1
|
Riva A, Sahin E, Volpedo G, Petretto A, Lavarello C, Di Sapia R, Barbarossa D, Zaniani NR, Craparotta I, Barbera MC, Sezerman U, Vezzani A, Striano P, Ravizza T. Identification of an epilepsy-linked gut microbiota signature in a pediatric rat model of acquired epilepsy. Neurobiol Dis 2024; 194:106469. [PMID: 38485093 DOI: 10.1016/j.nbd.2024.106469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 03/21/2024] Open
Abstract
A dysfunctional gut microbiota-brain axis is emerging as a potential pathogenic mechanism in epilepsy, particularly in pediatric forms of epilepsy. To add new insights into gut-related changes in acquired epilepsy that develops early in life, we used a multi-omics approach in a rat model with a 56% incidence of epilepsy. The presence of spontaneous seizures was assessed in adult rats (n = 46) 5 months after status epilepticus induced by intra-amygdala kainate at postnatal day 13, by 2 weeks (24/7) ECoG monitoring. Twenty-six rats developed epilepsy (Epi) while the remaining 20 rats (No-Epi) did not show spontaneous seizures. At the end of ECoG monitoring, all rats and their sham controls (n = 20) were sacrificed for quantitative histopathological and immunohistochemical analyses of the gut structure, glia and macrophages, as well as RTqPCR analysis of inflammation/oxidative stress markers. By comparing Epi, No-Epi rats, and sham controls, we found structural, cellular, and molecular alterations reflecting a dysfunctional gut, which were specifically associated with epilepsy. In particular, the villus height-to-crypt depth ratio and number of Goblet cells were reduced in the duodenum of Epi rats vs both No-Epi rats and sham controls (p < 0.01). Villus height and crypt depth in the duodenum and jejunum (p < 0.01) were increased in No-Epi vs both Epi and sham controls. We also detected enhanced Iba1-positive macrophages, together with increased IL1b and NFE2L2 transcripts and TNF protein, in the small intestine of Epi vs both No-Epi and sham control rats (p < 0.01), denoting the presence of inflammation and oxidative stress. Astroglial GFAP-immunostaining was similar in all experimental groups. Metagenomic analysis in the feces collected 5 months after status epilepticus showed that the ratio of two dominant phyla (Bacteroidota-to-Firmicutes) was similarly increased in Epi and No-Epi rats vs sham control rats. Notably, the relative abundance of families, genera, and species associated with SCFA production differed in Epi vs No-Epi rats, describing a bacterial imprint associated with epilepsy. Furthermore, Epi rats showed a blood metabolic signature characterized by changes in lipid metabolism compared to both No-Epi and sham control rats. Our study provides new evidence of long-term gut alterations, along with microbiota-related metabolic changes, occurring specifically in rats that develop epilepsy after brain injury early in life.
Collapse
Affiliation(s)
- Antonella Riva
- IRCCS Istituto Giannina Gaslini, Genova, Italy; Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Università degli Studi di Genova, Genova, Italy
| | - Eray Sahin
- Acıbadem University, Faculty of Medicine, Department of Biostatistics and Medical Informatics, Istanbul, Turkey
| | - Greta Volpedo
- IRCCS Istituto Giannina Gaslini, Genova, Italy; Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Università degli Studi di Genova, Genova, Italy
| | | | | | - Rossella Di Sapia
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Davide Barbarossa
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Nasibeh Riahi Zaniani
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Ilaria Craparotta
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Maria Chiara Barbera
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Uğur Sezerman
- Acıbadem University, Faculty of Medicine, Department of Biostatistics and Medical Informatics, Istanbul, Turkey
| | - Annamaria Vezzani
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Pasquale Striano
- IRCCS Istituto Giannina Gaslini, Genova, Italy; Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Università degli Studi di Genova, Genova, Italy
| | - Teresa Ravizza
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy.
| |
Collapse
|
2
|
Charalambous M, Muñana K, Patterson EE, Platt SR, Volk HA. ACVIM Consensus Statement on the management of status epilepticus and cluster seizures in dogs and cats. J Vet Intern Med 2024; 38:19-40. [PMID: 37921621 PMCID: PMC10800221 DOI: 10.1111/jvim.16928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 10/19/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Seizure emergencies (ie, status epilepticus [SE] and cluster seizures [CS]), are common challenging disorders with complex pathophysiology, rapidly progressive drug-resistant and self-sustaining character, and high morbidity and mortality. Current treatment approaches are characterized by considerable variations, but official guidelines are lacking. OBJECTIVES To establish evidence-based guidelines and an agreement among board-certified specialists for the appropriate management of SE and CS in dogs and cats. ANIMALS None. MATERIALS AND METHODS A panel of 5 specialists was formed to assess and summarize evidence in the peer-reviewed literature with the aim to establish consensus clinical recommendations. Evidence from veterinary pharmacokinetic studies, basic research, and human medicine also was used to support the panel's recommendations, especially for the interventions where veterinary clinical evidence was lacking. RESULTS The majority of the evidence was on the first-line management (ie, benzodiazepines and their various administration routes) in both species. Overall, there was less evidence available on the management of emergency seizure disorders in cats in contrast to dogs. Most recommendations made by the panel were supported by a combination of a moderate level of veterinary clinical evidence and pharmacokinetic data as well as studies in humans and basic research studies. CONCLUSIONS AND CLINICAL RELEVANCE Successful management of seizure emergencies should include an early, rapid, and stage-based treatment approach consisting of interventions with moderate to preferably high ACVIM recommendations; management of complications and underlying causes related to seizure emergencies should accompany antiseizure medications.
Collapse
Affiliation(s)
| | - Karen Muñana
- North Carolina State UniversityRaleighNorth CarolinaUSA
| | | | | | - Holger A. Volk
- University of Veterinary Medicine HannoverHannoverGermany
| |
Collapse
|
3
|
Schaper FLWVJ, Nordberg J, Cohen AL, Lin C, Hsu J, Horn A, Ferguson MA, Siddiqi SH, Drew W, Soussand L, Winkler AM, Simó M, Bruna J, Rheims S, Guenot M, Bucci M, Nummenmaa L, Staals J, Colon AJ, Ackermans L, Bubrick EJ, Peters JM, Wu O, Rost NS, Grafman J, Blumenfeld H, Temel Y, Rouhl RPW, Joutsa J, Fox MD. Mapping Lesion-Related Epilepsy to a Human Brain Network. JAMA Neurol 2023; 80:891-902. [PMID: 37399040 PMCID: PMC10318550 DOI: 10.1001/jamaneurol.2023.1988] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/03/2023] [Indexed: 07/04/2023]
Abstract
Importance It remains unclear why lesions in some locations cause epilepsy while others do not. Identifying the brain regions or networks associated with epilepsy by mapping these lesions could inform prognosis and guide interventions. Objective To assess whether lesion locations associated with epilepsy map to specific brain regions and networks. Design, Setting, and Participants This case-control study used lesion location and lesion network mapping to identify the brain regions and networks associated with epilepsy in a discovery data set of patients with poststroke epilepsy and control patients with stroke. Patients with stroke lesions and epilepsy (n = 76) or no epilepsy (n = 625) were included. Generalizability to other lesion types was assessed using 4 independent cohorts as validation data sets. The total numbers of patients across all datasets (both discovery and validation datasets) were 347 with epilepsy and 1126 without. Therapeutic relevance was assessed using deep brain stimulation sites that improve seizure control. Data were analyzed from September 2018 through December 2022. All shared patient data were analyzed and included; no patients were excluded. Main Outcomes and Measures Epilepsy or no epilepsy. Results Lesion locations from 76 patients with poststroke epilepsy (39 [51%] male; mean [SD] age, 61.0 [14.6] years; mean [SD] follow-up, 6.7 [2.0] years) and 625 control patients with stroke (366 [59%] male; mean [SD] age, 62.0 [14.1] years; follow-up range, 3-12 months) were included in the discovery data set. Lesions associated with epilepsy occurred in multiple heterogenous locations spanning different lobes and vascular territories. However, these same lesion locations were part of a specific brain network defined by functional connectivity to the basal ganglia and cerebellum. Findings were validated in 4 independent cohorts including 772 patients with brain lesions (271 [35%] with epilepsy; 515 [67%] male; median [IQR] age, 60 [50-70] years; follow-up range, 3-35 years). Lesion connectivity to this brain network was associated with increased risk of epilepsy after stroke (odds ratio [OR], 2.82; 95% CI, 2.02-4.10; P < .001) and across different lesion types (OR, 2.85; 95% CI, 2.23-3.69; P < .001). Deep brain stimulation site connectivity to this same network was associated with improved seizure control (r, 0.63; P < .001) in 30 patients with drug-resistant epilepsy (21 [70%] male; median [IQR] age, 39 [32-46] years; median [IQR] follow-up, 24 [16-30] months). Conclusions and Relevance The findings in this study indicate that lesion-related epilepsy mapped to a human brain network, which could help identify patients at risk of epilepsy after a brain lesion and guide brain stimulation therapies.
Collapse
Affiliation(s)
- Frederic L. W. V. J. Schaper
- Center for Brain Circuit Therapeutics, Departments of Neurology, Psychiatry and Radiology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Harvard University, Boston, Massachusetts
- Department of Neurology and School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Janne Nordberg
- Turku Brain and Mind Center, Department of Clinical Neurophysiology, Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland
| | - Alexander L. Cohen
- Center for Brain Circuit Therapeutics, Departments of Neurology, Psychiatry and Radiology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Harvard University, Boston, Massachusetts
- Department of Neurology, Boston Children’s Hospital, Boston, Massachusetts
- Computational Radiology Laboratory, Department of Radiology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Christopher Lin
- Center for Brain Circuit Therapeutics, Departments of Neurology, Psychiatry and Radiology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Joey Hsu
- Center for Brain Circuit Therapeutics, Departments of Neurology, Psychiatry and Radiology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Andreas Horn
- Center for Brain Circuit Therapeutics, Departments of Neurology, Psychiatry and Radiology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Michael A. Ferguson
- Center for Brain Circuit Therapeutics, Departments of Neurology, Psychiatry and Radiology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Shan H. Siddiqi
- Center for Brain Circuit Therapeutics, Departments of Neurology, Psychiatry and Radiology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Harvard University, Boston, Massachusetts
| | - William Drew
- Center for Brain Circuit Therapeutics, Departments of Neurology, Psychiatry and Radiology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Louis Soussand
- Center for Brain Circuit Therapeutics, Departments of Neurology, Psychiatry and Radiology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Harvard University, Boston, Massachusetts
- Department of Neurology, Boston Children’s Hospital, Boston, Massachusetts
| | - Anderson M. Winkler
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville
| | - Marta Simó
- Neuro-Oncology Unit, Hospital Universitari de Bellvitge - Institut Català d’Oncologia (IDIBELL), L’Hospitalet del Llobregat, Barcelona, Spain
| | - Jordi Bruna
- Neuro-Oncology Unit, Hospital Universitari de Bellvitge - Institut Català d’Oncologia (IDIBELL), L’Hospitalet del Llobregat, Barcelona, Spain
| | - Sylvain Rheims
- Department of Functional Neurology and Epileptology, Lyon Neurosciences Research Center, Hospices Civils de Lyon and University of Lyon, Lyon, France
- Institut national de la santé et de la recherche médicale, Lyon, France
| | - Marc Guenot
- Institut national de la santé et de la recherche médicale, Lyon, France
- Department of Functional Neurosurgery, Hospices Civils de Lyon and University of Lyon, Lyon, France
| | - Marco Bucci
- Turku PET Centre, University of Turku and Åbo Akademi University, Turku, Finland
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Lauri Nummenmaa
- Turku PET Centre, University of Turku and Åbo Akademi University, Turku, Finland
- Department of Psychology, University of Turku, Turku, Finland
| | - Julie Staals
- Department of Neurology and School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Albert J. Colon
- Academic Center for Epileptology Kempenhaeghe/Maastricht University Medical Center, Heeze & Maastricht, the Netherlands
- Department of Epileptology, Centre Hospitalier Universitaire Martinique, Fort-de-France, France
| | - Linda Ackermans
- Department of Neurosurgery and School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Ellen J. Bubrick
- Center for Brain Circuit Therapeutics, Departments of Neurology, Psychiatry and Radiology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Jurriaan M. Peters
- Harvard Medical School, Harvard University, Boston, Massachusetts
- Department of Neurology, Boston Children’s Hospital, Boston, Massachusetts
| | - Ona Wu
- Harvard Medical School, Harvard University, Boston, Massachusetts
- Athinoula A Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Natalia S. Rost
- Harvard Medical School, Harvard University, Boston, Massachusetts
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts
| | - Jordan Grafman
- Cognitive Neuroscience Laboratory, Think + Speak Lab, Shirley Ryan Ability Lab, Chicago, Illinois
- Department of Physical Medicine and Rehabilitation, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Hal Blumenfeld
- Departments of Neurology, Neuroscience and Neurosurgery, Yale School of Medicine, New Haven, Connecticut
| | - Yasin Temel
- Department of Neurosurgery and School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Rob P. W. Rouhl
- Department of Neurology and School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands
- Academic Center for Epileptology Kempenhaeghe/Maastricht University Medical Center, Heeze & Maastricht, the Netherlands
| | - Juho Joutsa
- Turku Brain and Mind Center, Department of Clinical Neurophysiology, Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland
- Turku PET Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Michael D. Fox
- Center for Brain Circuit Therapeutics, Departments of Neurology, Psychiatry and Radiology, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Harvard University, Boston, Massachusetts
- Athinoula A Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| |
Collapse
|
4
|
Zhang W, Wang T, Xue Y, Zhan B, Lai Z, Huang W, Peng X, Zhou Y. Research progress of extracellular vesicles and exosomes derived from mesenchymal stem cells in the treatment of oxidative stress-related diseases. Front Immunol 2023; 14:1238789. [PMID: 37646039 PMCID: PMC10461809 DOI: 10.3389/fimmu.2023.1238789] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/24/2023] [Indexed: 09/01/2023] Open
Abstract
There is growing evidence that mesenchymal stem cell-derived extracellular vesicles and exosomes can significantly improve the curative effect of oxidative stress-related diseases. Mesenchymal stem cell extracellular vesicles and exosomes (MSC-EVs and MSC-Exos) are rich in bioactive molecules and have many biological regulatory functions. In this review, we describe how MSC-EVs and MSC-Exos reduce the related markers of oxidative stress and inflammation in various systemic diseases, and the molecular mechanism of MSC-EVs and MSC-Exos in treating apoptosis and vascular injury induced by oxidative stress. The results of a large number of experimental studies have shown that both local and systemic administration can effectively inhibit the oxidative stress response in diseases and promote the survival and regeneration of damaged parenchymal cells. The mRNA and miRNAs in MSC-EVs and MSC-Exos are the most important bioactive molecules in disease treatment, which can inhibit the apoptosis, necrosis and oxidative stress of lung, heart, kidney, liver, bone, skin and other cells, and promote their survive and regenerate.
Collapse
Affiliation(s)
- Wenwen Zhang
- The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, China
- Department of Pathophysiology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Tingyu Wang
- The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, China
- Department of Pathophysiology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Yuanye Xue
- The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, China
- Department of Pathophysiology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Bingbing Zhan
- School of Pharmaceutical Sciences, Guangdong Medical University, Dongguan, China
| | - Zengjie Lai
- The Second Clinical Medical College of Guangdong Medical University, Dongguan, China
| | - Wenjie Huang
- School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Xinsheng Peng
- Biomedical Innovation Center, Guangdong Medical University, Dongguan, China
- Institute of Marine Medicine, Guangdong Medical University, Zhanjiang, China
| | - Yanfang Zhou
- The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, China
- Department of Pathophysiology, Guangdong Medical University, Dongguan, Guangdong, China
| |
Collapse
|
5
|
Cerovic M, Di Nunzio M, Craparotta I, Vezzani A. An in vitro model of drug-resistant seizures for selecting clinically effective antiseizure medications in Febrile Infection-Related Epilepsy Syndrome. Front Neurol 2023; 14:1129138. [PMID: 37034097 PMCID: PMC10074483 DOI: 10.3389/fneur.2023.1129138] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/21/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction FIRES is a rare epileptic encephalopathy induced by acute unremitting seizures that occur suddenly in healthy children or young adults after a febrile illness in the preceding 2 weeks. This condition results in high mortality, neurological disability, and drug-resistant epilepsy. The development of new therapeutics is hampered by the lack of validated experimental models. Our goal was to address this unmet need by providing a simple tool for rapid throughput screening of new therapies that target pathological inflammatory mechanisms in FIRES. The model was not intended to mimic the etiopathogenesis of FIRES which is still unknown, but to reproduce salient features of its clinical presentation such as the age, the cytokine storm and the refractoriness of epileptic activity to antiseizure medications (ASMs). Methods We refined an in vitro model of mouse hippocampal/temporal cortex acute slices where drug-resistant epileptic activity is induced by zero Mg2+/100 μM 4-aminopirydine. Clinical evidence suggests that acute unremitting seizures in FIRES are promoted by neuroinflammation triggered in the brain by the preceding infection. We mimicked this inflammatory component by exposing slices for 30 min to 10 μg/ml lipopolysaccharide (LPS). Results LPS induced a sustained neuroinflammatory response, as shown by increased mRNA levels of IL-1β, CXCL1 (IL-8), TNF, and increased IL-1β/IL-1Ra ratio. Epileptiform activity was exacerbated by neuroinflammation, also displaying increased resistance to maximal therapeutic concentrations of midazolam (100 μM), phenytoin (50 μM), sodium valproate (800 μM), and phenobarbital (100 μM). Treatment of LPS-exposed slices with two immunomodulatory drugs, a mouse anti-IL-6 receptor antibody (100 μM) corresponding to tocilizumab in humans, or anakinra (1.3 μM) which blocks the IL-1 receptor type 1, delayed the onset of epileptiform events and strongly reduced the ASM-resistant epileptiform activity evoked by neuroinflammation. These drugs were shown to reduce ASM-refractory seizures in FIRES patients. Discussion The neuroinflammatory component and the pharmacological responsiveness of epileptiform events provide a proof-of-concept validation of this in vitro model for the rapid selection of new treatments for acute ASM-refractory seizures in FIRES.
Collapse
Affiliation(s)
- Milica Cerovic
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- *Correspondence: Milica Cerovic
| | - Martina Di Nunzio
- Department of Acute Brain Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Ilaria Craparotta
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Annamaria Vezzani
- Department of Acute Brain Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Annamaria Vezzani
| |
Collapse
|
6
|
Erisken S, Nune G, Chung H, Kang JW, Koh S. Time and age dependent regulation of neuroinflammation in a rat model of mesial temporal lobe epilepsy: Correlation with human data. Front Cell Dev Biol 2022; 10:969364. [PMID: 36172274 PMCID: PMC9512631 DOI: 10.3389/fcell.2022.969364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/03/2022] [Indexed: 11/25/2022] Open
Abstract
Acute brain insults trigger diverse cellular and signaling responses and often precipitate epilepsy. The cellular, molecular and signaling events relevant to the emergence of the epileptic brain, however, remain poorly understood. These multiplex structural and functional alterations tend also to be opposing - some homeostatic and reparative while others disruptive; some associated with growth and proliferation while others, with cell death. To differentiate pathological from protective consequences, we compared seizure-induced changes in gene expression hours and days following kainic acid (KA)-induced status epilepticus (SE) in postnatal day (P) 30 and P15 rats by capitalizing on age-dependent differential physiologic responses to KA-SE; only mature rats, not immature rats, have been shown to develop spontaneous recurrent seizures after KA-SE. To correlate gene expression profiles in epileptic rats with epilepsy patients and demonstrate the clinical relevance of our findings, we performed gene analysis on four patient samples obtained from temporal lobectomy and compared to four control brains from NICHD Brain Bank. Pro-inflammatory gene expressions were at higher magnitudes and more sustained in P30. The inflammatory response was driven by the cytokines IL-1β, IL-6, and IL-18 in the acute period up to 72 h and by IL-18 in the subacute period through the 10-day time point. In addition, a panoply of other immune system genes was upregulated, including chemokines, glia markers and adhesion molecules. Genes associated with the mitogen activated protein kinase (MAPK) pathways comprised the largest functional group identified. Through the integration of multiple ontological databases, we analyzed genes belonging to 13 separate pathways linked to Classical MAPK ERK, as well as stress activated protein kinases (SAPKs) p38 and JNK. Interestingly, genes belonging to the Classical MAPK pathways were mostly transiently activated within the first 24 h, while genes in the SAPK pathways had divergent time courses of expression, showing sustained activation only in P30. Genes in P30 also had different regulatory functions than in P15: P30 animals showed marked increases in positive regulators of transcription, of signaling pathways as well as of MAPKKK cascades. Many of the same inflammation-related genes as in epileptic rats were significantly upregulated in human hippocampus, higher than in lateral temporal neocortex. They included glia-associated genes, cytokines, chemokines and adhesion molecules and MAPK pathway genes. Uniquely expressed in human hippocampus were adaptive immune system genes including immune receptors CDs and MHC II HLAs. In the brain, many immune molecules have additional roles in synaptic plasticity and the promotion of neurite outgrowth. We propose that persistent changes in inflammatory gene expression after SE leads not only to structural damage but also to aberrant synaptogenesis that may lead to epileptogenesis. Furthermore, the sustained pattern of inflammatory genes upregulated in the epileptic mature brain was distinct from that of the immature brain that show transient changes and are resistant to cell death and neuropathologic changes. Our data suggest that the epileptogenic process may be a result of failed cellular signaling mechanisms, where insults overwhelm the system beyond a homeostatic threshold.
Collapse
Affiliation(s)
- Sinem Erisken
- Department of Pediatrics, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University School of Medicine, Chicago, IL, United States
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, United States
| | - George Nune
- Department of Pediatrics, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University School of Medicine, Chicago, IL, United States
- Department of Neurology, University of Southern California, Los Angeles, CA, United States
| | - Hyokwon Chung
- Department of Pediatrics, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University School of Medicine, Chicago, IL, United States
- Department of Pediatrics, Children’s Hospital & Medical Center, University of Nebraska, Omaha, NE, United States
| | - Joon Won Kang
- Department of Pediatrics, Children’s Hospital & Medical Center, University of Nebraska, Omaha, NE, United States
- Department of Pediatrics & Medical Science, Brain Research Institute, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Sookyong Koh
- Department of Pediatrics, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University School of Medicine, Chicago, IL, United States
- Department of Pediatrics, Children’s Hospital & Medical Center, University of Nebraska, Omaha, NE, United States
- *Correspondence: Sookyong Koh,
| |
Collapse
|
7
|
Lekoubou A, Ba DM, Nguyen C, Liu G, Leslie DL, Bonilha L, Vernon CM. Poststroke Seizures and the Risk of Dementia Among Young Stroke Survivors. Neurology 2022; 99:e385-e392. [PMID: 35584925 PMCID: PMC9421769 DOI: 10.1212/wnl.0000000000200736] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 03/30/2022] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND AND OBJECTIVES The effect of new-onset seizures in young stroke survivors on the subsequent development of dementia is poorly understood. This study aimed to assess the association between new onset of seizure and dementia in a population-based study of patients with stroke. METHODS The IBM Watson Health MarketScan Commercial Claims and Encounters database for the years 2005-2014 served as the data source for this study. Using the International Classification of Diseases, Ninth Revision (ICD-9), we identified patients aged 18-60 years with ischemic strokes (ISs; 433.x1, 434.x1, and 436) and hemorrhagic strokes (HSs; 430, 431, 432.0, 432.1, and 432.9) between January 1, 2006, and December 31, 2009, which constituted our baseline study cohort. At baseline, all included participants were free of claims for dementia, brain tumors, toxin exposure, traumatic brain injury, and neuroinfectious diseases, identified using ICD-9 codes. They had at least 1-year continuous enrollment before the index stroke diagnosis and 5 years after, with no seizure claims within 1 year after the index date. The exposure of interest was seizures: a time-dependent variable. The study outcome of interest was dementia (ICD-9: 290.0, 290.10-13, 290.20-21, 290.3, 290.40-43, 291.2, 292.82, 294.10-11, 294.20-21, 294.8, 331.0, 331.11, 331.19, and 331.82), which occurred during the follow-up period from January 1, 2010, to December 31, 2014. A Cox proportional hazards regression model was applied to calculate the hazard ratio (HR) and 95% CI for the independent association of seizures with the occurrence of dementia. RESULTS At the end of the baseline period, we identified 23,680 patients with stroke (IS: 20,642 and HS: 3,038). The cumulative incidence of seizure was 6.7%, 6.4%, and 8.3% for all strokes, IS, and HS, respectively. The cumulative incidence of dementia was 1.3%, 1.4%, and 0.9% for all strokes, IS, and HS, respectively. After multivariable adjustment, young patients with stroke who developed seizures had a greater risk of dementia compared with those without seizures (all strokes adjusted HR: 2.53, 95% CI 1.84-3.48; IS: 2.52, 1.79-3.53; HS: 2.80, 1.05-7.43). DISCUSSION These findings suggest that the onset of seizures in young stroke survivors is associated with a 2.53 times increased risk of developing dementia. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that poststroke seizures increase the probability of dementia in young stroke survivors.
Collapse
Affiliation(s)
- Alain Lekoubou
- From the Department of Neurology (A.L., C.N.), Penn State University, Hershey Medical Center; Department of Public Health Sciences (D.M.B., G.L., D.L.L., C.M.V.), Penn State College of Medicine, Hershey; and Department of Neurology (L.B.), Medical University of South Carolina, Charleston.
| | - Djibril M Ba
- From the Department of Neurology (A.L., C.N.), Penn State University, Hershey Medical Center; Department of Public Health Sciences (D.M.B., G.L., D.L.L., C.M.V.), Penn State College of Medicine, Hershey; and Department of Neurology (L.B.), Medical University of South Carolina, Charleston
| | - Clever Nguyen
- From the Department of Neurology (A.L., C.N.), Penn State University, Hershey Medical Center; Department of Public Health Sciences (D.M.B., G.L., D.L.L., C.M.V.), Penn State College of Medicine, Hershey; and Department of Neurology (L.B.), Medical University of South Carolina, Charleston
| | - Guodong Liu
- From the Department of Neurology (A.L., C.N.), Penn State University, Hershey Medical Center; Department of Public Health Sciences (D.M.B., G.L., D.L.L., C.M.V.), Penn State College of Medicine, Hershey; and Department of Neurology (L.B.), Medical University of South Carolina, Charleston
| | - Douglas L Leslie
- From the Department of Neurology (A.L., C.N.), Penn State University, Hershey Medical Center; Department of Public Health Sciences (D.M.B., G.L., D.L.L., C.M.V.), Penn State College of Medicine, Hershey; and Department of Neurology (L.B.), Medical University of South Carolina, Charleston
| | - Leonardo Bonilha
- From the Department of Neurology (A.L., C.N.), Penn State University, Hershey Medical Center; Department of Public Health Sciences (D.M.B., G.L., D.L.L., C.M.V.), Penn State College of Medicine, Hershey; and Department of Neurology (L.B.), Medical University of South Carolina, Charleston
| | - Chinchilli M Vernon
- From the Department of Neurology (A.L., C.N.), Penn State University, Hershey Medical Center; Department of Public Health Sciences (D.M.B., G.L., D.L.L., C.M.V.), Penn State College of Medicine, Hershey; and Department of Neurology (L.B.), Medical University of South Carolina, Charleston
| |
Collapse
|
8
|
Han W, Jiang L, Song X, Li T, Chen H, Cheng L. VEGF Modulates Neurogenesis and Microvascular Remodeling in Epileptogenesis After Status Epilepticus in Immature Rats. Front Neurol 2022; 12:808568. [PMID: 35002944 PMCID: PMC8739962 DOI: 10.3389/fneur.2021.808568] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/06/2021] [Indexed: 12/17/2022] Open
Abstract
Neurogenesis and angiogenesis are widely recognized to occur during epileptogenesis and important in brain development. Because vascular endothelial growth factor (VEGF) is a critical neurovascular target in neurological diseases, its effect on neurogenesis, microvascular remodeling and epileptogenesis in the immature brain after lithium-pilocarpine-induced status epilepticus (SE) was investigated. The dynamic changes in and the correlation between hippocampal neurogenesis and microvascular remodeling after SE and the influence of VEGF or SU5416 injection into the lateral ventricles at different stages after SE on neurogenesis and microvascular remodeling through regulation of VEGF expression were assessed by immunofluorescence and immunohistochemistry. Western blot analysis revealed that the VEGFR2 signaling pathway promotes phosphorylated ERK and phosphorylated AKT expression. The effects of VEGF expression regulation at different stages after SE on pathological changes in hippocampal structure and spontaneous recurrent seizures (SRS) were evaluated by Nissl staining and electroencephalography (EEG). The results showed that hippocampal neurogenesis after SE is related to microvascular regeneration. VEGF promotion in the acute period and inhibition in the latent period after SE alleviates loss of hippocampal neuron, abnormal vascular regeneration and inhibits neural stem cells (NSCs) ectopic migration, which may effectively alleviate SRS severity. Interfering with VEGF via the AKT and ERK pathways in different phases after SE may be a promising strategy for treating and preventing epilepsy in children.
Collapse
Affiliation(s)
- Wei Han
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Li Jiang
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xiaojie Song
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Tianyi Li
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Hengsheng Chen
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Li Cheng
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
9
|
Luo Q, Xian P, Wang T, Wu S, Sun T, Wang W, Wang B, Yang H, Yang Y, Wang H, Liu W, Long Q. Antioxidant activity of mesenchymal stem cell-derived extracellular vesicles restores hippocampal neurons following seizure damage. Am J Cancer Res 2021; 11:5986-6005. [PMID: 33897894 PMCID: PMC8058724 DOI: 10.7150/thno.58632] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/15/2021] [Indexed: 12/24/2022] Open
Abstract
Oxidative stress is a critical event in neuronal damage following seizures. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have been shown to be promising nanotherapeutic agents in neurological disorders. However, the mechanism underlying MSC-EVs therapeutic efficacy for oxidative stress-induced neuronal damage remains poorly understood. Methods: We investigated the antioxidant and restoration activities of MSC-EVs on hippocampal neurons in response to H2O2 stimulation in vitro and seizures in vivo. We also explored the potential underlying mechanism by injecting adeno-associated virus (AAV)-nuclear factor erythroid-derived 2, like 2 (Nrf2), a key antioxidant mediator, in animal models. Results: MSC-EVs were enriched in antioxidant miRNAs and exhibited remarkable antioxidant activity evident by increased ferric ion-reducing antioxidant ability, catalase, superoxide dismutase, and glutathione peroxidase activities and decreased reactive oxygen species (ROS) generation, DNA/lipid/protein oxidation, and stress-associated molecular patterns in cultured cells and mouse models. Notably, EV administration exerted restorative effects on the hippocampal neuronal structure and associated functional impairments, including dendritic spine alterations, electrophysiological disturbances, calcium transients, mitochondrial changes, and cognitive decline after oxidative stress in vitro or in vivo. Mechanistically, we found that the Nrf2 signaling pathway was involved in the restorative effect of EV therapy against oxidative neuronal damage, while AAV-Nrf2 injection attenuated the antioxidant activity of MSC-EVs on the seizure-induced hippocampal injury. Conclusions: We have shown that MSC-EVs facilitate the reconstruction of hippocampal neurons associated with the Nrf2 defense system in response to oxidative insults. Our study highlights the clinical value of EV-therapy in neurological disorders such as seizures.
Collapse
|
10
|
Koh S, Wirrell E, Vezzani A, Nabbout R, Muscal E, Kaliakatsos M, Wickström R, Riviello JJ, Brunklaus A, Payne E, Valentin A, Wells E, Carpenter JL, Lee K, Lai Y, Eschbach K, Press CA, Gorman M, Stredny CM, Roche W, Mangum T. Proposal to optimize evaluation and treatment of Febrile infection-related epilepsy syndrome (FIRES): A Report from FIRES workshop. Epilepsia Open 2021; 6:62-72. [PMID: 33681649 PMCID: PMC7918329 DOI: 10.1002/epi4.12447] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 11/03/2020] [Accepted: 11/13/2020] [Indexed: 12/23/2022] Open
Abstract
Febrile infection-related epilepsy syndrome (FIRES) is a rare catastrophic epileptic encephalopathy that presents suddenly in otherwise normal children and young adults causing significant neurological disability, chronic epilepsy, and high rates of mortality. To suggest a therapy protocol to improve outcome of FIRES, workshops were held in conjunction with American Epilepsy Society annual meeting between 2017 and 2019. An international group of pediatric epileptologists, pediatric neurointensivists, rheumatologists and basic scientists with interest and expertise in FIRES convened to propose an algorithm for a standardized approach to the diagnosis and treatment of FIRES. The broad differential for refractory status epilepticus (RSE) should include FIRES, to allow empiric therapies to be started early in the clinical course. FIRES should be considered in all previously healthy patients older than two years of age who present with explosive onset of seizures rapidly progressing to RSE, following a febrile illness in the preceding two weeks. Once FIRES is suspected, early administrations of ketogenic diet and anakinra (the IL-1 receptor antagonist that blocks biologic activity of IL-1β) are recommended.
Collapse
Affiliation(s)
- Sookyong Koh
- Department of PediatricsEmory University School of MedicineAtlantaGAUSA
| | - Elaine Wirrell
- Child and Adolescent Neurology and EpilepsyMayo ClinicRochesterMNUSA
| | - Annamaria Vezzani
- Department of NeuroscienceInstituto di Ricerche Farmacologiche Mario Negri IRCCSMilanItaly
| | - Rima Nabbout
- Reference Centre for Rare EpilepsiesDepartment of Pediatric NeurologyNecker Enfants Malades Hospital, APHPImagine InstituteParis Descartes UniversityParisFrance
| | - Eyal Muscal
- Department of PediatricsSection of Pediatric, RheumatologyBaylor College of MedicineHoustonTXUSA
| | - Marios Kaliakatsos
- Department of NeurologyGreat Ormond Street Hospital for ChildrenLondonUK
| | - Ronny Wickström
- Neuropediatric UnitDepartment of Women's and Children's HealthKarolinska InstituteStockholmSweden
| | | | - Andreas Brunklaus
- Paediatric Neurosciences Research GroupRoyal Hospital for ChildrenGlasgowUK
| | - Eric Payne
- Child and Adolescent Neurology and EpilepsyMayo ClinicRochesterMNUSA
| | - Antonio Valentin
- Department of Basic and Clinical Neuroscience, Psychology and NeuroscienceDepartment of Clinical NeurophysiologyKing's College Hospital NHS TrustLondonUK
| | - Elizabeth Wells
- Center for Neuroscience and Behavioral MedicineChildren’s National Health SystemWashingtonDCUSA
| | - Jessica L. Carpenter
- Center for Neuroscience and Behavioral MedicineChildren’s National Health SystemWashingtonDCUSA
| | - Kihyeong Lee
- Comprehensive Epilepsy CenterAdvent Health for ChildrenOrlandoFLUSA
| | - Yi‐Chen Lai
- Jan and Dan Duncan Neurological Research InstituteBaylor College of MedicineHoustonTXUSA
| | - Krista Eschbach
- Department of PediatricsSection of NeurologyUniversity of Colorado DenverDenverCOUSA
| | - Craig A. Press
- Department of PediatricsSection of NeurologyUniversity of Colorado DenverDenverCOUSA
| | - Mark Gorman
- Department of NeurologyBoston Children’s HospitalBostonMAUSA
| | | | - William Roche
- Department of PediatricsEmory University School of MedicineAtlantaGAUSA
| | - Tara Mangum
- Department of PediatricsPhoenix Children’s HospitalPhoenixAZUSA
| |
Collapse
|
11
|
Pressler RM, Cilio MR, Mizrahi EM, Moshé SL, Nunes ML, Plouin P, Vanhatalo S, Yozawitz E, de Vries LS, Puthenveettil Vinayan K, Triki CC, Wilmshurst JM, Yamamoto H, Zuberi SM. The ILAE classification of seizures and the epilepsies: Modification for seizures in the neonate. Position paper by the ILAE Task Force on Neonatal Seizures. Epilepsia 2021; 62:615-628. [PMID: 33522601 DOI: 10.1111/epi.16815] [Citation(s) in RCA: 150] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 12/23/2022]
Abstract
Seizures are the most common neurological emergency in the neonatal period and in contrast to those in infancy and childhood, are often provoked seizures with an acute cause and may be electrographic-only. Hence, neonatal seizures may not fit easily into classification schemes for seizures and epilepsies primarily developed for older children and adults. A Neonatal Seizures Task Force was established by the International League Against Epilepsy (ILAE) to develop a modification of the 2017 ILAE Classification of Seizures and Epilepsies, relevant to neonates. The neonatal classification framework emphasizes the role of electroencephalography (EEG) in the diagnosis of seizures in the neonate and includes a classification of seizure types relevant to this age group. The seizure type is determined by the predominant clinical feature. Many neonatal seizures are electrographic-only with no evident clinical features; therefore, these are included in the proposed classification. Clinical events without an EEG correlate are not included. Because seizures in the neonatal period have been shown to have a focal onset, a division into focal and generalized is unnecessary. Seizures can have a motor (automatisms, clonic, epileptic spasms, myoclonic, tonic), non-motor (autonomic, behavior arrest), or sequential presentation. The classification allows the user to choose the level of detail when classifying seizures in this age group.
Collapse
Affiliation(s)
- Ronit M Pressler
- Clinical Neuroscience, UCL- Great Ormond Street Institute of Child Health, London, UK.,Department of Clinical Neurophysiology, Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Maria Roberta Cilio
- Division of Pediatric Neurology, Institute for Experimental and Clinical Research, Saint-Luc University Hospital, Université Catholique de Louvain, Brussels, Belgium
| | - Eli M Mizrahi
- Departments of Neurology and Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Solomon L Moshé
- Isabelle Rapin Division of Child Neurology, Saul R. Korey Department of Neurology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA.,Department of Pediatrics, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
| | - Magda L Nunes
- Pontificia Universidade Catolica do Rio Grande do Sul - PUCRS School of Medicine and the Brain Institute, Porto Alegre, RS, Brazil
| | - Perrine Plouin
- Department of Clinical Neurophysiology, Hospital Necker Enfant Malades, Paris, France
| | - Sampsa Vanhatalo
- Department of Clinical Neurophysiology and BABA center Children's Hospital, HUS Imaging, Neuroscience Center, Helsinki Institute of Life Science, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Elissa Yozawitz
- Isabelle Rapin Division of Child Neurology, Saul R. Korey Department of Neurology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA.,Department of Pediatrics, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
| | - Linda S de Vries
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Chahnez C Triki
- Department of Child Neurology, Hedi Chaker Hospital, LR19ES15 Sfax University, Sfax, Tunisia
| | - Jo M Wilmshurst
- Department of Paediatric Neurology, Red Cross War Memorial Children's Hospital, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Hitoshi Yamamoto
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Sameer M Zuberi
- Paediatric Neurosciences Research Group, Royal Hospital for Children & Institute of Health & Wellbeing, University of Glasgow, Glasgow, UK
| |
Collapse
|
12
|
Efficacy of Intravenous Hydrocortisone Treatment in Refractory Neonatal Seizures: A Report on Three Cases. Brain Sci 2020; 10:brainsci10110885. [PMID: 33233684 PMCID: PMC7699678 DOI: 10.3390/brainsci10110885] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 11/24/2022] Open
Abstract
Neonatal seizures are the most common neurological emergency, and neonatal status epilepticus (NSE) remains a controversial entity, with no general consensus about its definition and treatment. Here, we report on three newborns with NSE refractory to first- and second-line antiepileptic drugs successfully treated with intravenous (IV) hydrocortisone. The patients had previously failed therapy with levetiracetam, phenobarbital and midazolam, showing persistent clinical and electrical seizures. Modulation of brain inflammation triggered during prolonged epileptic activity has been thought to potentially explain the beneficial effects of anti-inflammatory treatment.
Collapse
|
13
|
Kang DH, Choi BY, Lee SH, Kho AR, Jeong JH, Hong DK, Kang BS, Park MK, Song HK, Choi HC, Lim MS, Suh SW. Effects of Cerebrolysin on Hippocampal Neuronal Death After Pilocarpine-Induced Seizure. Front Neurosci 2020; 14:568813. [PMID: 33177978 PMCID: PMC7596733 DOI: 10.3389/fnins.2020.568813] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022] Open
Abstract
Epilepsy is one of the most common and severe brain diseases. The exact cause of epilepsy is unclear. Epilepsy often occurs following brain damage, such as traumatic brain injury (TBI) and ischemia. Cerebrolysin is a porcine brain peptide that is a unique neurotropic and neuroprotective agent. Cerebrolysin has been reported to increase neuroprotective effects after TBI, ischemia, and other CNS diseases. However, the effects of cerebrolysin on seizures are not known. Therefore, this study aimed to investigate the effects of neuropeptide cerebrolysin on neuronal death in the hippocampus after a seizure. To confirm the effects of cerebrolysin, we used a pilocarpine-induced seizure animal model. Cerebrolysin (2.5 ml/kg, i.p., once per day for 7 days) was immediately injected after a seizure induction. After 1 week, we obtained brain tissues and performed staining to histologically evaluate the potentially protective effects of cerebrolysin on seizure-induced neuronal death in the hippocampus. We found that cerebrolysin decreased hippocampal neuronal death after a seizure. In addition, an increase in brain-derived neurotrophic factor (BDNF) was confirmed through Western blot analysis to further support our hypothesis. Therefore, the present study suggests that the administration of cerebrolysin can be a useful therapeutic tool for preventing neuronal death after a seizure.
Collapse
Affiliation(s)
- Dong Hyeon Kang
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
- Neurology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Bo Young Choi
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Song Hee Lee
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - A Ra Kho
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Jeong Hyun Jeong
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Dae Ki Hong
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Beom Seok Kang
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Min Kyu Park
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Hong Ki Song
- Neurology, College of Medicine, Hallym University, Chuncheon, South Korea
- Hallym Institute of Epilepsy Research, Chuncheon, South Korea
| | - Hui Chul Choi
- Neurology, College of Medicine, Hallym University, Chuncheon, South Korea
- Hallym Institute of Epilepsy Research, Chuncheon, South Korea
| | - Man-Sup Lim
- Department of Medical Education, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Sang Won Suh
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| |
Collapse
|
14
|
Huang JB, Hsu SP, Pan HY, Chen SD, Chen SF, Lin TK, Liu XP, Li JH, Chen NC, Liou CW, Hsu CY, Chuang HY, Chuang YC. Peroxisome Proliferator-Activated Receptor γ Coactivator 1α Activates Vascular Endothelial Growth Factor That Protects Against Neuronal Cell Death Following Status Epilepticus through PI3K/AKT and MEK/ERK Signaling. Int J Mol Sci 2020; 21:ijms21197247. [PMID: 33008083 PMCID: PMC7583914 DOI: 10.3390/ijms21197247] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022] Open
Abstract
Status epilepticus may cause molecular and cellular events, leading to hippocampal neuronal cell death. Peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) is an important regulator of vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR2), also known as fetal liver kinase receptor 1 (Flk-1). Resveratrol is an activator of PGC-1α. It has been suggested to provide neuroprotective effects in epilepsy, stroke, and neurodegenerative diseases. In the present study, we used microinjection of kainic acid into the left hippocampal CA3 region in Sprague Dawley rats to induce bilateral prolonged seizure activity. Upregulating the PGC-1α pathway will increase VEGF/VEGFR2 (Flk-1) signaling and further activate some survival signaling that includes the mitogen activated protein kinase kinase (MEK)/mitogen activated protein kinase (ERK) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathways and offer neuroprotection as a consequence of apoptosis in the hippocampal neurons following status epilepticus. Otherwise, downregulation of PGC-1α by siRNA against pgc-1α will inhibit VEGF/VEGFR2 (Flk-1) signaling and suppress pro-survival PI3K/AKT and MEK/ERK pathways that are also accompanied by hippocampal CA3 neuronal cell apoptosis. These results may indicate that the PGC-1α induced VEGF/VEGFR2 pathway may trigger the neuronal survival signaling, and the PI3K/AKT and MEK/ERK signaling pathways. Thus, the axis of PGC-1α/VEGF/VEGFR2 (Flk-1) and the triggering of downstream PI3K/AKT and MEK/ERK signaling could be considered an endogenous neuroprotective effect against apoptosis in the hippocampus following status epilepticus.
Collapse
Affiliation(s)
- Jyun-Bin Huang
- Department of Emergency Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (J.-B.H.); (H.-Y.P.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-D.C.); (S.-F.C.); (T.-K.L.); (N.-C.C.); (C.-W.L.)
| | - Shih-Pin Hsu
- Department of Neurology, E-Da Hospital/School of Medicine, I-Shou University, Kaohsiung 824, Taiwan;
| | - Hsiu-Yung Pan
- Department of Emergency Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (J.-B.H.); (H.-Y.P.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-D.C.); (S.-F.C.); (T.-K.L.); (N.-C.C.); (C.-W.L.)
| | - Shang-Der Chen
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-D.C.); (S.-F.C.); (T.-K.L.); (N.-C.C.); (C.-W.L.)
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (X.-P.L.); (J.-H.L.)
| | - Shu-Fang Chen
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-D.C.); (S.-F.C.); (T.-K.L.); (N.-C.C.); (C.-W.L.)
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Tsu-Kung Lin
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-D.C.); (S.-F.C.); (T.-K.L.); (N.-C.C.); (C.-W.L.)
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Mitochondrial Research Unit, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Xuan-Ping Liu
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (X.-P.L.); (J.-H.L.)
| | - Jie-Hau Li
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (X.-P.L.); (J.-H.L.)
| | - Nai-Ching Chen
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-D.C.); (S.-F.C.); (T.-K.L.); (N.-C.C.); (C.-W.L.)
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Chia-Wei Liou
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-D.C.); (S.-F.C.); (T.-K.L.); (N.-C.C.); (C.-W.L.)
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Mitochondrial Research Unit, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Chung-Yao Hsu
- Department of Neurology, School of Medicine, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Hung-Yi Chuang
- Department of Occupational and Environmental Medicine, Kaohsiung Medical University Hospital and School of Public Health, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Yao-Chung Chuang
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-D.C.); (S.-F.C.); (T.-K.L.); (N.-C.C.); (C.-W.L.)
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (X.-P.L.); (J.-H.L.)
- Department of Neurology, School of Medicine, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Biological Science, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Correspondence:
| |
Collapse
|
15
|
Which clinical and neuropsychological factors are responsible for cognitive impairment in patients with epilepsy? Int J Public Health 2020; 65:947-956. [DOI: 10.1007/s00038-020-01401-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 10/24/2022] Open
|
16
|
VanHaerents S, Gerard EE. Epilepsy Emergencies: Status Epilepticus, Acute Repetitive Seizures, and Autoimmune Encephalitis. Continuum (Minneap Minn) 2019; 25:454-476. [PMID: 30921018 DOI: 10.1212/con.0000000000000716] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW This article reviews epilepsy emergencies, including status epilepticus, acute repetitive seizures, autoimmune encephalitis, and the current perspective on their diagnosis and treatment. RECENT FINDINGS Recent guidelines on the treatment of status epilepticus from the Neurocritical Care Society in 2012 and the American Epilepsy Society in 2016 highlight areas of consensus in the treatment of status epilepticus as well as areas of uncertainty. The TRENdS (Treatment of Recurrent Electrographic Nonconvulsive Seizures) trial is the first prospective randomized clinical trial to evaluate the efficacy of IV antiseizure medications in controlling nonconvulsive seizures on continuous EEG. It demonstrated that IV lacosamide is noninferior to fosphenytoin in this setting. Autoimmune encephalitis is an increasingly recognized cause of new-onset seizures or status epilepticus. Recently described scoring systems, the Antibody Prevalence in Epilepsy score and the Response to Immunotherapy in Epilepsy score, can help in the assessment of autoimmune encephalitis. SUMMARY Status epilepticus, acute repetitive seizures, and autoimmune encephalitis are neurologic emergencies. For all these conditions, rapid and appropriate treatment may influence patient prognosis and mitigate neuronal injury. For convulsive status epilepticus, there is reasonable consensus on the initial steps that need to be taken. There is less agreement about the management of acute repetitive seizures and nonconvulsive status epilepticus. An increasingly recognized etiology of status epilepticus is autoimmune encephalitis, which may not be as rare as previously thought.
Collapse
|
17
|
Novel treatment approaches and pediatric research networks in status epilepticus. Epilepsy Behav 2019; 101:106564. [PMID: 31708430 DOI: 10.1016/j.yebeh.2019.106564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 09/11/2019] [Indexed: 11/22/2022]
Abstract
This paper contains five contributions which were presented as part of the novel therapies section of the 7th London-Innsbruck Colloquium on Status Epilepticus and Acute Seizures. These illustrate recent advances being made in the management and therapy of status epilepticus. The five contributions concern: genetic variations in Na + channel genes and their importance in status epilepticus; the European Reference Network for rare and complex epilepsies EpiCARE; the North American Pediatric Status Epilepticus Research Group (pSERG); Fenfluramine as a potential therapy for status epilepticus' and the valproate derivatives, valnoctamide and sec-butylpropylacetamide (SPD), as potential therapies for status epilepticus. This article is part of the Special Issue "Proceedings of the 7th London-Innsbruck Colloquium on Status Epilepticus and Acute Seizures".
Collapse
|
18
|
Status Epilepticus Increases Cell Proliferation and Neurogenesis in the Developing Rat Cerebellum. THE CEREBELLUM 2019; 19:48-57. [PMID: 31656012 DOI: 10.1007/s12311-019-01078-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Status epilepticus (SE) promotes neuronal proliferation and differentiation in the adult and developing rodent hippocampus. However, the effect of SE on other neurogenic brain regions such as the cerebellum has been less explored. To determine whether SE induced by pentylentetrazole (PTZ-SE) and lithium-pilocarpine (Li-Pilo-SE) increases cell proliferation and neurogenesis in the developing rat cerebellum. SE was induced in 14-day-old (P14) Wistar rat pups (both sexes). One hour after SE and the following day rats were injected intraperitoneally with 5-bromo-2'-deoxyuridine (BrdU, 50 mg/kg). Seven days after SE, immunohistochemistry was performed to detect BrdU-positive (BrdU+) cells or BrdU/NeuN+ cells in the cerebellar vermis. SE induced by PTZ or Li-Pilo statistically significant increased the number of cerebellar BrdU+ cells when compared with the control group (58% and 40%, respectively); maximal cell proliferation occurred in lobules II, III, VIb, VIc, VIII, IXa, and IXb of PTZ-SE group and II, V, VIc, VII, and X of Li-Pilo-SE group. An increased number of BrdU/NeuN+ cells was detected in lobules V (17 ± 1.9), VIc (25.8 ± 2.7), and VII (26.2 ± 3.4) after Li-Pilo-SE compared to their control group (9.8 ± 1.7, 12.8 ± 2.8, and 11 ± 1.7, respectively), while the number of BrdU/NeuN+ cells remained the same after PTZ-induced SE or control conditions. SE induced in the developing rat by different experimental models increases cell proliferation in the granular layer of the cerebellar vermis, but only SE of limbic seizures increases neurogenesis in specific cerebellar lobes.
Collapse
|
19
|
Bourel-Ponchel E, Mahmoudzadeh M, Adebimpe A, Wallois F. Functional and Structural Network Disorganizations in Typical Epilepsy With Centro-Temporal Spikes and Impact on Cognitive Neurodevelopment. Front Neurol 2019; 10:809. [PMID: 31555191 PMCID: PMC6727184 DOI: 10.3389/fneur.2019.00809] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 07/15/2019] [Indexed: 12/20/2022] Open
Abstract
Epilepsy with Centrotemporal Spikes (ECTS) is the most common form of self-limited focal epilepsy. The pathophysiological mechanisms by which ECTS induces neuropsychological impairment in 15-30% of affected children remain unclear. The objective of this study is to review the current state of knowledge concerning the brain structural and functional changes that may be involved in cognitive dysfunctions in ECTS. Structural brain imaging suggests the presence of subtle neurodevelopmental changes over the epileptogenic zone and over distant regions in ECTS. This structural remodeling likely occurs prior to the diagnosis and evolves over time, especially in patients with cognitive impairment, suggesting that the epileptogenic processes might interfere with the dynamics of the brain development and/or the normal maturation processes. Functional brain imaging demonstrates profound disorganization accentuated by interictal epileptic spikes (IES) in the epileptogenic zone and in remote networks in ECTS. Over the epileptogenic zone, the literature demonstrates changes in term of neuronal activity and synchronization, which are effective several hundred milliseconds before the IES. In the same time window, functional changes are also observed in bilateral distant networks, notably in the frontal and temporal lobes. Effective connectivity demonstrates that the epileptogenic zone constitutes the key area at the origin of IES propagation toward distant cortical regions, including frontal areas. Altogether, structural and functional network disorganizations, in terms of: (i) power spectral values, (ii) functional and effective connectivity, are likely to participate in the cognitive impairment commonly reported in children with ECTS. These results suggest a central and causal role of network disorganizations related to IES in the neuropsychological impairment described in ECTS children.
Collapse
Affiliation(s)
- Emilie Bourel-Ponchel
- INSERM UMR 1105, Research Group on Multimodal Analysis of Brain Function, University of Picardie Jules Verne, Amiens, France
- INSERM UMR 1105, EFSN Pediatric, Amiens University Hospital, Amiens, France
| | - Mahdi Mahmoudzadeh
- INSERM UMR 1105, Research Group on Multimodal Analysis of Brain Function, University of Picardie Jules Verne, Amiens, France
- INSERM UMR 1105, EFSN Pediatric, Amiens University Hospital, Amiens, France
| | - Azeez Adebimpe
- INSERM UMR 1105, Research Group on Multimodal Analysis of Brain Function, University of Picardie Jules Verne, Amiens, France
| | - Fabrice Wallois
- INSERM UMR 1105, Research Group on Multimodal Analysis of Brain Function, University of Picardie Jules Verne, Amiens, France
- INSERM UMR 1105, EFSN Pediatric, Amiens University Hospital, Amiens, France
| |
Collapse
|
20
|
Jia R, Jia N, Yang F, Liu Z, Li R, Jiang Y, Zhao J, Wang L, Zhang S, Zhang Z, Zhang H, Wu S, Gao F, Jiang W. Hydrogen Alleviates Necroptosis and Cognitive Deficits in Lithium-Pilocarpine Model of Status Epilepticus. Cell Mol Neurobiol 2019; 39:857-869. [PMID: 31089833 DOI: 10.1007/s10571-019-00685-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022]
Abstract
Status epilepticus without prompt seizure control always leads to neuronal death and long-term cognitive deficits, but effective intervention is still absent. Here, we found that hydrogen could alleviate the hippocampus-dependent spatial learning and memory deficit in lithium-pilocarpine model of status epilepticus in rats, as evidenced by the results in Morris water maze test. Hydrogen treatment downregulated the expression of necroptosis-related proteins, such as MLKL, phosphorylated-MLKL, and RIPK3 in hippocampus, and further protected neurons and astrocytes from necroptosis which was here first verified to occur in status epilepticus. Hydrogen also protected cells from apoptosis, which was indicated by the decreased cleaved-Caspase 3 expression. Meanwhile, Iba1+ microglial activation by status epilepticus was reduced by hydrogen treatment. These findings confirm the utility of hydrogen treatment in averting cell death including necroptosis and alleviating cognitive deficits caused by status epilepticus. Therefore, hydrogen may provide a potential and powerful clinical treatment for status epilepticus-related cognitive deficits.
Collapse
Affiliation(s)
- Ruihua Jia
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, 127 Chang Le Xi Road, Xi'an, 710032, Shaanxi, China
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, Shaanxi, China
| | - Ning Jia
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, Shaanxi, China
- The Medical College of Yan'an University, 19 Guanghua Street, Yan'an, 716000, Shaanxi, China
| | - Fang Yang
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, 127 Chang Le Xi Road, Xi'an, 710032, Shaanxi, China
| | - Zihe Liu
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, Shaanxi, China
| | - Rui Li
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, Shaanxi, China
| | - Yongli Jiang
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, 127 Chang Le Xi Road, Xi'an, 710032, Shaanxi, China
| | - Jingjing Zhao
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, 127 Chang Le Xi Road, Xi'an, 710032, Shaanxi, China
| | - Lu Wang
- The Medical College of Yan'an University, 19 Guanghua Street, Yan'an, 716000, Shaanxi, China
| | - Shuo Zhang
- Department of Diagnostic Radiology, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Zhengping Zhang
- Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, 555 Youyi Road, Xi'an, 710054, Shaanxi, China
| | - Haifeng Zhang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, Shaanxi, China
| | - Shengxi Wu
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, Shaanxi, China
| | - Fang Gao
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, 710032, Shaanxi, China.
| | - Wen Jiang
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, 127 Chang Le Xi Road, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
21
|
Sirtuin 1 Regulates Mitochondrial Biogenesis and Provides an Endogenous Neuroprotective Mechanism Against Seizure-Induced Neuronal Cell Death in the Hippocampus Following Status Epilepticus. Int J Mol Sci 2019; 20:ijms20143588. [PMID: 31340436 PMCID: PMC6678762 DOI: 10.3390/ijms20143588] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/21/2019] [Accepted: 07/22/2019] [Indexed: 12/20/2022] Open
Abstract
Status epilepticus may decrease mitochondrial biogenesis, resulting in neuronal cell death occurring in the hippocampus. Sirtuin 1 (SIRT1) functionally interacts with peroxisome proliferator-activated receptors and γ coactivator 1α (PGC-1α), which play a crucial role in the regulation of mitochondrial biogenesis. In Sprague-Dawley rats, kainic acid was microinjected unilaterally into the hippocampal CA3 subfield to induce bilateral seizure activity. SIRT1, PGC-1α, and other key proteins involving mitochondrial biogenesis and the amount of mitochondrial DNA were investigated. SIRT1 antisense oligodeoxynucleotide was used to evaluate the relationship between SIRT1 and mitochondrial biogenesis, as well as the mitochondrial function, oxidative stress, and neuronal cell survival. Increased SIRT1, PGC-1α, and mitochondrial biogenesis machinery were found in the hippocampus following experimental status epilepticus. Downregulation of SIRT1 decreased PGC-1α expression and mitochondrial biogenesis machinery, increased Complex I dysfunction, augmented the level of oxidized proteins, raised activated caspase-3 expression, and promoted neuronal cell damage in the hippocampus. The results suggest that the SIRT1 signaling pathway may play a pivotal role in mitochondrial biogenesis, and could be considered an endogenous neuroprotective mechanism counteracting seizure-induced neuronal cell damage following status epilepticus.
Collapse
|
22
|
López-Vázquez MÁ, Gama-García CE, Estrada-Reyes Y, Gaytán-Tocavén L, Alfaro JMC, Olvera-Cortés ME. Neonatal Monosodium Glutamate Administration Disrupts Place Learning and Alters Hippocampal-Prefrontal Learning-Related Theta Activity in the Adult Rat. Neuroscience 2019; 414:228-244. [PMID: 31299349 DOI: 10.1016/j.neuroscience.2019.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/23/2019] [Accepted: 07/01/2019] [Indexed: 01/30/2023]
Abstract
Neonatal treatment with monosodium glutamate causes profound deficits in place learning and memory in adult rats evaluated in the Morris maze. Theta activity has been related to hippocampal learning, and increased high-frequency theta activity occurs through efficient place learning training in the Morris maze. We wondered whether the place learning deficits observed in adult rats that had been neonatally treated with monosodium glutamate (MSG), were related to altered theta patterns in the hippocampus and prelimbic cortex, which were recorded during place learning training in the Morris maze. The MSG-treated group had a profound deficit in place learning ability, with a marginal reduction in escape latencies during the final days of training. Learning-related changes were observed in the relative power distribution in control and MSG-treated groups in the hippocampal EEG, but not in the prelimbic cortex. Increased prefrontal and reduced hippocampal absolute power that appeared principally during the final days of training, and reduced coherence between regions throughout the training (4-12 Hz), were observed in the MSG-treated rats, thereby suggesting a misfunction of the circuits rather than a hyperexcitable general state. In conclusion, neonatal administration of MSG, which caused a profound deficit in place learning at the adult age, also altered the theta pattern both in the hippocampus and prelimbic cortex.
Collapse
Affiliation(s)
- Miguel Ángel López-Vázquez
- Laboratorio de Neuroplasticidad de los Procesos Cognitivos, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Camino de la Arboleda 300, Ex-Hacienda de San José de la Huerta, C.P., 58341, Morelia, Michoacán, México.
| | - Carla Estefanía Gama-García
- Laboratorio de Neurofisiología Experimental, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Camino de la Arboleda 300, Ex-Hacienda de San José de la Huerta, C.P., 58341, Morelia, Michoacán, México
| | - Yoana Estrada-Reyes
- Laboratorio de Neuroplasticidad de los Procesos Cognitivos, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Camino de la Arboleda 300, Ex-Hacienda de San José de la Huerta, C.P., 58341, Morelia, Michoacán, México
| | - Lorena Gaytán-Tocavén
- Laboratorio de Neurofisiología Experimental, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Camino de la Arboleda 300, Ex-Hacienda de San José de la Huerta, C.P., 58341, Morelia, Michoacán, México
| | - José Miguel Cervantes Alfaro
- Laboratorio de Neurociencias, Departamento de Postgrado, Facultad de Medicina "Dr. Ignacio Chávez", Universidad Michoacana de San Nicolás de Hidalgo, Rafael Carrillo esq. Salvador González Herrejón S/N. C.P., 58000, Colonia Centro, Morelia, Michoacán, México
| | - María Esther Olvera-Cortés
- Laboratorio de Neurofisiología Experimental, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Camino de la Arboleda 300, Ex-Hacienda de San José de la Huerta, C.P., 58341, Morelia, Michoacán, México
| |
Collapse
|
23
|
Lauková M, Velíšková J, Velíšek L, Shakarjian MP. Tetramethylenedisulfotetramine neurotoxicity: What have we learned in the past 70 years? Neurobiol Dis 2019; 133:104491. [PMID: 31176716 DOI: 10.1016/j.nbd.2019.104491] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/27/2019] [Accepted: 06/05/2019] [Indexed: 12/21/2022] Open
Abstract
Tetramethylenedisulfotetramine (tetramine, TETS, TMDT) is a seizure-producing neurotoxic chemical formed by the condensation of sulfamide and formaldehyde. Serendipitously discovered through an occupational exposure in 1949, it was promoted as a rodenticide but later banned worldwide due to its danger to human health. However, exceptional activity of the agent against rodent pests resulted in its clandestine manufacture with large numbers of inadvertent, intentional, and mass poisonings, which continue to this day. Facile synthesis, extreme potency, persistence, lack of odor, color, and taste identify it as an effective food adulterant and potential chemical agent of terror. No known antidote or targeted treatment is currently available. In this review we examine the origins of tetramethylenedisulfotetramine, from its identification as a neurotoxicant 70 years ago, through early research, to the most recent findings including the risk it poses in the post-911 world. Included is the information known regarding its in vitro pharmacology as a GABAA receptor channel antagonist, the toxic syndrome it produces in vivo, and its effect upon vulnerable populations. We also summarize the available information about potential therapeutic countermeasures and treatment strategies as well as the contribution of clinical development of TMDT poisoning to our understanding of epileptogenesis. Finally we identify gaps in our knowledge and suggest potentially fruitful directions for continued research on this dangerous, yet intriguing compound.
Collapse
Affiliation(s)
- Marcela Lauková
- Department of Public Health, Division of Environmental Health Science, School of Health Sciences and Practice, New York Medical College, 40 Sunshine Cottage Rd, Valhalla, NY 10595, USA; Department of Pediatrics, New York Medical College, 40 Sunshine Cottage Rd, Valhalla, NY 10595, USA; Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava 84505, Slovakia
| | - Jana Velíšková
- Department of Cell Biology and Anatomy, New York Medical College, 40 Sunshine Cottage Rd, Valhalla, NY 10595, USA; Department of Obstetrics and Gynecology, New York Medical College, 40 Sunshine Cottage Rd, Valhalla, NY 10595, USA; Department of Neurology, New York Medical College, 40 Sunshine Cottage Rd, Valhalla, NY 10595, USA
| | - Libor Velíšek
- Department of Cell Biology and Anatomy, New York Medical College, 40 Sunshine Cottage Rd, Valhalla, NY 10595, USA; Department of Neurology, New York Medical College, 40 Sunshine Cottage Rd, Valhalla, NY 10595, USA; Department of Pediatrics, New York Medical College, 40 Sunshine Cottage Rd, Valhalla, NY 10595, USA
| | - Michael P Shakarjian
- Department of Public Health, Division of Environmental Health Science, School of Health Sciences and Practice, New York Medical College, 40 Sunshine Cottage Rd, Valhalla, NY 10595, USA; Department of Cell Biology and Anatomy, New York Medical College, 40 Sunshine Cottage Rd, Valhalla, NY 10595, USA; Department of Medicine, Division of Pulmonary and Critical Care Medicine, Rutgers-Robert Wood Johnson Medical School, 675 Hoes Ln W, Piscataway, NJ 08854, USA.
| |
Collapse
|
24
|
Noè F, Cattalini A, Vila Verde D, Alessi C, Colciaghi F, Figini M, Zucca I, de Curtis M. Epileptiform activity contralateral to unilateral hippocampal sclerosis does not cause the expression of brain damage markers. Epilepsia 2019; 60:1184-1199. [PMID: 31111475 DOI: 10.1111/epi.15611] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/24/2019] [Accepted: 04/24/2019] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Patients with epilepsy often ask if recurrent seizures harm their brain and aggravate their epileptic condition. This crucial question has not been specifically addressed by dedicated experiments. We analyze here if intense bilateral seizure activity induced by local injection of kainic acid (KA) in the right hippocampus produces brain damage in the left hippocampus. METHODS Adult guinea pigs were bilaterally implanted with hippocampal electrodes for continuous video-electroencephalography (EEG) monitoring. Unilateral injection of 1 μg KA in the dorsal CA1 area induced nonconvulsive status epilepticus (ncSE) characterized by bilateral hippocampal seizure discharges. This treatment resulted in selective unilateral sclerosis of the KA-injected hippocampus. Three days after KA injection, the animals were killed, and the brains were submitted to ex vivo magnetic resonance imaging (MRI) and were processed for immunohistochemical analysis. RESULTS During ncSE, epileptiform activity was recorded for 27.6 ± 19.1 hours in both the KA-injected and contralateral hippocampi. Enhanced T1-weighted MR signal due to gadolinium deposition, mean diffusivity reduction, neuronal loss, gliosis, and blood-brain barrier permeability changes was observed exclusively in the KA-injected hippocampus. Despite the presence of a clear unilateral hippocampal sclerosis at the site of KA injection, no structural alterations were detected by MR and immunostaining analysis performed in the hippocampus contralateral to KA injection 3 days and 2 months after ncSE induction. Fluoro-Jade and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining at the same time points confirmed the absence of degenerating cells in the hippocampi contralateral to KA injection. SIGNIFICANCE We demonstrate that intense epileptiform activity during ncSE does not cause obvious brain damage in the hippocampus contralateral to unilateral hippocampal KA injection. These findings argue against the hypothesis that epileptiform activity per se contributes to focal brain injury in previously undamaged cortical regions.
Collapse
Affiliation(s)
- Francesco Noè
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | - Diogo Vila Verde
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Camilla Alessi
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Francesca Colciaghi
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Matteo Figini
- Scientific Direction, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Ileana Zucca
- Scientific Direction, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Marco de Curtis
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
25
|
Bialer M, Johannessen SI, Koepp MJ, Levy RH, Perucca E, Tomson T, White HS. Progress report on new antiepileptic drugs: A summary of the Fourteenth Eilat Conference on New Antiepileptic Drugs and Devices (EILAT XIV). II. Drugs in more advanced clinical development. Epilepsia 2019; 59:1842-1866. [PMID: 30368788 DOI: 10.1111/epi.14555] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/05/2018] [Accepted: 08/06/2018] [Indexed: 12/24/2022]
Abstract
The Fourteenth Eilat Conference on New Antiepileptic Drugs and Devices (EILAT XIV) took place in Madrid, Spain, on May 13-16, 2018 and was attended by 168 delegates from 28 countries. The conference provided a forum for professionals involved in basic science, clinical research, regulatory affairs, and clinical care to meet and discuss the latest advances related to discovery and development of drugs and devices aimed at improving the management of people with epilepsy. This progress report provides a summary of findings on investigational compounds for which data from both preclinical studies and studies in patients were presented. The compounds reviewed include anakinra, cannabidiol, cannabidivarin, fenfluramine, ganaxolone, medium-chain fatty acids, padsevonil, and the valproic derivatives valnoctamide and sec-butylpropylacetamide. On June 25, 2018, the US Food and Drug Administration approved a standardized formulation of cannabidiol oral solution for the treatment of seizures associated with Lennox-Gastaut syndrome and Dravet syndrome in patients 2 years and older. The report shows that there continues to be a steady flow of potential antiepileptic drugs progressing to clinical development. Many of these compounds show innovative mechanisms of action, and some have already been tested in placebo-controlled randomized controlled trials, with promising efficacy and safety results.
Collapse
Affiliation(s)
- Meir Bialer
- Faculty of Medicine, School of Pharmacy and David R. Bloom Center for Pharmacy, Institute for Drug Research, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Svein I Johannessen
- National Center for Epilepsy, Sandvika, Norway.,Department of Pharmacology, Oslo University Hospital, Oslo, Norway
| | - Matthias J Koepp
- Department of Clinical and Experimental Epilepsy, University College London Institute of Neurology, London, United Kingdom
| | - René H Levy
- Department of Pharmaceutics and Neurological Surgery, University of Washington, Seattle, Washington
| | - Emilio Perucca
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy.,IRCCS Mondino Foundation, Pavia, Italy
| | - Torbjörn Tomson
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - H Steve White
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, Washington
| |
Collapse
|
26
|
Milovanovic M, Radivojevic V, Radosavljev-Kircanski J, Grujicic R, Toskovic O, Aleksić-Hil O, Pejovic-Milovancevic M. Epilepsy and interictal epileptiform activity in patients with autism spectrum disorders. Epilepsy Behav 2019; 92:45-52. [PMID: 30611007 DOI: 10.1016/j.yebeh.2018.12.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 12/14/2018] [Accepted: 12/15/2018] [Indexed: 02/06/2023]
Abstract
PURPOSE The purpose of this study was to determine the prevalence of epilepsy and subclinical epileptiform abnormalities in children with autism spectrum disorder (ASD), and to investigate its effects on core autistic symptoms and adaptive behavior skills. METHODS Patients with diagnosis of ASD who met full criteria on Autism Diagnostic Interview-Revised (ADI-R) were included in the study. Adaptive behavior skills were assessed by Vineland Adaptive Behavior Scale-II (VABS-II). Clinical assessment for epilepsy and video electroencephalography (EEG) (v-EEG) examinations during wakefulness and/or sleep were prospectively performed in all patients. RESULTS A total of 112 patients with diagnosis of ASD of mean age 6.58 ± 3.72 were included in the study. Based on clinical and v-EEG assessments, three groups of patients were defined: 1) patients with epilepsy (n = 17; 15.2%); 2) patients with epileptiform discharges in absence of clinical seizures (n = 14; 12.5%); 3) patients without epilepsy and without epileptiform discharges (n = 81; 72.3%). There were no significant differences between three groups of patients on ADI-R subscores. Speech development was also not significantly related to epilepsy. There was a slight tendency of the VABS-II motor skills score to be higher in the group of patients with autism without clinical diagnosis of epilepsy and without subclinical epileptiform discharges (p < 0.05) in comparison with the two other groups. According to this tendency, we might claim that patients with higher scores on motor skills could have 0.88 times lower odds for having epileptiform EEG activity. CONCLUSIONS According to our results, we were not able to detect differences in the ADI-R between the three populations with ASD, all with unknown etiology. Epilepsy, as well as subclinical epileptic discharges, showed small effects on Motor Skills in patients with autism, and had no effect on adaptive behavior Communication/Socialization/Daily Living Skills.
Collapse
Affiliation(s)
- Maja Milovanovic
- Institute of Mental Health, Belgrade, Serbia; Faculty of Special Education and Rehabilitation, University of Belgrade, Belgrade, Serbia
| | | | | | | | - Oliver Toskovic
- Department of Psychology, Faculty of Philosophy, University of Belgrade, Belgrade, Serbia
| | | | - Milica Pejovic-Milovancevic
- Institute of Mental Health, Belgrade, Serbia; Faculty of Medicine, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
27
|
Haines KM, Matson LM, Dunn EN, Ardinger CE, Lee-Stubbs R, Bibi D, McDonough JH, Bialer M. Comparative efficacy of valnoctamide and sec-butylpropylacetamide (SPD) in terminating nerve agent-induced seizures in pediatric rats. Epilepsia 2019; 60:315-321. [PMID: 30615805 DOI: 10.1111/epi.14630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 12/02/2018] [Accepted: 12/03/2018] [Indexed: 01/13/2023]
Abstract
OBJECTIVES Children and adults are likely to be among the casualties in a civilian nerve agent exposure. This study evaluated the efficacy of valnoctamide (racemic-VCD), sec-butylpropylacetamide (racemic-SPD), and phenobarbital for stopping nerve agent seizures in both immature and adult rats. METHODS Female and male postnatal day (PND) 21, 28, and 70 (adult) rats, previously implanted with electroencephalography (EEG) electrodes were exposed to seizure-inducing doses of the nerve agents sarin or VX and EEG was recorded continuously. Five minutes after seizure onset, animals were treated with SPD, VCD, or phenobarbital. The up-down method was used over successive animals to determine the anticonvulsant median effective dose (ED50 ) of the drugs. RESULTS SPD-ED50 values in the VX model were the following: PND21, 53 mg/kg (male) and 48 mg/kg (female); PND28, 108 mg/kg (male) and 43 mg/kg (female); and PND70, 101 mg/kg (male) and 40 mg/kg (female). SPD-ED50 values in the sarin model were the following: PND21, 44 mg/kg (male) and 28 mg/kg (female); PND28, 79 mg/kg (male) and 34 mg/kg (female); and PND70, 53 mg/kg (male) and 53 mg/kg (female). VCD-ED50 values in the VX model were the following: PND21, 34 mg/kg (male) and 43 mg/kg (female); PND28, 165 mg/kg (male) and 59 mg/kg (female); and PND70, 87 mg/kg (male) and 91 mg/kg (female). VCD-ED50 values in the sarin model were the following: PND21, 45 mg/kg (male), 48 mg/kg (female); PND28, 152 mg/kg (male) 79 mg/kg (female); and PND70, 97 mg/kg (male) 79 mg/kg (female). Phenobarbital-ED50 values in the VX model were the following: PND21, 43 mg/kg (male) and 18 mg/kg (female); PND28, 48 mg/kg (male) and 97 mg/kg (female). Phenobarbital-ED50 values in the sarin model were the following: PND21, 32 mg/kg (male) and 32 mg/kg (female); PND28, 58 mg/kg (male) and 97 mg/kg (female); and PND70, 65 mg/kg (female). SIGNIFICANCE SPD and VCD demonstrated anticonvulsant activity in both immature and adult rats in the sarin- and VX-induced status epilepticus models. Phenobarbital was effective in immature rats, whereas in adult rats, higher doses were required that were accompanied by toxicity. Overall, significantly less drug was required to stop seizures in PND21 animals than in the older animals, and overall, males required higher amounts of drug than females.
Collapse
Affiliation(s)
- Kari M Haines
- Nerve Agent Countermeasures, Medical Toxicology Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Liana M Matson
- Nerve Agent Countermeasures, Medical Toxicology Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Emily N Dunn
- Nerve Agent Countermeasures, Medical Toxicology Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Cherish E Ardinger
- Nerve Agent Countermeasures, Medical Toxicology Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Robyn Lee-Stubbs
- Research Support Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - David Bibi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - John H McDonough
- Nerve Agent Countermeasures, Medical Toxicology Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Meir Bialer
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.,David R. Bloom Center for Pharmacy, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
28
|
Rigas P, Sigalas C, Nikita M, Kaplanian A, Armaos K, Leontiadis LJ, Zlatanos C, Kapogiannatou A, Peta C, Katri A, Skaliora I. Long-Term Effects of Early Life Seizures on Endogenous Local Network Activity of the Mouse Neocortex. Front Synaptic Neurosci 2018; 10:43. [PMID: 30538627 PMCID: PMC6277496 DOI: 10.3389/fnsyn.2018.00043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 11/06/2018] [Indexed: 12/17/2022] Open
Abstract
Understanding the long term impact of early life seizures (ELS) is of vital importance both for researchers and clinicians. Most experimental studies of how seizures affect the developing brain have drawn their conclusions based on changes detected at the cellular or behavioral level, rather than on intermediate levels of analysis, such as the physiology of neuronal networks. Neurons work as part of networks and network dynamics integrate the function of molecules, cells and synapses in the emergent properties of brain circuits that reflect the balance of excitation and inhibition in the brain. Therefore, studying network dynamics could help bridge the cell-to-behavior gap in our understanding of the neurobiological effects of seizures. To this end we investigated the long-term effects of ELS on local network dynamics in mouse neocortex. By using the pentylenetetrazole (PTZ)-induced animal model of generalized seizures, single or multiple seizures were induced at two different developmental stages (P9-15 or P19-23) in order to examine how seizure severity and brain maturational status interact to affect the brain's vulnerability to ELS. Cortical physiology was assessed by comparing spontaneous network activity (in the form of recurring Up states) in brain slices of adult (>5 mo) mice. In these experiments we examined two distinct cortical regions, the primary motor (M1) and somatosensory (S1) cortex in order to investigate regional differences in vulnerability to ELS. We find that the effects of ELSs vary depending on (i) the severity of the seizures (e.g., single intermittent ELS at P19-23 had no effect on Up state activity, but multiple seizures induced during the same period caused a significant change in the spectral content of spontaneous Up states), (ii) the cortical area examined, and (iii) the developmental stage at which the seizures are administered. These results reveal that even moderate experiences of ELS can have long lasting age- and region-specific effects in local cortical network dynamics.
Collapse
Affiliation(s)
- Pavlos Rigas
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - Maria Nikita
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Ani Kaplanian
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | | | - Christos Zlatanos
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - Charoula Peta
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Anna Katri
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Irini Skaliora
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
29
|
Pisani F, Pavlidis E. What is new: Talk about status epilepticus in the neonatal period. Eur J Paediatr Neurol 2018; 22:757-762. [PMID: 29861333 DOI: 10.1016/j.ejpn.2018.05.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 04/30/2018] [Accepted: 05/20/2018] [Indexed: 10/16/2022]
Abstract
Nowadays, no general consensus was achieved regarding neonatal status epilepticus and its definition. Indeed, different criteria (mainly based on seizure duration) were used. Whereas a recent proposal has been developed to define status epilepticus in older ages, it seems that the peculiar characteristics of neonatal seizures and of the immature brain make difficult to find a tailored definition for this period of life. Achieving a consensus on this entity would mean to make the first step toward a targeted therapeutic strategy of intervention.
Collapse
Affiliation(s)
- Francesco Pisani
- Child Neuropsychiatry Unit, Medicine & Surgery Department, University of Parma, Italy
| | - Elena Pavlidis
- Child Neuropsychiatry Unit, Medicine & Surgery Department, University of Parma, Italy.
| |
Collapse
|
30
|
Hong S, Li T, Luo Y, Li W, Tang X, Ye Y, Wu P, Hu Q, Cheng L, Chen H, Jiang L. Dynamic Changes of Astrocytes and Adenosine Signaling in Rat Hippocampus in Post-status Epilepticus Model of Epileptogenesis. Cell Mol Neurobiol 2018; 38:1227-1234. [PMID: 29770956 DOI: 10.1007/s10571-018-0590-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 05/08/2018] [Indexed: 01/04/2023]
Abstract
It is of great importance to explore the development of epileptogenesis, and the adenosine and adenosine kinase (ADK) system seems to play a key role in this process. The aim of this study is to explore the dynamic changes of astrocytes and adenosine signaling during epileptogenesis in rat hippocampus in a post-status epileptogenesis (SE) model. Rat SE models were built and killed for experiments at 1 day (acute phase of epileptogenesis), 5 days (latent phase), 4 weeks (chronic phase), and 8 weeks (late chronic phase of epileptogenesis) after SE induction. Immunofluorescence staining, high-performance liquid chromatography, and Western blotting were performed to assess changes of astrocytes, adenosine, ADK, and ADK receptors (including A1R, A2aR, A2bR, and A3R) in hippocampus. The expression level of glial fibrillary acidic protein significantly increased from latent to late chronic phase. The concentration of adenosine sharply increased in acute phase and gradually decreased in the remaining phases of post-SE, being significantly lower than in the control group in late chronic phase. Protein levels of A1R and A2aR in post-SE models increased in acute phase, whereas A2bR and A3R protein expression decreased in latent phase, chronic phase, and late chronic phase following post-SE epileptogenesis. Protein expression of ADK significantly increased during latent phase, chronic phase, and late chronic phase of post-SE epileptogenesis. In conclusion, the levels of adenosine and protein expression of A1R and A2R significantly increased during acute phase of post-SE. During the remaining phases of post-SE epileptogenesis, there was imbalance among astrocytes, adenosine, adenosine receptors, and ADK. Regulation of the ADK/adenosine system may provide potential treatment strategies for epileptogenesis.
Collapse
Affiliation(s)
- Siqi Hong
- Lab of Pediatric Neurology, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, China International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Tingsong Li
- Lab of Pediatric Neurology, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, China International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yuanyuan Luo
- Lab of Pediatric Neurology, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, China International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Wenjuan Li
- Lab of Pediatric Neurology, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, China International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoju Tang
- Lab of Pediatric Neurology, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, China International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yuanzhen Ye
- Lab of Pediatric Neurology, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, China International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Wu
- Lab of Pediatric Neurology, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, China International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Qiao Hu
- Lab of Pediatric Neurology, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, China International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Li Cheng
- Lab of Pediatric Neurology, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, China International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hengsheng Chen
- Lab of Pediatric Neurology, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, China International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Li Jiang
- Lab of Pediatric Neurology, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, China International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| |
Collapse
|
31
|
Scholl EA, Miller-Smith SM, Bealer SL, Lehmkuhle MJ, Ekstrand JJ, Dudek FE, McDonough JH. Age-dependent behaviors, seizure severity and neuronal damage in response to nerve agents or the organophosphate DFP in immature and adult rats. Neurotoxicology 2018; 66:10-21. [PMID: 29510177 PMCID: PMC5996394 DOI: 10.1016/j.neuro.2018.02.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 02/07/2018] [Accepted: 02/27/2018] [Indexed: 01/15/2023]
Abstract
Exposure to nerve agents (NAs) and other organophosphates (OPs) can initiate seizures that rapidly progress to status epilepticus (SE). While the electrographic and neuropathological sequelae of SE evoked by NAs and OPs have been characterized in adult rodents, they have not been adequately investigated in immature animals. In this study postnatal day (PND) 14, 21 and 28 rat pups, along with PND70 animals as adult controls, were exposed to NAs (sarin, VX) or another OP (diisopropylfluorophosphate, DFP). We then evaluated behavioral and electrographic (EEG) correlates of seizure activity, and performed neuropathology using Fluoro-Jade B. Although all immature rats exhibited behaviors that are often characterized as seizures, the incidence, duration, and severity of the electrographic seizure activity were age-dependent. No (sarin and VX) or brief (DFP) EEG seizure activity was evoked in PND14 rats, while SE progressively increased in severity as a function of age in PND21, 28 and 70 animals. Fluoro-Jade B staining was observed in multiple brain regions of animals that exhibited prolonged seizure activity. Neuronal injury in PND14 animals treated with DFP was lower than in older animals and absent in rats exposed to sarin or VX. In conclusion, we found that NAs and an OP provoked robust SE and neuronal injury similar to adults in PND21 and PND28, but not in PND14, rat pups. Convulsive behaviors were often present independent of EEG seizures and were unaccompanied by neuronal damage. These differential responses should be considered when investigating medical countermeasures for NA and OP exposure in pediatric populations.
Collapse
Affiliation(s)
- Erika A Scholl
- Department of Neurosurgery, University of Utah, Salt Lake City, UT, 84108 USA
| | - Stephanie M Miller-Smith
- Neuroscience Branch, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, 21010-5400 USA
| | - Steven L Bealer
- Department of Neurosurgery, University of Utah, Salt Lake City, UT, 84108 USA; Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, 84108 USA
| | - Mark J Lehmkuhle
- Department of Neurosurgery, University of Utah, Salt Lake City, UT, 84108 USA; Epitel, Inc., Salt Lake City, UT, 84111 USA
| | - Jeffrey J Ekstrand
- Department of Pediatrics, University of Utah, Salt Lake City, UT, 84108 USA
| | - F Edward Dudek
- Department of Neurosurgery, University of Utah, Salt Lake City, UT, 84108 USA
| | - John H McDonough
- Neuroscience Branch, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, 21010-5400 USA.
| |
Collapse
|
32
|
Vezzani A, Dingledine R, Rossetti AO. Immunity and inflammation in status epilepticus and its sequelae: possibilities for therapeutic application. Expert Rev Neurother 2018; 15:1081-92. [PMID: 26312647 DOI: 10.1586/14737175.2015.1079130] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Status epilepticus (SE) is a life-threatening neurological emergency often refractory to available treatment options. It is a very heterogeneous condition in terms of clinical presentation and causes, which besides genetic, vascular and other structural causes also include CNS or severe systemic infections, sudden withdrawal from benzodiazepines or anticonvulsants and rare autoimmune etiologies. Treatment of SE is essentially based on expert opinions and antiepileptic drug treatment per se seems to have no major impact on prognosis. There is, therefore, urgent need of novel therapies that rely upon a better understanding of the basic mechanisms underlying this clinical condition. Accumulating evidence in animal models highlights that inflammation ensuing in the brain during SE may play a determinant role in ongoing seizures and their long-term detrimental consequences, independent of an infection or auto-immune cause; this evidence encourages reconsideration of the treatment flow in SE patients.
Collapse
Affiliation(s)
- Annamaria Vezzani
- a 1 Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milano, Italy
| | | | | |
Collapse
|
33
|
Álvarez-Croda DM, Santiago-García J, Medel-Matus JS, Martínez-Quiroz J, Puig-Lagunes AA, Beltrán-Parrazal L, López-Meraz ML. Hippocampal distribution of IL-1β and IL-1RI following lithium-pilocarpine-induced status epilepticus in the developing rat. AN ACAD BRAS CIENC 2018; 88 Suppl 1:653-63. [PMID: 27168372 DOI: 10.1590/0001-3765201620150296] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 06/19/2015] [Indexed: 12/27/2022] Open
Abstract
The contribution of Interleukin-1β (IL-1β) to neuronal injury induced by status epilepticus (SE) in the immature brain remains unclear. The goal of this study was to determine the hippocampal expression of IL-1β and its type 1 receptor (IL-1RI) following SE induced by the lithium-pilocarpine model in fourteen-days-old rat pups; control animals were given an equal volume of saline instead of the convulsant. IL-1β and IL-1RI mRNA hippocampal levels were assessed by qRT-PCR 6 and 24 h after SE or control conditions. IL-1β and IL-1RI expression was detected in the dorsal hippocampus by immunohistochemical procedures; Fluoro-Jade B staining was carried out in parallel sections in order to detect neuronal cell death. IL-1β mRNA expression was increased 6 h following SE, but not at 24 h; however IL-1RI mRNA expression was unaffected when comparing with the control group. IL-1β and IL-1RI immunoreactivity was not detected in control animals. IL-1β and IL-1RI were expressed in the CA1 pyramidal layer, the dentate gyrus granular layer and the hilus 6 h after SE, whereas injured cells were detected 24 h following seizures. Early expression of IL-1β and IL-1RI in the hippocampus could be associated with SE-induced neuronal cell death mechanisms in the developing rat.
Collapse
Affiliation(s)
- Dulce-Mariely Álvarez-Croda
- Centro de Investigaciones Cerebrales, Universidad Veracruzana, Unidad de Ciencias de la Salud, Médicos y Odontólogos, s/n, Col. Unidad del Bosque, C.P. 91010, Xalapa, Veracruz, México, Universidad Veracruzana, Universidad Veracruzana, Centro de Investigaciones Cerebrales, Xalapa Veracruz , México.,Doutorado em Neuroetologia, Universidad Veracruzana, Av. Luis Castelazo, s/n, Col. Industrial Ánimas, C.P. 91190, Xalapa, Veracruz, México, Universidad Veracruzana, Universidad Veracruzana, Xalapa Veracruz , México
| | - Juan Santiago-García
- Instituto de Investigaciones Biológicas, Universidad Veracruzana, Av. Luis Castelazom s/n, Col. Industrial Ánimas, C.P. 91190, Xalapa, Veracruz, México, Universidad Veracruzana, Universidad Veracruzana, Instituto de Investigaciones Biológicas, Xalapa Veracruz , México
| | - Jesús S Medel-Matus
- Centro de Investigaciones Cerebrales, Universidad Veracruzana, Unidad de Ciencias de la Salud, Médicos y Odontólogos, s/n, Col. Unidad del Bosque, C.P. 91010, Xalapa, Veracruz, México, Universidad Veracruzana, Universidad Veracruzana, Centro de Investigaciones Cerebrales, Xalapa Veracruz , México
| | - Joel Martínez-Quiroz
- Departamento de Farmacia, Escuela Nacional de Ciencias Biológicas-IPN, Wilfrido Massieu, s/n, Unidad Profesional Adolfo López Mateos, Gustavo A. Madero, C.P. 07738, Mexico D.F., México, Escuela Nacional de Ciencias Biológicas, Departamento de Farmacia, Mexico D.F. , México
| | - Angel A Puig-Lagunes
- Centro de Investigaciones Cerebrales, Universidad Veracruzana, Unidad de Ciencias de la Salud, Médicos y Odontólogos, s/n, Col. Unidad del Bosque, C.P. 91010, Xalapa, Veracruz, México, Universidad Veracruzana, Universidad Veracruzana, Centro de Investigaciones Cerebrales, Xalapa Veracruz , México
| | - Luis Beltrán-Parrazal
- Centro de Investigaciones Cerebrales, Universidad Veracruzana, Unidad de Ciencias de la Salud, Médicos y Odontólogos, s/n, Col. Unidad del Bosque, C.P. 91010, Xalapa, Veracruz, México, Universidad Veracruzana, Universidad Veracruzana, Centro de Investigaciones Cerebrales, Xalapa Veracruz , México
| | - María-Leonor López-Meraz
- Centro de Investigaciones Cerebrales, Universidad Veracruzana, Unidad de Ciencias de la Salud, Médicos y Odontólogos, s/n, Col. Unidad del Bosque, C.P. 91010, Xalapa, Veracruz, México, Universidad Veracruzana, Universidad Veracruzana, Centro de Investigaciones Cerebrales, Xalapa Veracruz , México
| |
Collapse
|
34
|
Lauková M, Velíšková J, Velíšek L, Shakarjian MP. Developmental and sex differences in tetramethylenedisulfotetramine (TMDT)-induced syndrome in rats. Dev Neurobiol 2018; 78:403-416. [PMID: 29411537 DOI: 10.1002/dneu.22582] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 01/15/2018] [Accepted: 02/01/2018] [Indexed: 12/21/2022]
Abstract
Tetramethylenedisulfotetramine (TMDT) is a synthetic neurotoxic rodenticide considered a chemical threat agent. Symptoms of intoxication include seizures leading to status epilepticus and death. While children and women have been often the victims, no studies exist investigating the neurotoxic effects of TMDT in developing individuals or females. Thus, we performed such an investigation in developing Sprague-Dawley rats of both sexes in order to identify potential age- or sex-dependent vulnerability to TMDT exposure. Subcutaneous injection was chosen as the preferred route of TMDT exposure. EEG recordings confirmed the seizure activity observed in both postnatal day 15 (P15) and adult rats. Additionally, P15 rats displayed greater sensitivity to TMDT than postnanatal day 25 or adult animals. Seizures were generally more severe in females compared to males. Barrel rotations accompanied convulsions in P25 and adult, but sparsely in P15 rats. Adults developed barrel rolling less frequently than P25 population. Neuronal cell death was not present in 24-h TMDT survivors at any age or sex tested. A seizure rechallenge with flurothyl 7 days following TMDT exposure demonstrated longer latencies to the first clonic seizure but a faster progression into the tonic-clonic seizure in P15 and adult survivors as compared to their vehicle-injected counterparts. In conclusion, the youngest age group represents the most vulnerable population to the TMDT-induced toxidrome. Females appear to be more vulnerable than males. TMDT exposure promotes seizure spread and progression in survivors. These findings will help to establish sex- and age-specific treatment strategies for TMDT-exposed individuals. © 2018 Wiley Periodicals, Inc. Develop Neurobiol 78: 403-416, 2018.
Collapse
Affiliation(s)
- Marcela Lauková
- Department of Public Health, Division of Environmental Health Science, School of Health Sciences and Practice, New York Medical College, Valhalla, New York.,Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Jana Velíšková
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York.,Department of Obstetrics and Gynecology, New York Medical College, Valhalla, New York.,Department of Neurology, New York Medical College, Valhalla, New York
| | - Libor Velíšek
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York.,Department of Neurology, New York Medical College, Valhalla, New York.,Department of Pediatrics, New York Medical College, Valhalla, New York
| | - Michael P Shakarjian
- Department of Public Health, Division of Environmental Health Science, School of Health Sciences and Practice, New York Medical College, Valhalla, New York.,Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York.,Department of Medicine, Division of Pulmonary and Critical Care Medicine, Rutgers-Robert Wood Johnson Medical School, Piscataway, New Jersey
| |
Collapse
|
35
|
Punnakkal P, Dominic D. NMDA Receptor GluN2 Subtypes Control Epileptiform Events in the Hippocampus. Neuromolecular Med 2018; 20:90-96. [PMID: 29335819 DOI: 10.1007/s12017-018-8477-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/03/2018] [Indexed: 12/19/2022]
Abstract
NMDA receptors (NMDARs) play a key role in synaptic plasticity and excitotoxicity. Subtype-specific role of NMDAR in neural disorders is an emerging area. Recent studies have revealed that mutations in NMDARs are a cause for epilepsy. Hippocampus is a known focal point for epilepsy. In hippocampus, expression of the NMDAR subtypes GluN1/GluN2A and GluN1/GluN2B is temporally regulated. However, the pharmacological significance of these subtypes is not well understood in epileptic context/models. To investigate this, epilepsy was induced in hippocampal slices by the application of artificial cerebrospinal fluid that contained high potassium but no magnesium. Epileptiform events (EFEs) were recorded from the CA1 and DG areas of hippocampus with or without subtype-specific antagonists. Irrespective of the age group, CA1 and DG showed epileptiform activity. The NMDAR antagonist AP5 was found to reduce the number of EFEs significantly. However, the application of subtype-specific antagonists (TCN 201 for GluN1/GluN2A and Ro 25-69811 for GluN1/GluN2B) revealed that EFEs had area-specific and temporal components. In slices from neonates, EFEs in CA1 were effectively reduced by Ro 25-69811, but were largely insensitive to TCN 201. In contrast, EFEs in DG were equally sensitive to both of the subtype-specific antagonists. However, the differential sensitivity for the antagonists observed in neonates was absent in later developmental stages. The study provides a functional insight into the NMDAR subtype-dependent contribution of EFEs in hippocampus of young rats, which may have implications in treating childhood epilepsy and avoiding unnecessary side effects of broad spectrum antagonists.
Collapse
Affiliation(s)
- Pradeep Punnakkal
- Molecular Medicine, Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695012, India.
| | - Deity Dominic
- Molecular Medicine, Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695012, India
| |
Collapse
|
36
|
Zhang X, Qu H, Wang Y, Zhao S, Xiao T, Zhao C, Teng W. Aberrant plasticity in the hippocampus after neonatal seizures. Int J Neurosci 2017; 128:384-391. [PMID: 28937832 DOI: 10.1080/00207454.2017.1384380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Xiaoqian Zhang
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Huiling Qu
- Department of Neurology, The People's Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Ying Wang
- Department of Neurology, The First Hospital of Dalian Medical University, Dalian, Liaoning, PR China
| | - Shanshan Zhao
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Ting Xiao
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning, PR China
- Key Laboratory of Immunodermatology, Ministry of Health, Ministry of Education, Shenyang, Liaoning, PR China
| | - Chuansheng Zhao
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Weiyu Teng
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning, PR China
| |
Collapse
|
37
|
Rossini L, Garbelli R, Gnatkovsky V, Didato G, Villani F, Spreafico R, Deleo F, Lo Russo G, Tringali G, Gozzo F, Tassi L, de Curtis M. Seizure activity per se does not induce tissue damage markers in human neocortical focal epilepsy. Ann Neurol 2017; 82:331-341. [PMID: 28749594 DOI: 10.1002/ana.25005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 07/10/2017] [Accepted: 07/17/2017] [Indexed: 12/31/2022]
Abstract
OBJECTIVE The contribution of recurring seizures to the progression of epileptogenesis is debated. Seizure-induced brain damage is not conclusively demonstrated either in humans or in animal models of epilepsy. We evaluated the expression of brain injury biomarkers on postsurgical brain tissue obtained from 20 patients with frequent seizures and a long history of drug-resistant focal epilepsy. METHODS The expression patterns of specific glial, neuronal, and inflammatory molecules were evaluated by immunohistochemistry in the core of type II focal cortical dysplasias (FCD-II), at the FCD boundary (perilesion), and in the adjacent normal-appearing area included in the epileptogenic region. We also analyzed surgical specimens from cryptogenic patients not presenting structural alterations at imaging. RESULTS Astroglial and microglial activation, reduced neuronal density, perivascular CD3-positive T-lymphocyte clustering, and fibrinogen extravasation were demonstrated in the core of FCD-II lesions. No pathological immunoreactivity was observed outside the FCD-II or in cryptogenetic specimens, where the occurrence of interictal and ictal epileptiform activity was confirmed by either stereo-electroencephalography or intraoperative electrocorticography. INTERPRETATION Recurrent seizures do not induce the expression of brain damage markers in nonlesional epileptogenic cortex studied in postsurgical tissue from cryptogenic and FCD patients. This evidence argues against the hypothesis that epileptiform activity per se contributes to focal brain injury, at least in the neocortical epilepsies considered here. Ann Neurol 2017;82:331-341.
Collapse
Affiliation(s)
- Laura Rossini
- Epilepsy Unit, C. Besta Neurological Institute Foundation
| | - Rita Garbelli
- Epilepsy Unit, C. Besta Neurological Institute Foundation
| | | | | | - Flavio Villani
- Epilepsy Unit, C. Besta Neurological Institute Foundation
| | | | | | | | - Giovanni Tringali
- Neurosurgery Unit, C. Besta Neurological Institute Foundation, Milan, Italy
| | | | - Laura Tassi
- C. Munari Epilepsy Surgery Center, Niguarda Hospital
| | | |
Collapse
|
38
|
Saniya K, Patil BG, Chavan MD, Prakash KG, Sailesh KS, Archana R, Johny M. Neuroanatomical Changes in Brain Structures Related to Cognition in Epilepsy: An Update. J Nat Sci Biol Med 2017; 8:139-143. [PMID: 28781476 PMCID: PMC5523517 DOI: 10.4103/0976-9668.210016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Understanding the microanatomical changes in brain structures is necessary for developing innovative therapeutic approaches to prevent/delay the cognitive impairment in epilepsy. We review here the microanatomical changes in the brain structures related to cognition in epilepsy. Here, we have presented the changes in major brain structures related to cognition, which helps the clinicians understand epilepsy more clearly and also helps researchers develop new treatment procedures.
Collapse
Affiliation(s)
- K Saniya
- Department of Anatomy, Azeezia Institute of Medical Sciences, Kollam, Kerala, India
| | - B G Patil
- Department of Anatomy, Shri B. M. Patil Medical College, Bijapur, Karnataka, India
| | - Madhavrao D Chavan
- Department of Pharmacology, Azeezia Institute of Medical Sciences, Kollam, Kerala, India
| | - K G Prakash
- Department of Anatomy, Azeezia Institute of Medical Sciences, Kollam, Kerala, India
| | - Kumar Sai Sailesh
- Department of Physiology, Little Flower Institute of Medical Sciences and Research, Angamaly, Kerala, India
| | - R Archana
- Department of Anatomy, Saveetha Medical College, Saveetha University, Chennai, Tamil Nadu, India
| | - Minu Johny
- Department of Physiology, Little Flower Institute of Medical Sciences and Research, Angamaly, Kerala, India
| |
Collapse
|
39
|
Han W, Song X, He R, Li T, Cheng L, Xie L, Chen H, Jiang L. VEGF regulates hippocampal neurogenesis and reverses cognitive deficits in immature rats after status epilepticus through the VEGF R2 signaling pathway. Epilepsy Behav 2017; 68:159-167. [PMID: 28193596 DOI: 10.1016/j.yebeh.2016.12.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/02/2016] [Accepted: 12/12/2016] [Indexed: 01/17/2023]
Abstract
Epilepsy is the most common chronic disease in children, who exhibit a higher risk for status epilepticus (SE) than adults. Hippocampal neurogenesis is altered by epilepsy, particularly in the immature brain, which may influence cognitive development. Vascular endothelial growth factor (VEGF) represents an attractive target to modulate brain function at the neurovascular interface and is a double-edged sword in seizures. We used the lithium-pilocarpine-induced epilepsy model in immature Sprague-Dawley rats to study the effects of VEGF on hippocampal neurogenesis in the acute phase and on long-term cognitive behaviors in immature rats following status epilepticus (SE). VEGF correlates with cell proliferation in the immature brain after SE. By preprocessing VEGF in the lateral ventricles prior to the induction of the SE model, we found that VEGF increased the proliferation of neural stem cells (NSCs) and promoted the migration of newly generated cells via the VEGF receptor 2 (VEGFR2) signaling pathway. VEGF also inhibited cell loss and reversed the cognitive deficits that accompany SE. Based on our results, VEGF positively contributes to the initial stages of neurogenesis and alleviates cognitive deficits following seizures; moreover, the VEGF/VEGFR2 signaling pathway may provide a novel treatment strategy for epilepsy.
Collapse
Affiliation(s)
- Wei Han
- Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Xiaojie Song
- Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Rong He
- Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Tianyi Li
- Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Li Cheng
- Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Lingling Xie
- Department of Neurology, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Chongqing 400014, China
| | - Hengsheng Chen
- Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Li Jiang
- Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China; Department of Neurology, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Chongqing 400014, China.
| |
Collapse
|
40
|
Cetica V, Chiari S, Mei D, Parrini E, Grisotto L, Marini C, Pucatti D, Ferrari A, Sicca F, Specchio N, Trivisano M, Battaglia D, Contaldo I, Zamponi N, Petrelli C, Granata T, Ragona F, Avanzini G, Guerrini R. Clinical and genetic factors predicting Dravet syndrome in infants with SCN1A mutations. Neurology 2017; 88:1037-1044. [PMID: 28202706 DOI: 10.1212/wnl.0000000000003716] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 12/22/2016] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To explore the prognostic value of initial clinical and mutational findings in infants with SCN1A mutations. METHODS Combining sex, age/fever at first seizure, family history of epilepsy, EEG, and mutation type, we analyzed the accuracy of significant associations in predicting Dravet syndrome vs milder outcomes in 182 mutation carriers ascertained after seizure onset. To assess the diagnostic accuracy of all parameters, we calculated sensitivity, specificity, receiver operating characteristic (ROC) curves, diagnostic odds ratios, and positive and negative predictive values and the accuracy of combined information. We also included in the study demographic and mutational data of the healthy relatives of mutation carrier patients. RESULTS Ninety-seven individuals (48.5%) had Dravet syndrome, 49 (23.8%) had generalized/genetic epilepsy with febrile seizures plus, 30 (14.8%) had febrile seizures, 6 (3.5%) had focal epilepsy, and 18 (8.9%) were healthy relatives. The association study indicated that age at first seizure and frameshift mutations were associated with Dravet syndrome. The risk of Dravet syndrome was 85% in the 0- to 6-month group, 51% in the 6- to 12-month range, and 0% after the 12th month. ROC analysis identified onset within the sixth month as the diagnostic cutoff for progression to Dravet syndrome (sensitivity = 83.3%, specificity = 76.6%). CONCLUSIONS In individuals with SCN1A mutations, age at seizure onset appears to predict outcome better than mutation type. Because outcome is not predetermined by genetic factors only, early recognition and treatment that mitigates prolonged/repeated seizures in the first year of life might also limit the progression to epileptic encephalopathy.
Collapse
Affiliation(s)
- Valentina Cetica
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Sara Chiari
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Davide Mei
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Elena Parrini
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Laura Grisotto
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Carla Marini
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Daniela Pucatti
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Annarita Ferrari
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Federico Sicca
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Nicola Specchio
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Marina Trivisano
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Domenica Battaglia
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Ilaria Contaldo
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Nelia Zamponi
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Cristina Petrelli
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Tiziana Granata
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Francesca Ragona
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Giuliano Avanzini
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Renzo Guerrini
- From the Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (V.C., S.C., D.M., E.P., C.M., D.P., R.G.), Neuroscience Department, A Meyer Children's Hospital, University of Florence; Department of Statistics, Computer Science and Applications (L.G.), University of Florence; Division of Child Neurology and Psychiatry Epilepsy and Clinical Neurophysiology Laboratory (A.F., F.S., R.G.), IRCCS Stella Maris Foundation, Pisa; Department of Neurosciences (N.S., M.T.), Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome; Child Neuropsichiatry Fondazione Policlinico Universitario Agostino Gemelli (D.B., I.C.), Università Cattolica del Sacro Cuore, Rome; Child Neuropsychiatry Unit (N.Z., C.P.), Ospedali Riuniti, Ancona; and Department of Pediatric Neuroscience (T.G., F.R., G.A.), Foundation IRCCS Neurological Institute C. Besta, Milan, Italy.
| |
Collapse
|
41
|
Rodriguez-Alvarez N, Jimenez-Mateos EM, Engel T, Quinlan S, Reschke CR, Conroy RM, Bhattacharya A, Boylan GB, Henshall DC. Effects of P2X7 receptor antagonists on hypoxia-induced neonatal seizures in mice. Neuropharmacology 2017; 116:351-363. [PMID: 28082183 DOI: 10.1016/j.neuropharm.2017.01.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 12/12/2016] [Accepted: 01/08/2017] [Indexed: 12/28/2022]
Abstract
Neonatal seizures are a common consequence of hypoxic/ischemic encephalopathy (HIE). Phenobarbital remains the frontline treatment for neonatal seizures but is often ineffective. The P2X7 receptor (P2X7R) is a cell surface-expressed ionotropic receptor activated by high amounts of ATP which may be released during seizures or as a consequence of tissue injury. Here, we explored the role of the P2X7R in a mouse model of neonatal seizures induced by hypoxia. Exposure of postnatal day 7 (P7) mouse pups to global hypoxia (5% O2 for 15 min) produced electrographically-defined seizures with behavioural correlates that persisted after restitution of normoxia. Expression of the P2X7R showed age-dependent increases in the hippocampus and neocortex of developing mice and was present in human neonatal brain. P2X7R transcript and protein levels were increased 24 h after neonatal hypoxia-induced seizures in mouse pups. EEG recordings in pups determined that injection of the P2X7R antagonist A-438079 (25 mg/kg-1, intraperitoneal) reduced electrographic seizure number, EEG power and spiking during hypoxia. A-438079 did not reduce post-hypoxia seizures. Caspase-1 processing and molecular markers of inflammation and microglia were reduced in A438079-treated mice. Electrographic seizure-suppressive effects were also observed with a second P2X7R antagonist, JNJ-47965567, in the same model. The present study shows hypoxia-induced seizures alter expression of purinergic and neuroinflammatory signalling components and suggest potential applications but also limitations of the P2X7R as a target for the treatment of HIE and other causes of neonatal seizures.
Collapse
Affiliation(s)
- Natalia Rodriguez-Alvarez
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Eva M Jimenez-Mateos
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Tobias Engel
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Sean Quinlan
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Cristina R Reschke
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Ronán M Conroy
- Division of Population Health Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Geraldine B Boylan
- Irish Centre for Fetal and Neonatal Translational Research (INFANT) Cork, Ireland
| | - David C Henshall
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland; Irish Centre for Fetal and Neonatal Translational Research (INFANT) Cork, Ireland.
| |
Collapse
|
42
|
Puig-Lagunes AA, Manzo J, Beltrán-Parrazal L, Morgado-Valle C, Toledo-Cárdenas R, López-Meraz ML. Pentylenetetrazole-induced seizures in developing rats prenatally exposed to valproic acid. PeerJ 2016; 4:e2709. [PMID: 27917314 PMCID: PMC5131616 DOI: 10.7717/peerj.2709] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 10/21/2016] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Epidemiological evidence indicates epilepsy is more common in patients with autism spectrum disorders (ASD) (20-25%) than in the general population. The aim of this project was to analyze seizure susceptibility in developing rats prenatally exposed to valproic acid (VPA) as autism model. METHODS Pregnant females were injected with VPA during the twelfth embryonic day. Seizures were induced in fourteen-days-old rat pups using two models of convulsions: pentylenetetrazole (PTZ) and lithium-pilocarpine (Li-Pilo). RESULTS Two subgroups with different PTZ-induced seizure susceptibility in rats exposed to VPA were found: a high susceptibility (VPA+) (28/42, seizure severity 5) and a low susceptibility (VPA-) (14/42, seizure severity 2). The VPA+ subgroup exhibited an increased duration of the generalized tonic-clonic seizure (GTCS; 45 ± 2.7 min), a higher number of rats showed several GTCS (14/28) and developed status epilepticus (SE) after PTZ injection (19/27) compared with control animals (36.6 ± 1.9 min; 10/39; 15/39, respectively). No differences in seizure severity, latency or duration of SE induced by Li-Pilo were detected between VPA and control animals. DISCUSSION Prenatal VPA modifies the susceptibility to PTZ-induced seizures in developing rats, which may be linked to an alteration in the GABAergic transmission. These findings contribute to a better understanding of the comorbidity between autism and epilepsy.
Collapse
Affiliation(s)
- Angel A. Puig-Lagunes
- Doctorado en Investigaciones Cerebrales, Universidad Veracruzana, Xalapa, Veracruz, Mexico
| | - Jorge Manzo
- Centro de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa, Veracruz, Mexico
| | - Luis Beltrán-Parrazal
- Centro de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa, Veracruz, Mexico
| | | | | | | |
Collapse
|
43
|
Friedman LK, Sharma A, Corcia M, Webster T, Qazi L, Simsovits D, Khalil Y, Hu S, Kantrowitz M, Hong H. Selective inhibition of metabotropic glutamate type 1 alpha receptor (mGluR1α) reduces cell proliferation and migration following status epilepticus in early development. Int J Dev Neurosci 2016; 54:6-21. [PMID: 27530811 DOI: 10.1016/j.ijdevneu.2016.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 08/09/2016] [Accepted: 08/11/2016] [Indexed: 12/16/2022] Open
Abstract
The present study examined whether a single or multiple episode(s) of status epilepticus induced with kainic acid (KA) during the first 3 weeks of postnatal (P) development would aberrantly stimulate proliferation zones that alters migration to potentially injured areas and whether they would be blocked by selective Group I mGluR antagonists. mGluR1α (LY367385) and mGluR5 (MPEP) antagonists were administered 2h following KA-induced status epilepticus and animals were examined after 7days. Proliferating cells of the subventricular zone (SVZ), third ventricle, hippocampus, amygdala cortical complex were analyzed with the proliferative marker, Ki67; and two complementary retrograde dye tracers. Proliferation increased in extrahippocampal limbic structures when KA was administered on P13 or P20 which correlated with number of injured cells at the older age. LY367385 post-treatment caused striking decreases in proliferation in all limbic structures in the presence and absence of injury, whereas a reduction with MPEP was observed only within the amygdala cortical complex (Amg/ERcx) in the presence of multiple seizures (3×KA). After 3×KA and LY367385 post-treatments, diminished co-staining of dye tracers with Ki67 was observed within the Amg/ERcx despite high levels of progenitors marked by the retrograde tracers in this region. This indicates that not only was local proliferation within the SVZ and distant structures inhibited, but also that migration itself was reduced indirectly since there were less cells to migrate from the SVZ. Co-labeling with biomarkers provided evidence for neuronal differentiation suggesting potential aberrant integration may occur in distant locations, and that targeting of mGluR1α receptors may be a potential therapeutic strategy for future development.
Collapse
Affiliation(s)
- L K Friedman
- New York Medical College, Valhalla, NY, United States; New York College of Osteopathic Medicine, Old Westbury, NY, United States.
| | - A Sharma
- New York College of Osteopathic Medicine, Old Westbury, NY, United States
| | - M Corcia
- New York College of Osteopathic Medicine, Old Westbury, NY, United States
| | - T Webster
- New York College of Osteopathic Medicine, Old Westbury, NY, United States
| | - L Qazi
- New York College of Osteopathic Medicine, Old Westbury, NY, United States
| | - D Simsovits
- New York College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Y Khalil
- New York College of Osteopathic Medicine, Old Westbury, NY, United States
| | - S Hu
- New York Medical College, Valhalla, NY, United States
| | - M Kantrowitz
- New York College of Osteopathic Medicine, Old Westbury, NY, United States
| | - H Hong
- New York College of Osteopathic Medicine, Old Westbury, NY, United States
| |
Collapse
|
44
|
A Time and Place for Everything: Early-Life Seizures at Different Developmental Epochs Have Distinct Effects on Adult Hippocampal Structure and Function. Epilepsy Curr 2016; 16:106-7. [DOI: 10.5698/1535-7511-16.2.106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
45
|
Heuser K, Nakken KO, Sandvig I, Taubøll E. [What causes febrile convulsions?]. TIDSSKRIFT FOR DEN NORSKE LEGEFORENING 2016; 136:36-38. [PMID: 26757658 DOI: 10.4045/tidsskr.14.1503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023] Open
Abstract
Febrile convulsions affect 2-5% of children in the age group from 6 months to 5 years. The convulsions seldom have negative consequences for the child's development, but may in rare cases constitute a debut symptom of epilepsy.
Collapse
Affiliation(s)
- Kjell Heuser
- Nevrologisk avdeling Oslo universitetssykehus, Rikshospitalet
| | - Karl O Nakken
- Spesialsykehuset for epilepsi - SSE Oslo universitetssykehus
| | - Inger Sandvig
- Barneavdeling for nevrofag Oslo universitetssykehus, Rikshospitalet
| | - Erik Taubøll
- Nevrologisk avdeling Oslo universitetssykehus, Rikshospitalet
| |
Collapse
|
46
|
Rodriguez-Alvarez N, Jimenez-Mateos EM, Dunleavy M, Waddington JL, Boylan GB, Henshall DC. Effects of hypoxia-induced neonatal seizures on acute hippocampal injury and later-life seizure susceptibility and anxiety-related behavior in mice. Neurobiol Dis 2015; 83:100-14. [PMID: 26341542 DOI: 10.1016/j.nbd.2015.08.023] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 08/06/2015] [Accepted: 08/21/2015] [Indexed: 12/30/2022] Open
Abstract
Seizures are common during the neonatal period, often due to hypoxic-ischemic encephalopathy and may contribute to acute brain injury and the subsequent development of cognitive deficits and childhood epilepsy. Here we explored short- and long-term consequences of neonatal hypoxia-induced seizures in 7 day old C57BL/6J mice. Seizure activity, molecular markers of hypoxia and histological injury were investigated acutely after hypoxia and response to chemoconvulsants and animal behaviour was explored at adulthood. Hypoxia was induced by exposing pups to 5% oxygen for 15 min (global hypoxia). Electrographically defined seizures with behavioral correlates occurred in 95% of these animals and seizures persisted for many minutes after restitution of normoxia. There was minimal morbidity or mortality. Pre- or post-hypoxia injection of phenobarbital (50mg/kg) had limited efficacy at suppressing seizures. The hippocampus from neonatal hypoxia-seizure mice displayed increased expression of vascular endothelial growth factor and the immediate early gene c-fos, minimal histological evidence of cell injury and activation of caspase-3 in scattered neurons. Behavioral analysis of mice five weeks after hypoxia-induced seizures detected novel anxiety-related and other behaviors, while performance in a spatial memory test was similar to controls. Seizure threshold tests with kainic acid at six weeks revealed that mice previously subject to neonatal hypoxia-induced seizures developed earlier, more frequent and longer-duration seizures. This study defines a set of electro-clinical, molecular, pharmacological and behavioral consequences of hypoxia-induced seizures that indicate short- and long-term deleterious outcomes and may be a useful model to investigate the pathophysiology and treatment of neonatal seizures in humans.
Collapse
Affiliation(s)
| | - Eva M Jimenez-Mateos
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Mark Dunleavy
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - John L Waddington
- Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Geraldine B Boylan
- Irish Centre for Fetal and Neonatal Translational Research (INFANT), Cork, Ireland
| | - David C Henshall
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland; Irish Centre for Fetal and Neonatal Translational Research (INFANT), Cork, Ireland.
| |
Collapse
|
47
|
P2X purinoceptors as a link between hyperexcitability and neuroinflammation in status epilepticus. Epilepsy Behav 2015; 49:8-12. [PMID: 25843343 DOI: 10.1016/j.yebeh.2015.02.031] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 12/11/2022]
Abstract
There remains a need for more efficacious treatments for status epilepticus. Prolonged seizures result in the release of ATP from cells which activates the P2 class of ionotropic and metabotropic purinoceptors. The P2X receptors gate depolarizing sodium and calcium entry and are expressed by both neurons and glia throughout the brain, and a number of subtypes are upregulated after status epilepticus. Recent studies have explored the in vivo effects of targeting ATP-gated P2X receptors in preclinical models of status epilepticus, with particular focus on the P2X7 receptor (P2X7R). The P2X7R mediates microglial activation and the release of the proepileptogenic inflammatory cytokine interleukin 1β. The receptor may also directly modulate neurotransmission and gliotransmission and promote the recruitment of immune cells into brain parenchyma. Data from our group and collaborators show that status epilepticus produced by intraamygdala microinjection of kainic acid increases P2X7R expression in the hippocampus and neocortex of mice. Antagonism of the P2X7R in the model reduced seizure severity, microglial activation and interleukin 1β release, and neuronal injury. Coadministration of a P2X7R antagonist with a benzodiazepine also provided seizure suppression in a model of drug-refractory status epilepticus when either treatment alone was minimally effective. More recently, we showed that status epilepticus in immature rats is also reduced by P2X7R antagonism. Together, these findings suggest that P2X receptors may be novel targets for seizure control and interruption of neuroinflammation after status epilepticus. This article is part of a Special Issue entitled "Status Epilepticus".
Collapse
|
48
|
Höller Y, Trinka E. Is There a Relation between EEG-Slow Waves and Memory Dysfunction in Epilepsy? A Critical Appraisal. Front Hum Neurosci 2015; 9:341. [PMID: 26124717 PMCID: PMC4463866 DOI: 10.3389/fnhum.2015.00341] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 05/28/2015] [Indexed: 12/12/2022] Open
Abstract
Is there a relationship between peri-ictal slow waves, loss of consciousness, memory, and slow-wave sleep, in patients with different forms of epilepsy? We hypothesize that mechanisms, which result in peri-ictal slow-wave activity as detected by the electroencephalogram, could negatively affect memory processes. Slow waves (≤4 Hz) can be found in seizures with impairment of consciousness and also occur in focal seizures without impairment of consciousness but with inhibited access to memory functions. Peri-ictal slow waves are regarded as dysfunctional and are probably caused by mechanisms, which are essential to disturb the consolidation of memory entries in these patients. This is in strong contrast to physiological slow-wave activity during deep sleep, which is thought to group memory-consolidating fast oscillatory activity. In patients with epilepsy, slow waves may not only correlate with the peri-ictal clouding of consciousness, but could be the epiphenomenon of mechanisms, which interfere with normal brain function in a wider range. These mechanisms may have transient impacts on memory, such as temporary inhibition of memory systems, altered patterns of hippocampal-neocortical interactions during slow-wave sleep, or disturbed cross-frequency coupling of slow and fast oscillations. In addition, repeated tonic-clonic seizures over the years in uncontrolled chronic epilepsy may cause a progressive cognitive decline. This hypothesis can only be assessed in long-term prospective studies. These studies could disentangle the reversible short-term impacts of seizures, and the impacts of chronic uncontrolled seizures. Chronic uncontrolled seizures lead to irreversible memory impairment. By contrast, short-term impacts do not necessarily lead to a progressive cognitive decline but result in significantly impaired peri-ictal memory performance.
Collapse
Affiliation(s)
- Yvonne Höller
- Department of Neurology, Christian Doppler Medical Centre and Centre for Cognitive Neuroscience, Paracelsus Medical University, Salzburg, Austria
| | - Eugen Trinka
- Department of Neurology, Christian Doppler Medical Centre and Centre for Cognitive Neuroscience, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
49
|
Miller SL, Aroniadou-Anderjaska V, Figueiredo TH, Prager EM, Almeida-Suhett CP, Apland JP, Braga MFM. A rat model of nerve agent exposure applicable to the pediatric population: The anticonvulsant efficacies of atropine and GluK1 antagonists. Toxicol Appl Pharmacol 2015; 284:204-16. [PMID: 25689173 DOI: 10.1016/j.taap.2015.02.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 01/30/2015] [Accepted: 02/07/2015] [Indexed: 12/27/2022]
Abstract
Inhibition of acetylcholinesterase (AChE) after nerve agent exposure induces status epilepticus (SE), which causes brain damage or death. The development of countermeasures appropriate for the pediatric population requires testing of anticonvulsant treatments in immature animals. In the present study, exposure of 21-day-old (P21) rats to different doses of soman, followed by probit analysis, produced an LD50 of 62μg/kg. The onset of behaviorally-observed SE was accompanied by a dramatic decrease in brain AChE activity; rats who did not develop SE had significantly less reduction of AChE activity in the basolateral amygdala than rats who developed SE. Atropine sulfate (ATS) at 2mg/kg, administered 20 min after soman exposure (1.2×LD50), terminated seizures. ATS at 0.5mg/kg, given along with an oxime within 1 min after exposure, allowed testing of anticonvulsants at delayed time-points. The AMPA/GluK1 receptor antagonist LY293558, or the specific GluK1 antagonist UBP302, administered 1h post-exposure, terminated SE. There were no degenerating neurons in soman-exposed P21 rats, but both the amygdala and the hippocampus were smaller than in control rats at 30 and 90days post-exposure; this pathology was not present in rats treated with LY293558. Behavioral deficits present at 30 days post-exposure, were also prevented by LY293558 treatment. Thus, in immature animals, a single injection of atropine is sufficient to halt nerve agent-induced seizures, if administered timely. Testing anticonvulsants at delayed time-points requires early administration of ATS at a low dose, sufficient to counteract only peripheral toxicity. LY293558 administered 1h post-exposure, prevents brain pathology and behavioral deficits.
Collapse
Affiliation(s)
- Steven L Miller
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; Program in Neuroscience, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.
| | - Vassiliki Aroniadou-Anderjaska
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; Department of Psychiatry, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; Program in Neuroscience, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.
| | - Taiza H Figueiredo
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.
| | - Eric M Prager
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; Program in Neuroscience, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.
| | - Camila P Almeida-Suhett
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; Program in Neuroscience, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.
| | - James P Apland
- Neurotoxicology Branch, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010, USA.
| | - Maria F M Braga
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; Department of Psychiatry, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; Program in Neuroscience, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.
| |
Collapse
|
50
|
Puskarjov M, Ahmad F, Khirug S, Sivakumaran S, Kaila K, Blaesse P. BDNF is required for seizure-induced but not developmental up-regulation of KCC2 in the neonatal hippocampus. Neuropharmacology 2015; 88:103-9. [PMID: 25229715 DOI: 10.1016/j.neuropharm.2014.09.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 09/01/2014] [Accepted: 09/04/2014] [Indexed: 01/21/2023]
Abstract
A robust increase in the functional expression of the neuronal K-Cl cotransporter KCC2 during CNS development is necessary for the emergence of hyperpolarizing ionotropic GABAergic transmission. BDNF-TrkB signaling has been implicated in the developmental up-regulation of KCC2 and, in mature animals, in fast activity-dependent down-regulation of KCC2 function following seizures and trauma. In contrast to the decrease in KCC2 expression observed in the adult hippocampus following trauma, seizures in the neonate trigger a TrkB-dependent up-regulation of neuronal Cl(-) extrusion capacity associated with enhanced surface expression of KCC2. Here, we show that this effect is transient, and impaired in the hippocampus of Bdnf(-/-) mice. Notably, however, a complete absence of BDNF does not compromise the increase in KCC2 protein or K-Cl transport functionality during neuronal development. Furthermore, we present data indicating that the functional up-regulation of KCC2 by neonatal seizures is temporally limited by calpain activity.
Collapse
Affiliation(s)
- Martin Puskarjov
- Department of Biosciences and Neuroscience Center, University of Helsinki, FI-00014 Helsinki, Finland
| | - Faraz Ahmad
- Department of Biosciences and Neuroscience Center, University of Helsinki, FI-00014 Helsinki, Finland
| | - Stanislav Khirug
- Department of Biosciences and Neuroscience Center, University of Helsinki, FI-00014 Helsinki, Finland
| | - Sudhir Sivakumaran
- Department of Biosciences and Neuroscience Center, University of Helsinki, FI-00014 Helsinki, Finland
| | - Kai Kaila
- Department of Biosciences and Neuroscience Center, University of Helsinki, FI-00014 Helsinki, Finland
| | - Peter Blaesse
- Department of Biosciences and Neuroscience Center, University of Helsinki, FI-00014 Helsinki, Finland.
| |
Collapse
|