Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2015

Open Access 01.12.2015 | Short report

Increased CCL2, CCL3, CCL5, and IL-1β cytokine concentration in piriform cortex, hippocampus, and neocortex after pilocarpine-induced seizures

verfasst von: Gabriel M. Arisi, Maira L. Foresti, Khurshed Katki, Lee A. Shapiro

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2015

Abstract

Background

Cytokines and chemokines play an important role in the neuroinflammatory response to an initial precipitating injury such as status epilepticus (SE). These signaling molecules participate in recruitment of immune cells, including brain macrophages (microglia), as well as neuroplastic changes, deterioration of damaged tissue, and epileptogenesis. This study describes the temporal and brain region pattern expression of numerous cytokines, including chemokines, after pilocarpine-induced seizures and discusses them in the larger context of their potential involvement in the changes that precede the development of epilepsy.

Findings

Adult rats received pilocarpine to induce SE and 90 min after seizure onset were treated with diazepam to mitigate seizures. Rats were subsequently deeply anesthetized and brain regions (hippocampus, piriform cortex, neocortex, and cerebellum) were freshly dissected at 2, 6, and 24 h or 5 days after seizures. Using methodology identical to our previous studies, simultaneous assay of multiple cytokines (CCL2, CCL3, CCL5, interleukin IL-1β, tumor necrosis factor (TNF-α)), and vascular endothelial growth factor (VEGF) was performed and compared to control rats. These proteins were selected based on existing evidence implicating them in the epileptogenic progression. A robust increase in CCL2 and CCL3 concentrations in the hippocampus, piriform cortex, and neocortex was observed at all time-points. The concentrations peaked with a ~200-fold increase 24 h after seizures and were two orders of magnitude greater than the significant increases observed for CCL5 and IL-1β in the same brain structures. TNF-α levels were altered in the piriform cortex and neocortex (24 h) and in the hippocampus (5 days) after SE.

Conclusions

Pilocarpine-induced status epilepticus causes a rapid increase of multiple cytokines in limbic and neocortical regions. Understanding the precise spatial and temporal pattern of cytokines and chemokine changes could provide more viable therapeutic targets to reduce, reverse, or prevent the development of epilepsy following a precipitating injury.
Hinweise

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

GMA carried out the experimental animal studies and statistical analyses and drafted the manuscript. MLF helped with the animal studies and drafting of the manuscript. KK carried out the cytokine immunoassays and analyses. LAS participated in the design and coordination of the study and drafted the manuscript with GMA. All authors read and approved the final manuscript.
Abkürzungen
CCL
C-C motif chemokines
CCL2/MCP-1
monocyte chemoattractant protein-1
CCL3/MIP-1α
macrophage inflammatory protein 1 alpha
CCL5/RANTES
regulated upon activation normal T cell expressed
CCR
CCL receptor
IL
interleukin
TNF
tumor necrosis factor
VEGF
vascular endothelial growth factor

Findings

Introduction

Secretion of cytokines is a fundamental component of the inflammatory response in the nervous system [14]. These proteins are important immunomodulators, and chemokines, a special class of cytokines, also possess chemoattractant properties. Following an initial precipitating injury such as status epilepticus (SE), alterations occur to the milieu of these neuroinflammatory proteins. Of the numerous functional outcomes attributed to these alterations, neuroplasticity has been implicated to play a prominent role in the epileptogenic progression. Other changes that have also been implicated in pathogenesis include vascular permeability, angiogenesis, and immune responses that contribute to tissue damage [2]. Considering that SE increases subsequent seizure susceptibility and the likelihood of developing epilepsy, immunomodulatory signaling proteins are garnering great interest as therapeutic targets to treat neuropathology and the epileptogenic progression [1, 3]. Further evidence in support of the potential for these proteins as therapeutic targets is found in patients with epilepsy. In these patients, studies have observed increased levels of chemokines CCL2, CCL3, and CCL5 (among others) in the hippocampus, other temporal lobe structures, and neocortex [4]. Moreover, elevated cytokines such as interleukin 6 (IL-6) and tumor necrosis factor (TNF) can be detected in serum [1], and previous experimental studies in rodents have yielded promising results regarding the potential therapeutic efficacy of targeting these particular inflammatory molecules. Considering these data, a greater understanding of the temporal and spatial changes of these proteins can yield more efficacious treatment options, as well as more precise targeting.
After the acute and early phases of SE, the brain typically enters into a latent period that precedes the development of spontaneous seizures. During this period, it has been suggested that neuroplastic and neuroinflammatory changes provide a substrate for hyperexcitable neural networks [5, 6]. Indeed, our previous studies have implicated the neuroinflammatory response in the hippocampus to provide a substrate for the aberrant sprouting of basal dendrites from immature and mature granule cells [7]. Considering the complex interactions that occur among the cytokines both in immune and nervous systems, the most efficacious method for examining their presence in epileptogenesis is through simultaneous multiplex immunoassay [8]. We previously demonstrated elevated hippocampal concentrations of chemokine C-C motif ligand 2 (CCL2) during the acute phase after SE [7]. This report extends these previous findings to further identify acute and early alterations to cytokine concentrations in the hippocampus, cerebellum, and piriform cortex, brain structures that are known to be involved in seizures and epilepsy.

Methods

Animals and seizure induction

Male Sprague-Dawley rats, weighing 230–235 g at the beginning of experiments were used in this study. The experimental procedure was approved by the IACUC of the Texas A&M University Health Science Center (protocol #2008-002-R).
SE was induced as previously described [7]. Animals were treated with pilocarpine hydrochloride in a single dose of 320 mg/kg i.p. or saline. The onset of SE was considered when animals exhibited a class 3 motor seizure in Racine’s scale of limbic seizures [9]. Only animals that experienced stage 5 seizures were used for analysis (n = 48). Ninety minutes after SE onset, rats were treated with diazepam (10 mg/kg) to mitigate seizures; control animals also received diazepam.

Tissue harvest

Animals were deeply anesthetized using Euthasol (pentobarbital sodium and phenytoin, 1 ml/kg) and decapitated 2, 6, and 24 h or 5 days (n = 12 per time-point) after diazepam treatment. The brain was rapidly removed from the skull, and the hippocampus, cerebellum, piriform cortex, and a large slice of neocortex close to the midline containing the retrosplenial, cingulate, and primary motor cortex were dissected and immediately frozen.

Cytokine analyses

Simultaneous measuring of different cytokines in brain regions was performed as previously described [8]. The following cytokines were assayed: CCL2, CCL3, CCL5, IL-1β, TNF-α, and vascular endothelial growth factor (VEGF) (Table 1) in tissue homogenates using a MILLIPLEX MAP kit (Millipore, Billerica, MA, USA) on a BIOPLEX 200 analyzer (BioRad, Hercules, CA, USA). The tissue concentration of each analyte was normalized to the total protein concentration measured with a Bradford assay and presented as a proportion of cytokines in picograms (pg) per microgram (μg) of total protein ± SEM. Student’s t test was employed to detect statistical differences in values of control and experimental groups.
Table 1
Average cytokine concentration in pg/μg of total protein
Analytes
Brain region
2 h
6 h
24 h
5 days
Cytokines
Chemokines
Control
SE
Control
SE
Control
SE
Control
SE
 
CCL2
Piriform
3.6
10.7
3.7
34.8
3.7
694.8
3.6
22.8
Hippocampus
3.7
16.3
4.4
81.6
2.2
243.8
4.4
26.8
Neocortex
2.9
21.7
3.5
97.0
3.7
933.3
3.8
15.5
Cerebellum
1.4
1.8
1.4
1.9
2.8
4.0
3.8
5.7
CCL3
Piriform
0.4
24.3
1.7
29.8
0.2
50.3
0.3
15.6
Hippocampus
0.5
31.4
1.2
30.6
0.2
42.2
0.1
29.3
Neocortex
0.9
32.3
0.9
23.3
0.1
37.7
0.3
22.1
Cerebellum
0.2
0.2
0.0
0.0
0.0
0.9
0.0
0.0
CCL5
Piriform
6.7
8.2
8.1
10.3
6.0
13.8
6.6
10.1
Hippocampus
4.9
5.8
5.8
5.9
6.6
10.6
4.5
6.7
Neocortex
8.6
8.9
7.9
10.5
8.2
14.2
8.4
8.2
Cerebellum
7.1
5.9
7.1
6.2
7.2
6.0
6.8
5.8
IL-1β
 
Piriform
5.0
7.7
5.2
7.8
6.2
12.2
6.1
6.7
 
Hippocampus
5.5
8.5
6.0
8.5
5.9
7.7
5.8
6.4
 
Neocortex
6.4
10.0
5.6
9.1
5.7
13.4
6.4
8.0
 
Cerebellum
6.0
6.4
5.0
5.7
5.3
6.5
5.7
6.7
TNF
 
Piriform
1.3
1.0
0.7
0.8
1.2
0.6
1.1
1.6
 
Hippocampus
1.6
1.6
1.3
1.9
2.5
1.6
1.5
2.8
 
Neocortex
1.2
1.5
1.0
1.5
0.7
1.5
2.0
1.5
 
Cerebellum
1.0
1.0
0.5
0.4
0.6
0.5
0.6
0.9
(Signal protein)
          
VEGF
 
Piriform
0.9
0.7
0.9
0.5
0.9
1.1
0.9
0.6
 
Hippocampus
0.8
0.7
0.6
0.5
0.9
0.9
0.7
0.7
 
Neocortex
0.7
0.5
0.7
0.6
0.6
0.7
0.9
0.9
 
Cerebellum
0.3
0.7
0.5
0.7
0.5
0.5
0.6
0.7

Results

The results revealed significant increases in the concentration of numerous cytokines at different time-points in several of the brain regions examined (Tables 1 and 2).
Table 2
Cytokine concentration ratio of SE/control animals
Analytes
Brain region
2 h
6 h
24 h
5 days
Cytokines
Chemokines
 
CCL2
Piriform
3**
9**
186**
6**
Hippocampus
4*
18**
108**
6**
Neocortex
7**
27**
252**
4
Cerebellum
1
1
1
1
CCL3
Piriform
60**
18**
228**
54**
Hippocampus
69**
25**
211**
238**
Neocortex
35**
25**
272**
86**
Cerebellum
1
1
1
1
CCL5
Piriform
1
1
2**
2*
Hippocampus
1
1
2*
1
Neocortex
1
1
2*
1
Cerebellum
1
1
1
1
IL-1β
 
Piriform
2*
1
2*
1
 
Hippocampus
2*
2*
1
1
 
Neocortex
2*
2*
2**
1
 
Cerebellum
1
1
1
1
TNF
 
Piriform
1
1
1/2*
1
 
Hippocampus
1
1
1/2*
2*
 
Neocortex
1
2
2
1
 
Cerebellum
1
1
1
1
Signal protein
      
VEGF
 
Piriform
1
1
1
1
 
Hippocampus
1
1
1
1
 
Neocortex
1
1
1
1
 
Cerebellum
1
1
1
1
*p < 0.05; **p < 0.001
Tissue concentration of CCL2 was significantly increased in seizure animals at all time-points, and in all brain regions, except in the cerebellum. Concentrations peaked at 24 h after SE in the piriform cortex hippocampus, and midline neocortex. Five days after seizures, CCL2 levels were still significantly elevated in the piriform cortex, hippocampus, and neocortex but had declined from the 24-h peak levels (Fig. 1).
The chemokine CCL3 presented a larger increase at earlier time-points relative to CCL2 protein observed 2 h after SE in the piriform cortex, hippocampus, and midline neocortex (Fig. 2). At 6 h after SE, increase in the same areas was followed by a peak concentration 24 h after SE in all regions, except the cerebellum. Five days after SE, concentrations of CCL3 remained significantly elevated in the hippocampus, piriform cortex, and midline neocortex, but to a lesser extent than at the 24-h peak (Fig. 2).
Concentration of CCL5 was significantly elevated at 24 h after SE in the piriform cortex and neocortex. No significant alterations were observed in either the hippocampus or the cerebellum. Five days after SE, the CCL5 concentration was still significantly elevated in the piriform cortex but returned close to control levels in other regions (Fig. 3).
The concentration of IL-1β was significantly elevated 2 h after SE in the neocortex, piriform cortex, and hippocampus, but not in the cerebellum (Fig. 4). Concentrations of IL-1β were significantly elevated 6 h after SE in the neocortex and hippocampus, but not in the piriform cortex or cerebellum (Fig. 4). The peak concentration was observed 24 h after SE in the piriform cortex and neocortex. Five days after seizures, IL-1β levels returned to levels similar to control rats in all brain regions (Fig. 4).
Analysis of TNF-α levels revealed significant concentration decrease in the piriform cortex and hippocampus 24 h after SE and a significant concentration increase in the hippocampus 5 days after SE (Table 1). No significant alterations in VEGF levels were observed at any of the time-points, nor in any of the regions analyzed (Tables 1 and 2).

Discussion

Simultaneous analysis of multiple cytokines in different brain regions, at different time-points after SE, revealed significant alterations in the concentration of CCL2, CCL3, CCL5, IL-1β, and TNF-α. CCL2 and CCL3 showed the largest concentration increases in the piriform cortex, hippocampus, and neocortex that peaked at 24 h after SE. CCL5 and IL-1β were also significantly elevated in these brain regions, although the increases were orders of magnitude lower relative to CCL2 and CCL3. These novel data in the rat pilocarpine SE model are consistent with findings in human epileptic patients, where microarray analyses showed upregulation of genes ccl3 and ccl4 in surgically removed hippocampus [10]. Similarly, animal studies showed increased CCL2 and CCL3 mRNA that peak 1 day after electrically induced SE [11], and following kainic acid-induced seizures in rats, the gene for ccl2 was upregulated in entorhinal, perirhinal, posterior piriform cortices and hippocampus, where its expression level was the highest relative to the other genes examined [12]. Therefore, future translational studies can selectively target one or more of these inflammatory proteins in the pilocarpine SE model.
Further highlighting the clinical relevance of these chemokines, elevated CCL2 protein was observed in the temporal lobe and hippocampus from patients with intractable epilepsy [13], and both CCL2 and CCL3 proteins were elevated in temporal lobe epilepsy patients measured with multiplex assay [4]. We have also previously shown that CCL2 and its receptor CCR2 were elevated in the rat hippocampus 5 days after seizures in rats [8], whereas other studies demonstrated increased CCL3 protein within the first 2 h after pilocarpine injection in mice [14]. Results consistent with these findings were also found in other experimental models of temporal lobe epilepsy, such as tetanic stimulation of the hippocampus [11] and following kainic acid-induced seizures [5]. Thus, there is substantial accumulating evidence that changes in these cytokines may play an important role in epileptogenesis and thus may represent viable therapeutic targets.
From a functional perspective, it is possible that CCL2 is involved in direct impairment of neuronal survival in epileptogenic structures. For example, CCL2 activates caspase signaling pathways [15], which can contribute directly to cell death. CCL2 can also influence glia activation and the ensuing inflammation [16], which results in uncoupling of gap junctions between astrocytes and thus a reduced calcium buffering potential by the astrocytic network observed in temporal lobe epilepsy patients [17]. Further evidence in support for direct functional impairment attributed to CCL2 elevation was reported in a study in which expression by astrocytes in transgenic mice impairs synaptic plasticity in the hippocampus [18]. Indeed, a study that inhibited CCL2 secretion with pyrrolidine dithiocarbamate prior to pilocarpine-induced SE demonstrated beneficial outcomes with less microglial activation and decreased neuronal damage [19]. While such experimentation may not be clinically viable, it does provide evidence for the therapeutic potential of targeting CCL2 to improve epileptogenic outcomes.
Targeting the seizure-induced increase in CCR5 may be another putative therapeutic target to ameliorate the epileptogenic progression. The chemokine CCL5 is chemotactic to T cells, eosinophils, and basophils. Consistent with the findings in the present study, there is an upregulation of CCR5 expression after kainic acid-induced seizures especially in hippocampal formation neurons and glial cells [20] and after pilocarpine-induced seizures in hippocampal astrocytes [21]. Interestingly, the use of RNA interference to lower CCR5 expression in rats protected against kainic acid-induced seizures by decreasing hippocampal neuronal loss and macrophage/microglial infiltration [22]. The fact that CCR5-knockout mice did not show the same protection against inflammatory hippocampal injury suggests that compensatory mechanisms may render it necessary to target multiple inflammatory molecules to achieve optimal benefit [23].
The result of elevated IL-1β following SE is consistent with previous studies showing that increased concentration of IL-1β after seizures exerts a pro-epileptogenic action, and blocking of IL-1β presented anticonvulsive effects while exogenous application of the cytokine worsened electrographic seizure development [2, 4]. In contrast, the results from the current study were inconsistent with previous reports of increased VEGF after pilocarpine-induced seizures [24]. One possible explanation for this discrepancy is that the current study examined total VEGF protein in brain homogenates, whereas the latter study observed elevated cellular VEGF in neurons and astrocytes [24]. Thus, it is possible that the altered VEGF expression observed is a qualitative rather than a quantitative one.

Conclusions

The data from this short report shows, for the first time, elevated CCL2 and CCL3, as well as increased CCL5, IL-1β, and differentially altered TNF-α. It is becoming increasingly clear that the neuroinflammatory response after an initial precipitating injury such as SE plays an important role in nervous tissue pathology. Understanding the temporal and spatial expression patterns of the neuroinflammatory milieu is essential to develop improved therapeutic interventions following these types of injury. Therefore, simultaneous measurement of multiple cytokines from different brain regions is especially important to elucidate the extensive, intertwined, and complex interactions among the inflammatory proteins. Such studies will enable subsequent studies that target multiple cytokine/chemokines at different time-points and possibly different brain regions to achieve optimal therapeutic efficacy.

Acknowledgements

This material is the result of the work supported by the use of facilities at the Central Texas Veterans Health Care System, Temple, TX, USA. The work in the authors’ laboratory is/was supported by grants from The American Heart Association (09SDG2370076) and Scott and White Hospital. We thank Amanda Ruch for the technical expertise.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
The Creative Commons Public Domain Dedication waiver (https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

GMA carried out the experimental animal studies and statistical analyses and drafted the manuscript. MLF helped with the animal studies and drafting of the manuscript. KK carried out the cytokine immunoassays and analyses. LAS participated in the design and coordination of the study and drafted the manuscript with GMA. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Sinha S, Patil SA, Jayalekshmy V, Satishchandra P. Do cytokines have any role in epilepsy? Epilepsy Res. 2008;82:171–6.PubMedCrossRef Sinha S, Patil SA, Jayalekshmy V, Satishchandra P. Do cytokines have any role in epilepsy? Epilepsy Res. 2008;82:171–6.PubMedCrossRef
2.
Zurück zum Zitat Minami M, Kuraishi Y, Satoh M. Effects of kainic acid on messenger RNA levels of IL-1 beta, IL-6, TNF alpha and LIF in the rat brain. Biochem Biophys Res Commun. 1991;176:593–8.PubMedCrossRef Minami M, Kuraishi Y, Satoh M. Effects of kainic acid on messenger RNA levels of IL-1 beta, IL-6, TNF alpha and LIF in the rat brain. Biochem Biophys Res Commun. 1991;176:593–8.PubMedCrossRef
3.
Zurück zum Zitat Fabene PF, Bramanti P, Constantin G. The emerging role for chemokines in epilepsy. J Neuroimmunol. 2010;224:22–7.PubMedCrossRef Fabene PF, Bramanti P, Constantin G. The emerging role for chemokines in epilepsy. J Neuroimmunol. 2010;224:22–7.PubMedCrossRef
4.
Zurück zum Zitat Kan AA, de Jager W, de Wit M, Heijnen C, van Zuiden M, Ferrier C, et al. Protein expression profiling of inflammatory mediators in human temporal lobe epilepsy reveals co-activation of multiple chemokines and cytokines. J Neuroinflammation. 2012;9:207.PubMedCentralPubMedCrossRef Kan AA, de Jager W, de Wit M, Heijnen C, van Zuiden M, Ferrier C, et al. Protein expression profiling of inflammatory mediators in human temporal lobe epilepsy reveals co-activation of multiple chemokines and cytokines. J Neuroinflammation. 2012;9:207.PubMedCentralPubMedCrossRef
5.
Zurück zum Zitat Somera-Molina KC, Nair S, Van Eldik LJ, Watterson DM, Wainwright MS. Enhanced microglial activation and proinflammatory cytokine upregulation are linked to increased susceptibility to seizures and neurologic injury in a ‘two-hit’ seizure model. Brain Res. 2009;1282:162–72.PubMedCentralPubMedCrossRef Somera-Molina KC, Nair S, Van Eldik LJ, Watterson DM, Wainwright MS. Enhanced microglial activation and proinflammatory cytokine upregulation are linked to increased susceptibility to seizures and neurologic injury in a ‘two-hit’ seizure model. Brain Res. 2009;1282:162–72.PubMedCentralPubMedCrossRef
6.
Zurück zum Zitat Foresti ML, Arisi GM, Shapiro LA. Role of glia in epilepsy-associated neuropathology, neuroinflammation and neurogenesis. Brain Res Rev. 2010;66:115–22.PubMedCrossRef Foresti ML, Arisi GM, Shapiro LA. Role of glia in epilepsy-associated neuropathology, neuroinflammation and neurogenesis. Brain Res Rev. 2010;66:115–22.PubMedCrossRef
7.
Zurück zum Zitat Foresti ML, Arisi GM, Katki K, Montañez A, Sanchez RM, Shapiro LA. Chemokine CCL2 and its receptor CCR2 are increased in the hippocampus following pilocarpine-induced status epilepticus. J Neuroinflammation. 2009;6:40.PubMedCentralPubMedCrossRef Foresti ML, Arisi GM, Katki K, Montañez A, Sanchez RM, Shapiro LA. Chemokine CCL2 and its receptor CCR2 are increased in the hippocampus following pilocarpine-induced status epilepticus. J Neuroinflammation. 2009;6:40.PubMedCentralPubMedCrossRef
8.
Zurück zum Zitat Mukherjee S, Katki K, Arisi GM, Foresti ML, Shapiro LA. Early TBI-induced cytokine alterations are similarly detected by two distinct methods of multiplex assay. Front Mol Neurosci. 2011;4:21.PubMedCentralPubMedCrossRef Mukherjee S, Katki K, Arisi GM, Foresti ML, Shapiro LA. Early TBI-induced cytokine alterations are similarly detected by two distinct methods of multiplex assay. Front Mol Neurosci. 2011;4:21.PubMedCentralPubMedCrossRef
9.
Zurück zum Zitat Racine RJ. Modification of seizure activity by electrical stimulation. II. Motor seizure. Electroencephalogr Clin Neurophysiol. 1972;32:281–94.PubMedCrossRef Racine RJ. Modification of seizure activity by electrical stimulation. II. Motor seizure. Electroencephalogr Clin Neurophysiol. 1972;32:281–94.PubMedCrossRef
10.
Zurück zum Zitat van Gassen KL, de Wit M, Koerkamp MJ, Rensen MG, van Rijen PC, Holstege FC, et al. Possible role of the innate immunity in temporal lobe epilepsy. Epilepsia. 2008;49:1055–65.PubMedCrossRef van Gassen KL, de Wit M, Koerkamp MJ, Rensen MG, van Rijen PC, Holstege FC, et al. Possible role of the innate immunity in temporal lobe epilepsy. Epilepsia. 2008;49:1055–65.PubMedCrossRef
11.
Zurück zum Zitat Gorter JA, van Vliet EA, Aronica E, Breit T, Rauwerda H, da Silva FHL, et al. Potential new antiepileptogenic targets indicated by microarray analysis in a rat model for temporal lobe epilepsy. J Neurosci. 2006;26:11083–110.PubMedCrossRef Gorter JA, van Vliet EA, Aronica E, Breit T, Rauwerda H, da Silva FHL, et al. Potential new antiepileptogenic targets indicated by microarray analysis in a rat model for temporal lobe epilepsy. J Neurosci. 2006;26:11083–110.PubMedCrossRef
12.
Zurück zum Zitat Sharma AK, Searfoss GH, Reams RY, Jordan WH, Snyder PW, Chiang AY, et al. Kainic acid-induced F-344 rat model of mesial temporal lobe epilepsy: gene expression and canonical pathways. Toxicol Pathol. 2009;37:776–89.PubMedCrossRef Sharma AK, Searfoss GH, Reams RY, Jordan WH, Snyder PW, Chiang AY, et al. Kainic acid-induced F-344 rat model of mesial temporal lobe epilepsy: gene expression and canonical pathways. Toxicol Pathol. 2009;37:776–89.PubMedCrossRef
13.
Zurück zum Zitat Wu Y, Wang X, Mo X, Xi Z, Xiao F, Li J, et al. Expression of monocyte chemoattractant protein-1 in brain tissue of patients with intractable epilepsy. Clin Neuropathol. 2008;27:55–63.PubMedCrossRef Wu Y, Wang X, Mo X, Xi Z, Xiao F, Li J, et al. Expression of monocyte chemoattractant protein-1 in brain tissue of patients with intractable epilepsy. Clin Neuropathol. 2008;27:55–63.PubMedCrossRef
14.
Zurück zum Zitat Xu JH, Long L, Tang YC, Zhang JT, Hut HT, Tang FR. CCR3, CCR2A and macrophage inflammatory protein (MIP)-1α, monocyte chemotactic protein-1 (MCP-1) in the mouse hippocampus during and after pilocarpine-induced status epilepticus (PISE). Neuropathol Appl Neurobiol. 2009;35:496–514.PubMedCrossRef Xu JH, Long L, Tang YC, Zhang JT, Hut HT, Tang FR. CCR3, CCR2A and macrophage inflammatory protein (MIP)-1α, monocyte chemotactic protein-1 (MCP-1) in the mouse hippocampus during and after pilocarpine-induced status epilepticus (PISE). Neuropathol Appl Neurobiol. 2009;35:496–514.PubMedCrossRef
15.
Zurück zum Zitat Sheehan JJ, Zhou C, Gravanis I, Rogove AD, Wu YP, Bogenhagen DF, et al. Proteolytic activation of monocyte chemoattractant protein-1 by plasmin underlies excitotoxic neurodegeneration in mice. J Neurosci. 2007;27:1738–45.PubMedCrossRef Sheehan JJ, Zhou C, Gravanis I, Rogove AD, Wu YP, Bogenhagen DF, et al. Proteolytic activation of monocyte chemoattractant protein-1 by plasmin underlies excitotoxic neurodegeneration in mice. J Neurosci. 2007;27:1738–45.PubMedCrossRef
16.
Zurück zum Zitat Kalehua AN, Nagel JE, Whelchel LM, Gides JJ, Pyle RS, Smith RJ, et al. Monocyte chemoattractant protein-1 and macrophage inflammatory protein-2 are involved in both excitotoxin-induced neurodegeneration and regeneration. Exp Cell Res. 2004;297:197–211.PubMedCrossRef Kalehua AN, Nagel JE, Whelchel LM, Gides JJ, Pyle RS, Smith RJ, et al. Monocyte chemoattractant protein-1 and macrophage inflammatory protein-2 are involved in both excitotoxin-induced neurodegeneration and regeneration. Exp Cell Res. 2004;297:197–211.PubMedCrossRef
17.
Zurück zum Zitat Bedner P, Dupper A, Hüttmann K, Müller J, Herde MK, Dublin P, et al. Astrocyte uncoupling as a cause of human temporal lobe epilepsy. Brain. 2015;138:1208–22.PubMedCrossRef Bedner P, Dupper A, Hüttmann K, Müller J, Herde MK, Dublin P, et al. Astrocyte uncoupling as a cause of human temporal lobe epilepsy. Brain. 2015;138:1208–22.PubMedCrossRef
18.
Zurück zum Zitat Nelson TE, Hao C, Manos J, Ransohoff RM, Gruol DL. Altered hippocampal synaptic transmission in transgenic mice with astrocyte-targeted enhanced CCL2 expression. Brain Behav Immun. 2011;25 Suppl 1:S106–19.PubMedCentralPubMedCrossRef Nelson TE, Hao C, Manos J, Ransohoff RM, Gruol DL. Altered hippocampal synaptic transmission in transgenic mice with astrocyte-targeted enhanced CCL2 expression. Brain Behav Immun. 2011;25 Suppl 1:S106–19.PubMedCentralPubMedCrossRef
19.
Zurück zum Zitat Lv R, Xu X, Luo Z, Shen N, Wang F, Zhao Y. Pyrrolidine dithiocarbamate (PDTC) inhibits the overexpression of MCP-1 and attenuates microglial activation in the hippocampus of a pilocarpine-induced status epilepticus rat model. Exp Ther Med. 2014;7:39–45.PubMedCentralPubMed Lv R, Xu X, Luo Z, Shen N, Wang F, Zhao Y. Pyrrolidine dithiocarbamate (PDTC) inhibits the overexpression of MCP-1 and attenuates microglial activation in the hippocampus of a pilocarpine-induced status epilepticus rat model. Exp Ther Med. 2014;7:39–45.PubMedCentralPubMed
20.
Zurück zum Zitat Mennicken F, Chabot JG, Quirion R. Systemic administration of kainic acid in adult rat stimulates expression of the chemokine receptor CCR5 in the forebrain. Glia. 2002;37:124–38.PubMedCrossRef Mennicken F, Chabot JG, Quirion R. Systemic administration of kainic acid in adult rat stimulates expression of the chemokine receptor CCR5 in the forebrain. Glia. 2002;37:124–38.PubMedCrossRef
21.
Zurück zum Zitat Kan AA, van der Hel WS, Kolk SM, Bos IW, Verlinde SA, van Nieuwenhuizen O, et al. Prolonged increase in rat hippocampal chemokine signalling after status epilepticus. J Neuroimmunol. 2012;245:15–22.PubMedCrossRef Kan AA, van der Hel WS, Kolk SM, Bos IW, Verlinde SA, van Nieuwenhuizen O, et al. Prolonged increase in rat hippocampal chemokine signalling after status epilepticus. J Neuroimmunol. 2012;245:15–22.PubMedCrossRef
22.
Zurück zum Zitat Louboutin JP, Chekmasova A, Marusich E, Agrawal L, Strayer DS. Role of CCR5 and its ligands in the control of vascular inflammation and leukocyte recruitment required for acute excitotoxic seizure induction and neural damage. FASEB J. 2010;25:737–53.PubMedCrossRef Louboutin JP, Chekmasova A, Marusich E, Agrawal L, Strayer DS. Role of CCR5 and its ligands in the control of vascular inflammation and leukocyte recruitment required for acute excitotoxic seizure induction and neural damage. FASEB J. 2010;25:737–53.PubMedCrossRef
23.
Zurück zum Zitat Chen Z, Yu S, Bakhiet M, Winblad B, Zhu J. The chemokine receptor CCR5 is not a necessary inflammatory mediator in kainic acid-induced hippocampal injury: evidence for a compensatory effect by increased CCR2 and CCR3. J Neurochem. 2003;86:61–8.PubMedCrossRef Chen Z, Yu S, Bakhiet M, Winblad B, Zhu J. The chemokine receptor CCR5 is not a necessary inflammatory mediator in kainic acid-induced hippocampal injury: evidence for a compensatory effect by increased CCR2 and CCR3. J Neurochem. 2003;86:61–8.PubMedCrossRef
24.
Zurück zum Zitat Croll SD, Goodman JH, Scharfman HE. Vascular endothelial growth factor (VEGF) in seizures: a double-edged sword. Adv Exp Med Biol. 2004;548:57–68.PubMedCentralPubMedCrossRef Croll SD, Goodman JH, Scharfman HE. Vascular endothelial growth factor (VEGF) in seizures: a double-edged sword. Adv Exp Med Biol. 2004;548:57–68.PubMedCentralPubMedCrossRef
Metadaten
Titel
Increased CCL2, CCL3, CCL5, and IL-1β cytokine concentration in piriform cortex, hippocampus, and neocortex after pilocarpine-induced seizures
verfasst von
Gabriel M. Arisi
Maira L. Foresti
Khurshed Katki
Lee A. Shapiro
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2015
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-015-0347-z

Weitere Artikel der Ausgabe 1/2015

Journal of Neuroinflammation 1/2015 Zur Ausgabe