1
|
Adam CD, Schellinger ED, White A, Joksimovic SM, Takano H, Coulter DA. Chemogenetic Breakdown of the Dentate Gate Causes Seizures and Spatial Memory Deficits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.12.623184. [PMID: 39605633 PMCID: PMC11601345 DOI: 10.1101/2024.11.12.623184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The dentate gyrus has often been posited to act as a gate that dampens highly active afferent input into the hippocampus. Effective gating is thought to prevent seizure initiation and propagation in the hippocampus and support learning and memory processes. Pathological changes to DG circuitry that occur in temporal lobe epilepsy (TLE) can increase DG excitability and impair its gating ability which can contribute to seizures and cognitive deficits. There is evidence that TLE pathologies and seizures may independently contribute to learning and memory deficits in TLE through distinct mechanisms. These two factors are difficult to untangle since TLE pathologies can drive seizures, and seizures can worsen TLE pathologies. Here we assessed whether chemogenetically increasing dentate granule cell (DGC) excitability was enough to break down the dentate gate in the absence of TLE pathologies. We found that increasing excitability specifically in DGCs caused seizures in non-epileptic mice. Importantly, due to the modulatory nature of DREADD effects, seizures were driven by intrinsic circuit activity rather than direct activation of DGCs. These seizures resulted in a spatial memory deficit when induced after training in the spatial object recognition task and showed stereotypical patterns of activity in miniscope calcium recordings. Our results provide direct support for the dentate gate hypothesis since seizures could be induced in non-epileptic animals by artificially degrading the dentate gate with chemogenetics in the absence of epilepsy pathologies.
Collapse
Affiliation(s)
- Christopher D Adam
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Emily D Schellinger
- The Research Institute of the Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Alicia White
- The Research Institute of the Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Srdjan M Joksimovic
- The Research Institute of the Children's Hospital of Philadelphia, Philadelphia, PA 19104
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Hajime Takano
- The Research Institute of the Children's Hospital of Philadelphia, Philadelphia, PA 19104
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Douglas A Coulter
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- The Research Institute of the Children's Hospital of Philadelphia, Philadelphia, PA 19104
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
2
|
Huang CS, Houser CR. Alterations and Imbalance of Dorsal and Ventral Mossy Cells in a Mouse Model of Epilepsy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.07.622490. [PMID: 39574697 PMCID: PMC11581000 DOI: 10.1101/2024.11.07.622490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2024]
Abstract
Mossy cells (MCs) in the hilus of the dentate gyrus (DG) are important for regulating activity of dentate granule cells and are particularly vulnerable to excitotoxic damage in epilepsy. Recent studies have demonstrated that MCs in the dorsal and ventral DG differ in the patterns of their axonal projections and neurochemical identities. Such differences raised questions about the vulnerability and plasticity of dorsal and ventral MCs in epilepsy and led to this study using a mouse pilocarpine model of epilepsy. Dorsal MCs were labeled by transfection of Cre-dependent eYFP in the dorsal DG of Calcrl-Cre mice that express Cre selectively in MCs. Ventral MCs were labeled with calretinin (CR), which labels ventral but not dorsal MCs. At 6-8 weeks after pilocarpine treatment, MC loss and axonal projections of remaining MCs were studied in control and pilocarpine-treated mice with confocal microscopy. Dorsal MCs were severely depleted, but many ventral MCs remained, and quantitative analysis of GluA2-labeled hilar neurons demonstrated a proportionally greater loss of dorsal MCs (77.6% loss) than ventral MCs (21.5% loss). Loss of dorsal MCs led to a marked reduction in the dorsal commissural pathway, while the remaining ventral MCs maintained a prominent, though reduced, ventral to dorsal association pathway. In pilocarpine-treated animals, a plexus of CR-labeled fibers extended into the middle molecular layer, suggesting axonal sprouting of remaining ventral MCs, with some of these fibers in contact with parvalbumin-labeled dendrites. These findings suggest that dorsal and ventral MCs differ in their vulnerability to seizure-induced damage in this animal model, creating an imbalance between the dorsal and ventral MC pathways that could alter the excitatory/inhibitory balance within the dentate gyrus.
Collapse
Affiliation(s)
- Christine S. Huang
- Departments of Neurobiology, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California 90095
| | - Carolyn R. Houser
- Departments of Neurobiology, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California 90095
- Brain Research Institute, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California 90095
| |
Collapse
|
3
|
Bakhtazad A, Kabbaj M, Garmabi B, Joghataei MT. The role of CART peptide in learning and memory: A potential therapeutic target in memory-related disorders. Peptides 2024; 181:171298. [PMID: 39317295 DOI: 10.1016/j.peptides.2024.171298] [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] [Received: 04/27/2024] [Revised: 08/19/2024] [Accepted: 09/20/2024] [Indexed: 09/26/2024]
Abstract
Cocaine and amphetamine-regulated transcript (CART) mRNA and peptide are vastly expressed in both cortical and subcortical brain areas and are involved in critical cognitive functions. CART peptide (CARTp), described in reward-related brain structures, regulates drug-induced learning and memory, and its role appears specific to psychostimulants. However, many other drugs of abuse, such as alcohol, opiates, nicotine, and caffeine, have been shown to alter the expression levels of CART mRNA and peptides in brain structures directly or indirectly associated with learning and memory processes. However, the number of studies demonstrating the contribution of CARTp in learning and memory is still minimal. Notably, the exact cellular and molecular mechanisms underlying CARTp effects are still unknown. The discoveries that CARTp effects are mediated through a putative G-protein coupled receptor and activation of cellular signaling cascades via NMDA receptor-coupled ERK have enhanced our knowledge about the action of this neuropeptide and allowed us to comprehend better CARTp exact cellular/molecular mechanisms that could mediate drug-induced changes in learning and memory functions. Unfortunately, these efforts have been impeded by the lack of suitable and specific CARTp receptor antagonists. In this review, following a short introduction about CARTp, we report on current knowledge about CART's roles in learning and memory processes and its recently described role in memory-related neurological disorders. We will also discuss the importance of further investigating how CARTp interacts with its receptor(s) and other neurotransmitter systems to influence learning and memory functions. This topic is sure to intrigue and motivate further exploration in the field of neuroscience.
Collapse
Affiliation(s)
- Atefeh Bakhtazad
- Cellular and Molecular Research Center, Deputy of Research and Technology, Iran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mohamed Kabbaj
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306-1270, United States; Program of Neuroscience, Florida State University, Tallahassee, FL 32306-1270, United States
| | - Behzad Garmabi
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mohammad Taghi Joghataei
- Cellular and Molecular Research Center, Deputy of Research and Technology, Iran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Jain S, LaFrancois JJ, Gerencer K, Botterill JJ, Kennedy M, Criscuolo C, Scharfman HE. Increasing adult-born neurons protects mice from epilepsy. eLife 2024; 12:RP90893. [PMID: 39446467 PMCID: PMC11501206 DOI: 10.7554/elife.90893] [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] [Indexed: 10/26/2024] Open
Abstract
Neurogenesis occurs in the adult brain in the hippocampal dentate gyrus, an area that contains neurons which are vulnerable to insults and injury, such as severe seizures. Previous studies showed that increasing adult neurogenesis reduced neuronal damage after these seizures. Because the damage typically is followed by chronic life-long seizures (epilepsy), we asked if increasing adult-born neurons would prevent epilepsy. Adult-born neurons were selectively increased by deleting the pro-apoptotic gene Bax from Nestin-expressing progenitors. Tamoxifen was administered at 6 weeks of age to conditionally delete Bax in Nestin-CreERT2Baxfl/fl mice. Six weeks after tamoxifen administration, severe seizures (status epilepticus; SE) were induced by injection of the convulsant pilocarpine. After mice developed epilepsy, seizure frequency was quantified for 3 weeks. Mice with increased adult-born neurons exhibited fewer chronic seizures. Postictal depression was reduced also. These results were primarily in female mice, possibly because they were more affected by Bax deletion than males, consistent with sex differences in Bax. The female mice with enhanced adult-born neurons also showed less neuronal loss of hilar mossy cells and hilar somatostatin-expressing neurons than wild-type females or males, which is notable because loss of these two hilar cell types is implicated in epileptogenesis. The results suggest that selective Bax deletion to increase adult-born neurons can reduce experimental epilepsy, and the effect shows a striking sex difference. The results are surprising in light of past studies showing that suppressing adult-born neurons can also reduce chronic seizures.
Collapse
Affiliation(s)
- Swati Jain
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - John J LaFrancois
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Kasey Gerencer
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Justin J Botterill
- Department of Anatomy, Physiology, & Pharmacology, College of Medicine, University of SaskatchewanSaskatoonCanada
| | - Meghan Kennedy
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Chiara Criscuolo
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
| | - Helen E Scharfman
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
- Departments of Neuroscience & Physiology, Psychiatry, and the Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| |
Collapse
|
5
|
Wang SH, Lee DS, Kim TH, Kim JE, Kang TC. Reciprocal regulation of oxidative stress and mitochondrial fission augments parvalbumin downregulation through CDK5-DRP1- and GPx1-NF-κB signaling pathways. Cell Death Dis 2024; 15:707. [PMID: 39349423 PMCID: PMC11443148 DOI: 10.1038/s41419-024-07050-5] [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: 02/25/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 10/02/2024]
Abstract
Loss of parvalbumin (PV) expressing neurons (PV neurons) is relevant to the underlying mechanisms of the pathogenesis of neurological and psychiatric diseases associated with the dysregulation of neuronal excitatory networks and brain metabolism. Although PV modulates mitochondrial morphology, volume and dynamics, it is largely unknown whether mitochondrial dynamics affect PV expression and what the molecular events are responsible for PV neuronal degeneration. In the present study, L-buthionine sulfoximine (BSO, an inhibitor of glutathione synthesis) did not degenerate PV neurons under physiological condition. However, BSO-induced oxidative stress decreased PV expression and facilitated cyclin-dependent kinase 5 (CDK5) tyrosine (Y) 15 phosphorylation, dynamin-related protein 1 (DRP1)-mediated mitochondrial fission and glutathione peroxidase-1 (GPx1) downregulation in PV neurons. Co-treatment of roscovitine (a CDK5 inhibitor) or mitochondrial division inhibitor-1 (Mdivi-1, an inhibitor of mitochondrial fission) attenuated BSO-induced PV downregulation. WY14643 (an inducer of mitochondrial fission) reduced PV expression without affecting CDK5 Y15 phosphorylation. Following status epilepticus (SE), CDK5 Y15 phosphorylation and mitochondrial fission were augmented in PV neurons. These were accompanied by reduced GPx1-mediated inhibition of NF-κB p65 serine (S) 536 phosphorylation. N-acetylcysteine (NAC), roscovitine and Mdivi-1 ameliorated SE-induced PV neuronal degeneration by mitigating CDK5 Y15 hyperphosphorylation, aberrant mitochondrial fragmentation and reduced GPx1-mediated NF-κB inhibition. Furthermore, SN50 (a NF-κB inhibitor) alleviated SE-induced PV neuronal degeneration, independent of dysregulation of mitochondrial fission, CDK5 hyperactivation and GPx1 downregulation. These findings provide an evidence that oxidative stress may activate CDK5-DRP1- and GPx1-NF-κB-mediated signaling pathways, which would be possible therapeutic targets for preservation of PV neurons in various diseases.
Collapse
Affiliation(s)
- Su Hyeon Wang
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Duk-Shin Lee
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Tae-Hyun Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Ji-Eun Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea.
| | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea.
| |
Collapse
|
6
|
Jain S, LaFrancois JJ, Gerencer K, Botterill JJ, Kennedy M, Criscuolo C, Scharfman HE. Increasing adult-born neurons protects mice from epilepsy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.08.548217. [PMID: 37502909 PMCID: PMC10369878 DOI: 10.1101/2023.07.08.548217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Neurogenesis occurs in the adult brain in the hippocampal dentate gyrus, an area that contains neurons which are vulnerable to insults and injury, such as severe seizures. Previous studies showed that increasing adult neurogenesis reduced neuronal damage after these seizures. Because the damage typically is followed by chronic life-long seizures (epilepsy), we asked if increasing adult-born neurons would prevent epilepsy. Adult-born neurons were selectively increased by deleting the pro-apoptotic gene Bax from Nestin-expressing progenitors. Tamoxifen was administered at 6 weeks of age to conditionally delete Bax in Nestin-CreERT2 Bax fl/fl mice. Six weeks after tamoxifen administration, severe seizures (status epilepticus; SE) were induced by injection of the convulsant pilocarpine. After mice developed epilepsy, seizure frequency was quantified for 3 weeks. Mice with increased adult-born neurons exhibited fewer chronic seizures. Postictal depression was reduced also. These results were primarily in female mice, possibly because they were the more affected by Bax deletion than males, consistent with sex differences in Bax. The female mice with enhanced adult-born neurons also showed less neuronal loss of hilar mossy cells and hilar somatostatin-expressing neurons than wild type females or males, which is notable because these two hilar cell types are implicated in epileptogenesis. The results suggest that selective Bax deletion to increase adult-born neurons can reduce experimental epilepsy, and the effect shows a striking sex difference. The results are surprising in light of past studies showing that suppressing adult-born neurons can also reduce chronic seizures.
Collapse
Affiliation(s)
- Swati Jain
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
| | - John J. LaFrancois
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
| | - Kasey Gerencer
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Current address: Department of Psychology, The University of Maine, Orono, ME 04469
| | - Justin J. Botterill
- Department of Anatomy, Physiology, & Pharmacology, College of Medicine, Saskatoon, SK S7N 5E5
| | - Meghan Kennedy
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
| | - Chiara Criscuolo
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY 10016
| | - Helen E. Scharfman
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY 10016
- Departments of Neuroscience & Physiology, Psychiatry, and the New York University, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016
| |
Collapse
|
7
|
Alves M, Gil B, Villegas-Salmerón J, Salari V, Martins-Ferreira R, Arribas Blázquez M, Menéndez Méndez A, Da Rosa Gerbatin R, Smith J, de Diego-Garcia L, Conte G, Sierra-Marquez J, Merino Serrais P, Mitra M, Fernandez Martin A, Wang Y, Kesavan J, Melia C, Parras A, Beamer E, Zimmer B, Heiland M, Cavanagh B, Parcianello Cipolat R, Morgan J, Teng X, Prehn JHM, Fabene PF, Bertini G, Artalejo AR, Ballestar E, Nicke A, Olivos-Oré LA, Connolly NMC, Henshall DC, Engel T. Opposing effects of the purinergic P2X7 receptor on seizures in neurons and microglia in male mice. Brain Behav Immun 2024; 120:121-140. [PMID: 38777288 DOI: 10.1016/j.bbi.2024.05.023] [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] [Received: 12/23/2023] [Revised: 04/28/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND The purinergic ATP-gated P2X7 receptor (P2X7R) is increasingly recognized to contribute to pathological neuroinflammation and brain hyperexcitability. P2X7R expression has been shown to be increased in the brain, including both microglia and neurons, in experimental models of epilepsy and patients. To date, the cell type-specific downstream effects of P2X7Rs during seizures remain, however, incompletely understood. METHODS Effects of P2X7R signaling on seizures and epilepsy were analyzed in induced seizure models using male mice including the kainic acid model of status epilepticus and pentylenetetrazole model and in male and female mice in a genetic model of Dravet syndrome. RNA sequencing was used to analyze P2X7R downstream signaling during seizures. To investigate the cell type-specific role of the P2X7R during seizures and epilepsy, we generated mice lacking exon 2 of the P2rx7 gene in either microglia (P2rx7:Cx3cr1-Cre) or neurons (P2rx7:Thy-1-Cre). To investigate the protective potential of overexpressing P2X7R in GABAergic interneurons, P2X7Rs were overexpressed using adeno-associated virus transduction under the mDlx promoter. RESULTS RNA sequencing of hippocampal tissue from wild-type and P2X7R knock-out mice identified both glial and neuronal genes, in particular genes involved in GABAergic signaling, under the control of the P2X7R following seizures. Mice with deleted P2rx7 in microglia displayed less severe acute seizures and developed a milder form of epilepsy, and microglia displayed an anti-inflammatory molecular profile. In contrast, mice lacking P2rx7 in neurons showed a more severe seizure phenotype when compared to epileptic wild-type mice. Analysis of single-cell expression data revealed that human P2RX7 expression is elevated in the hippocampus of patients with temporal lobe epilepsy in excitatory and inhibitory neurons. Functional studies determined that GABAergic interneurons display increased responses to P2X7R activation in experimental epilepsy. Finally, we show that viral transduction of P2X7R in GABAergic interneurons protects against evoked and spontaneous seizures in experimental temporal lobe epilepsy and in mice lacking Scn1a, a model of Dravet syndrome. CONCLUSIONS Our results suggest a dual and opposing action of P2X7R in epilepsy and suggest P2X7R overexpression in GABAergic interneurons as a novel therapeutic strategy for acquired and, possibly, genetic forms of epilepsy.
Collapse
Affiliation(s)
- Mariana Alves
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Beatriz Gil
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Javier Villegas-Salmerón
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland; The SFI Centre for Research Training in Genomics Data Science, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Valentina Salari
- Department of Neurosciences, Biomedicine and Movement Sciences, School of Medicine, University of Verona, 37134 Verona, Italy
| | - Ricardo Martins-Ferreira
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Barcelona, Spain; Immunogenetics Laboratory, Molecular Pathology and Immunology, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Autoimmunity and Neuroscience Group, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal; ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| | - Marina Arribas Blázquez
- Department of Pharmacology and Toxicology, Veterinary Faculty, Universidad Complutense de Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - Aida Menéndez Méndez
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670, Villaviciosa de Odon, Spain
| | - Rogerio Da Rosa Gerbatin
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| | - Jonathon Smith
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| | - Laura de Diego-Garcia
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; Ocupharm Research Group, Faculty of Optics and Optometry, Complutense University of Madrid, Avda. Arcos de Jalon, 118 (28037), Madrid, Spain
| | - Giorgia Conte
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Juan Sierra-Marquez
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany; Laboratorio Cajal de Circuitos Corticales (CTB), Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus Montegancedo S/N, Pozuelo de Alarcon, 28223 Madrid, Spain; Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28002, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid 28031, Spain
| | - Paula Merino Serrais
- Laboratorio Cajal de Circuitos Corticales (CTB), Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus Montegancedo S/N, Pozuelo de Alarcon, 28223 Madrid, Spain
| | - Meghma Mitra
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Ana Fernandez Martin
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Yitao Wang
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jaideep Kesavan
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| | - Ciara Melia
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; VivoArchitect, Route de la Corniche 5, 1066 Epalinges, Vaud, Switzerland
| | - Alberto Parras
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Edward Beamer
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Béla Zimmer
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Mona Heiland
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| | - Brenton Cavanagh
- Cellular and Molecular Imaging Core, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Rafael Parcianello Cipolat
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| | - James Morgan
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, UK
| | - Xinchen Teng
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jochen H M Prehn
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| | - Paolo F Fabene
- Department of Neurosciences, Biomedicine and Movement Sciences, School of Medicine, University of Verona, 37134 Verona, Italy; Section of Anatomy and Histology, Department of Neurosciences, Biomedicine, and Movement Science, Faculty of Medicine, University of Verona, Verona, Italy; Section of Innovation Biomedicine, Department of Engineering for Innovation Medicine, Faculty of Medicine, University of Verona, Verona, Italy
| | - Giuseppe Bertini
- Department of Neurosciences, Biomedicine and Movement Sciences, School of Medicine, University of Verona, 37134 Verona, Italy
| | - Antonio R Artalejo
- Department of Pharmacology and Toxicology, Veterinary Faculty, Universidad Complutense de Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - Esteban Ballestar
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Barcelona, Spain; Epigenetics in Inflammatory and Metabolic Diseases Laboratory, Health Science Center (HSC), East China Normal University (ECNU), Shanghai 200241, China
| | - Annette Nicke
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Luis A Olivos-Oré
- Department of Pharmacology and Toxicology, Veterinary Faculty, Universidad Complutense de Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - Niamh M C Connolly
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| | - David C Henshall
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| | - Tobias Engel
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland.
| |
Collapse
|
8
|
Noguchi H, Arela JC, Ngo T, Cocas L, Pleasure S. Shh from mossy cells contributes to preventing NSC pool depletion after seizure-induced neurogenesis and in aging. eLife 2023; 12:RP91263. [PMID: 38079471 PMCID: PMC10712957 DOI: 10.7554/elife.91263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
Epileptic seizures induce aberrant neurogenesis from resident neural stem cells (NSCs) in the dentate gyrus of the adult mouse hippocampus, which has been implicated in depletion of the NSC pool and impairment of hippocampal function. However, the mechanisms regulating neurogenesis after seizures remain unknown. Here, we demonstrate that Sonic hedgehog (Shh) from mossy cells is a major source of Shh signaling activity after seizures, by which mossy cells contribute to seizure-induced neurogenesis and maintenance of the NSC pool. Deletion of Shh from mossy cells attenuates seizure-induced neurogenesis. Moreover, in the absence of Shh from mossy cells, NSCs pool are prematurely depleted after seizure-induced proliferation, and NSCs have impaired self-renewal. Likewise, lack of Shh from mossy cells accelerates age-related decline of the NSC pool with accompanying reduction of self-renewal of NSCs outside the context of pathology such as seizures. Together, our findings indicate that Shh from mossy cells is critical to maintain NSCs and to prevent exhaustion from excessive consumption in aging and after seizures.
Collapse
Affiliation(s)
- Hirofumi Noguchi
- Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
| | - Jessica Chelsea Arela
- Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
| | - Thomas Ngo
- Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
| | - Laura Cocas
- Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
- Santa Clara University, Biology Department, Neuroscience ProgramSanta ClaraUnited States
| | - Samuel Pleasure
- Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
- Programs in Neuroscience and Developmental & Stem Cell Biology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San FranciscoSan FranciscoUnited States
| |
Collapse
|
9
|
Pelkey KA, Vargish GA, Pellegrini LV, Calvigioni D, Chapeton J, Yuan X, Hunt S, Cummins AC, Eldridge MAG, Pickel J, Chittajallu R, Averbeck BB, Tóth K, Zaghloul K, McBain CJ. Evolutionary conservation of hippocampal mossy fiber synapse properties. Neuron 2023; 111:3802-3818.e5. [PMID: 37776852 PMCID: PMC10841147 DOI: 10.1016/j.neuron.2023.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 07/03/2023] [Accepted: 09/06/2023] [Indexed: 10/02/2023]
Abstract
Various specialized structural/functional properties are considered essential for contextual memory encoding by hippocampal mossy fiber (MF) synapses. Although investigated to exquisite detail in model organisms, synapses, including MFs, have undergone minimal functional interrogation in humans. To determine the translational relevance of rodent findings, we evaluated MF properties within human tissue resected to treat epilepsy. Human MFs exhibit remarkably similar hallmark features to rodents, including AMPA receptor-dominated synapses with small contributions from NMDA and kainate receptors, large dynamic range with strong frequency facilitation, NMDA receptor-independent presynaptic long-term potentiation, and strong cyclic AMP (cAMP) sensitivity of release. Array tomography confirmed the evolutionary conservation of MF ultrastructure. The astonishing congruence of rodent and human MF core features argues that the basic MF properties delineated in animal models remain critical to human MF function. Finally, a selective deficit in GABAergic inhibitory tone onto human MF postsynaptic targets suggests that unrestrained detonator excitatory drive contributes to epileptic circuit hyperexcitability.
Collapse
Affiliation(s)
- Kenneth A Pelkey
- Eunice Kennedy Shriver National Institute of Child Health and Human Development Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Geoffrey A Vargish
- Eunice Kennedy Shriver National Institute of Child Health and Human Development Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Leonardo V Pellegrini
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Brain and Mind Research Institute, Ottawa, ON K1H 8M5, Canada
| | - Daniela Calvigioni
- Eunice Kennedy Shriver National Institute of Child Health and Human Development Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Julio Chapeton
- National Institute of Neurological Disorders and Stroke Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xiaoqing Yuan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Steven Hunt
- Eunice Kennedy Shriver National Institute of Child Health and Human Development Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alex C Cummins
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark A G Eldridge
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - James Pickel
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ramesh Chittajallu
- Eunice Kennedy Shriver National Institute of Child Health and Human Development Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bruno B Averbeck
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Katalin Tóth
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Brain and Mind Research Institute, Ottawa, ON K1H 8M5, Canada
| | - Kareem Zaghloul
- National Institute of Neurological Disorders and Stroke Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chris J McBain
- Eunice Kennedy Shriver National Institute of Child Health and Human Development Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
10
|
Hung YC, Wu YJ, Chien ME, Lin YT, Tsai CF, Hsu KS. Loss of oxytocin receptors in hilar mossy cells impairs social discrimination. Neurobiol Dis 2023; 187:106311. [PMID: 37769745 DOI: 10.1016/j.nbd.2023.106311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/02/2023] Open
Abstract
Hippocampal oxytocin receptor (OXTR) signaling is crucial for discrimination of social stimuli to guide social recognition, but circuit mechanisms and cell types involved remain incompletely understood. Here, we report a role for OXTR-expressing hilar mossy cells (MCs) of the dentate gyrus in social stimulus discrimination by regulating granule cell (GC) activity. Using a Cre-loxP recombination approach, we found that ablation of Oxtr from MCs impairs discrimination of social, but not object, stimuli in adult male mice. Ablation of MC Oxtr increases spontaneous firing rate of GCs, synaptic excitation to inhibition ratio of MC-to-GC circuit, and GC firing when temporally associated with the lateral perforant path inputs. Using mouse hippocampal slices, we found that bath application of OXTR agonist [Thr4,Gly7]-oxytocin causes membrane depolarization and increases MC firing activity. Optogenetic activation of MC-to-GC circuit ameliorates social discrimination deficit in MC OXTR deficient mice. Together, our results uncover a previously unknown role of MC OXTR signaling for discrimination of social stimuli and delineate a MC-to-GC circuit responsible for social information processing.
Collapse
Affiliation(s)
- Yu-Chieh Hung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yi-Jen Wu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70457, Taiwan; Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| | - Miao-Er Chien
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70457, Taiwan
| | - Yu-Ting Lin
- Institute of Systems Neuroscience, College of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Cheng-Fang Tsai
- Department of Physical Medicine and Rehabilitation, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
| | - Kuei-Sen Hsu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.
| |
Collapse
|
11
|
Noguchi H, Arela JC, Ngo TT, Cocas L, Pleasure SJ. Shh from mossy cells contributes to preventing NSC pool depletion after seizure-induced neurogenesis and in aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.21.554173. [PMID: 37662214 PMCID: PMC10473584 DOI: 10.1101/2023.08.21.554173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Epileptic seizures induce aberrant neurogenesis from resident neural stem cells (NSCs) in the dentate gyrus of the adult mouse hippocampus, which has been implicated in depletion of the NSC pool and impairment of hippocampal function. However, the mechanisms regulating neurogenesis after seizures remain unknown. Here we demonstrate that Shh from mossy cells is a major source of Shh signaling activity after seizures, by which mossy cells contribute to seizure-induced neurogenesis and maintenance of the NSC pool. Deletion of Shh from mossy cells attenuates seizure-induced neurogenesis. Moreover, in the absence of Shh from mossy cells, NSCs pool are prematurely depleted after seizure-induced proliferation, and NSCs have impaired self-renewal. Likewise, lack of Shh from mossy cells accelerates age-related decline of the NSC pool with accompanying reduction of self-renewal of NSCs outside the context of pathology such as seizures. Together, our findings indicate that Shh from mossy cells is critical to maintain NSCs and to prevent exhaustion from excessive consumption in aging and after seizures.
Collapse
|
12
|
Vivien J, El Azraoui A, Lheraux C, Lanore F, Aouizerate B, Herry C, Humeau Y, Bienvenu TCM. Axo-axonic cells in neuropsychiatric disorders: a systematic review. Front Cell Neurosci 2023; 17:1212202. [PMID: 37435048 PMCID: PMC10330806 DOI: 10.3389/fncel.2023.1212202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/09/2023] [Indexed: 07/13/2023] Open
Abstract
Imbalance between excitation and inhibition in the cerebral cortex is one of the main theories in neuropsychiatric disorder pathophysiology. Cortical inhibition is finely regulated by a variety of highly specialized GABAergic interneuron types, which are thought to organize neural network activities. Among interneurons, axo-axonic cells are unique in making synapses with the axon initial segment of pyramidal neurons. Alterations of axo-axonic cells have been proposed to be implicated in disorders including epilepsy, schizophrenia and autism spectrum disorder. However, evidence for the alteration of axo-axonic cells in disease has only been examined in narrative reviews. By performing a systematic review of studies investigating axo-axonic cells and axo-axonic communication in epilepsy, schizophrenia and autism spectrum disorder, we outline convergent findings and discrepancies in the literature. Overall, the implication of axo-axonic cells in neuropsychiatric disorders might have been overstated. Additional work is needed to assess initial, mostly indirect findings, and to unravel how defects in axo-axonic cells translates to cortical dysregulation and, in turn, to pathological states.
Collapse
Affiliation(s)
- Juliette Vivien
- Université de Bordeaux, Inserm Neurocentre Magendie U1215, Bordeaux, France
| | - Anass El Azraoui
- Université de Bordeaux, Inserm Neurocentre Magendie U1215, Bordeaux, France
- Univ Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Cloé Lheraux
- Université de Bordeaux, Inserm Neurocentre Magendie U1215, Bordeaux, France
| | - Frederic Lanore
- Centre Hospitalier Charles Perrens, Inserm Neurocentre Magendie U1215, Bordeaux, France
| | - Bruno Aouizerate
- Université de Bordeaux, Inserm Neurocentre Magendie U1215, Bordeaux, France
- Centre Hospitalier Charles Perrens, Inserm Neurocentre Magendie U1215, Bordeaux, France
- INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Cyril Herry
- Université de Bordeaux, Inserm Neurocentre Magendie U1215, Bordeaux, France
| | - Yann Humeau
- Univ Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Thomas C. M. Bienvenu
- Université de Bordeaux, Inserm Neurocentre Magendie U1215, Bordeaux, France
- Centre Hospitalier Charles Perrens, Inserm Neurocentre Magendie U1215, Bordeaux, France
| |
Collapse
|
13
|
Stöber TM, Batulin D, Triesch J, Narayanan R, Jedlicka P. Degeneracy in epilepsy: multiple routes to hyperexcitable brain circuits and their repair. Commun Biol 2023; 6:479. [PMID: 37137938 PMCID: PMC10156698 DOI: 10.1038/s42003-023-04823-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 04/06/2023] [Indexed: 05/05/2023] Open
Abstract
Due to its complex and multifaceted nature, developing effective treatments for epilepsy is still a major challenge. To deal with this complexity we introduce the concept of degeneracy to the field of epilepsy research: the ability of disparate elements to cause an analogous function or malfunction. Here, we review examples of epilepsy-related degeneracy at multiple levels of brain organisation, ranging from the cellular to the network and systems level. Based on these insights, we outline new multiscale and population modelling approaches to disentangle the complex web of interactions underlying epilepsy and to design personalised multitarget therapies.
Collapse
Affiliation(s)
- Tristan Manfred Stöber
- Frankfurt Institute for Advanced Studies, 60438, Frankfurt am Main, Germany
- Institute for Neural Computation, Faculty of Computer Science, Ruhr University Bochum, 44801, Bochum, Germany
- Epilepsy Center Frankfurt Rhine-Main, Department of Neurology, Goethe University, 60590, Frankfurt, Germany
| | - Danylo Batulin
- Frankfurt Institute for Advanced Studies, 60438, Frankfurt am Main, Germany
- CePTER - Center for Personalized Translational Epilepsy Research, Goethe University, 60590, Frankfurt, Germany
- Faculty of Computer Science and Mathematics, Goethe University, 60486, Frankfurt, Germany
| | - Jochen Triesch
- Frankfurt Institute for Advanced Studies, 60438, Frankfurt am Main, Germany
| | - Rishikesh Narayanan
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India
| | - Peter Jedlicka
- ICAR3R - Interdisciplinary Centre for 3Rs in Animal Research, Faculty of Medicine, Justus Liebig University Giessen, 35390, Giessen, Germany.
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University, 60590, Frankfurt am Main, Germany.
| |
Collapse
|
14
|
Kim JE, Lee DS, Kim TH, Kang TC. CDDO-Me Attenuates CA1 Neuronal Death by Facilitating RalBP1-Mediated Mitochondrial Fission and 4-HNE Efflux in the Rat Hippocampus Following Status Epilepticus. Antioxidants (Basel) 2022; 11:985. [PMID: 35624848 PMCID: PMC9137584 DOI: 10.3390/antiox11050985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 11/16/2022] Open
Abstract
Ras-related protein Ral-A (RalA)-binding protein 1 (RalBP1, also known as Ral-interacting protein of 76 kDa (RLIP76) or Ral-interacting protein 1 (RLIP1 or RIP1)) is involved in the efflux of 4-hydroxynonenal (4-HNE, an end product of lipid peroxidation), as well as mitochondrial fission. In the present study, we found that 2-cyano-3,12-dioxo-oleana-1,9(11)-dien-28-oic acid methyl ester (CDDO-Me) attenuated CA1 neuronal death and aberrant mitochondrial elongations in these neurons coupled with enhanced RalBP1 expression and reduced 4-HNE levels following status epilepticus (SE). RalBP1 knockdown did not affect mitochondrial dynamics and CA1 neuronal death under physiological and post-SE conditions. Following SE, however, cotreatment of RalBP1 siRNA diminished the effect of CDDO-Me on 4-HNE levels, mitochondrial hyperfusion in CA1 neurons, and CA1 neuronal death. These findings indicate that CDDO-Me may ameliorate CA1 neuronal death by facilitating RalBP1-mediated 4-HNE efflux and mitochondrial fission following SE. Therefore, our findings suggest that increased RalBP1 expression/activity may be one of the considerable targets to protect neurons from SE.
Collapse
Affiliation(s)
| | | | | | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology and Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon 24252, Korea; (J.-E.K.); (D.-S.L.); (T.-H.K.)
| |
Collapse
|
15
|
Yang C, Shi Y, Li X, Guan L, Li H, Lin J. Cadherins and the pathogenesis of epilepsy. Cell Biochem Funct 2022; 40:336-348. [PMID: 35393670 DOI: 10.1002/cbf.3699] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/22/2022] [Accepted: 03/12/2022] [Indexed: 12/13/2022]
Abstract
Epilepsy is a nervous system disease caused by abnormal discharge of brain neurons, which is characterized by recurrent seizures. The factors that induce epilepsy include genetic and environmental factors. Genetic factors are important pathogenic factors of epilepsy, such as epilepsy caused by protocadherin-19 (PCDH-19) mutation, which is an X-linked genetic disease. It is more common in female heterozygotes, which are caused by mutations in the PCDH-19 gene. Epilepsy caused by environmental factors is mainly caused by brain injury, which is commonly caused by brain tumors, brain surgery, or trauma to the brain. In addition, the pathogenesis of epilepsy is closely related to abnormalities in some signaling pathways. The Wnt/β-catenin signaling pathway is considered a new target for the treatment of epilepsy. This review summarizes these factors inducing epilepsy and the research hypotheses regarding the pathogenesis of epilepsy. The focus of this review centers on cadherins and the pathogenesis of epilepsy. We analyzed the pathogenesis of epilepsy induced by N-cadherin and PCDH-19 in the cadherin family members. Finally, we expect that in the future, new breakthroughs will be made in the study of the pathogenesis and mechanism of epilepsy at the cellular and molecular levels.
Collapse
Affiliation(s)
- Ciqing Yang
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang, China
| | - Yaping Shi
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Xiaoying Li
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Lihong Guan
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Han Li
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Juntang Lin
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang, China
| |
Collapse
|
16
|
Ríos JL, Schinella GR, Moragrega I. Phenolics as GABA A Receptor Ligands: An Updated Review. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27061770. [PMID: 35335130 PMCID: PMC8953830 DOI: 10.3390/molecules27061770] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 11/16/2022]
Abstract
Natural products can act as potential GABA modulators, avoiding the undesirable effects of traditional pharmacology used for the inhibition of the central nervous system such as benzodiazepines (BZD). Phenolics, especially flavonoids and phlorotannins, have been considered as modulators of the BZD-site of GABAA receptors (GABAARs), with sedative, anxiolytic or anticonvulsant effects. However, the wide chemical structural variability of flavonoids shows their potential action at more than one additional binding site on GABAARs, which may act either negatively, positively, by neutralizing GABAARs, or directly as allosteric agonists. Therefore, the aim of the present review is to compile and discuss an update of the role of phenolics, namely as pharmacological targets involving dysfunctions of the GABA system, analyzing both their different compounds and their mechanism as GABAergic modulators. We focus this review on articles written in English since the year 2010 until the present. Of course, although more research would be necessary to fully establish the type specificity of phenolics and their pharmacological activity, the evidence supports their potential as GABAAR modulators, thereby favoring their inclusion in the development of new therapeutic targets based on natural products. Specifically, the data compiled in this review allows for the directing of future research towards ortho-dihydroxy diterpene galdosol, the flavonoids isoliquiritigenin (chalcone), rhusflavone and agathisflavone (biflavonoids), as well as the phlorotannins, dieckol and triphlorethol A. Clinically, flavonoids are the most interesting phenolics due to their potential as anticonvulsant and anxiolytic drugs, and phlorotannins are also of interest as sedative agents.
Collapse
Affiliation(s)
- José-Luis Ríos
- Departament de Farmacologia, Facultat de Farmàcia, Universitat de València, Av. Vicent Andrés Estellés s/n, Burjassot, 46100 Valencia, Spain
- Correspondence:
| | - Guillermo R. Schinella
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata BA1900, Argentina;
- Instituto de Ciencias de la Salud, UNAJ-CICPBA, Florencio Varela BA1888, Argentina
| | - Inés Moragrega
- Departament de Psicobiologia, Facultat de Psicologia, Universitat de València, Av. Blasco Ibáñez 21, 46010 Valencia, Spain;
| |
Collapse
|
17
|
Smiley JF, Bleiwas C, Canals-Baker S, Williams SZ, Sears R, Teixeira CM, Wilson DA, Saito M. Neonatal ethanol causes profound reduction of cholinergic cell number in the basal forebrain of adult animals. Alcohol 2021; 97:1-11. [PMID: 34464696 DOI: 10.1016/j.alcohol.2021.08.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 05/24/2021] [Accepted: 08/12/2021] [Indexed: 11/26/2022]
Abstract
In animal models that mimic human third-trimester fetal development, ethanol causes substantial cellular apoptosis in the brain, but for most brain structures, the extent of permanent neuron loss that persists into adulthood is unknown. We injected ethanol into C57BL/6J mouse pups at postnatal day 7 (P7) to model human late-gestation ethanol toxicity, and then used stereological methods to investigate adult cell numbers in several subcortical neurotransmitter systems that project extensively in the forebrain to regulate arousal states. Ethanol treatment caused especially large reductions (34-42%) in the cholinergic cells of the basal forebrain, including cholinergic cells in the medial septal/vertical diagonal band nuclei (Ch1/Ch2) and in the horizontal diagonal band/substantia innominata/nucleus basalis nuclei (Ch3/Ch4). Cell loss was also present in non-cholinergic basal forebrain cells, as demonstrated by 34% reduction of parvalbumin-immunolabeled GABA cells and 25% reduction of total Nissl-stained neurons in the Ch1/Ch2 region. In contrast, cholinergic cells in the striatum were reduced only 12% by ethanol, and those of the brainstem pedunculopontine/lateral dorsal tegmental nuclei (Ch5/Ch6) were not significantly reduced. Similarly, ethanol did not significantly reduce dopamine cells of the ventral tegmental area/substantia nigra or serotonin cells in the dorsal raphe nucleus. Orexin (hypocretin) cells in the hypothalamus showed a modest reduction (14%). Our findings indicate that the basal forebrain is especially vulnerable to alcohol exposure in the late gestational period. Reduction of cholinergic and GABAergic projection neurons from the basal forebrain that regulate forebrain arousal may contribute to the behavioral and cognitive deficits associated with neonatal ethanol exposure.
Collapse
|
18
|
Wang X, Zhang Y, Cheng W, Gao Y, Li S. Decreased excitatory drive onto hilar neuronal nitric oxide synthase expressing interneurons in chronic models of epilepsy. Brain Res 2021; 1764:147467. [PMID: 33831408 DOI: 10.1016/j.brainres.2021.147467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 11/29/2022]
Abstract
Excitation-inhibition imbalance of GABAergic interneurons is predisposed to develop chronic temporal lobe epilepsy (TLE). We have previously shown that virtually every neuronal nitric oxide synthase (nNOS)-positive cell is a GABAergic inhibitory interneuron in the denate gyrus. The present study was designed to quantify the number of nNOS-containing hilar interneurons using stereology in pilocapine- and kainic acid (KA)-exposed transgenic adult mice that expressed GFP under the nNOS promoter. In addition, we studied the properties of miniature excitatory postsynaptic current (mEPSC) and paired-pulse response ratio (PPR) of evoked EPSC in nNOS interneurons using whole cell recording techniques. Results showed that there were fewer nNOS-immunoreactive interneurons of chronically epileptic animals. Importantly, patch-clamp recordings revealed reduction in mEPSC frequency, indicating diminished global excitatory input. In contrast, PPR of evoked EPSC following the granule cell layer stimulation was increased in epileptic animals suggesting reduced neurotransmitter release from granule cell input. In summary, we propose that impaired excitatory drive onto hippocampal nNOS interneurons may be implicated in the development of refractory epilepsy.
Collapse
Affiliation(s)
- Xiaona Wang
- Henan Neurodevelopment Engineering Research Center for Children, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, 33 Longhu Outer Circle Dong Road, Zhengzhou, 450018, Henan, China.
| | - Yaodong Zhang
- Henan Neurodevelopment Engineering Research Center for Children, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, 33 Longhu Outer Circle Dong Road, Zhengzhou, 450018, Henan, China
| | - Weyland Cheng
- Department of Orthopaedics, Children's Hospital Affiliated to Zhengzhou University, 33 Longhu Outer Circle Dong Road, Zhengzhou 450018, Henan, China
| | - Yinbo Gao
- Henan Neurodevelopment Engineering Research Center for Children, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, 33 Longhu Outer Circle Dong Road, Zhengzhou, 450018, Henan, China
| | - Shao Li
- Department of Physiology, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China
| |
Collapse
|
19
|
Mihály I, Molnár T, Berki ÁJ, Bod RB, Orbán-Kis K, Gáll Z, Szilágyi T. Short-Term Amygdala Low-Frequency Stimulation Does not Influence Hippocampal Interneuron Changes Observed in the Pilocarpine Model of Epilepsy. Cells 2021; 10:cells10030520. [PMID: 33804543 PMCID: PMC7998440 DOI: 10.3390/cells10030520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/19/2021] [Accepted: 02/25/2021] [Indexed: 11/23/2022] Open
Abstract
Temporal lobe epilepsy (TLE) is characterized by changes in interneuron numbers in the hippocampus. Deep brain stimulation (DBS) is an emerging tool to treat TLE seizures, although its mechanisms are not fully deciphered. We aimed to depict the effect of amygdala DBS on the density of the most common interneuron types in the CA1 hippocampal subfield in the lithium-pilocarpine model of epilepsy. Status epilepticus was induced in male Wistar rats. Eight weeks later, a stimulation electrode was implanted to the left basolateral amygdala of both pilocarpine-treated (Pilo, n = 14) and age-matched control rats (n = 12). Ten Pilo and 4 control animals received for 10 days 4 daily packages of 50 s 4 Hz regular stimulation trains. At the end of the stimulation period, interneurons were identified by immunolabeling for parvalbumin (PV), neuropeptide Y (NPY), and neuronal nitric oxide synthase (nNOS). Cell density was determined in the CA1 subfield of the hippocampus using confocal microscopy. We found that PV+ cell density was preserved in pilocarpine-treated rats, while the NPY+/nNOS+ cell density decreased significantly. The amygdala DBS did not significantly change the cell density in healthy or in epileptic animals. We conclude that DBS with low frequency applied for 10 days does not influence interneuron cell density changes in the hippocampus of epileptic rats.
Collapse
Affiliation(s)
- István Mihály
- Department of Physiology, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (T.M.); (Á.-J.B.); (R.-B.B.); (K.O.-K.); (T.S.)
- Correspondence: ; Tel.: +40-749-768-257
| | - Tímea Molnár
- Department of Physiology, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (T.M.); (Á.-J.B.); (R.-B.B.); (K.O.-K.); (T.S.)
| | - Ádám-József Berki
- Department of Physiology, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (T.M.); (Á.-J.B.); (R.-B.B.); (K.O.-K.); (T.S.)
| | - Réka-Barbara Bod
- Department of Physiology, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (T.M.); (Á.-J.B.); (R.-B.B.); (K.O.-K.); (T.S.)
| | - Károly Orbán-Kis
- Department of Physiology, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (T.M.); (Á.-J.B.); (R.-B.B.); (K.O.-K.); (T.S.)
| | - Zsolt Gáll
- Department of Pharmacology and Clinical Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Tibor Szilágyi
- Department of Physiology, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (T.M.); (Á.-J.B.); (R.-B.B.); (K.O.-K.); (T.S.)
| |
Collapse
|
20
|
Houser CR, Peng Z, Wei X, Huang CS, Mody I. Mossy Cells in the Dorsal and Ventral Dentate Gyrus Differ in Their Patterns of Axonal Projections. J Neurosci 2021; 41:991-1004. [PMID: 33268544 PMCID: PMC7880284 DOI: 10.1523/jneurosci.2455-20.2020] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/08/2020] [Accepted: 11/20/2020] [Indexed: 01/22/2023] Open
Abstract
Mossy cells (MCs) of the dentate gyrus (DG) are a major group of excitatory hilar neurons that are important for regulating activity of dentate granule cells. MCs are particularly intriguing because of their extensive longitudinal connections within the DG. It has generally been assumed that MCs in the dorsal and ventral DG have similar patterns of termination in the inner one-third of the dentate molecular layer. Here, we demonstrate that axonal projections of MCs in these two regions are considerably different. MCs in dorsal and ventral regions were labeled selectively with Cre-dependent eYFP or mCherry, using two transgenic mouse lines (including both sexes) that express Cre-recombinase in MCs. At four to six weeks following unilateral labeling of MCs in the ventral DG, a dense band of fibers was present in the inner one-fourth of the molecular layer and extended bilaterally throughout the rostral-caudal extent of the DG, replicating the expected distribution of MC axons. In contrast, following labeling of MCs in the dorsal DG, the projections were more diffusely distributed. At the level of transfection, fibers were present in the inner molecular layer, but they progressively expanded into the middle molecular layer and, most ventrally, formed a distinct band in this region. Optical stimulation of these caudal fibers expressing ChR2 demonstrated robust EPSCs in ipsilateral granule cells and enhanced the effects of perforant path stimulation in the ventral DG. These findings suggest that MCs in the dorsal and ventral DG differ in the distribution of their axonal projections and possibly their function.SIGNIFICANCE STATEMENT Mossy cells (MCs), a major cell type in the hilus of the dentate gyrus (DG), are unique in providing extensive longitudinal and commissural projections throughout the DG. Although it has been assumed that all MCs have similar patterns of termination in the inner molecular layer of the DG, we discovered that the axonal projections of dorsal and ventral MCs differ. While ventral MC projections exhibit the classical pattern, with dense innervation in the inner molecular layer, dorsal MCs have a more diffuse distribution and expand into the middle molecular layer where they overlap and interact with innervation from the perforant path. These distinct locations and patterns of axonal projections suggest that dorsal and ventral MCs may have different functional roles.
Collapse
Affiliation(s)
- Carolyn R Houser
- Department of Neurobiology
- Brain Research Institute, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California 90095
| | | | | | | | - Istvan Mody
- Department of Neurology
- Brain Research Institute, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California 90095
| |
Collapse
|
21
|
Bidirectional Regulation of Cognitive and Anxiety-like Behaviors by Dentate Gyrus Mossy Cells in Male and Female Mice. J Neurosci 2021; 41:2475-2495. [PMID: 33472828 DOI: 10.1523/jneurosci.1724-20.2021] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 12/04/2020] [Accepted: 01/04/2021] [Indexed: 02/08/2023] Open
Abstract
The dentate gyrus (DG) of the hippocampus is important for cognition and behavior. However, the circuits underlying these functions are unclear. DG mossy cells (MCs) are potentially important because of their excitatory synapses on the primary cell type, granule cells (GCs). However, MCs also activate GABAergic neurons, which inhibit GCs. We used viral delivery of designer receptors exclusively activated by designer drugs (DREADDs) in mice to implement a gain- and loss-of-function study of MCs in diverse behaviors. Using this approach, manipulations of MCs could bidirectionally regulate behavior. The results suggest that inhibiting MCs can reduce anxiety-like behavior and improve cognitive performance. However, not all cognitive or anxiety-related behaviors were influenced, suggesting specific roles of MCs in some, but not all, types of cognition and anxiety. Notably, several behaviors showed sex-specific effects, with females often showing more pronounced effects than the males. We also used the immediate early gene c-Fos to address whether DREADDs bidirectionally regulated MC or GC activity. We confirmed excitatory DREADDs increased MC c-Fos. However, there was no change in GC c-Fos, consistent with MC activation leading to GABAergic inhibition of GCs. In contrast, inhibitory DREADDs led to a large increase in GC c-Fos, consistent with a reduction in MC excitation of GABAergic neurons, and reduced inhibition of GCs. Together, these results suggest that MCs regulate anxiety and cognition in specific ways. We also raise the possibility that cognitive performance may be improved by reducing anxiety.SIGNIFICANCE STATEMENT The dentate gyrus (DG) has many important cognitive roles as well as being associated with affective behavior. This study addressed how a glutamatergic DG cell type called mossy cells (MCs) contributes to diverse behaviors, which is timely because it is known that MCs regulate the activity of the primary DG cell type, granule cells (GCs), but how MC activity influences behavior is unclear. We show, surprisingly, that activating MCs can lead to adverse behavioral outcomes, and inhibiting MCs have an opposite effect. Importantly, the results appeared to be task-dependent and showed that testing both sexes was important. Additional experiments indicated what MC and GC circuitry was involved. Together, the results suggest how MCs influence behaviors that involve the DG.
Collapse
|
22
|
Botterill JJ, Lu YL, LaFrancois JJ, Bernstein HL, Alcantara-Gonzalez D, Jain S, Leary P, Scharfman HE. An Excitatory and Epileptogenic Effect of Dentate Gyrus Mossy Cells in a Mouse Model of Epilepsy. Cell Rep 2020; 29:2875-2889.e6. [PMID: 31775052 PMCID: PMC6905501 DOI: 10.1016/j.celrep.2019.10.100] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 09/25/2019] [Accepted: 10/24/2019] [Indexed: 12/20/2022] Open
Abstract
The sparse activity of hippocampal dentate gyrus (DG) granule cells (GCs) is thought to be critical for cognition and behavior, whereas excessive DG activity may contribute to disorders such as temporal lobe epilepsy (TLE). Glutamatergic mossy cells (MCs) of the DG are potentially critical to normal and pathological functions of the DG because they can regulate GC activity through innervation of GCs or indirectly through GABAergic neurons. Here, we test the hypothesis that MC excitation of GCs is normally weak, but under pathological conditions, MC excitation of GCs is dramatically strengthened. We show that selectively inhibiting MCs during severe seizures reduced manifestations of those seizures, hippocampal injury, and chronic epilepsy. In contrast, selectively activating MCs was pro-convulsant. Mechanistic in vitro studies using optogenetics further demonstrated the unanticipated ability of MC axons to excite GCs under pathological conditions. These results demonstrate an excitatory and epileptogenic effect of MCs in the DG.
Collapse
Affiliation(s)
- Justin J Botterill
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Yi-Ling Lu
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - John J LaFrancois
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Hannah L Bernstein
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA; Department of Neuroscience & Physiology, New York University Langone Health, New York, NY 10016, USA
| | - David Alcantara-Gonzalez
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Swati Jain
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Paige Leary
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Helen E Scharfman
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA; Department of Neuroscience & Physiology, New York University Langone Health, New York, NY 10016, USA; Department of Psychiatry, New York University Langone Health, New York, NY 10016, USA.
| |
Collapse
|
23
|
Post-exposure environment modulates long-term developmental ethanol effects on behavior, neuroanatomy, and cortical oscillations. Brain Res 2020; 1748:147128. [PMID: 32950485 DOI: 10.1016/j.brainres.2020.147128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/25/2020] [Accepted: 09/12/2020] [Indexed: 11/23/2022]
Abstract
Developmental exposure to ethanol has a wide range of anatomical, cellular, physiological and behavioral impacts that can last throughout life. In humans, this cluster of effects is termed fetal alcohol spectrum disorder and is highly prevalent in western cultures. The ultimate expression of the effects of developmental ethanol exposure however can be influenced by post-exposure experience. Here we examined the effects of developmental binge exposure to ethanol (postnatal day 7) in C57BL/6By mice on a specific cohort of inter-related long-term outcomes including contextual memory, hippocampal parvalbumin-expressing neuron density, frontal cortex oscillations related to sleep-wake cycling including delta oscillation amplitude and sleep spindle density, and home-cage behavioral activity. When assessed in adults that were raised in standard housing, all of these factors were altered by early ethanol exposure compared to saline controls except home-cage activity. However, exposure to an enriched environment and exercise from weaning to postnatal day 90 reversed most of these ethanol-induced impairments including memory, CA1 but not dentate gyrus PV+ cell density, delta oscillations and sleep spindles, and enhanced home-cage behavioral activity in Saline- but not EtOH-treated mice. The results are discussed in terms of the inter-dependence of diverse developmental ethanol outcomes and potential mechanisms of post-exposure experiences to regulate those outcomes.
Collapse
|
24
|
Ábrahám H, Molnár JE, Sóki N, Gyimesi C, Horváth Z, Janszky J, Dóczi T, Seress L. Etiology-related Degree of Sprouting of Parvalbumin-immunoreactive Axons in the Human Dentate Gyrus in Temporal Lobe Epilepsy. Neuroscience 2020; 448:55-70. [PMID: 32931846 DOI: 10.1016/j.neuroscience.2020.09.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/22/2020] [Accepted: 09/05/2020] [Indexed: 11/16/2022]
Abstract
In the present study, we examined parvalbumin-immunoreactive cells and axons in the dentate gyrus of surgically resected tissues of therapy-resistant temporal lobe epilepsy (TLE) patients with different etiologies. Based on MRI results, five groups of patients were formed: (1) hippocampal sclerosis (HS), (2) malformation of cortical development, (3) malformation of cortical development + HS, (4) tumor-induced TLE, (5) patients with negative MRI result. Four control samples were also included in the study. Parvalbumin-immunoreactive cells were observed mostly in subgranular location in the dentate hilus in controls, in tumor-induced TLE, in malformation of cortical development and in MR-negative cases. In patients with HS, significant decrease in the number of hilar parvalbumin-immunoreactive cells and large numbers of ectopic parvalbumin-containing neurons were detected in the dentate gyrus' molecular layer. The ratio of ectopic/normally-located cells was significantly higher in HS than in other TLE groups. In patients with HS, robust sprouting of parvalbumin-immunoreactive axons were frequently visible in the molecular layer. The extent of sprouting was significantly higher in TLE patients with HS than in other groups. Strong sprouting of parvalbumin-immunoreactive axons were frequently observed in patients who had childhood febrile seizure. Significant correlation was found between the level of sprouting of axons and the ratio of ectopic/normally-located parvalbumin-containing cells. Electron microscopy demonstrated that sprouted parvalbumin-immunoreactive axons terminate on proximal and distal dendritic shafts as well as on dendritic spines of granule cells. Our results indicate alteration of target profile of parvalbumin-immunoreactive neurons in HS that contributes to the known synaptic remodeling in TLE.
Collapse
Affiliation(s)
- Hajnalka Ábrahám
- Department of Medical Biology and Central Electron Microscopic Laboratory, University of Pécs Medical School, Szigeti u 12., Pécs 7624, Hungary.
| | - Judit E Molnár
- Department of Medical Biology and Central Electron Microscopic Laboratory, University of Pécs Medical School, Szigeti u 12., Pécs 7624, Hungary
| | - Noémi Sóki
- Department of Medical Biology and Central Electron Microscopic Laboratory, University of Pécs Medical School, Szigeti u 12., Pécs 7624, Hungary
| | - Csilla Gyimesi
- Department of Neurology, University of Pécs Medical School, Rét u. 2., Pécs 7623, Hungary
| | - Zsolt Horváth
- Department of Neurosurgery, University of Pécs Medical School, Rét u. 2., Pécs 7623, Hungary
| | - József Janszky
- Department of Neurology, University of Pécs Medical School, Rét u. 2., Pécs 7623, Hungary
| | - Tamás Dóczi
- Department of Neurosurgery, University of Pécs Medical School, Rét u. 2., Pécs 7623, Hungary
| | - László Seress
- Department of Medical Biology and Central Electron Microscopic Laboratory, University of Pécs Medical School, Szigeti u 12., Pécs 7624, Hungary
| |
Collapse
|
25
|
Semilunar Granule Cells Are the Primary Source of the Perisomatic Excitatory Innervation onto Parvalbumin-Expressing Interneurons in the Dentate Gyrus. eNeuro 2020; 7:ENEURO.0323-19.2020. [PMID: 32571963 PMCID: PMC7340841 DOI: 10.1523/eneuro.0323-19.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 05/29/2020] [Accepted: 06/02/2020] [Indexed: 11/21/2022] Open
Abstract
We analyzed the origin and relevance of the perisomatic excitatory inputs on the parvalbumin interneurons of the granule cell layer in mouse. Confocal analysis of the glutamatergic innervation showed that it represents ∼50% of the perisomatic synapses that parvalbumin cells receive. This excitatory input may originate from granule cell collaterals, the mossy cells, or even supramammillary nucleus. First, we assessed the input from the mossy cells on parvalbumin interneurons. Axon terminals of mossy cells were visualized by their calretinin content. Using multicolor confocal microscopy, we observed that less than 10% of perisomatic excitatory innervation of parvalbumin cells could originate from mossy cells. Correlative light and electron microscopy revealed that innervation from mossy cells, although present, was indeed infrequent, except for those parvalbumin cells whose somata were located in the inner molecular layer. Second, we investigated the potential input from supramammillary nucleus on parvalbumin cell somata using anterograde tracing or immunocytochemistry against vesicular glutamate transporter 2 (VGLUT2) and found only occasional contacts. Third, we intracellularly filled dentate granule cells in acute slice preparations using whole-cell recording and examined whether their axon collaterals target parvalbumin interneurons. We found that typical granule cells do not innervate the perisomatic region of these GABAergic cells. In sharp contrast, semilunar granule cells (SGCs), a scarce granule cell subtype often contacted the parvalbumin cell soma and proximal dendrites. Our data, therefore, show that perisomatic excitatory drive of parvalbumin interneurons in the granular layer of the dentate gyrus is abundant and originates primarily from SGCs.
Collapse
|
26
|
Cui Y, Liu J, Luo Y, He S, Xia Y, Zhang Y, Yao D, Guo D. Aberrant Connectivity During Pilocarpine-Induced Status Epilepticus. Int J Neural Syst 2019; 30:1950029. [PMID: 31847633 DOI: 10.1142/s0129065719500291] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Status epilepticus (SE) is a common, life-threatening neurological disorder that may lead to permanent brain damage. In rodent models, SE is an acute phase of seizures that could be reproduced by injecting with pilocarpine and then induce chronic temporal lobe epilepsy (TLE) seizures. However, how SE disrupts brain activity, especially communications among brain regions, is still unclear. In this study, we aimed to identify the characteristic abnormalities of network connections among the frontal cortex, hippocampus and thalamus during the SE episodes in a pilocarpine model with functional and effective connectivity measurements. We showed that the coherence connectivity among these regions increased significantly during the SE episodes in almost all frequency bands (except the alpha band) and that the frequency band with enhanced connections was specific to different stages of SE episodes. Moreover, with the effective analysis, we revealed a closed neural circuit of bidirectional effective interactions between the frontal regions and the hippocampus and thalamus in both ictal and post-ictal stages, implying aberrant enhancement of communication across these brain regions during the SE episodes. Furthermore, an effective connection from the hippocampus to the thalamus was detected in the delta band during the pre-ictal stage, which shifted in an inverse direction during the ictal stage in the theta band and in the theta, alpha, beta and low-gamma bands during the post-ictal stage. This specificity of the effective connection between the hippocampus and thalamus illustrated that the hippocampal structure is critical for the initiation of SE discharges, while the thalamus is important for the propagation of SE discharges. Overall, our results demonstrated enhanced interaction among the frontal cortex, hippocampus and thalamus during the SE episodes and suggested the modes of information flow across these structures for the initiation and propagation of SE discharges. These findings may reveal an underlying mechanism of aberrant network communication during pilocarpine-induced SE discharges and deepen our knowledge of TLE seizures.
Collapse
Affiliation(s)
- Yan Cui
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
- Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
| | - Jie Liu
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
- Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
| | - Yan Luo
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
- Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
| | - Shan He
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
- Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
| | - Yang Xia
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
- Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
| | - Yangsong Zhang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
- Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
| | - Dezhong Yao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
- Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
| | - Daqing Guo
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
- Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
| |
Collapse
|
27
|
Function of local circuits in the hippocampal dentate gyrus-CA3 system. Neurosci Res 2018; 140:43-52. [PMID: 30408501 DOI: 10.1016/j.neures.2018.11.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/27/2018] [Accepted: 10/15/2018] [Indexed: 11/20/2022]
Abstract
Anatomical observations, theoretical work and lesioning experiments have supported the idea that the CA3 in the hippocampus is important for encoding, storage and retrieval of memory while the dentate gyrus (DG) is important for the pattern separation of the incoming inputs from the entorhinal cortex. Study of the presumed function of the dentate gyrus in pattern separation has been hampered by the lack of reliable methods to identify different excitatory cell types in the DG. Recent papers have identified different cell types in the DG, in awake behaving animals, with more reliable methods. These studies have revealed each cell type's spatial representation as well as their involvement in pattern separation. Moreover, chronic electrophysiological recording from sleeping and waking animals also provided more insights into the operation of the DG-CA3 system for memory encoding and retrieval. This article will review the local circuit architectures and physiological properties of the DG-CA3 system and discuss how the local circuit in the DG-CA3 may function, incorporating recent physiological findings in the DG-CA3 system.
Collapse
|
28
|
Bumanglag AV, Sloviter RS. No latency to dentate granule cell epileptogenesis in experimental temporal lobe epilepsy with hippocampal sclerosis. Epilepsia 2018; 59:2019-2034. [PMID: 30338519 DOI: 10.1111/epi.14580] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/19/2018] [Accepted: 09/19/2018] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To determine when spontaneous granule cell epileptiform discharges first occur after hippocampal injury, and to identify the postinjury "latent" period as either a "silent" gestational state of epileptogenesis or a subtle epileptic state in gradual transition to a more obvious epileptic state. METHODS Nonconvulsive status epilepticus evoked by perforant path stimulation in urethane-sedated rats produced selective and extensive hippocampal injury and a "latent" period that preceded the onset of the first clinically obvious epileptic seizures. Continuous granule cell layer depth recording and video monitoring assessed the time course of granule cell hyperexcitability and the onset/offset times of spontaneous epileptiform discharges and behavioral seizures. RESULTS One day postinjury, granule cells in awake rats were hyperexcitable to afferent input, and continuously generated spontaneous population spikes. During the ~2-4 week "latent" period, granule cell epileptiform discharges lasting ~30 seconds caused subtle focal seizures characterized by immobilization and facial automatisms that were undetected by behavioral assessment alone but identified post hoc. Granule cell layer epileptiform discharge duration eventually tripled, which caused the first clinically obvious seizure, ending the "latent" period. Behavioral seizure duration was linked tightly to spontaneous granule cell layer events. Granule cell epileptiform discharges preceded all behavioral seizure onsets, and clonic behaviors ended abruptly within seconds of the termination of each granule cell epileptiform discharge. Noninjurious hippocampal excitation produced no evidence of granule cell hyperexcitability or epileptogenesis. SIGNIFICANCE The latent period in this model is a subtle epileptic state in transition to a more clinically obvious epileptic state, not a seizure-free "gestational" state when an unidentified epileptogenic mechanism gradually develops. Based on the onset/offset times of electrographic and behavioral events, granule cell behavior may be the prime determinant of seizure onset, phenotype, duration, and offset in this model of hippocampal-onset epilepsy. Extensive hippocampal neuron loss could be the primary epileptogenic mechanism.
Collapse
Affiliation(s)
| | - Robert S Sloviter
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia.,Department of Neurobiology, Morehouse School of Medicine, Atlanta, Georgia.,Department of Pharmacology & Toxicology, Morehouse School of Medicine, Atlanta, Georgia
| |
Collapse
|
29
|
Cǎlin A, Stancu M, Zagrean AM, Jefferys JGR, Ilie AS, Akerman CJ. Chemogenetic Recruitment of Specific Interneurons Suppresses Seizure Activity. Front Cell Neurosci 2018; 12:293. [PMID: 30233328 PMCID: PMC6134067 DOI: 10.3389/fncel.2018.00293] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 08/16/2018] [Indexed: 12/22/2022] Open
Abstract
Current anti-epileptic medications that boost synaptic inhibition are effective in reducing several types of epileptic seizure activity. Nevertheless, these drugs can generate significant side-effects and even paradoxical responses due to the broad nature of their action. Recently developed chemogenetic techniques provide the opportunity to pharmacologically recruit endogenous inhibitory mechanisms in a selective and circuit-specific manner. Here, we use chemogenetics to assess the potential of suppressing epileptiform activity by enhancing the synaptic output from three major interneuron populations in the rodent hippocampus: parvalbumin (PV), somatostatin (SST), and vasoactive intestinal peptide (VIP) expressing interneurons. To target different neuronal populations, promoter-specific cre-recombinase mice were combined with viral-mediated delivery of chemogenetic constructs. Targeted electrophysiological recordings were then conducted in an in vitro model of chronic, drug-resistant epilepsy. In addition, behavioral video-scoring was performed in an in vivo model of acutely triggered seizure activity. Pre-synaptic and post-synaptic whole cell recordings in brain slices revealed that each of the three interneuron types increase their firing rate and synaptic output following chemogenetic activation. However, the interneuron populations exhibited different effects on epileptiform discharges. Recruiting VIP interneurons did not change the total duration of epileptiform discharges. In contrast, recruiting SST or PV interneurons produced robust suppression of epileptiform synchronization. PV interneurons exhibited the strongest effect per cell, eliciting at least a fivefold greater reduction in epileptiform activity than the other cell types. Consistent with this, we found that in vivo chemogenetic recruitment of PV interneurons suppressed convulsive behaviors by more than 80%. Our findings support the idea that selective chemogenetic enhancement of inhibitory synaptic pathways offers potential as an anti-seizure strategy.
Collapse
Affiliation(s)
- Alexandru Cǎlin
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Mihai Stancu
- Division of Physiology and Neuroscience, Department of Functional Sciences, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Ana-Maria Zagrean
- Division of Physiology and Neuroscience, Department of Functional Sciences, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Andrei S. Ilie
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Colin J. Akerman
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
30
|
Kim JE, Kang TC. Differential Roles of Mitochondrial Translocation of Active Caspase-3 and HMGB1 in Neuronal Death Induced by Status Epilepticus. Front Cell Neurosci 2018; 12:301. [PMID: 30233331 PMCID: PMC6133957 DOI: 10.3389/fncel.2018.00301] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/17/2018] [Indexed: 11/13/2022] Open
Abstract
Under pathophysiological conditions, aberrant mitochondrial dynamics lead to the different types of neuronal death: excessive mitochondrial fission provokes apoptosis and abnormal mitochondrial elongation induces necrosis. However, the underlying mechanisms how the different mitochondrial dynamics result in the distinct neuronal death patterns have been elusive. In the present study, status epilepticus (SE) evoked excessive mitochondrial fission in parvalbumin (PV) cells (one of GABAergic interneurons) and abnormal mitochondrial elongation in CA1 neurons in the rat hippocampus. These impaired mitochondrial dynamics were accompanied by mitochondrial translocations of active caspase-3 and high mobility group box 1 (HMGB1) in PV cells and CA1 neurons, respectively. WY14643 (an activator of mitochondrial fission) aggravated SE-induced PV cell loss by enhancing active caspase-3 induction and its mitochondrial translocation, which were attenuated by Mdivi-1 (an inhibitor of mitochondrial fission). Mitochondrial HMGB1 import was not observed in PV cell. In contrast to PV cells, Mdivi-1 deteriorated SE-induced CA1 neuronal death concomitant with mitochondrial HMGB1 translocation, which was abrogated by WY14643. These findings suggest that SE-induced aberrant mitochondrial dynamics may be involved in translocation of active caspase-3 and HMGB1 into mitochondria, which regulate neuronal apoptosis and necrosis, respectively.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, South Korea
| |
Collapse
|
31
|
Koyama R, Ikegaya Y. The Molecular and Cellular Mechanisms of Axon Guidance in Mossy Fiber Sprouting. Front Neurol 2018; 9:382. [PMID: 29896153 PMCID: PMC5986954 DOI: 10.3389/fneur.2018.00382] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/11/2018] [Indexed: 01/25/2023] Open
Abstract
The question of whether mossy fiber sprouting is epileptogenic has not been resolved; both sprouting-induced recurrent excitatory and inhibitory circuit hypotheses have been experimentally (but not fully) supported. Therefore, whether mossy fiber sprouting is a potential therapeutic target for epilepsy remains under debate. Moreover, the axon guidance mechanisms of mossy fiber sprouting have attracted the interest of neuroscientists. Sprouting of mossy fibers exhibits several uncommon axonal growth features in the basically non-plastic adult brain. For example, robust branching of axonal collaterals arises from pre-existing primary mossy fiber axons. Understanding the branching mechanisms in adulthood may contribute to axonal regeneration therapies in neuroregenerative medicine in which robust axonal re-growth is essential. Additionally, because granule cells are produced throughout life in the neurogenic dentate gyrus, it is interesting to examine whether the mossy fibers of newly generated granule cells follow the pre-existing trajectories of sprouted mossy fibers in the epileptic brain. Understanding these axon guidance mechanisms may contribute to neuron transplantation therapies, for which the incorporation of transplanted neurons into pre-existing neural circuits is essential. Thus, clarifying the axon guidance mechanisms of mossy fiber sprouting could lead to an understanding of central nervous system (CNS) network reorganization and plasticity. Here, we review the molecular and cellular mechanisms of axon guidance in mossy fiber sprouting by discussing mainly in vitro studies.
Collapse
Affiliation(s)
- Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
32
|
Bui AD, Nguyen TM, Limouse C, Kim HK, Szabo GG, Felong S, Maroso M, Soltesz I. Dentate gyrus mossy cells control spontaneous convulsive seizures and spatial memory. Science 2018; 359:787-790. [PMID: 29449490 DOI: 10.1126/science.aan4074] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 12/21/2017] [Indexed: 01/06/2023]
Abstract
Temporal lobe epilepsy (TLE) is characterized by debilitating, recurring seizures and an increased risk for cognitive deficits. Mossy cells (MCs) are key neurons in the hippocampal excitatory circuit, and the partial loss of MCs is a major hallmark of TLE. We investigated how MCs contribute to spontaneous ictal activity and to spatial contextual memory in a mouse model of TLE with hippocampal sclerosis, using a combination of optogenetic, electrophysiological, and behavioral approaches. In chronically epileptic mice, real-time optogenetic modulation of MCs during spontaneous hippocampal seizures controlled the progression of activity from an electrographic to convulsive seizure. Decreased MC activity is sufficient to impede encoding of spatial context, recapitulating observed cognitive deficits in chronically epileptic mice.
Collapse
Affiliation(s)
- Anh D Bui
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA. .,Department of Anatomy and Neurobiology, University of California, Irvine, CA 92697, USA
| | - Theresa M Nguyen
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Charles Limouse
- Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - Hannah K Kim
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Gergely G Szabo
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Sylwia Felong
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Mattia Maroso
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
33
|
França KLDA, de Almeida ACG, Saddow SE, Santos LEC, Scorza CA, Scorza FA, Rodrigues AM. GABA a excitation and synaptogenesis after Status Epilepticus - A computational study. Sci Rep 2018; 8:4193. [PMID: 29520076 PMCID: PMC5843660 DOI: 10.1038/s41598-018-22581-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 01/23/2018] [Indexed: 11/09/2022] Open
Abstract
The role of GABAergic neurotransmission on epileptogenesis has been the subject of speculation according to different approaches. However, it is a very complex task to specifically consider the action of the GABAa neurotransmitter, which, in its dependence on the intracellular level of Cl-, can change its effect from inhibitory to excitatory. We have developed a computational model that represents the dentate gyrus and is composed of three different populations of neurons (granule cells, interneurons and mossy cells) that are mutually interconnected. The interconnections of the neurons were based on compensation theory with Hebbian and anti-Hebbian rules. The model also incorporates non-synaptic mechanisms to control the ionic homeostasis and was able to reproduce ictal discharges. The goal of the work was to investigate the hypothesis that the observed aberrant sprouting is promoted by GABAa excitatory action. Conjointly with the abnormal sprouting of the mossy fibres, the simulations show a reduction of the mossy cells connections in the network and an increased inhibition of the interneurons as a response of the neuronal network to control the activity. This finding contributes to increasing the changes in the connectivity of the neuronal circuitry and to increasing the epileptiform activity occurrences.
Collapse
Affiliation(s)
- Keite Lira de Almeida França
- Laboratório de Neurociência Experimental e Computacional (LANEC), Departamento de Engenharia de Biossistemas, Universidade Federal de São João del-Rei (UFSJ), São João del-Rei, Brazil
| | - Antônio-Carlos Guimarães de Almeida
- Laboratório de Neurociência Experimental e Computacional (LANEC), Departamento de Engenharia de Biossistemas, Universidade Federal de São João del-Rei (UFSJ), São João del-Rei, Brazil
| | - Stephen E Saddow
- Electrical Engineering Department, University of South of Florida, Tampa, FL, USA
| | - Luiz Eduardo Canton Santos
- Laboratório de Neurociência Experimental e Computacional (LANEC), Departamento de Engenharia de Biossistemas, Universidade Federal de São João del-Rei (UFSJ), São João del-Rei, Brazil
| | | | | | - Antônio Márcio Rodrigues
- Laboratório de Neurociência Experimental e Computacional (LANEC), Departamento de Engenharia de Biossistemas, Universidade Federal de São João del-Rei (UFSJ), São João del-Rei, Brazil.
| |
Collapse
|
34
|
Hansen MG, Ledri LN, Kirik D, Kokaia M, Ledri M. Preserved Function of Afferent Parvalbumin-Positive Perisomatic Inhibitory Synapses of Dentate Granule Cells in Rapidly Kindled Mice. Front Cell Neurosci 2018; 11:433. [PMID: 29375319 PMCID: PMC5767181 DOI: 10.3389/fncel.2017.00433] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 12/22/2017] [Indexed: 11/13/2022] Open
Abstract
Parvalbumin- (PV-) containing basket cells constitute perisomatic GABAergic inhibitory interneurons innervating principal cells at perisomatic area, a strategic location that allows them to efficiently control the output and synchronize oscillatory activity at gamma frequency (30–90 Hz) oscillations. This oscillatory activity can convert into higher frequency epileptiform activity, and therefore could play an important role in the generation of seizures. However, the role of endogenous modulators of seizure activity, such as Neuropeptide Y (NPY), has not been fully explored in at PV input and output synapses. Here, using selective optogenetic activation of PV cells in the hippocampus, we show that seizures, induced by rapid kindling (RK) stimulations, enhance gamma-aminobutyric acid (GABA) release from PV cells onto dentate gyrus (DG) granule cells (GC). However, PV-GC synapses did not differ between controls and kindled animals in terms of GABA release probability, short-term plasticity and sensitivity to NPY. Kinetics of gamma-aminobutyric acid A (GABA-A) mediated currents in postsynaptic GC were also unaffected. When challenged by repetitive high-frequency optogenetic stimulations, PV synapses in kindled animals responded with enhanced GABA release onto GC. These results unveil a mechanism that might possibly contribute to the generation of abnormal synchrony and maintenance of epileptic seizures.
Collapse
Affiliation(s)
- Marita G Hansen
- Epilepsy Center, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Litsa N Ledri
- Epilepsy Center, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems (BRAINS) Unit, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Merab Kokaia
- Epilepsy Center, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Marco Ledri
- Epilepsy Center, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
35
|
Klein P, Dingledine R, Aronica E, Bernard C, Blümcke I, Boison D, Brodie MJ, Brooks-Kayal AR, Engel J, Forcelli PA, Hirsch LJ, Kaminski RM, Klitgaard H, Kobow K, Lowenstein DH, Pearl PL, Pitkänen A, Puhakka N, Rogawski MA, Schmidt D, Sillanpää M, Sloviter RS, Steinhäuser C, Vezzani A, Walker MC, Löscher W. Commonalities in epileptogenic processes from different acute brain insults: Do they translate? Epilepsia 2018; 59:37-66. [PMID: 29247482 PMCID: PMC5993212 DOI: 10.1111/epi.13965] [Citation(s) in RCA: 201] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2017] [Indexed: 12/12/2022]
Abstract
The most common forms of acquired epilepsies arise following acute brain insults such as traumatic brain injury, stroke, or central nervous system infections. Treatment is effective for only 60%-70% of patients and remains symptomatic despite decades of effort to develop epilepsy prevention therapies. Recent preclinical efforts are focused on likely primary drivers of epileptogenesis, namely inflammation, neuron loss, plasticity, and circuit reorganization. This review suggests a path to identify neuronal and molecular targets for clinical testing of specific hypotheses about epileptogenesis and its prevention or modification. Acquired human epilepsies with different etiologies share some features with animal models. We identify these commonalities and discuss their relevance to the development of successful epilepsy prevention or disease modification strategies. Risk factors for developing epilepsy that appear common to multiple acute injury etiologies include intracranial bleeding, disruption of the blood-brain barrier, more severe injury, and early seizures within 1 week of injury. In diverse human epilepsies and animal models, seizures appear to propagate within a limbic or thalamocortical/corticocortical network. Common histopathologic features of epilepsy of diverse and mostly focal origin are microglial activation and astrogliosis, heterotopic neurons in the white matter, loss of neurons, and the presence of inflammatory cellular infiltrates. Astrocytes exhibit smaller K+ conductances and lose gap junction coupling in many animal models as well as in sclerotic hippocampi from temporal lobe epilepsy patients. There is increasing evidence that epilepsy can be prevented or aborted in preclinical animal models of acquired epilepsy by interfering with processes that appear common to multiple acute injury etiologies, for example, in post-status epilepticus models of focal epilepsy by transient treatment with a trkB/PLCγ1 inhibitor, isoflurane, or HMGB1 antibodies and by topical administration of adenosine, in the cortical fluid percussion injury model by focal cooling, and in the albumin posttraumatic epilepsy model by losartan. Preclinical studies further highlight the roles of mTOR1 pathways, JAK-STAT3, IL-1R/TLR4 signaling, and other inflammatory pathways in the genesis or modulation of epilepsy after brain injury. The wealth of commonalities, diversity of molecular targets identified preclinically, and likely multidimensional nature of epileptogenesis argue for a combinatorial strategy in prevention therapy. Going forward, the identification of impending epilepsy biomarkers to allow better patient selection, together with better alignment with multisite preclinical trials in animal models, should guide the clinical testing of new hypotheses for epileptogenesis and its prevention.
Collapse
Affiliation(s)
- Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD, USA
| | | | - Eleonora Aronica
- Department of (Neuro) Pathology, Academic Medical Center and Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| | - Christophe Bernard
- Aix Marseille Univ, Inserm, INS, Instit Neurosci Syst, Marseille, 13005, France
| | - Ingmar Blümcke
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Detlev Boison
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, USA
| | - Martin J Brodie
- Epilepsy Unit, West Glasgow Ambulatory Care Hospital-Yorkhill, Glasgow, UK
| | - Amy R Brooks-Kayal
- Division of Neurology, Departments of Pediatrics and Neurology, University of Colorado School of Medicine, Aurora, CO, USA
- Children's Hospital Colorado, Aurora, CO, USA
- Neuroscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jerome Engel
- Departments of Neurology, Neurobiology, and Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, Brain Research Institute, University of California, Los Angeles, CA, USA
| | | | | | | | | | - Katja Kobow
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | | | - Phillip L Pearl
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Asla Pitkänen
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Noora Puhakka
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Michael A Rogawski
- Department of Neurology, University of California, Davis, Sacramento, CA, USA
| | | | - Matti Sillanpää
- Departments of Child Neurology and General Practice, University of Turku and Turku University Hospital, Turku, Finland
| | - Robert S Sloviter
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Annamaria Vezzani
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Institute for Pharmacological Research, Milan,, Italy
| | - Matthew C Walker
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
36
|
Excitatory Synaptic Input to Hilar Mossy Cells under Basal and Hyperexcitable Conditions. eNeuro 2017; 4:eN-NWR-0364-17. [PMID: 29214210 PMCID: PMC5714709 DOI: 10.1523/eneuro.0364-17.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 11/06/2017] [Accepted: 11/10/2017] [Indexed: 11/21/2022] Open
Abstract
Hilar mossy cells (HMCs) in the hippocampus receive glutamatergic input from dentate granule cells (DGCs) via mossy fibers (MFs) and back-projections from CA3 pyramidal neuron collateral axons. Many fundamental features of these excitatory synapses have not been characterized in detail despite their potential relevance to hippocampal cognitive processing and epilepsy-induced adaptations in circuit excitability. In this study, we compared pre- and postsynaptic parameters between MF and CA3 inputs to HMCs in young and adult mice of either sex and determined the relative contributions of the respective excitatory inputs during in vitro and in vivo models of hippocampal hyperexcitability. The two types of excitatory synapses both exhibited a modest degree of short-term plasticity, with MF inputs to HMCs exhibiting lower paired-pulse (PP) and frequency facilitation than was described previously for MF–CA3 pyramidal cell synapses. MF–HMC synapses exhibited unitary excitatory synaptic currents (EPSCs) of larger amplitude, contained postsynaptic kainate receptors, and had a lower NMDA/AMPA receptor ratio compared to CA3–HMC synapses. Pharmacological induction of hippocampal hyperexcitability in vitro transformed the abundant but relatively weak CA3–HMC connections to very large amplitude spontaneous bursts of compound EPSCs (cEPSCs) in young mice (∼P20) and, to a lesser degree, in adult mice (∼P70). CA3–HMC cEPSCs were also observed in slices prepared from mice with spontaneous seizures several weeks after intrahippocampal kainate injection. Strong excitation of HMCs during synchronous CA3 activity represents an avenue of significant excitatory network generation back to DGCs and might be important in generating epileptic networks.
Collapse
|
37
|
Kim JE, Kang TC. p47Phox/CDK5/DRP1-Mediated Mitochondrial Fission Evokes PV Cell Degeneration in the Rat Dentate Gyrus Following Status Epilepticus. Front Cell Neurosci 2017; 11:267. [PMID: 28919853 PMCID: PMC5585136 DOI: 10.3389/fncel.2017.00267] [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: 06/03/2017] [Accepted: 08/21/2017] [Indexed: 01/31/2023] Open
Abstract
Parvalbumin (PV) is one of the calcium-binding proteins, which plays an important role in the responsiveness of inhibitory neurons to an adaptation to repetitive spikes. Furthermore, PV neurons are highly vulnerable to status epilepticus (SE, prolonged seizure activity), although the underlining mechanism remains to be clarified. In the present study, we found that p47Phox expression was transiently and selectively increased in PV neurons 6 h after SE. This up-regulated p47Phox expression was accompanied by excessive mitochondrial fission. In this time point, CDK5-tyrosine 15 and dynamin-related protein 1 (DRP1)-serine 616 phosphorylations were also increased in PV cells. Apocynin (a p47Phox inhibitor) effectively mitigated PV cell loss via inhibition of CDK5/DRP1 phosphorylations and mitochondrial fragmentation induced by SE. Roscovitine (a CDK5 inhibitor) and Mdivi-1 (a DRP1 inhibitor) attenuated SE-induced PV cell loss by inhibiting aberrant mitochondrial fission. These findings suggest that p47Phox/CDK5/DRP1 may be one of the important upstream signaling pathways in PV cell degeneration induced by SE via excessive mitochondrial fragmentation.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym UniversityChuncheon, South Korea
| | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym UniversityChuncheon, South Korea
| |
Collapse
|
38
|
Remigio GJ, Loewen JL, Heuston S, Helgeson C, White HS, Wilcox KS, West PJ. Corneal kindled C57BL/6 mice exhibit saturated dentate gyrus long-term potentiation and associated memory deficits in the absence of overt neuron loss. Neurobiol Dis 2017; 105:221-234. [PMID: 28624414 PMCID: PMC5538573 DOI: 10.1016/j.nbd.2017.06.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 06/09/2017] [Indexed: 12/25/2022] Open
Abstract
Memory deficits have a significant impact on the quality of life of patients with epilepsy and currently no effective treatments exist to mitigate this comorbidity. While these cognitive comorbidities can be associated with varying degrees of hippocampal cell death and hippocampal sclerosis, more subtle changes in hippocampal physiology independent of cell loss may underlie memory dysfunction in many epilepsy patients. Accordingly, animal models of epilepsy or epileptic processes exhibiting memory deficits in the absence of cell loss could facilitate novel therapy discovery. Mouse corneal kindling is a cost-effective and non-invasive model of focal to bilateral tonic-clonic seizures that may exhibit memory deficits in the absence of cell loss. Therefore, we tested the hypothesis that corneal kindled C57BL/6 mice exhibit spatial pattern processing and memory deficits in a task reliant on DG function and that these impairments would be concurrent with physiological remodeling of the DG as opposed to overt neuron loss. Following corneal kindling, C57BL/6 mice exhibited deficits in a DG-associated spatial memory test - the metric task. Compatible with this finding, we also discovered saturated, and subsequently impaired, LTP of excitatory synaptic transmission at the perforant path to DGC synapse. This saturation of LTP was consistent with evidence suggesting that perforant path to DGC synapses in kindled mice had previously experienced LTP-like changes to their synaptic weights: increased postsynaptic depolarizations in response to equivalent presynaptic input and significantly larger amplitude AMPA receptor mediated spontaneous EPSCs. Additionally, there was evidence for kindling-induced changes in the intrinsic excitability of DGCs: reduced threshold to population spikes under extracellular recording conditions and significantly increased membrane resistances observed in DGCs. Importantly, quantitative immunohistochemical analysis revealed hippocampal astrogliosis in the absence of overt neuron loss. These changes in spatial pattern processing and memory deficits in corneal kindled mice represent a novel model of seizure-induced cognitive dysfunction associated with pathophysiological remodeling of excitatory synaptic transmission and granule cell excitability in the absence of overt cell loss.
Collapse
Affiliation(s)
- Gregory J Remigio
- Interdepartmental Neuroscience Program, University of Utah, Salt Lake City, UT 84108-1210, USA; Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84108-1210, USA
| | - Jaycie L Loewen
- Interdepartmental Neuroscience Program, University of Utah, Salt Lake City, UT 84108-1210, USA; Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84108-1210, USA
| | | | - Colin Helgeson
- Juan Diego Catholic High School, Draper, UT 84020-9035, USA
| | - H Steve White
- Interdepartmental Neuroscience Program, University of Utah, Salt Lake City, UT 84108-1210, USA; Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84108-1210, USA; Anticonvulsant Drug Development Program, University of Utah, Salt Lake City, UT 84108-1210, USA
| | - Karen S Wilcox
- Interdepartmental Neuroscience Program, University of Utah, Salt Lake City, UT 84108-1210, USA; Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84108-1210, USA; Anticonvulsant Drug Development Program, University of Utah, Salt Lake City, UT 84108-1210, USA
| | - Peter J West
- Interdepartmental Neuroscience Program, University of Utah, Salt Lake City, UT 84108-1210, USA; Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84108-1210, USA; Anticonvulsant Drug Development Program, University of Utah, Salt Lake City, UT 84108-1210, USA.
| |
Collapse
|
39
|
Buckmaster PS, Abrams E, Wen X. Seizure frequency correlates with loss of dentate gyrus GABAergic neurons in a mouse model of temporal lobe epilepsy. J Comp Neurol 2017; 525:2592-2610. [PMID: 28425097 PMCID: PMC5963263 DOI: 10.1002/cne.24226] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/11/2017] [Accepted: 04/12/2017] [Indexed: 01/19/2023]
Abstract
Epilepsy occurs in one of 26 people. Temporal lobe epilepsy is common and can be difficult to treat effectively. It can develop after brain injuries that damage the hippocampus. Multiple pathophysiological mechanisms involving the hippocampal dentate gyrus have been proposed. This study evaluated a mouse model of temporal lobe epilepsy to test which pathological changes in the dentate gyrus correlate with seizure frequency and help prioritize potential mechanisms for further study. FVB mice (n = 127) that had experienced status epilepticus after systemic treatment with pilocarpine 31-61 days earlier were video-monitored for spontaneous, convulsive seizures 9 hr/day every day for 24-36 days. Over 4,060 seizures were observed. Seizure frequency ranged from an average of one every 3.6 days to one every 2.1 hr. Hippocampal sections were processed for Nissl stain, Prox1-immunocytochemistry, GluR2-immunocytochemistry, Timm stain, glial fibrillary acidic protein-immunocytochemistry, glutamic acid decarboxylase in situ hybridization, and parvalbumin-immunocytochemistry. Stereological methods were used to measure hilar ectopic granule cells, mossy cells, mossy fiber sprouting, astrogliosis, and GABAergic interneurons. Seizure frequency was not significantly correlated with the generation of hilar ectopic granule cells, the number of mossy cells, the extent of mossy fiber sprouting, the extent of astrogliosis, or the number of GABAergic interneurons in the molecular layer or hilus. Seizure frequency significantly correlated with the loss of GABAergic interneurons in or adjacent to the granule cell layer, but not with the loss of parvalbumin-positive interneurons. These findings prioritize the loss of granule cell layer interneurons for further testing as a potential cause of temporal lobe epilepsy.
Collapse
Affiliation(s)
- Paul S. Buckmaster
- Department of Comparative Medicine, Stanford University, Stanford, California
- Department of Neurology & Neurological Sciences, Stanford University, Stanford, California
| | - Emily Abrams
- Department of Comparative Medicine, Stanford University, Stanford, California
| | - Xiling Wen
- Department of Comparative Medicine, Stanford University, Stanford, California
| |
Collapse
|
40
|
Farrell JS, Colangeli R, Wolff MD, Wall AK, Phillips TJ, George A, Federico P, Teskey GC. Postictal hypoperfusion/hypoxia provides the foundation for a unified theory of seizure-induced brain abnormalities and behavioral dysfunction. Epilepsia 2017. [DOI: 10.1111/epi.13827] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jordan S. Farrell
- Hotchkiss Brain Institute; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| | - Roberto Colangeli
- Hotchkiss Brain Institute; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| | - Marshal D. Wolff
- Hotchkiss Brain Institute; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| | - Alexandra K. Wall
- Hotchkiss Brain Institute; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| | - Thomas J. Phillips
- Hotchkiss Brain Institute; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| | - Antis George
- Hotchkiss Brain Institute; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| | - Paolo Federico
- Hotchkiss Brain Institute; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| | - G. Campbell Teskey
- Hotchkiss Brain Institute; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| |
Collapse
|
41
|
Li Z, You Z, Li M, Pang L, Cheng J, Wang L. Protective Effect of Resveratrol on the Brain in a Rat Model of Epilepsy. Neurosci Bull 2017; 33:273-280. [PMID: 28161868 PMCID: PMC5567521 DOI: 10.1007/s12264-017-0097-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/15/2016] [Indexed: 12/28/2022] Open
Abstract
Accumulating evidence has suggested resveratrol as a promising drug candidate for the treatment of epilepsy. To validate this, we tested the protective effect of resveratrol on a kainic acid (KA)-induced epilepsy model in rats and investigated the underlying mechanism. We found that acute resveratrol application partially inhibited evoked epileptiform discharges in the hippocampal CA1 region. During acute, silent and chronic phases of epilepsy, the expression of hippocampal kainate glutamate receptor (GluK2) and the GABAA receptor alpha1 subunit (GABAAR-alpha1) was up-regulated and down-regulated, respectively. Resveratrol reversed these effects and induced an antiepileptic effect. Furthermore, in the chronic phase, resveratrol treatment inhibited the KA-induced increased glutamate/GABA ratio in the hippocampus. The antiepileptic effects of resveratrol may be partially attributed to the reduction of glutamate-induced excitotoxicity and the enhancement in GABAergic inhibition.
Collapse
MESH Headings
- Animals
- Anticonvulsants/administration & dosage
- Anticonvulsants/pharmacology
- CA1 Region, Hippocampal/drug effects
- CA1 Region, Hippocampal/metabolism
- CA1 Region, Hippocampal/physiopathology
- Disease Models, Animal
- Down-Regulation
- Epilepsy, Temporal Lobe/chemically induced
- Epilepsy, Temporal Lobe/drug therapy
- Epilepsy, Temporal Lobe/metabolism
- Excitatory Amino Acid Agonists/pharmacology
- Glutamic Acid/drug effects
- Kainic Acid/pharmacology
- Male
- Neuroprotective Agents/administration & dosage
- Neuroprotective Agents/pharmacology
- Rats
- Rats, Wistar
- Receptors, GABA-A/drug effects
- Receptors, Kainic Acid/drug effects
- Resveratrol
- Stilbenes/administration & dosage
- Stilbenes/pharmacology
- Up-Regulation
- gamma-Aminobutyric Acid/drug effects
- GluK2 Kainate Receptor
Collapse
Affiliation(s)
- Zhen Li
- Department of Pharmacology, Anhui Medical University, Hefei, 230032, China
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Zhuyan You
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Min Li
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Liang Pang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Juan Cheng
- Department of Pharmacology, Anhui Medical University, Hefei, 230032, China
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Liecheng Wang
- Department of Pharmacology, Anhui Medical University, Hefei, 230032, China.
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
42
|
GoodSmith D, Chen X, Wang C, Kim SH, Song H, Burgalossi A, Christian KM, Knierim JJ. Spatial Representations of Granule Cells and Mossy Cells of the Dentate Gyrus. Neuron 2017; 93:677-690.e5. [PMID: 28132828 DOI: 10.1016/j.neuron.2016.12.026] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 11/01/2016] [Accepted: 12/12/2016] [Indexed: 01/12/2023]
Abstract
Granule cells in the dentate gyrus of the hippocampus are thought to be essential to memory function by decorrelating overlapping input patterns (pattern separation). A second excitatory cell type in the dentate gyrus, the mossy cell, forms an intricate circuit with granule cells, CA3c pyramidal cells, and local interneurons, but the influence of mossy cells on dentate function is often overlooked. Multiple tetrode recordings, supported by juxtacellular recording techniques, showed that granule cells fired very sparsely, whereas mossy cells in the hilus fired promiscuously in multiple locations and in multiple environments. The activity patterns of these cell types thus represent different environments through distinct computational mechanisms: sparse coding in granule cells and changes in firing field locations in mossy cells.
Collapse
Affiliation(s)
- Douglas GoodSmith
- Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Xiaojing Chen
- Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Cheng Wang
- Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sang Hoon Kim
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore MD 21205 USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore MD 21205 USA
| | - Hongjun Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore MD 21205 USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore MD 21205 USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore MD 21205 USA
| | - Andrea Burgalossi
- Werner-Reichardt Centre for Integrative Neuroscience, 72076 Tübingen, Germany
| | - Kimberly M Christian
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore MD 21205 USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore MD 21205 USA
| | - James J Knierim
- Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore MD 21205 USA.
| |
Collapse
|
43
|
Walker MC. Pathophysiology of status epilepticus. Neurosci Lett 2016; 667:84-91. [PMID: 28011391 DOI: 10.1016/j.neulet.2016.12.044] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/16/2016] [Accepted: 12/17/2016] [Indexed: 12/22/2022]
Abstract
Status epilepticus (SE) is the maximal expression of epilepsy with a high morbidity and mortality. It occurs due to the failure of mechanisms that terminate seizures. Both human and animal data indicate that the longer a seizure lasts, the less likely it is to stop. Recent evidence suggests that there is a critical transition from an ictal to a post-ictal state, associated with a transition from a spatio-temporally desynchronized state to a highly synchronized state, respectively. As SE continues, it becomes progressively resistant to drugs, in particular benzodiazepines due partly to NMDA receptor-dependent internalization of GABA(A) receptors. Moreover, excessive calcium entry into neurons through excessive NMDA receptor activation results in activation of nitric oxide synthase, calpains, and NADPH oxidase. The latter enzyme plays a critical part in the generation of seizure-dependent reactive oxygen species. Calcium also accumulates in mitochondria resulting in mitochondrial failure (decreased ATP production), and opening of the mitochondrial permeability transition pore. Together these changes result in status epilepticus-dependent neuronal death via several pathways. Multiple downstream mechanisms including inflammation, break down of the blood-brain barrier, and changes in gene expression can contribute to later pathological processes including chronic epilepsy and cognitive decline.
Collapse
Affiliation(s)
- Matthew C Walker
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London WC1N 3BG, United Kingdom.
| |
Collapse
|
44
|
Csabai D, Seress L, Varga Z, Ábrahám H, Miseta A, Wiborg O, Czéh B. Electron Microscopic Analysis of Hippocampal Axo-Somatic Synapses in a Chronic Stress Model for Depression. Hippocampus 2016; 27:17-27. [PMID: 27571571 PMCID: PMC5215622 DOI: 10.1002/hipo.22650] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2016] [Indexed: 01/01/2023]
Abstract
Stress can alter the number and morphology of excitatory synapses in the hippocampus, but nothing is known about the effect of stress on inhibitory synapses. Here, we used an animal model for depression, the chronic mild stress model, and quantified the number of perisomatic inhibitory neurons and their synapses. We found reduced density of parvalbumin‐positive (PV+) neurons in response to stress, while the density of cholecystokinin‐immunoreactive (CCK+) neurons was unaffected. We did a detailed electron microscopic analysis to quantify the frequency and morphology of perisomatic inhibitory synapses in the hippocampal CA1 area. We analyzed 1100 CA1 pyramidal neurons and 4800 perisomatic terminals in five control and four chronically stressed rats. In the control animals we observed the following parameters: Number of terminals/soma = 57; Number of terminals/100 µm cell perimeter = 10; Synapse/terminal ratio = 32%; Synapse number/100 terminal = 120; Average terminal length = 920nm. None of these parameters were affected by the stress exposure. Overall, these data indicate that despite the depressive‐like behavior and the decrease in the number of perisomatic PV+ neurons in the light microscopic preparations, the number of perisomatic inhibitory synapses on CA1 pyramidal cells was not affected by stress. In the electron microscope, PV+ neurons and the axon terminals appeared to be normal and we did not find any apoptotic or necrotic cells. This data is in sharp contrast to the remarkable remodeling of the excitatory synapses on spines that has been reported in response to stress and depressive‐like behavior. © 2016 The Authors Hippocampus Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Dávid Csabai
- MTA - PTE, Neurobiology of Stress Research Group, Szentágothai Research Center, Pécs, 7624, Hungary
| | - László Seress
- Central Electron Microscope Laboratory, University of Pécs, Medical School, Pécs, 7624, Hungary
| | - Zsófia Varga
- MTA - PTE, Neurobiology of Stress Research Group, Szentágothai Research Center, Pécs, 7624, Hungary
| | - Hajnalka Ábrahám
- Central Electron Microscope Laboratory, University of Pécs, Medical School, Pécs, 7624, Hungary.,Department of Medical Biology, University of Pécs, Medical School, Pécs, 7624, Hungary
| | - Attila Miseta
- Department of Laboratory Medicine, University of Pécs, Medical School, Pécs, 7624, Hungary
| | - Ove Wiborg
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
| | - Boldizsár Czéh
- MTA - PTE, Neurobiology of Stress Research Group, Szentágothai Research Center, Pécs, 7624, Hungary.,Department of Laboratory Medicine, University of Pécs, Medical School, Pécs, 7624, Hungary.,Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
| |
Collapse
|
45
|
Botterill JJ, Nogovitsyn N, Caruncho HJ, Kalynchuk LE. Selective plasticity of hippocampal GABAergic interneuron populations following kindling of different brain regions. J Comp Neurol 2016; 525:389-406. [PMID: 27362579 DOI: 10.1002/cne.24071] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 06/06/2016] [Accepted: 06/28/2016] [Indexed: 12/20/2022]
Abstract
The vulnerability and plasticity of hippocampal GABAergic interneurons is a topic of broad interest and debate in the field of epilepsy. In this experiment, we used the electrical kindling model of epilepsy to determine whether seizures that originate in different brain regions have differential effects on hippocampal interneuron subpopulations. Long-Evans rats received 99 electrical stimulations of the hippocampus, amygdala, or caudate nucleus, followed by sacrifice and immunohistochemical or western blot analyses. We analyzed markers of dendritic (somatostatin), perisomatic (parvalbumin), and interneuron-selective (calretinin) inhibition, as well as an overall indicator (GAD67) of interneuron distribution across all major hippocampal subfields. Our results indicate that kindling produces selective effects on the number and morphology of different functional classes of GABAergic interneurons. In particular, limbic kindling appears to enhance dendritic inhibition, indicated by a greater number of somatostatin-immunoreactive (-ir) cells in the CA1 pyramidal layer and robust morphological sprouting in the dentate gyrus. We also found a reduction in the number of interneuron-selective calretinin-ir cells in the dentate gyrus of hippocampal-kindled rats, which suggests a possible reduction of synchronized dendritic inhibition. In contrast, perisomatic inhibition indicated by parvalbumin immunoreactivity appears to be largely resilient to the effects of kindling. Finally, we found a significant induction in the number of GAD67-cells in caudate-kindled rats in the dentate gyrus and CA3 hippocampal subfields. Taken together, our results demonstrate that kindling has subfield-selective effects on the different functional classes of hippocampal GABAergic interneurons. J. Comp. Neurol. 525:389-406, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- J J Botterill
- Department of Psychology, University of Saskatchewan, Saskatoon, SK, Canada
| | - N Nogovitsyn
- Department of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - H J Caruncho
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - L E Kalynchuk
- Department of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
46
|
Impact of rapamycin on status epilepticus induced hippocampal pathology and weight gain. Exp Neurol 2016; 280:1-12. [PMID: 26995324 DOI: 10.1016/j.expneurol.2016.03.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 03/11/2016] [Accepted: 03/14/2016] [Indexed: 02/07/2023]
Abstract
Growing evidence implicates the dentate gyrus in temporal lobe epilepsy (TLE). Dentate granule cells limit the amount of excitatory signaling through the hippocampus and exhibit striking neuroplastic changes that may impair this function during epileptogenesis. Furthermore, aberrant integration of newly-generated granule cells underlies the majority of dentate restructuring. Recently, attention has focused on the mammalian target of rapamycin (mTOR) signaling pathway as a potential mediator of epileptogenic change. Systemic administration of the mTOR inhibitor rapamycin has promising therapeutic potential, as it has been shown to reduce seizure frequency and seizure severity in rodent models. Here, we tested whether mTOR signaling facilitates abnormal development of granule cells during epileptogenesis. We also examined dentate inflammation and mossy cell death in the dentate hilus. To determine if mTOR activation is necessary for abnormal granule cell development, transgenic mice that harbored fluorescently-labeled adult-born granule cells were treated with rapamycin following pilocarpine-induced status epilepticus. Systemic rapamycin effectively blocked phosphorylation of S6 protein (a readout of mTOR activity) and reduced granule cell mossy fiber axon sprouting. However, the accumulation of ectopic granule cells and granule cells with aberrant basal dendrites was not significantly reduced. Mossy cell death and reactive astrocytosis were also unaffected. These data suggest that anti-epileptogenic effects of mTOR inhibition may be mediated by mechanisms other than inhibition of these common dentate pathologies. Consistent with this conclusion, rapamycin prevented pathological weight gain in epileptic mice, suggesting that rapamycin might act on central circuits or even peripheral tissues controlling weight gain in epilepsy.
Collapse
|
47
|
Scharfman HE, Myers CE. Corruption of the dentate gyrus by "dominant" granule cells: Implications for dentate gyrus function in health and disease. Neurobiol Learn Mem 2016; 129:69-82. [PMID: 26391451 PMCID: PMC4792754 DOI: 10.1016/j.nlm.2015.09.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 09/02/2015] [Accepted: 09/06/2015] [Indexed: 12/31/2022]
Abstract
The dentate gyrus (DG) and area CA3 of the hippocampus are highly organized lamellar structures which have been implicated in specific cognitive functions such as pattern separation and pattern completion. Here we describe how the anatomical organization and physiology of the DG and CA3 are consistent with structures that perform pattern separation and completion. We then raise a new idea related to the complex circuitry of the DG and CA3 where CA3 pyramidal cell 'backprojections' play a potentially important role in the sparse firing of granule cells (GCs), considered important in pattern separation. We also propose that GC axons, the mossy fibers, already known for their highly specialized structure, have a dynamic function that imparts variance--'mossy fiber variance'--which is important to pattern separation and completion. Computational modeling is used to show that when a subset of GCs become 'dominant,' one consequence is loss of variance in the activity of mossy fiber axons and a reduction in pattern separation and completion in the model. Empirical data are then provided using an example of 'dominant' GCs--subsets of GCs that develop abnormally and have increased excitability. Notably, these abnormal GCs have been identified in animal models of disease where DG-dependent behaviors are impaired. Together these data provide insight into pattern separation and completion, and suggest that behavioral impairment could arise from dominance of a subset of GCs in the DG-CA3 network.
Collapse
Affiliation(s)
- Helen E Scharfman
- The Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY 10962, United States; Departments of Child & Adolescent Psychiatry, Physiology & Neuroscience, and Psychiatry, New York University Langone Medical Center, United States.
| | - Catherine E Myers
- VA New Jersey Health Care System, VA Medical Center, NeuroBehavioral Research Lab (Mail Stop 15a), 385 Tremont Avenue, East Orange, NJ 07018, United States; Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, United States
| |
Collapse
|
48
|
GABA-ergic cell therapy for epilepsy: Advances, limitations and challenges. Neurosci Biobehav Rev 2015; 62:35-47. [PMID: 26748379 DOI: 10.1016/j.neubiorev.2015.12.014] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 11/06/2015] [Accepted: 12/28/2015] [Indexed: 01/04/2023]
Abstract
Diminution in the number of gamma-amino butyric acid positive (GABA-ergic) interneurons and their axon terminals, and/or alterations in functional inhibition are conspicuous brain alterations believed to contribute to the persistence of seizures in acquired epilepsies such as temporal lobe epilepsy. This has steered a perception that replacement of lost GABA-ergic interneurons would improve inhibitory synaptic neurotransmission in the epileptic brain region and thereby reduce the occurrence of seizures. Indeed, studies using animal prototypes have reported that grafting of GABA-ergic progenitors derived from multiple sources into epileptic regions can reduce seizures. This review deliberates recent advances, limitations and challenges concerning the development of GABA-ergic cell therapy for epilepsy. The efficacy and limitations of grafts of primary GABA-ergic progenitors from the embryonic lateral ganglionic eminence and medial ganglionic eminence (MGE), neural stem/progenitor cells expanded from MGE, and MGE-like progenitors generated from human pluripotent stem cells for alleviating seizures and co-morbidities of epilepsy are conferred. Additional studies required for possible clinical application of GABA-ergic cell therapy for epilepsy are also summarized.
Collapse
|
49
|
Hendrickson PJ, Yu GJ, Song D, Berger TW. Interactions between Inhibitory Interneurons and Excitatory Associational Circuitry in Determining Spatio-Temporal Dynamics of Hippocampal Dentate Granule Cells: A Large-Scale Computational Study. Front Syst Neurosci 2015; 9:155. [PMID: 26635545 PMCID: PMC4647071 DOI: 10.3389/fnsys.2015.00155] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/27/2015] [Indexed: 11/16/2022] Open
Abstract
This paper reports on findings from a million-cell granule cell model of the rat dentate gyrus that was used to explore the contributions of local interneuronal and associational circuits to network-level activity. The model contains experimentally derived morphological parameters for granule cells, which each contain approximately 200 compartments, and biophysical parameters for granule cells, basket cells, and mossy cells that were based both on electrophysiological data and previously published models. Synaptic input to cells in the model consisted of glutamatergic AMPA-like EPSPs and GABAergic-like IPSPs from excitatory and inhibitory neurons, respectively. The main source of input to the model was from layer II entorhinal cortical neurons. Network connectivity was constrained by the topography of the system, and was derived from axonal transport studies, which provided details about the spatial spread of axonal terminal fields, as well as how subregions of the medial and lateral entorhinal cortices project to subregions of the dentate gyrus. Results of this study show that strong feedback inhibition from the basket cell population can cause high-frequency rhythmicity in granule cells, while the strength of feedforward inhibition serves to scale the total amount of granule cell activity. Results furthermore show that the topography of local interneuronal circuits can have just as strong an impact on the development of spatio-temporal clusters in the granule cell population as the perforant path topography does, both sharpening existing clusters and introducing new ones with a greater spatial extent. Finally, results show that the interactions between the inhibitory and associational loops can cause high frequency oscillations that are modulated by a low-frequency oscillatory signal. These results serve to further illustrate the importance of topographical constraints on a global signal processing feature of a neural network, while also illustrating how rich spatio-temporal and oscillatory dynamics can evolve from a relatively small number of interacting local circuits.
Collapse
Affiliation(s)
- Phillip J Hendrickson
- Department of Biomedical Engineering, University of Southern California Los Angeles, CA, USA
| | - Gene J Yu
- Department of Biomedical Engineering, University of Southern California Los Angeles, CA, USA
| | - Dong Song
- Department of Biomedical Engineering, University of Southern California Los Angeles, CA, USA
| | - Theodore W Berger
- Department of Biomedical Engineering, University of Southern California Los Angeles, CA, USA
| |
Collapse
|
50
|
Abstract
Seizure activity in the hippocampal region strongly affects stem cell-associated plasticity in the adult dentate gyrus. Here, we describe how seizures in rodent models of mesial temporal lobe epilepsy (mTLE) affect multiple steps in the developmental course from the dividing neural stem cell to the migrating and integrating newborn neuron. Furthermore, we discuss recent evidence indicating either that seizure-induced aberrant neurogenesis may contribute to the epileptic disease process or that altered neurogenesis after seizures may represent an attempt of the injured brain to repair itself. Last, we describe how dysfunction of adult neurogenesis caused by chronic seizures may play an important role in the cognitive comorbidities associated with mTLE.
Collapse
Affiliation(s)
| | - Jack M Parent
- Department of Neurology, University of Michigan Medical Center and VA Ann Arbor Healthcare System, Ann Arbor, Michigan 48109
| |
Collapse
|