS, Nouri N, Micheva KD, Mehalow AK, Huberman AD, Stafford B, Sher A, Litke AM, Lambris JD, Smith SJ, John SW, Barres BA. The classical complement cascade mediates cns synapse elimination. Cell. 2007;131(6):1164–78. doi: 10.1016/j.cell.2007.10.036. PubMed PMID: 18083105. [PubMed: 18083105]
- Sun FJ, Zhang CQ, Chen X, Wei YJ, Li S, Liu SY, Zang ZL, He JJ, Guo W, Yang H. Downregulation of cd47 and cd200 in patients with focal cortical dysplasia type iib and tuberous sclerosis complex. J Neuroinflammation. 2016;13(1):85. doi: 10.1186/s12974-016-0546-2. PubMed PMID: 27095555. [PMC free article: PMC4837553] [PubMed: 27095555]
- Sweeney MD, Ayyadurai S, Zlokovic BV. Pericytes of the neurovascular unit: Key functions and signaling pathways. Nat Neurosci. 2016;19(6):771–83. Epub 2016/05/27. doi: 10.1038/nn.4288. PubMed PMID: 27227366. [PMC free article: PMC5745011] [PubMed: 27227366]
- Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV. Blood-brain barrier: From physiology to disease and back. Physiol Rev. 2019;99(1):21–78. doi: 10.1152/physrev.00050.2017. PubMed PMID: 30280653. [PMC free article: PMC6335099] [PubMed: 30280653]
- Swissa E, Serlin Y, Vazana U, Prager O, Friedman A. Blood-brain barrier dysfunction in status epileptics: Mechanisms and role in epileptogenesis. Epilepsy Behav. 2019;101(Pt B):106285. Epub 2019/11/13. doi: 10.1016/j.yebeh.2019.04.038. PubMed PMID: 31711869. [PubMed: 31711869]
- Tay TL, Savage JC, Hui CW, Bisht K, Tremblay ME. Microglia across the lifespan: From origin to function in brain development, plasticity and cognition. J Physiol. 2017;595(6):1929–45. doi: 10.1113/JP272134. PubMed PMID: 27104646. [PMC free article: PMC5350449] [PubMed: 27104646]
- Tremblay ME, Lowery RL, Majewska AK. Microglial interactions with synapses are modulated by visual experience. PLoS Biol. 2010;8(11):e1000527. doi: 10.1371/journal.pbio.1000527. PubMed PMID: 21072242. [PMC free article: PMC2970556] [PubMed: 21072242]
- Ulland TK, Wang Y, Colonna M. Regulation of microglial survival and proliferation in health and diseases. Semin Immunol. 2015;27(6):410–5. doi: 10.1016/j.smim.2016.03.011. PubMed PMID: 27033414. [PMC free article: PMC5084693] [PubMed: 27033414]
- van Vliet EA, Aronica E, Vezzani A, Ravizza T. Review: Neuroinflammatory pathways as treatment targets and biomarker candidates in epilepsy: Emerging evidence from preclinical and clinical studies. Neuropath Appl Neuro. 2018;44(1):91–111. doi: 10.1111/nan.12444. PubMed PMID: WOS:000425522600007. [PubMed: 28977690]
- Varvel NH, Neher JJ, Bosch A, Wang W, Ransohoff RM, Miller RJ, Dingledine R. Infiltrating monocytes promote brain inflammation and exacerbate neuronal damage after status epilepticus. Proc Natl Acad Sci U S A. 2016;113(38):E5665–74. doi: 10.1073/pnas.1604263113. PubMed PMID: 27601660. [PMC free article: PMC5035862] [PubMed: 27601660]
- Vazana U, Veksler R, Pell GS, Prager O, Fassler M, Chassidim Y, Roth Y, Shahar H, Zangen A, Raccah R, Onesti E, Ceccanti M, Colonnese C, Santoro A, Salvati M, D’Elia A, Nucciarelli V, Inghilleri M, Friedman A. Glutamate-mediated blood-brain barrier opening: Implications for neuroprotection and drug delivery. J Neurosci. 2016;36(29):7727–39. Epub 2016/07/23. doi: 10.1523/JNEUROSCI.0587-16.2016. PubMed PMID: 27445149. [PMC free article: PMC4951577] [PubMed: 27445149]
- Vezzani A, Balosso S, Ravizza T. Neuroinflammatory pathways as treatment targets and biomarkers in epilepsy. Nat Rev Neurol. 2019;15(8):459–72. Epub 2019/07/03. doi: 10.1038/s41582-019-0217-x. PubMed PMID: 31263255. [PubMed: 31263255]
- Vezzani A, French J, Bartfai T, Baram TZ. The role of inflammation in epilepsy. Nat Rev Neurol. 2011;7(1):31–40. Epub 2010/12/08. doi: nrneurol.2010.178 [pii]10.1038/nrneurol.2010.178. PubMed PMID: 21135885. [PMC free article: PMC3378051] [PubMed: 21135885]
- Vezzani A, Lang B, Aronica E. Immunity and inflammation in epilepsy. Cold Spring Harb Perspect Med. 2015;6(2):a022699. Epub 2015/12/20. doi: 10.1101/cshperspect.a022699. PubMed PMID: 26684336. [PMC free article: PMC4743070] [PubMed: 26684336]
- Waisman A, Ginhoux F, Greter M, Bruttger J. Homeostasis of microglia in the adult brain: Review of novel microglia depletion systems. Trends Immunol. 2015;36(10):625–36. doi: 10.1016/j.it.2015.08.005. PubMed PMID: 26431940. [PubMed: 26431940]
- Wake H, Moorhouse AJ, Jinno S, Kohsaka S, Nabekura J. Resting microglia directly monitor the functional state of synapses in vivo and determine the fate of ischemic terminals. J Neurosci. 2009;29(13):3974–80. Epub 2009/04/03. doi: 29/13/3974 [pii] 10.1523/JNEUROSCI.4363-08.2009. PubMed PMID: 19339593. [PMC free article: PMC6665392] [PubMed: 19339593]
- Wan YS, Feng B, You Y, Yu J, Xu CL, Dai HB, Trapp BD, Shi P, Chen Z, Hu WW. Microglial displacement of gabaergic synapses is a protective event during complex febrile seizures. Cell Reports. 2020;33(5):108346. doi: ARTN 108346 10.1016/j.celrep.2020.108346. PubMed PMID: WOS:000587464900018. [PubMed: 33147450]
- Weinhard L, di Bartolomei G, Bolasco G, Machado P, Schieber NL, Neniskyte U, Exiga M, Vadisiute A, Raggioli A, Schertel A, Schwab Y, Gross CT. Microglia remodel synapses by presynaptic trogocytosis and spine head filopodia induction. Nat Commun. 2018;9(1):1228. doi: 10.1038/s41467-018-03566-5. PubMed PMID: 29581545. [PMC free article: PMC5964317] [PubMed: 29581545]
- Wirenfeldt M, Clare R, Tung S, Bottini A, Mathern GW, Vinters HV. Increased activation of iba1+ microglia in pediatric epilepsy patients with rasmussen’s encephalitis compared with cortical dysplasia and tuberous sclerosis complex. Neurobiol Dis. 2009;34(3):432–40. doi: 10.1016/j.nbd.2009.02.015. PubMed PMID: 19285133. [PMC free article: PMC2683191] [PubMed: 19285133]
- Wong M. The role of glia in epilepsy, intellectual disability, and other neurodevelopmental disorders in tuberous sclerosis complex. J Neurodev Disord. 2019;11(1):30. doi: 10.1186/s11689-019-9289-6. PubMed PMID: 31838997. [PMC free article: PMC6913020] [PubMed: 31838997]
- Wu W, Li Y, Wei Y, Bosco DB, Xie M, Zhao MG, Richardson JR, Wu LJ. Microglial depletion aggravates the severity of acute and chronic seizures in mice. Brain Behav Immun. 2020;89:245–55. doi: 10.1016/j.bbi.2020.06.028. PubMed PMID: 32621847. [PMC free article: PMC7572576] [PubMed: 32621847]
- Wyatt-Johnson SK, Brewster AL. Emerging roles for microglial phagocytic signaling in epilepsy. Epilepsy Curr. 2020;20(1):33–8. doi: 10.1177/1535759719890336. PubMed PMID: 31791133. [PMC free article: PMC7020537] [PubMed: 31791133]
- Wyatt-Johnson SK, Herr SA, Brewster AL. Status epilepticus triggers time-dependent alterations in microglia abundance and morphological phenotypes in the hippocampus. Front Neurol. 2017;8:700. doi: 10.3389/fneur.2017.00700. PubMed PMID: 29326654. [PMC free article: PMC5741821] [PubMed: 29326654]
- Wyatt-Johnson SK, Sommer AL, Shim KY, Brewster AL. Suppression of microgliosis with the colony-stimulating factor 1 receptor inhibitor plx3397 does not attenuate memory defects during epileptogenesis in the rat. Front Neurol. 2021;12:651096. Epub 2021/06/22. doi: 10.3389/fneur.2021.651096. PubMed PMID: 34149593. [PMC free article: PMC8209304] [PubMed: 34149593]
- Wyatt SK, Witt T, Barbaro NM, Cohen-Gadol AA, Brewster AL. Enhanced classical complement pathway activation and altered phagocytosis signaling molecules in human epilepsy. Exp Neurol. 2017;295:184–93. doi: 10.1016/j.expneurol.2017.06.009. PubMed PMID: 28601603. [PubMed: 28601603]
- Xu D, Robinson AP, Ishii T, Duncan DS, Alden TD, Goings GE, Ifergan I, Podojil JR, Penaloza-MacMaster P, Kearney JA, Swanson GT, Miller SD, Koh S. Peripherally derived t regulatory and gammadelta t cells have opposing roles in the pathogenesis of intractable pediatric epilepsy. J Exp Med. 2018;215(4):1169–86. doi: 10.1084/jem.20171285. PubMed PMID: 29487082. [PMC free article: PMC5881465] [PubMed: 29487082]
- Zehendner CM, Sebastiani A, Hugonnet A, Bischoff F, Luhmann HJ, Thal SC. Traumatic brain injury results in rapid pericyte loss followed by reactive pericytosis in the cerebral cortex. Sci Rep. 2015;5:13497. Epub 2015/09/04. doi: 10.1038/srep13497. PubMed PMID: 26333872. [PMC free article: PMC4558600] [PubMed: 26333872]
- Zhang B, Zou J, Han L, Beeler B, Friedman JL, Griffin E, Piao YS, Rensing NR, Wong M. The specificity and role of microglia in epileptogenesis in mouse models of tuberous sclerosis complex. Epilepsia. 2018;59(9):1796–806. doi: 10.1111/epi.14526. PubMed PMID: 30079598. [PMC free article: PMC6121723] [PubMed: 30079598]
- 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. Noninflammatory changes of microglia are sufficient to cause epilepsy. Cell Rep. 2018;22(8):2080–93. doi: 10.1016/j.celrep.2018.02.004. PubMed PMID: 29466735. [PMC free article: PMC5880308] [PubMed: 29466735]
- Zhao XF, Liao Y, Alam MM, Mathur R, Feustel P, Mazurkiewicz JE, Adamo MA, Zhu XC, Huang Y. Microglial mtor is neuronal protective and antiepileptogenic in the pilocarpine model of temporal lobe epilepsy. J Neurosci. 2020;40(40):7593–608. doi: 10.1523/JNEUROSCI.2754-19.2020. PubMed PMID: 32868461. [PMC free article: PMC7531547] [PubMed: 32868461]
- Zimmer TS, Broekaart DWM, Gruber VE, van Vliet EA, Muhlebner A, Aronica E. Tuberous sclerosis complex as disease model for investigating mtor-related gliopathy during epileptogenesis. Front Neurol. 2020;11:1028. doi: 10.3389/fneur.2020.01028. PubMed PMID: 33041976. [PMC free article: PMC7527496] [PubMed: 33041976]
- Zub E, Canet G, Garbelli R, Blaquiere M, Rossini L, Pastori C, Sheikh M, Reutelingsperger C, Klement W, de Bock F, Audinat E, Givalois L, Solito E, Marchi N. The gr-anxa1 pathway is a pathological player and a candidate target in epilepsy. FASEB J. 2019;33(12):13998–4009. Epub 2019/10/17. doi: 10.1096/fj.201901596R. PubMed PMID: 31618599. [PubMed: 31618599]
- Abstract
- Introduction: Neuro-Glio-Vascular Regulatory Cells in Seizure Networks
- What Is a Pericyte?
- Perivascular Inflammatory Cell Reactivity during Seizures: Focus on Pericytes
- What Are Microglial Cells?
- Microglia Surveillance and Neuronal Interactions
- Microglial Pro- and Anti-inflammatory Molecular Equilibriums in Experimental Epilepsy
- Microglia-Pericytes Perivascular Assembly and Reactivity during Seizures: Experimental and Clinical Evidence
- Microglial Profiles in Human Drug-Resistant Epilepsies
- Pharmacological Entry Points: Focus on Pericytes and Microglia
- Conclusion: Refining Timing and Targets for Pharmacological Interventions
- Acknowledgments
- Disclosure Statement
- References
- Seizure progression and inflammatory mediators promote pericytosis and pericyte-microglia clustering at the cerebrovasculature.[Neurobiol Dis. 2018]Seizure progression and inflammatory mediators promote pericytosis and pericyte-microglia clustering at the cerebrovasculature.Klement W, Garbelli R, Zub E, Rossini L, Tassi L, Girard B, Blaquiere M, Bertaso F, Perroy J, de Bock F, et al. Neurobiol Dis. 2018 May; 113:70-81. Epub 2018 Feb 9.
- Redistribution of PDGFRβ cells and NG2DsRed pericytes at the cerebrovasculature after status epilepticus.[Neurobiol Dis. 2014]Redistribution of PDGFRβ cells and NG2DsRed pericytes at the cerebrovasculature after status epilepticus.Milesi S, Boussadia B, Plaud C, Catteau M, Rousset MC, De Bock F, Schaeffer M, Lerner-Natoli M, Rigau V, Marchi N. Neurobiol Dis. 2014 Nov; 71:151-8. Epub 2014 Aug 1.
- PDGFRβ(+) cells in human and experimental neuro-vascular dysplasia and seizures.[Neuroscience. 2015]PDGFRβ(+) cells in human and experimental neuro-vascular dysplasia and seizures.Garbelli R, de Bock F, Medici V, Rousset MC, Villani F, Boussadia B, Arango-Lievano M, Jeanneteau F, Daneman R, Bartolomei F, et al. Neuroscience. 2015 Oct 15; 306:18-27. Epub 2015 Aug 15.
- Review The Neuroinflammatory Role of Pericytes in Epilepsy.[Biomedicines. 2021]Review The Neuroinflammatory Role of Pericytes in Epilepsy.Yamanaka G, Takata F, Kataoka Y, Kanou K, Morichi S, Dohgu S, Kawashima H. Biomedicines. 2021 Jun 30; 9(7). Epub 2021 Jun 30.
- Review From neurons to the neuro-glio-vascular unit: Seizures and brain homeostasis in networks.[Rev Neurol (Paris). 2023]Review From neurons to the neuro-glio-vascular unit: Seizures and brain homeostasis in networks.Cresto N, Janvier A, Marchi N. Rev Neurol (Paris). 2023 Apr; 179(4):308-315. Epub 2023 Feb 8.
- Pericytes and Microglia - Jasper's Basic Mechanisms of the EpilepsiesPericytes and Microglia - Jasper's Basic Mechanisms of the Epilepsies
Your browsing activity is empty.
Activity recording is turned off.
See more...