1
|
Follmann R, Jaswal T, Jacob G, de Oliveira JF, Herbert CB, Macau EEN, Rosa E. Temperature effects on neuronal synchronization in seizures. CHAOS (WOODBURY, N.Y.) 2024; 34:083141. [PMID: 39191247 DOI: 10.1063/5.0219836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/08/2024] [Indexed: 08/29/2024]
Abstract
We present a computational model of networked neurons developed to study the effect of temperature on neuronal synchronization in the brain in association with seizures. The network consists of a set of chaotic bursting neurons surrounding a core tonic neuron in a square lattice with periodic boundary conditions. Each neuron is reciprocally coupled to its four nearest neighbors via temperature dependent gap junctions. Incorporating temperature in the gap junctions makes the coupling stronger when temperature rises, resulting in higher likelihood for synchrony in the network. Raising the temperature eventually makes the network elicit waves of synchronization in circular ripples that propagate from the center outwardly. We suggest this process as a possible underlying mechanism for seizures induced by elevated brain temperatures.
Collapse
Affiliation(s)
- Rosangela Follmann
- School of Information Technology, Illinois State University, Normal, Illinois 61790, USA
| | - Twinkle Jaswal
- School of Information Technology, Illinois State University, Normal, Illinois 61790, USA
| | - George Jacob
- School of Information Technology, Illinois State University, Normal, Illinois 61790, USA
| | | | - Carter B Herbert
- Department of Physics, Illinois State University, Normal, Illinois 61790, USA
| | - Elbert E N Macau
- Federal University of São Paulo (UNIFESP), São José dos Campos, São Paulo, 12247-014 Brazil
| | - Epaminondas Rosa
- Department of Physics, Illinois State University, Normal, Illinois 61790, USA
- School of Biological Sciences, Illinois State University, Normal, Illinois 61790, USA
| |
Collapse
|
2
|
Sari S, Yurtoğlu S, Zengin M, Marcinkowska M, Siwek A, Saraç S. Azoles display promising anticonvulsant effects through possible PPAR-α activation. Neurosci Lett 2024; 828:137750. [PMID: 38548219 DOI: 10.1016/j.neulet.2024.137750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 04/19/2024]
Abstract
Azoles such as nafimidone, denzimol and loreclezole are known for their clinical efficacy against epilepsy, and loreclezole acts by potentiating γ-aminobutyric acid (GABA)-ergic currents. In the current study, we report a series of azole derivatives in alcohol ester and oxime ester structure showing promising anticonvulsant effects in 6 Hz and maximal electro shock (MES) models with minimal toxicity. The most promising of the series, 5f, was active in both 6 Hz and MES tests with a median effective dose (ED50) of 118.92 mg/kg in 6 Hz test and a median toxic dose (TD50) twice as high in mice. The compounds were predicted druglike and blood-brain barrier (BBB) penetrant in silico. Contrary to what was expected, the compounds showed no in vitro affinity to GABAA receptors (GABAARs) in radioligand binding assays; however, they were found structurally similar to peroxisome proliferator-activated receptors alpha (PPAR-α) agonists and predicted to show high affinity and agonist-like binding to PPAR-α in molecular docking studies. As a result, 5f emerged as a safe azole anticonvulsant with a wide therapeutic window and possible action through PPAR-α activation.
Collapse
Affiliation(s)
- Suat Sari
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.
| | - Sibel Yurtoğlu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Merve Zengin
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Monika Marcinkowska
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Agata Siwek
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Selma Saraç
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Baskent University, Ankara, Turkey
| |
Collapse
|
3
|
Lucasius C, Grigorovsky V, Nariai H, Galanopoulou AS, Gursky J, Moshe SL, Bardakjian BL. Biomimetic Deep Learning Networks With Applications to Epileptic Spasms and Seizure Prediction. IEEE Trans Biomed Eng 2024; 71:1056-1067. [PMID: 37851549 PMCID: PMC10979638 DOI: 10.1109/tbme.2023.3325762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
OBJECTIVE In this study, we present a novel biomimetic deep learning network for epileptic spasms and seizure prediction and compare its performance with state-of-the-art conventional machine learning models. METHODS Our proposed model incorporates modular Volterra kernel convolutional networks and bidirectional recurrent networks in combination with the phase amplitude cross-frequency coupling features derived from scalp EEG. They are applied to the standard CHB-MIT dataset containing focal epilepsy episodes as well as two other datasets from the Montefiore Medical Center and the University of California Los Angeles that provide data of patients experiencing infantile spasm (IS) syndrome. RESULTS Overall, in this study, the networks can produce accurate predictions (100%) and significant detection latencies (10 min). Furthermore, the biomimetic network outperforms conventional ones by producing no false positives. SIGNIFICANCE Biomimetic neural networks utilize extensive knowledge about processing and learning in the electrical networks of the brain. Predicting seizures in adults can improve their quality of life. Epileptic spasms in infants are part of a particular seizure type that needs identifying when suspicious behaviors are noticed in babies. Predicting epileptic spasms within a given time frame (the prediction horizon) suggests their existence and allows an epileptologist to flag an EEG trace for future review.
Collapse
|
4
|
Shao M, Yu H, Santhakumar V, Yu J. Antiepileptogenic and neuroprotective effect of mefloquine after experimental status epilepticus. Epilepsy Res 2023; 198:107257. [PMID: 37989006 DOI: 10.1016/j.eplepsyres.2023.107257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/30/2023] [Accepted: 11/06/2023] [Indexed: 11/23/2023]
Abstract
Acquired temporal lobe epilepsy (TLE) characterized by spontaneous recurrent seizures (SRS) and hippocampal inhibitory neuron dysfunction is often refractory to current therapies. Gap junctional or electrical coupling between inhibitory neurons has been proposed to facilitate network synchrony and intercellular molecular exchange suggesting a role in both seizures and neurodegeneration. While gap junction blockers can limit acute seizures, whether blocking neuronal gap junctions can modify development of chronic epilepsy has not been examined. This study examined whether mefloquine, a selective blocker of Connexin 36 gap junctions which are well characterized in inhibitory neurons, can limit epileptogenesis and related cellular and behavioral pathology in a model of acquired TLE. A single, systemic dose of mefloquine administered early after pilocarpine-induced status epilepticus (SE) in rat reduced both development of SRS and behavioral co-morbidities. Immunostaining for interneuron subtypes identified that mefloquine treatment likely reduced delayed inhibitory neuronal loss after SE. Uniquely, parvalbumin expressing neurons in the hippocampal dentate gyrus appeared relatively resistant to early cell loss after SE. Functionally, whole cell patch clamp recordings revealed that mefloquine treatment preserved inhibitory synaptic drive to projection neurons one week and one month after SE. These results demonstrate that mefloquine, a drug already approved for malaria prophylaxis, is potentially antiepileptogenic and can protect against progressive interneuron loss and behavioral co-morbidities of epilepsy.
Collapse
Affiliation(s)
- Mingting Shao
- Department of Neurosurgery, the First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Hang Yu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Vijayalakshmi Santhakumar
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA 92521, USA
| | - Jiandong Yu
- Department of Neurosurgery, the First Affiliated Hospital of Bengbu Medical College, Bengbu, China.
| |
Collapse
|
5
|
Ismail FS, Faustmann PM, Förster E, Corvace F, Faustmann TJ. Tiagabine and zonisamide differentially regulate the glial properties in an astrocyte-microglia co-culture model of inflammation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3253-3267. [PMID: 37231170 PMCID: PMC10567966 DOI: 10.1007/s00210-023-02538-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
Due to the role of astrocytes and microglia in the pathophysiology of epilepsy and limited studies of antiseizure medication (ASM) effects on glial cells, we studied tiagabine (TGB) and zonisamide (ZNS) in an astrocyte-microglia co-culture model of inflammation. Different concentrations of ZNS (10, 20, 40, 100 µg/ml) or TGB (1, 10, 20, 50 µg/ml) were added to primary rat astrocytes co-cultures with 5-10% (M5, physiological conditions) or 30-40% (M30, pathological inflammatory conditions) microglia for 24 h, aiming to study glial viability, microglial activation, connexin 43 (Cx43) expression and gap-junctional coupling. ZNS led to the reduction of glial viability by only 100 µg/ml under physiological conditions. By contrast, TGB revealed toxic effects with a significant, concentration-dependent reduction of glial viability under physiological and pathological conditions. After the incubation of M30 co-cultures with 20 µg/ml TGB, the microglial activation was significantly decreased and resting microglia slightly increased, suggesting possible anti-inflammatory features of TGB under inflammatory conditions. Otherwise, ZNS caused no significant changes of microglial phenotypes. The gap-junctional coupling was significantly decreased after the incubation of M5 co-cultures with 20 and 50 µg/ml TGB, which can be related to its anti-epileptic activity under noninflammatory conditions. A significant decrease of Cx43 expression and cell-cell coupling was found after the incubation of M30 co-cultures with 10 µg/ml ZNS, suggesting additional anti-seizure effects of ZNS with the disruption of glial gap-junctional communication under inflammatory conditions. TGB and ZNS differentially regulated the glial properties. Developing novel ASMs targeting glial cells may have future potential as an "add-on" therapy to classical ASMs targeting neurons.
Collapse
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, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Eckart Förster
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Franco Corvace
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Timo Jendrik Faustmann
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
6
|
Lia A, Di Spiezio A, Vitalini L, Tore M, Puja G, Losi G. Ion Channels and Ionotropic Receptors in Astrocytes: Physiological Functions and Alterations in Alzheimer's Disease and Glioblastoma. Life (Basel) 2023; 13:2038. [PMID: 37895420 PMCID: PMC10608464 DOI: 10.3390/life13102038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/03/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
The human brain is composed of nearly one hundred billion neurons and an equal number of glial cells, including macroglia, i.e., astrocytes and oligodendrocytes, and microglia, the resident immune cells of the brain. In the last few decades, compelling evidence has revealed that glial cells are far more active and complex than previously thought. In particular, astrocytes, the most abundant glial cell population, not only take part in brain development, metabolism, and defense against pathogens and insults, but they also affect sensory, motor, and cognitive functions by constantly modulating synaptic activity. Not surprisingly, astrocytes are actively involved in neurodegenerative diseases (NDs) and other neurological disorders like brain tumors, in which they rapidly become reactive and mediate neuroinflammation. Reactive astrocytes acquire or lose specific functions that differently modulate disease progression and symptoms, including cognitive impairments. Astrocytes express several types of ion channels, including K+, Na+, and Ca2+ channels, transient receptor potential channels (TRP), aquaporins, mechanoreceptors, and anion channels, whose properties and functions are only partially understood, particularly in small processes that contact synapses. In addition, astrocytes express ionotropic receptors for several neurotransmitters. Here, we provide an extensive and up-to-date review of the roles of ion channels and ionotropic receptors in astrocyte physiology and pathology. As examples of two different brain pathologies, we focus on Alzheimer's disease (AD), one of the most diffuse neurodegenerative disorders, and glioblastoma (GBM), the most common brain tumor. Understanding how ion channels and ionotropic receptors in astrocytes participate in NDs and tumors is necessary for developing new therapeutic tools for these increasingly common neurological conditions.
Collapse
Affiliation(s)
- Annamaria Lia
- Department Biomedical Science, University of Padova, 35131 Padova, Italy; (A.L.); (A.D.S.)
| | - Alessandro Di Spiezio
- Department Biomedical Science, University of Padova, 35131 Padova, Italy; (A.L.); (A.D.S.)
- Neuroscience Institute (CNR-IN), Padova Section, 35131 Padova, Italy
| | - Lorenzo Vitalini
- Department Life Science, University of Modena and Reggio Emilia, 41125 Modena, Italy; (L.V.); (G.P.)
| | - Manuela Tore
- Institute of Nanoscience (CNR-NANO), Modena Section, 41125 Modena, Italy;
- Department Biomedical Science, Metabolic and Neuroscience, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giulia Puja
- Department Life Science, University of Modena and Reggio Emilia, 41125 Modena, Italy; (L.V.); (G.P.)
| | - Gabriele Losi
- Institute of Nanoscience (CNR-NANO), Modena Section, 41125 Modena, Italy;
- Department Biomedical Science, Metabolic and Neuroscience, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
7
|
Dapino A, Davoine F, Curti S. D-type K+ current rules the function of electrically coupled neurons in a species-specific fashion. J Gen Physiol 2023; 155:e202313353. [PMID: 37378665 PMCID: PMC10308032 DOI: 10.1085/jgp.202313353] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/17/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Electrical synapses supported by gap junctions are known to form networks of electrically coupled neurons in many regions of the mammalian brain, where they play relevant functional roles. Yet, how electrical coupling supports sophisticated network operations and the contribution of the intrinsic electrophysiological properties of neurons to these operations remain incompletely understood. Here, a comparative analysis of electrically coupled mesencephalic trigeminal (MesV) neurons uncovered remarkable difference in the operation of these networks in highly related species. While spiking of MesV neurons might support the recruitment of coupled cells in rats, this rarely occurs in mice. Using whole-cell recordings, we determined that the higher efficacy in postsynaptic recruitment in rat's MesV neurons does not result from coupling strength of larger magnitude, but instead from the higher excitability of coupled neurons. Consistently, MesV neurons from rats present a lower rheobase, more hyperpolarized threshold, as well as a higher ability to generate repetitive discharges, in comparison to their counterparts from mice. This difference in neuronal excitability results from a significantly higher magnitude of the D-type K+ current (ID) in MesV neurons from mice, indicating that the magnitude of this current gates the recruitment of postsynaptic-coupled neurons. Since MesV neurons are primary afferents critically involved in the organization of orofacial behaviors, activation of a coupled partner could support lateral excitation, which by amplifying sensory inputs may significantly contribute to information processing and the organization of motor outputs.
Collapse
Affiliation(s)
- Antonella Dapino
- Laboratorio de Neurofisiología Celular, Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Federico Davoine
- Instituto de Ingeniería Eléctrica, Facultad de Ingeniería, Universidad de la República, Montevideo, Uruguay
| | - Sebastian Curti
- Laboratorio de Neurofisiología Celular, Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
8
|
Andrioli A, Fabene PF, Mudò G, Barresi V, Di Liberto V, Frinchi M, Bentivoglio M, Condorelli DF. Downregulation of the Astroglial Connexin Expression and Neurodegeneration after Pilocarpine-Induced Status Epilepticus. Int J Mol Sci 2022; 24:ijms24010023. [PMID: 36613467 PMCID: PMC9819917 DOI: 10.3390/ijms24010023] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Astrocytic networks and gap junctional communication mediated by connexins (Cxs) have been repeatedly implicated in seizures, epileptogenesis, and epilepsy. However, the effect of seizures on Cx expression is controversial. The present study focused on the response of Cxs to status epilepticus (SE), which is in turn an epileptogenic insult. The expression of neuronal Cx36 and astrocytic Cx30 and Cx43 mRNAs was investigated in the brain of rats in the first day after pilocarpine-induced SE. In situ hybridization revealed a progressive decrease in Cx43 and Cx30 mRNA levels, significantly marked 24 h after SE onset in neocortical areas and the hippocampus, and in most thalamic domains, whereas Cx36 mRNA did not exhibit obvious changes. Regional evaluation with quantitative real-time-RT-PCR confirmed Cx43 and Cx30 mRNA downregulation 24 h after SE, when ongoing neuronal cell death was found in the same brain regions. Immunolabeling showed at the same time point marked a decrease in Cx43, microglia activation, and interleukin-1β induction in some microglial cells. The data showed a transient downregulation of astroglial Cxs in the cortical and thalamic areas in which SE triggers neurodegenerative events in concomitance with microglia activation and cytokine expression. This could potentially represent a protective response of neuroglial networks to SE-induced acute damage.
Collapse
Affiliation(s)
- Anna Andrioli
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy
| | - Paolo Francesco Fabene
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy
- Verona Unit, National Institute of Neuroscience (INN), 37129 Verona, Italy
| | - Giuseppa Mudò
- Department of Biomedicine, Neuroscience and Advanced Diagnostic (BiND), University of Palermo, 90133 Palermo, Italy
| | - Vincenza Barresi
- Unit of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Valentina Di Liberto
- Department of Biomedicine, Neuroscience and Advanced Diagnostic (BiND), University of Palermo, 90133 Palermo, Italy
| | - Monica Frinchi
- Department of Biomedicine, Neuroscience and Advanced Diagnostic (BiND), University of Palermo, 90133 Palermo, Italy
| | - Marina Bentivoglio
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy
- Verona Unit, National Institute of Neuroscience (INN), 37129 Verona, Italy
| | - Daniele Filippo Condorelli
- Unit of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
- Correspondence:
| |
Collapse
|
9
|
Ismail FS, Faustmann PM, Kümmel ML, Förster E, Faustmann TJ, Corvace F. Brivaracetam exhibits mild pro-inflammatory features in an in vitro astrocyte-microglia co-culture model of inflammation. Front Cell Neurosci 2022; 16:995861. [PMID: 36406753 PMCID: PMC9670320 DOI: 10.3389/fncel.2022.995861] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 10/12/2022] [Indexed: 10/23/2024] Open
Abstract
Implications of glia in the pathophysiology of epilepsy raise the question of how these cells besides neurons are responsive to antiseizure medications (ASMs). Understanding ASM effects on glia and glia-mediated inflammation may help to explore astrocytes and microglia as potential targets for alternative anti-epileptogenic therapies. The aim of this study was to investigate the effects of the new generation ASM brivaracetam (BRV) in an astrocyte-microglia co-culture model of inflammation. Primary rat astrocytes co-cultures containing 5%-10% (M5, "physiological" conditions) or 30%-40% (M30, "pathological inflammatory" conditions) of microglia were treated with different concentrations of BRV (0.5, 2, 10, and 20 μg/ml) for 24 h. Glial cell viability was measured by MTT assay. Microglial activation states were analyzed by immunocytochemistry and astroglial connexin 43 (Cx43) expression by Western blot analysis and immunocytochemistry. Gap-junctional coupling was studied via Scrape Loading. Incubation with high, overdose concentration (20 μg/ml) of BRV significantly reduced the glial cell viability under physiological conditions (p < 0.01: **). Treatment with BRV in therapeutic concentrations (0.5 and 2 μg/ml) reduced the resting microglia (p < 0.05: *) and increased the microglial activation under inflammatory conditions (p < 0.01: **). Astroglial Cx43 expression was not affected. The gap-junctional coupling significantly increased only by 0.5 μg/ml BRV under physiological conditions (p < 0.05: *). Our findings suggest mild pro-inflammatory, in vitro features of BRV with regard to microglia morphology. BRV showed no effects on Cx43 expression and only limited effects on gap-junctional coupling. Reduction of glial viability by overdose BRV indicates possible toxic effects.
Collapse
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
| | - Marie-Luise Kümmel
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Bochum, Germany
| | - Eckart Förster
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Bochum, Germany
| | - Timo Jendrik Faustmann
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Franco Corvace
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
10
|
Liu B, Ran X, Yi Y, Zhang X, Chen H, Hu Y. Anticonvulsant Effect of Carbenoxolone on Chronic Epileptic Rats and Its Mechanism Related to Connexin and High-Frequency Oscillations. Front Mol Neurosci 2022; 15:870947. [PMID: 35615064 PMCID: PMC9125185 DOI: 10.3389/fnmol.2022.870947] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/04/2022] [Indexed: 12/03/2022] Open
Abstract
Objective This study was designed to investigate the influence and mechanism of gap junction carbenoxolone (CBX) on dynamic changes in the spectral power of ripples and fast ripples (FRs) in the hippocampus of chronic epileptic rats. Methods The lithium-pilocarpine (PILO) status epilepticus (SE) model (PILO group) and the CBX pretreatment model (CBX + PILO group) were established to analyze dynamic changes in the spectral power of ripples and FRs, and the dynamic expression of connexin (CX)26, CX32, CX36, and CX43 in the hippocampus of chronic epileptic rats. Results Within 28 days after SE, the number of spontaneous recurrent seizures (SRSs) in the PILO group was significantly higher than that in the CBX + PILO group. The average spectral power of FRs in the PILO group was significantly higher than the baseline level at 1 and 7 days after SE. The average spectral power of FRs in the PILO group was significantly higher than that in the CBX + PILO group at 1, 7, and 14 days after SE. Seizures induced an increase in CX43 expression at 1 and 7 days after SE, but had no significant effect on CX26, CX36, or CX32. CBX pretreatment did not affect the expression of CXs in the hippocampus of normal rats, but it inhibited the expression of CX43 in epileptic rats. The number of SRSs at 2 and 4 weeks after SE had the highest correlation with the average spectral power of FRs; the average spectral power of FRs was moderately correlated with the expression of CX43. Conclusion The results of this study indicate that the energy of FRs may be regulated by its interference with the expression of CX43, and thus, affect seizures. Blocking the expression of CX43 thereby reduces the formation of pathological high-frequency oscillations (HFOs), making it a promising strategy for the treatment of chronic epilepsy.
Collapse
Affiliation(s)
- Benke Liu
- Department of Neurology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Shenzhen Baoan Women’s and Children’s Hospital, Jinan University, Shenzhen, China
| | - Xiao Ran
- Department of Neurology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Yanjun Yi
- Department of Neurology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Xinyu Zhang
- Department of Neurology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Hengsheng Chen
- Department of Neurology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Yue Hu
- Department of Neurology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- *Correspondence: Yue Hu,
| |
Collapse
|
11
|
Corvace F, Faustmann TJ, Faustmann PM, Ismail FS. Anti-inflammatory properties of lacosamide in an astrocyte-microglia co-culture model of inflammation. Eur J Pharmacol 2022; 915:174696. [PMID: 34902360 DOI: 10.1016/j.ejphar.2021.174696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 11/03/2022]
Abstract
PURPOSE Understanding the effects of antiepileptic drugs on glial cells and glia-mediated inflammation is a new approach to future treatment of epilepsy. Little is known about direct effects of the antiepileptic drug lacosamide (LCM) on glial cells. Therefore, we aimed to study the LCM effects on glial viability, microglial activation, expression of gap-junctional (GJ) protein Cx43 as well as intercellular communication in an in vitro astrocyte-microglia co-culture model of inflammation. METHODS Primary rat astrocytes co-cultures containing 5% (M5, "physiological" conditions) or 30% (M30, "pathological inflammatory" conditions) of microglia were treated with different concentrations of LCM [5, 15, 30, and 90 μg/ml] for 24 h. Glial cell viability was measured by MTT assay. Immunocytochemistry was performed to analyze the microglial activation state. Western blot analysis was used to quantify the astroglial Cx43 expression. The GJ cell communication was studied via Scrape Loading. RESULTS A concentration-dependent incubation with LCM did not affect the glial cell viability both under physiological and pathological conditions. LCM induced a significant concentration-dependent decrease of activated microglia with parallel increase of ramified microglia under pathological inflammatory conditions. This correlated with an increase in astroglial Cx43 expression. Nevertheless, the functional coupling via GJs was significantly reduced after incubation with LCM. CONCLUSION LCM has not shown effects on the glial cell viability. The reduced GJ coupling by LCM could be related to its anti-epileptic activity. The anti-inflammatory glial features of LCM with inhibition of microglial activation under inflammatory conditions support beneficial role in epilepsy associated with neuroinflammation.
Collapse
Affiliation(s)
- Franco Corvace
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Bochum, Germany
| | - Timo Jendrik Faustmann
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University, Düsseldorf, 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
| | - Fatme Seval Ismail
- Department of Neurology, University Hospital Knappschaftskrankenhaus Bochum, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
12
|
Choudhary A, Mu C, Barrett KT, Charkhand B, Williams-Dyjur C, Marks WN, Shearer J, Rho JM, Scantlebury MH. The link between brain acidosis, breathing and seizures: a novel mechanism of action for the ketogenic diet in a model of infantile spasms. Brain Commun 2021; 3:fcab189. [PMID: 34734183 DOI: 10.1093/braincomms/fcab189] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2021] [Indexed: 11/12/2022] Open
Abstract
Infantile spasms (IS) syndrome is a catastrophic, epileptic encephalopathy of infancy that is often refractory to current antiepileptic therapies. The ketogenic diet (KD) has emerged as an alternative treatment for patients with medically intractable epilepsy, though the prospective validity and mechanism of action for IS remains largely unexplored. We investigated the KD's efficacy as well as its mechanism of action in a rodent model of intractable IS. The spasms were induced using the triple-hit paradigm and the animals were then artificially reared and put on either the KD (4:1 fats: carbohydrate + protein) or a control milk diet (CM; 1.7:1). 31Phosphorus magnetic resonance spectroscopy (31P MRS) and head-out plethysmography were examined in conjunction with continuous video-EEG behavioural recordings in lesioned animals and sham-operated controls. The KD resulted in a peripheral ketosis observed both in the blood and urine. The KD led to a robust reduction in the frequency of spasms observed, with approximately a 1.5-fold increase in the rate of survival. Intriguingly, the KD resulted in an intracerebral acidosis as measured with 31P MRS. In addition, the respiratory profile of the lesioned rats on the KD was significantly altered with slower, deeper and longer breathing, resulting in decreased levels of expired CO2. Sodium bicarbonate supplementation, acting as a pH buffer, partially reversed the KD's protective effects on spasm frequency. There were no differences in the mitochondrial respiratory profiles in the liver and brain frontal cortex measured between the groups, supporting the notion that the effects of the KD on breathing are not entirely due to changes in intermediary metabolism. Together, our results indicate that the KD produces its anticonvulsant effects through changes in respiration leading to intracerebral acidosis. These findings provide a novel understanding of the mechanisms underlying the anti-seizure effects of the KD in IS. Further research is required to determine whether the effects of the KD on breathing and intracerebral acid-base balance are seen in other paediatric models of epilepsy.
Collapse
Affiliation(s)
- Anamika Choudhary
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary Alberta, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Chunlong Mu
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary Alberta, Calgary, Alberta, Canada.,Department of Biochemistry & Molecular Biology, Cumming School of Medicine and Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Karlene T Barrett
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary Alberta, Calgary, Alberta, Canada
| | - Behshad Charkhand
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Christine Williams-Dyjur
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Wendie N Marks
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary Alberta, Calgary, Alberta, Canada.,Department of Biochemistry & Molecular Biology, Cumming School of Medicine and Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Jane Shearer
- Alberta Children's Hospital Research Institute, University of Calgary Alberta, Calgary, Alberta, Canada.,Department of Biochemistry & Molecular Biology, Cumming School of Medicine and Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Jong M Rho
- Departments of Neurosciences and Pediatrics, University of California San Diego (UCSD), San Diego, CA, USA
| | - Morris H Scantlebury
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary Alberta, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
13
|
Yi YJ, Ran X, Xiang J, Li XY, Jiang L, Chen HS, Hu Y. Effect of gap junction blockers on hippocampal ripple energy expression in rats with status epilepticus. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2021; 23:848-853. [PMID: 34511176 DOI: 10.7499/j.issn.1008-8830.2103162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
OBJECTIVES To study the effect of gap junction blockers, quinine (QUIN) and carbenoxolone (CBX), on hippocampal ripple energy expression in rats with status epilepticus (SE). METHODS A total of 24 rats were randomly divided into four groups: model, QUIN, valproic acid (VPA), and CBX (n=6 each). A rat model of SE induced by lithium-pilocarpine (PILO) was prepared. The QUIN, VPA, and CBX groups were given intraperitoneal injection of QUIN (50 mg/kg), VPA by gavage (200 mg/kg), and intraperitoneal injection of CBX (50 mg/kg) respectively, at 3 days before PILO injection. Electroencephalography was used to analyze the change in hippocampal ripple energy before and after modeling, as well as before and after chloral hydrate injection to control seizures. RESULTS Ripple expression was observed in the hippocampal CA1, CA3, and dentate gyrus regions of normal rats. After 10 minutes of PILO injection, all groups had a gradual increase in mean ripple energy expression compared with 1 day before modeling, with the highest expression level before chloral hydrate injection in the model, VPA and CBX groups (P<0.05). The QUIN group had the highest expression level of mean ripple energy 60 minutes after PILO injection. The mean ripple energy returned to normal levels in the three intervention groups immediately after chloral hydrate injection, while in the model group, the mean ripple energy returned to normal levels 1 hour after chloral hydrate injection. The mean ripple energy remained normal till to day 3 after SE in the four groups. The changing trend of maximum ripple energy was similar to that of mean ripple energy. CONCLUSIONS The change in ripple energy can be used as a quantitative indicator for early warning of seizures, while it cannot predict seizures in the interictal period. Gap junction blockers can reduce ripple energy during seizures.
Collapse
Affiliation(s)
- Yan-Jun Yi
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing 400136
| | - Xiao Ran
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing 400136
| | | | | | - Li Jiang
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing 400136
| | - Heng-Sheng Chen
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing 400136
| | - Yue Hu
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing 400136
| |
Collapse
|
14
|
Leite JP, Peixoto-Santos JE. Glia and extracellular matrix molecules: What are their importance for the electrographic and MRI changes in the epileptogenic zone? Epilepsy Behav 2021; 121:106542. [PMID: 31884121 DOI: 10.1016/j.yebeh.2019.106542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 12/22/2022]
Abstract
Glial cells and extracellular matrix (ECM) molecules are crucial for the maintenance of brain homeostasis. Especially because of their actions regarding neurotransmitter and ionic control, and synaptic function, these cells can potentially contribute to the hyperexcitability seen in the epileptogenic, while ECM changes are linked to synaptic reorganization. The present review will explore glial and ECM homeostatic roles and their potential contribution to tissue plasticity. Finally, we will address how glial, and ECM changes in the epileptogenic zone can be seen in magnetic resonance imaging (MRI), pointing out their importance as markers for the extension of the epileptogenic area. This article is part of the Special Issue "NEWroscience 2018".
Collapse
Affiliation(s)
- Joao Pereira Leite
- Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| | - Jose Eduardo Peixoto-Santos
- Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Department of Neurology and Neurosurgery, Paulista School of Medicine, UNIFESP, Sao Paulo, Brazil
| |
Collapse
|
15
|
Gobbo D, Scheller A, Kirchhoff F. From Physiology to Pathology of Cortico-Thalamo-Cortical Oscillations: Astroglia as a Target for Further Research. Front Neurol 2021; 12:661408. [PMID: 34177766 PMCID: PMC8219957 DOI: 10.3389/fneur.2021.661408] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/11/2021] [Indexed: 12/21/2022] Open
Abstract
The electrographic hallmark of childhood absence epilepsy (CAE) and other idiopathic forms of epilepsy are 2.5-4 Hz spike and wave discharges (SWDs) originating from abnormal electrical oscillations of the cortico-thalamo-cortical network. SWDs are generally associated with sudden and brief non-convulsive epileptic events mostly generating impairment of consciousness and correlating with attention and learning as well as cognitive deficits. To date, SWDs are known to arise from locally restricted imbalances of excitation and inhibition in the deep layers of the primary somatosensory cortex. SWDs propagate to the mostly GABAergic nucleus reticularis thalami (NRT) and the somatosensory thalamic nuclei that project back to the cortex, leading to the typical generalized spike and wave oscillations. Given their shared anatomical basis, SWDs have been originally considered the pathological transition of 11-16 Hz bursts of neural oscillatory activity (the so-called sleep spindles) occurring during Non-Rapid Eye Movement (NREM) sleep, but more recent research revealed fundamental functional differences between sleep spindles and SWDs, suggesting the latter could be more closely related to the slow (<1 Hz) oscillations alternating active (Up) and silent (Down) cortical activity and concomitantly occurring during NREM. Indeed, several lines of evidence support the fact that SWDs impair sleep architecture as well as sleep/wake cycles and sleep pressure, which, in turn, affect seizure circadian frequency and distribution. Given the accumulating evidence on the role of astroglia in the field of epilepsy in the modulation of excitation and inhibition in the brain as well as on the development of aberrant synchronous network activity, we aim at pointing at putative contributions of astrocytes to the physiology of slow-wave sleep and to the pathology of SWDs. Particularly, we will address the astroglial functions known to be involved in the control of network excitability and synchronicity and so far mainly addressed in the context of convulsive seizures, namely (i) interstitial fluid homeostasis, (ii) K+ clearance and neurotransmitter uptake from the extracellular space and the synaptic cleft, (iii) gap junction mechanical and functional coupling as well as hemichannel function, (iv) gliotransmission, (v) astroglial Ca2+ signaling and downstream effectors, (vi) reactive astrogliosis and cytokine release.
Collapse
Affiliation(s)
- Davide Gobbo
- 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
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| |
Collapse
|
16
|
In vivo calcium imaging reveals disordered interictal network dynamics in epileptic stxbp1b zebrafish. iScience 2021; 24:102558. [PMID: 34142057 PMCID: PMC8184515 DOI: 10.1016/j.isci.2021.102558] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/29/2021] [Accepted: 05/17/2021] [Indexed: 12/19/2022] Open
Abstract
STXBP1 mutations are associated with encephalopathy, developmental delay, intellectual disability, and epilepsy. While neural networks are known to operate at a critical state in the healthy brain, network behavior during pathological epileptic states remains unclear. Examining activity during periods between well-characterized ictal-like events (i.e., interictal period) could provide a valuable step toward understanding epileptic networks. To study these networks in the context of STXBP1 mutations, we combine a larval zebrafish model with in vivo fast confocal calcium imaging and extracellular local field potential recordings. Stxbp1b mutants display transient periods of elevated activity among local clusters of interacting neurons. These network "cascade" events were significantly larger in size and duration in mutants. At mesoscale resolution, cascades exhibit neurodevelopmental abnormalities. At single-cell scale, we describe spontaneous hyper-synchronized neuronal ensembles. That calcium imaging reveals uniquely disordered brain states during periods between pathological ictal-like seizure events is striking and represents a potential interictal biomarker.
Collapse
|
17
|
Byrne Á, Ross J, Nicks R, Coombes S. Mean-Field Models for EEG/MEG: From Oscillations to Waves. Brain Topogr 2021; 35:36-53. [PMID: 33993357 PMCID: PMC8813727 DOI: 10.1007/s10548-021-00842-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 04/21/2021] [Indexed: 11/24/2022]
Abstract
Neural mass models have been used since the 1970s to model the coarse-grained activity of large populations of neurons. They have proven especially fruitful for understanding brain rhythms. However, although motivated by neurobiological considerations they are phenomenological in nature, and cannot hope to recreate some of the rich repertoire of responses seen in real neuronal tissue. Here we consider a simple spiking neuron network model that has recently been shown to admit an exact mean-field description for both synaptic and gap-junction interactions. The mean-field model takes a similar form to a standard neural mass model, with an additional dynamical equation to describe the evolution of within-population synchrony. As well as reviewing the origins of this next generation mass model we discuss its extension to describe an idealised spatially extended planar cortex. To emphasise the usefulness of this model for EEG/MEG modelling we show how it can be used to uncover the role of local gap-junction coupling in shaping large scale synaptic waves.
Collapse
Affiliation(s)
- Áine Byrne
- School of Mathematics and Statistics, Science Centre, University College Dublin, South Belfield, Dublin 4, Ireland.
| | - James Ross
- School of Mathematical Sciences, Centre for Mathematical Medicine and Biology, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Rachel Nicks
- School of Mathematical Sciences, Centre for Mathematical Medicine and Biology, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Stephen Coombes
- School of Mathematical Sciences, Centre for Mathematical Medicine and Biology, University of Nottingham, Nottingham, NG7 2RD, UK
| |
Collapse
|
18
|
Obot P, Velíšek L, Velíšková J, Scemes E. The Contribution of Astrocyte and Neuronal Panx1 to Seizures Is Model and Brain Region Dependent. ASN Neuro 2021; 13:17590914211007273. [PMID: 33910381 PMCID: PMC8718119 DOI: 10.1177/17590914211007273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Pannexin1 (Panx1) is an ATP release channel expressed in neurons and astrocytes that plays important roles in CNS physiology and pathology. Evidence for the involvement of Panx1 in seizures includes the reduction of epileptiform activity and ictal discharges following Panx1 channel blockade or deletion. However, very little is known about the relative contribution of astrocyte and neuronal Panx1 channels to hyperexcitability. To this end, mice with global and cell type specific deletion of Panx1 were used in one in vivo and two in vitro seizure models. In the low-Mg2+ in vitro model, global deletion but not cell-type specific deletion of Panx1 reduced the frequency of epileptiform discharges. This reduced frequency of discharges did not impact the overall power spectra obtained from local field potentials. In the in vitro KA model, in contrast, global or cell type specific deletion of Panx1 did not affect the frequency of discharges, but reduced the overall power spectra. EEG recordings following KA-injection in vivo revealed that although global deletion of Panx1 did not affect the onset of status epilepticus (SE), SE onset was delayed in mice lacking neuronal Panx1 and accelerated in mice lacking astrocyte Panx1. EEG power spectral analysis disclosed a Panx1-dependent cortical region effect; while in the occipital region, overall spectral power was reduced in all three Panx1 genotypes; in the frontal cortex, the overall power was not affected by deletion of Panx1. Together, our results show that the contribution of Panx1 to ictal activity is model, cell-type and brain region dependent.
Collapse
Affiliation(s)
- Price Obot
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York, United States
| | - Libor Velíšek
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York, United States.,Department of Neurology, New York Medical College, Valhalla, New York, United States.,Department of Pediatrics, New York Medical College, Valhalla, New York, United States
| | - Jana Velíšková
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York, United States.,Department of Neurology, New York Medical College, Valhalla, New York, United States.,Department of Obstetrics & Gynecology, New York Medical College, Valhalla, New York, United States
| | - Eliana Scemes
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York, United States
| |
Collapse
|
19
|
A Review on Potential Footprints of Ferulic Acid for Treatment of Neurological Disorders. Neurochem Res 2021; 46:1043-1057. [PMID: 33547615 DOI: 10.1007/s11064-021-03257-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 01/16/2021] [Accepted: 01/22/2021] [Indexed: 02/06/2023]
Abstract
Ferulic acid is being screened in preclinical settings to combat various neurological disorders. It is a naturally occurring dietary flavonoid commonly found in grains, fruits, and vegetables such as rice, wheat, oats, tomatoes, sweet corn etc., which exhibits protective effects against a number of neurological diseases such as epilepsy, depression, ischemia-reperfusion injury, Alzheimer's disease, and Parkinson's disease. Ferulic acid prevents and treats different neurological diseases pertaining to its potent anti-oxidative and anti-inflammatory effects, beside modulating unique neuro-signaling pathways. It stays in the bloodstream for longer periods than other dietary polyphenols and antioxidants and easily crosses blood brain barrier. The use of novel drug delivery systems such as solid-lipid nanoparticles (SLNs) or its salt forms (sodium ferulate, ethyl ferulate, and isopentyl ferulate) further enhance its bioavailability and cerebral penetration. Based on reported studies, ferulic acid appears to be a promising molecule for treatment of neurological disorders; however, more preclinical (in vitro and in vivo) mechanism-based studies should be planned and conceived followed by its testing in clinical settings.
Collapse
|
20
|
Abstract
One in three epilepsy cases is drug resistant, and seizures often begin in infancy, when they are life-threatening and when therapeutic options are highly limited. An important tool for prioritizing and validating genes associated with epileptic conditions, which is suitable for large-scale screening, is disease modeling in Drosophila. Approximately two-thirds of disease genes are conserved in Drosophila, and gene-specific fly models exhibit behavioral changes that are related to symptoms of epilepsy. Models are based on behavior readouts, seizure-like attacks and paralysis following stimulation, and neuronal, cell-biological readouts that are in the majority based on changes in nerve cell activity or morphology. In this review, we focus on behavioral phenotypes. Importantly, Drosophila modeling is independent of, and complementary to, other approaches that are computational and based on systems analysis. The large number of known epilepsy-associated gene variants indicates a need for efficient research strategies. We will discuss the status quo of epilepsy disease modelling in Drosophila and describe promising steps towards the development of new drugs to reduce seizure rates and alleviate other epileptic symptoms.
Collapse
Affiliation(s)
- Paul Lasko
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, Netherlands
- Department of Biology, McGill University, Montréal, Québec, Canada
| | - Kevin Lüthy
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| |
Collapse
|
21
|
Rosiles-Abonce A, Rubio C, Taddei E, Rosiles D, Rubio-Osornio M. Antiepileptogenic Effect of Retinoic Acid. Curr Neuropharmacol 2021; 19:383-391. [PMID: 32351181 PMCID: PMC8033965 DOI: 10.2174/1570159x18666200429232104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/17/2020] [Accepted: 04/24/2020] [Indexed: 11/24/2022] Open
Abstract
Retinoic acid, a metabolite of vitamin A, acts through either genomic or nongenomic actions. The genomic action of retinoids exerts effects on gene transcription through interaction with retinoid receptors such as retinoic acid receptors (RARα, β, and γ) and retinoid X receptors (RXRα, β, and γ) that are primarily concentrated in the amygdala, pre-frontal cortex, and hippocampal areas in the brain. In response to retinoid binding, RAR/RXR heterodimers undergo major conformational changes and orchestrate the transcription of specific gene networks. Previous experimental studies have reported that retinoic acid exerts an antiepileptogenic effect through diverse mechanisms, including the modulation of gap junctions, neurotransmitters, long-term potentiation, calcium channels and some genes. To our knowledge, there are no previous or current clinical trials evaluating the use of retinoic acid for seizure control.
Collapse
Affiliation(s)
| | | | | | | | - Moisés Rubio-Osornio
- Address correspondence to this author at the Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico; E-mail:
| |
Collapse
|
22
|
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: 27] [Impact Index Per Article: 6.8] [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
|
23
|
Aquilino MS, Whyte-Fagundes P, Lukewich MK, Zhang L, Bardakjian BL, Zoidl GR, Carlen PL. Pannexin-1 Deficiency Decreases Epileptic Activity in Mice. Int J Mol Sci 2020; 21:ijms21207510. [PMID: 33053775 PMCID: PMC7589538 DOI: 10.3390/ijms21207510] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/02/2020] [Accepted: 10/03/2020] [Indexed: 02/07/2023] Open
Abstract
Objective: Pannexin-1 (Panx1) is suspected of having a critical role in modulating neuronal excitability and acute neurological insults. Herein, we assess the changes in behavioral and electrophysiological markers of excitability associated with Panx1 via three distinct models of epilepsy. Methods Control and Panx1 knockout C57Bl/6 mice of both sexes were monitored for their behavioral and electrographic responses to seizure-generating stimuli in three epilepsy models—(1) systemic injection of pentylenetetrazol, (2) acute electrical kindling of the hippocampus and (3) neocortical slice exposure to 4-aminopyridine. Phase-amplitude cross-frequency coupling was used to assess changes in an epileptogenic state resulting from Panx1 deletion. Results: Seizure activity was suppressed in Panx1 knockouts and by application of Panx1 channel blockers, Brilliant Blue-FCF and probenecid, across all epilepsy models. In response to pentylenetetrazol, WT mice spent a greater proportion of time experiencing severe (stage 6) seizures as compared to Panx1-deficient mice. Following electrical stimulation of the hippocampal CA3 region, Panx1 knockouts had significantly shorter evoked afterdischarges and were resistant to kindling. In response to 4-aminopyridine, neocortical field recordings in slices of Panx1 knockout mice showed reduced instances of electrographic seizure-like events. Cross-frequency coupling analysis of these field potentials highlighted a reduced coupling of excitatory delta–gamma and delta-HF rhythms in the Panx1 knockout. Significance: These results suggest that Panx1 plays a pivotal role in maintaining neuronal hyperexcitability in epilepsy models and that genetic or pharmacological targeting of Panx1 has anti-convulsant effects.
Collapse
Affiliation(s)
- Mark S. Aquilino
- IBME, University of Toronto, 164 College Street, Rosebrugh Building, Room 407, Toronto, ON M5S 3G9, Canada; (B.L.B.); (P.L.C.)
- Krembil Research Institute, University Health Network, 135 Nassau Street, Toronto, ON M5T 1M8, Canada; (M.K.L.); (L.Z.)
- Correspondence:
| | - Paige Whyte-Fagundes
- Department of Biology, York University, 4700 Keele Street, Toronto, ON M5S 3G9, Canada; (P.W.-F.); (G.R.Z.)
| | - Mark K. Lukewich
- Krembil Research Institute, University Health Network, 135 Nassau Street, Toronto, ON M5T 1M8, Canada; (M.K.L.); (L.Z.)
| | - Liang Zhang
- Krembil Research Institute, University Health Network, 135 Nassau Street, Toronto, ON M5T 1M8, Canada; (M.K.L.); (L.Z.)
| | - Berj L. Bardakjian
- IBME, University of Toronto, 164 College Street, Rosebrugh Building, Room 407, Toronto, ON M5S 3G9, Canada; (B.L.B.); (P.L.C.)
| | - Georg R. Zoidl
- Department of Biology, York University, 4700 Keele Street, Toronto, ON M5S 3G9, Canada; (P.W.-F.); (G.R.Z.)
| | - Peter L. Carlen
- IBME, University of Toronto, 164 College Street, Rosebrugh Building, Room 407, Toronto, ON M5S 3G9, Canada; (B.L.B.); (P.L.C.)
- Krembil Research Institute, University Health Network, 135 Nassau Street, Toronto, ON M5T 1M8, Canada; (M.K.L.); (L.Z.)
| |
Collapse
|
24
|
Choi EJ, Palacios-Prado N, Sáez JC, Lee J. Identification of Cx45 as a Major Component of GJs in HeLa Cells. Biomolecules 2020; 10:biom10101389. [PMID: 33003547 PMCID: PMC7650549 DOI: 10.3390/biom10101389] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/23/2020] [Accepted: 09/28/2020] [Indexed: 01/06/2023] Open
Abstract
Gap junctions (GJs) are intercellular channels that connect adjacent cells electrically and metabolically. The iodide-yellow fluorescent protein (I-YFP) gap junctional intercellular communication (GJIC) assay is a recently developed method with high sensitivity. HeLa cells have been widely used as GJ-deficient cells for GJ-related research. Herein, we present evidence showing that HeLa cells have functional GJs comprising connexin (Cx) 45 using the I-YFP GJ assay and CRISPR/Cas9 system. We conducted the I-YFP GJIC assay in HeLa cells, which revealed a weak level of GJIC that could not be detected by the Lucifer yellow scrape-loading assay. The mRNA expression of GJB5 (Cx31.1), GJA1 (Cx43), and GJC1 (Cx45) was detected in HeLa cells by RT-PCR analysis. Knocking out GJC1 (Cx45) abolished GJIC, as analyzed by the I-YFP assay and dual whole-cell patch-clamp assay. These results suggest that HeLa cells express Cx45-based GJs and that the I-YFP GJIC assay can be used for cells with weak GJIC, such as Cx45-expressing HeLa cells. Further, GJC1 (Cx45)-knockout HeLa cells are more suitable as a GJ-null cell model for transfection experiments than wild-type HeLa cells. This experimental design was successfully applied to knock out Cx43 expression and GJIC in A549 lung cancer cells and can thus be used to identify major Cxs in other cell types and to establish GJ assay systems for different Cxs.
Collapse
Affiliation(s)
- Eun Ju Choi
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, Korea;
| | - Nicolás Palacios-Prado
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago 6513677, Chile; (N.P.-P.); (J.C.S.)
| | - Juan C. Sáez
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago 6513677, Chile; (N.P.-P.); (J.C.S.)
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso 2360103, Chile
| | - Jinu Lee
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, Korea;
- Correspondence: ; Tel.: +82-32-749-4161
| |
Collapse
|
25
|
Upregulated and Hyperactivated Thalamic Connexin 43 Plays Important Roles in Pathomechanisms of Cognitive Impairment and Seizure of Autosomal Dominant Sleep-Related Hypermotor Epilepsy with S284L-Mutant α4 Subunit of Nicotinic ACh Receptor. Pharmaceuticals (Basel) 2020; 13:ph13050099. [PMID: 32443400 PMCID: PMC7280967 DOI: 10.3390/ph13050099] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 04/26/2020] [Accepted: 05/08/2020] [Indexed: 01/07/2023] Open
Abstract
To understand the pathomechanism and pathophysiology of autosomal dominant sleep-related hypermotor epilepsy (ADSHE), we studied functional abnormalities of glutamatergic transmission in thalamocortical pathway from reticular thalamic nucleus (RTN), mediodorsal thalamic nucleus (MDTN) to orbitofrontal cortex (OFC) associated with S286L-mutant α4β2-nicotinic acetylcholine receptor (nAChR), and connexin43 (Cx43) hemichannel of transgenic rats bearing rat S286L-mutant Chrna4 gene (S286L-TG), corresponding to the human S284L-mutant CHRNA4 gene using simple Western analysis and multiprobe microdialysis. Cx43 expression in the thalamic plasma membrane fraction of S286L-TG was upregulated compared with that of wild-type. Subchronic administrations of therapeutic-relevant doses of zonisamide (ZNS) and carbamazepine (CBZ) decreased and did not affect Cx43 expression of S286L-TG, respectively. Upregulated Cx43 enhanced glutamatergic transmission during both resting and hyperexcitable stages in S286L-TG. Furthermore, activation of GABAergic transmission RTN-MDTN pathway conversely enhanced, but not inhibited, l-glutamate release in the MDTN via upregulated/activated Cx43. Local administration of therapeutic-relevant concentration of ZNS and CBZ acutely supressed and did not affect glutamatergic transmission in the thalamocortical pathway, respectively. These results suggest that pathomechanisms of ADSHE seizure and its cognitive deficit comorbidity, as well as pathophysiology of CBZ-resistant/ZNS-sensitive ADSHE seizures of patients with S284L-mutation.
Collapse
|
26
|
Giaume C, Naus CC, Sáez JC, Leybaert L. Glial Connexins and Pannexins in the Healthy and Diseased Brain. Physiol Rev 2020; 101:93-145. [PMID: 32326824 DOI: 10.1152/physrev.00043.2018] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Over the past several decades a large amount of data have established that glial cells, the main cell population in the brain, dynamically interact with neurons and thus impact their activity and survival. One typical feature of glia is their marked expression of several connexins, the membrane proteins forming intercellular gap junction channels and hemichannels. Pannexins, which have a tetraspan membrane topology as connexins, are also detected in glial cells. Here, we review the evidence that connexin and pannexin channels are actively involved in dynamic and metabolic neuroglial interactions in physiological as well as in pathological situations. These features of neuroglial interactions open the way to identify novel non-neuronal aspects that allow for a better understanding of behavior and information processing performed by neurons. This will also complement the "neurocentric" view by facilitating the development of glia-targeted therapeutic strategies in brain disease.
Collapse
Affiliation(s)
- Christian Giaume
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France; University Pierre et Marie Curie, Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France; Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile; Instituo de Neurociencias, Centro Interdisciplinario de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile; Physiology Group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Christian C Naus
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France; University Pierre et Marie Curie, Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France; Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile; Instituo de Neurociencias, Centro Interdisciplinario de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile; Physiology Group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Juan C Sáez
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France; University Pierre et Marie Curie, Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France; Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile; Instituo de Neurociencias, Centro Interdisciplinario de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile; Physiology Group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Luc Leybaert
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France; University Pierre et Marie Curie, Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France; Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile; Instituo de Neurociencias, Centro Interdisciplinario de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile; Physiology Group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
27
|
Upregulated Connexin 43 Induced by Loss-of-Functional S284L-Mutant α4 Subunit of Nicotinic ACh Receptor Contributes to Pathomechanisms of Autosomal Dominant Sleep-Related Hypermotor Epilepsy. Pharmaceuticals (Basel) 2020; 13:ph13040058. [PMID: 32235384 PMCID: PMC7243124 DOI: 10.3390/ph13040058] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 01/07/2023] Open
Abstract
To study the pathomechanism and pathophysiology of autosomal dominant sleep-related hypermotor epilepsy (ADSHE), this study determined functional abnormalities of glutamatergic transmission in the thalamocortical motor pathway, from the reticular thalamic nucleus (RTN), motor thalamic nuclei (MoTN) tosecondary motor cortex (M2C) associated with the S286L-mutant α4β2-nicotinic acetylcholine receptor (nAChR) and the connexin43 (Cx43) hemichannel of transgenic rats bearing the rat S286L-mutant Chrna4 gene (S286L-TG), which corresponds to the human S284L-mutant CHRNA4 gene using multiprobe microdialysis, primary cultured astrocytes and a Simple Western system. Expression of Cx43 in the M2C plasma membrane fraction of S286L-TG was upregulated compared with wild-type rats. Subchronic nicotine administration decreased Cx43 expression of wild-type, but did not affect that of S286L-TG; however, zonisamide (ZNS) decreased Cx43 in both wild-type and S286L-TG. Primary cultured astrocytes of wild-type were not affected by subchronic administration of nicotine but was decreased by ZNS. Upregulated Cx43 enhanced glutamatergic transmission during both resting and hyperexcitable stages in S286L-TG. Furthermore, activation of glutamatergic transmission associated with upregulated Cx43 reinforced the prolonged Cx43 hemichannel activation. Subchronic administration of therapeutic-relevant doses of ZNS compensated the upregulation of Cx43 and prolonged reinforced activation of Cx43 hemichannel induced by physiological hyperexcitability during the non-rapid eye movement phase of sleep. The present results support the primary pathomechanisms and secondary pathophysiology of ADSHE seizures of patients with S284L-mutation.
Collapse
|
28
|
Lévesque M, Ragsdale D, Avoli M. Evolving Mechanistic Concepts of Epileptiform Synchronization and their Relevance in Curing Focal Epileptic Disorders. Curr Neuropharmacol 2020; 17:830-842. [PMID: 30479217 PMCID: PMC7052840 DOI: 10.2174/1570159x17666181127124803] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/26/2018] [Accepted: 11/17/2018] [Indexed: 01/01/2023] Open
Abstract
The synchronized activity of neuronal networks under physiological conditions is mirrored by specific oscillatory patterns of the EEG that are associated with different behavioral states and cognitive functions. Excessive synchronization can, however, lead to focal epileptiform activity characterized by interictal and ictal discharges in epileptic patients and animal models. This review focusses on studies that have addressed epileptiform synchronization in temporal lobe regions by employing in vitro and in vivo recording techniques. First, we consider the role of ionotropic and metabotropic excitatory glutamatergic transmission in seizure generation as well as the paradoxical role of GABAA signaling in initiating and perhaps maintaining focal seizure activity. Second, we address non-synaptic mechanisms (which include voltage-gated ionic currents and gap junctions) in the generation of epileptiform synchronization. For each mechanism, we discuss the actions of antiepileptic drugs that are presumably modulating excitatory or inhibitory signaling and voltage-gated currents to prevent seizures in epileptic patients. These findings provide insights into the mechanisms of seizure initiation and maintenance, thus leading to the development of specific pharmacological treatments for focal epileptic disorders.
Collapse
Affiliation(s)
- Maxime Lévesque
- Montreal Neurological Institute, McGill University, Montreal, H3A 2B4 Quebec, Canada
| | - David Ragsdale
- Montreal Neurological Institute, McGill University, Montreal, H3A 2B4 Quebec, Canada
| | - Massimo Avoli
- Montreal Neurological Institute, McGill University, Montreal, H3A 2B4 Quebec, Canada.,Departments of Neurology & Neurosurgery, and of Physiology, McGill University, Montréal, H3A 2B4 Québec, Canada.,Department of Experimental Medicine, Facoltà di Medicina e Odontoiatria, Sapienza University of Rome, 00185 Roma, Italy
| |
Collapse
|
29
|
Activation of Astroglial Connexin is Involved in Concentration-Dependent Double-Edged Sword Clinical Action of Clozapine. Cells 2020; 9:cells9020414. [PMID: 32054069 PMCID: PMC7072131 DOI: 10.3390/cells9020414] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/08/2020] [Accepted: 02/10/2020] [Indexed: 01/07/2023] Open
Abstract
Clozapine (CLZ) is a gold-standard antipsychotic against treatment-refractory schizophrenia, but is one of the most toxic antipsychotic agents. Pharmacological mechanisms of the double-edged sword clinical action of CLZ remain to be clarified. To explore the mechanisms of CLZ, the present study determined the astroglial transmission associated with connexin43 (Cx43), which is the most principal expression in astrocytes and myocardial cells, and expression of Cx43 in primary cultured astrocytes. Both acute and subchronic administrations of CLZ concentration-dependently increased Cx43-associated astroglial release of l-glutamate and d-serine, whereas therapeutic-relevant concentration of CLZ acutely did not affect but subchronically increased astroglial release. In contrast, after the subchronic administration of therapeutic-relevant concentration of valproate (VPA), acute administration of therapeutic-relevant concentration of CLZ drastically increased Cx43-associated astroglial releases. VPA increased Cx43 expression in cytosol fraction without affecting plasma membrane fraction, whereas CLZ increased Cx43 expression in both fractions. Acute administration of therapeutic-relevant concentration of CLZ drastically increased Cx43 expression in the plasma membrane fraction of astrocytes subchronically treated with VPA. The present findings suggest that CLZ-induced the activation of Cx43-associated channel activity and transported Cx43 to plasma membrane, probably contribute to the double-edged sword clinical action of CLZ, such as improvement of cognitive dysfunction and CLZ-induced myocarditis.
Collapse
|
30
|
Mat Nor MN, Rupenthal ID, Green CR, Acosta ML. Connexin Hemichannel Block Using Orally Delivered Tonabersat Improves Outcomes in Animal Models of Retinal Disease. Neurotherapeutics 2020; 17:371-387. [PMID: 31637594 PMCID: PMC7007471 DOI: 10.1007/s13311-019-00786-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Increased Connexin43 hemichannel opening is associated with inflammasome pathway activation and inflammation in a range of pathologies including ocular disorders, such as age-related macular degeneration (AMD) and diabetic retinopathy (DR). In this study, the effect on retinal function and morphology of clinically safe doses of orally delivered tonabersat, a small molecule connexin hemichannel blocker, was investigated in the light-damaged retina animal model of dry AMD and in a spontaneous rat model of DR. Clinical parameters (fundus imaging, optical coherence tomography (OCT), and electroretinography) and inflammatory markers (immunohistochemistry for Iba-1 microglial marker, astrocyte marker glial fibrillary acidic protein, and Connexin43 protein expression) were assessed. Tonabersat treatment reduced inflammation in the retina in parallel with preservation of retinal photoreceptor function when assessed up to 3 months post light damage in the dry AMD model. In the DR model, clinical signs, including the presence of aneurysms confirmed using Evans blue dye perfusion, were reduced after daily tonabersat treatment for 2 weeks. Inflammation was also reduced and retinal electrical function restored. Tonabersat regulates assembly of the inflammasome (NLRP3) through Connexin43 hemichannel block, with the potential to reduce inflammation, restore vascular integrity and improve anatomical along with some functional outcomes in retinal disease.
Collapse
Affiliation(s)
- Mohd Nasir Mat Nor
- School of Optometry and Vision Science and New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
- Faculty of Medicine, University of Sultan Zainal Abidin, Kuala Terengganu, Malaysia
| | - Ilva D Rupenthal
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, and New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
- Department of Ophthalmology and New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Colin R Green
- Department of Ophthalmology and New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Monica L Acosta
- School of Optometry and Vision Science and New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
31
|
Patel DC, Tewari BP, Chaunsali L, Sontheimer H. Neuron-glia interactions in the pathophysiology of epilepsy. Nat Rev Neurosci 2019; 20:282-297. [PMID: 30792501 DOI: 10.1038/s41583-019-0126-4] [Citation(s) in RCA: 234] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Epilepsy is a neurological disorder afflicting ~65 million people worldwide. It is caused by aberrant synchronized firing of populations of neurons primarily due to imbalance between excitatory and inhibitory neurotransmission. Hence, the historical focus of epilepsy research has been neurocentric. However, the past two decades have enjoyed an explosion of research into the role of glia in supporting and modulating neuronal activity, providing compelling evidence of glial involvement in the pathophysiology of epilepsy. The mechanisms by which glia, particularly astrocytes and microglia, may contribute to epilepsy and consequently could be harnessed therapeutically are discussed in this Review.
Collapse
Affiliation(s)
- Dipan C Patel
- Fralin Biomedical Research Institute, Glial Biology in Health, Disease, and Cancer Center, Roanoke, VA, USA
| | - Bhanu P Tewari
- Fralin Biomedical Research Institute, Glial Biology in Health, Disease, and Cancer Center, Roanoke, VA, USA
| | - Lata Chaunsali
- Fralin Biomedical Research Institute, Glial Biology in Health, Disease, and Cancer Center, Roanoke, VA, USA
| | - Harald Sontheimer
- Fralin Biomedical Research Institute, Glial Biology in Health, Disease, and Cancer Center, Roanoke, VA, USA. .,School of Neuroscience, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
32
|
González-Ramírez LR, Mauro AJ. Investigating the role of gap junctions in seizure wave propagation. BIOLOGICAL CYBERNETICS 2019; 113:561-577. [PMID: 31696304 DOI: 10.1007/s00422-019-00809-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 10/13/2019] [Indexed: 06/10/2023]
Abstract
The effect of gap junctions as well as the biological mechanisms behind seizure wave propagation is not completely understood. In this work, we use a simple neural field model to study the possible influence of gap junctions specifically on cortical wave propagation that has been observed in vivo preceding seizure termination. We consider a voltage-based neural field model consisting of an excitatory and an inhibitory population as well as both chemical and gap junction-like synapses. We are able to approximate important properties of cortical wave propagation previously observed in vivo before seizure termination. This model adds support to existing evidence from models and clinical data suggesting a key role of gap junctions in seizure wave propagation. In particular, we found that in this model gap junction-like connectivity determines the propagation of one-bump or two-bump traveling wave solutions with features consistent with the clinical data. For sufficiently increased gap junction connectivity, wave solutions cease to exist. Moreover, gap junction connectivity needs to be sufficiently low or moderate to permit the existence of linearly stable solutions of interest.
Collapse
Affiliation(s)
- Laura R González-Ramírez
- Departamento de Formación Básica Disciplinaria, Instituto Politécnico Nacional, Unidad Profesional Interdisciplinaria de Ingeniería Campus Hidalgo, San Agustín Tlaxiaca, Hidalgo, Mexico.
| | - Ava J Mauro
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
33
|
Shu Y, Zhu C, Zeng M, Zhan Q, Hu Z, Wu X. The protective effect of carbenoxolone on gap junction damage in the hippocampal CA1 area of a temporal lobe epilepsy rat model. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:624. [PMID: 31930025 DOI: 10.21037/atm.2019.11.04] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Astrocytes are one of the most important types of neural cells in the central nervous system (CNS). Dysfunctional gap junction (GJ) communication could play an underlying role in epileptogenesis. Carbenoxolone (CBX) is a conventional chemical GJ blocker, and its target is connexin 43 (Cx43). Previous studies have shown that CBX can inhibit status epilepticus (SE) and spontaneous epileptic seizures (SESs). However, there is little information about the direct interaction between CBX and Cxs in temporal lobe epilepsy (TLE). Methods The behavior of epileptic rats was observed. Moreover, micromorphological changes in the hippocampal cornu ammonis 1 (CA1) area of epileptic rats following CBX injection were determined through transmission electron microscopy (TEM). To illustrate the possible mechanism of these changes, the Western blot method was used. Results After the injection of CBX, the seizure frequency, seizure duration, latency period to the first instance of SES, SESs behavioral score according to a scoring system developed by Velíšková and microstructures in the CA1 area were shown to be improved 60 days after SE by TEM. Furthermore, the dynamic expression patterns of Cx43 and Cx43 phosphorylated at Ser368 continuously declined after the injection of CBX until 60 days after SE. Conclusions CBX may contribute to the improvement of GJ dysfunction during epileptogenesis in the hippocampal CA1 area in a TLE rat model.
Collapse
Affiliation(s)
- Yi Shu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Can Zhu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Min Zeng
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Qiong Zhan
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Zhiping Hu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xiaomei Wu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| |
Collapse
|
34
|
Men C, Wang Z, Zhou L, Qi M, An D, Xu W, Zhan Y, Chen L. Transient receptor potential vanilloid 4 is involved in the upregulation of connexin expression following pilocarpine-induced status epilepticus in mice. Brain Res Bull 2019; 152:128-133. [PMID: 31299321 DOI: 10.1016/j.brainresbull.2019.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/03/2019] [Accepted: 07/05/2019] [Indexed: 10/26/2022]
Abstract
Epilepsy is characterized by spontaneous seizures. Changes in the expression of the connexins (Cxs) have been reported to be involved in epileptogenesis. It has previously been shown that the transient receptor potential vanilloid 4 (TRPV4) plays an important role in the modulation of neuronal excitability, and that application of a TRPV4 antagonist blocks hyperthermia-induced seizures. Accordingly, in the present study, we sought to explore whether TRPV4 is involved in the regulation of Cx expression following pilocarpine-induced status epilepticus (PISE) in mice. We observed that TRPV4 protein levels in hippocampi increased 3 h to 30 d following PISE, peaking 1-3 d after induction, and that pre-application of the TRPV4 antagonist HC-067047 increased the latency to develop SE induced by pilocarpine and reduced the success rate of PISE preparation. We demonstrated that Cx43 protein levels followed a time profile similar to that of TRPV4, and further showed that the increase in Cx43 protein levels on 3 d post-PISE was markedly attenuated by HC-067047. In contrast, the corresponding increase in Cx32 protein levels lagged substantially behind, and these levels were unaffected by HC-067047. Similarly, the TRPV4 agonist GSK1016790A increased the mRNA and protein levels of Cx43, but not those of Cx32. We thus conclude that the upregulation of Cx43 expression by TRPV4 may be involved in the pathophysiology of epilepsy.
Collapse
Affiliation(s)
- Chen Men
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Zhouqing Wang
- Department of Physiology, Nanjing Medical University, Nanjing, PR China
| | - Li Zhou
- Department of Physiology, Nanjing Medical University, Nanjing, PR China
| | - Mengwen Qi
- Department of Physiology, Nanjing Medical University, Nanjing, PR China
| | - Dong An
- Department of Physiology, Nanjing Medical University, Nanjing, PR China
| | - Weixing Xu
- Department of Physiology, Nanjing Medical University, Nanjing, PR China
| | - Yiyang Zhan
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China.
| | - Lei Chen
- Department of Physiology, Nanjing Medical University, Nanjing, PR China; Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Nanjing, PR China.
| |
Collapse
|
35
|
Dossi E, Blauwblomme T, Moulard J, Chever O, Vasile F, Guinard E, Le Bert M, Couillin I, Pallud J, Capelle L, Huberfeld G, Rouach N. Pannexin-1 channels contribute to seizure generation in human epileptic brain tissue and in a mouse model of epilepsy. Sci Transl Med 2019; 10:10/443/eaar3796. [PMID: 29848662 DOI: 10.1126/scitranslmed.aar3796] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 11/02/2017] [Accepted: 05/10/2018] [Indexed: 01/08/2023]
Abstract
Epilepsies are characterized by recurrent seizures, which disrupt normal brain function. Alterations in neuronal excitability and excitation-inhibition balance have been shown to promote seizure generation, yet molecular determinants of such alterations remain to be identified. Pannexin channels are nonselective, large-pore channels mediating extracellular exchange of neuroactive molecules. Recent data suggest that these channels are activated under pathological conditions and regulate neuronal excitability. However, whether pannexin channels sustain or counteract chronic epilepsy in human patients remains unknown. We studied the impact of pannexin-1 channel activation in postoperative human tissue samples from patients with epilepsy displaying epileptic activity ex vivo. These samples were obtained from surgical resection of epileptogenic zones in patients suffering from lesional or drug-resistant epilepsy. We found that pannexin-1 channel activation promoted seizure generation and maintenance through adenosine triphosphate signaling via purinergic 2 receptors. Pharmacological inhibition of pannexin-1 channels with probenecid or mefloquine-two medications currently used for treating gout and malaria, respectively-blocked ictal discharges in human cortical brain tissue slices. Genetic deletion of pannexin-1 channels in mice had anticonvulsant effects when the mice were exposed to kainic acid, a model of temporal lobe epilepsy. Our data suggest a proepileptic role of pannexin-1 channels in chronic epilepsy in human patients and that pannexin-1 channel inhibition might represent an alternative therapeutic strategy for treating lesional and drug-resistant epilepsies.
Collapse
Affiliation(s)
- Elena Dossi
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNR UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, 75005 Paris, France
| | - Thomas Blauwblomme
- Assistance Publique-Hopitaux de Paris (AP-HP), Department of Pediatric Neurosurgery, Necker Hospital, INSERM U1129, Paris Descartes University, PRES Sorbonne Paris Cité, 75015 Paris, France.,INSERM U1129, Paris Descartes University, PRES Sorbonne Paris Cité, CEA, 75015 Paris, France
| | - Julien Moulard
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNR UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, 75005 Paris, France
| | - Oana Chever
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNR UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, 75005 Paris, France
| | - Flora Vasile
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNR UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, 75005 Paris, France
| | - Eleonore Guinard
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNR UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, 75005 Paris, France.,INSERM U1129, Paris Descartes University, PRES Sorbonne Paris Cité, CEA, 75015 Paris, France
| | - Marc Le Bert
- CNRS UMR7355, Experimental and Molecular Immunology and Neurogenetics, 45067 Orléans, France
| | - Isabelle Couillin
- CNRS UMR7355, Experimental and Molecular Immunology and Neurogenetics, 45067 Orléans, France
| | - Johan Pallud
- Department of Neurosurgery, Sainte-Anne Hospital and IMA-BRAIN, INSERM U894, Paris Descartes University, PRES Sorbonne Paris Cité, 75014 Paris, France
| | - Laurent Capelle
- Department of Neurosurgery, University Pierre and Marie Curie, La Pitié-Salpêtrière Hospital, AP-HP, Sorbonne Universités, 75013 Paris, France
| | - Gilles Huberfeld
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNR UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, 75005 Paris, France.,INSERM U1129, Paris Descartes University, PRES Sorbonne Paris Cité, CEA, 75015 Paris, France.,Department of Neurophysiology, La Pitié-Salpetrière Hospital, Sorbonne Universités, University Pierre and Marie Curie, AP-HP, 75013 Paris, France
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNR UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, 75005 Paris, France.
| |
Collapse
|
36
|
Sarrouilhe D, Mesnil M, Dejean C. Targeting Gap Junctions: New Insights into the Treatment of Major Depressive Disorder. Curr Med Chem 2019; 26:3775-3791. [DOI: 10.2174/0929867325666180327103530] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 12/22/2017] [Accepted: 03/21/2018] [Indexed: 01/05/2023]
Abstract
Background:Major depressive disorder (MDD) is a multifactorial chronic and debilitating mood disease with high lifetime prevalence and associated with excess mortality. Treatments for this disease are not effective in all patients showing the need to find new therapeutic targets.Objective:This review aims to update our knowledge on the involvement of astroglial gap junctions and hemichannels in MDD and to show how they have become potential targets for the treatment of this pathology.Methods:The method applied in this review includes a systematic compilation of the relevant literature.Results and Conclusion:The use of rodent models of depression, gene analysis of hippocampal tissues of MDD patients and post-mortem studies on the brains from MDD patients suggest that astrocytic gap junction dysfunction may be a part of MDD etiologies. Chronic antidepressant treatments of rats, rat cultured cortical astrocytes and human astrocytoma cell lines support the hypothesis that the up-regulation of gap junctional coupling between astrocytes could be an underlying mechanism for the therapeutic effect of antidepressants. However, two recent functional studies suggest that connexin43 hemichannel activity is a part of several antidepressants’ mode of action and that astrocyte gap junctional intercellular communication and hemichannels exert different effects on antidepressant drug response. Even if they emerge as new therapeutic targets for new and more active treatments, further studies are needed to decipher the sophisticated and respective role of astrocytic gap junctions and hemichannels in MDD.
Collapse
Affiliation(s)
- Denis Sarrouilhe
- Laboratoire de Physiologie Humaine, Faculte de Medecine et Pharmacie, Universite de Poitiers, 6 rue de la Miletrie, Bat D1, TSA 51115, 86073 Poitiers, Cedex 9, France
| | - Marc Mesnil
- STIM, ERL 7003, CNRS-Universite de Poitiers, Pole Biologie Sante, Bat B36, TSA 51106, 1 rue Georges Bonnet, 86073 Poitiers, Cedex 9, France
| | - Catherine Dejean
- Service Pharmacie, Pavillon Janet, Centre Hospitalier Henri Laborit, 370 avenue Jacques Coeur, 86021 Poitiers Cedex, France
| |
Collapse
|
37
|
Liou JY, Ma H, Wenzel M, Zhao M, Baird-Daniel E, Smith EH, Daniel A, Emerson R, Yuste R, Schwartz TH, Schevon CA. Role of inhibitory control in modulating focal seizure spread. Brain 2019; 141:2083-2097. [PMID: 29757347 DOI: 10.1093/brain/awy116] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 03/04/2018] [Indexed: 11/12/2022] Open
Abstract
Focal seizure propagation is classically thought to be spatially contiguous. However, distribution of seizures through a large-scale epileptic network has been theorized. Here, we used a multielectrode array, wide field calcium imaging, and two-photon calcium imaging to study focal seizure propagation pathways in an acute rodent neocortical 4-aminopyridine model. Although ictal neuronal bursts did not propagate beyond a 2-3-mm region, they were associated with hemisphere-wide field potential fluctuations and parvalbumin-positive interneuron activity outside the seizure focus. While bicuculline surface application enhanced contiguous seizure propagation, focal bicuculline microinjection at sites distant to the 4-aminopyridine focus resulted in epileptic network formation with maximal activity at the two foci. Our study suggests that both classical and epileptic network propagation can arise from localized inhibition defects, and that the network appearance can arise in the context of normal brain structure without requirement for pathological connectivity changes between sites.
Collapse
Affiliation(s)
- Jyun-You Liou
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Hongtao Ma
- Department of Neurological Surgery, Feil Family Brain and Mind Research Institute, Sackler Brain and Spine Institute, Weill Cornell Medical College, New York, NY, USA
| | - Michael Wenzel
- Neurotechnology Center, Department of Biological Sciences, Columbia University, New York, NY, USA; Department of Neuroscience, Columbia University, New York, NY, USA
| | - Mingrui Zhao
- Department of Neurological Surgery, Feil Family Brain and Mind Research Institute, Sackler Brain and Spine Institute, Weill Cornell Medical College, New York, NY, USA
| | - Eliza Baird-Daniel
- Department of Neurological Surgery, Feil Family Brain and Mind Research Institute, Sackler Brain and Spine Institute, Weill Cornell Medical College, New York, NY, USA
| | - Elliot H Smith
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, USA
| | - Andy Daniel
- Department of Neurological Surgery, Feil Family Brain and Mind Research Institute, Sackler Brain and Spine Institute, Weill Cornell Medical College, New York, NY, USA
| | - Ronald Emerson
- Hospital for Special Surgery, Weill Cornell Medical College, 535 East 70th Street, New York, NY, USA
| | - Rafael Yuste
- Neurotechnology Center, Department of Biological Sciences, Columbia University, New York, NY, USA; Department of Neuroscience, Columbia University, New York, NY, USA
| | - Theodore H Schwartz
- Department of Neurological Surgery, Feil Family Brain and Mind Research Institute, Sackler Brain and Spine Institute, Weill Cornell Medical College, New York, NY, USA
| | - Catherine A Schevon
- Department of Neurology, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
38
|
Effects of GLP-1 Receptor Activation on a Pentylenetetrazole-Kindling Rat Model. Brain Sci 2019; 9:brainsci9050108. [PMID: 31091715 PMCID: PMC6562858 DOI: 10.3390/brainsci9050108] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 04/26/2019] [Accepted: 05/06/2019] [Indexed: 12/12/2022] Open
Abstract
Objectives: To study the possible anti-seizure and neuroprotective effect of glucagon like peptide 1 (GLP1) analogue (liraglutide) in a pentylenetetrazole (PTZ) induced kindled rat model and its underlying mechanisms. Methods: Thirty Sprague Dawley rats were allocated into 3 equal groups; i) Normal group: normal rats received normal saline, ii) PTZ (kindling) group: received PTZ (50 mg/Kg intraperitoneally (i.p.)) every other day for 2 weeks and iii) PTZ + GLP1 group: same as the PTZ group but rats received liraglutide (75 µg/kg i.p. daily) for 2 weeks before PTZ injection. Seizure severity score, seizure latency and duration were assessed. Also, the expression of caspase-3 (apoptotic marker) and β-catenin (Wnt pathway) by western blotting, markers of oxidative stress (GSH, CAT and MDA) by biochemical assay and the expression of LC3 (marker of autophagy) and heat shock protein 70 (Hsp70) by immunostaining were assessed in hippocampal regions of brain tissues. Results: PTZ caused a significant increase in Racine score and seizure duration with a significant decrease in seizure latency. These effects were associated with a significant increase in MDA, β-catenin, caspase-3, Hsp70 and LC3 in brain tissues (p < 0.05). Meanwhile, liraglutide treatment caused significant attenuation in PTZ-induced seizures, which were associated with significant improvement in markers of oxidative stress, reduction in LC3, caspase-3 and β-catenin and marked increase in Hsp70 in hippocampal regions (p < 0.05). Conclusion: Activation of GLP1R might have anticonvulsant and neuroprotective effects against PTZ-induced epilepsy. These effects could be due to suppression of oxidative stress, apoptosis and autophagy and upregulation of Hsp70.
Collapse
|
39
|
Wellmann M, Álvarez-Ferradas C, Maturana CJ, Sáez JC, Bonansco C. Astroglial Ca 2+-Dependent Hyperexcitability Requires P2Y 1 Purinergic Receptors and Pannexin-1 Channel Activation in a Chronic Model of Epilepsy. Front Cell Neurosci 2018; 12:446. [PMID: 30542266 PMCID: PMC6277884 DOI: 10.3389/fncel.2018.00446] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 11/06/2018] [Indexed: 12/31/2022] Open
Abstract
Astrocytes from the hippocampus of chronic epileptic rats exhibit an abnormal pattern of intracellular calcium oscillations, characterized by an augmented frequency of long lasting spontaneous Ca2+ transients, which are sensitive to purinergic receptor antagonists but resistant to tetrodotoxin. The above suggests that alterations in astroglial Ca2+-dependent excitability observed in the epileptic tissue could arise from changes in astrocyte-to-astrocyte signaling, which is mainly mediated by purines in physiological and pathological conditions. In spite of that, how purinergic signaling contributes to astrocyte dysfunction in epilepsy remains unclear. Here, we assessed the possible contribution of P2Y1R as well as pannexin1 and connexin43 hemichannels—both candidates for non-vesicular ATP-release—by performing astroglial Ca2+ imaging and dye uptake experiments in hippocampal slices from control and fully kindled rats. P2Y1R blockade with MRS2179 decreased the mean duration of astroglial Ca2+ oscillations by reducing the frequency of slow Ca2+ transients, and thereby restoring the balance between slow (ST) and fast transients (FT) in the kindled group. The potential contribution of astroglial pannexin1 and connexin43 hemichannels as pathways for purine release (e.g., ATP) was assessed through dye uptake experiments. Astrocytes from kindled hippocampi exhibit three-fold more EtBr uptake than controls, whereby pannexin1 hemichannels (Panx1 HCs) accounts for almost all dye uptake with only a slight contribution from connexin43 hemichannels (Cx43 HCs). Confirming its functional involvement, Panx1 HCs inhibition decreased the mean duration of astroglial Ca2+ transients and the frequency of slow oscillations in kindled slices, but had no noticeable effects on the control group. As expected, Cx43 HCs blockade did not have any effects over the mean duration of astroglial Ca2+ oscillations. These findings suggest that P2Y1R and Panx1 HCs play a pivotal role in astroglial pathophysiology, which would explain the upregulation of glutamatergic neurotransmission in the epileptic brain and thus represents a new potential pharmacological target for the treatment of drug-refractory epilepsy.
Collapse
Affiliation(s)
- Mario Wellmann
- Centro de Neurobiología y Plasticidad Cerebral CNPC, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.,Escuela de Fonoaudiología, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Carla Álvarez-Ferradas
- Centro de Neurobiología y Plasticidad Cerebral CNPC, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.,Escuela de Ciencias de la Salud, Universidad Viña del Mar, Valparaíso, Chile
| | - Carola J Maturana
- Departamento de Ciencias Fisiológicas, Facultad Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Juan C Sáez
- Departamento de Ciencias Fisiológicas, Facultad Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Christian Bonansco
- Centro de Neurobiología y Plasticidad Cerebral CNPC, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
40
|
Li Q, Li QQ, Jia JN, Liu ZQ, Zhou HH, Mao XY. Targeting gap junction in epilepsy: Perspectives and challenges. Biomed Pharmacother 2018; 109:57-65. [PMID: 30396092 DOI: 10.1016/j.biopha.2018.10.068] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/08/2018] [Accepted: 10/12/2018] [Indexed: 12/21/2022] Open
Abstract
Gap junctions (GJs) are multiple cellular intercellular connections that allow ions to pass directly into the cytoplasm of neighboring cells. Electrical coupling mediated by GJs plays a role in the generation of highly synchronous electrical activity. Accumulative investigations show that GJs in the brain are involved in the generation, synchronization and maintenance of seizure events. At the same time, GJ blockers exert potent curative potential on epilepsy in vivo or in vitro. This review aims to shed light on the role of GJs in epileptogenesis. Targeting GJs is likely to be served as a novel therapeutic approach on epileptic patients.
Collapse
Affiliation(s)
- Qin Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, Hunan, China
| | - Qiu-Qi Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, Hunan, China
| | - Ji-Ning Jia
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, Hunan, China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, Hunan, China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, Hunan, China
| | - Xiao-Yuan Mao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, Hunan, China.
| |
Collapse
|
41
|
Ran X, Xiang J, Song PP, Jiang L, Liu BK, Hu Y. Effects of gap junctions blockers on fast ripples and connexin in rat hippocampi after status epilepticus. Epilepsy Res 2018; 146:28-35. [DOI: 10.1016/j.eplepsyres.2018.07.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 06/28/2018] [Accepted: 07/20/2018] [Indexed: 12/17/2022]
|
42
|
Walrave L, Pierre A, Albertini G, Aourz N, De Bundel D, Van Eeckhaut A, Vinken M, Giaume C, Leybaert L, Smolders I. Inhibition of astroglial connexin43 hemichannels with TAT-Gap19 exerts anticonvulsant effects in rodents. Glia 2018; 66:1788-1804. [PMID: 29683209 DOI: 10.1002/glia.23341] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 04/04/2018] [Accepted: 04/04/2018] [Indexed: 12/14/2022]
Abstract
Accumulating evidence shows a key function for astrocytic connexin43 (Cx43) signaling in epilepsy. However, the lack of experimental distinction between Cx43 gap junction channels (GJCs) and hemichannels (HCs) has impeded the identification of the exact contribution of either channel configurations to epilepsy. We therefore investigated whether TAT-Gap19, a Cx mimetic peptide that inhibits Cx43 HCs but not the corresponding Cx43 GJCs, influences experimentally induced seizures in rodents. Dye uptake experiments in acute hippocampal slices of mice demonstrated that astroglial Cx43 HCs open in response to the chemoconvulsant pilocarpine and this was inhibited by TAT-Gap19. In vivo, pilocarpine-induced seizures as well as the accompanying increase in D-serine microdialysate levels were suppressed by Cx43 HC inhibition. Moreover, the anticonvulsant action of TAT-Gap19 was reversed by exogenous D-serine administration, suggesting that Cx43 HC inhibition protects against seizures by lowering extracellular D-serine levels. The anticonvulsive properties of Cx43 HC inhibition were further confirmed in electrical seizure mouse models, i.e. an acute 6 Hertz (Hz) model of refractory seizures and a chronic 6 Hz corneal kindling model. Collectively, these results indicate that Cx43 HCs play a role in seizures and underscore their potential as a novel and druggable target in epilepsy treatment.
Collapse
Affiliation(s)
- Laura Walrave
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Anouk Pierre
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Giulia Albertini
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Najat Aourz
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Dimitri De Bundel
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Ann Van Eeckhaut
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Christian Giaume
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, Paris, 75005, France
| | - Luc Leybaert
- Physiology group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185, Ghent, 9000, Belgium
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| |
Collapse
|
43
|
Abstract
Major depressive disorder (MDD) is a chronic and debilitating illness that affects over 350 million people worldwide; however, current treatments have failed to cure or prevent the progress of depression. Increasing evidence suggests a crucial role for connexins in MDD. In this review, we have summarised recent accomplishments regarding the role of connexins, gap junctions, and hemichannels in the aetiology of MDD, and discussed the limitations of current research. A blockage of gap junctions or hemichannels induces depressive behaviour. Possible underlying mechanisms include the regulation of neurosecretory functions and synaptic activity by gap junctions and hemichannels. Gap junctions are functionally inhibited under stress conditions. Conversely, hemichannel permeability is increased. Antidepressants inhibit hemichannel permeability; however, they have contrasting effects on the function of gap junctions under normal conditions and can protect them against stress. In conclusion, the blockage of hemichannels concurrent with improvements in gap junction functionality might be potential targets for depression treatment.
Collapse
Affiliation(s)
- Cong-Yuan Xia
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhen-Zhen Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Tohru Yamakuni
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China.
| |
Collapse
|
44
|
Sheng J, Liu S, Qin H, Li B, Zhang X. Drug-Resistant Epilepsy and Surgery. Curr Neuropharmacol 2018; 16:17-28. [PMID: 28474565 PMCID: PMC5771378 DOI: 10.2174/1570159x15666170504123316] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/23/2017] [Accepted: 04/25/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Epilepsy is a chronic brain disease that is caused by various factors and characterized by recurrent, episodic and temporary central nervous system dysfunction which results due to excessive discharge of brain neurons. In the past decades, despite the continuous development of antiepileptic drugs, there are still many patients with epilepsy progressing to drugresistant epilepsy. Currently, surgical treatment is one of important way to cure drug-resistant epilepsy. METHODS Data were collected from Web of Science, Medline, Pubmed, through searching of these keywords: "surgery" and "drug-resistant epilepsy". RESULTS An increasing number of studies have shown that surgery plays an important role in the treatment of drug-resistant epilepsy. Moreover, the comprehensive treatment mainly based on surgery can achieve the remission and even cure of drug-resistant epilepsy. CONCLUSION In this review, we discuss the pathogenesis of drug-resistant epilepsy and the comprehensive treatment mainly based on surgery; this review may provide a reference for the clinical treatment of drug-resistant epilepsy.
Collapse
Affiliation(s)
- Jiyao Sheng
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun130041, P.R. China
| | - Shui Liu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun130041, P.R. China
| | - Hanjiao Qin
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun130041, P.R. China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun130041, P.R. China
| | - Xuewen Zhang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun130041, P.R. China
| |
Collapse
|
45
|
Ren Q, Wang ZZ, Chu SF, Xia CY, Chen NH. Gap junction channels as potential targets for the treatment of major depressive disorder. Psychopharmacology (Berl) 2018; 235:1-12. [PMID: 29178009 DOI: 10.1007/s00213-017-4782-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 11/05/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Major depressive disorder (MDD) remains a major public health problem worldwide. The association between MDD and the dysfunction of gap junction channels (GJCs) in glial cells, especially astrocytes, is still controversial. OBJECTIVE This review provides an overview of the role of astrocyte GJCs in LMDD. RESULTS Exposure to chronic unpredictable stress caused a reduction in connexin expression in the rat prefrontal cortex, a result that is consistent with clinical findings reported in postmortem studies of brains from MDD patients. Chronic antidepressant treatment in these rats increased the expression of connexins. However, pharmacological GJC blockade in normal rodents decreased connexin expression and caused depressive-like behaviors. Furthermore, GJC dysfunction affects electrical conductance, metabolic coupling and secondary messengers, and inflammatory responses, which are consistent with current hypotheses on MDD. All these results provide a comprehensive overview of the neurobiology of MDD. CONCLUSION This review supports the hypothesis that the regulation of GJCs between astrocytes could be an underlying mechanism for the therapeutic effect of antidepressants.
Collapse
Affiliation(s)
- Qian Ren
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Zhen-Zhen Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shi-Feng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Cong-Yuan Xia
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China. .,College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| |
Collapse
|
46
|
Barros-Barbosa AR, Oliveira Â, Lobo MG, Cordeiro JM, Correia-de-Sá P. Under stressful conditions activation of the ionotropic P2X7 receptor differentially regulates GABA and glutamate release from nerve terminals of the rat cerebral cortex. Neurochem Int 2017; 112:81-95. [PMID: 29154812 DOI: 10.1016/j.neuint.2017.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 11/08/2017] [Accepted: 11/10/2017] [Indexed: 01/08/2023]
Abstract
γ-Aminobutyric acid (GABA) and glutamate (Glu) are the main inhibitory and excitatory neurotransmitters in the central nervous system (CNS), respectively. Fine tuning regulation of extracellular levels of these amino acids is essential for normal brain activity. Recently, we showed that neocortical nerve terminals from patients with epilepsy express higher amounts of the non-desensitizing ionotropic P2X7 receptor. Once activated by ATP released from neuronal cells, the P2X7 receptor unbalances GABAergic vs. glutamatergic neurotransmission by differentially interfering with GABA and Glu uptake. Here, we investigated if activation of the P2X7 receptor also affects [3H]GABA and [14C]Glu release measured synchronously from isolated nerve terminals (synaptosomes) of the rat cerebral cortex. Data show that activation of the P2X7 receptor consistently increases [14C]Glu over [3H]GABA release from cortical nerve terminals, but the GABA/Glu ratio depends on extracellular Ca2+ concentrations. While the P2X7-induced [3H]GABA release is operated by a Ca2+-dependent pathway when external Ca2+ is available, this mechanism shifts towards the reversal of the GAT1 transporter in low Ca2+ conditions. A different scenario is verified regarding [14C]Glu outflow triggered by the P2X7 receptor, since the amino acid seems to be consistently released through the recruitment of connexin-containing hemichannels upon P2X7 activation, both in the absence and in the presence of external Ca2+. Data from this study add valuable information suggesting that ATP, via P2X7 activation, not only interferes with the high-affinity uptake of GABA and Glu but actually favors the release of these amino acids through distinct molecular mechanisms amenable to differential therapeutic control.
Collapse
Affiliation(s)
- Aurora R Barros-Barbosa
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
| | - Ângela Oliveira
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
| | - M Graça Lobo
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
| | - J Miguel Cordeiro
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal.
| |
Collapse
|
47
|
Sarrouilhe D, Dejean C, Mesnil M. Connexin43- and Pannexin-Based Channels in Neuroinflammation and Cerebral Neuropathies. Front Mol Neurosci 2017; 10:320. [PMID: 29066951 PMCID: PMC5641369 DOI: 10.3389/fnmol.2017.00320] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/20/2017] [Indexed: 12/19/2022] Open
Abstract
Connexins (Cx) are largely represented in the central nervous system (CNS) with 11 Cx isoforms forming intercellular channels. Moreover, in the CNS, Cx43 can form hemichannels (HCs) at non-junctional membrane as does the related channel-forming Pannexin1 (Panx1) and Panx2. Opening of Panx1 channels and Cx43 HCs appears to be involved in inflammation and has been documented in various CNS pathologies. Over recent years, evidence has accumulated supporting a link between inflammation and cerebral neuropathies (migraine, Alzheimer’s disease (AD), Parkinson’s disease (PD), major depressive disorder, autism spectrum disorder (ASD), epilepsy, schizophrenia, bipolar disorder). Involvement of Panx channels and Cx43 HCs has been also proposed in pathophysiology of neurological diseases and psychiatric disorders. Other studies showed that following inflammatory injury of the CNS, Panx1 activators are released and prolonged opening of Panx1 channels triggers neuronal death. In neuropsychiatric diseases, comorbidities are frequently present and can aggravate the symptoms and make therapeutic management more complex. The high comorbidity between some neuropathies can be partially understood by the fact that these diseases share a common etiology involving inflammatory pathways and Panx1 channels or Cx43 HCs. Thus, anti-inflammatory therapy opens perspectives of targets for new treatments and could have real potential in controlling a cerebral neuropathy and some of its comorbidities. The purpose of this mini review is to provide information of our knowledge on the link between Cx43- and Panx-based channels, inflammation and cerebral neuropathies.
Collapse
Affiliation(s)
- Denis Sarrouilhe
- Laboratoire de Physiologie Humaine, Faculté de Médecine et Pharmacie, Université de Poitiers, Poitiers, France
| | - Catherine Dejean
- Service Pharmacie, Pavillon Janet, Centre Hospitalier Henri Laborit, Poitiers, France
| | - Marc Mesnil
- STIM Laboratory, ERL 7368-CNRS, Université de Poitiers, Pôle Biologie Santé, Poitiers, France
| |
Collapse
|
48
|
Dong H, Zhou XW, Wang X, Yang Y, Luo JW, Liu YH, Mao Q. Complex role of connexin 43 in astrocytic tumors and possible promotion of glioma‑associated epileptic discharge (Review). Mol Med Rep 2017; 16:7890-7900. [PMID: 28983585 DOI: 10.3892/mmr.2017.7618] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 06/19/2017] [Indexed: 02/05/2023] Open
Abstract
Connexin (Cx)43 is a multifunction protein which forms gap junction channels and hemi‑channels. It also contains abundant binding domains which possess the ability to interact with certain Cx43‑associated proteins and therefore serve a fundamental role in various physiological and pathological functions. However, the understanding of the association between cancer and Cx43 along with Cx43‑gap junctions (GJ) remains unclear. All available data illustrate that Cx43 and its associated GJ serve important functions in cancers. The expression levels of Cx43 demonstrate a downward trend and an increase in the levels of malignancy, particularly in astrocytomas. The GJ intercellular communication activity in glioma cells can be adjusted via Cx43 phosphorylation and through the combination of Cx43 and its associated protein. Available evidence reveals Cx43 as a tumor‑inhibiting factor that suppresses glioma growth and proliferation. However, its mechanism is also regarded as complicated and ambiguous. Furthermore, it is apparent that Cx43‑GJ and the carboxyl tail may contribute to glioma growth and proliferation too. However, this valuable role could be weakened by its effects on migration and invasiveness. The detailed mechanism remains unclear and full of controversies. Cx43 can enhance the motor ability and invasiveness of astrocytic glioma cells. It is also able to influence glioma cells to detach from the tumor core to the peritumoral neocortex. This peritumoral region has recently been regarded as the basic focus of glioma‑associated seizure. Thus, Cx43 may take part in the onset and development of glioma‑associated epileptic discharge. In addition, change and increase of Cx43 expression in GJs has been observed in seizure perilesional tissue, which is associated with brain tumors. Cx43 or GJ/hemi‑channels exert enduring effects in the promotion of glioma‑associated epileptic release through direct mass effects and change of the tumor microenvironment. However, there are still a number of issues concerning this aspect that require further exploration. Cx43, as a potential treatment target against this incurable disease and its common symptom of epilepsy, requires further investigation.
Collapse
Affiliation(s)
- Hui Dong
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xing-Wang Zhou
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiang Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yuan Yang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jie-Wen Luo
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yan-Hui Liu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qing Mao
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
49
|
Ni X, Wang A, Zhang L, Shan LY, Zhang HC, Li L, Si JQ, Luo J, Li XZ, Ma KT. Up-regulation of gap junction in peripheral blood T lymphocytes contributes to the inflammatory response in essential hypertension. PLoS One 2017; 12:e0184773. [PMID: 28910394 PMCID: PMC5599050 DOI: 10.1371/journal.pone.0184773] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 08/30/2017] [Indexed: 12/22/2022] Open
Abstract
Inflammation has been shown to play an important role in the mechanisms involved in the pathogenesis of hypertension. Connexins (Cxs)-based gap junction channels (GJCs) or hemichannels (HCs) are involved in the maintenance of homeostasis in the immune system. However, the role of Cx43-based channels in T-lymphocytes in mediating the immune response in essential hypertension is not fully understand. The present study was designed to investigate the role of Cxs-based channels in T lymphocytes in the regulation of hypertension-mediated inflammation. The surface expressions of T lymphocyte subtypes, Cx40/Cx43, and inflammatory cytokines (IFN-γ (interferon-gamma) and TNF-ɑ (tumor necrosis factor alpha)) in T cells, as well as gap junction communication of peripheral blood lymphocytes from essential hypertensive patients (EHs) and normotensive healthy subjects (NTs) were detected by flow cytometry. Expression levels and phosphorylation of Cx43 protein in peripheral blood lymphocytes of EHs and NTs were analyzed by Western blot. The proliferation rate of peripheral blood mononuclear cells (PBMCs) after treatment with a Cxs inhibitor was examined by a CCK-8 assay. The levels of inflammatory cytokines were detected using ELISA. Within the CD3+ T cell subsets, we found a significant trend toward an increase in the percentage of CD4+ T cells and CD4+/CD8+ ratio as well as in serum levels of IFN-γ and TNF-ɑ in the peripheral blood of EHs compared with those in NTs. Moreover, the peripheral blood lymphocytes of EH patients exhibited enhanced GJCs formation, increased Cx43 protein level and Cx43 phosphorylation at Ser368, and a significant increase in Cx40/Cx43 surface expressions levels in CD4+ or CD8+ T lymphocytes. Cx43-based channel inhibition by a mimetic peptide greatly reduced the exchange of dye between lymphocytes, proliferation of stimulated lymphocytes and the pro-inflammatory cytokine levels of EHs and NTs. Our data suggest that Cx40/Cx43-based channels in lymphocytes may be involved in the regulation of T lymphocyte proliferation and the production of pro-inflammatory cytokines, which contribute to the hypertensive inflammatory response.
Collapse
Affiliation(s)
- Xin Ni
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, China.,Key Laboratory of Xingjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang, China
| | - Ai Wang
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, China.,Key Laboratory of Xingjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang, China
| | - Liang Zhang
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, China.,Key Laboratory of Xingjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang, China
| | - Li-Ya Shan
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, China.,Key Laboratory of Xingjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang, China
| | - Hai-Chao Zhang
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, China.,Key Laboratory of Xingjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang, China
| | - Li Li
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, China.,Key Laboratory of Xingjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang, China
| | - Jun-Qiang Si
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, China.,Key Laboratory of Xingjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang, China
| | - Jian Luo
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xin-Zhi Li
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, China.,Department of Pathophysiology, Medical College of Shihezi University, Shihezi, Xinjiang, China
| | - Ke-Tao Ma
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, China.,Key Laboratory of Xingjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|
50
|
Zhang H, Cohen AE. Optogenetic Approaches to Drug Discovery in Neuroscience and Beyond. Trends Biotechnol 2017; 35:625-639. [PMID: 28552428 PMCID: PMC5495001 DOI: 10.1016/j.tibtech.2017.04.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/08/2017] [Accepted: 04/10/2017] [Indexed: 12/11/2022]
Abstract
Recent advances in optogenetics have opened new routes to drug discovery, particularly in neuroscience. Physiological cellular assays probe functional phenotypes that connect genomic data to patient health. Optogenetic tools, in particular tools for all-optical electrophysiology, now provide a means to probe cellular disease models with unprecedented throughput and information content. These techniques promise to identify functional phenotypes associated with disease states and to identify compounds that improve cellular function regardless of whether the compound acts directly on a target or through a bypass mechanism. This review discusses opportunities and unresolved challenges in applying optogenetic techniques throughout the discovery pipeline - from target identification and validation, to target-based and phenotypic screens, to clinical trials.
Collapse
Affiliation(s)
- Hongkang Zhang
- Department of Chemistry and Chemical Biology, Howard Hughes Medical Institute, Department of Physics, Harvard University, Cambridge, MA 02138, USA
| | - Adam E Cohen
- Department of Chemistry and Chemical Biology, Howard Hughes Medical Institute, Department of Physics, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|