Abstract
Epilepsy is a major neurological condition that affects millions of people globally. While a number of interventions have been developed to mitigate this condition, a significant number of patients are refractory to these treatments. Consequently, other avenues of research are needed. One such avenue is modulation of the immune system response to this condition, which has mostly focused on microglia, the resident immune cells of the central nervous system (CNS). However, other immune cells can impact neurological conditions, principally bloodborne monocytes that can infiltrate into brain parenchyma after seizures. As such, this review will first discuss how monocytes can be recruited to the CNS and how they can be distinguished from there immunological cousins, microglia. Then we will explore what is known about the role monocytes have within seizure pathogenesis and epilepsy. Considering how little is known about monocyte function in seizure and epilepsy related pathologies, further studies are warranted that investigate infiltrated bloodborne monocytes as a potential therapeutic target for epilepsy treatment.
Keywords: monocytes, monocyte recruitment, microglia, seizures, epilepsy, blood-brain-barrier, inflammation
1. Introduction:
Epilepsy is a condition affecting upwards of 65 million people worldwide, and is the fourth most common neurological disorder in the United States [1, 2]. A major hallmark of epilepsy is the development of spontaneous seizures due to dysregulation of neuronal activity, synchronization, and excitability [1]. Although a number of medications have been developed to treat epilepsy, up to a third of patients are refractory to treatment [3]. Consequently, exploration of new avenues of epileptic intervention is needed. Epilepsy research has predominantly had a “neurocentric” focus, while the contributions of immunological systems have been largely understudied.
Within the central nervous system (CNS), microglia play the role of the resident immune cell type. Indeed, microglia contribute to the development of a variety of pathologies (e.g., pain, stroke, Alzheimer’s disease), and that modulation of their activity can impact the progression of these conditions [4–8]. In regards to epilepsy, microglia become readily activated taking on a larger, less ramified, amoeboid morphology. The pathological consequences of this activation include promotion of inflammation (i.e., expression of pro-inflammatory cytokines), regulation of neuronal activity, phagocytic clearance of neurons, and promotion of chronic-seizures [1, 9–11]. Moreover, it has been suggested that non-inflammatory alterations to microglia are sufficient to induce epilepsy [12]. However, microglia are not the only immune cell type that can influence epilepsy. Monocytes, an immunological relative to microglia, have also been suggested to affect a number of neurological disorders, including pain, stroke, amyotrophic lateral sclerosis (ALS), and epilepsy [13–17]. Monocytes are bloodborne myeloid-progenitor-derived cells that can respond rapidly to infected and damaged tissues. Monocyte infiltration also contributes significantly to the apparent microgliosis following status epilepticus (SE) [18]. However, the contributions of infiltrating monocytes and subsequent macrophages to neurological disorders, including epilepsy, have often been poorly defined. As such, this review seeks to explore the current literature regarding the role of monocytes within epileptic pathogenesis and promote novel directions for research into epileptic intervention that targets infiltrating monocytes.
2. Distinguishing Infiltrating Monocytes from Resident Microglia:
One key consideration when discussion monocyte activity within the CNS is how to distinguish them from resident microglia under pathological conditions. It is currently hypothesized that during early development, primitive macrophages contained within the yolk sac infiltrate into the nascent CNS prior to blood-blain-barrier (BBB) closure and give rise to microglia [19, 20]. Microglia are then thought to maintain their numbers via self-replication, albeit slowly [21, 22]. However, it has been postulated monocytes can also differentiate into “microglia-like” cells, which would make it difficult to distinguish these infiltrated “microglia-like” cells from true resident microglia [19, 23] (Figure 1). Nevertheless, this remains a controversial hypothesis, as many argue that monocytes can still be distinguished for microglia and are not a source of microglia in the adult CNS [18, 22, 24–26]. Further confounding this controversy, recent microglial ablation studies have argued both for and against monocytes as a source of microglial repopulation [27–29]. Thus, to fully characterize the role of monocytes within CNS pathologies, great care is needed when distinguishing between resident microglia and infiltrated monocytes. The following are some of the ways in which these types of investigations have been conducted.
Figure 1: Comparison of monocyte and macrophage characteristics.
One of the key difficulties in delineating the function of monocytes within any neurological condition is separating them from resident microglia. Differentiated macrophages, which arise from infiltrated monocytes, display similar morphologies and phenotypes as activated microglia. However, monocyte and microglia specific proteins may still be able to separate these populations. Finally, the controversial hypothesis that infiltrating monocytes can also differentiate into resident microglia within the adult CNS should be considered. Abbreviations: CCL2, chemokine (C-C motif) ligand 2; CCR2, C-C chemokine receptor type 2; CNS, central nervous system; CX3CR1, C-X3-C motif chemokine receptor 1; IBA1, ionized calcium binding adaptor molecule 1; TMEM119, transmembrane protein 119.
Some experiments have utilized bone-marrow chimeras to generate and trace labeled monocytes infiltrating into the CNS [23, 30, 31]. However, it is now accepted that utilization of lethal irradiation, which is used in the generation of these chimeric animals, is damaging to the BBB [32–34]. Therefore, the degree to which monocytes infiltrate into the CNS and the level of inflammation described in these models should be considered. However, other methods are being developed that can be used to distinguish infiltrating monocytes from resident microglia, without damaging radiation.
Flow cytometry remains a relatively popular method to investigate monocyte populations. Typical monocytes in humans can be identified by their expression of, CD14, CD16, CD64, and C-C chemokine receptor type 2 (CCR2; CD192) [35, 36]. For mice, the expression of Ly6C, CD43, CD11b, and CCR2 is used to help identify monocyte populations [37–39]. However, microglia have also been shown to express CD11b, CD14, CD16, and CD64 [40, 41]. Consequently, in disease contexts, like epilepsy, that requires separation of these two populations, more specific markers are needed. Traditionally, when microglia and monocytes need to be separated, relative expression of CD45 has often been employed [39]. However, with the recent advances in genetic profiling, markers specific to microglia have been identified (e.g., P2Y12, Transmembrane protein 119 (TMEM119), Hexosaminidase B (HexB), Olfactomedin-like protein 3 (Olfml3)) [42]. These markers are now being incorporated into fluorescence-activated cell sorting (FACS) activities [39]. Conversely, CCR2 has been shown to lacking on microglia and could also be used to separate these two cell types [43] (Figure 1).
While separation of microglia and monocytes by relative protein expression is possible with flow cytometry, direct visualization methods require more specific means. Under normal brain conditions, the small number of monocytes residing in blood vessels typically take on a small round morphology with little to no processes, while microglia are highly ramified with numerous long processes. However, under pathological conditions these differences in morphology fade away as infiltrated monocytes differentiate into macrophages, and microglia soma size increases and processes shorten [44] (Figure 1). Nevertheless, other means of investigation can be used to distinguish monocytes/macrophages from microglia, such as serial block-face scanning electron microscopy (SBF-SEM) [45]. It has also recently shown that electrophysiology approaches can be used to separate microglia and monocyte. Specifically, following SE induction activated microglia were observed to have both inward and outward potassium currents, while infiltrated monocytes displayed principally inwards currents [18].
However, genetic modulation may offer a more substantive means to tag and investigate monocytes and microglia independently. For example, CX3CR1CreER/+:R26tdTomato/+ (Ai14) mice have previously been used to separate microglia from monocytes [13, 46, 47]. By taking advantage of the fact that monocytes have a rapid turn-over rate (>90% after 3 weeks) when compared to microglia (<10% after 1 year), microglia could then be specifically labeled with tdTomato [13, 22, 47]. Then staining for a common marker, like ionized calcium binding adaptor molecule 1 (IBA1) could distinguish monocytes from microglia. Recently, more straightforward labeling methods have been developed. The use of CCR2-RFP mice has become a popular means to identify monocytes in vivo in a variety of contexts [43, 48–51]. This genetic labeling method has also been used to investigate monocytes within epilepsy [14, 15, 18]. For instance, when coupled to 2-photon imaging, CCR2-RFP labeling allowed for the identification of newly infiltrated monocytes following SE that displayed typical round morphologies [18]. It should be noted however, that CCR2 is suggested to be expressed by a number of cell types in the CNS (e.g., astrocytes, microglia) [52–54]. However, CCR2-RFP labeling seems to be specific to monocytes when investigating CNS pathologies [55].
Alternatively, microglia specific markers can be harnessed to separate monocytes from microglia. For instance, the microglial marker TMEM119 has recently been used to generate microglia specific transgenic reporter mice [56–58]. Similarly, P2Y12 has also been shown to distinguish resident microglia from infiltrated monocytes [25, 59]. As such, this marker is also being targeted for the development of genetic tools that can be utilize for the labeling and separation of microglia from monocytes [60]. However, it should be noted that under certain pathological conditions, microglia specific markers (i.e., P2Y12) can be down-regulated following activation [61, 62]. These markers, including TMEM119 and P2Y12, are also rapidly lost by microglia in culture [63]. While in vitro expression levels may not be of issue, for in vivo studies the observation that expression of these markers is not entirely consistent should be considered when designing experiments.
3. Epilepsy, Blood-Brain-Barrier Disruption, and Monocyte Infiltration:
While many studies do not attempt to separate microglia from infiltrating monocytes and subsequent macrophages, recent studies has shown that monocyte infiltration and microglial proliferation both contribute to observed hippocampal microgliosis following kainic acid (KA)-induced seizures [18]. Consequently, since it is possible that both microglia and monocytes contribute to seizure and epilepsy related pathologies, it is important to first understand how monocytes gain entry into the CNS. A key contributor to the immune privilege of the CNS is the blood-brain-barrier (BBB) [64]. The BBB is a selectively permeable boundary composed of brain microvascular endothelial cells (BMECs) supported by pericytes and the end-feet of astrocytes [65–67]. Generally, the BBB will only allow the passage of water, glucose, and other essential nutrients while excluding most other serum factors [68]. Moreover, under normal physiological conditions the BBB limits the passage of peripheral immune cells into the CNS [69]. However, under pathological condition the BBB can become compromised allowing previously excluded serum components and immune cells into the CNS inducing numerous adverse effects (Figure 2).
Figure 2: Blood-brain-barrier under physiological and pathological conditions.
The BBB is an integral part of healthy brain function. Under typical conditions the BBB is composed of tightly associated BMECs, supported by basement membrane, pericytes, and astrocyte endfeet. It will strictly limit passage into the greater brain to only select factors. However, under pathological conditions this tight barrier can become permeable allowing for previously excluded factors into the brain. Moreover, damage to the BBB can facilitate release of chemoattractants such as CCL2 into the bloodstream leading to the recruitment of monocytes to the CNS. Abbreviations: BBB, blood-brain-barrier; BMEC, brain microvascular endothelial cell; CCL2, chemokine (C-C motif) ligand 2; CNS, central nervous system.
The most notable example of BBB disruption is in stroke, particularly ischemic stroke. Specifically, endothelial tight junction degradation, breakdown of the underlying basement membrane, and modulation of cellular transport mechanism have been observed within the first few hours following ischemic stroke [70, 71]. In regards to epilepsy, there is significant evidence indicating that during the acute phase there is significant opening of the BBB similar to that observed during stroke [72]. This has been seen in humans and both infection-related (e.g., viral encephalitis) and “sterile” (e.g., pilocarpine, KA) animal models of epilepsy [15, 73, 74]. Moreover, studies have also shown that BBB disruption can directly induce seizure activity and exacerbate epileptogenesis [75–78].
However, while under normal circumstances the BBB regulates monocyte entry into then CNS, BBB disruption does not necessitate immediate infiltration of monocytes into the CNS. For example, following SE induction via intraperitoneal (i.p.) KA monocyte infiltration was not observed till 24 hours later [14, 15]. Our own preliminary investigations also observed that monocyte infiltration is delayed into the hippocampus following seizure induced BBB disruption, even though BBB permeability is rapidly disrupted (<6 hours). This can also be seen in other types of injury, such as traumatic brain injury (TBI), where neutrophils are observed to rapidly infiltrate into areas where the BBB is disrupted, while monocytes/macrophages only begin to appear 24 hours later [79]. Furthermore, MRI studies of experimental allergic encephalomyelitis (EAE) demonstrated that BBB disruption and monocyte infiltration are two distinct events occurring days apart [80]. While BBB disruption does have notable pathological consequences (e.g., leakage of plasma proteins, ion imbalance, neuronal dysfunction), it alone may not result in significant monocyte infiltration [79–81]. However, BBB disruption could facilitate the release of chemoattractants into circulation. Thus, it is important to discuss the possible factors that may facilitate monocyte attraction into seizure-injured brain regions.
4. Importance of the CCL2-CCR2 Axis to Monocyte Recruitment:
It is becoming increasingly apparent that the chemokine (C-C motif) ligand 2 (CCL2)-CCR2 axis is of particular importance to the activities of monocytes within the CNS and elsewhere. Expression of CCL2 seems to be related to a number of neuropathologies including cerebral ischemia, Alzheimer’s disease, and traumatic brain injury [82–84]. Furthermore, CCL2 is highly up-regulated in patients suffering from human immunodeficiency virus (HIV) associated neurocognitive disorders, which facilitates the attraction of monocytes into the brain [85, 86]. In regards to seizures and epilepsy, several studies have shown that CCL2 is extremely up-regulated, as much as 200 fold, in the brain following SE and in epileptic patients [14, 15, 87–89]. It has also been shown that cerebral CCL2 expression is elevated in patient’s suffering from intractable epilepsy [90–92]. Furthermore, systematic administration of the CCL2 transcription inhibitor, bindarit, or CCR2 antagonist, RS102895, suppressed the seizure exacerbating effects of lipopolysaccharide (LPS) [87]. A number of sources of CNS CCL2 have been described in the literature, including astrocytes, microglia, and endothelial cells. However, considering that CCL2 released from damaged neurons has been shown to be principal to the development and progression of neuropathic pain and encephalopathy, it is likely also the driver of the apparent increase following seizure. Indeed, it was found that CCL2 expression was mostly upregulated in microglia and neurons following SE [14] (Figure 3A).
Figure 3: CCL2 drives monocytes infiltration and activation after seizures.
(A) It has been shown that following seizures, neurons and microglia will secrete high levels of CCL2. This chemokine may then pass into circulation, attracting monocytes into the CNS via CCR2 signaling. (B) It has also been demonstrated that CCL2 may induce monocyte-derived macrophages to secrete the inflammatory cytokine IL-1β via STAT3 signaling mechanisms. IL-1β could then contribute to neuroinflammation and seizure related pathology. Abbreviations: CCL2, chemokine (C-C motif) ligand 2; CCR2, C-C chemokine receptor type 2; CNS, central nervous system; IL-1β, Interleukin 1 beta; STAT3, signal transducer and activator of transcription 3.
As a chemoattractant, CCL2 expression significantly influences monocyte recruitment via CCR2 signaling [93]. So much so that an alternative name for CCL2 is monocyte chemoattractant protein 1 (MCP-1). Additionally, CCR2+ monocytes are typically considered to be inflammatory [94]. Studies using CCR2 KO mice have shown stunted infiltration of monocytes following ischemia, traumatic brain injury, and experimental autoimmune encephalomyelitis [74, 95–97]. It has also been shown that neuronal expression of CCL2 following Theiler’s Murine Encephalomyelitis Virus (TMEV) infections will direct monocytes to infiltrate into the CNS [98]. Further demonstrating the importance of CCL2-CCR2, when CCR2 was knocked-out there was a significant reduction in monocytes infiltration into the hippocampus following kainic-acid-induced SE [14, 15]. Interestingly, one report showed that in La Crosse Virus encephalitis entry of Ly6Chi monocytes into the CNS was CCR2 independent, while still being necessary for their movement within the CNS [99].
In addition to providing a chemoattractant signal for monocytes, CCL2 may also support monocyte recruitment by increasing the permeability of the BBB. Several studies have shown that direct injection of CCL2 into the brain will result in BBB disruption and that this effect is plasmin-dependent [100–104]. Studies investigation the plasmin-dependence of CCL2-mediated BBB disruption also observed increased monocyte infiltration that correlated with increased plasmin activity [105, 106]. However, CCL2-mediate BBB disruption also seems to be CCR2-dependent. For instance, CCL2 biding to CCR2 receptors located on BMECs will disrupt the integrity of BBB tight junctions [107]. It has also demonstrated that CCR2 deficiency will reduced BBB disruption due to both seizures and stroke [15, 108]. There is also indication that monocytes will participate in CCL2 mediated BBB disruption, likely because of CCR2 [103]. Consequently, it is possible that following an initial neuropathological event, like a seizure, BBB disruption will allow for release of upregulated CCL2, thereby attracting monocytes, which in turn can further exacerbate BBB permeability.
However, it is unclear if sustained CCL2-CCR2 signaling is necessary for monocyte activities once they are recruited to the CNS. Following SE some infiltrated CCR2+ monocytes within the hippocampus were observed to also express C-X3-C motif chemokine receptor 1 (CX3CR1), and display morphological alterations (i.e., development of processes) [18]. It has also been shown that CCR2+ monocytes are only a portion of the total circulating monocyte population [109–111]. Moreover, inflammation related CCR2+ monocytes typically express low levels of CX3CR1, while non-inflammatory monocytes express low CCR2 but high CX3CR1 [111]. This dual CCR2+:CX3CR1+ population may represent an intermediate cell type that is transitioning away from a pro-inflammatory phenotype. This is also suggested to occur with infiltrated monocytes responding to ischemic stroke [112]. Nevertheless, even if monocytes can transition after arriving into the CNS, the initially responding CCR2+ monocytes would still be considered inflammatory. Thus, it is important to consider the role monocytes may have in promoting inflammation within epilepsy.
5. CNS Inflammation and Epilepsy:
CNS inflammation is now recognized as a key mediator of SE and epilepsy pathogenesis [1, 113–115]. It has also been suggested that neuroinflammatory pathways and factors are important targets for therapeutic intervention [116]. For instance, reducing oxidative stress and reactive oxygen species levels can improve epilepsy pathology [117, 118]. The effects of inflammation can be further magnified if there is also a pre-conditioning source (e.g., LPS stimulation) [119, 120]. Extrinsic inflammatory conditions such as obesity have also been linked to more sever seizure effects [121].
Additionally, a number of immunological cell types have been related to seizure and epilepsy induced inflammation. Several clinical reports have shown increased neutrophil levels within epileptic patients, which is a noted indicator of inflammation [122, 123]. Moreover, pro-inflammatory CD4+ and CD8+ T cells have been shown to significantly infiltrate into both the neocortex and hippocampus of mice shortly after seizure (<24h) and within the hippocampus of epilepsy patients [124, 125]. Production of pro-inflammatory IL-17 by these cells may also contribute to refractory pediatric epilepsy [126]. Interestingly though, it has also been observed that within recombination activating gene 1 (RAG1) KO mice, which lack T and B cells, there was significantly more neurodegeneration and an earlier onset of spontaneous recurrent seizures following KA treatment [127]. Thus, there are clear implications for innate immune response in seizure and epilepsy pathology.
While infiltrating monocytes have also been suggested to promote inflammation after SE, relatively little is known about the mechanisms by which monocytes can promote inflammation under epileptic conditions [15]. Besides CCL2, several other pro-inflammatory cytokines have been linked to epileptogenesis such as interleukin 6 (Il-6), IL-1β, and tumor necrosis factor alpha (TNF-α) [128–130]. Monocytes have been shown to express IL-6, while IL-1β and TNF-α will affect monocyte recruitment and function [14, 131–133]. Moreover, SE-induced expression of pro-inflammatory IL-1β was almost completely abolished within CCR2 KO mice [14]. It was then determined that IL-1β was being produced by both microglia and monocyte-derived macrophages in a signal transducer and activator of transcription 3 (STAT3) dependent manner (Figure 3B). With CCL2-CCR2 being possibly linked to neuroinflammation after infiltration, it further highlights the question as to the role of CCR2 dependent mechanisms beyond recruitment.
6. Effect of Monocytes upon Status Epilepticus and Epilepsy:
While there is evidence that the immune system has a role in aggregate epileptogenesis, there are several causes of epilepsy that are directly related to immunological effectors. Notably, epilepsy can arises due to viral encephalitis (VE), a viral infection that produces inflammation within the brain parenchyma. VE has been shown to induce seizures during the acute infection period, and later unprovoked epileptic events following infection resolution. In regards to monocytes there is clear evidence demonstrating a significant CCL2-dependent infiltration of monocyte into the CNS following viral infections, specifically into the hippocampus [98]. Studies utilizing CCR2 KO mice showed fewer activated myeloid cells, ostensibly monocytes, correlated with reduced hippocampal damage, but not seizure presentation [46]. Additionally, immunodepletion and adaptive bone marrow transfer experiments demonstrated a potential role for inflammatory monocytes in hippocampal damage [134, 135].
However, other studies involving depletion of macrophages, which are derived from monocytes, reduced the number of acute seizures and epileptic events due to VE, but not hippocampal damage [136–138]. It has also been suggested that this effect on seizures is dependent upon macrophage IL-6 expression [136, 139]. Furthermore, direct injection of monocytes into the brain seemed to exacerbate the seizure burden of TMEV infected mice [138, 140]. Consequently, it would seem the role monocytes play is still inconclusive. It is possible that alternative monocyte-derived populations, like differentiated macrophages, may affect seizure severity and hippocampal damage in different ways. Another possible explanation may be that microglia contribute to hippocampal damage, and that this activity is somehow affected by CCR2 KO, while infiltrating monocytes can affect seizure burden in a CCR2 independent fashion. Regardless, more research is needed to conclusively determine the role of monocytes in VE-induced seizures.
Viral infection is not the only means by which epilepsy and seizures can occur, consequently investigations into the role of monocytes in “sterile” models of SE and epilepsy are also important. Most, non-infection related models of SE and epilepsy utilize either kainic acid (KA) or pilocarpine to induce seizures and have often been correlated with temporal lobe epilepsy (TLB) [141–143]. As mentioned previously, CCR2+ monocyte infiltration has been suggested to promote inflammation following KA-induced SE, which in turn was correlated with neuronal damage but not seizure severity [14, 15]. Further, studies utilizing CCR2 KO mice revealed similar results indicating that monocyte infiltration exacerbates the pathological effects of KA-induced SE [14, 15]. However, it has also been shown that i.v. transplantation of bone marrow mononuclear cells (BMMCs) into lithium–pilocarpine-treated rats reduced seizure frequency and improved spatial memory deficits [144]. It should be noted though that the identity of these BMMCs was not determined, so this effect may not be due specifically to monocytes. Moreover, if these cells did contain monocytes, they may present alternative anti-inflammatory phenotypes not typically associated with monocytes that naturally respond to CNS pathologies (i.e., inflammatory CCR2+ monocytes).
It is also possible that differences in the pathological mechanisms governing the various models of epileptogenesis could result in the observed discrepancies in monocyte-related activity. When blood monocytes from patients suffering from Neurocysticercosis (NCC), a disease related to Taenia solium (T. solium) infection and a leading cause of epilepsy in the developing world, were compared to those derived from patients with idiopathic epilepsy via microarray analysis, it was revealed that NCC-related monocytes had higher association with inflammatory/immune pathways than idiopathic epilepsy patients [145]. It has also been suggested that NCC will induce inflammatory monocyte-astrocyte crosstalk [146]. Consequently, when determining the role monocytes have within epilepsy it is important to not only consider their relation to CNS factors, but also the root cause of the condition.
Finally, seizure and epileptic conditions have been liked to a number of co-morbidities including anxiety, depression, and memory deficits [147, 148]. Infiltration of monocytes into the CNS has also been shown to affect these behavioral and memory effects in other conditions such as prolonged stress and Alzheimer’s disease [149–151]. Furthermore, stress-induced anxiety may be related to recruitment of IL-1β producing monocytes [152]. In regards to epilepsy, when CCR2 KO mice were evaluated for anxiety and memory following KA-induced SE, it was found that CCR2 KO mice had better performance in these evaluations than did wild-type mice. These improvements were also correlated with reduced monocyte infiltration and microglial activation [14]. Consequently, while more work needs to be done to evaluate the contribution of monocytes to epilepsy-related co-morbidities, there are indications that infiltrating monocytes can modulate a wide range of effects (Table 1).
Table 1:
Selected references and key findings on monocytes in seizures and epilepsy.
Model | Administration | Species | Key Findings | Reference |
---|---|---|---|---|
Clinical | N/a | Humans |
|
[145] |
Kainic Acid | Intracerebroventricle (ICV) | Mice |
|
[14] |
Kainic Acid | ICV | Mice |
|
[18] |
Kainic Acid | ICV | Rats |
|
[155] |
Kainic Acid, Pilocarpine |
Intraperitoneal (IP) | Mice |
|
[15] |
Pilocarpine | IP | Rats |
|
[144] |
Pilocarpine | IP | Rats |
|
[154] |
Viral Encephalitis | Theiler’s murine encephalomyelitis virus (TMEV) Daniels (DA) strain Parietal cortex injection |
Mice |
|
[136] |
Viral Encephalitis | TMEV DA strain Intracerebral (IC) |
Mice |
|
[137] |
Viral Encephalitis | TMEV DA strain Intracerebral (IC) |
Mice |
|
[138] |
Viral Encephalitis | TMEV DA strain Intracerebral (IC) |
Mice |
|
[139] |
Viral Encephalitis | TMEV DA strain Intracerebral (IC) |
Mice |
|
[46] |
7. Interplay between Monocytes and Microglia within Seizure and Epilepsy Pathology:
Finally, it is important to note the possibility of combinatory and compensatory effects of monocytes and microglia within the context of seizure and epilepsy pathologies. As previously noted, monocyte recruitment and microglial proliferation both contribute to the apparent microgliosis following seizure [18]. This may result in a possibly compensatory effect when either microglia or monocytes are targeted by an intervention. Moreover, it has been suggested that microglia can act as a source of CCL2, which may facilitate monocyte recruitment [153]. This could mean that activation of microglia could result in further infiltration of monocytes and microgliosis. Yet, the degree to which either cell type contributes to seizure and epilepsy pathology remains largely unknown. Our own work indicates however, that both monocyte-derived macrophages and microglia may contribute to pathological IL-1β production after seizure [14]. Consequently, future explorations into the role of monocytes and microglia should consider how each cell type contributes to the observations. With the novel molecular tools currently being developed these investigations should become easier to perform.
8. Conclusion and Future Directions:
In summary, lines of evidence have shown that monocytes play a substantive role in pathogenesis of seizures and epilepsy. Now that new tools are being developed that can separate monocyte populations from resident microglia, there is significant opportunity in trying to investigate and target these cells for therapeutic purposes. For instance several of the previously cited studies have shown modulation of the CCL2-CCR2 axis will reduce monocyte infiltration and improve seizure-related outcomes. Additionally, Roscovitine, a cyclin-dependent kinase 5 (CDK5) inhibitor, was shown to mitigate monocyte infiltration and suppress SE-induced neuroinflammation [154]. Another possible avenue is directing infiltrated monocytes towards anti-inflammatory and restorative activities. It has been recently reported that 2-Deoxyglucose treatment will shift monocytes towards a restorative macrophage phenotype following KA induced seizures, which resulted in reduced hippocampal neuron loss [155]. However, several questions still remain. For example, the exact role of monocytes in epilepsy is still relatively unclear as there are conflicting reports depending on the animal model of epilepsy. It also remains unclear whether monocytes that do not express high levels of CCR2 have a role in epileptogenesis. Nevertheless, infiltrating monocytes offer novel possible research directions for the treatment of seizures and epilepsy.
Acknowledgements:
This work is supported by National Institute of Health (R01NS088627, R01NS112144).
Abbreviations:
- ALS
amyotrophic lateral sclerosis
- BBB
blood-brain-barrier
- BMEC
brain microvascular endothelial cell
- BMMC
bone marrow mononuclear cells
- CCL2
chemokine (C-C motif) ligand 2
- CCR2
C-C chemokine receptor type 2
- CCR2-RFP
CCR2 knock-in red fluorescent protein
- CNS
central nervous system
- CX3CR1
C-X3-C motif chemokine receptor 1
- FACS
fluorescence-activated cell sorting
- HexB
hexosaminidase B
- HIV
human immunodeficiency virus
- IBA1
ionized calcium binding adaptor molecule 1
- IL-1β
Interleukin 1 beta
- IL-6
Interleukin 6
- KA
kainic acid
- LPS
lipopolysaccharide
- NCC
neurocysticercosis
- Olfml3
olfactomedin-like protein 3
- RAG1
recombination activating gene 1
- SBF-SEM
serial block-face scanning electron microscopy
- SE
status epilepticus
- STAT3
signal transducer and activator of transcription 3
- TBI
traumatic brain injury
- TMEM119
transmembrane protein 119
- TMEV
Theiler’s murine encephalomyelitis virus
- TNF-α
tumor necrosis factor alpha
- VE
viral encephalitis
Footnotes
Conflict of Interest:
The authors declare no competing interests.
References:
- 1.Eyo UB, Murugan M & Wu LJ (2017) Microglia-Neuron Communication in Epilepsy, Glia. 65, 5–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Hirtz D, Thurman DJ, Gwinn-Hardy K, Mohamed M, Chaudhuri AR & Zalutsky R (2007) How common are the “common” neurologic disorders?, Neurology. 68, 326–37. [DOI] [PubMed] [Google Scholar]
- 3.Laxer KD, Trinka E, Hirsch LJ, Cendes F, Langfitt J, Delanty N, Resnick T & Benbadis SR (2014) The consequences of refractory epilepsy and its treatment, Epilepsy & Behavior. 37, 59–70. [DOI] [PubMed] [Google Scholar]
- 4.Inoue K & Tsuda M (2018) Microglia in neuropathic pain: cellular and molecular mechanisms and therapeutic potential, Nat Rev Neurosci. 19, 138–152. [DOI] [PubMed] [Google Scholar]
- 5.Kim E & Cho S (2016) Microglia and Monocyte-Derived Macrophages in Stroke, Neurotherapeutics. 13, 702–718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Wes PD, Sayed FA, Bard F & Gan L (2016) Targeting microglia for the treatment of Alzheimer’s Disease, Glia. 64, 1710–32. [DOI] [PubMed] [Google Scholar]
- 7.Zhuo M, Wu G & Wu L-J (2011) Neuronal and microglial mechanisms of neuropathic pain, Molecular Brain. 4, 31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Qin C, Zhou L-Q, Ma X-T, Hu Z-W, Yang S, Chen M, Bosco DB, Wu L-J & Tian D-S (2019) Dual Functions of Microglia in Ischemic Stroke, Neuroscience Bulletin. 35, 921–933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Benson MJ, Manzanero S & Borges K (2015) Complex alterations in microglial M1/M2 markers during the development of epilepsy in two mouse models, Epilepsia. 56, 895–905. [DOI] [PubMed] [Google Scholar]
- 10.Luo C, Koyama R & Ikegaya Y (2016) Microglia engulf viable newborn cells in the epileptic dentate gyrus, Glia. 64, 1508–1517. [DOI] [PubMed] [Google Scholar]
- 11.Hiragi T, Ikegaya Y & Koyama R (2018) Microglia after Seizures and in Epilepsy, Cells. 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Zhao X, Liao Y, Morgan S, Mathur R, Feustel P, Mazurkiewicz J, Qian J, Chang J, Mathern GW, Adamo MA, Ritaccio AL, Gruenthal M, Zhu X & Huang Y (2018) Noninflammatory Changes of Microglia Are Sufficient to Cause Epilepsy, Cell Rep. 22, 2080–2093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Peng J, Gu N, Zhou L, B Eyo U, Murugan M, Gan W-B & Wu L-J (2016) Microglia and monocytes synergistically promote the transition from acute to chronic pain after nerve injury, Nature Communications. 7, 12029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Tian D-S, Peng J, Murugan M, Feng L-J, Liu J-L, Eyo UB, Zhou L-J, Mogilevsky R, Wang W & Wu L-J (2017) Chemokine CCL2–CCR2 Signaling Induces Neuronal Cell Death via STAT3 Activation and IL-1β Production after Status Epilepticus, The Journal of Neuroscience. 37, 7878–7892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Varvel NH, Neher JJ, Bosch A, Wang W, Ransohoff RM, Miller RJ & Dingledine R (2016) Infiltrating monocytes promote brain inflammation and exacerbate neuronal damage after status epilepticus, Proceedings of the National Academy of Sciences of the United States of America. 113, E5665–E5674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.ElAli A & Jean LeBlanc N (2016) The Role of Monocytes in Ischemic Stroke Pathobiology: New Avenues to Explore, Front Aging Neurosci. 8, 29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Zondler L, Muller K, Khalaji S, Bliederhauser C, Ruf WP, Grozdanov V, Thiemann M, Fundel-Clemes K, Freischmidt A, Holzmann K, Strobel B, Weydt P, Witting A, Thal DR, Helferich AM, Hengerer B, Gottschalk KE, Hill O, Kluge M, Ludolph AC, Danzer KM & Weishaupt JH (2016) Peripheral monocytes are functionally altered and invade the CNS in ALS patients, Acta Neuropathol. 132, 391–411. [DOI] [PubMed] [Google Scholar]
- 18.Feng L, Murugan M, Bosco DB, Liu Y, Peng J, Worrell GA, Wang HL, Ta LE, Richardson JR, Shen Y & Wu LJ (2019) Microglial proliferation and monocyte infiltration contribute to microgliosis following status epilepticus, Glia. 67, 1434–1448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ginhoux F, Lim S, Hoeffel G, Low D & Huber T (2013) Origin and differentiation of microglia, Front Cell Neurosci. 7, 45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Menassa DA & Gomez-Nicola D (2018) Microglial Dynamics During Human Brain Development, Front Immunol. 9, 1014–1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Réu P, Khosravi A, Bernard S, Mold JE, Salehpour M, Alkass K, Perl S, Tisdale J, Possnert G, Druid H & Frisén J (2017) The Lifespan and Turnover of Microglia in the Human Brain, Cell reports. 20, 779–784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ajami B, Bennett JL, Krieger C, Tetzlaff W & Rossi FMV (2007) Local self-renewal can sustain CNS microglia maintenance and function throughout adult life, Nature Neuroscience. 10, 1538–1543. [DOI] [PubMed] [Google Scholar]
- 23.Djukic M, Mildner A, Schmidt H, Czesnik D, Brück W, Priller J, Nau R & Prinz M (2006) Circulating monocytes engraft in the brain, differentiate into microglia and contribute to the pathology following meningitis in mice, Brain. 129, 2394–2403. [DOI] [PubMed] [Google Scholar]
- 24.London A, Cohen M & Schwartz M (2013) Microglia and monocyte-derived macrophages: functionally distinct populations that act in concert in CNS plasticity and repair, Frontiers in Cellular Neuroscience. 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Gu N, Peng J, Murugan M, Wang X, Eyo Ukpong B., Sun D, Ren Y, DiCicco-Bloom E, Young W, Dong H & Wu L-J (2016) Spinal Microgliosis Due to Resident Microglial Proliferation Is Required for Pain Hypersensitivity after Peripheral Nerve Injury, Cell Reports. 16, 605–614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Cronk JC, Filiano AJ, Louveau A, Marin I, Marsh R, Ji E, Goldman DH, Smirnov I, Geraci N, Acton S, Overall CC & Kipnis J (2018) Peripherally derived macrophages can engraft the brain independent of irradiation and maintain an identity distinct from microglia, The Journal of Experimental Medicine. 215, 1627–1647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Lund H, Pieber M, Parsa R, Han J, Grommisch D, Ewing E, Kular L, Needhamsen M, Espinosa A, Nilsson E, Överby AK, Butovsky O, Jagodic M, Zhang X-M & Harris RA (2018) Competitive repopulation of an empty microglial niche yields functionally distinct subsets of microglia-like cells, Nature communications. 9, 4845–4845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Huang Y, Xu Z, Xiong S, Sun F, Qin G, Hu G, Wang J, Zhao L, Liang YX, Wu T, Lu Z, Humayun MS, So KF, Pan Y, Li N, Yuan TF, Rao Y & Peng B (2018) Repopulated microglia are solely derived from the proliferation of residual microglia after acute depletion, Nat Neurosci. 21, 530–540. [DOI] [PubMed] [Google Scholar]
- 29.Varvel NH, Grathwohl SA, Baumann F, Liebig C, Bosch A, Brawek B, Thal DR, Charo IF, Heppner FL, Aguzzi A, Garaschuk O, Ransohoff RM & Jucker M (2012) Microglial repopulation model reveals a robust homeostatic process for replacing CNS myeloid cells, Proceedings of the National Academy of Sciences of the United States of America. 109, 18150–18155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Flugel A, Bradl M, Kreutzberg GW & Graeber MB (2001) Transformation of donor-derived bone marrow precursors into host microglia during autoimmune CNS inflammation and during the retrograde response to axotomy, J Neurosci Res. 66, 74–82. [DOI] [PubMed] [Google Scholar]
- 31.Priller J, Flügel A, Wehner T, Boentert M, Haas CA, Prinz M, Fernández-Klett F, Prass K, Bechmann I, de Boer BA, Frotscher M, Kreutzberg GW, Persons DA & Dirnagl U (2001) Targeting gene-modified hematopoietic cells to the central nervous system: Use of green fluorescent protein uncovers microglial engraftment, Nat Med. 7, 1356–1361. [DOI] [PubMed] [Google Scholar]
- 32.Kierdorf K, Katzmarski N, Haas CA & Prinz M (2013) Bone marrow cell recruitment to the brain in the absence of irradiation or parabiosis bias, PLoS One. 8, e58544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Larochelle A, Bellavance M-A, Michaud J-P & Rivest S (2016) Bone marrow-derived macrophages and the CNS: An update on the use of experimental chimeric mouse models and bone marrow transplantation in neurological disorders, Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 1862, 310–322. [DOI] [PubMed] [Google Scholar]
- 34.Tashima R, Mikuriya S, Tomiyama D, Shiratori-Hayashi M, Yamashita T, Kohro Y, Tozaki-Saitoh H, Inoue K & Tsuda M (2016) Bone marrow-derived cells in the population of spinal microglia after peripheral nerve injury, Scientific Reports. 6, 23701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ziegler-Heitbrock HWL (2000) Definition of human blood monocytes, Journal of Leukocyte Biology. 67, 603–606. [DOI] [PubMed] [Google Scholar]
- 36.Ong S-M, Teng K, Newell E, Chen H, Chen J, Loy T, Yeo T-W, Fink K & Wong S-C (2019) A Novel, Five-Marker Alternative to CD16–CD14 Gating to Identify the Three Human Monocyte Subsets, Front Immunol. 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ritzel RM, Patel AR, Grenier JM, Crapser J, Verma R, Jellison ER & McCullough LD (2015) Functional differences between microglia and monocytes after ischemic stroke, Journal of Neuroinflammation. 12, 106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Sunderkötter C, Nikolic T, Dillon MJ, van Rooijen N, Stehling M, Drevets DA & Leenen PJM (2004) Subpopulations of Mouse Blood Monocytes Differ in Maturation Stage and Inflammatory Response, The Journal of Immunology. 172, 4410–4417. [DOI] [PubMed] [Google Scholar]
- 39.Li Q, Lan X, Han X & Wang J (2019) Expression of Tmem119/Sall1 and Ccr2/CD69 in FACS-Sorted Microglia- and Monocyte/Macrophage-Enriched Cell Populations After Intracerebral Hemorrhage, Frontiers in cellular neuroscience. 12, 520–520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Jeong H-K, Ji K, Min K & Joe E-H (2013) Brain inflammation and microglia: facts and misconceptions, Exp Neurobiol. 22, 59–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Walker DG & Lue L-F (2015) Immune phenotypes of microglia in human neurodegenerative disease: challenges to detecting microglial polarization in human brains, Alzheimers Res Ther. 7, 56–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Hickman SE, Kingery ND, Ohsumi TK, Borowsky ML, Wang LC, Means TK & El Khoury J (2013) The microglial sensome revealed by direct RNA sequencing, Nat Neurosci. 16, 1896–905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Mizutani M, Pino PA, Saederup N, Charo IF, Ransohoff RM & Cardona AE (2012) The fractalkine receptor but not CCR2 is present on microglia from embryonic development throughout adulthood, Journal of immunology (Baltimore, Md : 1950). 188, 29–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kettenmann H, Hanisch UK, Noda M & Verkhratsky A (2011) Physiology of microglia, Physiol Rev. 91, 461–553. [DOI] [PubMed] [Google Scholar]
- 45.Yamasaki R, Lu H, Butovsky O, Ohno N, Rietsch AM, Cialic R, Wu PM, Doykan CE, Lin J, Cotleur AC, Kidd G, Zorlu MM, Sun N, Hu W, Liu L, Lee J-C, Taylor SE, Uehlein L, Dixon D, Gu J, Floruta CM, Zhu M, Charo IF, Weiner HL & Ransohoff RM (2014) Differential roles of microglia and monocytes in the inflamed central nervous system, The Journal of experimental medicine. 211, 1533–1549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kaufer C, Chhatbar C, Broer S, Waltl I, Ghita L, Gerhauser I, Kalinke U & Loscher W (2018) Chemokine receptors CCR2 and CX3CR1 regulate viral encephalitis-induced hippocampal damage but not seizures, Proc Natl Acad Sci U S A. 115, E8929–e8938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Parkhurst Christopher N., Yang G, Ninan I, Savas Jeffrey N., Yates John R., Lafaille Juan J., Hempstead Barbara L., Littman Dan R. & Gan W-B (2013) Microglia Promote Learning-Dependent Synapse Formation through Brain-Derived Neurotrophic Factor, Cell. 155, 1596–1609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Kuroda N, Masuya M, Tawara I, Tsuboi J, Yoneda M, Nishikawa K, Kageyama Y, Hachiya K, Ohishi K, Miwa H, Yamada R, Hamada Y, Tanaka K, Kato T, Takei Y & Katayama N (2019) Infiltrating CCR2+ monocytes and their progenies, fibrocytes, contribute to colon fibrosis by inhibiting collagen degradation through the production of TIMP-1, Scientific Reports. 9, 8568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Garcia-Bonilla L, Faraco G, Moore J, Murphy M, Racchumi G, Srinivasan J, Brea D, Iadecola C & Anrather J (2016) Spatio-temporal profile, phenotypic diversity, and fate of recruited monocytes into the post-ischemic brain, Journal of neuroinflammation. 13, 285–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Madsen DH, Jurgensen HJ, Siersbaek MS, Kuczek DE, Grey Cloud L, Liu S, Behrendt N, Grontved L, Weigert R & Bugge TH (2017) Tumor-Associated Macrophages Derived from Circulating Inflammatory Monocytes Degrade Collagen through Cellular Uptake, Cell Rep. 21, 3662–3671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kim AJ, Xu N, Umeyama K, Hulin A, Ponny SR, Vagnozzi RJ, Green EA, Hanson P, McManus BM, Nagashima H & Yutzey KE (2020) Deficiency of Circulating Monocytes Ameliorates the Progression of Myxomatous Valve Degeneration in Marfan Syndrome, Circulation. 141, 132–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Banisadr G, Gosselin RD, Mechighel P, Rostene W, Kitabgi P & Melik Parsadaniantz S (2005) Constitutive neuronal expression of CCR2 chemokine receptor and its colocalization with neurotransmitters in normal rat brain: functional effect of MCP-1/CCL2 on calcium mobilization in primary cultured neurons, J Comp Neurol. 492, 178–92. [DOI] [PubMed] [Google Scholar]
- 53.El Khoury J, Toft M, Hickman SE, Means TK, Terada K, Geula C & Luster AD (2007) Ccr2 deficiency impairs microglial accumulation and accelerates progression of Alzheimer-like disease, Nat Med. 13, 432–438. [DOI] [PubMed] [Google Scholar]
- 54.Kawaguchi-Niida M, Yamamoto T, Kato Y, Inose Y & Shibata N (2013) MCP-1/CCR2 signaling-mediated astrocytosis is accelerated in a transgenic mouse model of SOD1-mutated familial ALS, Acta Neuropathologica Communications. 1, 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Saederup N, Cardona AE, Croft K, Mizutani M, Cotleur AC, Tsou C-L, Ransohoff RM & Charo IF (2010) Selective chemokine receptor usage by central nervous system myeloid cells in CCR2-red fluorescent protein knock-in mice, PloS one. 5, e13693–e13693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Kaiser T & Feng G (2019) Tmem119-EGFP and Tmem119-CreERT2 Transgenic Mice for Labeling and Manipulating Microglia, eNeuro. 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Ruan C, Sun L, Kroshilina A, Beckers L, De Jager P, Bradshaw EM, Hasson SA, Yang G & Elyaman W (2020) A novel Tmem119-tdTomato reporter mouse model for studying microglia in the central nervous system, Brain, Behavior, and Immunity. 83, 180–191. [DOI] [PubMed] [Google Scholar]
- 58.Bennett ML, Bennett FC, Liddelow SA, Ajami B, Zamanian JL, Fernhoff NB, Mulinyawe SB, Bohlen CJ, Adil A, Tucker A, Weissman IL, Chang EF, Li G, Grant GA, Hayden Gephart MG & Barres BA (2016) New tools for studying microglia in the mouse and human CNS, Proceedings of the National Academy of Sciences. 113, E1738–E1746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Butovsky O, Jedrychowski MP, Moore CS, Cialic R, Lanser AJ, Gabriely G, Koeglsperger T, Dake B, Wu PM, Doykan CE, Fanek Z, Liu L, Chen Z, Rothstein JD, Ransohoff RM, Gygi SP, Antel JP & Weiner HL (2014) Identification of a unique TGF-β–dependent molecular and functional signature in microglia, Nature Neuroscience. 17, 131–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.McKinsey GL, Lizama CO, Keown-Lang AE, Niu A, Chee E, Santander N & Arnold TD (2019) New Tools for Genetically Targeting Myeloid Populations in the Central Nervous System, bioRxiv, 2019.12.20.885244. [Google Scholar]
- 61.Haynes SE, Hollopeter G, Yang G, Kurpius D, Dailey ME, Gan WB & Julius D (2006) The P2Y12 receptor regulates microglial activation by extracellular nucleotides, Nat Neurosci. 9, 1512–9. [DOI] [PubMed] [Google Scholar]
- 62.Gelosa P, Lecca D, Fumagalli M, Wypych D, Pignieri A, Cimino M, Verderio C, Enerback M, Nikookhesal E, Tremoli E, Abbracchio MP & Sironi L (2014) Microglia is a key player in the reduction of stroke damage promoted by the new antithrombotic agent ticagrelor, J Cereb Blood Flow Metab. 34, 979–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Bohlen CJ, Bennett FC, Tucker AF, Collins HY, Mulinyawe SB & Barres BA (2017) Diverse Requirements for Microglial Survival, Specification, and Function Revealed by Defined-Medium Cultures, Neuron. 94, 759–773 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Pachter JS, de Vries HE & Fabry Z (2003) The blood-brain barrier and its role in immune privilege in the central nervous system, J Neuropathol Exp Neurol. 62, 593–604. [DOI] [PubMed] [Google Scholar]
- 65.Armulik A, Genové G, Mäe M, Nisancioglu MH, Wallgard E, Niaudet C, He L, Norlin J, Lindblom P, Strittmatter K, Johansson BR & Betsholtz C (2010) Pericytes regulate the blood–brain barrier, Nature. 468, 557–561. [DOI] [PubMed] [Google Scholar]
- 66.Alvarez JI, Katayama T & Prat A (2013) Glial influence on the blood brain barrier, Glia. 61, 1939–1958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Abbott NJ (2002) Astrocyte-endothelial interactions and blood-brain barrier permeability, J Anat. 200, 629–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Blanchette M & Daneman R (2015) Formation and maintenance of the BBB, Mech Dev. 138 Pt 1, 8–16. [DOI] [PubMed] [Google Scholar]
- 69.Muldoon LL, Alvarez JI, Begley DJ, Boado RJ, Del Zoppo GJ, Doolittle ND, Engelhardt B, Hallenbeck JM, Lonser RR, Ohlfest JR, Prat A, Scarpa M, Smeyne RJ, Drewes LR & Neuwelt EA (2013) Immunologic privilege in the central nervous system and the blood-brain barrier, Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism. 33, 13–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Jiang X, Andjelkovic AV, Zhu L, Yang T, Bennett MVL, Chen J, Keep RF & Shi Y (2018) Blood-brain barrier dysfunction and recovery after ischemic stroke, Progress in Neurobiology. 163-164, 144–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Yao Y (2019) Basement membrane and stroke, J Cereb Blood Flow Metab. 39, 3–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Oby E & Janigro D (2006) The Blood–Brain Barrier and Epilepsy, Epilepsia. 47, 1761–1774. [DOI] [PubMed] [Google Scholar]
- 73.Mendes NF, Pansani AP, Carmanhaes ERF, Tange P, Meireles JV, Ochikubo M, Chagas JR, da Silva AV, Monteiro de Castro G & Le Sueur-Maluf L (2019) The Blood-Brain Barrier Breakdown During Acute Phase of the Pilocarpine Model of Epilepsy Is Dynamic and Time-Dependent, Front Neurol. 10, 382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Johnson HL, Jin F, Pirko I & Johnson AJ (2014) Theiler’s murine encephalomyelitis virus as an experimental model system to study the mechanism of blood–brain barrier disruption, Journal of NeuroVirology. 20, 107–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Marchi N, Angelov L, Masaryk T, Fazio V, Granata T, Hernandez N, Hallene K, Diglaw T, Franic L, Najm I & Janigro D (2007) Seizure-promoting effect of blood-brain barrier disruption, Epilepsia. 48, 732–742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.van Vliet EA, da Costa Araújo S, Redeker S, van Schaik R, Aronica E & Gorter JA (2006) Blood–brain barrier leakage may lead to progression of temporal lobe epilepsy, Brain. 130, 521–534. [DOI] [PubMed] [Google Scholar]
- 77.Seiffert E, Dreier JP, Ivens S, Bechmann I, Tomkins O, Heinemann U & Friedman A (2004) Lasting Blood-Brain Barrier Disruption Induces Epileptic Focus in the Rat Somatosensory Cortex, The Journal of Neuroscience. 24, 7829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Frigerio F, Frasca A, Weissberg I, Parrella S, Friedman A, Vezzani A & Noe’ FM (2012) Long-lasting pro-ictogenic effects induced in vivo by rat brain exposure to serum albumin in the absence of concomitant pathology, Epilepsia. 53, 1887–1897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Soares H, Hicks R, Smith D & McIntosh T (1995) Inflammatory leukocytic recruitment and diffuse neuronal degeneration are separate pathological processes resulting from traumatic brain injury, The Journal of Neuroscience. 15, 8223–8233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Floris S, Blezer ELA, Schreibelt G, Döpp E, van der Pol SMA, Schadee‐Eestermans IL, Nicolay K, Dijkstra CD & de Vries HE (2004) Blood–brain barrier permeability and monocyte infiltration in experimental allergic encephalomyelitis: A quantitative MRI study, Brain. 127, 616–627. [DOI] [PubMed] [Google Scholar]
- 81.Obermeier B, Daneman R & Ransohoff RM (2013) Development, maintenance and disruption of the blood-brain barrier, Nat Med. 19, 1584–1596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Losy J & Zaremba J (2001) Monocyte chemoattractant protein-1 is increased in the cerebrospinal fluid of patients with ischemic stroke, Stroke. 32, 2695–6. [DOI] [PubMed] [Google Scholar]
- 83.Westin K, Buchhave P, Nielsen H, Minthon L, Janciauskiene S & Hansson O (2012) CCL2 Is Associated with a Faster Rate of Cognitive Decline during Early Stages of Alzheimer’s Disease, PLOS ONE. 7, e30525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Semple BD, Bye N, Rancan M, Ziebell JM & Morganti-Kossmann MC (2010) Role of CCL2 (MCP-1) in traumatic brain injury (TBI): evidence from severe TBI patients and CCL2−/− mice, Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism. 30, 769–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Gras G & Kaul M (2010) Molecular mechanisms of neuroinvasion by monocytes-macrophages in HIV-1 infection, Retrovirology. 7, 30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Yuan L, Qiao L, Wei F, Yin J, Liu L, Ji Y, Smith D, Li N & Chen D (2013) Cytokines in CSF correlate with HIV-associated neurocognitive disorders in the post-HAART era in China, J Neurovirol. 19, 144–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Cerri C, Genovesi S, Allegra M, Pistillo F, Puntener U, Guglielmotti A, Perry VH, Bozzi Y & Caleo M (2016) The Chemokine CCL2 Mediates the Seizure-enhancing Effects of Systemic Inflammation, J Neurosci. 36, 3777–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Arisi GM, Foresti ML, Katki K & Shapiro LA (2015) Increased CCL2, CCL3, CCL5, and IL-1beta cytokine concentration in piriform cortex, hippocampus, and neocortex after pilocarpine-induced seizures, J Neuroinflammation. 12, 129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Broekaart DWM, Anink JJ, Baayen JC, Idema S, de Vries HE, Aronica E, Gorter JA & van Vliet EA (2018) Activation of the innate immune system is evident throughout epileptogenesis and is associated with blood-brain barrier dysfunction and seizure progression, Epilepsia. 59, 1931–1944. [DOI] [PubMed] [Google Scholar]
- 90.Choi J, Nordli DR Jr., Alden TD, DiPatri A Jr., Laux L, Kelley K, Rosenow J, Schuele SU, Rajaram V & Koh S (2009) Cellular injury and neuroinflammation in children with chronic intractable epilepsy, J Neuroinflammation. 6, 38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wu Y, Wang X, Mo X, Xi Z, Xiao F, Li J, Zhu X, Luan G, Wang Y, Li Y & Zhang J (2008) Expression of monocyte chemoattractant protein-1 in brain tissue of patients with intractable epilepsy, Clin Neuropathol. 27, 55–63. [DOI] [PubMed] [Google Scholar]
- 92.de Vries EE, van den Munckhof B, Braun KP, van Royen-Kerkhof A, de Jager W & Jansen FE (2016) Inflammatory mediators in human epilepsy: A systematic review and meta-analysis, Neurosci Biobehav Rev. 63, 177–90. [DOI] [PubMed] [Google Scholar]
- 93.Deshmane SL, Kremlev S, Amini S & Sawaya BE (2009) Monocyte chemoattractant protein-1 (MCP-1): an overview, J Interferon Cytokine Res. 29, 313–326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Jiang W, St-Pierre S, Roy P, Morley BJ, Hao J & Simard AR (2016) Infiltration of CCR2+Ly6Chigh Proinflammatory Monocytes and Neutrophils into the Central Nervous System Is Modulated by Nicotinic Acetylcholine Receptors in a Model of Multiple Sclerosis, Journal of immunology (Baltimore, Md : 1950). 196, 2095–2108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Dimitrijevic OB, Stamatovic SM, Keep RF & Andjelkovic AV (2007) Absence of the chemokine receptor CCR2 protects against cerebral ischemia/reperfusion injury in mice, Stroke. 38, 1345–53. [DOI] [PubMed] [Google Scholar]
- 96.Hsieh CL, Niemi EC, Wang SH, Lee CC, Bingham D, Zhang J, Cozen ML, Charo I, Huang EJ, Liu J & Nakamura MC (2014) CCR2 deficiency impairs macrophage infiltration and improves cognitive function after traumatic brain injury, J Neurotrauma. 31, 1677–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Izikson L, Klein RS, Charo IF, Weiner HL & Luster AD (2000) Resistance to experimental autoimmune encephalomyelitis in mice lacking the CC chemokine receptor (CCR)2, J Exp Med. 192, 1075–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Howe CL, LaFrance-Corey RG, Goddery EN, Johnson RK & Mirchia K (2017) Neuronal CCL2 expression drives inflammatory monocyte infiltration into the brain during acute virus infection, J Neuroinflammation. 14, 238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Winkler CW, Woods TA, Robertson SJ, McNally KL, Carmody AB, Best SM & Peterson KE (2018) Cutting Edge: CCR2 Is Not Required for Ly6C(hi) Monocyte Egress from the Bone Marrow but Is Necessary for Migration within the Brain in La Crosse Virus Encephalitis, J Immunol. 200, 471–476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Dimitrijevic OB, Stamatovic SM, Keep RF & Andjelkovic AV (2006) Effects of the chemokine CCL2 on blood-brain barrier permeability during ischemia-reperfusion injury, J Cereb Blood Flow Metab. 26, 797–810. [DOI] [PubMed] [Google Scholar]
- 101.Stamatovic SM, Dimitrijevic OB, Keep RF & Andjelkovic AV (2006) Protein kinase Calpha-RhoA cross-talk in CCL2-induced alterations in brain endothelial permeability, J Biol Chem. 281, 8379–88. [DOI] [PubMed] [Google Scholar]
- 102.Stamatovic SM, Keep RF, Kunkel SL & Andjelkovic AV (2003) Potential role of MCP-1 in endothelial cell tight junction ‘opening’: signaling via Rho and Rho kinase, J Cell Sci. 116, 4615–28. [DOI] [PubMed] [Google Scholar]
- 103.Stamatovic SM, Shakui P, Keep RF, Moore BB, Kunkel SL, Van Rooijen N & Andjelkovic AV (2005) Monocyte chemoattractant protein-1 regulation of blood-brain barrier permeability, J Cereb Blood Flow Metab. 25, 593–606. [DOI] [PubMed] [Google Scholar]
- 104.Yao Y & Tsirka SE (2014) Monocyte chemoattractant protein-1 and the blood-brain barrier, Cell Mol Life Sci. 71, 683–697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Reijerkerk A, Kooij G, van der Pol SM, Leyen T, van Het Hof B, Couraud PO, Vivien D, Dijkstra CD & de Vries HE (2008) Tissue-type plasminogen activator is a regulator of monocyte diapedesis through the brain endothelial barrier, J Immunol. 181, 3567–74. [DOI] [PubMed] [Google Scholar]
- 106.Kataoka K, Asai T, Taneda M, Ueshima S, Matsuo O, Kuroda R, Kawabata A & Carmeliet P (2000) Roles of urokinase type plasminogen activator in a brain stab wound, Brain Res. 887, 187–90. [DOI] [PubMed] [Google Scholar]
- 107.Yao Y & Tsirka SE (2011) Truncation of monocyte chemoattractant protein 1 by plasmin promotes blood-brain barrier disruption, J Cell Sci. 124, 1486–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Strecker JK, Minnerup J, Schutte-Nutgen K, Gess B, Schabitz WR & Schilling M (2013) Monocyte chemoattractant protein-1-deficiency results in altered blood-brain barrier breakdown after experimental stroke, Stroke. 44, 2536–44. [DOI] [PubMed] [Google Scholar]
- 109.Franca CN, Izar MCO, Hortencio MNS, do Amaral JB, Ferreira CES, Tuleta ID & Fonseca FAH (2017) Monocyte subtypes and the CCR2 chemokine receptor in cardiovascular disease, Clin Sci (Lond). 131, 1215–1224. [DOI] [PubMed] [Google Scholar]
- 110.Strauss-Ayali D, Conrad SM & Mosser DM (2007) Monocyte subpopulations and their differentiation patterns during infection, Journal of Leukocyte Biology. 82, 244–252. [DOI] [PubMed] [Google Scholar]
- 111.Geissmann F, Jung S & Littman DR (2003) Blood monocytes consist of two principal subsets with distinct migratory properties, Immunity. 19, 71–82. [DOI] [PubMed] [Google Scholar]
- 112.Gliem M, Mausberg AK, Lee JI, Simiantonakis I, van Rooijen N, Hartung HP & Jander S (2012) Macrophages prevent hemorrhagic infarct transformation in murine stroke models, Ann Neurol. 71, 743–52. [DOI] [PubMed] [Google Scholar]
- 113.Vezzani A (2014) Epilepsy and inflammation in the brain: overview and pathophysiology, Epilepsy Curr. 14, 3–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Vezzani A, French J, Bartfai T & Baram TZ (2011) The role of inflammation in epilepsy, Nat Rev Neurol. 7, 31–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Vezzani A (2008) Innate immunity and inflammation in temporal lobe epilepsy: new emphasis on the role of complement activation, Epilepsy Curr. 8, 75–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Vezzani A, Balosso S & Ravizza T (2019) Neuroinflammatory pathways as treatment targets and biomarkers in epilepsy, Nature Reviews Neurology. 15, 459–472. [DOI] [PubMed] [Google Scholar]
- 117.Terrone G, Balosso S, Pauletti A, Ravizza T & Vezzani A (2019) Inflammation and reactive oxygen species as disease modifiers in epilepsy, Neuropharmacology, 107742. [DOI] [PubMed] [Google Scholar]
- 118.Pauletti A, Terrone G, Shekh-Ahmad T, Salamone A, Ravizza T, Rizzi M, Pastore A, Pascente R, Liang L-P, Villa BR, Balosso S, Abramov AY, van Vliet EA, Del Giudice E, Aronica E, Patel M, Walker MC & Vezzani A (2019) Targeting oxidative stress improves disease outcomes in a rat model of acquired epilepsy, Brain. 142, e39–e39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Chen YH, Kuo TT, Chu MT, Ma HI, Chiang YH & Huang EY (2013) Postnatal systemic inflammation exacerbates impairment of hippocampal synaptic plasticity in an animal seizure model, Neuroimmunomodulation. 20, 223–32. [DOI] [PubMed] [Google Scholar]
- 120.Auvin S, Shin D, Mazarati A, Nakagawa J, Miyamoto J & Sankar R (2007) Inflammation exacerbates seizure-induced injury in the immature brain, Epilepsia. 48 Suppl 5, 27–34. [DOI] [PubMed] [Google Scholar]
- 121.Kang DH, Heo RW, Yi C. o., Kim H, Choi CH & Roh GS (2015) High-fat diet-induced obesity exacerbates kainic acid-induced hippocampal cell death, BMC Neuroscience. 16, 72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Güneş M & Büyükgöl H (2020) Relationship between generalized epileptic seizure and neutrophil/lymphocyte ratio, platelet/lymphocyte ratio, and neutrophil mediated inflammation, International Journal of Neuroscience, 1–6. [DOI] [PubMed] [Google Scholar]
- 123.Özdemir HH, Akil E, Acar A, Tamam Y, Varol S, Cevik MU & Arikanoglu A (2017) Changes in serum albumin levels and neutrophil–lymphocyte ratio in patients with convulsive status epilepticus, International Journal of Neuroscience. 127, 417–420. [DOI] [PubMed] [Google Scholar]
- 124.Silverberg J, Ginsburg D, Orman R, Amassian V, Durkin HG & Stewart M (2010) Lymphocyte infiltration of neocortex and hippocampus after a single brief seizure in mice, Brain, Behavior, and Immunity. 24, 263–272. [DOI] [PubMed] [Google Scholar]
- 125.Nakahara H, Konishi Y,G Beach T, Yamada N, Makino S & Tooyama I (2010) Infiltration of T Lymphocytes and Expression of ICAM-1 in the Hippocampus of Patients with Hippocampal Sclerosis, ACTA HISTOCHEMICA ET CYTOCHEMICA. 43, 157–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Kumar P, Shih DCW, Lim A, Paleja B, Ling S, Li Yun L, Li Poh S, Ngoh A, Arkachaisri T, Yeo JG & Albani S (2019) Pro-inflammatory, IL-17 pathways dominate the architecture of the immunome in pediatric refractory epilepsy, JCI Insight. 5, e126337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Zattoni M, Mura ML, Deprez F, Schwendener RA, Engelhardt B, Frei K & Fritschy J-M (2011) Brain Infiltration of Leukocytes Contributes to the Pathophysiology of Temporal Lobe Epilepsy, The Journal of Neuroscience. 31, 4037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Patel DC, Wallis G, Dahle EJ, McElroy PB, Thomson KE, Tesi RJ, Szymkowski DE, West PJ, Smeal RM, Patel M, Fujinami RS, White HS & Wilcox KS (2017) Hippocampal TNFα Signaling Contributes to Seizure Generation in an Infection-Induced Mouse Model of Limbic Epilepsy, eNeuro. 4, ENEURO.0105–17.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Li G, Bauer S, Nowak M, Norwood B, Tackenberg B, Rosenow F, Knake S, Oertel WH & Hamer HM (2011) Cytokines and epilepsy, Seizure. 20, 249–256. [DOI] [PubMed] [Google Scholar]
- 130.Hoda U, Agarwal NB, Vohora D, Parvez S & Raisuddin S (2017) Resveratrol suppressed seizures by attenuating IL-1β, IL1-Ra, IL-6, and TNF-α in the hippocampus and cortex of kindled mice, Nutritional Neuroscience. 20, 497–504. [DOI] [PubMed] [Google Scholar]
- 131.D’Mello C, Le T & Swain MG (2009) Cerebral microglia recruit monocytes into the brain in response to tumor necrosis factoralpha signaling during peripheral organ inflammation, J Neurosci. 29, 2089–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Seleme MC, Kosmac K, Jonjic S & Britt WJ (2017) Tumor Necrosis Factor Alpha-Induced Recruitment of Inflammatory Mononuclear Cells Leads to Inflammation and Altered Brain Development in Murine Cytomegalovirus-Infected Newborn Mice, J Virol. 91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Pare A, Mailhot B, Levesque SA, Juzwik C, Ignatius Arokia Doss PM, Lecuyer MA, Prat A, Rangachari M, Fournier A & Lacroix S (2018) IL-1beta enables CNS access to CCR2(hi) monocytes and the generation of pathogenic cells through GM-CSF released by CNS endothelial cells, Proc Natl Acad Sci U S A. 115, E1194–e1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Howe CL, Lafrance-Corey RG, Sundsbak RS & Lafrance SJ (2012) Inflammatory monocytes damage the hippocampus during acute picornavirus infection of the brain, J Neuroinflammation. 9, 50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Howe CL, Lafrance-Corey RG, Sundsbak RS, Sauer BM, Lafrance SJ, Buenz EJ & Schmalstieg WF (2012) Hippocampal protection in mice with an attenuated inflammatory monocyte response to acute CNS picornavirus infection, Sci Rep. 2, 545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Cusick MF, Libbey JE, Patel DC, Doty DJ & Fujinami RS (2013) Infiltrating macrophages are key to the development of seizures following virus infection, J Virol. 87, 1849–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Waltl I, Kaufer C, Broer S, Chhatbar C, Ghita L, Gerhauser I, Anjum M, Kalinke U & Loscher W (2018) Macrophage depletion by liposome-encapsulated clodronate suppresses seizures but not hippocampal damage after acute viral encephalitis, Neurobiol Dis. 110, 192–205. [DOI] [PubMed] [Google Scholar]
- 138.DePaula-Silva AB, Sonderegger FL, Libbey JE, Doty DJ & Fujinami RS (2018) The immune response to picornavirus infection and the effect of immune manipulation on acute seizures, J Neurovirol. 24, 464–477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Libbey JE, Kennett NJ, Wilcox KS, White HS & Fujinami RS (2011) Interleukin-6, produced by resident cells of the central nervous system and infiltrating cells, contributes to the development of seizures following viral infection, J Virol. 85, 6913–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.DePaula-Silva AB, Hanak TJ, Libbey JE & Fujinami RS (2017) Theiler’s murine encephalomyelitis virus infection of SJL/J and C57BL/6J mice: Models for multiple sclerosis and epilepsy, Journal of neuroimmunology. 308, 30–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Bertoglio D, Amhaoul H, Van Eetveldt A, Houbrechts R, Van De Vijver S, Ali I & Dedeurwaerdere S (2017) Kainic Acid-Induced Post-Status Epilepticus Models of Temporal Lobe Epilepsy with Diverging Seizure Phenotype and Neuropathology, Frontiers in neurology. 8, 588–588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Levesque M & Avoli M (2013) The kainic acid model of temporal lobe epilepsy, Neurosci Biobehav Rev. 37, 2887–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Curia G, Longo D, Biagini G, Jones RSG & Avoli M (2008) The pilocarpine model of temporal lobe epilepsy, J Neurosci Methods. 172, 143–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Venturin GT, Greggio S, Marinowic DR, Zanirati G, Cammarota M, Machado DC & DaCosta JC (2011) Bone marrow mononuclear cells reduce seizure frequency and improve cognitive outcome in chronic epileptic rats, Life Sci. 89, 229–34. [DOI] [PubMed] [Google Scholar]
- 145.Prabhakaran V, Drevets DA, Ramajayam G, Manoj JJ, Anderson MP, Hanas JS, Rajshekhar V, Oommen A & Carabin H (2017) Comparison of monocyte gene expression among patients with neurocysticercosis-associated epilepsy, Idiopathic Epilepsy and idiopathic headaches in India, PLoS Negl Trop Dis. 11, e0005664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Uddin J, Garcia HH, Gilman RH, Gonzalez AE & Friedland JS (2005) Monocyte-astrocyte networks and the regulation of chemokine secretion in neurocysticercosis, J Immunol. 175, 3273–81. [DOI] [PubMed] [Google Scholar]
- 147.Seidenberg M, Pulsipher DT & Hermann B (2009) Association of epilepsy and comorbid conditions, Future Neurol. 4, 663–668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Butler CR & Zeman AZ (2008) Recent insights into the impairment of memory in epilepsy: transient epileptic amnesia, accelerated long-term forgetting and remote memory impairment, Brain. 131, 2243–2263. [DOI] [PubMed] [Google Scholar]
- 149.Wohleb ES, McKim DB, Sheridan JF & Godbout JP (2015) Monocyte trafficking to the brain with stress and inflammation: a novel axis of immune-to-brain communication that influences mood and behavior, Front Neurosci. 8, 447–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Minogue AM (2017) Role of infiltrating monocytes/macrophages in acute and chronic neuroinflammation: Effects on cognition, learning and affective behaviour, Progress in Neuro-Psychopharmacology and Biological Psychiatry. 79, 15–18. [DOI] [PubMed] [Google Scholar]
- 151.Guedes JR, Lao T, Cardoso AL & El Khoury J (2018) Roles of Microglial and Monocyte Chemokines and Their Receptors in Regulating Alzheimer’s Disease-Associated Amyloid-β and Tau Pathologies, Frontiers in Neurology. 9, 549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.McKim DB, Weber MD, Niraula A, Sawicki CM, Liu X, Jarrett BL, Ramirez-Chan K, Wang Y, Roeth RM, Sucaldito AD, Sobol CG, Quan N, Sheridan JF & Godbout JP (2018) Microglial recruitment of IL-1β-producing monocytes to brain endothelium causes stress-induced anxiety, Molecular Psychiatry. 23, 1421–1431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Mélik-Parsadaniantz S & Rostène W (2008) Chemokines and neuromodulation, Journal of Neuroimmunology. 198, 62–68. [DOI] [PubMed] [Google Scholar]
- 154.Kim JE, Park H, Choi SH, Kong MJ & Kang TC (2019) Roscovitine Attenuates Microglia Activation and Monocyte Infiltration via p38 MAPK Inhibition in the Rat Frontoparietal Cortex Following Status Epilepticus, Cells. 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Nikbakht F, Belali R, Rasoolijazi H & Mohammad Khanizadeh A (2019) 2-Deoxyglucose protects hippocampal neurons against kainate-induced temporal lobe epilepsy by modulating monocyte-derived macrophages (mo-MPhi) and progranulin production in the hippocampus, Neuropeptides. 76, 101932. [DOI] [PubMed] [Google Scholar]