1
|
Chen J, Li Y, Huo X, Huang Z, Li S, Cao W, Zhou C, Xu Y. Targeted suppression of BRD7 with BI-9564 prevents seizure behaviors in pentylenetetrazol and pilocarpine-induced mouse model of epilepsy. Behav Brain Res 2025; 485:115549. [PMID: 40120945 DOI: 10.1016/j.bbr.2025.115549] [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: 02/04/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Epilepsy is a serious neurological disorder, but its underlying cellular and molecular mechanisms remains incomplete. As a member of the bromodomain-containing protein (BCP) family, BRD7 has been implicated in a variety of cellular processes, including chromatin remodeling, transcriptional regulation, and cell cycle progression. However, the role of BRD7 in epilepsy in vivo is still poorly understood. In the present study, we found that pentylenetetrazole (PTZ)-induced epilepsy increased hippocampal BRD7, which was mainly localized in neurons. In addition, the enhanced expression of hippocampal BRD7 was normalized by using the anti-epilepsy drug valproic acid (VPA). Furthermore, we identified that the BRD7 inhibitor BI-9564 could dose dependently alleviated the seizure behavior in PTZ treated mice, which was also validated in pilocarpine mouse model. Mechanistically, the anti-seizure effect of BI- 9564 might be due to its negative-regulation of hippocampal TRPV4 that downregulated neuronal over-excitability. Importantly, BRD7 blockade retained its antiepileptic activity over chronic dosing that was not related to psychomotor or cognitive effects. To our knowledge, these results are the first evidence to demonstrate that BRD7 inhibitor can down-regulate neuronal over-excitation caused by epilepsy possible by regulating TRPV4. Targeting BRD7 through the development of selective inhibitors may lead to disease-modifying therapies that reduce seizure behavior.
Collapse
Affiliation(s)
- Jie Chen
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yujia Li
- Institute of Neuroscience, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiaofei Huo
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Ziqiang Huang
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Suyun Li
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Wenyu Cao
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Cuilan Zhou
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - Yang Xu
- Institute of Neuroscience, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
2
|
Liang H, Qin L, Feng R, Shim J, Huang X, Xu X, Zhao D, Yu Z, Boczek T, Li M, Tong Y, Huang J, Gao Q, Wang L, Cao X, Liu D, Du K, Xu J, Zhao Y, Wang W, Seehus CR, Zhao W, Guo F. Increased Na V1.2 expression and its interaction with CaM contribute to the hyperexcitability induced by prolonged inhibition of CaMKII. Epilepsia 2025. [PMID: 40119845 DOI: 10.1111/epi.18377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 03/24/2025]
Abstract
OBJECTIVE Dysfunction of calcium/calmodulin (CaM)-dependent kinase II (CaMKII) has been involved in hyperexcitability-related disorders including epilepsy. However, the relationship between CaMKII and neuronal excitability remains unclear. METHODS Neuronal excitability was detected in vivo and in vitro by electroencephalography (EEG), patch clamp and multi-electrode array (MEA), respectively. Next, we assessed the currents of voltage-gated sodium channels (VGSCs) by patch clamp, and mRNA and protein expressions of VGSCs were determined by real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and western blot, respectively. Meanwhile, the association between the nuclear receptor subfamily 4 group A member 2 (NR4A2) and promoters of Scn2a, was determined by chromatin immunoprecipitation (ChIP)-qPCR. In addition, we utilized co-immunoprecipitation (Co-IP), immunofluorescence labeling, and pull-down to determine the interaction between VGSCs and CaM. RESULTS Prolonged CaMKII inhibition by KN93, an inhibitor of CaMKII, for 24 h and CaMKII knockdown induced more seizure-like events in Wistar rats, TRM rats and C57BL/6 mice, and led to hyperexcitability in primary hippocampal neurons and human induced-pluripotent stem cell (hiPSC)-derived cortical neurons. In addition, prolonged CaMKII inhibition resulted in elevated persistent sodium current (INaP)/transient sodium current (INaT) and increased mRNA and protein expressions of NaV1.2. Meanwhile, prolonged CaMKII inhibition by KN93 decreased NR4A2 expression and contributed to a transcriptional repression role of NR4A2 in Scn2a regulation, leading to increased NaV1.2 expression. Moreover, an increased interaction between NaV1.2 and CaM was attributable to enhanced binding of CaM to the isoleucine-glutamine (IQ) domain at the C-terminus of the NaV1.2 channel, which may also lead to the potentiation in INaP/INaT and channel activity. Furthermore, a peptide that antagonized CaM binding to NaV1.2 IQ domain (ACNp) rescued hyperexcitability following prolonged CaMKII inhibition. SIGNIFICANCE We unveiled that prolonged CaMKII inhibition induced hyperexcitability through increasing the expression of NaV1.2 and its association with CaM. Thus, our study uncovers a novel signaling mechanism by which CaMKII maintains appropriate neuronal excitability.
Collapse
Affiliation(s)
- Hongyue Liang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Ling Qin
- Department of Physiology, China Medical University, Shenyang, China
| | - Rui Feng
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Jaehoon Shim
- Department of Neurobiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Xuan Huang
- Department of Neurobiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Xiaoxue Xu
- Department of Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Dongyi Zhao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Zhiyi Yu
- Division of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland
| | - Meixuan Li
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Yu Tong
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Junwei Huang
- Department of Neurobiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Qinghua Gao
- Department of Developmental Cell Biology, China Medical University, Shenyang, China
| | - Li Wang
- Department of Developmental Cell Biology, China Medical University, Shenyang, China
| | - Xinyu Cao
- Department of Developmental Cell Biology, China Medical University, Shenyang, China
| | - Dongxin Liu
- Department of Developmental Cell Biology, China Medical University, Shenyang, China
| | - Ke Du
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Jianjun Xu
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Yue Zhao
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang City, Liaoning Province, China
| | - Wuyang Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Corey Ray Seehus
- Department of Neurobiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Weidong Zhao
- Department of Developmental Cell Biology, China Medical University, Shenyang, China
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang City, Liaoning Province, China
| | - Feng Guo
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
3
|
Xie C, Kessi M, Yin F, Peng J. Roles of KCNA2 in Neurological Diseases: from Physiology to Pathology. Mol Neurobiol 2024; 61:8491-8517. [PMID: 38517617 DOI: 10.1007/s12035-024-04120-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 03/10/2024] [Indexed: 03/24/2024]
Abstract
Potassium voltage-gated channel subfamily a member 2 (Kv1.2, encoded by KCNA2) is highly expressed in the central and peripheral nervous systems. Based on the patch clamp studies, gain-of function (GOF), loss-of-function (LOF), and a mixed type (GOF/LOF) variants can cause different conditions/disorders. KCNA2-related neurological diseases include epilepsy, intellectual disability (ID), attention deficit/hyperactive disorder (ADHD), autism spectrum disorder (ASD), pain as well as autoimmune and movement disorders. Currently, the molecular mechanisms for the reported variants in causing diverse disorders are unknown. Consequently, this review brings up to date the related information regarding the structure and function of Kv1.2 channel, expression patterns, neuronal localizations, and tetramerization as well as important cell and animal models. In addition, it provides updates on human genetic variants, genotype-phenotype correlations especially highlighting the deep insight into clinical prognosis of KCNA2-related developmental and epileptic encephalopathy, mechanisms, and the potential treatment targets for all KCNA2-related neurological disorders.
Collapse
Affiliation(s)
- Changning Xie
- Department of Pediatrics, Xiangya Hospital, Central South University, Xiangya Road 87, Hunan, Changsha, 410008, China
| | - Miriam Kessi
- Department of Pediatrics, Xiangya Hospital, Central South University, Xiangya Road 87, Hunan, Changsha, 410008, China
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Xiangya Road 87, Hunan, Changsha, 410008, China
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Xiangya Road 87, Hunan, Changsha, 410008, China.
- Hunan Intellectual and Development Disabilities Research Center, Hunan, Changsha, 410008, China.
| |
Collapse
|
4
|
Zeng ML, Kong S, Chen TX, Peng BW. Transient Receptor Potential Vanilloid 4: a Double-Edged Sword in the Central Nervous System. Mol Neurobiol 2023; 60:1232-1249. [PMID: 36434370 DOI: 10.1007/s12035-022-03141-6] [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: 08/09/2022] [Accepted: 11/17/2022] [Indexed: 11/26/2022]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a nonselective cation channel that can be activated by diverse stimuli, such as heat, mechanical force, hypo-osmolarity, and arachidonic acid metabolites. TRPV4 is widely expressed in the central nervous system (CNS) and participates in many significant physiological processes. However, accumulative evidence has suggested that deficiency, abnormal expression or distribution, and overactivation of TRPV4 are involved in pathological processes of multiple neurological diseases. Here, we review the latest studies concerning the known features of this channel, including its expression, structure, and its physiological and pathological roles in the CNS, proposing an emerging therapeutic strategy for CNS diseases.
Collapse
Affiliation(s)
- Meng-Liu Zeng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Rd185#, Wuhan, 430071, Hubei, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Shuo Kong
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Rd185#, Wuhan, 430071, Hubei, China
| | - Tao-Xiang Chen
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Rd185#, Wuhan, 430071, Hubei, China
| | - Bi-Wen Peng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Rd185#, Wuhan, 430071, Hubei, China.
| |
Collapse
|
5
|
Zou Y, Zhang M, Wu Q, Zhao N, Chen M, Yang C, Du Y, Han B. Activation of transient receptor potential vanilloid 4 is involved in pressure overload-induced cardiac hypertrophy. eLife 2022; 11:e74519. [PMID: 35731090 PMCID: PMC9224988 DOI: 10.7554/elife.74519] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 06/02/2022] [Indexed: 11/13/2022] Open
Abstract
Previous studies, including our own, have demonstrated that transient receptor potential vanilloid 4 (TRPV4) is expressed in hearts and implicated in cardiac remodeling and dysfunction. However, the effects of TRPV4 on pressure overload-induced cardiac hypertrophy remain unclear. In this study, we found that TRPV4 expression was significantly increased in mouse hypertrophic hearts, human failing hearts, and neurohormone-induced hypertrophic cardiomyocytes. Deletion of TRPV4 attenuated transverse aortic constriction (TAC)-induced cardiac hypertrophy, cardiac dysfunction, fibrosis, inflammation, and the activation of NFκB - NOD - like receptor pyrin domain-containing protein 3 (NLRP3) in mice. Furthermore, the TRPV4 antagonist GSK2193874 (GSK3874) inhibited cardiac remodeling and dysfunction induced by TAC. In vitro, pretreatment with GSK3874 reduced the neurohormone-induced cardiomyocyte hypertrophy and intracellular Ca2+ concentration elevation. The specific TRPV4 agonist GSK1016790A (GSK790A) triggered Ca2+ influx and evoked the phosphorylation of Ca2+/calmodulin-dependent protein kinase II (CaMKII). But these effects were abolished by removing extracellular Ca2+ or GSK3874. More importantly, TAC or neurohormone stimulation-induced CaMKII phosphorylation was significantly blocked by TRPV4 inhibition. Finally, we show that CaMKII inhibition significantly prevented the phosphorylation of NFκB induced by GSK790A. Our results suggest that TRPV4 activation contributes to pressure overload-induced cardiac hypertrophy and dysfunction. This effect is associated with upregulated Ca2+/CaMKII mediated activation of NFκB-NLRP3. Thus, TRPV4 may represent a potential therapeutic drug target for cardiac hypertrophy and dysfunction after pressure overload.
Collapse
Affiliation(s)
- Yan Zou
- Department of Cardiology, Xuzhou Central HospitalXuzhouChina
- Xuzhou Institute of Cardiovascular Disease, Xuzhou Central HospitalXuzhouChina
| | - Miaomiao Zhang
- Department of Cardiology, Xuzhou Central HospitalXuzhouChina
| | - Qiongfeng Wu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Ning Zhao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Minwei Chen
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Cui Yang
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Yimei Du
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Bing Han
- Department of Cardiology, Xuzhou Central HospitalXuzhouChina
| |
Collapse
|
6
|
Meza RC, Ancatén-González C, Chiu CQ, Chávez AE. Transient Receptor Potential Vanilloid 1 Function at Central Synapses in Health and Disease. Front Cell Neurosci 2022; 16:864828. [PMID: 35518644 PMCID: PMC9062234 DOI: 10.3389/fncel.2022.864828] [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: 01/28/2022] [Accepted: 03/29/2022] [Indexed: 12/11/2022] Open
Abstract
The transient receptor potential vanilloid 1 (TRPV1), a ligand-gated nonselective cation channel, is well known for mediating heat and pain sensation in the periphery. Increasing evidence suggests that TRPV1 is also expressed at various central synapses, where it plays a role in different types of activity-dependent synaptic changes. Although its precise localizations remain a matter of debate, TRPV1 has been shown to modulate both neurotransmitter release at presynaptic terminals and synaptic efficacy in postsynaptic compartments. In addition to being required in these forms of synaptic plasticity, TRPV1 can also modify the inducibility of other types of plasticity. Here, we highlight current evidence of the potential roles for TRPV1 in regulating synaptic function in various brain regions, with an emphasis on principal mechanisms underlying TRPV1-mediated synaptic plasticity and metaplasticity. Finally, we discuss the putative contributions of TRPV1 in diverse brain disorders in order to expedite the development of next-generation therapeutic treatments.
Collapse
Affiliation(s)
- Rodrigo C Meza
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Carlos Ancatén-González
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Chiayu Q Chiu
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Andrés E Chávez
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
7
|
Du K, He M, Zhao D, Wang Y, Ma C, Liang H, Wang W, Min D, Xue L, Guo F. Mechanism of cell death pathways in status epilepticus and related therapeutic agents. Biomed Pharmacother 2022; 149:112875. [PMID: 35367755 DOI: 10.1016/j.biopha.2022.112875] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/15/2022] [Accepted: 03/23/2022] [Indexed: 11/30/2022] Open
Abstract
The most severe form of epilepsy, status epilepticus (SE), causes brain damage and results in the development of recurring seizures. Currently, the management of SE remains a clinical challenge because patients do not respond adequately to conventional treatments. Evidence suggests that neural cell death worsens the occurrence and progression of SE. The main forms of cell death are apoptosis, necroptosis, pyroptosis, and ferroptosis. Herein, these mechanisms of neuronal death in relation to SE and the alleviation of SE by potential modulators that target neuronal death have been reviewed. An understanding of these pathways and their possible roles in SE may assist in the development of SE therapies and in the discovery of new agents.
Collapse
Affiliation(s)
- Ke Du
- Department of Pharmacology, School of Pharmaceutical Science, China Medical University, Shenyang 110001, China
| | - Miao He
- Department of Pharmaceutical Toxicology, School of Pharmaceutical Science, China Medical University, Shenyang 110001, China
| | - Dongyi Zhao
- Department of Pharmaceutical Toxicology, School of Pharmaceutical Science, China Medical University, Shenyang 110001, China
| | - Yuting Wang
- Department of Pharmaceutical Toxicology, School of Pharmaceutical Science, China Medical University, Shenyang 110001, China
| | - Chao Ma
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hongyue Liang
- Department of Pharmaceutical Toxicology, School of Pharmaceutical Science, China Medical University, Shenyang 110001, China
| | - Wuyang Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, 209Tongshan Rd, Xuzhou 221002, China
| | - Dongyu Min
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang 110032, China.
| | - Lei Xue
- China Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Shenyang, China.
| | - Feng Guo
- Department of Pharmaceutical Toxicology, School of Pharmaceutical Science, China Medical University, Shenyang 110001, China.
| |
Collapse
|
8
|
Zeng ML, Cheng JJ, Kong S, Yang XL, Jia XL, Cheng XL, Chen L, He FG, Liu YM, Fan YT, Gongga L, Chen TX, Liu WH, He XH, Peng BW. Inhibition of Transient Receptor Potential Vanilloid 4 (TRPV4) Mitigates Seizures. Neurotherapeutics 2022; 19:660-681. [PMID: 35182379 PMCID: PMC9226259 DOI: 10.1007/s13311-022-01198-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2022] [Indexed: 02/08/2023] Open
Abstract
Astrocytes are critical regulators of the immune/inflammatory response in several human central nervous system (CNS) diseases. Emerging evidence suggests that dysfunctional astrocytes are crucial players in seizures. The objective of this study was to investigate the role of transient receptor potential vanilloid 4 (TRPV4) in 4-aminopyridine (4-AP)-induced seizures and the underlying mechanism. We also provide evidence for the role of Yes-associated protein (YAP) in seizures. 4-AP was administered to mice or primary cultured astrocytes. YAP-specific small interfering RNA (siRNA) was administered to primary cultured astrocytes. Mouse brain tissue and surgical specimens from epileptic patient brains were examined, and the results showed that TRPV4 was upregulated, while astrocytes were activated and polarized to the A1 phenotype. The levels of glial fibrillary acidic protein (GFAP), cytokine production, YAP, signal transducer activator of transcription 3 (STAT3), intracellular Ca2+([Ca2+]i) and the third component of complement (C3) were increased in 4-AP-induced mice and astrocytes. Perturbations in the immune microenvironment in the brain were balanced by TRPV4 inhibition or the manipulation of [Ca2+]i in astrocytes. Knocking down YAP with siRNA significantly inhibited 4-AP-induced pathological changes in astrocytes. Our study demonstrated that astrocytic TRPV4 activation promoted neuroinflammation through the TRPV4/Ca2+/YAP/STAT3 signaling pathway in mice with seizures. Astrocyte TRPV4 inhibition attenuated neuroinflammation, reduced neuronal injury, and improved neurobehavioral function. Targeting astrocytic TRPV4 activation may provide a promising therapeutic approach for managing epilepsy.
Collapse
Affiliation(s)
- Meng-liu Zeng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Donghu Rd185#, 430071 Wuhan, Hubei China
| | - Jing-jing Cheng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Donghu Rd185#, 430071 Wuhan, Hubei China
| | - Shuo Kong
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Donghu Rd185#, 430071 Wuhan, Hubei China
| | - Xing-liang Yang
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Donghu Rd185#, 430071 Wuhan, Hubei China
| | - Xiang-lei Jia
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Donghu Rd185#, 430071 Wuhan, Hubei China
| | - Xue-lei Cheng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Donghu Rd185#, 430071 Wuhan, Hubei China
| | - Ling Chen
- Institute of Forensic Medicine, School of Basic Medical Sciences, Wuhan University, 430071 Wuhan, Hubei China
| | - Fang-gang He
- Institute of Forensic Medicine, School of Basic Medical Sciences, Wuhan University, 430071 Wuhan, Hubei China
| | - Yu-min Liu
- Department of Neurology, Zhongnan Hospital, Wuhan University, Donghu Road 169#, 430071 Wuhan, Hubei China
| | - Yuan-teng Fan
- Department of Neurology, Zhongnan Hospital, Wuhan University, Donghu Road 169#, 430071 Wuhan, Hubei China
| | - Lanzi Gongga
- Tibet University Medical College, 850000 Lhasa, Tibet China
| | - Tao-xiang Chen
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Donghu Rd185#, 430071 Wuhan, Hubei China
| | - Wan-hong Liu
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, 430071 Wuhan, Hubei China
| | - Xiao-hua He
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, 430071 Wuhan, Hubei China
| | - Bi-wen Peng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Donghu Rd185#, 430071 Wuhan, Hubei China
| |
Collapse
|