1
|
Riva M, Moriceau S, Morabito A, Dossi E, Sanchez-Bellot C, Azzam P, Navas-Olive A, Gal B, Dori F, Cid E, Ledonne F, David S, Trovero F, Bartolomucci M, Coppola E, Rebola N, Depaulis A, Rouach N, de la Prida LM, Oury F, Pierani A. Aberrant survival of hippocampal Cajal-Retzius cells leads to memory deficits, gamma rhythmopathies and susceptibility to seizures in adult mice. Nat Commun 2023; 14:1531. [PMID: 36934089 PMCID: PMC10024761 DOI: 10.1038/s41467-023-37249-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 03/08/2023] [Indexed: 03/20/2023] Open
Abstract
Cajal-Retzius cells (CRs) are transient neurons, disappearing almost completely in the postnatal neocortex by programmed cell death (PCD), with a percentage surviving up to adulthood in the hippocampus. Here, we evaluate CR's role in the establishment of adult neuronal and cognitive function using a mouse model preventing Bax-dependent PCD. CRs abnormal survival resulted in impairment of hippocampus-dependent memory, associated in vivo with attenuated theta oscillations and enhanced gamma activity in the dorsal CA1. At the cellular level, we observed transient changes in the number of NPY+ cells and altered CA1 pyramidal cell spine density. At the synaptic level, these changes translated into enhanced inhibitory currents in hippocampal pyramidal cells. Finally, adult mutants displayed an increased susceptibility to lethal tonic-clonic seizures in a kainate model of epilepsy. Our data reveal that aberrant survival of a small proportion of postnatal hippocampal CRs results in cognitive deficits and epilepsy-prone phenotypes in adulthood.
Collapse
Affiliation(s)
- Martina Riva
- Université Paris Cité, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015, Paris, France
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014, Paris, France
| | - Stéphanie Moriceau
- Platform for Neurobehavioral and metabolism, Structure Fédérative de Recherche Necker, 26 INSERM US24/CNRS UAR, 3633, Paris, France
| | - Annunziato Morabito
- Sorbonne Université, Institut Du Cerveau-Paris Brain Institute-ICM, Inserm U1127, CNRS UMR 7225, 47 Boulevard de l'Hopital, 75013, Paris, France
| | - Elena Dossi
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | | | - Patrick Azzam
- Université Paris Cité, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015, Paris, France
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014, Paris, France
| | | | - Beatriz Gal
- Instituto Cajal, CSIC, Madrid, Spain
- Universidad Camilo José Cela, Madrid, Spain
| | - Francesco Dori
- Université Paris Cité, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015, Paris, France
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014, Paris, France
| | - Elena Cid
- Instituto Cajal, CSIC, Madrid, Spain
| | - Fanny Ledonne
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014, Paris, France
| | - Sabrina David
- Key-Obs SAS, 13 avenue Buffon, 45100, Orléans, France
| | | | - Magali Bartolomucci
- Université Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Eva Coppola
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014, Paris, France
| | - Nelson Rebola
- Sorbonne Université, Institut Du Cerveau-Paris Brain Institute-ICM, Inserm U1127, CNRS UMR 7225, 47 Boulevard de l'Hopital, 75013, Paris, France
| | - Antoine Depaulis
- Université Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Nathalie Rouach
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | | | - Franck Oury
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, 75015, Paris, France
| | - Alessandra Pierani
- Université Paris Cité, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015, Paris, France.
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014, Paris, France.
- GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, 75014, Paris, France.
| |
Collapse
|
2
|
Kasahara Y, Nakashima H, Nakashima K. Seizure-induced hilar ectopic granule cells in the adult dentate gyrus. Front Neurosci 2023; 17:1150283. [PMID: 36937666 PMCID: PMC10017466 DOI: 10.3389/fnins.2023.1150283] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
Epilepsy is a chronic neurological disorder characterized by hypersynchronous spontaneous recurrent seizures, and affects approximately 50 million people worldwide. Cumulative evidence has revealed that epileptogenic insult temporarily increases neurogenesis in the hippocampus; however, a fraction of the newly generated neurons are integrated abnormally into the existing neural circuits. The abnormal neurogenesis, including ectopic localization of newborn neurons in the hilus, formation of abnormal basal dendrites, and disorganization of the apical dendrites, rewires hippocampal neural networks and leads to the development of spontaneous seizures. The central roles of hilar ectopic granule cells in regulating hippocampal excitability have been suggested. In this review, we introduce recent findings about the migration of newborn granule cells to the dentate hilus after seizures and the roles of seizure-induced ectopic granule cells in the epileptic brain. In addition, we delineate possible intrinsic and extrinsic mechanisms underlying this abnormality. Finally, we suggest that the regulation of seizure-induced ectopic cells can be a promising target for epilepsy therapy and provide perspectives on future research directions.
Collapse
|
3
|
Leifeld J, Förster E, Reiss G, Hamad MIK. Considering the Role of Extracellular Matrix Molecules, in Particular Reelin, in Granule Cell Dispersion Related to Temporal Lobe Epilepsy. Front Cell Dev Biol 2022; 10:917575. [PMID: 35733853 PMCID: PMC9207388 DOI: 10.3389/fcell.2022.917575] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
The extracellular matrix (ECM) of the nervous system can be considered as a dynamically adaptable compartment between neuronal cells, in particular neurons and glial cells, that participates in physiological functions of the nervous system. It is mainly composed of carbohydrates and proteins that are secreted by the different kinds of cell types found in the nervous system, in particular neurons and glial cells, but also other cell types, such as pericytes of capillaries, ependymocytes and meningeal cells. ECM molecules participate in developmental processes, synaptic plasticity, neurodegeneration and regenerative processes. As an example, the ECM of the hippocampal formation is involved in degenerative and adaptive processes related to epilepsy. The role of various components of the ECM has been explored extensively. In particular, the ECM protein reelin, well known for orchestrating the formation of neuronal layer formation in the cerebral cortex, is also considered as a player involved in the occurrence of postnatal granule cell dispersion (GCD), a morphologically peculiar feature frequently observed in hippocampal tissue from epileptic patients. Possible causes and consequences of GCD have been studied in various in vivo and in vitro models. The present review discusses different interpretations of GCD and different views on the role of ECM protein reelin in the formation of this morphological peculiarity.
Collapse
Affiliation(s)
- Jennifer Leifeld
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum, Germany
- Department of Biochemistry I—Receptor Biochemistry, Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Bochum, Germany
- *Correspondence: Jennifer Leifeld, ; Eckart Förster,
| | - Eckart Förster
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum, Germany
- *Correspondence: Jennifer Leifeld, ; Eckart Förster,
| | - Gebhard Reiss
- Institute for Anatomy and Clinical Morphology, School of Medicine, Faculty of Health, Witten/ Herdecke University, Witten, Germany
| | - Mohammad I. K. Hamad
- Institute for Anatomy and Clinical Morphology, School of Medicine, Faculty of Health, Witten/ Herdecke University, Witten, Germany
| |
Collapse
|
4
|
Prume M, Rollenhagen A, Yakoubi R, Sätzler K, Lübke JH. Quantitative Three-Dimensional Reconstructions of Excitatory Synaptic Boutons in the "Barrel Field" of the Adult "Reeler" Mouse Somatosensory Neocortex: A Comparative Fine-Scale Electron Microscopic Analysis with the Wild Type Mouse. Cereb Cortex 2020; 30:3209-3227. [PMID: 31813963 DOI: 10.1093/cercor/bhz304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Synapses are key structural determinants for information processing and computations in the normal and pathologically altered brain. Here, the quantitative morphology of excitatory synaptic boutons in the "reeler" mutant, a model system for various neurological disorders, was investigated and compared with wild-type (WT) mice using high-resolution, fine-scale electron microscopy (EM) and quantitative three-dimensional (3D) models of synaptic boutons. Beside their overall geometry, the shape and size of presynaptic active zones (PreAZs) and postsynaptic densities (PSDs) forming the active zones and the three pools of synaptic vesicles (SVs), namely the readily releasable pool (RRP), the recycling pool (RP), and the resting pool, were quantified. Although the reeler mouse neocortex is severely disturbed, no significant differences were found in most of the structural parameters investigated: the size of boutons (~3 μm2), size of the PreAZs and PSDs (~0.17 μm2), total number of SVs, and SVs within a perimeter (p) of 10 nm and p20 nm RRP; the p60 nm, p100 nm, and p60-p200 nm RP; and the resting pool, except the synaptic cleft width. Taken together, the synaptic organization and structural composition of synaptic boutons in the reeler neocortex remain comparably "normal" and may thus contribute to a "correct" wiring of neurons within the reeler cortical network.
Collapse
Affiliation(s)
- Miriam Prume
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, 52425 Jülich, Germany
| | - Astrid Rollenhagen
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, 52425 Jülich, Germany
| | - Rachida Yakoubi
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, 52425 Jülich, Germany
| | - Kurt Sätzler
- School of Biomedical Sciences, University of Ulster, Londonderry BT52 1SA, UK
| | - Joachim Hr Lübke
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, 52425 Jülich, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH University Hospital Aachen, 52074 Aachen, Germany.,JARA Translational Brain Medicine, Jülich/Aachen, Germany
| |
Collapse
|
5
|
Machado RA, Benjumea-Cuartas V, Zapata Berruecos JF, Agudelo-Flóres PM, Salazar-Peláez LM. Reelin, tau phosphorylation and psychiatric complications in patients with hippocampal sclerosis and structural abnormalities in temporal lobe epilepsy. Epilepsy Behav 2019; 96:192-199. [PMID: 31150999 DOI: 10.1016/j.yebeh.2019.04.052] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/17/2019] [Accepted: 04/27/2019] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Temporal lobe epilepsy (TLE) is the most common adult epileptic syndrome. About 30-70% of those cases have neuropsychiatric complications. More than 10% of patients have TLE because of focal cortical dysplasia (FCD) type IIIa. OBJECTIVES The objective of this study was to review the evidence of reelin (RELN) deficiency and tau phosphorylation role in the histopathological, neuropsychiatric, and hyperexcitability features in TLE because of dysplasia type IIIa. METHODS The current literature was reviewed using Cochrane, EMBASE, PROSPERO, MEDLINE, and PubMed from 1995 to July 2018. Articles of interest were reviewed by one investigator (RAM). RESULTS Reelin deficit is related to an abnormal migration of neurons in dentate gyrus, and its deficit causes dentate gyrus abnormalities, which in turn has been associated with memory deficits in patients with TLE. A decreased in the expression of RELN ribonucleic acid (RNA) was found in patients with TLE and dysplasia type IIIa compared with patients with TLE and isolated hippocampal sclerosis (HS). Reelin might affect the distribution and dynamic instability of microtubules within neurons in the cerebral cortex and their phosphorylation. Amyloid pathology, tauopathy, or phosphorylated tau (p-tau) overexpression has been reported in epileptic human brain and in animal models of epilepsy. CONCLUSION Reelin deficit may determine an abnormal cortical lamination and dentate gyrus dispersion and might be associated with an abnormal tau phosphorylation. These processes can be associated with an abnormal hyperexcitability, neuropsychiatric complications, and a myriad of typical histopathological features seen in patients with TLE because of dysplasia type IIIa.
Collapse
Affiliation(s)
| | - Vanesa Benjumea-Cuartas
- Neurologist-epileptologist at Neurology Institute of Colombia. Grupo de Investigación en Ciencias Básicas, Escuela de Graduados, Universidad CES
| | - José Fernando Zapata Berruecos
- Neurology at Neurology Institute of Colombia, Grupo de Investigación en Ciencias Básicas, Escuela de Graduados, Universidad CES
| | | | | |
Collapse
|
6
|
Prume M, Rollenhagen A, Lübke JHR. Structural and Synaptic Organization of the Adult Reeler Mouse Somatosensory Neocortex: A Comparative Fine-Scale Electron Microscopic Study of Reeler With Wild Type Mice. Front Neuroanat 2018; 12:80. [PMID: 30344480 PMCID: PMC6182073 DOI: 10.3389/fnana.2018.00080] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 09/13/2018] [Indexed: 11/17/2022] Open
Abstract
The reeler mouse has been widely used to study various aspects of cortico- and synaptogenesis, but also as a model for several neurological and neurodegenerative disorders. In contrast to development, comparably little is known about the neuronal composition and synaptic organization of the adult reeler mouse neocortex, in particular at the fine-scale electron microscopic level, which was investigated here and compared with wild type (WT) mice. In this study, the “barrel field” of the adult reeler and WT mouse somatosensory neocortex is used as a model system. In reeler the characteristic six-layered structure is no longer existent, but replaced by a conglomerate of neurons organized in homologous clusters with maintained morphological identity and heterologous clusters between neurons and/or oligodendrocytes. These clusters are loosely scattered throughout the neocortical mass between the pial surface and the white matter. In contrast to WT, layer 1 (L1), if existent, seems to be diluted into the volume of the neocortical mass with no clear boundary. L1 also contains clusters of migrated or persistent neurons, oligodendro- and astrocytes. As in WT, myelinated and unmyelinated axons were found throughout the neocortical mass, but in reeler they were organized in massive fiber bundles with a high fiber packing density. A prominent and massive thalamocortical projection traverses through the neocortical mass, always accompanied by numerous “active” oligodendrocytes whereas in WT no such projections were found and “silent” oligodendrocytes were restricted to the white matter. In the adult reeler mouse neocortex, synaptic boutons terminate on somata, dendritic shafts, spines of different types and axon initial segments with no signs of structural distortion and/or degeneration, indicating a “normal” postsynaptic innervation pattern of neurons. In addition, synaptic complexes between boutons and their postsynaptic targets are tightly ensheathed by fine astrocytic processes, as in WT. In conclusion, the neuronal clusters may represent a possible alternative organization principle in adult reeler mice “replacing” layer formation. If so, these homologous clusters may represent individual “functional units” where neurons are highly interconnected and may function as the equivalent of neurons integrated in a cortical layer. The structural composition and postsynaptic innervation pattern of neurons by synaptic boutons provide the structural basis for the establishment of a functional although altered cortical network in the adult reeler mouse.
Collapse
Affiliation(s)
- Miriam Prume
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Jülich, Germany
| | - Astrid Rollenhagen
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Jülich, Germany
| | - Joachim H R Lübke
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Jülich, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH University Hospital Aachen, Aachen, Germany.,JARA Translational Brain Medicine, Jülich, Germany
| |
Collapse
|
7
|
Katsarou A, Moshé SL, Galanopoulou AS. INTERNEURONOPATHIES AND THEIR ROLE IN EARLY LIFE EPILEPSIES AND NEURODEVELOPMENTAL DISORDERS. Epilepsia Open 2017; 2:284-306. [PMID: 29062978 PMCID: PMC5650248 DOI: 10.1002/epi4.12062] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2017] [Indexed: 12/22/2022] Open
Abstract
GABAergic interneurons control the neural circuitry and network activity in the brain. The advances in genetics have identified genes that control the development, maturation and integration of GABAergic interneurons and implicated them in the pathogenesis of epileptic encephalopathies or neurodevelopmental disorders. For example, mutations of the Aristaless-Related homeobox X-linked gene (ARX) may result in defective GABAergic interneuronal migration in infants with epileptic encephalopathies like West syndrome (WS), Ohtahara syndrome or X-linked lissencephaly with abnormal genitalia (XLAG). The concept of "interneuronopathy", i.e. impaired development, migration or function of interneurons, has emerged as a possible etiopathogenic mechanism for epileptic encephalopathies. Treatments that enhance GABA levels, may help seizure control but do not necessarily show disease modifying effect. On the other hand, interneuronopathies can be seen in other conditions in which epilepsy may not be the primary manifestation, such as autism. In this review, we plan to outline briefly the current state of knowledge on the origin, development, and migration and integration of GABAergic interneurons, present neurodevelopmental conditions, with or without epilepsy, that have been associated with interneuronopathies and discuss the evidence linking certain types of interneuronal dysfunction with epilepsy and/or cognitive or behavioral deficits.
Collapse
Affiliation(s)
- Anna‐Maria Katsarou
- Laboratory of Developmental EpilepsySaul R. Korey Department of NeurologyAlbert Einstein College of MedicineBronxNew YorkU.S.A.
| | - Solomon L. Moshé
- Laboratory of Developmental EpilepsySaul R. Korey Department of NeurologyAlbert Einstein College of MedicineBronxNew YorkU.S.A.
- Dominick P. Purpura Department of NeuroscienceMontefiore/Einstein Epilepsy CenterAlbert Einstein College of MedicineBronxNew YorkU.S.A.
- Department of PediatricsAlbert Einstein College of MedicineBronxNew YorkU.S.A.
| | - Aristea S. Galanopoulou
- Laboratory of Developmental EpilepsySaul R. Korey Department of NeurologyAlbert Einstein College of MedicineBronxNew YorkU.S.A.
- Dominick P. Purpura Department of NeuroscienceMontefiore/Einstein Epilepsy CenterAlbert Einstein College of MedicineBronxNew YorkU.S.A.
| |
Collapse
|
8
|
Bozzi Y, Provenzano G, Casarosa S. Neurobiological bases of autism-epilepsy comorbidity: a focus on excitation/inhibition imbalance. Eur J Neurosci 2017; 47:534-548. [PMID: 28452083 DOI: 10.1111/ejn.13595] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/18/2017] [Accepted: 04/21/2017] [Indexed: 12/13/2022]
Abstract
Autism spectrum disorders (ASD) and epilepsy are common neurological diseases of childhood, with an estimated incidence of approximately 0.5-1% of the worldwide population. Several genetic, neuroimaging and neuropathological studies clearly showed that both ASD and epilepsy have developmental origins and a substantial degree of heritability. Most importantly, ASD and epilepsy frequently coexist in the same individual, suggesting a common neurodevelopmental basis for these disorders. Genome-wide association studies recently allowed for the identification of a substantial number of genes involved in ASD and epilepsy, some of which are mutated in syndromes presenting both ASD and epilepsy clinical features. At the cellular level, both preclinical and clinical studies indicate that the different genetic causes of ASD and epilepsy may converge to perturb the excitation/inhibition (E/I) balance, due to the dysfunction of excitatory and inhibitory circuits in various brain regions. Metabolic and immune dysfunctions, as well as environmental causes also contribute to ASD pathogenesis. Thus, an E/I imbalance resulting from neurodevelopmental deficits of multiple origins might represent a common pathogenic mechanism for both diseases. Here, we will review the most significant studies supporting these hypotheses. A deeper understanding of the molecular and cellular determinants of autism-epilepsy comorbidity will pave the way to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Yuri Bozzi
- Neurodevelopmental Disorders Research Group, Centre for Mind/Brain Sciences, University of Trento, via Sommarive 9, 38123, Povo, Trento, Italy.,CNR Neuroscience Institute, Pisa, Italy
| | - Giovanni Provenzano
- Laboratory of Molecular Neuropathology, Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Simona Casarosa
- CNR Neuroscience Institute, Pisa, Italy.,Laboratory of Neural Development and Regeneration, Centre for Integrative Biology, University of Trento, Trento, Italy
| |
Collapse
|
9
|
Kim KC, Gonzales EL, Lázaro MT, Choi CS, Bahn GH, Yoo HJ, Shin CY. Clinical and Neurobiological Relevance of Current Animal Models of Autism Spectrum Disorders. Biomol Ther (Seoul) 2016; 24:207-43. [PMID: 27133257 PMCID: PMC4859786 DOI: 10.4062/biomolther.2016.061] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 04/05/2016] [Indexed: 12/24/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social and communication impairments, as well as repetitive and restrictive behaviors. The phenotypic heterogeneity of ASD has made it overwhelmingly difficult to determine the exact etiology and pathophysiology underlying the core symptoms, which are often accompanied by comorbidities such as hyperactivity, seizures, and sensorimotor abnormalities. To our benefit, the advent of animal models has allowed us to assess and test diverse risk factors of ASD, both genetic and environmental, and measure their contribution to the manifestation of autistic symptoms. At a broader scale, rodent models have helped consolidate molecular pathways and unify the neurophysiological mechanisms underlying each one of the various etiologies. This approach will potentially enable the stratification of ASD into clinical, molecular, and neurophenotypic subgroups, further proving their translational utility. It is henceforth paramount to establish a common ground of mechanistic theories from complementing results in preclinical research. In this review, we cluster the ASD animal models into lesion and genetic models and further classify them based on the corresponding environmental, epigenetic and genetic factors. Finally, we summarize the symptoms and neuropathological highlights for each model and make critical comparisons that elucidate their clinical and neurobiological relevance.
Collapse
Affiliation(s)
- Ki Chan Kim
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Edson Luck Gonzales
- Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Seoul 05029, Republic of Korea.,School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - María T Lázaro
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chang Soon Choi
- Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Seoul 05029, Republic of Korea.,School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Geon Ho Bahn
- Department of Neuropsychiatry, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hee Jeong Yoo
- Department of Neuropsychiatry, Seoul National University Bungdang Hospital, Seongnam 13620, Republic of Korea
| | - Chan Young Shin
- Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Seoul 05029, Republic of Korea.,School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
10
|
Abstract
The name of Jan Evangelista Purkyně and the cerebellum belong inseparably together. He was the first who saw and described the largest nerve cells in the brain, de facto in the cerebellum. The most distinguished researchers of the nervous system then showed him the highest recognition by naming these neurons as Purkinje cells. Through experiments by J. E. Purkyně and his followers properly functionally was attributed to the cerebellum share in precision of motor skills. Despite ongoing and fruitful research, after a relatively long time, especially in the last two decades, scientists had to constantly replenish and re-evaluate the traditional conception of the cerebellum and formulate a new one. It started in the early 1990s, when it was found that cerebellar cortex contains more neurons than the cerebral cortex. Shortly thereafter it was gradually revealed that such enormous numbers of neural cells are not without an impact on brain functions and that the cerebellum, except its traditional role in the motor skills, also participates in higher nervous activity. These new findings were obtained thanks to the introduction of modern methods of examination into the clinical praxis, and experimental procedures using animal models of cerebellar disorders described below.
Collapse
Affiliation(s)
- F Vožeh
- Department of Pathophysiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.
| |
Collapse
|
11
|
Ching AS, Ahmad-Annuar A. A Perspective on the Role of microRNA-128 Regulation in Mental and Behavioral Disorders. Front Cell Neurosci 2015; 9:465. [PMID: 26696825 PMCID: PMC4677093 DOI: 10.3389/fncel.2015.00465] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 11/16/2015] [Indexed: 12/18/2022] Open
Abstract
MiRNAs are short, non-coding RNA molecules that regulate gene expression post-transcriptionally. Over the past decade, misregulated miRNA pathways have been associated with various diseases such as cancer, neurodegenerative diseases, and neurodevelopmental disorders. In this article, we aim to discuss the role played by miR-128 in neuropsychiatric disorders, and highlight potential target genes from an in silico analysis of predicted miR-128 targets. We also discuss the differences of target gene determination based on a bioinformatics or empirical approach. Using data from TargetScan and published reports, we narrowed the miR-128 target gene list to those that are known to be associated with neuropsychiatric disorders, and found that these genes can be classified into 29 gene clusters and are mostly enriched in cancer and MAPK signaling pathways. We also highlight some recent studies on several of the miR-128 targets which should be investigated further as potential candidate genes for therapeutic interventions.
Collapse
Affiliation(s)
- Ai-Sze Ching
- Department of Biomedical Science, Faculty of Medicine, University of Malaya Kuala Lumpur, Malaysia
| | - Azlina Ahmad-Annuar
- Department of Biomedical Science, Faculty of Medicine, University of Malaya Kuala Lumpur, Malaysia
| |
Collapse
|
12
|
Stouffer MA, Golden JA, Francis F. Neuronal migration disorders: Focus on the cytoskeleton and epilepsy. Neurobiol Dis 2015; 92:18-45. [PMID: 26299390 DOI: 10.1016/j.nbd.2015.08.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 08/05/2015] [Accepted: 08/12/2015] [Indexed: 01/28/2023] Open
Abstract
A wide spectrum of focal, regional, or diffuse structural brain abnormalities, collectively known as malformations of cortical development (MCDs), frequently manifest with intellectual disability (ID), epilepsy, and/or autistic spectrum disorder (ASD). As the acronym suggests, MCDs are perturbations of the normal architecture of the cerebral cortex and hippocampus. The pathogenesis of these disorders remains incompletely understood; however, one area that has provided important insights has been the study of neuronal migration. The amalgamation of human genetics and experimental studies in animal models has led to the recognition that common genetic causes of neurodevelopmental disorders, including many severe epilepsy syndromes, are due to mutations in genes regulating the migration of newly born post-mitotic neurons. Neuronal migration genes often, though not exclusively, code for proteins involved in the function of the cytoskeleton. Other cellular processes, such as cell division and axon/dendrite formation, which similarly depend on cytoskeletal functions, may also be affected. We focus here on how the susceptibility of the highly organized neocortex and hippocampus may be due to their laminar organization, which involves the tight regulation, both temporally and spatially, of gene expression, specialized progenitor cells, the migration of neurons over large distances and a birthdate-specific layering of neurons. Perturbations in neuronal migration result in abnormal lamination, neuronal differentiation defects, abnormal cellular morphology and circuit formation. Ultimately this results in disorganized excitatory and inhibitory activity leading to the symptoms observed in individuals with these disorders.
Collapse
Affiliation(s)
- Melissa A Stouffer
- INSERM UMRS 839, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Jeffrey A Golden
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Fiona Francis
- INSERM UMRS 839, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie, Paris, France; Institut du Fer à Moulin, Paris, France.
| |
Collapse
|
13
|
Ogden KK, Ozkan ED, Rumbaugh G. Prioritizing the development of mouse models for childhood brain disorders. Neuropharmacology 2015; 100:2-16. [PMID: 26231830 DOI: 10.1016/j.neuropharm.2015.07.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 07/18/2015] [Accepted: 07/22/2015] [Indexed: 12/20/2022]
Abstract
Mutations in hundreds of genes contribute to cognitive and behavioral dysfunction associated with developmental brain disorders (DBDs). Due to the sheer number of risk factors available for study combined with the cost of developing new animal models, it remains an open question how genes should be prioritized for in-depth neurobiological investigations. Recent reviews have argued that priority should be given to frequently mutated genes commonly found in sporadic DBD patients. Intrigued by this idea, we explored to what extent "high priority" risk factors have been studied in animals in an effort to assess their potential for generating valuable preclinical models capable of advancing the neurobiological understanding of DBDs. We found that in-depth whole animal studies are lacking for many high priority genes, with relatively few neurobiological studies performed in construct valid animal models aimed at understanding the pathological substrates associated with disease phenotypes. However, some high priority risk factors have been extensively studied in animal models and they have generated novel insights into DBD patho-neurobiology while also advancing early pre-clinical therapeutic treatment strategies. We suggest that prioritizing model development toward genes frequently mutated in non-specific DBD populations will accelerate the understanding of DBD patho-neurobiology and drive novel therapeutic strategies. This article is part of the Special Issue entitled 'Synaptopathy--from Biology to Therapy'.
Collapse
Affiliation(s)
- Kevin K Ogden
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Emin D Ozkan
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Gavin Rumbaugh
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| |
Collapse
|
14
|
Hester MS, Danzer SC. Hippocampal granule cell pathology in epilepsy - a possible structural basis for comorbidities of epilepsy? Epilepsy Behav 2014; 38:105-16. [PMID: 24468242 PMCID: PMC4110172 DOI: 10.1016/j.yebeh.2013.12.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 12/17/2013] [Accepted: 12/21/2013] [Indexed: 01/31/2023]
Abstract
Temporal lobe epilepsy in both animals and humans is characterized by abnormally integrated hippocampal dentate granule cells. Among other abnormalities, these cells make axonal connections with inappropriate targets, grow dendrites in the wrong direction, and migrate to ectopic locations. These changes promote the formation of recurrent excitatory circuits, leading to the appealing hypothesis that these abnormal cells may by epileptogenic. While this hypothesis has been the subject of intense study, less attention has been paid to the possibility that abnormal granule cells in the epileptic brain may also contribute to comorbidities associated with the disease. Epilepsy is associated with a variety of general findings, such as memory disturbances and cognitive dysfunction, and is often comorbid with a number of other conditions, including schizophrenia and autism. Interestingly, recent studies implicate disruption of common genes and gene pathways in all three diseases. Moreover, while neuropsychiatric conditions are associated with changes in a variety of brain regions, granule cell abnormalities in temporal lobe epilepsy appear to be phenocopies of granule cell deficits produced by genetic mouse models of autism and schizophrenia, suggesting that granule cell dysmorphogenesis may be a common factor uniting these seemingly diverse diseases. Disruption of common signaling pathways regulating granule cell neurogenesis may begin to provide mechanistic insight into the cooccurrence of temporal lobe epilepsy and cognitive and behavioral disorders.
Collapse
Affiliation(s)
- Michael S Hester
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Molecular and Developmental Biology Graduate Program, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Anesthesia, University of Cincinnati, Cincinnati, OH 45267, USA; Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45267, USA; Molecular and Developmental Biology Graduate Program, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
15
|
Cendelin J. From mice to men: lessons from mutant ataxic mice. CEREBELLUM & ATAXIAS 2014; 1:4. [PMID: 26331028 PMCID: PMC4549131 DOI: 10.1186/2053-8871-1-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/21/2014] [Indexed: 01/01/2023]
Abstract
Ataxic mutant mice can be used to represent models of cerebellar degenerative disorders. They serve for investigation of cerebellar function, pathogenesis of degenerative processes as well as of therapeutic approaches. Lurcher, Hot-foot, Purkinje cell degeneration, Nervous, Staggerer, Weaver, Reeler, and Scrambler mouse models and mouse models of SCA1, SCA2, SCA3, SCA6, SCA7, SCA23, DRPLA, Niemann-Pick disease and Friedreich ataxia are reviewed with special regard to cerebellar pathology, pathogenesis, functional changes and possible therapeutic influences, if any. Finally, benefits and limitations of mouse models are discussed.
Collapse
Affiliation(s)
- Jan Cendelin
- Department of Pathophysiology, Faculty of Medicine in Pilsen, Charles University in Prague, Lidicka 1, 301 66 Plzen, Czech Republic ; Biomedical Centre, Faculty of Medicine in Pilsen, Charles University in Prague, Plzen, Czech Republic
| |
Collapse
|
16
|
Miller AR, Hawkins NA, McCollom CE, Kearney JA. Mapping genetic modifiers of survival in a mouse model of Dravet syndrome. GENES BRAIN AND BEHAVIOR 2013; 13:163-72. [PMID: 24152123 DOI: 10.1111/gbb.12099] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 09/06/2013] [Accepted: 10/21/2013] [Indexed: 12/26/2022]
Abstract
Epilepsy is a common neurological disorder affecting approximately 1% of the population. Mutations in voltage-gated sodium channels are responsible for several monogenic epilepsy syndromes. More than 800 mutations in the voltage-gated sodium channel SCN1A have been reported in patients with generalized epilepsy with febrile seizures plus and Dravet syndrome. Heterozygous loss-of-function mutations in SCN1A result in Dravet syndrome, a severe infant-onset epileptic encephalopathy characterized by intractable seizures, developmental delays and increased mortality. A common feature of monogenic epilepsies is variable expressivity among individuals with the same mutation, suggesting that genetic modifiers may influence clinical severity. Mice with heterozygous deletion of Scn1a (Scn1a(+/-) ) model a number of Dravet syndrome features, including spontaneous seizures and premature lethality. Phenotype severity in Scn1a(+/-) mice is strongly dependent on strain background. On the 129S6/SvEvTac strain Scn1a(+/-) mice exhibit no overt phenotype, whereas on the (C57BL/6J × 129S6/SvEvTac)F1 strain Scn1a(+/-) mice exhibit spontaneous seizures and early lethality. To systematically identify loci that influence premature lethality in Scn1a(+/-) mice, we performed genome scans on reciprocal backcrosses. Quantitative trait locus mapping revealed modifier loci on mouse chromosomes 5, 7, 8 and 11. RNA-seq analysis of strain-dependent gene expression, regulation and coding sequence variation provided a list of potential functional candidate genes at each locus. Identification of modifier genes that influence survival in Scn1a(+/-) mice will improve our understanding of the pathophysiology of Dravet syndrome and may suggest novel therapeutic strategies for improved treatment of human patients.
Collapse
Affiliation(s)
- A R Miller
- Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | | | | | | |
Collapse
|
17
|
Mei X, Wu S, Bassuk AG, Slusarski DC. Mechanisms of prickle1a function in zebrafish epilepsy and retinal neurogenesis. Dis Model Mech 2013; 6:679-88. [PMID: 23324328 PMCID: PMC3634651 DOI: 10.1242/dmm.010793] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Epilepsy is a complex neurological disorder characterized by unprovoked seizures. The etiology is heterogeneous with both genetic and environmental causes. Genes that regulate neurotransmitters and ion channels in the central nervous system have been associated with epilepsy. However, a recent screening in human epilepsy patients identified mutations in the PRICKLE1 (PK1) locus, highlighting a potentially novel mechanism underlying seizures. PK1 is a core component of the planar cell polarity network that regulates tissue polarity. Zebrafish studies have shown that Pk1 coordinates cell movement, neuronal migration and axonal outgrowth during embryonic development. Yet how dysfunction of Pk1 relates to epilepsy is unknown. To address the mechanism underlying epileptogenesis, we used zebrafish to characterize Pk1a function and epilepsy-related mutant forms. We show that knockdown of pk1a activity sensitizes zebrafish larva to a convulsant drug. To model defects in the central nervous system, we used the retina and found that pk1a knockdown induces neurite outgrowth defects; yet visual function is maintained. Furthermore, we characterized the functional and biochemical properties of the PK1 mutant forms identified in human patients. Functional analyses demonstrate that the wild-type Pk1a partially suppresses the gene knockdown retinal defects but not the mutant forms. Biochemical analysis reveals increased ubiquitylation of one mutant form and decreased translational efficiency of another mutant form compared with the wild-type Pk1a. Taken together, our results indicate that mutation of human PK1 could lead to defects in neurodevelopment and signal processing, providing insight into seizure predisposition in these patients.
Collapse
Affiliation(s)
- Xue Mei
- Department of Biology, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
18
|
Sebe JY, Bershteyn M, Hirotsune S, Wynshaw-Boris A, Baraban SC. ALLN rescues an in vitro excitatory synaptic transmission deficit in Lis1 mutant mice. J Neurophysiol 2012; 109:429-36. [PMID: 23100132 DOI: 10.1152/jn.00431.2012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
LIS1 gene mutations lead to a rare neurological disorder, classical lissencephaly, characterized by brain malformations, mental retardation, seizures, and premature death. Mice heterozygous for Lis1 (Lis1(+/-)) exhibit cortical malformations, defects in neuronal migration, increased glutamate-mediated synaptic transmission, and spontaneous electrographic seizures. Recent work demonstrated that in utero treatment of Lis1(+/-) mutant dams with ALLN, a calpain inhibitor, partially rescues neuronal migration defects in the offspring. Given the challenges of in utero drug administration, we examined the therapeutic potential of ALLN on postnatal lissencephalic cells. Voltage- and current-clamp studies were performed with acute hippocampal slices obtained from Lis1 mutant mice and age-matched littermate control mice. Specifically, we determined whether postnatal ALLN treatment can reverse excitatory synaptic transmission deficits, namely, an increase in spontaneous and miniature excitatory postsynaptic current (EPSC) frequency, on CA1 pyramidal neurons observed in tissue slices from Lis1(+/-) mice. We found that acute application of ALLN restored spontaneous and miniature EPSC frequencies to wild-type levels without affecting inhibitory postsynaptic synaptic current. Furthermore, Western blot analysis of protein expression, including proteins involved in excitatory synaptic transmission, demonstrated that ALLN blocks the cleavage of the calpain substrate αII-spectrin but does not rescue Lis1 protein levels in Lis1(+/-) mutants.
Collapse
Affiliation(s)
- Joy Y Sebe
- Epilepsy Research Laboratory, Department of Neurological Surgery, University of California, San Francisco, California 94143, USA
| | | | | | | | | |
Collapse
|
19
|
Abstract
Epilepsy is characterized by spontaneous recurrent seizures and comprises a diverse group of syndromes with different etiologies. Epileptogenesis refers to the process whereby the brain becomes epileptic and can be related to several factors, such as acquired structural brain lesions, inborn brain malformations, alterations in neuronal signaling, and defects in maturation and plasticity of neuronal networks. In this review, we will focus on alterations of brain development that lead to an hyperexcitability phenotype in adulthood, providing examples from both animal and human studies. Malformations of cortical development (including focal cortical dysplasia, lissencephaly, heterotopia, and polymicrogyria) are frequently epileptogenic and result from defects in cell proliferation in the germinal zone and/or impaired neuronal migration and differentiation. Delayed or reduced arrival of inhibitory interneurons into the cortical plate is another possible cause of epileptogenesis. GABAergic neurons are generated during early development in the ganglionic eminences, and failure to pursue migration toward the cortex alters the excitatory/inhibitory balance resulting in aberrant network hyperexcitability. More subtle defects in the developmental assembly of excitatory and inhibitory synapses are also involved in epilepsy. For example, mutations in the presynaptic proteins synapsins and SNAP-25 cause derangements of synaptic transmission and plasticity which underlie appearance of an epileptic phenotype. Finally, there is evidence that defects in synapse elimination and remodeling during early "critical periods" can trigger hyperexcitability later in life. Further clarification of the developmental pathways to epilepsy has important implications for disease prevention and therapy.
Collapse
Affiliation(s)
- Yuri Bozzi
- Laboratory of Molecular Neuropathology, Centre for Integrative Biology, University of Trento Trento, Italy
| | | | | |
Collapse
|
20
|
Kowalski J, Geuting M, Paul S, Dieni S, Laurens J, Zhao S, Drakew A, Haas CA, Frotscher M, Vida I. Proper layering is important for precisely timed activation of hippocampal mossy cells. ACTA ACUST UNITED AC 2010; 20:2043-54. [PMID: 20053714 DOI: 10.1093/cercor/bhp267] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The mammalian cortex exhibits a laminated structure that may underlie optimal synaptic connectivity and support temporally precise activation of neurons. In 'reeler' mice, the lack of the extracellular matrix protein Reelin leads to abnormal positioning of cortical neurons and disrupted layering. To address how these structural changes impact neuronal function, we combined electrophysiological and neuroanatomical techniques to investigate the synaptic activation of hippocampal mossy cells (MCs), the cell type that integrates the output of dentate gyrus granule cells (GCs). While somatodendritic domains of wild-type (WT) MCs were confined to the hilus, the somata and dendrites of reeler MCs were often found in the molecular layer, where the perforant path (PP) terminates. Most reeler MCs received aberrant monosynaptic excitatory input from the PP, whereas the disynaptic input to MCs via GCs was decreased and inhibition was increased. In contrast to the uniform disynaptic discharge of WT MCs, many reeler cells discharged with short, monosynaptic latencies, while others fired with long latencies over a broad temporal window in response to PP activation. Thus, disturbed lamination results in aberrant synaptic connectivity and altered timing of action potential generation. These results highlight the importance of a layered cortical structure for information processing.
Collapse
Affiliation(s)
- Janina Kowalski
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, University of Freiburg, D-79104 Freiburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Gardiner J, Marc J. Disruption of normal cytoskeletal dynamics may play a key role in the pathogenesis of epilepsy. Neuroscientist 2009; 16:28-39. [PMID: 19429889 DOI: 10.1177/1073858409334422] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Epilepsy, a common disease affecting 1% to 2% of the population, is characterized by seizures, hyperexcitability at synapses, and aberrant extension of neurons following seizures. Much work has been done on the role of synaptic components in the pathogenesis of epilepsy, but relatively little attention has been given to the potential role of the cytoskeleton. The neuronal cytoskeleton consists of microtubules, actin filaments, intermediate filaments, and associated proteins. A number of mutations in both microtubule-associated proteins (MAPs) and actin-binding proteins, as well as altered expression levels of several cytoskeletal proteins, are known to be involved in epilepsy. These changes will affect the dynamics of the neuronal cytoskeleton and therefore are likely to contribute to the pathogenesis of epilepsy through mechanisms such as increased neurotrophic support to neurons and increased sprouting of mossy fibers. These changes may also contribute to hyperexcitability of neurons through an as yet unidentified mechanism.
Collapse
Affiliation(s)
- John Gardiner
- School of Biological Sciences, The University of Sydney, Camperdown, Australia.
| | | |
Collapse
|
22
|
Increased reelin promoter methylation is associated with granule cell dispersion in human temporal lobe epilepsy. J Neuropathol Exp Neurol 2009; 68:356-64. [PMID: 19287316 DOI: 10.1097/nen.0b013e31819ba737] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Mesial temporal sclerosis (MTS) is the most common lesion in chronic, intractable temporal lobe epilepsies (TLE) and characterized by segmental neuronal cell loss in major hippocampal segments. Another histopathological hallmark includes granule cell dispersion (GCD), an architectural disturbance of the dentate gyrus encountered in approximately 50% of patients with mesial temporal sclerosis. Reelin, which plays a key role during hippocampal development and maintenance of laminar organization, is synthesized and released by Cajal-Retzius cells of the dentate molecular layer, and previous studies have shown that Reelin transcript levels are downregulated in human temporal lobe epilepsies specimens. To investigate whether epigenetic silencing by Reelin promoter methylation may be an underlying pathogenetic mechanism of GCD, DNA was harvested from 3 microdissected hippocampal subregions (i.e. molecular and granule cell layers of the dentate gyrus and presubiculum) from 8 MTS specimens with GCD, 5 TLE samples without GCD, and 3 autopsy controls. Promoter methylation was analyzed after bisulfite treatment, cloning, and direct sequencing; immunohistochemistry was performed to identify Cajal-Retzius cells. Reelin promoter methylation was found to be greater in TLE specimens than in controls; promoter methylation correlated with GCD among TLE specimens (p < 0.0002). No other clinical or histopathological parameter (i.e. sex, age, seizure duration, medication or extent, of MTS) correlated with promoter methylation. These data support a compromised Reelin-signaling pathway and identify promoter methylation as an epigenetic mechanism in the pathogenesis of TLE.
Collapse
|
23
|
Hunt RF, Scheff SW, Smith BN. Posttraumatic epilepsy after controlled cortical impact injury in mice. Exp Neurol 2008; 215:243-52. [PMID: 19013458 DOI: 10.1016/j.expneurol.2008.10.005] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2008] [Revised: 10/06/2008] [Accepted: 10/10/2008] [Indexed: 11/28/2022]
Abstract
Many patients develop temporal lobe epilepsy after trauma, but basic mechanisms underlying the development of chronic seizures after head injury remain poorly understood. Using the controlled cortical impact injury model we examined whether mice developed spontaneous seizures after mild (0.5 mm injury depth) or severe (1.0 mm injury depth) brain injury and how subsequent posttraumatic mossy fiber sprouting was associated with excitability in the dentate gyrus 42-71 d after injury. After several weeks, spontaneous behavioral seizures were observed in 20% of mice with mild and 36% of mice with severe injury. Mossy fiber sprouting was typically present in septal slices of the dentate gyrus ipsilateral to the injury, but not in control mice. In slices with mossy fiber sprouting, perforant path stimulation revealed a significant reduction (P<0.01) in paired-pulse ratios in dentate granule cells at 20 ms and 40 ms interpulse intervals, but not at 80 ms or 160 ms intervals. These slices were also characterized by spontaneous and hilar-evoked epileptiform activity in the dentate gyrus in the presence of Mg(2+)-free ACSF containing 100 microM picrotoxin. In contrast, paired-pulse and hilar-evoked responses in slices from injured animals that did not display mossy fiber sprouting were not different from controls. These data suggest the development of spontaneous posttraumatic seizures as well as structural and functional network changes associated with temporal lobe epilepsy in the mouse dentate gyrus by 71 d after CCI injury. Identifying experimental injury models that exhibit similar pathology to injury-induced epilepsy in humans should help to elucidate the mechanisms by which the injured brain becomes epileptic.
Collapse
Affiliation(s)
- Robert F Hunt
- Department of Physiology, University of Kentucky, MS-508 Chandler Medical Center, 800 Rose St., Lexington, KY 40536-0298, USA
| | | | | |
Collapse
|
24
|
Veronesi MC, Kubek DJ, Kubek MJ. Isoflurane exacerbates electrically evoked seizures in amygdala-kindled rats during recovery. Epilepsy Res 2008; 82:15-20. [PMID: 18674885 DOI: 10.1016/j.eplepsyres.2008.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2007] [Revised: 05/21/2008] [Accepted: 06/18/2008] [Indexed: 11/29/2022]
Abstract
Neuroexcitatory effects of isoflurane during or following anesthesia are controversial, particularly in epileptic patients. In contrast, halothane is generally considered to be highly anticonvulsant. Kindling is an animal model of epilepsy suitable for studying the effects of anesthetic agents on the epileptic brain. Fully kindled, Sprague-Dawley rats were either untreated or received a 5 min exposure to isoflurane or halothane 30 min prior to a seizure and compared to seizures in the absence of prior anesthesia. Afterdischarge duration was assessed via electroencephalographs recorded from electrodes implanted in the basolateral amygdala and behavioral seizure stereotypy (stages I-V) was simultaneously recorded and analyzed using digital video for all seizures. Total seizure duration and clonus duration were significantly (P<0.05) increased 30 min after isoflurane but not halothane exposure relative to pre-treatment control. These results are the first to demonstrate that isoflurane exacerbates electrically evoked secondarily generalized seizures in fully kindled animals during recovery. These results also show that the kindling paradigm is useful for evaluating the mechanism of anesthetic agents that may be proconvulsant in epileptic subjects.
Collapse
Affiliation(s)
- Michael C Veronesi
- Program in Medical Neuroscience, Indiana University School of Medicine, 635 Barnhill Drive, Room MS5022, Indianapolis, IN 46202, United States
| | | | | |
Collapse
|
25
|
Gant JC, Thibault O, Blalock EM, Yang J, Bachstetter A, Kotick J, Schauwecker PE, Hauser KF, Smith GM, Mervis R, Li Y, Barnes GN. Decreased number of interneurons and increased seizures in neuropilin 2 deficient mice: implications for autism and epilepsy. Epilepsia 2008; 50:629-45. [PMID: 18657176 DOI: 10.1111/j.1528-1167.2008.01725.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
PURPOSE Clinically, perturbations in the semaphorin signaling system have been associated with autism and epilepsy. The semaphorins have been implicated in guidance, migration, differentiation, and synaptic plasticity of neurons. The semaphorin 3F (Sema3F) ligand and its receptor, neuropilin 2 (NPN2) are highly expressed within limbic areas. NPN2 signaling may intimately direct the apposition of presynaptic and postsynaptic locations, facilitating the development and maturity of hippocampal synaptic function. To further understand the role of NPN2 signaling in central nevous system (CNS) plasticity, structural and functional alterations were assessed in NPN2 deficient mice. METHODS In NPN2 deficient mice, we measured seizure susceptibility after kainic acid or pentylenetetrazol, neuronal excitability and synaptic throughput in slice preparations, principal and interneuron cell counts with immunocytochemical protocols, synaptosomal protein levels with immunoblots, and dendritic morphology with Golgi-staining. RESULTS NPN2 deficient mice had shorter seizure latencies, increased vulnerability to seizure-related death, were more likely to develop spontaneous recurrent seizure activity after chemical challenge, and had an increased slope on input/output curves. Principal cell counts were unchanged, but GABA, parvalbumin, and neuropeptide Y interneuron cell counts were significantly reduced. Synaptosomal NPN2 protein levels and total number of GABAergic synapses were decreased in a gene dose-dependent fashion. CA1 pyramidal cells showed reduced dendritic length and complexity, as well as an increased number of dendritic spines. DISCUSSION These data suggest the novel hypothesis that the Sema 3F signaling system's role in appropriate placement of subsets of hippocampal interneurons has critical downstream consequences for hippocampal function, resulting in a more seizure susceptible phenotype.
Collapse
Affiliation(s)
- John C Gant
- Departments of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, KY, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Nosten-Bertrand M, Kappeler C, Dinocourt C, Denis C, Germain J, Dinh Tuy FP, Verstraeten S, Alvarez C, Métin C, Chelly J, Giros B, Miles R, Depaulis A, Francis F. Epilepsy in Dcx knockout mice associated with discrete lamination defects and enhanced excitability in the hippocampus. PLoS One 2008; 3:e2473. [PMID: 18575605 PMCID: PMC2429962 DOI: 10.1371/journal.pone.0002473] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2008] [Accepted: 05/08/2008] [Indexed: 12/24/2022] Open
Abstract
Patients with Doublecortin (DCX) mutations have severe cortical malformations associated with mental retardation and epilepsy. Dcx knockout (KO) mice show no major isocortical abnormalities, but have discrete hippocampal defects. We questioned the functional consequences of these defects and report here that Dcx KO mice are hyperactive and exhibit spontaneous convulsive seizures. Changes in neuropeptide Y and calbindin expression, consistent with seizure occurrence, were detected in a large proportion of KO animals, and convulsants, including kainate and pentylenetetrazole, also induced seizures more readily in KO mice. We show that the dysplastic CA3 region in KO hippocampal slices generates sharp wave-like activities and possesses a lower threshold for epileptiform events. Video-EEG monitoring also demonstrated that spontaneous seizures were initiated in the hippocampus. Similarly, seizures in human patients mutated for DCX can show a primary involvement of the temporal lobe. In conclusion, seizures in Dcx KO mice are likely to be due to abnormal synaptic transmission involving heterotopic cells in the hippocampus and these mice may therefore provide a useful model to further study how lamination defects underlie the genesis of epileptiform activities.
Collapse
Affiliation(s)
- Marika Nosten-Bertrand
- INSERM, U513, Université Pierre et Marie Curie, Paris, France
- UMPC Université Paris 06, Neurobiologie et Psychiatrie, Paris, France
| | - Caroline Kappeler
- Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Paris, France
- INSERM, U567, Paris, France
| | | | - Cécile Denis
- INSERM, U513, Université Pierre et Marie Curie, Paris, France
- UMPC Université Paris 06, Neurobiologie et Psychiatrie, Paris, France
| | - Johanne Germain
- INSERM, U513, Université Pierre et Marie Curie, Paris, France
- UMPC Université Paris 06, Neurobiologie et Psychiatrie, Paris, France
| | - Françoise Phan Dinh Tuy
- Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Paris, France
- INSERM, U567, Paris, France
| | - Soraya Verstraeten
- INSERM, U513, Université Pierre et Marie Curie, Paris, France
- UMPC Université Paris 06, Neurobiologie et Psychiatrie, Paris, France
| | - Chantal Alvarez
- UPMC, Paris, France
- INSERM, U839, Institut du Fer à Moulin, Paris, France
| | - Christine Métin
- UPMC, Paris, France
- INSERM, U839, Institut du Fer à Moulin, Paris, France
| | - Jamel Chelly
- Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Paris, France
- INSERM, U567, Paris, France
| | - Bruno Giros
- INSERM, U513, Université Pierre et Marie Curie, Paris, France
- UMPC Université Paris 06, Neurobiologie et Psychiatrie, Paris, France
- Douglas Hospital Research Center, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Richard Miles
- INSERM, U739, UPMC, CHU Pitié Salpêtrière, Paris, France
| | - Antoine Depaulis
- Grenoble Institute of Neurosciences, Inserm U836-UJF-CEA-CHU, Université Joseph Fourier, Grenoble, France
| | - Fiona Francis
- Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Paris, France
- INSERM, U567, Paris, France
- * E-mail:
| |
Collapse
|
27
|
Moroni RF, Inverardi F, Regondi MC, Panzica F, Spreafico R, Frassoni C. Altered spatial distribution of PV-cortical cells and dysmorphic neurons in the somatosensory cortex of BCNU-treated rat model of cortical dysplasia. Epilepsia 2008; 49:872-87. [DOI: 10.1111/j.1528-1167.2007.01440.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
28
|
Patrylo PR, Willingham A. Anatomic and electrophysiologic evidence for a proconvulsive circuit in the dentate gyrus of reeler mutant mice, an animal model of diffuse cortical malformation. Dev Neurosci 2007; 29:73-83. [PMID: 17148950 DOI: 10.1159/000096212] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2006] [Accepted: 03/24/2006] [Indexed: 11/19/2022] Open
Abstract
Although cortical malformations (CMs) are often associated with epilepsy, the underlying mechanisms are unknown. The reeler mouse is a model of CM with enhanced susceptibility to epileptiform activity, including the in vitro dentate gyrus, a region normally resistant to seizures. In this study, field potential recordings in hippocampal slices and the Timm stain were used to examine mossy fiber distribution in the dentate gyrus. In artificial cerebrospinal fluid containing bicuculline, 100% of reeler slices and 0% of control slices had spontaneous and antidromic evoked prolonged negative field potential shifts that were blocked by glutamate receptor antagonists. Sections from reeler mice, but not controls, exhibited a dark band of Timm's stain at the molecular layer/granule cell layer border. These data reveal that mossy fiber distribution is altered in reeler mice and coincides with the presence of an abnormal proconvulsive glutamatergic circuit.
Collapse
Affiliation(s)
- Peter R Patrylo
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA.
| | | |
Collapse
|
29
|
Scharfman H, Goodman J, McCloskey D. Ectopic granule cells of the rat dentate gyrus. Dev Neurosci 2007; 29:14-27. [PMID: 17148946 PMCID: PMC1934347 DOI: 10.1159/000096208] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2006] [Accepted: 04/05/2006] [Indexed: 01/19/2023] Open
Abstract
Granule cells of the mammalian dentate gyrus normally form a discrete layer, and virtually all granule cells migrate to this location. Exceptional granule cells that are positioned incorrectly, in 'ectopic' locations, are rare. Although the characteristics of such ectopic granule cells appear similar in many respects to granule cells located in the granule cell layer, their rare occurrence has limited a full evaluation of their structure and function. More information about ectopic granule cells has been obtained by studying those that develop after experimental manipulations that increase their number. For example, after severe seizures, the number of ectopic granule cells located in the hilus increases dramatically. These experimentally-induced ectopic granule cells may not be equivalent to normal ectopic granule cells necessarily, but the vastly increased numbers have allowed much more information to be obtained. Remarkably, the granule cells that are positioned ectopically develop intrinsic properties and an axonal projection that are similar to granule cells that are located normally, i.e., in the granule cell layer. However, dendritic structure and synaptic structure/function appear to differ. These studies have provided new insight into a rare type of granule cell in the dentate gyrus, and the plastic characteristics of dentate granule cells that appear to depend on the location of the cell body.
Collapse
Affiliation(s)
- Helen Scharfman
- Department of Pharmacology, Columbia University, New York, NY, USA.
| | | | | |
Collapse
|
30
|
Marrone MC, Marinelli S, Biamonte F, Keller F, Sgobio CA, Ammassari-Teule M, Bernardi G, Mercuri NB. Altered cortico-striatal synaptic plasticity and related behavioural impairments in reeler mice. Eur J Neurosci 2006; 24:2061-70. [PMID: 17067303 DOI: 10.1111/j.1460-9568.2006.05083.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Reelin-deficient mice have been used to investigate the role of this extracellular protein in cortico-striatal plasticity and striatum-related behaviours. Here we show that a repetitive electrical stimulation of the cortico-striatal pathway elicited long-term potentiation (LTP) in homozygous reeler (rl/rl) mice, while causing long-term depression in their wild-type (+/+) littermates. The N-methyl-D-aspartic acid (NMDA) receptor antagonist D-(-)-2 amino-5-phosphonopentanoic acid prevented the induction of LTP in (rl/rl) mice, thus confirming that this form of synaptic plasticity was NMDA receptor-dependent. Interestingly, in the presence of tiagabine, a blocker of gamma-aminobutyric acid (GABA) re-uptake system, the probability that (rl/rl) mice showed LTP decreased significantly, thus suggesting an impaired GABAergic transmission in reeler mutants. Consistent with this view, a decreased density of parvalbumin-positive GABAergic striatal interneurons was found in (rl/rl) mice in comparison to (+/+) mice. Finally, compatible with their abnormal striatal function (rl/rl) mice exhibited procedural learning deficits. Our data, showing alterations in cortico-striatal plasticity largely depending on a depressed GABAergic tone, delineate a mechanism whereby the lack of reelin may affect cognitive functions.
Collapse
|
31
|
Kopjas NN, Jones RT, Bany B, Patrylo PR. Reeler mutant mice exhibit seizures during recovery from isoflurane-induced anesthesia. Epilepsy Res 2006; 69:87-91. [PMID: 16466907 DOI: 10.1016/j.eplepsyres.2005.12.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2005] [Revised: 12/13/2005] [Accepted: 12/13/2005] [Indexed: 11/28/2022]
Abstract
Reeler mice are a model of cortical malformation with enhanced seizure susceptibility. Data suggest that the propensity to anesthesia-induced seizures may be enhanced in animal models with developmental anomalies. Consequently, reeler mice were monitored behaviorally before, during and after isoflurane anesthesia. During recovery, 12% of reeler homozygotes had class I/II seizures while the remaining 88% exhibited convulsive seizures entailing opisthotonus and forepaw drumming. Similar behavior was not observed in controls. These data reveal that reeler mice display isoflurane-induced seizures and provide support for the hypothesis that developmental anomalies may predispose the central nervous system to anesthesia-induced seizures.
Collapse
Affiliation(s)
- Nicholas N Kopjas
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA
| | | | | | | |
Collapse
|