251
|
Simeone TA, Simeone KA, Stafstrom CE, Rho JM. Do ketone bodies mediate the anti-seizure effects of the ketogenic diet? Neuropharmacology 2018; 133:233-241. [PMID: 29325899 PMCID: PMC5858992 DOI: 10.1016/j.neuropharm.2018.01.011] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 11/27/2017] [Accepted: 01/07/2018] [Indexed: 01/01/2023]
Abstract
Although the mechanisms underlying the anti-seizure effects of the high-fat ketogenic diet (KD) remain unclear, a long-standing question has been whether ketone bodies (i.e., β-hydroxybutyrate, acetoacetate and acetone), either alone or in combination, contribute mechanistically. The traditional belief has been that while ketone bodies reflect enhanced fatty acid oxidation and a general shift toward intermediary metabolism, they are not likely to be the key mediators of the KD's clinical effects, as blood levels of β-hydroxybutyrate do not correlate consistently with improved seizure control. Against this unresolved backdrop, new data support ketone bodies as having anti-seizure actions. Specifically, β-hydroxybutyrate has been shown to interact with multiple novel molecular targets such as histone deacetylases, hydroxycarboxylic acid receptors on immune cells, and the NLRP3 inflammasome. Clearly, as a diet-based therapy is expected to render a broad array of biochemical, molecular, and cellular changes, no single mechanism can explain how the KD works. Specific metabolic substrates or enzymes are only a few of many important factors influenced by the KD that can collectively influence brain hyperexcitability and hypersynchrony. This review summarizes recent novel experimental findings supporting the anti-seizure and neuroprotective properties of ketone bodies.
Collapse
Affiliation(s)
- Timothy A Simeone
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, USA
| | - Kristina A Simeone
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, USA
| | - Carl E Stafstrom
- Department of Neurology, and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jong M Rho
- Department of Pediatrics, Department of Clinical Neurosciences, and Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
252
|
Dysregulated long non-coding RNAs in the temporal lobe epilepsy mouse model. Seizure 2018; 58:110-119. [DOI: 10.1016/j.seizure.2018.04.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 04/05/2018] [Accepted: 04/11/2018] [Indexed: 02/07/2023] Open
|
253
|
A mechanistic approach to explore the neuroprotective potential of zonisamide in seizures. Inflammopharmacology 2018; 26:1125-1131. [PMID: 29644555 DOI: 10.1007/s10787-018-0478-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/30/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Epilepsy, a disease of the brain, is one of the most common serious neurological conditions. It is associated with a group of processes which alter energy metabolism, interrupt cellular ionic homeostasis, cause receptor dysfunction, activate inflammatory cascade, alter neurotransmitter uptake and result in neuronal damage. The increasing knowledge and understanding about the basis of neuronal changes in epilepsy lead to investigate the mechanistic pathway of neuroprotective agents in epilepsy. With this background, the present study is designed to reveal the molecular and biochemical mechanisms involved in the neuroprotective potential of zonisamide in epilepsy. METHODS Seizure-induced neuronal damage was produced by maximal electroshock seizures in animals. The oxidative stress and neuroinflammatory and apoptotic markers were assessed in the brain tissue of animals. RESULTS AND DISCUSSION The present findings revealed that zonisamide treatment prevented the development of seizures in animals. Seizures-induced free radicals production and neuroinflammation were markedly ameliorated by zonisamide administration. In conclusion, the present study demonstrated the mechanisms behind the strong neuroprotective potential of zonisamide against seizures by attenuating the oxidative stress, inflammatory cascade and neuronal death associated with progression of seizures. It can be further developed as a neuroprotective agent for epilepsy and other neurodegenerative disorders.
Collapse
|
254
|
Chronic agomelatine treatment prevents comorbid depression in the post-status epilepticus model of acquired epilepsy through suppression of inflammatory signaling. Neurobiol Dis 2018; 115:127-144. [PMID: 29653194 DOI: 10.1016/j.nbd.2018.04.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 03/08/2018] [Accepted: 04/04/2018] [Indexed: 12/22/2022] Open
Abstract
Inflammatory signal molecules are suggested to be involved in the mechanism underlying comorbid depression in epilepsy. In the present study, we tested the hypothesis that the novel antidepressant agomelatine, a potent melatonin MT1 and MT2 receptor agonist and serotonin 5HT2C receptor antagonist, can prevent depressive symptoms developed during the chronic epileptic phase by suppressing an inflammatory response. Chronic treatment with agomelatine (40 mg/kg, i.p.) was initiated an hour after the kainate acid (KA)-induced status epilepticus (SE) and maintained for a period of 10 weeks in Wistar rats. Registration of spontaneous motor seizures was performed through a video (24 h/day) and EEG monitoring. Antidepressant activity of agomelatine was explored in the splash test, sucrose preference test (SPT) and forced swimming test (FST) while anxiolytic effect was observed through the novelty suppression-feeding test (NSFT) during chronic phase in epileptic rats. The frequency of motor seizures detected by video and EEG recording did not differ between vehicle and Ago group. Rats with registered spontaneous motor seizures showed symptoms typical for depressive behavior that included decreased grooming, anhedonia during the dark period and hopeless-like behavior. Epileptic rats exhibited also anxiety with novelty-induced hypophagia. This behavioral deficit correlated with increased signal markers of inflammation (plasma levels of interleukin (IL)-1β and activated glia in brain), while plasma corticosterone levels were not changed. Agomelatine treatment during epileptogenesis exerted a clear antidepressant effect by suppressing all behavioral hallmarks, reducing plasma IL-1β levels and preventing microgliosis and astrogliosis in specific limbic regions. The present results suggest that agomelatine treatment starting after SE can provide an effective therapy of comorbid depression in chronic epileptic condition through suppression of inflammatory signaling.
Collapse
|
255
|
Matrix Metalloproteinase-Mediated Blood-Brain Barrier Dysfunction in Epilepsy. J Neurosci 2018; 38:4301-4315. [PMID: 29632167 DOI: 10.1523/jneurosci.2751-17.2018] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 03/12/2018] [Accepted: 03/21/2018] [Indexed: 01/08/2023] Open
Abstract
The blood-brain barrier is dysfunctional in epilepsy, thereby contributing to seizure genesis and resistance to antiseizure drugs. Previously, several groups reported that seizures increase brain glutamate levels, which leads to barrier dysfunction. One critical component of barrier dysfunction is brain capillary leakage. Based on our preliminary data, we hypothesized that glutamate released during seizures mediates an increase in matrix-metalloproteinase (MMP) expression and activity levels, thereby contributing to barrier leakage. To test this hypothesis, we exposed isolated brain capillaries from male Sprague Dawley rats to glutamate ex vivo and used an in vivo/ex vivo approach of isolated brain capillaries from female Wistar rats that experienced status epilepticus as an acute seizure model. We found that exposing isolated rat brain capillaries to glutamate increased MMP-2 and MMP-9 protein and activity levels, and decreased tight junction protein levels, which resulted in barrier leakage. We confirmed these findings in vivo in rats after status epilepticus and in brain capillaries from male mice lacking cytosolic phospholipase A2 Together, our data support the hypothesis that glutamate released during seizures signals an increase in MMP-2 and MMP-9 protein expression and activity levels, resulting in blood-brain barrier leakage.SIGNIFICANCE STATEMENT The mechanism leading to seizure-mediated blood-brain barrier dysfunction in epilepsy is poorly understood. In the present study, we focused on defining this mechanism in the brain capillary endothelium. We demonstrate that seizures trigger a pathway that involves glutamate signaling through cytosolic phospholipase A2, which increases MMP levels and decreases tight junction protein expression levels, resulting in barrier leakage. These findings may provide potential therapeutic avenues within the blood-brain barrier to limit barrier dysfunction in epilepsy and decrease seizure burden.
Collapse
|
256
|
Abstract
OBJECTIVE Several cross-sectional studies have reported a relationship between posttraumatic stress disorder (PTSD) and epilepsy. However, the temporal association between PTSD and epilepsy has rarely been investigated. We hypothesized that the risk of developing epilepsy later in life would be higher in patients with PTSD than in those without PTSD. METHODS Using the Taiwan National Health Insurance Research Database, 6425 individuals with PTSD and 24,980 age-/sex-matched controls were enrolled between 2002 and 2009 in our study and followed up to the end of 2011. Those who developed epilepsy during the follow-up period were identified. RESULTS Individuals with PTSD had a higher incidence of developing epilepsy (2.65 versus 0.33 per 1000 person-years, p < .001), with an earlier onset of epilepsy (37.53 years [15.80 years] versus 48.11 years [23.97 years], p = .002) than did the controls. Individuals with PTSD had an elevated risk of developing epilepsy (hazard ratio [HR] = 3.72, 95% confidence interval [CI] = 2.27-6.11) during the follow-up after adjustment for demographic data and medical and psychiatric comorbidities. Sensitivity analyses after excluding the observation in the first year (HR = 2.53, 95% CI = 1.44-4.47) and the first 3 years (HR = 2.14, 95% CI = 1.15-4.01) revealed consistent results. CONCLUSIONS These results supported a temporal association between PTSD and the development of epilepsy. Further studies are warranted to investigate the underlying pathophysiological pathways that explain the longitudinal association of PTSD with subsequent epilepsy.
Collapse
|
257
|
Khadrawy YA, Sawie HG, Hosny EN. Neuroprotective effect of curcumin nanoparticles against rat model of status epilepticus induced by pilocarpine. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2018; 15:jcim-2017-0117. [PMID: 29570450 DOI: 10.1515/jcim-2017-0117] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 02/20/2018] [Indexed: 11/15/2022]
Abstract
Abstract
Background
The present study aims to investigate the neuroprotective effect of curcumin nanoparticles (Cur-NP) on the rat model of status epilepticus (SE) induced by pilocarpine.
Methods
In the present study, animals were divided into three groups: control animals, rat model of SE induced by a single dose of pilocarpine (380 mg/kg) injected intraperitoneally, and rat model of SE that received a daily intraperitoneal injection of Cur-NP (50 mg/kg) for four consecutive days prior to pilocarpine administration.
Results
The present results revealed a state of oxidative stress in the cortex and hippocampus of rat model of SE as compared to control. This was evident from the significant increase in lipid peroxidation and the significant decrease in reduced glutathione and nitric oxide. In addition, a significant increase in the levels of tumor necrosis factor-alpha (TNF-α) and caspase-3 was detected in the two studied brain regions of rat model of SE. The activities of acetylcholinesterase (AchE) and Na+/K+-ATPase decreased significantly in the cortex and hippocampus of rat model of SE. Protection with Cur-NP prevented oxidative stress and improved the elevated level of caspase-3 in the hippocampus and cortex and the hippocampal TNF-α to nonsignificant changes. Although Cur-NP prevented the decrease in AchE activity in the two studied brain regions, it failed to return Na+/K+-ATPase activity to its normal value.
Conclusions
It is clear from the present findings that Cur-NP could prevent the oxidative stress and neuroinflammation and cell death that were induced during SE. This in turn may help in ameliorating the subsequent cascades of events that follow SE and its development into epileptogenesis.
Collapse
Affiliation(s)
- Yasser A Khadrawy
- Medical Physiology Department, Medical Division, National Research Center, El-Behouth St., Giza, Egypt
| | - Hussein G Sawie
- Medical Physiology Department, Medical Division, National Research Center, El-Behouth St., Giza, Egypt
| | - Eman N Hosny
- Medical Physiology Department, Medical Division, National Research Center, El-Behouth St., Giza, Egypt
| |
Collapse
|
258
|
Li TR, Jia YJ, Ma C, Qiu WY, Wang Q, Shao XQ, Lv RJ. The role of the microRNA-146a/complement factor H/interleukin-1β-mediated inflammatory loop circuit in the perpetuate inflammation of chronic temporal lobe epilepsy. Dis Model Mech 2018; 11:11/3/dmm031708. [PMID: 29590637 PMCID: PMC5897725 DOI: 10.1242/dmm.031708] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 02/19/2018] [Indexed: 01/28/2023] Open
Abstract
Increasing evidence indicates that neuroinflammation plays a crucial role in the pathogenesis of temporal lobe epilepsy (TLE). However, it is unclear how the perpetuate inflammation develops. Some recent studies have suggested the possible involvement of microRNA-146a (miR-146a) in the modulation of inflammatory signaling occurring in TLE. To understand how miR-146a modulates inflammatory signaling in TLE, we investigated the role of interleukin-1β (IL-1β), miR-146a and human complement factor H (CFH) in the perpetuate inflammation in rat models of chronic TLE and U251 cells. We found that enhancive miR-146a could upregulate the expression of IL-1β and downregulate the expression of CFH, whereas reductive miR-146a could downregulate the expression of IL-1β and upregulate the expression of CFH, in hippocampi of chronic TLE rat models. Meanwhile, enhancive miR-146a could increase the abnormal wave forms in the chronic TLE rat models. Additionally, enhancive IL-1β could feedback downregulate the expression of CFH, upregulate the expression of miR-146a and increase the abnormal wave forms in chronic TLE rat models. After CFH gene knockdown in U251 cells, enhancive miR-146a did not upregulate the expression of IL-1β. In summary, this study shows that enhancive miR-146a can upregulate the inflammatory factor IL-1β in chronic TLE by downregulating CFH, and that upregulation of IL-1β plays an important feedback-regulating role in the expression of miR-146a and CFH, forming a miR-146a–CFH–IL-1β loop circuit that initiates a cascade of inflammation and then leads to the perpetuate inflammation in TLE. Therefore, modulation of the miR-146a–CFH–IL-1β loop circuit could be a novel therapeutic target for TLE. Summary: The microRNA-146a–complement factor H–interleukin-1β loop circuit might initiate a cascade of inflammation, leading to the perpetuate inflammation in temporal lobe epilepsy.
Collapse
Affiliation(s)
- Tao-Ran Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases, 6 TianTanXiLi, Dongcheng District, Beijing, 100050, China.,Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 1 East Road of JianShe, Erqi District, Zhengzhou, 450052, China
| | - Yan-Jie Jia
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases, 6 TianTanXiLi, Dongcheng District, Beijing, 100050, China.,Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 1 East Road of JianShe, Erqi District, Zhengzhou, 450052, China
| | - Chao Ma
- Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Peking Union Medical College,1 Shuai Fu Yuan, Dongcheng District, Beijing, 100730, China
| | - Wen-Ying Qiu
- Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Peking Union Medical College,1 Shuai Fu Yuan, Dongcheng District, Beijing, 100730, China
| | - Qun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases, 6 TianTanXiLi, Dongcheng District, Beijing, 100050, China
| | - Xiao-Qiu Shao
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases, 6 TianTanXiLi, Dongcheng District, Beijing, 100050, China
| | - Rui-Juan Lv
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases, 6 TianTanXiLi, Dongcheng District, Beijing, 100050, China
| |
Collapse
|
259
|
Shoji H, Irino Y, Yoshida M, Miyakawa T. Behavioral effects of long-term oral administration of aluminum ammonium sulfate in male and female C57BL/6J mice. Neuropsychopharmacol Rep 2018; 38:18-36. [PMID: 30106265 PMCID: PMC7292291 DOI: 10.1002/npr2.12002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/27/2017] [Accepted: 11/27/2017] [Indexed: 12/18/2022] Open
Abstract
Background Aluminum (Al) is considered to be a neurotoxic metal, and excessive exposure to Al has been reported to be a potential risk factor for neurodegenerative diseases. Al ammonium sulfate is one of the Al compounds that is widely used as a food additive. However, the effects of the oral administration of Al ammonium sulfate on physical development and behavior remain to be examined. Methods In this study, we investigated the effects of the administration of Al ammonium sulfate 12‐water dissolved in drinking water (0.075 mg/mL) beginning in adolescence on various types of behavior in adult female C57BL/6J mice through a battery of behavioral tests (low‐dose experiment; Experiment 1). We further examined the behavioral effects of the oral administration of a higher dose of the Al compound in drinking water (1 mg/mL) beginning in the prenatal period on behavior in adult male and female mice (high‐dose experiment; Experiment 2). Results In the low‐dose experiment, in which females’ oral intake of Al was estimated to be 0.97 mg Al/kg/d as adults, Al‐treated females exhibited an increase in total arm entries in the elevated plus maze test, an initial decrease and subsequent increase in immobility in the forced swim test, and reduced freezing in the fear conditioning test approximately 1 month after the conditioning session compared with vehicle‐treated females (uncorrected P < .05). However, the behavioral differences did not reach a statistically significant level after correction for multiple testing. In the high‐dose experiment, in which animals’ oral intakes were estimated to be about ten times higher than those in the low‐dose experiment, behavioral differences found in the low‐dose experiment were not observed in high‐dose Al‐treated mice, suggesting that the results of the low‐dose experiment might be false positives. Additionally, although high‐dose Al‐treated females exhibited increased social contacts with unfamiliar conspecifics and impaired reference memory performance, and high‐dose Al‐treated mice exhibited decreases in prepulse inhibition and in correct responses in the working memory task (uncorrected P < .05), the differences in any of the behavioral measures did not reach the significance level after correction for multiple testing. Conclusion Our results show that long‐term oral exposure to Al ammonium sulfate at the doses used in this study may have the potential to induce some behavioral changes in C57BL/6J mice. However, the behavioral effects of Al were small and statistically weak, as indicated by the fact that the results failed to reach the study‐wide significance level. Thus, further study will be needed to replicate the results and reevaluate the behavioral outcomes of oral intake of Al ammonium sulfate. Aluminum (Al) ammonium sulfate was orally administered to C57BL/6J mice (estimated dose of 0.97‐9.78 Al mg/kg/d). Behavioral effects of Al were assessed in a battery of behavioral tests in mice in adulthood. Statistically significant behavioral differences were not found between Al‐ and vehicle‐treated mice.
![]()
Collapse
Affiliation(s)
- Hirotaka Shoji
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Yasuhiro Irino
- Division of Evidence-Based Laboratory Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masaru Yoshida
- Division of Metabolomics Research, Department of Internal Related, Kobe University Graduate School of Medicine, Kobe, Japan.,Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| |
Collapse
|
260
|
Chen L, Deng H, Cui H, Fang J, Zuo Z, Deng J, Li Y, Wang X, Zhao L. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 2018; 9:7204-7218. [PMID: 29467962 PMCID: PMC5805548 DOI: 10.18632/oncotarget.23208] [Citation(s) in RCA: 2742] [Impact Index Per Article: 391.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 11/03/2017] [Indexed: 02/07/2023] Open
Abstract
Inflammation is a biological response of the immune system that can be triggered by a variety of factors, including pathogens, damaged cells and toxic compounds. These factors may induce acute and/or chronic inflammatory responses in the heart, pancreas, liver, kidney, lung, brain, intestinal tract and reproductive system, potentially leading to tissue damage or disease. Both infectious and non-infectious agents and cell damage activate inflammatory cells and trigger inflammatory signaling pathways, most commonly the NF-κB, MAPK, and JAK-STAT pathways. Here, we review inflammatory responses within organs, focusing on the etiology of inflammation, inflammatory response mechanisms, resolution of inflammation, and organ-specific inflammatory responses.
Collapse
Affiliation(s)
- Linlin Chen
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
| | - Huidan Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
| | - Hengmin Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Jing Fang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Zhicai Zuo
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Junliang Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Yinglun Li
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Xun Wang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Ling Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| |
Collapse
|
261
|
Zhang H, Qu Y, Wang A. Antagonist targeting microRNA-146a protects against lithium-pilocarpine-induced status epilepticus in rats by nuclear factor-κB pathway. Mol Med Rep 2018; 17:5356-5361. [PMID: 29363732 DOI: 10.3892/mmr.2018.8465] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 12/05/2017] [Indexed: 11/05/2022] Open
Abstract
Previous studies have indicated that nuclear factor-κB (NF-κB) has an important role in the pathogenesis of epilepsy. The aim of the present study was to evaluate the expression of microRNA (miRNA)‑146a, phosphorylated (p)‑P65/P65, B‑cell lymphoma‑2(Bcl‑2)/Bcl‑2‑associated X protein (Bax) and pro‑inflammatory cytokines, such as interleukin (IL)‑6, IL‑1β and tumor necrosis factor (TNF‑α) in the brain tissue of rats with epilepsy. Sprague‑Dawley rats were used to establish the epilepsy model using the lithium‑pilocarpine method. The expression of miR‑146a, pro‑inflammatory cytokines, P‑glycoprotein (P‑gp), Bcl‑2/Bax and p‑P65/P65 were assessed by reverse transcription‑semi‑quantitative polymerase chain reaction, enzyme‑linked immunosorbent assay and western blotting, respectively. Hematoxylin and eosin staining was used to determine the pathology of epilepsy. The current findings revealed that the expression of miR‑146a was greater in the model group compared with the control group, and that the expression of miR‑146a reached a maximum at 7 days post‑treatment. The expression levels of IL‑1β, IL‑6 and TNF‑α were significantly reduced in the miR‑146a antagonist group when compared with the model group. Additionally, the expression levels of P‑gp and p‑P65/P65 were significantly reduced following the addition of the miR‑146a antagonist, whereas the expression levels of Bcl‑2/Bax significantly increased under the same conditions. Therefore, the NF‑κB pathway and miR‑146a may be potential therapeutic targets in the treatment of epilepsy.
Collapse
Affiliation(s)
- Huilong Zhang
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Yun Qu
- Department of Emergency, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Aihua Wang
- Department of Neurology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
262
|
Miratashi Yazdi SA, Abbasi M, Miratashi Yazdi SM. Epilepsy and vitamin D: a comprehensive review of current knowledge. Rev Neurosci 2018; 28:185-201. [PMID: 27988507 DOI: 10.1515/revneuro-2016-0044] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 09/24/2016] [Indexed: 12/29/2022]
Abstract
Vitamin D has been considered as neurosteroid, and its pivotal role in neuroprotection, brain development, and immunomodulation has been noticed in studies; however, our knowledge regarding its role in neurological disorders is still developing. The potential role of vitamin D in the pathophysiology and treatment of epilepsy, as one the most prevalent neurological disorders, has received less attention in recent years. In this article, we review the possible relationship between vitamin D and epilepsy from different aspects, including the action mechanism of vitamin D in the central nervous system and ecological and epidemiological findings. We also present the outcome of studies that evaluated the level of vitamin D and the impact of administrating vitamin D in epileptic patients or animal subjects. We also review the current evidence on interactions between vitamin D and antiepileptic drugs.
Collapse
|
263
|
Abstract
Until a decade ago, epilepsy research had focused mainly on alterations of neuronal activities and excitability. Such neurocentric emphasis has neglected the role of glia and involvement of inflammation in the pathogenesis of epilepsy. It is becoming clear that immune and inflammatory reactions do occur in the brain despite the brain's lack of conventional lymphatic drainage and graft acceptance and the presence of vascular brain barrier that tightly regulates infiltration of blood monocytes and lymphocytes. The critical roles of brain-resident immune mediators and of brain-infiltrating peripheral leukocytes are increasingly recognized. Inflammatory processes, including activation of microglia and astrocytes and production of proinflammatory cytokines and related molecules, occur in human epilepsy as well as in experimental models of epilepsy. Immune mechanism that underlies evolution of drug-resistant epilepsy and epileptic encephalopathy represents a new target and will aid in development of novel immunotherapeutic drugs and therapies against the key constituents in immune pathways.
Collapse
Affiliation(s)
- Sookyong Koh
- 1 Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
264
|
Liu Z, Yang C, Meng X, Li Z, Lv C, Cao P. Neuroprotection of edaravone on the hippocampus of kainate-induced epilepsy rats through Nrf2/HO-1 pathway. Neurochem Int 2018; 112:159-165. [DOI: 10.1016/j.neuint.2017.07.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/11/2017] [Accepted: 07/07/2017] [Indexed: 10/19/2022]
|
265
|
Oliveira CVD, Grigoletto J, Canzian JM, Duarte MMMF, Duarte T, Furian AF, Oliveira MS. Effect of atorvastatin on behavioral alterations and neuroinflammation during epileptogenesis. Epilepsy Behav 2018; 78:109-117. [PMID: 29186698 DOI: 10.1016/j.yebeh.2017.10.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/06/2017] [Accepted: 10/13/2017] [Indexed: 01/16/2023]
Abstract
Temporal lobe epilepsy (TLE) is the most frequent and medically refractory type of epilepsy in humans. In addition to seizures, patients with TLE suffer from behavioral alterations and cognitive deficits. Poststatus epilepticus model of TLE induced by pilocarpine in rodents has enhanced the understanding of the processes leading to epilepsy and thus, of potential targets for antiepileptogenic therapies. Clinical and experimental evidence suggests that inflammatory processes in the brain may critically contribute to epileptogenesis. Statins are inhibitors of cholesterol synthesis, and present pleiotropic effects that include antiinflammatory properties. We aimed the present study to test the hypothesis that atorvastatin prevents behavioral alterations and proinflammatory state in the early period after pilocarpine-induced status epilepticus. Male and female C57BL/6 mice were subjected to status epilepticus induced by pilocarpine and treated with atorvastatin (10 or 100mg/kg) for 14days. Atorvastatin slightly improved the performance of mice in the open-field and object recognition tests. In addition, atorvastatin dose-dependently decreased basal and status epilepticus-induced levels of interleukin-1β (IL-1β), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and interferon-γ (INF-γ) and increased interleukin-10 (IL-10) levels in the hippocampus and cerebral cortex. The antiinflammatory effects of atorvastatin were qualitatively identical in both sexes. Altogether, these findings extend the range of beneficial actions of atorvastatin and indicate that its antiinflammatory effects may be useful after an epileptogenic insult.
Collapse
Affiliation(s)
| | - Jéssica Grigoletto
- Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Julia Marion Canzian
- Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | | | - Thiago Duarte
- Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Ana Flávia Furian
- Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil; Graduate Program in Food and Science Technology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | | |
Collapse
|
266
|
Volumetric response of the adult brain to seizures depends on the developmental stage when systemic inflammation was induced. Epilepsy Behav 2018; 78:280-287. [PMID: 29128467 DOI: 10.1016/j.yebeh.2017.09.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 09/15/2017] [Accepted: 09/15/2017] [Indexed: 01/07/2023]
Abstract
Inflammation has detrimental influences on the developing brain including triggering the epileptogenesis. On the other hand, seizure episodes may induce inflammatory processes and further increase of brain excitability. The present study focuses on the problem whether transitory systemic inflammation during developmental period may have critical importance to functional and/or structural features of the adult brain. An inflammatory status was induced with lipopolysaccharide (LPS) in 6- or 30-day-old rats. Two-month-old rats which experienced the inflammation and untreated controls received injections of pilocarpine, and the intensity of their seizure behavior was rated during a 6-hour period. Three days thereafter, the animals were perfused; their brains were postfixed and subjected to magnetic resonance imaging (MRI) scans. Then, volumes of the brain and of its main regions were assessed. LPS injections alone performed at different developmental stages led to different changes in the volume of adult brain and also to different susceptibility to seizures induced in adulthood. Moreover, the LPS pretreatments modified different volumetric responses of the brain and of its regions to seizures. The responses showed strong inverse correlations with the intensity of seizures but exclusively in rats treated with LPS on postnatal day 30. It could be concluded that generalized inflammation elicited at developmental stages may have strong age-dependent effects on the adult brain regarding not only its susceptibility to action of a seizuregenic agent but also its volumetric reactivity to seizures.
Collapse
|
267
|
Zheng F, Yang Y, Lu S, Yang Q, Li Y, Xu X, Zhang Y, Liu F, Tian X, Wang X. CD36 Deficiency Suppresses Epileptic Seizures. Neuroscience 2017; 367:110-120. [PMID: 29111364 DOI: 10.1016/j.neuroscience.2017.10.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 10/16/2017] [Accepted: 10/18/2017] [Indexed: 01/01/2023]
Abstract
Cluster of differentiation 36 (CD36) belongs to the class B scavenger receptor family. CD36 is a glycoprotein found on the surface of various cell types and has been implicated in the mechanism of numerous central nervous system (CNS) diseases. However, the relationship between CD36 and epilepsy remains unknown. In this study, we aimed to detect the expression of CD36 in two different chronic epileptic mouse models and determine whether CD36 deficiency leads to suppressive neuronal hyperexcitability and decreased susceptibility of epileptic seizures. Here, we found that CD36 was expressed in the neurons and that CD36 expression was significantly elevated in epileptic mice induced by pentylenetetrazol (PTZ) and kainic acid (KA). Behavioral studies revealed that CD36 deletion in mice (CD36-/- mice) resulted in an attenuated progression of chronic epilepsy compared with wild-type (WT) mice. Whole-cell patch-clamp technique exhibited a decreased frequency of action potentials (APs) in the hippocampal slices of CD36-/- mice. In addition, local field potential (LFP) analysis further indicated that CD36 deletion reduced the frequency and duration of epileptiform-like discharges. These results revealed that CD36 deficiency could produce an antiepileptic effect and could provide new insight into antiepileptic treatment.
Collapse
Affiliation(s)
- Fangshuo Zheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China.
| | - Yong Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China.
| | - Shanshan Lu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China.
| | - Qin Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China.
| | - Yun Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China.
| | - Xin Xu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China.
| | - Yanke Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China.
| | - Feng Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China.
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China.
| | - Xuefeng Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China; Center of Epilepsy, Beijing Institute for Brain Disorders, 10 Xitoutiao, Youanmen, Fengtai District, Beijing 100069, China.
| |
Collapse
|
268
|
Gao Y, Luo CL, Li LL, Ye GH, Gao C, Wang HC, Huang WW, Wang T, Wang ZF, Ni H, Chen XP, Tao LY. IL-33 Provides Neuroprotection through Suppressing Apoptotic, Autophagic and NF-κB-Mediated Inflammatory Pathways in a Rat Model of Recurrent Neonatal Seizure. Front Mol Neurosci 2017; 10:423. [PMID: 29311813 PMCID: PMC5742123 DOI: 10.3389/fnmol.2017.00423] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/04/2017] [Indexed: 12/19/2022] Open
Abstract
Interleukin-33 (IL-33) is a novel identified chromatin-associated cytokine of IL-1 family cytokines. It signals through a heterodimer comprised of ST2L and IL-1RAcp, and plays a crucial role in many diseases. However, very little is known about the role and underlying intricate mechanisms of IL-33 in recurrent neonatal seizure (RNS). To determine whether IL-33 plays an important regulatory role, we established a neonatal seizure model in this study. Rats were subjected to recurrent seizures induced by inhaling volatile flurothyl. Recombinant IL-33 or PBS were also administered by intraperitoneally (IP) before surgery, respectively. Here, our current results indicated that RNS contributed to a significant reduction in IL-33 and its specific receptor (ST2L) expressions in cortex. While, in hippocampus, RNS induced an increase in IL-33 and ST2L evidently, compared with Sham group. After injection with IL-33, however, a remarkable increase in total IL-33 was detected both in brain cortex and hippocampus. In addition, IL-33 was mainly co-localized in the nuclear of GFAP+ astrocytes and the cytoplasm of the Iba-1+ microglia and IL-33+/NeuN+ merged cells. In parallel, ST2L was expressed mainly in the membrane of GFAP+ astrocytes, Iba-1+ microglia and NeuN+ neurons, respectively. Furthermore, administration of IL-33 improved RNS-induced behavioral deficits, promoted bodyweight gain, and ameliorated spatial learning and memory ability. Moreover, IL-33 pretreatment blocked the activation of NF-κB, resisted inflammatory cytokines IL-1β and TNF-α increase, as well as suppressed apoptosis and autophagy activation after RNS. Collectively, IL-33 provides potential neuroprotection through suppressing apoptosis, autophagy and at least in part by NF-κB-mediated inflammatory pathways after RNS.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Forensic Medicine, Wenzhou Medical University, Wenzhou, China.,Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Cheng-Liang Luo
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Li-Li Li
- Department of Neurology Laboratory, Children's Hospital of Soochow University, Suzhou, China
| | - Guang-Hua Ye
- Department of Forensic Medicine, Wenzhou Medical University, Wenzhou, China
| | - Cheng Gao
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Hao-Chen Wang
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Wen-Wen Huang
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Tao Wang
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Zu-Feng Wang
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Hong Ni
- Department of Neurology Laboratory, Children's Hospital of Soochow University, Suzhou, China
| | - Xi-Ping Chen
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Lu-Yang Tao
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| |
Collapse
|
269
|
Gumus E, Taskıran AS, Toptas HA, Güney Ö, Kutlu R, Gunes H, Ozdemir E, Arslan G. Effect of the cyclooxygenase-2 inhibitor tenoxicam on pentylenetetrazole-induced epileptic seizures in rats. ACTA ACUST UNITED AC 2017. [DOI: 10.7197/223.v39i32356.369027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
270
|
Amorim RP, Araújo MGL, Valero J, Lopes-Cendes I, Pascoal VDB, Malva JO, da Silva Fernandes MJ. Silencing of P2X7R by RNA interference in the hippocampus can attenuate morphological and behavioral impact of pilocarpine-induced epilepsy. Purinergic Signal 2017; 13:467-478. [PMID: 28707031 PMCID: PMC5714836 DOI: 10.1007/s11302-017-9573-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 06/28/2017] [Indexed: 12/01/2022] Open
Abstract
Cell signaling mediated by P2X7 receptors (P2X7R) has been suggested to be involved in epileptogenesis, via modulation of intracellular calcium levels, excitotoxicity, activation of inflammatory cascades, and cell death, among other mechanisms. These processes have been described to be involved in pilocarpine-induced status epilepticus (SE) and contribute to hyperexcitability, resulting in spontaneous and recurrent seizures. Here, we aimed to investigate the role of P2X7R in epileptogenesis in vivo using RNA interference (RNAi) to inhibit the expression of this receptor. Small interfering RNA (siRNA) targeting P2X7R mRNA was injected into the lateral ventricles (icv) 6 h after SE. Four groups were studied: Saline-Vehicle, Saline-siRNA, Pilo-Vehicle, and Pilo-siRNA. P2X7R was quantified by western blotting and neuronal death assessed by Fluoro-Jade B histochemistry. The hippocampal volume (edema) was determined 48 h following RNAi. Behavioral parameters as latency to the appearance of spontaneous seizures and the number of seizures were determined until 60 days after the SE onset. The Saline-siRNA and Pilo-siRNA groups showed a 43 and 37% reduction, respectively, in P2X7R protein levels compared to respective vehicle groups. Neuroprotection was observed in CA1 and CA3 of the Pilo-siRNA group compared to Pilo-Vehicle. P2X7R silencing in pilocarpine group reversed the increase in the edema detected in the hilus, suprapyramidal dentate gyrus, CA1, and CA3; reduced mortality rate following SE; increased the time to onset of spontaneous seizure; and reduced the number of seizures, when compared to the Pilo-Vehicle group. Therefore, our data highlights the potential of P2X7R as a therapeutic target for the adjunct treatment of epilepsy.
Collapse
Affiliation(s)
- Rebeca Padrão Amorim
- Departamento de Neurologia e Neurocirurgia, Disciplina de Neurociência, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, 2° andar, São Paulo, SP, CEP 04039-032, Brazil
| | - Michelle Gasparetti Leão Araújo
- Departamento de Neurologia e Neurocirurgia, Disciplina de Neurociência, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, 2° andar, São Paulo, SP, CEP 04039-032, Brazil
| | - Jorge Valero
- Centro de Neurociências e Biologia Celular, Universidade de Coimbra, Coimbra, Portugal
- Achucarro Basque Center for Neuroscience, Zamudio, Bizkaia, Spain
- Ikerbasque Basque Foundation for Science, Bilbao, Bizkaia, Spain
| | - Iscia Lopes-Cendes
- Departamento de Genética Médica, Faculdade de Medicina da Unicamp, Campinas, SP, Brazil
| | | | - João Oliveira Malva
- Institute of Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Maria José da Silva Fernandes
- Departamento de Neurologia e Neurocirurgia, Disciplina de Neurociência, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, 2° andar, São Paulo, SP, CEP 04039-032, Brazil.
| |
Collapse
|
271
|
mTOR-related neuropathology in mutant tsc2 zebrafish: Phenotypic, transcriptomic and pharmacological analysis. Neurobiol Dis 2017; 108:225-237. [DOI: 10.1016/j.nbd.2017.09.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 08/03/2017] [Accepted: 09/05/2017] [Indexed: 12/15/2022] Open
|
272
|
Neumann AM, Abele J, Kirschstein T, Engelmann R, Sellmann T, Köhling R, Müller-Hilke B. Mycophenolate mofetil prevents the delayed T cell response after pilocarpine-induced status epilepticus in mice. PLoS One 2017; 12:e0187330. [PMID: 29182639 PMCID: PMC5705158 DOI: 10.1371/journal.pone.0187330] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 10/17/2017] [Indexed: 01/01/2023] Open
Abstract
Growing clinical and laboratory evidence corroborates a role for the immune system in the pathophysiology of epilepsy. In order to delineate the immune response following pilocarpine-induced status epilepticus (SE) in the mouse, we monitored the kinetics of leukocyte presence in the hippocampus over the period of four weeks. SE was induced following a ramping protocol of pilocarpine injection into 4–5 weeks old C57BL/6 mice. Brains were removed at days 1–4, 14 or 28 after SE, and the hippocampi were analyzed via flow cytometry, via quantitative reverse transcriptase PCR (qRT-PCR) and via immunohistochemistry. Epileptogenesis was confirmed by Timm staining of mossy fiber sprouting in the inner molecular layer of the dentate gyrus. The flow cytometry data revealed a biphasic immune response following pilocarpine-induced SE with a transient increase in activated CD11b+ and F4/80+ macrophages within the first four days replaced by an increase in CD3+ T-lymphocytes around day 28. This delayed T cell response was confirmed via qRT-PCR and via immunohistochemistry. In addition, qRT-PCR data could show that the delayed T cell response was associated with an increased CD8/CD4 ratio indicating a cytotoxic T cell response after SE. Intriguingly, early intervention with mycophenolate mofetil administration on days 0–3 after SE prevented this delayed T cell response. These results show an orchestrated immunological sequela and provide evidence that the delayed T cell response is sensitive to early immunomodulatory intervention.
Collapse
Affiliation(s)
- Anne-Marie Neumann
- Institute of Immunology, University of Rostock, Rostock, Germany
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Julia Abele
- Institute of Immunology, University of Rostock, Rostock, Germany
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Robby Engelmann
- Institute of Immunology, University of Rostock, Rostock, Germany
| | - Tina Sellmann
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | | |
Collapse
|
273
|
Rossi A, Murta V, Auzmendi J, Ramos AJ. Early Gabapentin Treatment during the Latency Period Increases Convulsive Threshold, Reduces Microglial Activation and Macrophage Infiltration in the Lithium-Pilocarpine Model of Epilepsy. Pharmaceuticals (Basel) 2017; 10:ph10040093. [PMID: 29182533 PMCID: PMC5748648 DOI: 10.3390/ph10040093] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/14/2017] [Accepted: 11/19/2017] [Indexed: 12/20/2022] Open
Abstract
The lithium-pilocarpine model of epilepsy reproduces several features of temporal lobe epilepsy in humans, including the chronological timeline of an initial latency period followed by the development of spontaneous seizures. Epilepsy therapies in humans are implemented, as a rule, after the onset of the spontaneous seizures. We here studied the potential effect on epileptogenesis of starting an early treatment during the latency period, in order to prevent the development of spontaneous seizures. Adult male Wistar rats were treated with 3 mEq/kg LiCl, and 20 h later 30 mg/kg pilocarpine. Once status epilepticus (SE) was achieved, it was allowed to last for 20 min, and then motor seizures were controlled with the administration of 20 mg/kg diazepam. At 1DPSE (DPSE, days post-status epilepticus), animals started to receive 400 mg/kg/day gabapentin or saline for 4 days. At 5DPSE, we observed that SE induced an early profuse microglial and astroglial reactivity, increased synaptogenic trombospondin-1 expression and reduced AQP4 expression in astroglial ending feet. Blood brain barrier (BBB) integrity seemed to be compromised, as infiltrating NG2+ macrophages and facilitated access to the CNS was observed by transplanting eGFP+ blood cells and bone marrow-derived progenitors in the SE animals. The early 4-day gabapentin treatment successfully reduced microglial cell reactivity and blood-borne cell infiltration, without significantly altering the mRNA of proinflammatory cytokines IL-1β and TNFα immediately after the treatment. After 21DSPE, another group of animals that developed SE and received 4 days of gabapentin treatment, were re-exposed to subconvulsive accumulative doses of pilocarpine (10 mg/kg/30 min) and were followed by recording the Racine scale reached. Early 4-day gabapentin treatment reduced the Racine scale reached by the animals, reduced animal mortality, and reduced the number of animals that achieved SE (34% vs. 72%). We conclude that early gabapentin treatment following SE, during the latency period, is able to reduce neuroinflammation and produces a persistent effect that limits seizures and increases convulsive threshold, probably by restricting microglial reactivity and spurious synaptogenesis.
Collapse
Affiliation(s)
- Alicia Rossi
- Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires CP1121, Argentina.
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Profesor E. De Robertis" IBCN UBA-CONICET, Buenos Aires CP1121, Argentina.
| | - Veronica Murta
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Profesor E. De Robertis" IBCN UBA-CONICET, Buenos Aires CP1121, Argentina.
| | - Jerónimo Auzmendi
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Profesor E. De Robertis" IBCN UBA-CONICET, Buenos Aires CP1121, Argentina.
| | - Alberto Javier Ramos
- Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires CP1121, Argentina.
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Profesor E. De Robertis" IBCN UBA-CONICET, Buenos Aires CP1121, Argentina.
| |
Collapse
|
274
|
Tröscher AR, Klang A, French M, Quemada-Garrido L, Kneissl SM, Bien CG, Pákozdy Á, Bauer J. Selective Limbic Blood-Brain Barrier Breakdown in a Feline Model of Limbic Encephalitis with LGI1 Antibodies. Front Immunol 2017; 8:1364. [PMID: 29093718 PMCID: PMC5651237 DOI: 10.3389/fimmu.2017.01364] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/05/2017] [Indexed: 12/30/2022] Open
Abstract
Human leucine-rich glioma-inactivated protein 1 encephalitis (LGI1) is an autoimmune limbic encephalitis in which serum and cerebrospinal fluid contain antibodies targeting LGI1, a protein of the voltage gated potassium channel (VGKC) complex. Recently, we showed that a feline model of limbic encephalitis with LGI1 antibodies, called feline complex partial seizures with orofacial involvement (FEPSO), is highly comparable to human LGI1 encephalitis. In human LGI1 encephalitis, neuropathological investigations are difficult because very little material is available. Taking advantage of this natural animal model to study pathological mechanisms will, therefore, contribute to a better understanding of its human counterpart. Here, we present a brain-wide histopathological analysis of FEPSO. We discovered that blood–brain barrier (BBB) leakage was present not only in all regions of the hippocampus but also in other limbic structures such as the subiculum, amygdale, and piriform lobe. However, in other regions, such as the cerebellum, no leakage was observed. In addition, this brain-region-specific immunoglobulin leakage was associated with the breakdown of endothelial tight junctions. Brain areas affected by BBB dysfunction also revealed immunoglobulin and complement deposition as well as neuronal cell death. These neuropathological findings were supported by magnetic resonance imaging showing signal and volume increase in the amygdala and the piriform lobe. Importantly, we could show that BBB disturbance in LGI1 encephalitis does not depend on T cell infiltrates, which were present brain-wide. This finding points toward another, so far unknown, mechanism of opening the BBB. The limbic predilection sites of immunoglobulin antibody leakage into the brain may explain why most patients with LGI1 antibodies have a limbic phenotype even though LGI1, the target protein, is ubiquitously distributed across the central nervous system.
Collapse
Affiliation(s)
- Anna R Tröscher
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Andrea Klang
- Department for Pathobiology, Institute of Pathology and Forensic Veterinary Medicine, University of Veterinary Medicine, Vienna, Austria
| | - Maria French
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Lucía Quemada-Garrido
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Sibylle Maria Kneissl
- Diagnostic Imaging, Department for Companion Animals and Horses, University of Veterinary Medicine, Vienna, Austria
| | | | - Ákos Pákozdy
- Clinical Unit of Internal Medicine Small Animals, University of Veterinary Medicine, Vienna, Austria
| | - Jan Bauer
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
275
|
Rong X, Yin J, Wang H, Zhang X, Peng Y. Statin treatment may lower the risk of postradiation epilepsy in patients with nasopharyngeal carcinoma. Epilepsia 2017; 58:2172-2177. [PMID: 29034463 DOI: 10.1111/epi.13924] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2017] [Indexed: 01/22/2023]
Affiliation(s)
- Xiaoming Rong
- Department of Neurology; Sun Yat-Sen Memorial Hospital; Sun Yat-Sen University; Guangzhou China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation; Medical Research Center; Sun Yat-Sen Memorial Hospital; Sun Yat-Sen University; Guangzhou China
| | - Jing Yin
- Department of Neurology; Sun Yat-Sen Memorial Hospital; Sun Yat-Sen University; Guangzhou China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation; Medical Research Center; Sun Yat-Sen Memorial Hospital; Sun Yat-Sen University; Guangzhou China
| | - Hongxuan Wang
- Department of Neurology; Sun Yat-Sen Memorial Hospital; Sun Yat-Sen University; Guangzhou China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation; Medical Research Center; Sun Yat-Sen Memorial Hospital; Sun Yat-Sen University; Guangzhou China
| | - Xiaoni Zhang
- Department of Neurology; Sun Yat-Sen Memorial Hospital; Sun Yat-Sen University; Guangzhou China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation; Medical Research Center; Sun Yat-Sen Memorial Hospital; Sun Yat-Sen University; Guangzhou China
| | - Ying Peng
- Department of Neurology; Sun Yat-Sen Memorial Hospital; Sun Yat-Sen University; Guangzhou China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation; Medical Research Center; Sun Yat-Sen Memorial Hospital; Sun Yat-Sen University; Guangzhou China
| |
Collapse
|
276
|
Gales JM, Prayson RA. Chronic inflammation in refractory hippocampal sclerosis-related temporal lobe epilepsy. Ann Diagn Pathol 2017; 30:12-16. [DOI: 10.1016/j.anndiagpath.2017.05.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 05/15/2017] [Indexed: 02/07/2023]
|
277
|
Vilela LR, Lima IV, Kunsch ÉB, Pinto HPP, de Miranda AS, Vieira ÉLM, de Oliveira ACP, Moraes MFD, Teixeira AL, Moreira FA. Anticonvulsant effect of cannabidiol in the pentylenetetrazole model: Pharmacological mechanisms, electroencephalographic profile, and brain cytokine levels. Epilepsy Behav 2017; 75:29-35. [PMID: 28821005 DOI: 10.1016/j.yebeh.2017.07.014] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 07/01/2017] [Accepted: 07/07/2017] [Indexed: 11/26/2022]
Abstract
Cannabidiol (CBD), the main nonpsychotomimetic compound from Cannabis sativa, inhibits experimental seizures in animal models and alleviates certain types of intractable epilepsies in patients. Its pharmacological profile, however, is still uncertain. Here we tested the hypothesis that CBD anticonvulsant mechanisms are prevented by cannabinoid (CB1 and CB2) and vanilloid (TRPV1) receptor blockers. We also investigated its effects on electroencephalographic (EEG) activity and hippocampal cytokines in the pentylenetetrazole (PTZ) model. Pretreatment with CBD (60mg/kg) attenuated seizures induced by intraperitoneal, subcutaneous, and intravenous PTZ administration in mice. The effects were reversed by CB1, CB2, and TRPV1 selective antagonists (AM251, AM630, and SB366791, respectively). Additionally, CBD delayed seizure sensitization resulting from repeated PTZ administration (kindling). This cannabinoid also prevented PTZ-induced EEG activity and interleukin-6 increase in prefrontal cortex. In conclusion, the robust anticonvulsant effects of CBD may result from multiple pharmacological mechanisms, including facilitation of endocannabinoid signaling and TRPV1 mechanisms. These findings advance our understanding on CBD inhibition of seizures, EEG activity, and cytokine actions, with potential implications for the development of new treatments for certain epileptic syndromes.
Collapse
Affiliation(s)
- Luciano R Vilela
- Graduate School in Neuroscience, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Isabel V Lima
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Érica B Kunsch
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Hyorrana Priscila P Pinto
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Aline S de Miranda
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Érica Leandro M Vieira
- Graduate School in Neuroscience, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | | | - Marcio Flávio D Moraes
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Antônio L Teixeira
- Department of Internal Medicine, School of Medicine, Universidade Federal de Minas Gerais, Brazil.
| | - Fabricio A Moreira
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil.
| |
Collapse
|
278
|
Complement system dysregulation in patients affected by Idiopathic Generalized Epilepsy and the effect of antiepileptic treatment. Epilepsy Res 2017; 137:107-111. [PMID: 28985614 DOI: 10.1016/j.eplepsyres.2017.09.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 07/28/2017] [Accepted: 09/15/2017] [Indexed: 11/22/2022]
Abstract
Complement system dysregulation has been hypothesized as a possible pathogenetic factor triggering epileptogenesis in both animal models and human studies. The aim of the present study is to evaluate the complement system in adult patients affected by idiopathic generalized epilepsy (IGE), either untreated or treated by antiepileptic drugs (AEDs). Thirty-seven IGE patients were compared to a population of 20 matched healthy controls. IGE patients underwent neurological investigation, epilepsy diary, 24-h EEG recording, and blood sample for the assessment of the complement factors C3 and C4, fibrinogen, and C-reactive protein (CRP) serum levels. We excluded patients with clinical and subclinical seizures in the 24h before obtaining the blood sample. We observed decreased C3 and C4 serum levels in IGE patients with respect to controls (p<0.05), and in untreated compared to treated IGE patients (p<0.05). We found significant correlations in the IGE group linking C3 to C4 (R=0.34), CRP (R=0.49), and fibrinogen serum levels (R=0.61). This study proved a significant alteration of the complement system in IGE patients not related to ictal conditions. The hyperactivation of the complement cascade was more significant in untreated than in treated IGE patients. Hence, this study documented the complement factors dysregulation in patients affected by IGE. However, the impact of complement system alteration in the epileptogenetic process needs to be clarified.
Collapse
|
279
|
Wang N, Han X, Liu H, Zhao T, Li J, Feng Y, Mi X, Zhang Y, Chen Y, Wang X. Myeloid differentiation factor 88 is up-regulated in epileptic brain and contributes to experimental seizures in rats. Exp Neurol 2017; 295:23-35. [DOI: 10.1016/j.expneurol.2017.05.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 04/27/2017] [Accepted: 05/16/2017] [Indexed: 01/30/2023]
|
280
|
Activation of the peripheral immune system regulates neuronal aromatase in the adult zebra finch brain. Sci Rep 2017; 7:10191. [PMID: 28860515 PMCID: PMC5579002 DOI: 10.1038/s41598-017-10573-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 08/10/2017] [Indexed: 11/23/2022] Open
Abstract
Estradiol provision via neural aromatization decreases neuro-inflammation and –degeneration, but almost nothing is known about the interactions between the peripheral immune system and brain aromatase. Given the vulnerability of the CNS we reasoned that brain aromatization may protect circuits from the threats of peripheral infection; perhaps shielding cells that are less resilient from the degeneration associated with peripheral infection or trauma. Lipopolysaccharide (LPS) or vehicle was administered peripherally to adult zebra finches and sickness behavior was recorded 2 or 24 hours later. The central transcription of cytokines and aromatase was measured, as were telencephalic aromatase activity and immunoreactive aromatase (24 hour time point only). Two hours post LPS, sickness-like behaviors increased, the transcription of IL-1β was higher in both sexes, and TNFα was elevated in females. 24 hours post-LPS, the behavior of LPS birds was similar to controls, and cytokines had returned to baseline, but aromatase mRNA and activity were elevated in both sexes. Immunocytochemistry revealed greater numbers of aromatase-expressing neurons in LPS birds. These data suggest that the activation of the immune system via peripheral endotoxin increases neuronal aromatase; a mechanism that may rapidly generate a potent anti-neuroinflammatory steroid in response to peripheral activation of the immune system.
Collapse
|
281
|
Scavenging reactive oxygen species inhibits status epilepticus-induced neuroinflammation. Exp Neurol 2017; 298:13-22. [PMID: 28822838 DOI: 10.1016/j.expneurol.2017.08.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 07/26/2017] [Accepted: 08/15/2017] [Indexed: 02/07/2023]
Abstract
Inflammation has been identified as an important mediator of seizures and epileptogenesis. Understanding the mechanisms underlying seizure-induced neuroinflammation could lead to the development of novel therapies for the epilepsies. Reactive oxygen species (ROS) are recognized as mediators of seizure-induced neuronal damage and are known to increase in models of epilepsies. ROS are also known to contribute to inflammation in several disease states. We hypothesized that ROS are key modulators of neuroinflammation i.e. pro-inflammatory cytokine production and microglial activation in acquired epilepsy. The role of ROS in modulating seizure-induced neuroinflammation was investigated in the pilocarpine model of temporal lobe epilepsy (TLE). Pilocarpine-induced status epilepticus (SE) resulted in a time-dependent increase in pro-inflammatory cytokine production in the hippocampus and piriform cortex. Scavenging ROS with a small-molecule catalytic antioxidant decreased SE-induced pro-inflammatory cytokine production and microglial activation, suggesting that ROS contribute to SE-induced neuroinflammation. Scavenging ROS also attenuated phosphorylation of ribosomal protein S6, the downstream target of the mammalian target of rapamycin (mTOR) pathway indicating that this pathway might provide one mechanistic link between SE-induced ROS production and inflammation. Together, these results demonstrate that ROS contribute to SE-induced cytokine production and antioxidant treatment may offer a novel approach to control neuroinflammation in epilepsy.
Collapse
|
282
|
Deshpande T, Li T, Herde MK, Becker A, Vatter H, Schwarz MK, Henneberger C, Steinhäuser C, Bedner P. Subcellular reorganization and altered phosphorylation of the astrocytic gap junction protein connexin43 in human and experimental temporal lobe epilepsy. Glia 2017; 65:1809-1820. [DOI: 10.1002/glia.23196] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 01/12/2023]
Affiliation(s)
- Tushar Deshpande
- Institute of Cellular Neurosciences, Medical Faculty; University of Bonn Germany
| | - Tingsong Li
- Institute of Cellular Neurosciences, Medical Faculty; University of Bonn Germany
- Department of Neurology; Children's Hospital, Chongqing Medical University; Chongqing China
| | - Michel K. Herde
- Institute of Cellular Neurosciences, Medical Faculty; University of Bonn Germany
| | - Albert Becker
- Department of Neuropathology; Medical Faculty, University of Bonn; Bonn Germany
| | - Hartmut Vatter
- Department of Neurosurgery; Medical Faculty, University of Bonn; Bonn Germany
| | - Martin K. Schwarz
- Department of Epileptology, Medical Faculty; University of Bonn; Bonn Germany
| | - Christian Henneberger
- Institute of Cellular Neurosciences, Medical Faculty; University of Bonn Germany
- Institute of Neurology, University College London; London UK
- German Center for Degenerative Diseases (DZNE); Bonn Germany
| | | | - Peter Bedner
- Institute of Cellular Neurosciences, Medical Faculty; University of Bonn Germany
| |
Collapse
|
283
|
Lee YC, Huang YJ, Hung MC, Hung SC, Hsiao CY, Cho HL, Lai LF, Tong SH, Wang JT. Risk factors associated with the development of seizures among adult patients treated with ertapenem: A matched case-control study. PLoS One 2017; 12:e0182046. [PMID: 28759588 PMCID: PMC5536326 DOI: 10.1371/journal.pone.0182046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 07/11/2017] [Indexed: 01/01/2023] Open
Abstract
Objective The purpose of this study is to compare the characteristics of those ertapenem-treated adult patients with and without development of seizures, and identify the associated factors for the development of seizures. Methods This retrospective study was conducted at Chia-Yi Christian Hospital from January 2012 to December 2014. Patients developing seizures during their ertapenem treatment course were identified as case patients. Those without seizures who had received ertapenem for at least five days were considered as the pool of control patients. For each case patient, four matched patients from the control pool were randomly selected as the final control group, based on age, gender, and the date of ertapenem prescription. Results A total of 1706 ertapenem-treated patients were identified, 33 (1.9%) individuals developed seizures with the enrollment of 132 matched control patients. Among these 33 patients, the average age was 79.3 ± 7.5 years, and 20 (60.6%) were male. The mean Charlson co-morbidity score was 4.5 ± 2.4, and the first episode of seizure happened 3.3 ± 2.6 days after receiving ertapenem. In multivariate logistic regression analysis, the independent predictors associated with the development of ertapenem-associated seizures were old stroke (OR, 14.36; 95% CI, 4.38–47.02; p < 0.0001), undergoing brain images within one year prior to the admission (OR, 5.73; 95% CI, 1.78–18.43; p = 0.0034), low hemoglobin level (OR, 3.88; 95% CI, 1.28–12.75; p = 0.0165) and low platelet count (OR, 4,94; 95% CI, 1.56–15.68; p = 0.0067) at presentations, and protective factors against the development of seizures were heart failure (OR, 0.04; 95% CI, 0.00–0.63; p = 0.0222), concomitant use of steroids (OR, 0.19; 95% CI, 0.05–0.77; p = 0.0201), or antiplatelet agents (OR, 0.12; 95% CI, 0.02–0.63, p = 0.0123) with ertapenem. Conclusions The development of ertapenem-associated seizures may occur more frequently and much earlier due to its widespread use in treating drug-resistant pathogens, especially when these pathogens emerged worldwide.Our study would help physician to estimate the risk of developing seizure among patients receiving ertapenem.
Collapse
Affiliation(s)
- Yi-Chien Lee
- Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, Taiwan
- Department of Sports Management, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Yun-Jhong Huang
- Department of Colorectal Surgery, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, Taiwan
| | - Miao-Chiu Hung
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Sheng-Che Hung
- Department of Radiology, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Yen Hsiao
- Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, Taiwan
- Department of Hospital and Health Care Administration, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Hui-Ling Cho
- Department of Nursing, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, Taiwan
| | - Li-Fen Lai
- Department of Nursing, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, Taiwan
| | - Show-Hwa Tong
- Department of Pharmacy, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, Taiwan
| | - Jann-Tay Wang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
284
|
Cerri C, Caleo M, Bozzi Y. Chemokines as new inflammatory players in the pathogenesis of epilepsy. Epilepsy Res 2017; 136:77-83. [PMID: 28780154 DOI: 10.1016/j.eplepsyres.2017.07.016] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/13/2017] [Accepted: 07/25/2017] [Indexed: 12/22/2022]
Abstract
A large series of clinical and experimental studies supports a link between inflammation and epilepsy, indicating that inflammatory processes within the brain are important contributors to seizure recurrence and precipitation. Systemic inflammation can precipitate seizures in children suffering from epileptic encephalopathies, and hallmarks of a chronic inflammatory state have been found in patients with temporal lobe epilepsy. Research performed on animal models of epilepsy further corroborates the idea that seizures upregulate inflammatory mediators, which in turn may enhance brain excitability and neuronal degeneration. Several inflammatory molecules and their signaling pathways have been implicated in epilepsy. Among these, the chemokine pathway has increasingly gained attention. Chemokines are small cytokines secreted by blood cells, which act as chemoattractants for leukocyte migration. Recent studies indicate that chemokines and their receptors are also produced by brain cells, and are involved in various neurological disorders including epilepsy. In this review, we will focus on a subset of pro-inflammatory chemokines (namely CCL2, CCL3, CCL5, CX3CL1) and their receptors, and their increasingly recognized role in seizure control.
Collapse
Affiliation(s)
- Chiara Cerri
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124, Pisa, Italy; Fondazione Umberto Veronesi, Piazza Velasca 5, 20122 Milano, Italy.
| | - Matteo Caleo
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124, Pisa, Italy.
| | - Yuri Bozzi
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124, Pisa, Italy; Neurodevelopmental Disorders Research Group, Centre for Mind/Brain Sciences, University of Trento, via Sommarive 9, 38123 Povo, Trento, Italy.
| |
Collapse
|
285
|
Kumar B, Modi M, Saikia B, Medhi B. Evaluation of Brain Pharmacokinetic and Neuropharmacodynamic Attributes of an Antiepileptic Drug, Lacosamide, in Hepatic and Renal Impairment: Preclinical Evidence. ACS Chem Neurosci 2017; 8:1589-1597. [PMID: 28324653 DOI: 10.1021/acschemneuro.7b00084] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The knowledge of pharmacokinetic and pharmacodynamic properties of antiepileptic drugs is helpful in optimizing drug therapy for epilepsy. This study was designed to evaluate the pharmacokinetic and pharmacodynamic properties of lacosamide in experimentally induced hepatic and renal impairment in seizure animals. Hepatic or renal impairment was induced by injection of carbon tetrachloride or diclofenac sodium, respectively. After induction, the animals were administered a single dose of lacosamide. At different time points, maximal electroshock (MES) seizure recordings were made followed by isolation of plasma and brain samples for drug quantification and pharmacodynamic measurements. Our results showed a significant increase in the area under the curve of lacosamide in hepatic and renal impairment groups. Reduced clearance of lacosamide was observed in animals with renal impairment. Along with pharmacokinetic alterations, the changes in pharmacodynamic effects of lacosamide were also observed in all the groups. Lacosamide showed a significant protection against MES-induced seizures, oxidative stress, and neuroinflammatory cytokines. These findings revealed that experimentally induced hepatic or renal impairment could alter the pharmacokinetic as well as pharmacodynamic properties of lacosamide. Hence, these conditions may affect the safety and efficacy of lacosamide.
Collapse
Affiliation(s)
- Baldeep Kumar
- Department
of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Manish Modi
- Department
of Neurology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Biman Saikia
- Department
of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Bikash Medhi
- Department
of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| |
Collapse
|
286
|
Mahfoz AM, Abdel-Wahab AF, Afify MA, Shahzad N, Ibrahim IAA, ElSawy NA, Bamagous GA, Al Ghamdi SS. Neuroprotective effects of vitamin D alone or in combination with lamotrigine against lithium-pilocarpine model of status epilepticus in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2017; 390:977-985. [PMID: 28687854 DOI: 10.1007/s00210-017-1400-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/28/2017] [Indexed: 10/19/2022]
Abstract
Status epilepticus (SE) is considered one of the major serious forms of epilepsy with high mortality rate. Since the currently available antiepileptic drugs have low efficacy and high adverse effects, new more efficient and safe therapies are critically needed. There is increasing evidence supporting dietary and alternative therapies for epilepsy, including the ketogenic diet, modified Atkins diet, and omega-3 fatty acids. Recent studies have shown significant prophylactic and therapeutic potential of vitamin D (vit-D) use in many neurological disorders. Therefore, in the present study, the neuroprotective effects and mechanisms of vit-D alone or in combination with lamotrigine have been evaluated in the lithium-pilocarpine model of SE in rats. Rats were divided into five groups: normal group, SE group, lamotrigine (25 mg/kg/day) pretreated group, vit-D (1.5 mcg/kg/day) pretreated group, and group pretreated with vit-D and lamotrigine for 2 weeks. At the end of treatment, SE was induced by single intraperitoneal injection of LiCl (127 mg/kg), followed 24 h later by pilocarpine (30 mg/kg). Seizures' latency, cognitive performance in Morris water maze, brain oxidative stress biomarkers (glutathione, lipid peroxides, and nitric oxide), brain neurochemistry (γ-aminobutyric acid and glutamate), and brain histopathology have been evaluated. Vit-D prevented pilocarpine-induced behavioral impairments and oxidative stress in the brain; these results were improved in combination with lamotrigine. Vit-D has a promising antiepileptic, neuroprotective, and antioxidant effects. It can be provided to patients as a supportive treatment besides antiepileptic drugs. However, clinical trials are needed to establish its efficacy and safety.
Collapse
Affiliation(s)
- Amal M Mahfoz
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Ali F Abdel-Wahab
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia.,Department of Clinical Pharmacology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed A Afify
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Naiyer Shahzad
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ibrahim A A Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Naser A ElSawy
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ghazi A Bamagous
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Saeed S Al Ghamdi
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
287
|
Leal B, Chaves J, Carvalho C, Bettencourt A, Brito C, Boleixa D, Freitas J, Brás S, Lopes J, Ramalheira J, Costa PP, da Silva BM, da Silva AM. Immunogenetic predisposing factors for mesial temporal lobe epilepsy with hippocampal sclerosis. Int J Neurosci 2017; 128:305-310. [PMID: 28675059 DOI: 10.1080/00207454.2017.1349122] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE Neuroinflammation appears as an important epileptogenic mechanism. Experimental and clinical studies have demonstrated an upregulation of pro-inflammatory cytokines such as IL-1β and TNF-α, in mesial temporal lobe epilepsy with hippocampal sclerosis (MTLE-HS). Expression of these cytokines can be modulated by polymorphisms such as rs16944 and rs1800629, respectively, both of which have been associated with febrile seizures (FS) and MTLE-HS development. The human leukocyte antigen (HLA) system has also been implicated in diverse epileptic entities, suggesting a variable role of this system in epilepsy. Our aim was to analyse the association between immunogenetic factors and MTLE-HS development. For that rs16944 (-511 T>C, IL-1β), rs1800629 (-308 G>A, TNF-α) polymorphisms and HLA-DRB1 locus were genotyped in a Portuguese Population. METHODS We studied 196 MTLE-HS patients (108 females, 88 males, 44.7 ± 12.0 years, age of onset = 13.6 ± 10.3 years, 104 with FS antecedents) and 282 healthy controls in a case-control study. RESULTS The frequency of rs16944 TT genotype was higher in MTLE-HS patients compared to controls (14.9% in MTLE-HS vs. 7.7% in controls, p = 0.021, OR [95% CI] = 2.20 [1.13-4.30]). This association was independent of FS antecedents. No association was observed between rs1800629 genotypes or HLA-DRB1 alleles and MTLE-HS susceptibility. Also, no correlation was observed between the studied polymorphisms and disease age of onset. CONCLUSION The rs16944 TT genotype is associated with MTLE-HS development what may be explained by the higher IL-1β levels produced by this genotype. High IL-1β levels may have neurotoxic effects or imbalance neurotransmission leading to seizures.
Collapse
Affiliation(s)
- Bárbara Leal
- a UMIB - Instituto de Ciências Biomédicas Abel Salazar [ICBAS] - Universidade do Porto - Rua Jorge Viterbo Ferreira , Porto , Portugal.,b Lab. Imunogenética - DPIM, ICBAS-UPorto - Rua Jorge Viterbo Ferreira , Porto , Portugal
| | - João Chaves
- c Serviço de Neurologia, Hospital de Santo António - Centro Hospitalar do Porto - Largo Prof. Abel Salazar , Porto , Portugal
| | - Cláudia Carvalho
- a UMIB - Instituto de Ciências Biomédicas Abel Salazar [ICBAS] - Universidade do Porto - Rua Jorge Viterbo Ferreira , Porto , Portugal.,b Lab. Imunogenética - DPIM, ICBAS-UPorto - Rua Jorge Viterbo Ferreira , Porto , Portugal
| | - Andreia Bettencourt
- a UMIB - Instituto de Ciências Biomédicas Abel Salazar [ICBAS] - Universidade do Porto - Rua Jorge Viterbo Ferreira , Porto , Portugal.,b Lab. Imunogenética - DPIM, ICBAS-UPorto - Rua Jorge Viterbo Ferreira , Porto , Portugal
| | - Cláudia Brito
- b Lab. Imunogenética - DPIM, ICBAS-UPorto - Rua Jorge Viterbo Ferreira , Porto , Portugal
| | - Daniela Boleixa
- b Lab. Imunogenética - DPIM, ICBAS-UPorto - Rua Jorge Viterbo Ferreira , Porto , Portugal
| | - Joel Freitas
- c Serviço de Neurologia, Hospital de Santo António - Centro Hospitalar do Porto - Largo Prof. Abel Salazar , Porto , Portugal
| | - Sandra Brás
- b Lab. Imunogenética - DPIM, ICBAS-UPorto - Rua Jorge Viterbo Ferreira , Porto , Portugal
| | - João Lopes
- d Serviço de Neurofisiologia, Hospital de Santo António - Centro Hospitalar do Porto - Largo Prof. Abel Salazar , Porto , Portugal
| | - João Ramalheira
- d Serviço de Neurofisiologia, Hospital de Santo António - Centro Hospitalar do Porto - Largo Prof. Abel Salazar , Porto , Portugal
| | - Paulo P Costa
- a UMIB - Instituto de Ciências Biomédicas Abel Salazar [ICBAS] - Universidade do Porto - Rua Jorge Viterbo Ferreira , Porto , Portugal.,e Departamento de Genética , Instituto Nacional de Saúde Dr. Ricardo Jorge - Porto , Porto , Portugal
| | - Berta Martins da Silva
- a UMIB - Instituto de Ciências Biomédicas Abel Salazar [ICBAS] - Universidade do Porto - Rua Jorge Viterbo Ferreira , Porto , Portugal.,b Lab. Imunogenética - DPIM, ICBAS-UPorto - Rua Jorge Viterbo Ferreira , Porto , Portugal
| | - António Martins da Silva
- a UMIB - Instituto de Ciências Biomédicas Abel Salazar [ICBAS] - Universidade do Porto - Rua Jorge Viterbo Ferreira , Porto , Portugal.,d Serviço de Neurofisiologia, Hospital de Santo António - Centro Hospitalar do Porto - Largo Prof. Abel Salazar , Porto , Portugal
| |
Collapse
|
288
|
Abstract
BACKGROUND Epilepsy is a common neurological condition, with an estimated incidence of 50 per 100,000 persons. People with epilepsy may present with various types of immunological abnormalities, such as low serum immunoglobulin A (IgA) levels, lack of the immunoglobulin G (IgG) subclass and identification of certain types of antibodies. Intravenous immunoglobulin (IVIg) treatment may represent a valuable approach and its efficacy has important implications for epilepsy management. This is an updated version of the original Cochrane review published in Issue 1, 2011. OBJECTIVES To examine the effects of IVIg on the frequency and duration of seizures, quality of life and adverse effects when used as monotherapy or as add-on treatment for people with epilepsy. SEARCH METHODS For the latest update, we searched the Cochrane Epilepsy Group Specialized Register (2 February 2017), the Cochrane Central Register of Controlled Trials (CENTRAL) via the Cochrane Register of Studies Online (2 February 2017), MEDLINE (Ovid, 1946 to 2 February 2017), Web of Science (1898 to 2 February 2017), ISRCTN registry (2 February 2017), WHO International Clinical Trials Registry Platform (ICTRP, 2 February 2017), the US National Institutes of Health ClinicalTrials.gov (2 February 2017), and reference lists of articles. SELECTION CRITERIA Randomized or quasi-randomized controlled trials of IVIg as monotherapy or add-on treatment in people with epilepsy. DATA COLLECTION AND ANALYSIS Two review authors independently assessed the trials for inclusion and extracted data. We contacted study authors for additional information. Outcomes included percentage of people rendered seizure-free, 50% or greater reduction in seizure frequency, adverse effects, treatment withdrawal and quality of life. MAIN RESULTS We included one study (61 participants). The included study was a randomized, double-blind, placebo-controlled, multi-centre trial which compared the treatment efficacy of IVIg as an add-on with a placebo add-on in patients with refractory epilepsy. There was no significant difference between IVIg and placebo in 50% or greater reduction in seizure frequency. The study reported a statistically significant effect for global assessment in favour of IVIg. No adverse effects were demonstrated. We found no randomized controlled trials that investigated the effects of IVIg monotherapy for epilepsy. Overall, the included study was rated as low/unclear risk of bias. Using GRADE methodology, the quality of the evidence was rated as low. AUTHORS' CONCLUSIONS We cannot draw any reliable conclusions regarding the efficacy of IVIg as a treatment for epilepsy. Further randomized controlled trials are needed.
Collapse
Affiliation(s)
- JinSong Geng
- Medical School of Nantong UniversityEvidence‐based Medicine Center19 Qixiu Road, Medical School of NantongNantongChina226001
| | - JianCheng Dong
- Medical School of Nantong UniversityEvidence‐based Medicine Center19 Qixiu Road, Medical School of NantongNantongChina226001
| | - Youping Li
- West China Hospital, Sichuan UniversityChinese Cochrane Centre, Chinese Evidence‐Based Medicine CentreNo. 37, Guo Xue XiangChengduChina610041
| | - Hengjian Ni
- Medical School of Nantong UniversityEvidence‐based Medicine Center19 Qixiu Road, Medical School of NantongNantongChina226001
| | - Kui Jiang
- Medical School of Nantong UniversityEvidence‐based Medicine Center19 Qixiu Road, Medical School of NantongNantongChina226001
| | - Li Li Shi
- Evidence‐based Medicine Center, Medical School of Nantong UniversityNantongChina
| | - GuoHua Wang
- Nantong UniversityInstitute of Nautical Medicine19 Qixiu RoadNantongChina226001
| |
Collapse
|
289
|
Broekaart DWM, van Scheppingen J, Geijtenbeek KW, Zuidberg MRJ, Anink JJ, Baayen JC, Mühlebner A, Aronica E, Gorter JA, van Vliet EA. Increased expression of (immuno)proteasome subunits during epileptogenesis is attenuated by inhibition of the mammalian target of rapamycin pathway. Epilepsia 2017. [DOI: 10.1111/epi.13823] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Diede W. M. Broekaart
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Jackelien van Scheppingen
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Karlijne W. Geijtenbeek
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Mark R. J. Zuidberg
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Jasper J. Anink
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Johannes C. Baayen
- Department of Neurosurgery; VU University Medical Center; Vrije Universiteit; Amsterdam The Netherlands
| | - Angelika Mühlebner
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
- Swammerdam Institute for Life Sciences; Center for Neuroscience; University of Amsterdam; Amsterdam The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN); Heemstede The Netherlands
| | - Jan A. Gorter
- Swammerdam Institute for Life Sciences; Center for Neuroscience; University of Amsterdam; Amsterdam The Netherlands
| | - Erwin A. van Vliet
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| |
Collapse
|
290
|
Tohidpour A, Morgun AV, Boitsova EB, Malinovskaya NA, Martynova GP, Khilazheva ED, Kopylevich NV, Gertsog GE, Salmina AB. Neuroinflammation and Infection: Molecular Mechanisms Associated with Dysfunction of Neurovascular Unit. Front Cell Infect Microbiol 2017; 7:276. [PMID: 28676848 PMCID: PMC5476750 DOI: 10.3389/fcimb.2017.00276] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 06/06/2017] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation is a complex inflammatory process in the central nervous system, which is sought to play an important defensive role against various pathogens, toxins or factors that induce neurodegeneration. The onset of neurodegenerative diseases and various microbial infections are counted as stimuli that can challenge the host immune system and trigger the development of neuroinflammation. The homeostatic nature of neuroinflammation is essential to maintain the neuroplasticity. Neuroinflammation is regulated by the activity of neuronal, glial, and endothelial cells within the neurovascular unit, which serves as a “platform” for the coordinated action of pro- and anti-inflammatory mechanisms. Production of inflammatory mediators (cytokines, chemokines, reactive oxygen species) by brain resident cells or cells migrating from the peripheral blood, results in the impairment of blood-brain barrier integrity, thereby further affecting the course of local inflammation. In this review, we analyzed the most recent data on the central nervous system inflammation and focused on major mechanisms of neurovascular unit dysfunction caused by neuroinflammation and infections.
Collapse
Affiliation(s)
- Abolghasem Tohidpour
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| | - Andrey V Morgun
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia.,Department of Paediatrics, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| | - Elizaveta B Boitsova
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia.,Department of Children Infectious Diseases, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| | - Natalia A Malinovskaya
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| | - Galina P Martynova
- Department of Children Infectious Diseases, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| | - Elena D Khilazheva
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| | - Natalia V Kopylevich
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| | - Galina E Gertsog
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| | - Alla B Salmina
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| |
Collapse
|
291
|
Cyclooxygenase-2 inhibitors differentially attenuate pentylenetetrazol-induced seizures and increase of pro- and anti-inflammatory cytokine levels in the cerebral cortex and hippocampus of mice. Eur J Pharmacol 2017; 810:15-25. [PMID: 28583427 DOI: 10.1016/j.ejphar.2017.05.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 04/24/2017] [Accepted: 05/08/2017] [Indexed: 12/31/2022]
Abstract
Seizures increase prostaglandin and cytokine levels in the brain. However, it remains to be determined whether cyclooxygenase-2 (COX-2) derived metabolites play a role in seizure-induced cytokine increase in the brain and whether anticonvulsant activity is shared by all COX-2 inhibitors. In this study we investigated whether three different COX-2 inhibitors alter pentylenetetrazol (PTZ)-induced seizures and increase of interleukin-1β (IL-1β), interleukin-6 (IL-6), interferon-γ (INF-γ), tumor necrosis factor-α (TNF-α) and interleukin-10 (IL-10) levels in the hippocampus and cerebral cortex of mice. Adult male albino Swiss mice received nimesulide, celecoxib or etoricoxib (0.2, 2 or 20mg/kg in 0.1% carboxymethylcellulose (CMC) in 5% Tween 80, p.o.). Sixty minutes thereafter the animals were injected with PTZ (50mg/kg, i.p.) and the latency to myoclonic jerks and to generalized tonic-clonic seizures were recorded. Twenty minutes after PTZ injection animals were killed and cytokine levels were measured. PTZ increased cytokine levels in the cerebral cortex and hippocampus. While celecoxib and nimesulide attenuated PTZ -induced increase of proinflammatory cytokines in the cerebral cortex, etoricoxib did not. Nimesulide was the only COX-2 inhibitors that attenuated PTZ-induced seizures. This effect coincided with an increase of IL-10 levels in the cerebral cortex and hippocampus, constituting circumstantial evidence that IL-10 increase may be involved in the anticonvulsant effect of nimesulide.
Collapse
|
292
|
Association between NLPR1, NLPR3, and P2X7R Gene Polymorphisms with Partial Seizures. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9547902. [PMID: 28503575 PMCID: PMC5414494 DOI: 10.1155/2017/9547902] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 04/06/2017] [Indexed: 11/17/2022]
Abstract
Objectives. Clinical and experimental evidence has clarified that the inflammatory processes within the brain play a pivotal role in the pathophysiology of seizures and epilepsy. Inflammasomes and P2X7 purinergic receptor (P2X7R) are important mediators during the inflammatory process. Therefore, we investigated the possible association between partial seizures and inflammasomes NLPR1, NLRP3, and P2X7R gene polymorphisms in the present study. Method. A total of 163 patients and 201 health controls were enrolled in this study and polymorphisms of NLPR1, NLRP3, and P2X7R genes were detected using polymerase chain reaction- (PCR-) ligase detection reaction method. Result. The frequency of rs878329 (G>C) genotype with C (CG + CC) was significantly lower among patients with partial seizures relative to controls (OR = 2.033, 95% CI = 1.290-3.204, p = 0.002 for GC + CC versus GG). Intriguingly, we found that the significant difference of rs878329 (G>C) genotype and allele frequency only existed among males (OR = 2.542, 95% CI = 1.344-4.810, p = 0.004 for GC + CC versus GG), while there was no statistically significant difference among females. However, no significant results were presented for the genotype distributions of rs8079034, rs4612666, rs10754558, rs2027432, rs3751143, and rs208294 polymorphisms between patients and controls. Conclusion. Our study demonstrated the potentially significant role of NLRP1 rs878329 (G>C) in developing susceptibility to the partial seizures in a Chinese Han population.
Collapse
|
293
|
Wang FX, Xiong XY, Zhong Q, Meng ZY, Yang H, Yang QW. Foxp3 exhibits antiepileptic effects in ictogenesis involved in TLR4 signaling. FASEB J 2017; 31:2948-2962. [PMID: 28386044 DOI: 10.1096/fj.201600989r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 03/13/2017] [Indexed: 11/11/2022]
Abstract
Inflammatory processes play critical roles in epileptogenesis, but the exact mechanisms that underlie these processes are still not completely understood. In this study, we investigated the role of forkhead transcription factor 3 (Foxp3), a transcription factor that is involved in T-cell differentiation, in epileptogenesis. In both human epileptic tissues and experimental seizure models, we found significant up-regulation of Foxp3 in neurons and glial cells. Of importance, Foxp3-/- mice were susceptible to kainic acid-induced seizures, whereas overexpression of Foxp3 reduced acute seizure occurrence and decreased chronic seizure recurrence. In addition, in vitro experiments revealed that Foxp3 inhibited neuronal excitability via glial cells and not neurons. The protective effects of Foxp3 were manifested as a reduction in glial cell activation and proinflammatory cytokine production and increased neuronal survival. Moreover, we showed that beneficial effects of Foxp3 involved the attenuation of TLR4 signaling and inflammation, which led to the inactivation of NR2B-containing NMDA receptors. These results suggest that Foxp3 in glial cells may play an antiepileptic role in epileptogenesis and may act as a modulator of TLR4. Taken together, our results indicate that Foxp3 may represent a novel therapeutic target for achieving anticonvulsant effects in patients with epilepsy that is currently resistant to drugs.-Wang, F.-X., Xiong, X.-Y., Zhong, Q., Meng, Z.-Y., Yang, H., Yang, Q.-W. Foxp3 exhibits antiepileptic effects in ictogenesis involved in TLR4 signaling.
Collapse
Affiliation(s)
- Fa-Xiang Wang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xiao-Yi Xiong
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Qi Zhong
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Zhao-You Meng
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Hui Yang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Chongqing, China;
| |
Collapse
|
294
|
Ghaemi A, Alizadeh L, Babaei S, Jafarian M, Khaleghi Ghadiri M, Meuth SG, Kovac S, Gorji A. Astrocyte-mediated inflammation in cortical spreading depression. Cephalalgia 2017; 38:626-638. [PMID: 28372497 DOI: 10.1177/0333102417702132] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Background Cortical spreading depression (CSD) related diseases such as migraine, cerebrovascular diseases, and epilepsy have been associated with reactive astrocytosis, yet the mechanisms of these tissue changes remain unclear. CSD-induced inflammatory response has been proposed to play a role in some neurological disorders and thus may also contribute to reactive astrocytosis. Methods Using ex vivo brain slices and in vitro astrocytic cultures, we aimed to characterize CSD related changes in astrocytes and markers of inflammation by immunocyto- and immunohistochemistry. CSD was induced by application of KCl (3 mol/l) on neocortical tissues. The application of KCl was repeated weekly over the course of four weeks. Results CSD induced an increase in the mean number and volume of astrocytes in rat brain tissue when compared to controls, whereas no changes in neuronal numbers and volumes were seen. These cell-type specific changes, suggestive of reactive astrocytosis, were paralleled by an increased expression of protein markers indicative of astrocytes and neuroinflammation in ex vivo brain slices of animals undergoing CSD when compared to sham-treated controls. Cultured astrocytes showed an increased expression of the immune modulatory enzyme indoleamine 2,3-dioxygenase and an elevated expression of the pro-inflammatory markers, IL-6, IL-1β, and TNFα in addition to increased levels of toll like receptors (TLR3 and TLR4) and astrocytic markers after induction of CSD. Conclusion These findings indicate that CSD related reactive astrocytosis is linked to an upregulation of inflammatory markers. Targeting inflammation with already approved and available immunomodulatory treatments may thus represent a strategy to combat or ameliorate CSD-related disease.
Collapse
Affiliation(s)
- Amir Ghaemi
- 1 Shefa Neuroscience Research Center, Khatam-Alanbia Hospital, Tehran, Iran.,2 Department of Virology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Leila Alizadeh
- 1 Shefa Neuroscience Research Center, Khatam-Alanbia Hospital, Tehran, Iran
| | - Shahnaz Babaei
- 1 Shefa Neuroscience Research Center, Khatam-Alanbia Hospital, Tehran, Iran
| | - Maryam Jafarian
- 1 Shefa Neuroscience Research Center, Khatam-Alanbia Hospital, Tehran, Iran
| | | | - Sven G Meuth
- 4 Department of Neurology, WestfälischeWilhelms-Universität Münster, Germany
| | - Stjepana Kovac
- 4 Department of Neurology, WestfälischeWilhelms-Universität Münster, Germany
| | - Ali Gorji
- 1 Shefa Neuroscience Research Center, Khatam-Alanbia Hospital, Tehran, Iran.,3 Department of Neurosurgery, WestfälischeWilhelms-Universität Münster, Germany.,4 Department of Neurology, WestfälischeWilhelms-Universität Münster, Germany.,5 Department of Neuroscience, Mashhad University of Medical Sciences, Mashhad, Iran.,6 Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Germany
| |
Collapse
|
295
|
Abd-Elghafour BA, El-Sayed NM, Ahmed AA, Zaitone SA, Moustafa YM. Aspirin and (or) omega-3 polyunsaturated fatty acids protect against corticohippocampal neurodegeneration and downregulate lipoxin A4 production and formyl peptide receptor-like 1 expression in pentylenetetrazole-kindled rats. Can J Physiol Pharmacol 2017; 95:340-348. [DOI: 10.1139/cjpp-2016-0060] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
There is evidence for a relationship between inflammation and seizures because epilepsy can be caused by or result in inflammation. This study aimed to investigate the effect of aspirin and (or) omega-3 polyunsaturated fatty acids (PUFAs) on seizure activity and neurodegeneration in pentylenetetrazole (PTZ)-kindled rats focusing on their effect on corticohippocampal production of lipoxin A4 (LXA4) and expression of formyl peptide receptor-like 1 (FPRL1) receptors. Male rats were injected with PTZ (35 mg/kg, i.p.) 3 times per week for a total of 15 doses. Rats were treated daily with aspirin (20 mg/kg, i.p.), omega-3 PUFAs (85 mg/kg, p.o.), or a combination of them for 35 days. Both LXA4 level and expression of FPRL1 receptor in the cortices and hippocampi of rats’ brains were greater in PTZ-kindled rats compared to a saline control group. Cotreatment with aspirin and (or) omega-3 PUFAs reduced convulsive behaviour; reduced levels of LXA4, interleukin-1β, and nuclear factor-κB; and showed a lower percentage of corticohippocampal degenerative cells compared to PTZ-kindled rats. The combination of the 2 therapeutic agents did not provide significant improvement in comparison with the monotherapies. These findings suggest the use of aspirin or omega-3 PUFAs may delay the development of seizures and provide neuroprotection in a clinical setting.
Collapse
Affiliation(s)
| | - Norhan M. El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Amal A.M. Ahmed
- Department of Cytology & Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Sawsan A. Zaitone
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Yasser M. Moustafa
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
296
|
Systemic inflammation enhances stimulant-induced striatal dopamine elevation. Transl Psychiatry 2017; 7:e1076. [PMID: 28350401 PMCID: PMC5404612 DOI: 10.1038/tp.2017.18] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 12/10/2016] [Accepted: 01/09/2017] [Indexed: 12/25/2022] Open
Abstract
Changes in the mesolimbic dopamine (DA) system are implicated in a range of neuropsychiatric conditions including addiction, depression and schizophrenia. Dysfunction of the neuroimmune system is often comorbid with such conditions and affects similar areas of the brain. The goal of this study was to use positron emission tomography with the dopamine D2 antagonist tracer, 11C-raclopride, to explore the effect of acute immune activation on striatal DA levels. DA transmission was modulated by an oral methylphenidate (MP) challenge in order to reliably elicit DA elevation. Elevation in DA concentration due to MP was estimated via change in 11C-raclopride binding potential from the baseline scan. Prior to the post-MP scan, subjects were pre-treated with either the immune activator lipopolysaccharide (LPS) or placebo (PBO) in a cross-over design. Immune activation was confirmed by measuring tumor necrosis factor alpha (TNFα), interleukin (IL)-6 and IL-8 concentration in plasma. Eight healthy subjects were scanned four times each to determine the MP-induced DA elevation under both LPS and PBO pre-treatment conditions. MP-induced DA elevation in the striatum was significantly greater (P<0.01) after LPS pre-treatment compared to PBO pre-treatment. Seven of eight subjects responded similarly. This effect was observed in the caudate and putamen (P<0.02), but was not present in ventral striatum. DA elevation induced by MP was significantly greater when subjects were pre-treated with LPS compared to PBO. The amplification of stimulant-induced DA signaling in the presence of systemic inflammation may have important implications for our understanding of addiction and other diseases of DA dysfunction.
Collapse
|
297
|
Gaikwad S, Patel D, Agrawal-Rajput R. CD40 Negatively Regulates ATP-TLR4-Activated Inflammasome in Microglia. Cell Mol Neurobiol 2017; 37:351-359. [PMID: 26961545 PMCID: PMC11482137 DOI: 10.1007/s10571-016-0358-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 02/29/2016] [Indexed: 01/01/2023]
Abstract
During acute brain injury and/or sterile inflammation, release of danger-associated molecular patterns (DAMPs) activates pattern recognition receptors (PRRs). Microglial toll-like receptor (TLR)-4 activated by DAMPs potentiates neuroinflammation through inflammasome-induced IL-1β and pathogenic Th17 polarization which critically influences brain injury. TLR4 activation accompanies increased CD40, a cognate costimulatory molecule, involved in microglia-mediated immune responses in the brain. During brain injury, excessive release of extracellular ATP (DAMPs) is involved in promoting the damage. However, the regulatory role of CD40 in microglia during ATP-TLR4-mediated inflammasome activation has never been explored. We report that CD40, in the absence of ATP, synergizes TLR4-induced proinflammatory cytokines but not IL-1β, suggesting that the response is independent of inflammasome. The presence of ATP during TLR4 activation leads to NLRP3 inflammasome activation and caspase-1-mediated IL-1β secretion which was inhibited during CD40 activation, accompanied with inhibition of ERK1/2 and reactive oxygen species (ROS), and elevation in p38 MAPK phosphorylation. Experiments using selective inhibitors prove indispensability of ERK 1/2 and ROS for inflammasome activation. The ATP-TLR4-primed macrophages polarize the immune response toward pathogenic Th17 cells, whereas CD40 activation mediates Th1 response. Exogenous supplementation of IFN-γ (a Th1 cytokine and CD40 inducer) results in decreased IL-1β, suggesting possible feedback loop mechanism of inflammasome inhibition, whereby IFN-γ-mediated increase in CD40 expression and activation suppress neurotoxic inflammasome activation required for Th17 response. Collectively, the findings indicate that CD40 is a novel negative regulator of ATP-TLR4-mediated inflammasome activation in microglia, thus providing a checkpoint to regulate excessive inflammasome activation and Th17 response during DAMP-mediated brain injury.
Collapse
Affiliation(s)
- Sagar Gaikwad
- Department of Immunology, School of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382 007, India
| | - Divyesh Patel
- Department of Immunology, School of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382 007, India
| | - Reena Agrawal-Rajput
- Department of Immunology, School of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382 007, India.
| |
Collapse
|
298
|
Scorza CA, Marques MJG, Gomes da Silva S, Naffah-Mazzacoratti MDG, Scorza FA, Cavalheiro EA. Status epilepticus does not induce acute brain inflammatory response in the Amazon rodent Proechimys, an animal model resistant to epileptogenesis. Neurosci Lett 2017; 668:169-173. [PMID: 28235602 DOI: 10.1016/j.neulet.2017.02.049] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 01/31/2017] [Accepted: 02/20/2017] [Indexed: 12/23/2022]
Abstract
Mesial temporal lobe epilepsy is a serious brain disorder in adults that is often preceded by an initial brain insult, such as status epilepticus (SE), that after a latent period leads to recurrent seizures. Post-SE models are widely used for studies on epileptogenic processes. Previous findings of our laboratory suggested that the Neotropical rodents Proechimys exhibit endogenous antiepileptogenic mechanisms in post-SE models. Strong body of research supports that SE triggers a rapid and dramatic upregulation of inflammatory mediators and vascular endothelial growth factor (VEGF). In this work we found that, in the epilepsy-resistant Proechimys, hippocampal and cortical levels of inflammatory cytokines (IL-1β, IL-6, IL-10, TNF-α) and VEGF remained unchanged 24h after SE, strongly contrasting to the high levels of post-SE changes observed in Wistar rats. Furthermore, substantial differences in the brain baseline levels of these proteins were encountered between animal species studied. Since inflammatory cytokines and VEGF have been recognized as major orchestrators of the epileptogenic process, our results suggest their role in the antiepileptogenic mechanisms previously described in Proechimys.
Collapse
Affiliation(s)
- Carla A Scorza
- Disciplina de Neurociência, Escola Paulista de Medicina/Universidade Federal de São Paulo, Brazil.
| | - Marcia J G Marques
- Disciplina de Neurociência, Escola Paulista de Medicina/Universidade Federal de São Paulo, Brazil
| | - Sérgio Gomes da Silva
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil; Núcleo de Pesquisas Tecnológicas, Universidade de Mogi das Cruzes, Mogi das Cruzes, SP, Brazil.
| | | | - Fulvio A Scorza
- Disciplina de Neurociência, Escola Paulista de Medicina/Universidade Federal de São Paulo, Brazil
| | - Esper A Cavalheiro
- Disciplina de Neurociência, Escola Paulista de Medicina/Universidade Federal de São Paulo, Brazil
| |
Collapse
|
299
|
McElroy PB, Sri Hari A, Day BJ, Patel M. Post-translational Activation of Glutamate Cysteine Ligase with Dimercaprol: A NOVEL MECHANISM OF INHIBITING NEUROINFLAMMATION IN VITRO. J Biol Chem 2017; 292:5532-5545. [PMID: 28202547 DOI: 10.1074/jbc.m116.723700] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 01/25/2017] [Indexed: 11/06/2022] Open
Abstract
Neuroinflammation and oxidative stress are hallmarks of various neurological diseases. However, whether and how the redox processes control neuroinflammation is incompletely understood. We hypothesized that increasing cellular glutathione (GSH) levels would inhibit neuroinflammation. A series of thiol compounds were identified to elevate cellular GSH levels by a novel approach (i.e. post-translational activation of glutamate cysteine ligase (GCL), the rate-limiting enzyme in GSH biosynthesis). These small thiol-containing compounds were examined for their ability to increase intracellular GSH levels in a murine microglial cell line (BV2), of which dimercaprol (2,3-dimercapto-1-propanol (DMP)) was found to be the most effective compound. DMP increased GCL activity and decreased LPS-induced production of pro-inflammatory cytokines and inducible nitric-oxide synthase induction in BV2 cells in a concentration-dependent manner. The ability of DMP to elevate GSH levels and attenuate LPS-induced pro-inflammatory cytokine production was inhibited by buthionine sulfoximine, an inhibitor of GCL. DMP increased the expression of GCL holoenzyme without altering the expression of its subunits or Nrf2 target proteins (NQO1 and HO-1), suggesting a post-translational mechanism. DMP attenuated LPS-induced MAPK activation in BV2 cells, suggesting the MAPK pathway as the signaling mechanism underlying the effect of DMP. Finally, the ability of DMP to increase GSH via GCL activation was observed in mixed cerebrocortical cultures and N27 dopaminergic cells. Together, the data demonstrate a novel mechanism of GSH elevation by post-translational activation of GCL. Post-translational activation of GCL offers a novel targeted approach to control inflammation in chronic neuronal disorders associated with impaired adaptive responses.
Collapse
Affiliation(s)
- Pallavi B McElroy
- From the Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045 and
| | - Ashwini Sri Hari
- From the Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045 and
| | - Brian J Day
- the Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - Manisha Patel
- From the Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045 and
| |
Collapse
|
300
|
Abstract
OBJECTIVE Epilepsy is a chronic neurological disease characterised with seizures. The aetiology of the most generalised epilepsies cannot be explicitly determined and the seizures are pronounced to be genetically determined by disturbances of receptors in central nervous system. Besides, neurotransmitter distributions or other metabolic problems are supposed to involve in epileptogenesis. Lack of adequate data about pharmacological agents that have antiepileptogenic effects point to need of research on this field. Thus, in this review, inflammatory aspects of epileptogenesis has been focussed via considering several concepts like role of immune system, blood-brain barrier and antibody involvement in epileptogenesis. METHODS We conducted an evidence-based review of the literatures in order to evaluate the possible participation of inflammatory processes to epileptogenesis and also, promising agents which are effective to these processes. We searched PubMed database up to November 2015 with no date restrictions. RESULTS In the present review, 163 appropriate articles were included. Obtained data suggests that inflammatory processes participate to epileptogenesis in several ways like affecting fibroblast growth factor-2 and tropomyosin receptor kinase B signalling pathways, detrimental proinflammatory pathways [such as the interleukin-1 beta (IL-1β)-interleukin-1 receptor type 1 (IL-1R1) system], mammalian target of rapamycin pathway, microglial activities, release of glial inflammatory proteins (such as macrophage inflammatory protein, interleukin 6, C-C motif ligand 2 and IL-1β), adhesion molecules that are suggested to function in signalling pathways between neurons and microglia and also linkage between these molecules and proinflammatory cytokines. CONCLUSION The literature research indicated that inflammation is a part of epileptogenesis. For this reason, further studies are necessary for assessing agents that will be effective in clinical use for therapeutic treatment of epileptogenesis.
Collapse
|