1
|
Eberhard J, Henning L, Fülle L, Knöpper K, Böhringer J, Graelmann FJ, Hänschke L, Kenzler J, Brosseron F, Heneka MT, Domingos AI, Eyerich S, Lochner M, Weighardt H, Bedner P, Steinhäuser C, Förster I. Ablation of CCL17-positive hippocampal neurons induces inflammation-dependent epilepsy. Epilepsia 2024. [PMID: 39607395 DOI: 10.1111/epi.18200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024]
Abstract
OBJECTIVE Neuronal cell death and neuroinflammation are characteristic features of epilepsy, but it remains unclear whether neuronal cell death as such is causative for the development of epileptic seizures. To test this hypothesis, we established a novel mouse line permitting inducible ablation of pyramidal neurons by inserting simian diphtheria toxin (DT) receptor (DTR) cDNA into the Ccl17 locus. The chemokine CCL17 is expressed in pyramidal CA1 neurons in adult mice controlling microglial quiescence. METHODS Seizure activity in CCL17-DTR mice was analyzed by electroencephalographic recordings following treatment with DT for 3 consecutive days. Neuroinflammation and neuronal cell death were evaluated by (immuno)histochemistry. Pharmacological inhibition of TNFR1 signaling was achieved by treatment with XPro1595, a dominant-negative inhibitor of soluble tumor necrosis factor. RESULTS Neuronal cell death was detectable 7 days (d7) after the first DT injection in heterozygous CCL17-DTR mice. Spontaneous epileptic seizures were observed in the vast majority of mice, often with an initial peak at d6-9, followed by a period of reduced activity and a gradual increase during the 1-month observation period. Microglial reactivity was overt from d5 after DT administration not only in the CA1 region but also in the CA2/CA3 area, shortly followed by astrogliosis. Reactive microgliosis and astrogliosis persisted until d30 and, together with neuronal loss and stratum radiatum shrinkage, reflected important features of human hippocampal sclerosis. Granule cell dispersion was detectable only 3 months after DT treatment. Application of XPro1595 significantly reduced chronic seizure burden without affecting the development of hippocampal sclerosis. SIGNIFICANCE In conclusion, our data demonstrate that sterile pyramidal neuronal death is sufficient to cause epilepsy in the absence of other pathological processes. The CCL17-DTR mouse line may thus be a valuable model for further mechanistic studies on epilepsy and assessment of antiseizure medication.
Collapse
Affiliation(s)
- Judith Eberhard
- Immunology & Environment, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Lukas Henning
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
- Deutsche Forschungsgemeinschaft, Bonn, Germany
| | - Lorenz Fülle
- Immunology & Environment, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
- Business Development Europe Research Services, WuXi Biologics, Leverkusen, Germany
| | - Konrad Knöpper
- Immunology & Environment, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California, USA
| | - Jana Böhringer
- Immunology & Environment, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Frederike J Graelmann
- Immunology & Environment, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Lea Hänschke
- Molecular Developmental Biology, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Julia Kenzler
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
- Sanofi-Aventis Deutschland, Medical Operations General Medicines in Germany, Switzerland, Austria (GSA), Berlin, Germany
| | | | - Michael T Heneka
- German Center for Neurodegenerative Diseases, Bonn, Germany
- Luxembourg Center for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Ana I Domingos
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, UK
| | - Stefanie Eyerich
- Institute for Medical Microbiology, Immunology, and Hygiene, Technical University of Munich, Munich, Germany
- ZAUM-Center of Allergy and Environment, Technical University and Helmholtz Center Munich, Munich, Germany
| | - Matthias Lochner
- Institute for Medical Microbiology, Immunology, and Hygiene, Technical University of Munich, Munich, Germany
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Heike Weighardt
- Immunology & Environment, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Peter Bedner
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Irmgard Förster
- Immunology & Environment, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
2
|
Pelz L, Dossou L, Kompier N, Jüttner R, Siemonsmeier G, Meyer N, Lowenstein ED, Lahmann I, Kettenmann H, Birchmeier C, Rathjen FG. The IgCAM BT-IgSF (IgSF11) is essential for connexin43-mediated astrocyte-astrocyte coupling in mice. eNeuro 2024; 11:ENEURO.0283-23.2024. [PMID: 38388443 PMCID: PMC10957231 DOI: 10.1523/eneuro.0283-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
The type I transmembrane protein BT-IgSF is predominantly localized in the brain and testes. It belongs to the CAR subgroup of Ig cell adhesion proteins, that are hypothesized to regulate connexin expression or localization. Here, we studied the putative link between BT-IgSF and connexins in astrocytes, ependymal cells and neurons of the mouse. Global knockout of BT-IgSF caused an increase in the clustering of connexin43 (Gja1), but not of connexin30 (Gjb6), on astrocytes and ependymal cells. Additionally, knockout animals displayed reduced expression levels of connexin43 protein in the cortex and hippocampus. Importantly, analysis of biocytin spread in hippocampal or cortical slices from mature mice of either sex revealed a decrease in astrocytic cell-cell coupling in the absence of BT-IgSF. Blocking either protein biosynthesis or proteolysis showed that the lysosomal pathway increased connexin43 degradation in astrocytes. Localization of connexin43 in subcellular compartments was not impaired in astrocytes of BT-IgSF mutants. In contrast to connexin43 the localization and expression of connexin36 (Gjd2) on neurons was not affected by the absence of BT-IgSF. Overall, our data indicate that the IgCAM BT-IgSF is essential for correct gap junction-mediated astrocyte-to-astrocyte cell communication.Significance Statement Astrocytes regulate a variety of physiological processes in the developing and adult brain that are essential for proper brain function. Astrocytes form extensive networks in the brain and communicate via gap junctions. Disruptions of gap junction coupling are found in several diseases such as neurodegeneration or epilepsy. Here, we demonstrate that the cell adhesion protein BT-IgSF is essential for gap junction mediated coupling between astrocytes in the cortex and hippocampus.
Collapse
Affiliation(s)
- Laura Pelz
- Max-Delbrück-Center for Molecular Medicine, Berlin DE-13092, Germany
| | - Laura Dossou
- Max-Delbrück-Center for Molecular Medicine, Berlin DE-13092, Germany
| | - Nine Kompier
- Max-Delbrück-Center for Molecular Medicine, Berlin DE-13092, Germany
| | - René Jüttner
- Max-Delbrück-Center for Molecular Medicine, Berlin DE-13092, Germany
| | | | - Niklas Meyer
- Max-Delbrück-Center for Molecular Medicine, Berlin DE-13092, Germany
| | | | - Ines Lahmann
- Max-Delbrück-Center for Molecular Medicine, Berlin DE-13092, Germany
| | - Helmut Kettenmann
- Max-Delbrück-Center for Molecular Medicine, Berlin DE-13092, Germany
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Carmen Birchmeier
- Max-Delbrück-Center for Molecular Medicine, Berlin DE-13092, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Fritz G. Rathjen
- Max-Delbrück-Center for Molecular Medicine, Berlin DE-13092, Germany
| |
Collapse
|
3
|
Bedner P, Steinhäuser C. Role of Impaired Astrocyte Gap Junction Coupling in Epileptogenesis. Cells 2023; 12:1669. [PMID: 37371139 DOI: 10.3390/cells12121669] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/25/2023] [Accepted: 06/19/2023] [Indexed: 06/29/2023] Open
Abstract
The gap-junction-coupled astroglial network plays a central role in the regulation of neuronal activity and synchronisation, but its involvement in the pathogenesis of neuronal diseases is not yet understood. Here, we present the current state of knowledge about the impact of impaired glial coupling in the development and progression of epilepsy and discuss whether astrocytes represent alternative therapeutic targets. We focus mainly on temporal lobe epilepsy (TLE), which is the most common form of epilepsy in adults and is characterised by high therapy resistance. Functional data from TLE patients and corresponding experimental models point to a complete loss of astrocytic coupling, but preservation of the gap junction forming proteins connexin43 and connexin30 in hippocampal sclerosis. Several studies further indicate that astrocyte uncoupling is a causal event in the initiation of TLE, as it occurs very early in epileptogenesis, clearly preceding dysfunctional changes in neurons. However, more research is needed to fully understand the role of gap junction channels in epilepsy and to develop safe and effective therapeutic strategies targeting astrocytes.
Collapse
Affiliation(s)
- Peter Bedner
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| |
Collapse
|
4
|
Pathak D, Sriram K. Neuron-astrocyte omnidirectional signaling in neurological health and disease. Front Mol Neurosci 2023; 16:1169320. [PMID: 37363320 PMCID: PMC10286832 DOI: 10.3389/fnmol.2023.1169320] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/09/2023] [Indexed: 06/28/2023] Open
Abstract
Astrocytes are an abundantly distributed population of glial cells in the central nervous system (CNS) that perform myriad functions in the normal and injured/diseased brain. Astrocytes exhibit heterogeneous phenotypes in response to various insults, a process known as astrocyte reactivity. The accuracy and precision of brain signaling are primarily based on interactions involving neurons, astrocytes, oligodendrocytes, microglia, pericytes, and dendritic cells within the CNS. Astrocytes have emerged as a critical entity within the brain because of their unique role in recycling neurotransmitters, actively modulating the ionic environment, regulating cholesterol and sphingolipid metabolism, and influencing cellular crosstalk in diverse neural injury conditions and neurodegenerative disorders. However, little is known about how an astrocyte functions in synapse formation, axon specification, neuroplasticity, neural homeostasis, neural network activity following dynamic surveillance, and CNS structure in neurological diseases. Interestingly, the tripartite synapse hypothesis came to light to fill some knowledge gaps that constitute an interaction of a subpopulation of astrocytes, neurons, and synapses. This review highlights astrocytes' role in health and neurological/neurodegenerative diseases arising from the omnidirectional signaling between astrocytes and neurons at the tripartite synapse. The review also recapitulates the disruption of the tripartite synapse with a focus on perturbations of the homeostatic astrocytic function as a key driver to modulate the molecular and physiological processes toward neurodegenerative diseases.
Collapse
|
5
|
Henning L, Unichenko P, Bedner P, Steinhäuser C, Henneberger C. Overview Article Astrocytes as Initiators of Epilepsy. Neurochem Res 2023; 48:1091-1099. [PMID: 36244037 PMCID: PMC10030460 DOI: 10.1007/s11064-022-03773-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 08/22/2022] [Accepted: 09/27/2022] [Indexed: 10/17/2022]
Abstract
Astrocytes play a dual role in the brain. On the one hand, they are active signaling partners of neurons and can for instance control synaptic transmission and its plasticity. On the other hand, they fulfill various homeostatic functions such as clearance of glutamate and K+ released from neurons. The latter is for instance important for limiting neuronal excitability. Therefore, an impairment or failure of glutamate and K+ clearance will lead to increased neuronal excitability, which could trigger or aggravate brain diseases such as epilepsy, in which neuronal hyperexcitability plays a role. Experimental data indicate that astrocytes could have such a causal role in epilepsy, but the role of astrocytes as initiators of epilepsy and the relevant mechanisms are under debate. In this overview, we will discuss the potential mechanisms with focus on K+ clearance, glutamate uptake and homoeostasis and related mechanisms, and the evidence for their causative role in epilepsy.
Collapse
Affiliation(s)
- Lukas Henning
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127, Bonn, Germany
| | - Petr Unichenko
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127, Bonn, Germany
| | - Peter Bedner
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127, Bonn, Germany
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127, Bonn, Germany.
| | - Christian Henneberger
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127, Bonn, Germany.
- German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany.
| |
Collapse
|
6
|
Khan D, Bedner P, Müller J, Lülsberg F, Henning L, Prinz M, Steinhäuser C, Muhammad S. TGF-β Activated Kinase 1 (TAK1) Is Activated in Microglia After Experimental Epilepsy and Contributes to Epileptogenesis. Mol Neurobiol 2023; 60:3413-3422. [PMID: 36862288 PMCID: PMC10122619 DOI: 10.1007/s12035-023-03290-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/19/2023] [Indexed: 03/03/2023]
Abstract
Increasing evidence suggests that inflammation promotes epileptogenesis. TAK1 is a central enzyme in the upstream pathway of NF-κB and is known to play a central role in promoting neuroinflammation in neurodegenerative diseases. Here, we investigated the cellular role of TAK1 in experimental epilepsy. C57Bl6 and transgenic mice with inducible and microglia-specific deletion of Tak1 (Cx3cr1CreER:Tak1fl/fl) were subjected to the unilateral intracortical kainate mouse model of temporal lobe epilepsy (TLE). Immunohistochemical staining was performed to quantify different cell populations. The epileptic activity was monitored by continuous telemetric electroencephalogram (EEG) recordings over a period of 4 weeks. The results show that TAK1 was activated predominantly in microglia at an early stage of kainate-induced epileptogenesis. Tak1 deletion in microglia resulted in reduced hippocampal reactive microgliosis and a significant decrease in chronic epileptic activity. Overall, our data suggest that TAK1-dependent microglial activation contributes to the pathogenesis of chronic epilepsy.
Collapse
Affiliation(s)
- Dilaware Khan
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany.,Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Peter Bedner
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Julia Müller
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Fabienne Lülsberg
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Lukas Henning
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Sajjad Muhammad
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany. .,Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
7
|
Henning L, Antony H, Breuer A, Müller J, Seifert G, Audinat E, Singh P, Brosseron F, Heneka MT, Steinhäuser C, Bedner P. Reactive microglia are the major source of tumor necrosis factor alpha and contribute to astrocyte dysfunction and acute seizures in experimental temporal lobe epilepsy. Glia 2023; 71:168-186. [PMID: 36373840 DOI: 10.1002/glia.24265] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/12/2022] [Accepted: 08/17/2022] [Indexed: 11/09/2022]
Abstract
Extensive microglia reactivity has been well described in human and experimental temporal lobe epilepsy (TLE). To date, however, it is not clear whether and based on which molecular mechanisms microglia contribute to the development and progression of focal epilepsy. Astroglial gap junction coupled networks play an important role in regulating neuronal activity and loss of interastrocytic coupling causally contributes to TLE. Here, we show in the unilateral intracortical kainate (KA) mouse model of TLE that reactive microglia are primary producers of tumor necrosis factor (TNF)α and contribute to astrocyte dysfunction and severity of status epilepticus (SE). Immunohistochemical analyses revealed pronounced and persistent microglia reactivity, which already started 4 h after KA-induced SE. Partial depletion of microglia using a colony stimulating factor 1 receptor inhibitor prevented early astrocyte uncoupling and attenuated the severity of SE, but increased the mortality of epileptic mice following surgery. Using microglia-specific inducible TNFα knockout mice we identified microglia as the major source of TNFα during early epileptogenesis. Importantly, microglia-specific TNFα knockout prevented SE-induced gap junction uncoupling in astrocytes. Continuous telemetric EEG recordings revealed that during the first 4 weeks after SE induction, microglial TNFα did not significantly contribute to spontaneous generalized seizure activity. Moreover, the absence of microglial TNFα did not affect the development of hippocampal sclerosis but attenuated gliosis. Taken together, these data implicate reactive microglia in astrocyte dysfunction and network hyperexcitability after an epileptogenic insult.
Collapse
Affiliation(s)
- Lukas Henning
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Henrike Antony
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Annika Breuer
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Julia Müller
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Gerald Seifert
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Etienne Audinat
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | | | | | | | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Peter Bedner
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
8
|
Shao H, Wu W, Wang P, Han T, Zhuang C. Role of Necroptosis in Central Nervous System Diseases. ACS Chem Neurosci 2022; 13:3213-3229. [PMID: 36373337 DOI: 10.1021/acschemneuro.2c00405] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Necroptosis is a type of precisely regulated necrotic cell death activated in caspase-deficient conditions. Multiple factors initiate the necroptotic signaling pathway, including toll-like receptor 3/4, tumor necrosis factor (TNF), dsRNA viruses, and T cell receptors. Presently, TNF-induced necroptosis via the phosphorylation of three key proteins, receptor-interacting protein kinase 1, receptor-interacting protein kinase 3, and mixed lineage kinase domain-like protein, is the best-characterized process. Necroptosis induced by Z-DNA-binding protein 1 (ZBP-1) and toll/interleukin-1 receptor (TIR)-domain-containing adapter-inducing interferon (TRIF) plays a significant role in infectious diseases, such as influenza A virus, Zika virus, and herpesvirus infection. An increasing number of studies have demonstrated the close association of necroptosis with multiple diseases, and disrupting necroptosis has been confirmed to be effective for treating (or managing) these diseases. The central nervous system (CNS) exhibits unique physiological structures and immune characteristics. Necroptosis may occur without the sequential activation of signal proteins, and the necroptosis of supporting cells has more important implications in disease development. Additionally, necroptotic signals can be activated in the absence of necroptosis. Here, we summarize the role of necroptosis and its signal proteins in CNS diseases and characterize typical necroptosis regulators to provide a basis for the further development of therapeutic strategies for treating such diseases. In the present review, relevant information has been consolidated from recent studies (from 2010 until the present), excluding the patents in this field.
Collapse
Affiliation(s)
- Hongming Shao
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Wenbin Wu
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Pei Wang
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Ting Han
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Chunlin Zhuang
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China.,School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|
9
|
Vezzani A, Ravizza T, Bedner P, Aronica E, Steinhäuser C, Boison D. Astrocytes in the initiation and progression of epilepsy. Nat Rev Neurol 2022; 18:707-722. [PMID: 36280704 PMCID: PMC10368155 DOI: 10.1038/s41582-022-00727-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2022] [Indexed: 11/09/2022]
Abstract
Epilepsy affects ~65 million people worldwide. First-line treatment options include >20 antiseizure medications, but seizure control is not achieved in approximately one-third of patients. Antiseizure medications act primarily on neurons and can provide symptomatic control of seizures, but do not alter the onset and progression of epilepsy and can cause serious adverse effects. Therefore, medications with new cellular and molecular targets and mechanisms of action are needed. Accumulating evidence indicates that astrocytes are crucial to the pathophysiological mechanisms of epilepsy, raising the possibility that these cells could be novel therapeutic targets. In this Review, we discuss how dysregulation of key astrocyte functions - gliotransmission, cell metabolism and immune function - contribute to the development and progression of hyperexcitability in epilepsy. We consider strategies to mitigate astrocyte dysfunction in each of these areas, and provide an overview of how astrocyte activation states can be monitored in vivo not only to assess their contribution to disease but also to identify markers of disease processes and treatment effects. Improved understanding of the roles of astrocytes in epilepsy has the potential to lead to novel therapies to prevent the initiation and progression of epilepsy.
Collapse
Affiliation(s)
- Annamaria Vezzani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy.
| | - Teresa Ravizza
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Peter Bedner
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam, Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, Netherlands
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
10
|
Baracaldo-Santamaría D, Corrales-Hernández MG, Ortiz-Vergara MC, Cormane-Alfaro V, Luque-Bernal RM, Calderon-Ospina CA, Cediel-Becerra JF. Connexins and Pannexins: Important Players in Neurodevelopment, Neurological Diseases, and Potential Therapeutics. Biomedicines 2022; 10:2237. [PMID: 36140338 PMCID: PMC9496069 DOI: 10.3390/biomedicines10092237] [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: 06/30/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Cell-to-cell communication is essential for proper embryonic development and its dysfunction may lead to disease. Recent research has drawn attention to a new group of molecules called connexins (Cxs) and pannexins (Panxs). Cxs have been described for more than forty years as pivotal regulators of embryogenesis; however, the exact mechanism by which they provide this regulation has not been clearly elucidated. Consequently, Cxs and Panxs have been linked to congenital neurodegenerative diseases such as Charcot-Marie-Tooth disease and, more recently, chronic hemichannel opening has been associated with adult neurodegenerative diseases (e.g., Alzheimer's disease). Cell-to-cell communication via gap junctions formed by hexameric assemblies of Cxs, known as connexons, is believed to be a crucial component in developmental regulation. As for Panxs, despite being topologically similar to Cxs, they predominantly seem to form channels connecting the cytoplasm to the extracellular space and, despite recent research into Panx1 (Pannexin 1) expression in different regions of the brain during the embryonic phase, it has been studied to a lesser degree. When it comes to the nervous system, Cxs and Panxs play an important role in early stages of neuronal development with a wide span of action ranging from cellular migration during early stages to neuronal differentiation and system circuitry formation. In this review, we describe the most recent available evidence regarding the molecular and structural aspects of Cx and Panx channels, their role in neurodevelopment, congenital and adult neurological diseases, and finally propose how pharmacological modulation of these channels could modify the pathogenesis of some diseases.
Collapse
Affiliation(s)
- Daniela Baracaldo-Santamaría
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - María Gabriela Corrales-Hernández
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Maria Camila Ortiz-Vergara
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Valeria Cormane-Alfaro
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Ricardo-Miguel Luque-Bernal
- Anatomy and Embriology Units, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Carlos-Alberto Calderon-Ospina
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
- GENIUROS Research Group, Center for Research in Genetics and Genomics (CIGGUR), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Juan-Fernando Cediel-Becerra
- Histology and Embryology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| |
Collapse
|
11
|
Berger TC, Taubøll E, Heuser K. The potential role of DNA methylation as preventive treatment target of epileptogenesis. Front Cell Neurosci 2022; 16:931356. [PMID: 35936496 PMCID: PMC9353008 DOI: 10.3389/fncel.2022.931356] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/27/2022] [Indexed: 11/23/2022] Open
Abstract
Pharmacological therapy of epilepsy has so far been limited to symptomatic treatment aimed at neuronal targets, with the result of an unchanged high proportion of patients lacking seizure control. The dissection of the intricate pathological mechanisms that transform normal brain matter to a focus for epileptic seizures—the process of epileptogenesis—could yield targets for novel treatment strategies preventing the development or progression of epilepsy. While many pathological features of epileptogenesis have been identified, obvious shortcomings in drug development are now believed to be based on the lack of knowledge of molecular upstream mechanisms, such as DNA methylation (DNAm), and as well as a failure to recognize glial cell involvement in epileptogenesis. This article highlights the potential role of DNAm and related gene expression (GE) as a treatment target in epileptogenesis.
Collapse
Affiliation(s)
- Toni Christoph Berger
- Department of Neurology, Oslo University Hospital, Oslo, Norway
- *Correspondence: Toni Christoph Berger
| | - Erik Taubøll
- Department of Neurology, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kjell Heuser
- Department of Neurology, Oslo University Hospital, Oslo, Norway
- Kjell Heuser
| |
Collapse
|
12
|
Chen KN, Guan QW, Yin XX, Wang ZJ, Zhou HH, Mao XY. Ferrostatin-1 obviates seizures and associated cognitive deficits in ferric chloride-induced posttraumatic epilepsy via suppressing ferroptosis. Free Radic Biol Med 2022; 179:109-118. [PMID: 34952157 DOI: 10.1016/j.freeradbiomed.2021.12.268] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/28/2021] [Accepted: 12/16/2021] [Indexed: 12/16/2022]
Abstract
Posttraumatic epilepsy (PTE) is a prevalent complication of brain trauma. Current anti-epileptic drugs available do not have satisfactory response to PTE. It is of desperate need to explore novel therapeutic approaches for curing PTE. Our prior work revealed that ferroptosis, a recently discovered mode of cell death, occurs in rodent model of PTE. In the present study, we aimed to further investigate the effect of ferrostatin-1 (Fer-1), a specific ferroptosis inhibitor, on seizure behavior and cognitive deficit in a mouse model of PTE. The preparation of PTE was performed by stereotaxical injection in the somatosensory cortex region of 50 mM FeCl3. Seizure activity was assessed via Racine scoring and electroencephalogram analysis. PTE-related cognitive function was evaluated by novel object recognition and Morris water maze tests. Ferroptosis-related indices including glutathione peroxidase (GPx) activity and protein expressions of 4-hydroxynonenal (4-HNE) were detected using a commercial kit and immunofluorescence, respectively. It was found that treatment with Fer-1 significantly exerted protective effects against acute seizure and memory decline, although no evident effect on epileptic progression. Fer-1 also exhibited good tolerability and safety as we observed that it hardly influenced the body weight. Furthermore, it was noted that administration of Fer-1 suppressed ferroptosis-related indices including GPx activity and protein expressions of 4-HNE in hippocampus. These data altogether indicate that Fer-1 has potent therapeutic effects against seizures and cognitive impairment following PTE-induced brain insult. Fer-1 may act as a promising drug for curing PTE patients.
Collapse
Affiliation(s)
- Kang-Ni Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, PR China
| | - Qi-Wen Guan
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, PR China
| | - Xi-Xi Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Zhao-Jun Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, PR China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, PR China
| | - Xiao-Yuan Mao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, PR China.
| |
Collapse
|
13
|
Wagnon JL. Back to Basics: A Role for Astrocyte Alkalization in Epileptogenesis. Epilepsy Curr 2021; 21:298-299. [PMID: 34690573 PMCID: PMC8512908 DOI: 10.1177/15357597211018687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
[Box: see text]
Collapse
|
14
|
Binder DK, Steinhäuser C. Astrocytes and Epilepsy. Neurochem Res 2021; 46:2687-2695. [PMID: 33661442 DOI: 10.1007/s11064-021-03236-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/12/2022]
Abstract
Changes in astrocyte channels, transporters, and metabolism play a critical role in seizure generation and epilepsy. In particular, alterations in astrocyte potassium, glutamate, water and adenosine homeostasis and gap junctional coupling have all been associated with hyperexcitability and epileptogenesis (largely in temporal lobe epilepsy). Distinct astrocytic changes have also been identified in other types of epilepsy, such as tuberous sclerosis, tumor-associated epilepsy and post-traumatic epilepsy. Together, the emerging literature on astrocytes and epilepsy provides powerful rationale for distinct new therapeutic targets that are astrocyte-specific.
Collapse
Affiliation(s)
- Devin K Binder
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA.
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
15
|
Schweigmann M, Caudal LC, Stopper G, Scheller A, Koch KP, Kirchhoff F. Versatile Surface Electrodes for Combined Electrophysiology and Two-Photon Imaging of the Mouse Central Nervous System. Front Cell Neurosci 2021; 15:720675. [PMID: 34447299 PMCID: PMC8383317 DOI: 10.3389/fncel.2021.720675] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/19/2021] [Indexed: 11/23/2022] Open
Abstract
Understanding and modulating CNS function in physiological as well as pathophysiological contexts remains a significant ambition in research and clinical applications. The investigation of the multifaceted CNS cell types including their interactions and contributions to neural function requires a combination of the state-of-the-art in vivo electrophysiology and imaging techniques. We developed a novel type of liquid crystal polymer (LCP) surface micro-electrode manufactured in three customized designs with up to 16 channels for recording and stimulation of brain activity. All designs include spare central spaces for simultaneous 2P-imaging. Nanoporous platinum-plated contact sites ensure a low impedance and high current transfer. The epidural implantation of the LCP micro-electrodes could be combined with standard cranial window surgery. The epidurally positioned electrodes did not only display long-term biocompatibility, but we also observed an additional stabilization of the underlying CNS tissue. We demonstrate the electrode’s versatility in combination with in vivo 2P-imaging by monitoring anesthesia-awake cycles of transgenic mice with GCaMP3 expression in neurons or astrocytes. Cortical stimulation and simultaneous 2P Ca2+ imaging in neurons or astrocytes highlighted the astrocytes’ integrative character in neuronal activity processing. Furthermore, we confirmed that spontaneous astroglial Ca2+ signals are dampened under anesthesia, while evoked signals in neurons and astrocytes showed stronger dependency on stimulation intensity rather than on various levels of anesthesia. Finally, we show that the electrodes provide recordings of the electrocorticogram (ECoG) with a high signal-to noise ratio and spatial signal differences which help to decipher brain activity states during experimental procedures. Summarizing, the novel LCP surface micro-electrode is a versatile, convenient, and reliable tool to investigate brain function in vivo.
Collapse
Affiliation(s)
- Michael Schweigmann
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany.,Department of Electrical Engineering, Trier University of Applied Sciences, Trier, Germany
| | - Laura C Caudal
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Gebhard Stopper
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Anja Scheller
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Klaus P Koch
- Department of Electrical Engineering, Trier University of Applied Sciences, Trier, Germany
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| |
Collapse
|
16
|
Heuser K, Enger R. Astrocytic Ca 2+ Signaling in Epilepsy. Front Cell Neurosci 2021; 15:695380. [PMID: 34335188 PMCID: PMC8320018 DOI: 10.3389/fncel.2021.695380] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/16/2021] [Indexed: 01/10/2023] Open
Abstract
Epilepsy is one of the most common neurological disorders – estimated to affect at least 65 million worldwide. Most of the epilepsy research has so far focused on how to dampen neuronal discharges and to explain how changes in intrinsic neuronal activity or network function cause seizures. As a result, pharmacological therapy has largely been limited to symptomatic treatment targeted at neurons. Given the expanding spectrum of functions ascribed to the non-neuronal constituents of the brain, in both physiological brain function and in brain disorders, it is natural to closely consider the roles of astrocytes in epilepsy. It is now widely accepted that astrocytes are key controllers of the composition of the extracellular fluids, and may directly interact with neurons by releasing gliotransmitters. A central tenet is that astrocytic intracellular Ca2+ signals promote release of such signaling substances, either through synaptic or non-synaptic mechanisms. Accruing evidence suggests that astrocytic Ca2+ signals play important roles in both seizures and epilepsy, and this review aims to highlight the current knowledge of the roles of this central astrocytic signaling mechanism in ictogenesis and epileptogenesis.
Collapse
Affiliation(s)
- Kjell Heuser
- Department of Neurology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Rune Enger
- Letten Centre and GliaLab, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
17
|
Ismail FS, Faustmann PM. Experimental investigations of antiepileptic drugs in astrocytes-microglia co-cultures suggest possible protective effects on astrocytes during early epileptogenesis. Epilepsia 2021; 62:2297-2298. [PMID: 34169508 DOI: 10.1111/epi.16968] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 01/15/2023]
Affiliation(s)
- Fatme Seval Ismail
- Department of Neurology, University Hospital Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany
| | - Pedro M Faustmann
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
18
|
Henning L, Steinhäuser C, Bedner P. Initiation of Experimental Temporal Lobe Epilepsy by Early Astrocyte Uncoupling Is Independent of TGFβR1/ALK5 Signaling. Front Neurol 2021; 12:660591. [PMID: 34025561 PMCID: PMC8137820 DOI: 10.3389/fneur.2021.660591] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/24/2021] [Indexed: 12/22/2022] Open
Abstract
Blood–brain barrier (BBB) dysfunction following brain insults has been associated with the development and progression of focal epilepsy, although the underlying molecular mechanisms are not fully elucidated yet. Activation of transforming growth factor beta (TGFβ) signaling in astrocytes by extravasated albumin impairs the ability of astrocytes to properly interact with neurons, eventually leading to epileptiform activity. We used the unilateral intracortical kainate mouse model of temporal lobe epilepsy (TLE) with hippocampal sclerosis (HS) to gain further insights into the role of BBB leakage in status epilepticus (SE)-induced epileptogenesis. Immunohistochemical examination revealed pronounced albumin extravasation already 4 h after SE induction. Astrocytes were virtually devoid of albumin immunoreactivity (IR), indicating the lack of uptake by this time point. Inhibition of the TGFβ pathway by the specific TGFβ receptor 1 (TGFβR1) kinase inhibitor IPW-5371 did not prevent seizure-induced reduction of astrocytic gap junction coupling. Thus, loss of coupling, which is thought to play a causative role in triggering TLE-HS, is most likely not mediated by extravasated albumin. Continuous telemetric EEG recordings and video monitoring performed over a period of 4 weeks after epilepsy induction revealed that inhibition of the TGFβ pathway during the initial phase of epileptogenesis slightly attenuated acute and chronic epileptiform activity, but did not reduce the extent of HS. Together, these data indicate that albumin extravasation due to increased BBB permeability and TGFβ pathway activation during the first hours after SE induction are not significantly involved in initiating TLE.
Collapse
Affiliation(s)
- Lukas Henning
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Peter Bedner
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
19
|
Wu Z, Deshpande T, Henning L, Bedner P, Seifert G, Steinhäuser C. Cell death of hippocampal CA1 astrocytes during early epileptogenesis. Epilepsia 2021; 62:1569-1583. [PMID: 33955001 DOI: 10.1111/epi.16910] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/06/2021] [Accepted: 04/06/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Growing evidence suggests that dysfunctional astrocytes are crucial players in the development of mesial temporal lobe epilepsy (MTLE). Using a mouse model closely recapitulating key alterations of chronic human MTLE with hippocampal sclerosis, here we asked whether death of astrocytes contributes to the initiation of the disease and investigated potential underlying molecular mechanisms. METHODS Antibody staining was combined with confocal imaging and semiquantitative real-time polymerase chain reaction analysis to identify markers of different cellular death mechanisms between 4 h and 3 days after epilepsy induction. RESULTS Four hours after kainate-mediated induction of status epilepticus (SE), we found a significant reduction in the density of astrocytes in the CA1 stratum radiatum (SR) of the ipsilateral hippocampus. This reduction was transient, as within the next 3 days, astrocyte cell numbers recovered to the initial values, which was accompanied by enhanced proliferation. Four hours after SE induction, a small proportion of astrocytes in the ipsilateral CA1 SR expressed autophagy-related genes and proteins, whereas we did not find astrocytes positive for cleaved caspase 3 or terminal deoxynucleotide transferase-mediated deoxyuridine triphosphate nick-end labeling, ruling out apoptosis-related astrocytic death. Importantly, at the same early time point post-SE, many astrocytes in the ipsilateral CA1 SR showed strong expression of genes encoding pro-necroptosis factors, including receptor-interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain-like protein (MLKL). Phosphorylation of MLKL (pMLKL), formation of necrosome complexes composed of RIPK3 and pMLKL, and translocation of pMLKL to the nucleus and to the plasma membrane were often observed in astrocytes of the ipsilateral hippocampus 4 h post-SE. SIGNIFICANCE The present study revealed that astrocytes die shortly after induction of SE. Our expression data and immunohistochemistry suggest that necroptosis and autophagy contribute to astrocytic death. These findings help to better understand how dysfunctional and pathological remodeling of astrocytes contributes to the initiation of temporal lobe epilepsy.
Collapse
Affiliation(s)
- Zhou Wu
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Tushar Deshpande
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany.,Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Lukas Henning
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Peter Bedner
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Gerald Seifert
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
20
|
Wei Y, Chen T, Bosco DB, Xie M, Zheng J, Dheer A, Ying Y, Wu Q, Lennon VA, Wu LJ. The complement C3-C3aR pathway mediates microglia-astrocyte interaction following status epilepticus. Glia 2021; 69:1155-1169. [PMID: 33314324 PMCID: PMC7936954 DOI: 10.1002/glia.23955] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 12/19/2022]
Abstract
Gliosis is a histopathological characteristic of epilepsy that comprises activated microglia and astrocytes. It is unclear whether or how crosstalk occurs between microglia and astrocytes in the evolution of epilepsy. Here, we report in a mouse model of status epilepticus, induced by intracerebroventricular injection of kainic acid (KA), sequential activation of microglia and astrocytes and their close spatial interaction in the hippocampal CA3 region. Microglial ablation reduced astrocyte activation and their upregulation of complement C3. When compared to wild-type mice, both C3-/- and C3aR-/- mice had significantly less microglia-astrocyte interaction in response to KA-induced status epilepticus. Additionally, KA-injected C3-/- mice had significantly less histochemical evidence of neurodegeneration. The results suggest that the C3-C3aR pathway contributes to KA-induced neurodegeneration by mediating microglia-astrocyte communication. The C3-C3aR pathway may prove to be a potential therapeutic target for epilepsy treatment.
Collapse
Affiliation(s)
- Yujia Wei
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Neurosurgery, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Tingjun Chen
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Dale B. Bosco
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Manling Xie
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Aastha Dheer
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yanlu Ying
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Qian Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Vanda A. Lennon
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| |
Collapse
|
21
|
Sadanandan N, Saft M, Gonzales-Portillo B, Borlongan CV. Multipronged Attack of Stem Cell Therapy in Treating the Neurological and Neuropsychiatric Symptoms of Epilepsy. Front Pharmacol 2021; 12:596287. [PMID: 33815100 PMCID: PMC8010689 DOI: 10.3389/fphar.2021.596287] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 01/15/2021] [Indexed: 11/13/2022] Open
Abstract
Epilepsy stands as a life-threatening disease that is characterized by unprovoked seizures. However, an important characteristic of epilepsy that needs to be examined is the neuropsychiatric aspect. Epileptic patients endure aggression, depression, and other psychiatric illnesses. Therapies for epilepsy can be divided into two categories: antiepileptic medications and surgical resection. Antiepileptic drugs are used to attenuate heightened neuronal firing and to lessen seizure frequency. Alternatively, surgery can also be conducted to physically cut out the area of the brain that is assumed to be the root cause for the anomalous firing that triggers seizures. While both treatments serve as viable approaches that aim to regulate seizures and ameliorate the neurological detriments spurred by epilepsy, they do not serve to directly counteract epilepsy's neuropsychiatric traits. To address this concern, a potential new treatment involves the use of stem cells. Stem cell therapy has been employed in experimental models of neurological maladies, such as Parkinson's disease, and neuropsychiatric illnesses like depression. Cell-based treatments for epilepsy utilizing stem cells such as neural stem cells (NSCs), mesenchymal stem cells (MSCs), and interneuron grafts have been explored in preclinical and clinical settings, highlighting both the acute and chronic stages of epilepsy. However, it is difficult to create an animal model to capitalize on all the components of epilepsy due to the challenges in delineating the neuropsychiatric aspect. Therefore, further preclinical investigation into the safety and efficacy of stem cell therapy in addressing both the neurological and the neuropsychiatric components of epilepsy is warranted in order to optimize cell dosage, delivery, and timing of cell transplantation.
Collapse
Affiliation(s)
| | | | | | - Cesar V. Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| |
Collapse
|
22
|
Exacerbation of Epilepsy by Astrocyte Alkalization and Gap Junction Uncoupling. J Neurosci 2021; 41:2106-2118. [PMID: 33478985 DOI: 10.1523/jneurosci.2365-20.2020] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/02/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022] Open
Abstract
Seizures invite seizures. At the initial stage of epilepsy, seizures intensify with each episode; however, the mechanisms underlying this exacerbation remain to be solved. Astrocytes have a strong control over neuronal excitability and the mode of information processing. This control is accomplished by adjusting the levels of various ions in the extracellular space. The network of astrocytes connected via gap junctions allows a wider or more confined distribution of these ions depending on the open probability of the gap junctions. K+ clearance relies on the K+ uptake by astrocytes and the subsequent diffusion of K+ through the astrocyte network. When astrocytes become uncoupled, K+ clearance becomes hindered. Accumulation of extracellular K+ leads to hyperexcitability of neurons. Here, using acute hippocampal slices from mice, we uncovered that brief periods of epileptiform activity result in gap junction uncoupling. In slices that experienced short-term epileptiform activity, extracellular K+ transients in response to glutamate became prolonged. Na+ imaging with a fluorescent indicator indicated that intercellular diffusion of small cations in the astrocytic syncytium via gap junctions became rapidly restricted after epileptiform activity. Using a transgenic mouse with astrocyte-specific expression of a pH sensor (Lck-E2GFP), we confirmed that astrocytes react to epileptiform activity with intracellular alkalization. Application of Na+/HCO3 - cotransporter blocker led to the suppression of intracellular alkalization of astrocytes and to the prevention of astrocyte uncoupling and hyperactivity intensification both in vitro and in vivo Therefore, the inhibition of astrocyte alkalization could become a promising therapeutic strategy for countering epilepsy development.SIGNIFICANCE STATEMENT We aimed to understand the mechanisms underlying the plastic change of forebrain circuits associated with the intensification of epilepsy. Here, we demonstrate that first-time exposure to only brief periods of epileptiform activity results in acute disturbance of the intercellular astrocyte network formed by gap junctions in hippocampal tissue slices from mice. Moreover, rapid clearance of K+ from the extracellular space was impaired. Epileptiform activity activated inward Na+/HCO3 - cotransport in astrocytes by cell depolarization, resulting in their alkalization. Our data suggest that alkaline pH shifts in astrocytes lead to gap junction uncoupling, hampering K+ clearance, and thereby to exacerbation of epilepsy. Pharmacological intervention could become a promising new strategy to dampen neuronal hyperexcitability and epileptogenesis.
Collapse
|
23
|
Caudal LC, Gobbo D, Scheller A, Kirchhoff F. The Paradox of Astroglial Ca 2 + Signals at the Interface of Excitation and Inhibition. Front Cell Neurosci 2020; 14:609947. [PMID: 33324169 PMCID: PMC7726216 DOI: 10.3389/fncel.2020.609947] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/03/2020] [Indexed: 12/15/2022] Open
Abstract
Astroglial networks constitute a non-neuronal communication system in the brain and are acknowledged modulators of synaptic plasticity. A sophisticated set of transmitter receptors in combination with distinct secretion mechanisms enables astrocytes to sense and modulate synaptic transmission. This integrative function evolved around intracellular Ca2+ signals, by and large considered as the main indicator of astrocyte activity. Regular brain physiology meticulously relies on the constant reciprocity of excitation and inhibition (E/I). Astrocytes are metabolically, physically, and functionally associated to the E/I convergence. Metabolically, astrocytes provide glutamine, the precursor of both major neurotransmitters governing E/I in the central nervous system (CNS): glutamate and γ-aminobutyric acid (GABA). Perisynaptic astroglial processes are structurally and functionally associated with the respective circuits throughout the CNS. Astonishingly, in astrocytes, glutamatergic as well as GABAergic inputs elicit similar rises in intracellular Ca2+ that in turn can trigger the release of glutamate and GABA as well. Paradoxically, as gliotransmitters, these two molecules can thus strengthen, weaken or even reverse the input signal. Therefore, the net impact on neuronal network function is often convoluted and cannot be simply predicted by the nature of the stimulus itself. In this review, we highlight the ambiguity of astrocytes on discriminating and affecting synaptic activity in physiological and pathological state. Indeed, aberrant astroglial Ca2+ signaling is a key aspect of pathological conditions exhibiting compromised network excitability, such as epilepsy. Here, we gather recent evidence on the complexity of astroglial Ca2+ signals in health and disease, challenging the traditional, neuro-centric concept of segregating E/I, in favor of a non-binary, mutually dependent perspective on glutamatergic and GABAergic transmission.
Collapse
Affiliation(s)
- Laura C Caudal
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| | - Davide Gobbo
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| | - Anja Scheller
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| | - Frank Kirchhoff
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| |
Collapse
|
24
|
Astrocytic Connexin43 Channels as Candidate Targets in Epilepsy Treatment. Biomolecules 2020; 10:biom10111578. [PMID: 33233647 PMCID: PMC7699773 DOI: 10.3390/biom10111578] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/15/2022] Open
Abstract
In epilepsy research, emphasis is put on exploring non-neuronal targets such as astrocytic proteins, since many patients remain pharmacoresistant to current treatments, which almost all target neuronal mechanisms. This paper reviews available data on astrocytic connexin43 (Cx43) signaling in seizures and epilepsy. Cx43 is a widely expressed transmembrane protein and the constituent of gap junctions (GJs) and hemichannels (HCs), allowing intercellular and extracellular communication, respectively. A plethora of research papers show altered Cx43 mRNA levels, protein expression, phosphorylation state, distribution and/or functional coupling in human epileptic tissue and experimental models. Human Cx43 mutations are linked to seizures as well, as 30% of patients with oculodentodigital dysplasia (ODDD), a rare genetic condition caused by mutations in the GJA1 gene coding for Cx43 protein, exhibit neurological symptoms including seizures. Cx30/Cx43 double knock-out mice show increased susceptibility to evoked epileptiform events in brain slices due to impaired GJ-mediated redistribution of K+ and glutamate and display a higher frequency of spontaneous generalized chronic seizures in an epilepsy model. Contradictory, Cx30/Cx43 GJs can traffic nutrients to high-energy demanding neurons and initiate astrocytic Ca2+ waves and hyper synchronization, thereby supporting proconvulsant effects. The general connexin channel blocker carbenoxolone and blockers from the fenamate family diminish epileptiform activity in vitro and improve seizure outcome in vivo. In addition, interventions with more selective peptide inhibitors of HCs display anticonvulsant actions. To conclude, further studies aiming to disentangle distinct roles of HCs and GJs are necessary and tools specifically targeting Cx43 HCs may facilitate the search for novel epilepsy treatments.
Collapse
|
25
|
Kim YS, Choi J, Yoon BE. Neuron-Glia Interactions in Neurodevelopmental Disorders. Cells 2020; 9:cells9102176. [PMID: 32992620 PMCID: PMC7601502 DOI: 10.3390/cells9102176] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022] Open
Abstract
Recent studies have revealed synaptic dysfunction to be a hallmark of various psychiatric diseases, and that glial cells participate in synapse formation, development, and plasticity. Glial cells contribute to neuroinflammation and synaptic homeostasis, the latter being essential for maintaining the physiological function of the central nervous system (CNS). In particular, glial cells undergo gliotransmission and regulate neuronal activity in tripartite synapses via ion channels (gap junction hemichannel, volume regulated anion channel, and bestrophin-1), receptors (for neurotransmitters and cytokines), or transporters (GLT-1, GLAST, and GATs) that are expressed on glial cell membranes. In this review, we propose that dysfunction in neuron-glia interactions may contribute to the pathogenesis of neurodevelopmental disorders. Understanding the mechanisms of neuron-glia interaction for synapse formation and maturation will contribute to the development of novel therapeutic targets of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Yoo Sung Kim
- Department of Molecular Biology, Dankook University, Cheonan 31116, Korea; (Y.S.K.); (J.C.)
| | - Juwon Choi
- Department of Molecular Biology, Dankook University, Cheonan 31116, Korea; (Y.S.K.); (J.C.)
| | - Bo-Eun Yoon
- Department of Molecular Biology, Dankook University, Cheonan 31116, Korea; (Y.S.K.); (J.C.)
- Department of Nanobiomedical science, Dankook University, Cheonan 31116, Korea
- Correspondence: ; Tel.: +82-41-529-6085
| |
Collapse
|
26
|
Courtney CD, Christian-Hinman C. Assessin' the Vexin' Connexin Between Severity of Epilepsy and Hippocampal Gliosis. Epilepsy Curr 2020; 20:294-296. [PMID: 34025243 PMCID: PMC7653651 DOI: 10.1177/1535759720944924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Constitutive Deletion of Astrocytic Connexins Aggravates Kainate-Induced Epilepsy Deshpande T, Li T, Henning L, Wu Z, Mueller JAL, Seifert G, Steinhäuser C, Bedner P. Glia. 2020. doi:10.1002/glia.23832 The astroglial gap junctional network formed by connexin (Cx) channels plays a central role in regulating neuronal activity and network synchronization. However, its involvement in the development and progression of epilepsy is not yet understood. Loss of interastrocytic gap junction (GJ) coupling has been observed in the sclerotic hippocampus of patients with mesial temporal lobe epilepsy (MTLE) and in mouse models of MTLE, leading to the suggestion that it plays a causative role in the pathogenesis. To further elucidate this clinically relevant question, we investigated consequences of astrocyte disconnection on the time course and severity of kainate-induced MTLE with hippocampal sclerosis (HS) by comparing mice deficient for astrocytic Cx proteins with wild-type (WT) mice. Continuous telemetric EEG recordings and video monitoring performed over a period of 4 weeks after epilepsy induction revealed substantially higher seizure and interictal spike activity during the chronic phase in Cx deficient versus WT mice, while the severity of status epilepticus was not different. Immunohistochemical analysis showed that, despite the elevated chronic seizure activity, astrocyte disconnection did not aggravate the severity of HS. Indeed, the extent of CA1 pyramidal cell loss was similar between the experimental groupsx, while astrogliosis, granule cell dispersion, angiogenesis, and microglia activation were even reduced in Cx deficient as compared to WT mice. Interestingly, seizure-induced neurogenesis in the adult dentate gyrus was also independent of astrocytic Cxs. Together, our data indicate that constitutive loss of GJ coupling between astrocytes promotes neuronal hyperexcitability and attenuates seizure-induced histopathological outcomes.
Collapse
|