1
|
De Meulemeester AS, Reid C, Auvin S, Carlen PL, Andrew CJ, Szlendak R, Di Sapia R, Moshé SL, Sankar R, O’Brien TJ, Baulac S, Henshall DC, Akman Ö, Galanopoulou AS. WONOEP appraisal: Modeling early onset epilepsies. Epilepsia 2024; 65:2553-2566. [PMID: 39042520 PMCID: PMC11534511 DOI: 10.1111/epi.18063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/25/2024]
Abstract
Epilepsy has a peak incidence during the neonatal to early childhood period. These early onset epilepsies may be severe conditions frequently associated with comorbidities such as developmental deficits and intellectual disability and, in a significant percentage of patients, may be medication-resistant. The use of adult rodent models in the exploration of mechanisms and treatments for early life epilepsies is challenging, as it ignores significant age-specific developmental differences. More recently, models developed in immature animals, such as rodent pups, or in three-dimensional organoids may more closely model aspects of the immature brain and could result in more translatable findings. Although models are not perfect, they may offer a more controlled screening platform in studies of mechanisms and treatments, which cannot be done in pediatric patient cohorts. On the other hand, more simplified models with higher throughput capacities are required to deal with the large number of epilepsy candidate genes and the need for new treatment options. Therefore, a combination of different modeling approaches will be beneficial in addressing the unmet needs of pediatric epilepsy patients. In this review, we summarize the discussions on this topic that occurred during the XVI Workshop on Neurobiology of Epilepsy, organized in 2022 by the Neurobiology Commission of the International League Against Epilepsy. We provide an overview of selected models of early onset epilepsies, discussing their advantages and disadvantages. Heterologous expression models provide initial functional insights, and zebrafish, rodent models, and brain organoids present increasingly complex platforms for modeling and validating epilepsy-related phenomena. Together, these models offer valuable insights into early onset epilepsies and accelerate hypothesis generation and therapy discovery.
Collapse
Affiliation(s)
- Ann-Sofie De Meulemeester
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, Inserm, CNRS, Paris, France
- Laboratory for Molecular Biodiscovery, KU Leuven, Leuven, Belgium
| | - Christopher Reid
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia
- Department of Medicine, Epilepsy Research Centre, University of Melbourne, Austin Health, Heidelberg, VIC 3084, Australia
| | - Stéphane Auvin
- AP-HP, Robert-Debré University Hospital, Pediatric Neurology Department, CRMR épilepsies rares, EpiCARE member, Paris, France
- Université Paris Cité, INSERM NeuroDiderot, Paris, France
- Institut Universitaire de France (IUF), Paris, France
| | - Peter L. Carlen
- Krembil Research Institute, 60 Leonard Ave, 7KDT430, Toronto, ON, Canada M5T 0S8
- Institute of Biomedical Engineering, University of Toronto, Rosebrugh Building, 164 College St Room 407, Toronto, ON, Canada, M5S 3G9
- Departments of Medicine and Physiology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Toronto, ON, Canada, M5S 1A8
| | - Cole J. Andrew
- MGH Epilepsy Service, Division of Clinical Neurophysiology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Roza Szlendak
- Department of Medical Genetics, Institute of Mother and Child, Kasprzaka 17A, 01-211 Warsaw, Poland
- Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Rossella Di Sapia
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Solomon L. Moshé
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York
- Isabelle Rapin Division of Child Neurology, Albert Einstein College of Medicine, Bronx, New York, USA
- Dominick P Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Raman Sankar
- Department of Neurology and Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Terence J. O’Brien
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Royal Parade, Parkville, Victoria 3050, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, 3004 Victoria, Australia
- Department of Neurology, The Alfred Hospital, Commercial Road, Melbourne, Victoria, 3004 Victoria, Australia
| | - Stéphanie Baulac
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, Inserm, CNRS, Paris, France
| | - David C. Henshall
- FutureNeuro SFI Research Centre, RCSI University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, D02 YN7
| | - Özlem Akman
- Demiroglu Bilim University, Faculty of Medicine Department of Physiology, Istanbul, Turkey
| | - Aristea S. Galanopoulou
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York
- Isabelle Rapin Division of Child Neurology, Albert Einstein College of Medicine, Bronx, New York, USA
- Dominick P Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
2
|
Rana M, Vega Gonzales-Portillo JD, Hahn C, Dutt M, Sanchez-Fernandez I, Jonas R, Douglass L, Torres AR. Current Evidence: Seizures in Extremely Low Gestational Age Newborns (ELGANs). J Child Neurol 2024; 39:285-291. [PMID: 38836290 DOI: 10.1177/08830738241259052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Extremely low gestational age newborns (ELGANs) are born at or below 28 weeks of gestational age. Despite improved obstetric care, the incidence of preterm birth continues to rise in advanced countries. Preterm birth remains a major cause of infant mortality, and for infants who survive, neonatal seizures are a significant predictor of later neurologic morbidity. However, little is known about risk factors for neonatal seizures in ELGANs. Understanding the association between neonatal seizures and the development of other neurologic disorders is important given the increasing prevalence of ELGANs. Identifying risk factors that contribute to the development of neonatal seizures in ELGANs may offer insights into novel mechanisms of epileptogenesis in the developing brain and improvements in the prevention or treatment of seizures in preterm infants, including ELGANs. In this literature review, we outline the limitations of epidemiologic studies of neonatal seizures in ELGANs and discuss risk factors for neonatal seizures.
Collapse
Affiliation(s)
- Mandeep Rana
- Division of Pediatric Neurology and Sleep Medicine, Department of Pediatrics, Boston University School of Medicine, Boston Medical Center, Boston, MA, USA
| | - Juan Diego Vega Gonzales-Portillo
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Cecil Hahn
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Monideep Dutt
- Division of Pediatric Neurology, Children's Healthcare of Atlanta: Pediatric Institute, Emory University, Atlanta, GA, USA
| | - Ivan Sanchez-Fernandez
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Rinat Jonas
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Laurie Douglass
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Alcy R Torres
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
3
|
Franco R, Lillo A, Rivas-Santisteban R, Reyes-Resina I, Navarro G. Microglial Adenosine Receptors: From Preconditioning to Modulating the M1/M2 Balance in Activated Cells. Cells 2021; 10:1124. [PMID: 34066933 PMCID: PMC8148598 DOI: 10.3390/cells10051124] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/19/2021] [Accepted: 05/01/2021] [Indexed: 12/14/2022] Open
Abstract
Neuronal survival depends on the glia, that is, on the astroglial and microglial support. Neurons die and microglia are activated not only in neurodegenerative diseases but also in physiological aging. Activated microglia, once considered harmful, express two main phenotypes: the pro-inflammatory or M1, and the neuroprotective or M2. When neuroinflammation, i.e., microglial activation occurs, it is important to achieve a good M1/M2 balance, i.e., at some point M1 microglia must be skewed into M2 cells to impede chronic inflammation and to afford neuronal survival. G protein-coupled receptors in general and adenosine receptors in particular are potential targets for increasing the number of M2 cells. This article describes the mechanisms underlying microglial activation and analyzes whether these cells exposed to a first damaging event may be ready to be preconditioned to better react to exposure to more damaging events. Adenosine receptors are relevant due to their participation in preconditioning. They can also be overexpressed in activated microglial cells. The potential of adenosine receptors and complexes formed by adenosine receptors and cannabinoids as therapeutic targets to provide microglia-mediated neuroprotection is here discussed.
Collapse
Affiliation(s)
- Rafael Franco
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain;
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Alejandro Lillo
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain;
| | - Rafael Rivas-Santisteban
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain;
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Irene Reyes-Resina
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain;
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Gemma Navarro
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain;
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain;
| |
Collapse
|
4
|
Serotonin Pretreatment Abolishes Sex-specific NMDA-induced Seizure Behavior in Developing Rats. Neuroscience 2021; 463:184-196. [PMID: 33838289 DOI: 10.1016/j.neuroscience.2021.03.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 11/24/2022]
Abstract
Neuronal excitability and susceptibility to excitotoxic damage can be sex-specific, with neurons from males usually being more 'easily excitable' compared to neurons from females, especially during development. Increased excitability at an individual neuronal level can lead to the formation of hyperexcitable neuronal networks, which, consequently can make the brain more seizure prone. Both animal and clinical data suggest that males experience more frequent and severe seizures than do females. Serotonin (5-hydroxytryptamine; 5-HT) can mediate neuronal excitability and seizure behavior, often serving as an anticonvulsant. Importantly, 5-HT signaling during parts of the perinatal period is sexually dimorphic. Sex differences during development have been reported in both serotonin levels and receptor type (excitatory vs. inhibitory) expression in a manner that may leave the male brain more vulnerable to over-excitation. Thus, we aimed to determine if the anticonvulsant effects of 5-HT were sex- and/or age-dependent in juvenile animals. We report a baseline sex difference in N-methyl-d-aspartate (NMDA)-induced seizure behavior and hippocampal neuronal loss, with postnatal day (PND) 14 males exhibiting more severe seizure behavior compared to females. Pretreatment with the general 5-HT receptor agonist 5-methoxytryptamine (5-MT) abolishes baseline sex differences, providing an anticonvulsant effect for males only. These sex differences appear to be at least in part organized by testosterone, as females given neonatal androgen exhibit a seizure behavior profile in between that of males and females.
Collapse
|
5
|
Cho KHT, Fraser M, Wassink G, Dhillon SJ, Davidson JO, Dean JM, Gunn AJ, Bennet L. TLR7 agonist modulation of postasphyxial neurophysiological and cardiovascular adaptations in preterm fetal sheep. Am J Physiol Regul Integr Comp Physiol 2020; 318:R369-R378. [PMID: 31913689 DOI: 10.1152/ajpregu.00295.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Activation of Toll-like receptors (TLRs) after hypoxic-ischemic brain injury can exacerbate injury but also alleviate cell loss, as recently demonstrated with the TLR7 agonist Gardiquimod (GDQ). However, TLR agonists also modulate vascular function and neuronal excitability. Thus, we examined the effects of TLR7 activation with GDQ on cardiovascular function and seizures after asphyxia in preterm fetal sheep at 0.7 gestation (104 days, term ∼147 days). Fetuses received sham asphyxia or asphyxia induced by umbilical cord occlusion for 25 min or asphyxia followed by a continuous intracerebroventricular infusion of 3.34 mg of GDQ from 1 to 4 h after asphyxia. Fetuses were monitored continuously for 72 h postasphyxia. GDQ treatment was associated with sustained, moderate hypertension for 72 h (P < 0.05), with a transient increase in heart rate. Electroencephalographic (EEG) power was suppressed for the entire postasphyxial period in both groups, whereas EEG spectral edge transiently increased during the GDQ infusion compared with asphyxia alone (P < 0.05), with higher β- and lower δ-EEG frequencies (P < 0.05). This increase in EEG frequency was not related to epileptiform activity. After the GDQ infusion, there was earlier onset of high-amplitude stereotypic evolving seizures, with increased numbers of seizures and seizure burden (P < 0.05). Hemodynamic function and seizure activity are important indices of preterm wellbeing. These data highlight the importance of physiological monitoring during preclinical testing of potential neuroprotective strategies.
Collapse
Affiliation(s)
- Kenta H T Cho
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Mhoyra Fraser
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Guido Wassink
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | | | - Joanne O Davidson
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Justin M Dean
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- Department of Physiology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
6
|
Petrenko V, van de Looij Y, Mihhailova J, Salmon P, Hüppi PS, Sizonenko SV, Kiss JZ. Multimodal MRI Imaging of Apoptosis-Triggered Microstructural Alterations in the Postnatal Cerebral Cortex. Cereb Cortex 2019; 28:949-962. [PMID: 28158611 DOI: 10.1093/cercor/bhw420] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Indexed: 12/17/2022] Open
Abstract
Prematurely born children often develop neurodevelopmental delay that has been correlated with reduced growth and microstructural alterations in the cerebral cortex. Much research has focused on apoptotic neuronal cell death as a key neuropathological features following preterm brain injuries. How scattered apoptotic death of neurons may contribute to microstructural alterations remains unknown. The present study investigated in a rat model the effects of targeted neuronal apoptosis on cortical microstructure using in vivo MRI imaging combined with neuronal reconstruction and histological analysis. We describe that mild, targeted death of layer IV neurons in the developing rat cortex induces MRI-defined metabolic and microstructural alterations including increased cortical fractional anisotropy. Delayed architectural modifications in cortical gray matter and myelin abnormalities in the subcortical white matter such as hypomyelination and microglia activation follow the acute phase of neuronal death and axonal degeneration. These results establish the link between mild cortical apoptosis and MRI-defined microstructure changes that are reminiscent to those previously observed in preterm babies.
Collapse
Affiliation(s)
- Volodymyr Petrenko
- Department of Neurosciences, University of Geneva Medical School, Geneva, Switzerland
| | - Yohan van de Looij
- Division of Child Growth & Development, Department of Pediatrics, University of Geneva, Geneva, Switzerland.,Laboratory for Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Jevgenia Mihhailova
- Department of Neurosciences, University of Geneva Medical School, Geneva, Switzerland
| | - Patrick Salmon
- Department of Neurosciences, University of Geneva Medical School, Geneva, Switzerland
| | - Petra S Hüppi
- Division of Child Growth & Development, Department of Pediatrics, University of Geneva, Geneva, Switzerland
| | - Stéphane V Sizonenko
- Division of Child Growth & Development, Department of Pediatrics, University of Geneva, Geneva, Switzerland
| | - Jozsef Z Kiss
- Department of Neurosciences, University of Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
7
|
Sugimoto J, Tanaka M, Sugiyama K, Ito Y, Aizawa H, Soma M, Shimizu T, Mitani A, Tanaka K. Region-specific deletions of the glutamate transporter GLT1 differentially affect seizure activity and neurodegeneration in mice. Glia 2017; 66:777-788. [PMID: 29214672 DOI: 10.1002/glia.23281] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/21/2017] [Accepted: 11/24/2017] [Indexed: 11/06/2022]
Abstract
Glial glutamate transporter GLT1 plays a key role in the maintenance of extracellular glutamate homeostasis. Recent human genetic studies have suggested that de novo mutations in GLT1 (EAAT2) cause early-onset epilepsy with multiple seizure types. Consistent with these findings, global GLT1 null mice show lethal spontaneous seizures. The consequences of GLT1 dysfunction vary between different brain regions, suggesting that the role of GLT1 dysfunction in epilepsy may also vary with brain regions. In this study, we generated region-specific GLT1 knockout mice by crossing floxed-GLT1 mice with mice that express the Cre recombinase in a particular domain of the ventricular zone. Selective deletion of GLT1 in the diencephalon, brainstem and spinal cord is sufficient to reproduce the phenotypes (excess mortality, decreased body weight, and lethal spontaneous seizure) of the global GLT1 null mice. By contrast, dorsal forebrain-specific GLT1 knockout mice showed nonlethal complex seizures including myoclonic jerks, hyperkinetic running, spasm and clonic convulsion via the activation of NMDA receptors during a limited period from P12 to P14 and selective neuronal death in cortical layer II/III and the hippocampus. Thus, GLT1 dysfunction in the dorsal forebrain is involved in the pathogenesis of infantile epilepsy and GLT1 in the diencephalon, brainstem and spinal cord may play a critical role in preventing seizure-induced sudden death.
Collapse
Affiliation(s)
- Junya Sugimoto
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Moeko Tanaka
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kaori Sugiyama
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Yukiko Ito
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Hidenori Aizawa
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Miho Soma
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Tomoko Shimizu
- Laboratory of Physiology, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Akira Mitani
- Laboratory of Physiology, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Kohichi Tanaka
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.,Center for Brain Integration Research (CBIR), TMDU, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| |
Collapse
|
8
|
Folbergrová J, Ješina P, Kubová H, Druga R, Otáhal J. Status Epilepticus in Immature Rats Is Associated with Oxidative Stress and Mitochondrial Dysfunction. Front Cell Neurosci 2016; 10:136. [PMID: 27303267 PMCID: PMC4881382 DOI: 10.3389/fncel.2016.00136] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 05/10/2016] [Indexed: 01/01/2023] Open
Abstract
Epilepsy is a neurologic disorder, particularly frequent in infants and children where it can lead to serious consequences later in life. Oxidative stress and mitochondrial dysfunction are implicated in the pathogenesis of many neurological disorders including epilepsy in adults. However, their role in immature epileptic brain is unclear since there have been two contrary opinions: oxidative stress is age-dependent and does not occur in immature brain during status epilepticus (SE) and, on the other hand, evidence of oxidative stress in immature brain during a specific model of SE. To solve this dilemma, we have decided to investigate oxidative stress following SE induced in immature 12-day-old rats by three substances with a different mechanism of action, namely 4-aminopyridine, LiCl-pilocarpine or kainic acid. Fluoro-Jade-B staining revealed mild brain damage especially in hippocampus and thalamus in each of the tested models. Decrease of glucose and glycogen with parallel rises of lactate clearly indicate high rate of glycolysis, which was apparently not sufficient in 4-AP and Li-Pilo status, as evident from the decreases of PCr levels. Hydroethidium method revealed significantly higher levels of superoxide anion (by ∼60%) in the hippocampus, cerebral cortex and thalamus of immature rats during status. SE lead to mitochondrial dysfunction with a specific pronounced decrease of complex I activity that persisted for a long period of survival. Complexes II and IV activities remained in the control range. Antioxidant treatment with SOD mimetic MnTMPYP or peroxynitrite scavenger FeTPPS significantly attenuated oxidative stress and inhibition of complex I activity. These findings bring evidence that oxidative stress and mitochondrial dysfunction are age and model independent, and may thus be considered a general phenomenon. They can have a clinical relevance for a novel approach to the treatment of epilepsy, allowing to target the mechanisms which play a crucial or additive role in the pathogenesis of epilepsies in infants and children.
Collapse
|
9
|
Jensen FE. Developmental factors in the pathogenesis of neonatal seizures. JOURNAL OF PEDIATRIC NEUROLOGY 2015; 7:5-12. [PMID: 20191097 DOI: 10.3233/jpn-2009-0270] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neonatal seizures are inherently different from seizures in the child and the adult. The phenotype, often exhibiting electroclinical dissociation, is unique: neonatal seizures can be refractory to antiepileptic drugs otherwise effect for older patients. Recent experimental and human-based research reveals that the mechanism of neonatal seizures, as well as their long-term sequelae on later brain development, appears to involve a large number of age-specific factors. These observations help explain the resistance of neonatal seizures to conventional therapy as well as identify potential areas of risk for later neurocognitive development. Emerging targets from this research may suggest new therapies for this unique population of patients.
Collapse
Affiliation(s)
- Frances E Jensen
- Department of Neurology, Children's Hospital, and Program in Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Pourcyrous M, Basuroy S, Tcheranova D, Arheart KL, Elabiad MT, Leffler CW, Parfenova H. Brain-derived circulating endothelial cells in peripheral blood of newborn infants with seizures: a potential biomarker for cerebrovascular injury. Physiol Rep 2015; 3:3/3/e12345. [PMID: 25804265 PMCID: PMC4393173 DOI: 10.14814/phy2.12345] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Neonatal seizures have been associated with cerebrovascular endothelial injury and neurological disabilities. In a piglet model, the long-term loss of endothelial regulation of cerebral blood flow coincides with the surge of brain-derived circulating endothelial cells (BCECs) in blood. We hypothesized that BCECs could serve as a noninvasive biomarker of cerebrovascular injury in neonates with seizures. In a prospective pilot feasibility study, we enrolled newborn infants with confirmed diagnoses of perinatal asphyxia and intraventricular hemorrhage (IVH); both are commonly associated with seizures. Infants without clinical evidence of cerebrovascular injuries were representative of the control group. BCECs were detected in the CD45-negative fraction of peripheral blood mononuclear cells by coexpression of CD31 (common endothelial antigen) and GLUT1 (blood-brain barrier antigen) via automated flow cytometry method. In Infants with asphyxia (n = 12) and those with IVH grade III/IV (n = 5), the BCEC levels were 9.9 ± 0.9% and 19.0 ± 2.0%, respectively. These levels were significantly higher than the control group (n = 27), 0.9 ± 0.2%, P < 0.001. BCECs in infants with cerebrovascular insults with documented clinical seizures (n = 10; 16.8 ± 1.3%) were significantly higher than infants with cerebrovascular insults with subclinical or no seizures (n = 7; 9.5 ± 1.2%); P < 0.001. BCEC levels decreased with seizure control. BCECs levels were elevated in infants with seizures caused by severe IVH and perinatal asphyxia. We suggest that monitoring BCEC levels in peripheral blood can potentially offer a biological marker that reflects cerebrovascular insult and recovery. Further studies with a larger number of patients are required to support these findings.
Collapse
Affiliation(s)
- Massroor Pourcyrous
- Department of Pediatrics, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee Department of Physiology, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee Department of Neuroscience Institute, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee Department of Obstetrics and Gynecology, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee
| | - Shyamali Basuroy
- Department of Physiology, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee
| | - Dilyara Tcheranova
- Department of Physiology, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee
| | - Kristopher L Arheart
- Division of Biostatistics and Pediatrics, Department of Public Health Sciences, Miller School of Medicine University of Miami, Coral Gables, Florida
| | - Mohamad T Elabiad
- Department of Pediatrics, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee
| | - Charles W Leffler
- Department of Pediatrics, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee Department of Physiology, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee Department of Neuroscience Institute, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee
| | - Helena Parfenova
- Department of Physiology, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee Department of Neuroscience Institute, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee
| |
Collapse
|
11
|
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
|
12
|
Tan CC, Zhang JG, Tan MS, Chen H, Meng DW, Jiang T, Meng XF, Li Y, Sun Z, Li MM, Yu JT, Tan L. NLRP1 inflammasome is activated in patients with medial temporal lobe epilepsy and contributes to neuronal pyroptosis in amygdala kindling-induced rat model. J Neuroinflammation 2015; 12:18. [PMID: 25626361 PMCID: PMC4314732 DOI: 10.1186/s12974-014-0233-0] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 12/28/2014] [Indexed: 12/19/2022] Open
Abstract
Background Temporal lobe epilepsy (TLE) is often characterized pathologically by severe neuronal loss in the hippocampus. Understanding the mechanisms of neuron death is key to preventing the neurodegeneration associated with TLE. However, the involvement of neuronal loss to the epileptogenic process has yet to be fully determined. Recent studies have shown that the activation of NLRP1 can generate a functional caspase-1-containing inflammasome in vivo to drive the proinflammatory programmed cell death termed ‘pyroptosis’, which has a key role in the pathogenesis of neurological disorders. To the best of our knowledge, there are no reported studies that performed detailed identification and validation of NLRP1 inflammasome during the epileptogenic process. Methods We first compared expression of NLRP1 and caspase-1 in resected hippocampus from patients with intractable mesial temporal lobe epilepsy (mTLE) with that of matched control samples. To further examine whether the activation of NLRP1 inflammasome contributes to neuronal pyroptosis, we employed a nonviral strategy to knock down the expression of NLRP1 and caspase-1 in the amygdala kindling-induced rat model. Proinflammatory cytokines levels and hippocampal neuronal loss were evaluated after 6 weeks of treatment in these NLRP1 or caspase-1 deficiency TLE rats. Results Western blotting detected upregulated NLRP1 levels and active caspase-1 in mTLE patients in comparison to those levels seen in the controls, suggesting a role for this inflammasome in mTLE. Moreover, we employed direct in vivo infusion of nonviral small interfering RNA to knockdown NLRP1 or caspase-1 in the amygdala kindling-induced rat model, and discovered that these NLRP1 or caspase-1 silencing rats resulted in significantly reduced neuronal pyroptosis. Conclusions Our data suggest that NLRP1/caspase-1 signaling participates in the seizure-induced degenerative process in humans and in the animal model of TLE and points to the silencing of NLRP1 inflammasome as a promising strategy for TLE therapy. Electronic supplementary material The online version of this article (doi:10.1186/s12974-014-0233-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chen-Chen Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, 266071, China.
| | - Jian-Guo Zhang
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, No.6, Tiantan Xili, Beijing, 100050, China. .,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.6, Tiantan Xili, Beijing, 100050, China.
| | - Meng-Shan Tan
- College of Medicine and Pharmaceutics, Ocean University of China, No.5 Yushan Road, Qingdao, 266003, China.
| | - Hua Chen
- Department of Pathology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, 266071, China.
| | - Da-Wei Meng
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, No.6, Tiantan Xili, Beijing, 100050, China. .,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.6, Tiantan Xili, Beijing, 100050, China.
| | - Teng Jiang
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, No.5 Donghai Middle Road, Qingdao, 266071, China.
| | - Xiang-Fei Meng
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, 266071, China.
| | - Ying Li
- Department of Pathology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, 266071, China.
| | - Zhen Sun
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, 266071, China.
| | - Meng-Meng Li
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, 266071, China.
| | - Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, 266071, China. .,College of Medicine and Pharmaceutics, Ocean University of China, No.5 Yushan Road, Qingdao, 266003, China. .,Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, No.5 Donghai Middle Road, Qingdao, 266071, China. .,Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, 94158, USA.
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, 266071, China. .,College of Medicine and Pharmaceutics, Ocean University of China, No.5 Yushan Road, Qingdao, 266003, China. .,Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, No.5 Donghai Middle Road, Qingdao, 266071, China.
| |
Collapse
|
13
|
Abstract
Glutamate is the main excitatory neurotransmitter in the brain and ionotropic glutamate receptors mediate the majority of excitatory neurotransmission (Dingeldine et al. 1999). The high level of glutamatergic excitation allows the neonatal brain (the 2(nd) postnatal week in rat) to develop quickly but it also makes it highly prone to age-specific seizures that can cause lifelong neurological and cognitive disability (Haut et al. 2004). There are three types of ionotropic glutamate receptors (ligand-gated ion channels) named according to their prototypic agonists: N-methyl-D-aspartate (NMDA), 2-amino-3-(3-hydroxy-5-methyl-isoxazol-4-yl) propanoic acid (AMPA) and kainate (KA). During early stages of postnatal development glutamate receptors of NMDA and AMPA type undergo intensive functional changes owing to modifications in their subunit composition (Carter et al. 1988, Watanabe et al. 1992, Monyer et al. 1994, Wenzel et al. 1997, Sun et al. 1998, Lilliu et al. 2001, Kumar et al. 2002, Matsuda et al. 2002, Wee et al. 2008, Henson et al. 2010, Pachernegg et al. 2012, Paoletti et al. 2013). Participation and role of these receptors in mechanisms of seizures and epilepsy became one of the main targets of intensive investigation (De Sarro et al. 2005, Di Maio et al. 2012, Rektor 2013). LiCl/Pilocarpine (LiCl/Pilo) induced status epilepticus is a model of severe seizures resulting in development temporal lobe epilepsy (TLE). This review will consider developmental changes and contribution of NMDA and AMPA receptors in LiCl/Pilo model of status epilepticus in immature rats.
Collapse
Affiliation(s)
- E Szczurowska
- Institute of Physiology AS CR, Prague, Czech Republic.
| | | |
Collapse
|
14
|
Wang SQ, Li XJ, Zhou S, Sun DX, Wang H, Cheng PF, Ma XR, Liu L, Liu JX, Wang FF, Liang YF, Wu JM. Intervention effects of ganoderma lucidum spores on epileptiform discharge hippocampal neurons and expression of neurotrophin-4 and N-cadherin. PLoS One 2013; 8:e61687. [PMID: 23637882 PMCID: PMC3634853 DOI: 10.1371/journal.pone.0061687] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 03/13/2013] [Indexed: 01/15/2023] Open
Abstract
Epilepsy can cause cerebral transient dysfunctions. Ganoderma lucidum spores (GLS), a traditional Chinese medicinal herb, has shown some antiepileptic effects in our previous studies. This was the first study of the effects of GLS on cultured primary hippocampal neurons, treated with Mg2+ free medium. This in vitro model of epileptiform discharge hippocampal neurons allowed us to investigate the anti-epileptic effects and mechanism of GLS activity. Primary hippocampal neurons from <1 day old rats were cultured and their morphologies observed under fluorescence microscope. Neurons were confirmed by immunofluorescent staining of neuron specific enolase (NSE). Sterile method for GLS generation was investigated and serial dilutions of GLS were used to test the maximum non-toxic concentration of GLS on hippocampal neurons. The optimized concentration of GLS of 0.122 mg/ml was identified and used for subsequent analysis. Using the in vitro model, hippocampal neurons were divided into 4 groups for subsequent treatment i) control, ii) model (incubated with Mg2+ free medium for 3 hours), iii) GLS group I (incubated with Mg2+ free medium containing GLS for 3 hours and replaced with normal medium and incubated for 6 hours) and iv) GLS group II (neurons incubated with Mg2+ free medium for 3 hours then replaced with a normal medium containing GLS for 6 hours). Neurotrophin-4 and N-Cadherin protein expression were detected using Western blot. The results showed that the number of normal hippocampal neurons increased and the morphologies of hippocampal neurons were well preserved after GLS treatment. Furthermore, the expression of neurotrophin-4 was significantly increased while the expression of N-Cadherin was decreased in the GLS treated group compared with the model group. This data indicates that GLS may protect hippocampal neurons by promoting neurotrophin-4 expression and inhibiting N-Cadherin expression.
Collapse
Affiliation(s)
- Shu-Qiu Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang Province, P. R. China
- Children Neural Rehabilitation Laboratory of Jiamusi University, Jiamusi, Heilongjiang Province, P. R. China
- * E-mail:
| | - Xiao-Jie Li
- School of Rehabilitation Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang Province, P. R. China
- Children Neural Rehabilitation Laboratory of Jiamusi University, Jiamusi, Heilongjiang Province, P. R. China
| | - Shaobo Zhou
- Department of Pathophysiology, School of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang Province, P. R. China
- Department of Life Science, Institute of Biomedical and Environmental Science and Technology, University of Bedfordshire, Luton, United Kingdom
| | - Di-Xiang Sun
- Department of Pathophysiology, School of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang Province, P. R. China
| | - Hui Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang Province, P. R. China
| | - Peng-Fei Cheng
- Department of Pathophysiology, School of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang Province, P. R. China
| | - Xiao-Ru Ma
- Department of Pathophysiology, School of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang Province, P. R. China
| | - Lei Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang Province, P. R. China
| | - Jun-Xing Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang Province, P. R. China
| | - Fang-Fang Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang Province, P. R. China
| | - Yan-Feng Liang
- Department of Pathophysiology, School of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang Province, P. R. China
| | - Jia-Mei Wu
- Department of Pathophysiology, School of Basic Medical Sciences, Jiamusi University, Jiamusi, Heilongjiang Province, P. R. China
| |
Collapse
|
15
|
Abstract
Benign epilepsy with centrotemporal spikes, early-onset childhood occipital epilepsy (Panayiotopoulos syndrome [PS]) and late-onset childhood occipital epilepsy (Gastaut type [LOCE-G]) are the principal pediatric focal epilepsy syndromes. They share major common characteristics: the appearance and resolution of electroclinical features are age related, there is a strong genetic predisposition, the clinical course is often mild with infrequent and easy to control seizures, interictal epileptiform activity is disproportionately abundant when compared with the clinical correlate, and tends to potentiate and generalize during sleep. In this review, we outline the relevant pathophysiology underlying this electroclinical spectrum. Then, the initial description of individual syndromes is followed by a summary of overlapping features and intermediate presentations that question the boundaries between these entities and provide the basis for the concept of a childhood seizure susceptibility syndrome. Additionally, we outline the main features of the related epileptic encephalopathies. An outlook on potential future lines of research completes this review.
Collapse
|
16
|
Low-intensity physical training recovers object recognition memory impairment in rats after early-life induced Status epilepticus. Int J Dev Neurosci 2013; 31:196-201. [PMID: 23318691 DOI: 10.1016/j.ijdevneu.2013.01.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 01/02/2013] [Accepted: 01/03/2013] [Indexed: 12/18/2022] Open
Abstract
When it occurs early in life, Status epilepticus (SE) can cause behavioural and cognitive impairments in adulthood. Here, we evaluated the putative benefits of low-intensity treadmill training on long-standing cognitive impairment in rats submitted to SE early in life. Wistar rats were submitted to LiCl-pilocarpine-induced SE at P16. Animals from the trained group underwent a low-intensity treadmill protocol for 5 days per week for 4 weeks. At adulthood, rats subjected to early-life SE displayed impairment in long-term memory in an object recognition task, while the training protocol completely reversed this deficit. This result was associated with neither locomotor alterations nor changes in emotional behaviour; there were no differences between groups in the distance travelled, grooming or rearing in the open field test; there were also no differences between groups in the number of risk assessment, time spent in open arms in an elevated plus maze and number of entries into the open arms. These data suggest that physical exercise can ameliorate the long-standing recognition memory deficit induced by early-life SE, suggesting that it may be useful as a putative intervention for patients who suffered SE during infancy.
Collapse
|
17
|
Wasterlain CG, Gloss DS, Niquet J, Wasterlain AS. Epileptogenesis in the developing brain. HANDBOOK OF CLINICAL NEUROLOGY 2013; 111:427-39. [PMID: 23622191 DOI: 10.1016/b978-0-444-52891-9.00046-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The neonatal brain has poorly developed GABAergic circuits, and in many of them GABA is excitatory, favoring ictogenicity. Frequently repeated experimental seizures impair brain development in an age-dependent manner. At critical ages, they delay developmental milestones, permanently lower seizure thresholds, and can cause very specific cognitive and learning deficits, such as the permanent impairment of neuronal spatial maps. Some types of experimental status epilepticus cause neuronal necrosis and apoptosis, and are followed by chronic epilepsy with spontaneous recurrent seizures, others appear relatively benign, so that seizure-induced neuronal injury and epileptogenesis are highly age-, seizure model-, and species-dependent. Experimental febrile seizures can be epileptogenic, and hyperthermia aggravates both neuronal injury and epileptogenicity. Antiepileptic drugs, the mainstay of treatment, have major risks of their own, and can, at therapeutic or near-therapeutic doses, trigger neuronal apoptosis, which is also age-, drug-, cell type-, and species-dependent. The relevance of these experimental results to human disease is still uncertain, but while their brains are quite different, the basic biology of neurons in rodents and humans is strikingly similar. Further research is needed to elucidate the molecular mechanisms of epileptogenesis and of seizure- or drug-induced neuronal injury, in order to prevent their long-term consequences.
Collapse
Affiliation(s)
- Claude G Wasterlain
- Department of Neurology, VA Greater Los Angeles Health Care System, and David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| | | | | | | |
Collapse
|
18
|
Roy H, Lippé S, Lussier F, Sauerwein HC, Lortie A, Lacroix J, Lassonde M. Developmental outcome after a single episode of status epilepticus. Epilepsy Behav 2011; 21:430-6. [PMID: 21705280 DOI: 10.1016/j.yebeh.2011.05.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 05/06/2011] [Accepted: 05/07/2011] [Indexed: 10/18/2022]
Abstract
Consequences of status epilepticus (SE) on psychomotor development and the specific impact of the convulsive event on emerging executive functions remain controversial. Infants treated for a single episode of SE, those treated for a single febrile seizure, and healthy infants were tested with respect to motor development, language, personal, and social skills and self-regulation. The children were divided into two age groups to investigate the impact of the convulsive event at different windows of brain maturation. We found that infants who had had SE were inferior to healthy controls on the development scales. Age differentiated SE impact on visuomotor development versus sociolinguistic development. Children who had been treated for SE had significantly more difficulties delaying a response to an attractive stimulus in one of the long-delay conditions. A single episode of SE can interfere with psychomotor and cognitive development in children without previous developmental delay, and it seems that the functions that are emerging at the time of insult are most vulnerable.
Collapse
Affiliation(s)
- Hélène Roy
- University Hospital Mother/Child (Sainte Justine Hospital), Montréal, QC, Canada
| | | | | | | | | | | | | |
Collapse
|
19
|
Folbergrová J, Kunz WS. Mitochondrial dysfunction in epilepsy. Mitochondrion 2011; 12:35-40. [PMID: 21530687 DOI: 10.1016/j.mito.2011.04.004] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Revised: 02/16/2011] [Accepted: 04/14/2011] [Indexed: 11/29/2022]
Abstract
Mitochondrial dysfunction has been identified as one potential cause of epileptic seizures. Impaired mitochondrial function has been reported for the seizure focus of patients with temporal lobe epilepsy and Ammon's horn sclerosis and of adult and immature animal models of epilepsy. Since mitochondrial oxidative phosphorylation provides the major source of ATP in neurons and mitochondria participate in cellular Ca(2+) homeostasis and generation of reactive oxygen species, their dysfunction strongly affects neuronal excitability and synaptic transmission. Therefore, mitochondrial dysfunction is proposed to be highly relevant for seizure generation. Additionally, mitochondrial dysfunction is known to trigger neuronal cell death, which is a prominent feature of therapy-resistant epilepsy. For this reason mitochondria have to be considered as promising targets for neuroprotective strategies in epilepsy.
Collapse
Affiliation(s)
- Jaroslava Folbergrová
- Department of Developmental Epileptology, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | |
Collapse
|
20
|
Reid WM, Rolfe A, Register D, Levasseur JE, Churn SB, Sun D. Strain-related differences after experimental traumatic brain injury in rats. J Neurotrauma 2011; 27:1243-53. [PMID: 20392137 DOI: 10.1089/neu.2010.1270] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The present study directly compares the effects of experimental brain injury in two commonly used rat strains: Fisher 344 and Sprague-Dawley. We previously found that Fisher rats have a higher mortality rate and more frequent seizure attacks at the same injury level than Sprague-Dawley rats. Although strain differences in rats are commonly accepted as contributing to variability among studies, there is a paucity of literature addressing strain influence in experimental neurotrauma. Therefore this study compares outcome measures in two rat strains following lateral fluid percussion injury. Fisher 344 and Sprague-Dawley rats were monitored for changes in physiological measurements, intracranial pressure, and electroencephalographic activity. We further analyzed neuronal degeneration and cell death in the injured brain using Fluoro-Jade-B (FJB) histochemistry and caspase-3 immunostaining. Behavioral studies using the beam walk and Morris water maze were conducted to characterize strain differences in both motor and cognitive functional recovery following injury. We found that Fisher rats had significantly higher intracranial pressure, prolonged seizure activity, increased FJB-positive staining in the injured cortex and thalamus, and increased caspase-3 expression than Sprague-Dawley rats. On average, Fisher rats displayed a greater amount of total recording time in seizure activity and had longer ictal durations. The Fisher rats also had increased motor deficits, correlating with the above results. In spite of these results, Fisher rats performed better on cognitive tests following injury. The results demonstrate that different rat strains respond to injury differently, and thus in preclinical neurotrauma studies strain influence is an important consideration when evaluating outcomes.
Collapse
Affiliation(s)
- Wendy Murdock Reid
- Department of Neurosurgery, Virginia Commonwealth University, Richmond, Virginia 23298-0631, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Long-term consequences of a prolonged febrile seizure in a dual pathology model. Neurobiol Dis 2011; 43:312-21. [PMID: 21406232 DOI: 10.1016/j.nbd.2011.02.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Revised: 01/26/2011] [Accepted: 02/27/2011] [Indexed: 12/14/2022] Open
Abstract
Clinical evidence suggests that febrile status epilepticus (SE) in children can lead to acute hippocampal injury and subsequent temporal lobe epilepsy. The contribution of febrile SE to the mechanisms underlying temporal lobe epilepsy are however poorly understood. A rat model of temporal lobe epilepsy following hyperthermic SE was previously established in our laboratory, wherein a focal cortical lesion induced at postnatal day 1 (P1), followed by a hyperthermic SE (more than 30 min) at P10, leads to hippocampal atrophy at P22 (dual pathology model) and spontaneous recurrent seizures (SRS) with mild visuospatial memory deficits in adult rats. The goal of this study was to identify the long term electrophysiological, anatomical and molecular changes in this model. Following hyperthermic SE, all cortically lesioned pups developed progressive SRS as adults, characterized by the onset of highly rhythmic activity in the hippocampus. A reduction of hippocampal volume on the side of the lesion preceded the SRS and was associated with a loss of hippocampal neurons, a marked decrease in pyramidal cell spine density, an increase in the hippocampal levels of NMDA receptor NR2A subunit, but no significant change in GABA receptors. These findings suggest that febrile SE in the abnormal brain leads to hippocampal injury that is followed by progressive network reorganization and molecular changes that contribute to the epileptogenesis as well as the observed memory deficits.
Collapse
|
22
|
Wasterlain CG, Thompson KW, Suchomelova L, Niquet J. Brain energy metabolism during experimental neonatal seizures. Neurochem Res 2010; 35:2193-8. [PMID: 21136154 PMCID: PMC3002164 DOI: 10.1007/s11064-010-0339-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2010] [Accepted: 11/19/2010] [Indexed: 10/25/2022]
Abstract
During flurothyl seizures in 4-day-old rats, cortical concentration of ATP, phosphocreatine and glucose fell while lactate rose. Cortical energy use rate more than doubled, while glycolytic rate increased fivefold. Calculation of the cerebral metabolic balance during sustained seizures suggests that energy balance could be maintained in hyperglycemic animals, and would decline slowly in normoglycemia, but would be compromised by concurrent hypoglycemia, hyperthermia or hypoxia. These results suggest that the metabolic challenge imposed on the brain by this model of experimental neonatal seizures is milder than that seen at older ages, but can become critical when associated with other types of metabolic stress.
Collapse
Affiliation(s)
- Claude G Wasterlain
- Epilepsy Research Laboratory, VA Greater Los Angeles Health Care System, 11301 Wilshire Boulevard, West Los Angeles, CA 90073, USA.
| | | | | | | |
Collapse
|
23
|
Golbs A, Nimmervoll B, Sun JJ, Sava IE, Luhmann HJ. Control of programmed cell death by distinct electrical activity patterns. Cereb Cortex 2010; 21:1192-202. [PMID: 20966045 DOI: 10.1093/cercor/bhq200] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Electrical activity and sufficient supply with survival factors play a major role in the control of apoptosis in the developing cortex. Coherent high-frequency neuronal activity, which efficiently releases neurotrophins, is essential for the survival of immature neurons. We studied the influence of neuronal activity on apoptosis in the developing cortex. Dissociated cultures of the newborn mouse cerebral cortex were grown on multielectrode arrays to determine the activity patterns that promote neuronal survival. Cultures were transfected with a plasmid coding for a caspase-3-sensitive fluorescent protein allowing real-time analysis of caspase-3-dependent apoptosis in individual neurons. Elevated extracellular potassium concentrations (5 and 8 mM), application of 4-aminopyridine or the γ-aminobutyric acid-A receptor antagonist Gabazine induced a shift in the frequency distribution of activity toward high-frequency bursts. Under these conditions, a reduction or delay in caspase-3 activation and an overall increase in neuronal survival could be observed. This effect was dependent on the activity of phosphatidylinositol-3 kinase, as blockade of this enzyme abolished the survival-promoting effect of high extracellular potassium concentrations. Our data indicate that increased network activity can prevent apoptosis in developing cortical neurons.
Collapse
Affiliation(s)
- Antje Golbs
- Institute of Physiology and Pathophysiology, University Medical Center, Johannes Gutenberg University, D-55128 Mainz, Germany
| | | | | | | | | |
Collapse
|
24
|
Parfenova H, Leffler CW, Tcheranova D, Basuroy S, Zimmermann A. Epileptic seizures increase circulating endothelial cells in peripheral blood as early indicators of cerebral vascular damage. Am J Physiol Heart Circ Physiol 2010; 298:H1687-98. [PMID: 20363895 PMCID: PMC2886638 DOI: 10.1152/ajpheart.00032.2010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 03/29/2010] [Indexed: 12/24/2022]
Abstract
Circulating endothelial cells (CECs) are nonhematopoetic mononuclear cells in peripheral blood that are dislodged from injured vessels during cardiovascular disease, systemic vascular disease, and inflammation. Their occurrence during cerebrovascular insults has not been previously described. Epileptic seizures cause the long-term loss of cerebrovascular endothelial dilator function. We hypothesized that seizures cause endothelial sloughing from cerebral vessels and the appearance of brain-derived CECs (BCECs), possible early indicators of cerebral vascular damage. Epileptic seizures were induced by bicuculline in newborn pigs; venous blood was then sampled during a 4-h period. CECs were identified in the fraction of peripheral blood mononuclear cells by the expression of endothelial antigens (CD146, CD31, and endothelial nitric oxide synthase) and by Ulex europeaus lectin binding. In control animals, few CECs were detected. Seizures caused a time-dependent increase in CECs 2-4 h after seizure onset. Seizure-induced CECs coexpress glucose transporter-1, a blood-brain barrier-specific glucose transporter, indicating that these cells originate in the brain vasculature and are thus BCECs. Seizure-induced BCECs cultured in EC media exhibited low proliferative potential and abnormal cell contacts. BCEC appearance during seizures was blocked by a CO-releasing molecule (CORM-A1) or cobalt protoporphyrin (heme oxygenase-1 inducer), which prevented apoptosis in cerebral arterioles and the loss of cerebral vascular endothelial function during the late postictal period. These findings suggest that seizure-induced BCECs are injured ECs dislodged from cerebral microvessels during seizures. The correlation between the appearance of BCECs in peripheral blood, apoptosis in cerebral vessels, and the loss of postictal cerebral vascular function suggests that BCECs are early indicators of late cerebral vascular damage.
Collapse
Affiliation(s)
- Helena Parfenova
- Dept. of Physiology, Univ. of Tennessee Health Science Center, 894 Union Ave., Memphis, TN 38163, USA.
| | | | | | | | | |
Collapse
|
25
|
Laurén HB, Lopez-Picon FR, Brandt AM, Rios-Rojas CJ, Holopainen IE. Transcriptome analysis of the hippocampal CA1 pyramidal cell region after kainic acid-induced status epilepticus in juvenile rats. PLoS One 2010; 5:e10733. [PMID: 20505763 PMCID: PMC2873964 DOI: 10.1371/journal.pone.0010733] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 04/28/2010] [Indexed: 11/19/2022] Open
Abstract
Molecular mechanisms involved in epileptogenesis in the developing brain remain poorly understood. The gene array approach could reveal some of the factors involved by allowing the identification of a broad scale of genes altered by seizures. In this study we used microarray analysis to reveal the gene expression profile of the laser microdissected hippocampal CA1 subregion one week after kainic acid (KA)-induced status epilepticus (SE) in 21-day-old rats, which are developmentally roughly comparable to juvenile children. The gene expression analysis with the Chipster software generated a total of 1592 differently expressed genes in the CA1 subregion of KA-treated rats compared to control rats. The KEGG database revealed that the identified genes were involved in pathways such as oxidative phosporylation (26 genes changed), and long-term potentiation (LTP; 18 genes changed). Also genes involved in Ca2+ homeostasis, gliosis, inflammation, and GABAergic transmission were altered. To validate the microarray results we further examined the protein expression for a subset of selected genes, glial fibrillary protein (GFAP), apolipoprotein E (apo E), cannabinoid type 1 receptor (CB1), Purkinje cell protein 4 (PEP-19), and interleukin 8 receptor (CXCR1), with immunohistochemistry, which confirmed the transcriptome results. Our results showed that SE resulted in no obvious CA1 neuronal loss, and alterations in the expression pattern of several genes during the early epileptogenic phase were comparable to previous gene expression studies of the adult hippocampus of both experimental epileptic animals and patients with temporal lobe epilepsy (TLE). However, some changes seem to occur after SE specifically in the juvenile rat hippocampus. Insight of the SE-induced alterations in gene expression and their related pathways could give us hints for the development of new target-specific antiepileptic drugs that interfere with the progression of the disease in the juvenile age group.
Collapse
Affiliation(s)
- Hanna B. Laurén
- Department of Pharmacology, Drug Development, and Therapeutics, Institute of Biomedicine, University of Turku, Turku, Finland
- MediCity Research Laboratory, Turku, Finland
| | - Francisco R. Lopez-Picon
- Department of Pharmacology, Drug Development, and Therapeutics, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Annika M. Brandt
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Clarissa J. Rios-Rojas
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Irma E. Holopainen
- Department of Pharmacology, Drug Development, and Therapeutics, Institute of Biomedicine, University of Turku, Turku, Finland
- MediCity Research Laboratory, Turku, Finland
- * E-mail:
| |
Collapse
|
26
|
Prigol M, Wilhelm EA, Nogueira CW, Zeni G. Diphenyl diselenide-induced seizures in rat pups: possible interaction with GABAergic system. Neurol Res 2010; 32:1002-8. [PMID: 20433775 DOI: 10.1179/016164110x12670144737738] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES The involvement of the GABAergic system in seizures induced by diphenyl diselenide (PhSe)₂ in rat pups was investigated. METHODS To this end, the effect of aminooxyacetic acid hemihydrochloride (AOAA, 20 mg/kg; by intraperitoneal route, i.p.), a GABA-T inhibitor; DL-2,4-diamino-n-butyric acid hydrochloride (DABA, 16 mg/kg; i.p.), an inhibitor of GABA uptake; and γ-aminobutyric acid (GABA, 10 and 40 mg/kg; i.p.), diazepam (3 mg/kg; i.p.) and phenobarbital (40 mg/kg; i.p.), GABAergic agonists as well as picrotoxin (1 mg/kg; i.p.), a GABAA receptor antagonist on (PhSe)₂ (50 and 500 mg/kg, by oral route, p.o.)-induced seizures, were studied. The [(3)H]GABA uptake levels by cortical and hippocampal slices in rat pups exposed to (PhSe)₂ were also carried out. RESULTS Pre-treatment with GABA (40 mg/kg), diazepam, phenobarbital, AOAA and DABA abolished the appearance of seizures induced by 50 mg/kg (PhSe)₂ in rat pups. Picrotoxin increased the percentage of convulsing rat pups from 42 to 100% and reduced significantly the onset for the first convulsive episode induced by (PhSe)₂ at the dose of 50 mg/kg. Diazepam and phenobarbital prolonged significantly the latency for the onset of the first convulsive episode caused by 500 mg/kg (PhSe)₂ in rat pups. [(3)H]GABA uptake levels were stimulated in cerebral cortical and hippocampal slices of convulsing rat pups administered with both doses of (PhSe)₂. DISCUSSION Our findings demonstrated that seizures induced by (PhSe)₂ are mediated, at least in part, by an interaction with GABAergic system.
Collapse
Affiliation(s)
- Marina Prigol
- Departamento de Química, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Brazil
| | | | | | | |
Collapse
|
27
|
Abstract
Epileptogenesis is defined as the process of developing epilepsy-a disorder characterized by recurrent seizures-following an initial insult. Seizure incidence during the human lifespan is at its highest in infancy and childhood. Animal models of epilepsy and human tissue studies suggest that epileptogenesis involves a cascade of molecular, cellular and neuronal network alterations. Within minutes to days following the initial insult, there are acute early changes in neuronal networks, which include rapid alterations to ion channel kinetics as a result of membrane depolarization, post-translational modifications to existing functional proteins, and activation of immediate early genes. Subacute changes occur over hours to weeks, and include transcriptional events, neuronal death and activation of inflammatory cascades. The chronic changes that follow over weeks to months include anatomical changes, such as neurogenesis, mossy fiber sprouting, network reorganization, and gliosis. These epileptogenic processes are developmentally regulated and might contribute to differences in epileptogenesis between adult and developing brains. Here we review the factors responsible for enhanced seizure susceptibility in the developing brain, and consider age-specific mechanisms of epileptogenesis. An understanding of these factors could yield potential therapeutic targets for the prevention of epileptogenesis and also provide biomarkers for identifying patients at risk of developing epilepsy or for monitoring disease progression.
Collapse
|
28
|
HAMED SHERIFAA. THE RATIONALE FOR NEUROPROTECTION IN EPILEPSY: STEPS FORWARD FOR NEW THERAPEUTIC AND PREVENTIVE STRATEGIES. J Integr Neurosci 2010. [DOI: 10.1142/s0219635210002378] [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
|
29
|
Abstract
The lifespan risk of seizures is highest in the neonatal period. Current therapies have limited efficacy. Although the treatment of neonatal seizures has not changed significantly in the last several decades, there has been substantial progress in understanding developmental mechanisms that influence seizure generation and responsiveness to anticonvulsants. This article provides an overview of current approaches to the diagnosis and treatment of neonatal seizures, and some of the recent insights about the pathophysiology of neonatal seizures that may provide the foundation for better treatment are identified.
Collapse
Affiliation(s)
- Frances E Jensen
- Children's Hospital Boston, CLS 14073, 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
30
|
Dunleavy M, Shinoda S, Schindler C, Ewart C, Dolan R, Gobbo OL, Kerskens CM, Henshall DC. Experimental neonatal status epilepticus and the development of temporal lobe epilepsy with unilateral hippocampal sclerosis. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 176:330-42. [PMID: 19948825 DOI: 10.2353/ajpath.2010.090119] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hippocampal sclerosis is a common pathological finding in patients with temporal lobe epilepsy, including children, but a causal relationship to early-life seizures remains in question. Neonatal status epilepticus in animals can result in neuronal death within the hippocampus, although macroscopic features of hippocampal shrinkage are not evident at adulthood. Here, we examined electrophysiological and pathological consequences of focally evoked status epilepticus triggered by intra-amygdala microinjection of kainic acid in postnatal day 10 rat pups. Neonatal status epilepticus resulted in extensive neuronal death in the ipsilateral hippocampal CA1 and CA3 subfields and hilus, as assessed by DNA fragmentation and Fluoro-Jade B staining 72 hours later. The contralateral hippocampus was not significantly damaged. Histopathology at P55/P65 revealed unilateral hippocampal sclerosis (grade IV, modified Wyler/Watson scale) comprising >50% CA1 and CA3 neuron loss and astrogliosis. Additional features included hydrocephalus ex vacuo, modest dentate granule cell layer widening, and altered neuropeptide Y immunoreactivity indicative of synaptic rearrangement. Hippocampal atrophy was also evident on magnetic resonance imaging. Depth electrode recordings at adulthood detected spontaneous seizures that involved the ipsilateral hippocampus and amygdala. A significant positive correlation was found between hippocampal pathology grade and both frequency and duration of epileptic seizures at adulthood. The current study demonstrates that experimental neonatal status epilepticus can result in classical unilateral hippocampal sclerosis and temporal lobe epilepsy.
Collapse
Affiliation(s)
- Mark Dunleavy
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Glenn OA, Quiroz EM, Berman JI, Studholme C, Xu D. Diffusion-weighted imaging in fetuses with unilateral cortical malformations and callosal agenesis. AJNR Am J Neuroradiol 2009; 31:1100-2. [PMID: 19942706 DOI: 10.3174/ajnr.a1863] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
DWI was performed in fetuses with callosal agenesis and unilateral cortical malformations. ADC values were retrospectively measured in the developing white matter underlying the cortical malformation and compared with the corresponding contralateral white matter. In all 3 patients, ADC values were lower under the areas of cortical malformation compared with the normal contralateral side. Our findings suggest that there are structural differences in the developing white matter underlying areas of cortical malformation.
Collapse
Affiliation(s)
- O A Glenn
- Department of Radiology and Biomedical Imaging, University of California-San Francisco, 505 Parnassus Avenue, San Francisco, CA 94143, USA.
| | | | | | | | | |
Collapse
|
32
|
Thibeault-Eybalin MP, Lortie A, Carmant L. Neonatal seizures: do they damage the brain? Pediatr Neurol 2009; 40:175-80. [PMID: 19218030 DOI: 10.1016/j.pediatrneurol.2008.10.026] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 10/01/2008] [Accepted: 10/01/2008] [Indexed: 01/10/2023]
Abstract
Seizures are an early sign of brain injury in newborns. These seizures are in most cases repetitive or associated with asymptomatic electrographic seizures. Despite the relative resistance of the immature brain to seizure-induced brain damage, there is more and more evidence that neonatal seizures impair normal brain development. This review addresses the changes associated with neonatal seizures and discusses current and future potential neuroprotective strategies.
Collapse
|
33
|
Gibbs SA, Scantlebury MH, Awad P, Lema P, Essouma JB, Parent M, Descarries L, Carmant L. Hippocampal atrophy and abnormal brain development following a prolonged hyperthermic seizure in the immature rat with a focal neocortical lesion. Neurobiol Dis 2008; 32:176-82. [PMID: 18678257 DOI: 10.1016/j.nbd.2008.07.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 07/03/2008] [Accepted: 07/08/2008] [Indexed: 12/15/2022] Open
Abstract
In rats subjected to a focal cortical lesion soon after birth, hyperthermia at P10 induces a prolonged epileptic seizure, often followed by temporal lobe epilepsy in the adult. To determine whether brain damage and notably hippocampal atrophy occur early on in this model, whole brain as well as hemispheric, cortical, subcortical and hippocampal volumes was measured in non-lesioned and lesioned rat pups, 2 days (P12) and 12 days (P22) after the hyperthermic seizure. All pups with a cortical lesion showed reductions in whole brain and in ipsilateral hemispheric, cortical and hippocampal volumes at P12, which persisted at P22 in pups having also sustained a prolonged hyperthermic seizure at P10. Limiting the duration of the seizure with Diazepam prevented the hippocampal atrophy. Thus, a prolonged hyperthermic seizure in immature brain with a subtle neocortical lesion impairs normal brain development, and the duration of the seizure appears to be a key factor in generating hippocampal atrophy.
Collapse
Affiliation(s)
- Steve A Gibbs
- Sainte-Justine Hospital Research Centre, Université de Montréal, Montreal, QC, Canada
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Dean JM, George S, Naylor AS, Mallard C, Gunn AJ, Bennet L. Partial neuroprotection with low-dose infusion of the alpha2-adrenergic receptor agonist clonidine after severe hypoxia in preterm fetal sheep. Neuropharmacology 2008; 55:166-74. [PMID: 18572205 DOI: 10.1016/j.neuropharm.2008.05.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Revised: 04/12/2008] [Accepted: 05/07/2008] [Indexed: 10/22/2022]
Abstract
We have previously shown that brief alpha(2)-adrenergic receptor blockade increased neuronal injury after severe hypoxia in preterm fetal sheep. We now examine whether infusion of an alpha(2)-adrenergic receptor agonist, clonidine, is neuroprotective. Preterm fetal sheep (70% gestation) received either saline-vehicle or clonidine at either 10 microg/kg/h (low-dose) or 100 microg/kg/h (high-dose) from 15 min until 4 h after 25 min of umbilical cord occlusion. Both low- and high-dose clonidine infusions after sham-occlusion were associated with transient EEG suppression but no neuronal loss. Low-dose but not high-dose clonidine infusions after umbilical cord occlusion were associated with a significant overall increase in numbers of surviving neurons after three days' recovery. High-dose clonidine was associated with transient hyperglycemia and increased numbers of delayed electrographic seizures. These results provide further evidence that alpha(2)-adrenergic receptor activation shortly after perinatal hypoxia-ischemia can promote neural recovery, but highlight the complex dose-response of exogenous therapy.
Collapse
Affiliation(s)
- Justin M Dean
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | | | | | | | | | | |
Collapse
|
35
|
Järvelä JT, Lopez-Picon FR, Holopainen IE. Age-dependent cyclooxygenase-2 induction and neuronal damage after status epilepticus in the postnatal rat hippocampus. Epilepsia 2008; 49:832-41. [DOI: 10.1111/j.1528-1167.2007.01454.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
36
|
Abstract
The constitutive isoform of heme oxygenase, HO-2, is highly expressed in the brain and in cerebral vessels. HO-2 functions in the brain have been evaluated using pharmacological inhibitors of the enzyme and HO-2 gene deletion in in vivo animal models and in cultured cells (neurons, astrocytes, cerebral vascular endothelial cells). Rapid activation of HO-2 via post-translational modifications without upregulation of HO-2 expression or HO-1 induction coincides with the increase in cerebral blood flow aimed at maintaining brain homeostasis and neuronal survival during seizures, hypoxia, and hypotension. Pharmacological inhibition or gene deletion of brain HO-2 exacerbates oxidative stress induced by seizures, glutamate, and inflammatory cytokines, and causes cerebral vascular injury. Carbon monoxide (CO) and bilirubin, the end products of HO-catalyzed heme degradation, have distinct cytoprotective functions. CO, by binding to a heme prosthetic group, regulates the key components of cell signaling, including BK(Ca) channels, guanylyl cyclase, NADPH oxidase, and the mitochondria respiratory chain. Cerebral vasodilator effects of CO are mediated via activation of BK(Ca) channels and guanylyl cyclase. CO, by inhibiting the major components of endogenous oxidant-generating machinery, NADPH oxidase and the cytochrome C oxidase of the mitochondrial respiratory chain, blocks formation of reactive oxygen species. Bilirubin, via redox cycling with biliverdin, is a potent oxidant scavenger that removes preformed oxidants. Overall, HO-2 has dual housekeeping cerebroprotective functions by maintaining autoregulation of cerebral blood flow aimed at improving neuronal survival in a changing environment, and by providing an effective defense mechanism that blocks oxidant formation and prevents cell death caused by oxidative stress.
Collapse
Affiliation(s)
- Helena Parfenova
- Laboratory for Research in Neonatal Physiology, Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA.
| | | |
Collapse
|
37
|
Seizures in the developing brain: cellular and molecular mechanisms of neuronal damage, neurogenesis and cellular reorganization. Neurochem Int 2007; 52:935-47. [PMID: 18093696 DOI: 10.1016/j.neuint.2007.10.021] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2007] [Revised: 10/29/2007] [Accepted: 10/31/2007] [Indexed: 12/30/2022]
Abstract
Epilepsy is a common neurological disorder that occurs more frequently in children than in adults. The extent that prolonged seizure activity, i.e. status epilepticus (SE), and repeated, brief seizures affect neuronal structure and function in both the immature and mature brain has been the subject of increasing clinical and experimental research. Earlier studies suggest that seizure-induced effects in the immature brain compared with the adult brain are different. This is manifested as differences in neuronal vulnerability, cellular and synaptic reorganization and regenerative processes. The focus of this review is first to give a short overview of currently used experimental models of epilepsy in immature rats, and then discuss more thoroughly seizure-induced acute and sub-acute cellular and molecular alterations, highlight the contribution of inflammatory-like reactions and intracellular cytoskeleton to the insult, and reveal changes in the structure and function of inhibitory GABA(A) and excitatory glutamate receptors. The role of seizure-activated reparative, plastic processes, synaptic remodelling, neurogenesis as well as the long-term consequences of seizures are briefly outlined. The main emphasis is put on studies carried out in experimental animals, and the focus of interest is the hippocampus, the brain area of great vulnerability in epilepsy. In vitro studies are discussed only to limited extent. Collectively, recent studies suggest that the deleterious effects of seizures may not solely be a consequence of neuronal damage and loss per se, but could be due to the fact that seizures interfere with the highly regulated developmental processes in the immature brain.
Collapse
|
38
|
Abstract
In childhood, the risk for seizures is greatest in the neonatal period. Currently used therapies have limited efficacy. Although the treatment of neonatal seizures has not significantly changed in the past several decades, there has been substantial progress in understanding developmental mechanisms that influence seizure generation and responsiveness to anticonvulsants. This review includes an overview of current approaches to the diagnosis and treatment of neonatal seizures, identifies some of the critical factors that have limited progress, and highlights recent insights about the pathophysiology of neonatal seizures that may provide the foundation for better treatment.
Collapse
Affiliation(s)
- Faye S Silverstein
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109-0646, USA.
| | | |
Collapse
|
39
|
Xu S, Pang Q, Liu Y, Shang W, Zhai G, Ge M. Neuronal apoptosis in the resected sclerotic hippocampus in patients with mesial temporal lobe epilepsy. J Clin Neurosci 2007; 14:835-40. [PMID: 17660056 DOI: 10.1016/j.jocn.2006.08.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2006] [Revised: 08/23/2006] [Accepted: 08/26/2006] [Indexed: 01/06/2023]
Abstract
To further confirm at the molecular level that neuronal apoptosis occurs in mesial temporal sclerosis (MTS), the main substrate of mesial temporal lobe epilepsy (MTLE), 24 resected sclerotic hippocampi from 24 patients with drug-resistant MTLE associated with MTS were studied microscopically, electronmicroscopically and immunohistochemically, with detection of expression of apoptosis-associated genes including bcl-2, p53, bax, fas and caspase-3. Early apoptosis changes were found morphologically in hippocampi from three patients with MTLE using transmission electron microscopy. Positive immunostained neurons for bcl-2, p53, fas and caspase-3 were found in the sclerotic hippocampi of 19/24, 14/24, 22/24 and 20/24 patients respectively, which was statistically different from controls. Correlative analysis showed the expression of p53, fas and caspase-3 were positively correlated with seizure frequency. Apoptosis may contribute to MTS, and seizures may induce apoptosis, and thus contribute to neuronal loss in MTS.
Collapse
Affiliation(s)
- Shangchen Xu
- Department of Neurosurgery, Shandong Provincial Hospital of Shandong University, Jinan, 250021, PR China
| | | | | | | | | | | |
Collapse
|
40
|
Humphreys P, Deonandan R, Whiting S, Barrowman N, Matzinger MA, Briggs V, Hurteau J, Wallace E. Factors associated with epilepsy in children with periventricular leukomalacia. J Child Neurol 2007; 22:598-605. [PMID: 17690068 DOI: 10.1177/0883073807302599] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Children with cerebral palsy associated with periventricular leukomalacia frequently develop unprovoked epileptic seizures. This article reports an analysis of risk factors for epilepsy in children with radiologically confirmed periventricular leukomalacia. This cohort was screened for epilepsy and for an array of clinical and demographic factors. Of 154 subjects with radiologically confirmed periventricular leukomalacia, 40 (26.0%) had epilepsy. In the epileptic group, radiologic pathology other than periventricular leukomalacia was uncommon. Significant associations were found between epilepsy and cerebral palsy patterns other than spastic diparesis, mental handicap, cortical visual impairment, neonatal seizures, and severe periventricular leukomalacia. Only the presence of neonatal seizures was significantly associated with epilepsy once other risk factors were controlled in the regression model. Some previous studies have shown an association between neonatal seizures and later epilepsy for cerebral palsy in general. This is the first report of such an association for a single predominant type of cerebral pathology.
Collapse
Affiliation(s)
- Peter Humphreys
- Division of Neurology, Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa Children's Treatment Centre Ottawa, Ontario, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Scantlebury MH, Heida JG, Hasson HJ, Velísková J, Velísek L, Galanopoulou AS, Moshé SL. Age-Dependent Consequences of Status Epilepticus: Animal Models. Epilepsia 2007; 48 Suppl 2:75-82. [PMID: 17571355 DOI: 10.1111/j.1528-1167.2007.01069.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Status epilepticus (SE) is a significant neurological emergency that occurs most commonly in children. Although SE has been associated with an elevated risk of brain injury, it is unclear from clinical studies in whom and under what circumstances brain injury will occur. The purpose of this review is to evaluate the effects of age on the consequences of SE. In this review, we focus mainly on the animal data that describe the consequences of a single episode of SE induced in the adult and immature rat brain. The experimental data suggest that the risk of developing SE-induced brain damage, subsequent epilepsy and cognitive deficits in large part depends on the age in which the SE occurs. Younger rats are more resistant to seizure-induced brain damage than older rats; however, when SE occurs in immature rats with abnormal brains, there is an increase in the severity of seizure-induced brain injury. Better understanding of the pathophysiologic mechanisms underlying the age-specific alterations to the brain induced by SE will lead to the development of novel and effective strategies to improve the deleterious consequences.
Collapse
Affiliation(s)
- Morris H Scantlebury
- Korey Department of Neurology, Albert Einstein College of Medicine, Pelham Parkway South, Kennedy Center, Bronx, NY, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
Sohya K, Kitamura A, Akaneya Y. Chronic membrane depolarization-induced morphological alteration of developing neurons. Neuroscience 2007; 145:232-40. [PMID: 17222518 DOI: 10.1016/j.neuroscience.2006.11.045] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2006] [Revised: 10/23/2006] [Accepted: 11/26/2006] [Indexed: 11/30/2022]
Abstract
During development of CNS, young neurons experience various stimuli, and thereafter differentiate to mature neurons in an activity-dependent manner. Membrane depolarization acts as an inducer of excitability and various signals in the neurons, which can be used as a model of neuronal activity. However, the mechanisms of the influence of membrane depolarization on neuronal differentiation have not been fully understood. Therefore, we investigated the effect of membrane depolarization on morphology of spines and generation of valid electrical activity. Using rat hippocampal cultures treated from the plating day with or without high KCl (35 mM, termed HK), we directly observed living neurons transfected with green fluorescence protein-expressing plasmid through a two-photon laser scanning confocal microscope and electrophysiological recording using a patch-clamp technique. Compared with controls, the neurons cultured with HK for 3 days in vitro (DIV) showed marked filopodia-like protrusions as well as an increase in the number of spines, but those cultured with HK for 6 DIV profoundly lost these spines, resulting in a small number of fine filopodia-like protrusions proximally and on the cell body, and a smooth surface of distal dendrites. Electrophysiological recordings showed no spontaneous responses in 6 DIV HK-treated neurons. Moreover, addition of an N-methyl-D-aspartate receptor (NMDAR) antagonist to HK-treated neurons blocked the shrinkage and decrease in the number of filopodia-like protrusions significantly. These findings suggest that membrane depolarization of developing neurons induces synaptogenesis in the early stages of development but chronic treatment with HK causes pathological changes through NMDAR, and that there may be alternative mechanisms for the physiological differentiation of neurons in later developmental stages.
Collapse
Affiliation(s)
- K Sohya
- Division of Neurophysiology, Department of Neuroscience, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita 565-0871 Japan
| | | | | |
Collapse
|
43
|
Bennet L, Dean JM, Wassink G, Gunn AJ. Differential effects of hypothermia on early and late epileptiform events after severe hypoxia in preterm fetal sheep. J Neurophysiol 2006; 97:572-8. [PMID: 17093117 DOI: 10.1152/jn.00957.2006] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Moderate cerebral hypothermia is consistently neuroprotective after experimental hypoxia-ischemia; however, its mechanisms remain poorly defined. Using a model of complete umbilical cord occlusion for 25 min in 0.7 gestation fetal sheep, we examined the effects of cerebral hypothermia (fetal extradural temperature reduced from 39.5 +/- 0.2 degrees C to <34 degrees C; mean +/- SD), from 90 min to 70 h after the end of the insult, on postocclusion epileptiform activity. In the first 6 h after the end of occlusion, fetal electroencephalographic (EEG) activity was abnormal with a mixture of fast and slow epileptiform transients superimposed on a suppressed background; seizures started a mean of 8 h after occlusion. There was a close correlation between numbers of these EEG transients and subsequent neuronal loss in the striatum after 3 days recovery (r(2) = 0.65, P = 0.008). Hypothermia was associated with a marked reduction in numbers of epileptiform transients in the first 6 h, reduced amplitude of seizures, and reduced striatal neuronal loss. In conclusion, neuroprotection with delayed, prolonged head cooling after a severe asphyxial insult in the preterm fetus was associated with potent, specific suppression of epileptiform transients in the early recovery phase but not of numbers of delayed seizures.
Collapse
Affiliation(s)
- L Bennet
- Department of Physiology, Faculty of Medicine and Health Science, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | | | | | | |
Collapse
|
44
|
Abstract
Prognosis for seizure control and cognitive development varies considerably among syndromes. Several factors may interact to influence outcome of an epilepsy including a causative etiology, ictal and interictal discharges, seizure-related trauma or systemic perturbations, and antiepileptic drug (AED) effects. Clinical evidence convincingly supporting Gowers' hypothesis that seizures beget seizures is lacking. Short-term seizure suppression by early treatment does not appear to influence long-term prognosis. Malignant epilepsy syndromes usually begin in infancy or childhood, have a high seizure frequency, resist the initial AED, and are often associated with progressive cognitive dysfunction. Prompt management of some severe epilepsy syndromes may lessen cognitive decline. However, aggressive AEDs therapy must be balanced against the potential for cognitive side effects, particularly if multiple AEDs are used. Several experimental paradigms closely parallel human TLE as both have an initial precipitating injury (IPI), a latent period, then recurrent spontaneous seizures. In humans, an IPI is any medical event with neurological implications. Although transition from a latent period to a seizure disorder certainly constitutes "progression" of the disorder, convincing clinical evidence of subsequent worsening has not emerged. Substantial clinical and experimental evidence indicates some cognitive regression and focal atrophy with time for TLE and other intractable syndromes. However, seizure frequency and severity, established early in the disorder, appear stable in most patients, and even regress in benign syndromes. Factors mitigating or extinguishing epilepsies need to be further sought.
Collapse
Affiliation(s)
- Warren T Blume
- Department of Clinical Neurological Sciences, London Health Sciences Centre London, Ontario, Canada.
| |
Collapse
|
45
|
Bennet L, Roelfsema V, Pathipati P, Quaedackers JS, Gunn AJ. Relationship between evolving epileptiform activity and delayed loss of mitochondrial activity after asphyxia measured by near-infrared spectroscopy in preterm fetal sheep. J Physiol 2006; 572:141-54. [PMID: 16484298 PMCID: PMC1779651 DOI: 10.1113/jphysiol.2006.105197] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Early onset cerebral hypoperfusion after birth is highly correlated with neurological injury in premature infants, but the relationship with the evolution of injury remains unclear. We studied changes in cerebral oxygenation, and cytochrome oxidase (CytOx) using near-infrared spectroscopy in preterm fetal sheep (103-104 days of gestation, term is 147 days) during recovery from a profound asphyxial insult (n= 7) that we have shown produces severe subcortical injury, or sham asphyxia (n= 7). From 1 h after asphyxia there was a significant secondary fall in carotid blood flow (P < 0.001), and total cerebral blood volume, as reflected by total haemoglobin (P < 0.005), which only partially recovered after 72 h. Intracerebral oxygenation (difference between oxygenated and deoxygenated haemoglobin concentrations) fell transiently at 3 and 4 h after asphyxia (P < 0.01), followed by a substantial increase to well over sham control levels (P < 0.001). CytOx levels were normal in the first hour after occlusion, was greater than sham control values at 2-3 h (P < 0.05), but then progressively fell, and became significantly suppressed from 10 h onward (P < 0.01). In the early hours after reperfusion the fetal EEG was highly suppressed, with a superimposed mixture of fast and slow epileptiform transients; overt seizures developed from 8 +/- 0.5 h. These data strongly indicate that severe asphyxia leads to delayed, evolving loss of mitochondrial oxidative metabolism, accompanied by late seizures and relative luxury perfusion. In contrast, the combination of relative cerebral deoxygenation with evolving epileptiform transients in the early recovery phase raises the possibility that these early events accelerate or worsen the subsequent mitochondrial failure.
Collapse
Affiliation(s)
- L Bennet
- Department of Physiology, University of Auckland, Private Bag 92019, Auckland, New Zealand.
| | | | | | | | | |
Collapse
|
46
|
Abstract
We examined the mechanism of neuronal necrosis induced by hypoxia in dentate gyrus cultures or by status epilepticus (SE) in adult mice. Our observations showed that hypoxic necrosis can be an active process starting with early mitochondrial swelling and loss of the mitochondrial membrane potential, followed by cytochrome c release and caspase-9-dependent activation of caspase-3. This sequence of events (or program) was independent of protein synthesis and may be induced by energy failure and/or calcium overloading of mitochondria. We called this form of necrosis "programmed necrosis." After SE in adult mice, CA1 and CA3 pyramidal neurons displayed a necrotic morphology, associated with caspase-3 immunoreactivity and with double-stranded DNA breaks, suggesting that "programmed necrosis" may be involved in SE-induced neuronal loss.
Collapse
Affiliation(s)
- Jerome Niquet
- Epilepsy Research Laboratory, VA Greater Los Angeles Healthcare System, West Los Angeles, CA 90073, USA.
| | | | | |
Collapse
|
47
|
Thom M, Zhou J, Martinian L, Sisodiya S. Quantitative post-mortem study of the hippocampus in chronic epilepsy: seizures do not inevitably cause neuronal loss. Brain 2005; 128:1344-57. [PMID: 15758032 DOI: 10.1093/brain/awh475] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Hippocampal sclerosis describes a pattern of neuronal loss and gliosis involving the medial temporal structures most often encountered in patients with epilepsy. It is still a matter for debate as to whether this lesion is acquired during the course of the patient's seizure history or is present at the outset. Early febrile seizures, episodes of status epilepticus as well as repetitive brief seizures may all contribute to the evolution of hippocampal sclerosis. In addition, genetic factors and developmental abnormalities of the hippocampus may both increase vulnerability to seizures and hippocampal injury. Recent human studies have addressed neuropathological changes in young adults and children undergoing surgery for refractory seizures with hippocampal sclerosis. Post-mortem examination, however, provides the opportunity to evaluate the effect of a lifetime of seizures on both left and right hippocampi, and the presence of any co-existing malformation. Post-mortem stereological analysis of 28 patients with poorly controlled seizures has confirmed a subgroup with absence of significant hippocampal neuronal loss despite decades of generalized seizures, including status epilepticus. The presence of granule cell dispersion correlated to the severity of hippocampal neuronal loss. Furthermore, in patients with confirmed hippocampal sclerosis at post-mortem examination, stereological assessment of the neocortex failed to confirm significant white matter neuronal heterotopia that might indicate an underlying developmental abnormality. In conclusion, seizures do not invariably lead to hippocampal injury and white matter heterotopia is not invariably associated with hippocampal sclerosis.
Collapse
Affiliation(s)
- Maria Thom
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College, London, UK.
| | | | | | | |
Collapse
|
48
|
Sadgrove MP, Chad JE, Gray WP. Kainic acid induces rapid cell death followed by transiently reduced cell proliferation in the immature granule cell layer of rat organotypic hippocampal slice cultures. Brain Res 2005; 1035:111-9. [PMID: 15722051 DOI: 10.1016/j.brainres.2004.11.059] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2004] [Revised: 11/19/2004] [Accepted: 11/19/2004] [Indexed: 11/26/2022]
Abstract
Brain injury due to seizures results in transiently increased cell proliferation and neurogenesis in the subgranular zone of the adult dentate gyrus. In contrast, the immature postnatal brain appears to be more resistant to cell death after seizure-induced brain injury and paradoxically reacts to seizures by reducing SGZ proliferation. Organotypic hippocampal slice cultures are a useful paradigm for modelling the early postnatal hippocampus. We have investigated the temporal relationship between cell death and cell proliferation after kainate in the granule cell layer of rat organotypic hippocampal slice cultures equivalent to post natal day 11 animals. We found stable numbers and densities of mature thionine stained cells in the granule cell layer over 72 h in control cultures grown in defined medium. We also found a slowly declining cell proliferation rate over the same time period under control conditions. We report evidence of early cell death in the granule cell layer after just 2 h exposure to 5 microM kainate, followed by a significant decrease in cell proliferation in the granule cell layer at 24 h. In contrast to control conditions, cell proliferation rose significantly in the kainate exposed cultures by 72 h back to levels seen at 2 h. There were no significant changes in cell labelling with antibody to activated caspase-3 between kainate treated and control cultures at any time point examined. Our results suggest that kainate-induced injury in the early postnatal hippocampus damages precursor cells contributing to a reduction in granule layer cell proliferation.
Collapse
Affiliation(s)
- Matthew Paul Sadgrove
- Division of Clinical Neurosciences, Southampton Neurosciences Group, School of Medicine, University of Southampton, Room 6207, Level 6, Biomedical Sciences Building, Bassett Crescent East, Southampton SO16 7PX, UK
| | | | | |
Collapse
|
49
|
Parfenova H, Carratu P, Tcheranova D, Fedinec A, Pourcyrous M, Leffler CW. Epileptic seizures cause extended postictal cerebral vascular dysfunction that is prevented by HO-1 overexpression. Am J Physiol Heart Circ Physiol 2005; 288:H2843-50. [PMID: 15681702 DOI: 10.1152/ajpheart.01274.2004] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The extended postictal state is characterized by neurological problems in patients. Inadequate blood supply to the brain and impaired cerebral autoregulation may contribute to seizure-induced neuronal damage. Recent evidence in newborn pigs indicates that activation of the antioxidative enzyme heme oxygenase (HO) at the onset of seizures is necessary for increased cerebral blood flow during the ictal episode and for normal cerebral vascular functioning during the immediate postictal period. We hypothesized that seizures cause prolonged postictal cerebral vascular dysfunction that can be accentuated by HO inhibition and rescued by HO overexpression. Cerebral vascular responses to endothelium-dependent (hypercapnia, bradykinin) and -independent (isoproterenol, sodium nitroprusside) stimuli were assessed 48 h after bicuculline-induced seizures in: 1) saline-control newborn piglets, 2) HO-inhibited animals (HO was inhibited by tin protoporphyrin, SnPP, 3 mg/kg iv), and 3) HO-overexpressing piglets (HO-1 was upregulated by cobalt protoporphyrin, CoPP, 50 mg/kg ip). Extended alterations of HO expression in cerebral microvessels were confirmed by measuring CO production and inducible HO (HO-1) and constitutive HO (HO-2) proteins. Our data provide evidence that seizures cause a severe, sustained, postictal cerebral vascular dysfunction as reflected by impaired vascular reactivity to physiologically relevant dilators. During the delayed postictal state, vascular reactivity to all dilator stimuli was reduced in saline control and, to a greater extent, in HO-inhibited animals. In CoPP-treated piglets, no reduction in postictal cerebral vascular reactivity was observed. These findings may indicate that CoPP prevents postictal cerebral vascular dysfunction by upregulating HO-1, a finding that might have implications for preventing postictal neurological complications.
Collapse
Affiliation(s)
- Helena Parfenova
- Dept. of Physiology, Univ. of Tennessee Health Science Center, 894 Union Ave., Memphis, TN 38163, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
The extent that status epilepticus (SE), but also brief seizures, affects neuronal structure and function has been the subject of much clinical and experimental research. There is a reliance on findings from animal research because there have been few prospective clinical studies. This review suggests that the features of seizure-induced injury in the immature brain compared with the adult brain are different and that duration of seizures (SE versus brief), number of seizures, cause of seizures, presence of pre-existing abnormalities, and genetics affect the injury. Increased awareness of age-specific injuries from seizure has promoted research to determine the circumstances under which seizures may produce permanent detrimental effects. Together with recent advances in functional neuroimaging, genomic investigation, and prospective human data, these studies are likely to substantially increase our knowledge of seizure-induced injury, leading to the development of improved algorithms for prevention and treatment of epilepsy.
Collapse
Affiliation(s)
- Sheryl R Haut
- Department of Neurology, Comprehensive Epilepsy Management Center, NY, USA
| | | | | |
Collapse
|