1
|
Santarriaga S, Gerlovin K, Layadi Y, Karmacharya R. Human stem cell-based models to study synaptic dysfunction and cognition in schizophrenia: A narrative review. Schizophr Res 2024; 273:78-97. [PMID: 36925354 PMCID: PMC10500041 DOI: 10.1016/j.schres.2023.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023]
Abstract
Cognitive impairment is the strongest predictor of functional outcomes in schizophrenia and is hypothesized to result from synaptic dysfunction. However, targeting synaptic plasticity and cognitive deficits in patients remains a significant clinical challenge. A comprehensive understanding of synaptic plasticity and the molecular basis of learning and memory in a disease context can provide specific targets for the development of novel therapeutics targeting cognitive impairments in schizophrenia. Here, we describe the role of synaptic plasticity in cognition, summarize evidence for synaptic dysfunction in schizophrenia and demonstrate the use of patient derived induced-pluripotent stem cells for studying synaptic plasticity in vitro. Lastly, we discuss current advances and future technologies for bridging basic science research of synaptic dysfunction with clinical and translational research that can be used to predict treatment response and develop novel therapeutics.
Collapse
Affiliation(s)
- Stephanie Santarriaga
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Kaia Gerlovin
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yasmine Layadi
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chimie ParisTech, Université Paris Sciences et Lettres, Paris, France
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
2
|
Cai Y, Li J, Fan K, Zhang D, Lu H, Chen G. Downregulation of chloride voltage-gated channel 7 contributes to hyperalgesia following spared nerve injury. J Biol Chem 2024; 300:107779. [PMID: 39276933 PMCID: PMC11490881 DOI: 10.1016/j.jbc.2024.107779] [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: 06/26/2024] [Revised: 08/22/2024] [Accepted: 08/30/2024] [Indexed: 09/17/2024] Open
Abstract
Alterations in anion balance potential, along with the involvement of cation-chloride cotransporters, play pivotal roles in the development of hyperalgesia after peripheral nerve injury. Chloride voltage-gated channel seven (CLCN7) is the predominant member of the CLC protein family. Investigations on CLCN7 have focused primarily on its involvement in osteosclerosis and lysosomal storage disorders; nevertheless, its contribution to neuropathic pain has not been determined. In this investigation, we noted high expression of CLCN7 in neurons situated within the spinal dorsal horns and dorsal root ganglions (DRGs). Immunofluorescence analysis revealed that CLCN7 was predominantly distributed among IB4-positive and CGRP-positive neurons. Furthermore, the expression of CLCN7 was observed to be mainly reduced in neurons within the spinal dorsal horns and in small- and medium-sized neurons located in the DRGs of spared nerve injury mice. Knockdown of CLCN7 via siRNA in the DRGs resulted in increased mechanical and thermal hyperalgesia in naïve mice. Furthermore, the excitability of cultured DRG neurons in vitro was augmented upon treatment with CLCN7 siRNA. These findings suggested that CLCN7 downregulation following SNI was crucial for the manifestation of mechanical and thermal hyperalgesia, highlighting potential targeting strategies for treating neuropathic pain.
Collapse
Affiliation(s)
- Yunyun Cai
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu, China
| | - Jiajie Li
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Kewei Fan
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu, China
| | - Dongmei Zhang
- Department of Rehabilitation Medicine, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China; Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit of Immunology, Nantong First People's Hospital, Nantong, Jiangsu, China
| | - Hongjian Lu
- Department of Rehabilitation Medicine, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China; Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit of Immunology, Nantong First People's Hospital, Nantong, Jiangsu, China; Medical Research Center, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China.
| | - Gang Chen
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu, China; Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
3
|
Wan Y, Guo S, Zhen W, Xu L, Chen X, Liu F, Shen Y, Liu S, Hu L, Wang X, Ye F, Wang Q, Wen H, Yang F. Structural basis of adenine nucleotides regulation and neurodegenerative pathology in ClC-3 exchanger. Nat Commun 2024; 15:6654. [PMID: 39107281 PMCID: PMC11303396 DOI: 10.1038/s41467-024-50975-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
The ClC-3 chloride/proton exchanger is both physiologically and pathologically critical, as it is potentiated by ATP to detect metabolic energy level and point mutations in ClC-3 lead to severe neurodegenerative diseases in human. However, why this exchanger is differentially modulated by ATP, ADP or AMP and how mutations caused gain-of-function remains largely unknow. Here we determine the high-resolution structures of dimeric wildtype ClC-3 in the apo state and in complex with ATP, ADP and AMP, and the disease-causing I607T mutant in the apo and ATP-bounded state by cryo-electron microscopy. In combination with patch-clamp recordings and molecular dynamic simulations, we reveal how the adenine nucleotides binds to ClC-3 and changes in ion occupancy between apo and ATP-bounded state. We further observe I607T mutation induced conformational changes and augments in current. Therefore, our study not only lays the structural basis of adenine nucleotides regulation in ClC-3, but also clearly indicates the target region for drug discovery against ClC-3 mediated neurodegenerative diseases.
Collapse
Affiliation(s)
- Yangzhuoqun Wan
- Department of Biophysics and Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Shuangshuang Guo
- Department of Biophysics and Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Wenxuan Zhen
- Department of Biophysics and Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Lizhen Xu
- Department of Biophysics and Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Xiaoying Chen
- Department of Biophysics and Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Fangyue Liu
- Department of Neurobiology and Department of General Intensive Care Unit of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Shen
- Department of Neurobiology and Department of General Intensive Care Unit of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuangshuang Liu
- Core Facilities, Zhejiang University School of Medicine, Hangzhou, China
| | - Lidan Hu
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | | | | | | | - Han Wen
- DP Technology, Beijing, China.
- Institute for Advanced Algorithms Research, Shanghai, China.
- State Key Laboratory of Medical Proteomics, Shanghai, China.
- AI for Science Institute, Beijing, China.
- National Key Laboratory of Lead Druggability Research, Beijing, China.
| | - Fan Yang
- Department of Biophysics and Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.
| |
Collapse
|
4
|
Zheng J, Peng S, Cui L, Liu X, Li T, Zhao Z, Li Y, Hu Y, Zhang M, Xu L, Zhang J. Enriched environment attenuates hippocampal theta and gamma rhythms dysfunction in chronic cerebral hypoperfusion via improving imbalanced neural afferent levels. Front Cell Neurosci 2023; 17:985246. [PMID: 37265581 PMCID: PMC10231328 DOI: 10.3389/fncel.2023.985246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 02/27/2023] [Indexed: 06/03/2023] Open
Abstract
Chronic cerebral hypoperfusion (CCH) is increasingly recognized as a common cognitive impairment-causing mechanism. However, no clinically effective drugs to treat cognitive impairment due to CCH have been identified. An abnormal distribution of neural oscillations was found in the hippocampus of CCH rats. By releasing various neurotransmitters, distinct afferent fibers in the hippocampus influence neuronal oscillations in the hippocampus. Enriched environments (EE) are known to improve cognitive levels by modulating neurotransmitter homeostasis. Using EE as an intervention, we examined the levels of three classical neurotransmitters and the dynamics of neural oscillations in the hippocampus of the CCH rat model. The results showed that EE significantly improved the balance of three classical neurotransmitters (acetylcholine, glutamate, and GABA) in the hippocampus, enhanced the strength of theta and slow-gamma (SG) rhythms, and dramatically improved neural coupling across frequency bands in CCH rats. Furthermore, the expression of the three neurotransmitter vesicular transporters-vesicular acetylcholine transporters (VAChT) and vesicular GABA transporters (VGAT)-was significantly reduced in CCH rats, whereas the expression of vesicular glutamate transporter 1 (VGLUT1) was abnormally elevated. EE partially restored the expression of the three protein levels to maintain the balance of hippocampal afferent neurotransmitters. More importantly, causal mediation analysis showed EE increased the power of theta rhythm by increasing the level of VAChT and VGAT, which then enhanced the phase amplitude coupling of theta-SG and finally led to an improvement in the cognitive level of CCH. These findings shed light on the role of CCH in the disruption of hippocampal afferent neurotransmitter balance and neural oscillations. This study has implications for our knowledge of disease pathways.
Collapse
Affiliation(s)
- Jiaxin Zheng
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Sisi Peng
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lingling Cui
- Department of Anesthesiology, Tongren Hospital of Wuhan University, Wuhan, China
| | - Xi Liu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tian Li
- Clinical Medical Research Center for Dementia and Cognitive Impairment in Hubei Province, Wuhan, China
| | - Zhenyu Zhao
- Clinical Medical Research Center for Dementia and Cognitive Impairment in Hubei Province, Wuhan, China
| | - Yaqing Li
- Clinical Medical Research Center for Dementia and Cognitive Impairment in Hubei Province, Wuhan, China
| | - Yuan Hu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Miao Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Linling Xu
- Clinical Medical Research Center for Dementia and Cognitive Impairment in Hubei Province, Wuhan, China
| | - JunJian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
5
|
Howes OD, Shatalina E. Integrating the Neurodevelopmental and Dopamine Hypotheses of Schizophrenia and the Role of Cortical Excitation-Inhibition Balance. Biol Psychiatry 2022; 92:501-513. [PMID: 36008036 DOI: 10.1016/j.biopsych.2022.06.017] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/16/2022] [Accepted: 06/04/2022] [Indexed: 12/23/2022]
Abstract
The neurodevelopmental and dopamine hypotheses are leading theories of the pathoetiology of schizophrenia, but they were developed in isolation. However, since they were originally proposed, there have been considerable advances in our understanding of the normal neurodevelopmental refinement of synapses and cortical excitation-inhibition (E/I) balance, as well as preclinical findings on the interrelationship between cortical and subcortical systems and new in vivo imaging and induced pluripotent stem cell evidence for lower synaptic density markers in patients with schizophrenia. Genetic advances show that schizophrenia is associated with variants linked to genes affecting GABA (gamma-aminobutyric acid) and glutamatergic signaling as well as neurodevelopmental processes. Moreover, in vivo studies on the effects of stress, particularly during later development, show that it leads to synaptic elimination. We review these lines of evidence as well as in vivo evidence for altered cortical E/I balance and dopaminergic dysfunction in schizophrenia. We discuss mechanisms through which frontal cortex circuitry may regulate striatal dopamine and consider how frontal E/I imbalance may cause dopaminergic dysregulation to result in psychotic symptoms. This integrated neurodevelopmental and dopamine hypothesis suggests that overpruning of synapses, potentially including glutamatergic inputs onto frontal cortical interneurons, disrupts the E/I balance and thus underlies cognitive and negative symptoms. It could also lead to disinhibition of excitatory projections from the frontal cortex and possibly other regions that regulate mesostriatal dopamine neurons, resulting in dopamine dysregulation and psychotic symptoms. Together, this explains a number of aspects of the epidemiology and clinical presentation of schizophrenia and identifies new targets for treatment and prevention.
Collapse
Affiliation(s)
- Oliver D Howes
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Hammersmith Hospital, Imperial College London, United Kingdom; Department of Psychosis, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom.
| | - Ekaterina Shatalina
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Hammersmith Hospital, Imperial College London, United Kingdom
| |
Collapse
|
6
|
Riazanski V, Mauleon G, Zimnicka AM, Chen S, Nelson DJ. Phagosomal chloride dynamics in the alveolar macrophage. iScience 2022; 25:103636. [PMID: 35024579 PMCID: PMC8733233 DOI: 10.1016/j.isci.2021.103636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 10/11/2021] [Accepted: 12/14/2021] [Indexed: 12/04/2022] Open
Abstract
Acidification in intracellular organelles is tightly linked to the influx of Cl- counteracting proton translocation by the electrogenic V-ATPase. We quantified the dynamics of Cl- transfer accompanying cargo incorporation into single phagosomes in alveolar macrophages (AMs). Phagosomal Cl- concentration and acidification magnitude were followed in real time with maximal acidification achieved at levels of approximately 200 mM. Live cell confocal microscopy verified that phagosomal Cl- influx utilized predominantly the Cl- channel CFTR. Relative levels of elemental chlorine (Cl) in hard X-ray fluorescence microprobe (XFM) analysis within single phagosomes validated the increase in Cl- content. XFM revealed the complex interplay between elemental K content inside the phagosome and changes in Cl- during phagosomal particle uptake. Cl- -dependent changes in phagosomal membrane potential were obtained using second harmonic generation (SHG) microscopy. These studies provide a mechanistic insight for screening studies in drug development targeting pulmonary inflammatory disease.
Collapse
Affiliation(s)
- Vladimir Riazanski
- The University of Chicago, Department of Pharmacological and Physiological Sciences, 947 E. 58th Street, MC 0926, Chicago, IL 60637, USA
| | - Gerardo Mauleon
- The University of Chicago, Department of Pharmacological and Physiological Sciences, 947 E. 58th Street, MC 0926, Chicago, IL 60637, USA
| | - Adriana M. Zimnicka
- The University of Chicago, Department of Pharmacological and Physiological Sciences, 947 E. 58th Street, MC 0926, Chicago, IL 60637, USA
| | - Si Chen
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Argonne, IL 60439, USA
| | - Deborah J. Nelson
- The University of Chicago, Department of Pharmacological and Physiological Sciences, 947 E. 58th Street, MC 0926, Chicago, IL 60637, USA
| |
Collapse
|
7
|
Riazanski V, Mauleon G, Lucas K, Walker S, Zimnicka AM, McGrath JL, Nelson DJ. Real time imaging of single extracellular vesicle pH regulation in a microfluidic cross-flow filtration platform. Commun Biol 2022; 5:13. [PMID: 35013561 PMCID: PMC8748679 DOI: 10.1038/s42003-021-02965-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 12/13/2021] [Indexed: 11/22/2022] Open
Abstract
Extracellular vesicles (EVs) are cell-derived membranous structures carrying transmembrane proteins and luminal cargo. Their complex cargo requires pH stability in EVs while traversing diverse body fluids. We used a filtration-based platform to capture and stabilize EVs based on their size and studied their pH regulation at the single EV level. Dead-end filtration facilitated EV capture in the pores of an ultrathin (100 nm thick) and nanoporous silicon nitride (NPN) membrane within a custom microfluidic device. Immobilized EVs were rapidly exposed to test solution changes driven across the backside of the membrane using tangential flow without exposing the EVs to fluid shear forces. The epithelial sodium-hydrogen exchanger, NHE1, is a ubiquitous plasma membrane protein tasked with the maintenance of cytoplasmic pH at neutrality. We show that NHE1 identified on the membrane of EVs is functional in the maintenance of pH neutrality within single vesicles. This is the first mechanistic description of EV function on the single vesicle level. Riazanski et al describe a platform to capture extracellular vesicles (EVs) using a nanoporous silicon nitride membrane, investigate the expression of NHE1 protein on the surface of EVs and monitor the transport of Na+ and H+ at the single EV level. The authors report a mechanistic function of the proteins found in EVs and specifically identify NHE1 on a single EV, where it maintains pH neutrality within single vesicles.
Collapse
Affiliation(s)
- Vladimir Riazanski
- Department of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, IL, 60637, USA
| | - Gerardo Mauleon
- Department of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, IL, 60637, USA
| | - Kilean Lucas
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Samuel Walker
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Adriana M Zimnicka
- Department of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, IL, 60637, USA
| | - James L McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Deborah J Nelson
- Department of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
8
|
Difference of pain vulnerability in adult and juvenile rodents: the role of SIRT1-mediated ClC-3 trafficking in sensory neurons. Pain 2021; 162:1882-1896. [PMID: 33433144 DOI: 10.1097/j.pain.0000000000002176] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/15/2020] [Indexed: 12/16/2022]
Abstract
ABSTRACT Adults are more likely to suffer from chronic pain than minors, and its underlying mechanism remains unclear. SIRT1 an important age-related protein with function of lifespan extension; whether SIRT1 plays a role in the different pain vulnerability of adult and juvenile remains unclear. Here, we found that the expression level of SIRT1 in dorsal root ganglia (DRG) was related to the pain vulnerability. After nerve injury, the expression of SIRT1 in DRG was decreased in adult rodents whereas increased in juvenile rodents. Differential manipulation of SIRT1 abolished the different pain vulnerability between adult and juvenile rodents. Furthermore, SIRT1 interacted with ClC-3 channel and mediated ClC-3 membrane trafficking and Cl- current in DRG neurons. Differential manipulation of ClC-3 also abolished the difference in pain vulnerability between adult and juvenile rodents. The different anti-inflammatory ability determined the different change trends of SIRT1 and ClC-3 trafficking contributed to the different pain vulnerability in adult and juvenile rodents. In addition, the serum SIRT1 level was negatively correlated with the pain score in patients with chronic pain. These findings revealed the mechanism of the difference in pain vulnerability between adult and juvenile rodents and provided evidence for age-specific treatment of chronic pain.
Collapse
|
9
|
Bose S, He H, Stauber T. Neurodegeneration Upon Dysfunction of Endosomal/Lysosomal CLC Chloride Transporters. Front Cell Dev Biol 2021; 9:639231. [PMID: 33708769 PMCID: PMC7940362 DOI: 10.3389/fcell.2021.639231] [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: 12/08/2020] [Accepted: 02/03/2021] [Indexed: 12/15/2022] Open
Abstract
The regulation of luminal ion concentrations is critical for the function of, and transport between intracellular organelles. The importance of the acidic pH in the compartments of the endosomal-lysosomal pathway has been well-known for decades. Besides the V-ATPase, which pumps protons into their lumen, a variety of ion transporters and channels is involved in the regulation of the organelles' complex ion homeostasis. Amongst these are the intracellular members of the CLC family, ClC-3 through ClC-7. They localize to distinct but overlapping compartments of the endosomal-lysosomal pathway, partially with tissue-specific expression. Functioning as 2Cl−/H+ exchangers, they can support the vesicular acidification and accumulate luminal Cl−. Mutations in the encoding genes in patients and mouse models underlie severe phenotypes including kidney stones with CLCN5 and osteopetrosis or hypopigmentation with CLCN7. Dysfunction of those intracellular CLCs that are expressed in neurons lead to neuronal defects. Loss of endosomal ClC-3, which heteromerizes with ClC-4, results in neurodegeneration. Mutations in ClC-4 are associated with epileptic encephalopathy and intellectual disability. Mice lacking the late endosomal ClC-6 develop a lysosomal storage disease with reduced pain sensitivity. Human gene variants have been associated with epilepsy, and a gain-of-function mutation causes early-onset neurodegeneration. Dysfunction of the lysosomal ClC-7 leads to a lysosomal storage disease and neurodegeneration in mice and humans. Reduced luminal chloride, as well as altered calcium regulation, has been associated with lysosomal storage diseases in general. This review discusses the properties of endosomal and lysosomal Cl−/H+ exchange by CLCs and how various alterations of ion transport by CLCs impact organellar ion homeostasis and function in neurodegenerative disorders.
Collapse
Affiliation(s)
- Shroddha Bose
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Hailan He
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Tobias Stauber
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Department of Human Medicine and Institute for Molecular Medicine, MSH Medical School Hamburg, Hamburg, Germany
| |
Collapse
|
10
|
Weinert S, Gimber N, Deuschel D, Stuhlmann T, Puchkov D, Farsi Z, Ludwig CF, Novarino G, López-Cayuqueo KI, Planells-Cases R, Jentsch TJ. Uncoupling endosomal CLC chloride/proton exchange causes severe neurodegeneration. EMBO J 2020; 39:e103358. [PMID: 32118314 PMCID: PMC7196918 DOI: 10.15252/embj.2019103358] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 01/24/2020] [Accepted: 01/27/2020] [Indexed: 01/02/2023] Open
Abstract
CLC chloride/proton exchangers may support acidification of endolysosomes and raise their luminal Cl− concentration. Disruption of endosomal ClC‐3 causes severe neurodegeneration. To assess the importance of ClC‐3 Cl−/H+ exchange, we now generate Clcn3unc/unc mice in which ClC‐3 is converted into a Cl− channel. Unlike Clcn3−/− mice, Clcn3unc/unc mice appear normal owing to compensation by ClC‐4 with which ClC‐3 forms heteromers. ClC‐4 protein levels are strongly reduced in Clcn3−/−, but not in Clcn3unc/unc mice because ClC‐3unc binds and stabilizes ClC‐4 like wild‐type ClC‐3. Although mice lacking ClC‐4 appear healthy, its absence in Clcn3unc/unc/Clcn4−/− mice entails even stronger neurodegeneration than observed in Clcn3−/− mice. A fraction of ClC‐3 is found on synaptic vesicles, but miniature postsynaptic currents and synaptic vesicle acidification are not affected in Clcn3unc/unc or Clcn3−/− mice before neurodegeneration sets in. Both, Cl−/H+‐exchange activity and the stabilizing effect on ClC‐4, are central to the biological function of ClC‐3.
Collapse
Affiliation(s)
- Stefanie Weinert
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Niclas Gimber
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Dorothea Deuschel
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Till Stuhlmann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Dmytro Puchkov
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Zohreh Farsi
- Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Carmen F Ludwig
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Gaia Novarino
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Karen I López-Cayuqueo
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Rosa Planells-Cases
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Thomas J Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany.,NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
11
|
Hall LS, Medway CW, Pain O, Pardiñas AF, Rees EG, Escott-Price V, Pocklington A, Bray NJ, Holmans PA, Walters JTR, Owen MJ, O’Donovan MC. A transcriptome-wide association study implicates specific pre- and post-synaptic abnormalities in schizophrenia. Hum Mol Genet 2020; 29:159-167. [PMID: 31691811 PMCID: PMC7416679 DOI: 10.1093/hmg/ddz253] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/31/2019] [Accepted: 09/02/2019] [Indexed: 12/25/2022] Open
Abstract
Schizophrenia is a complex highly heritable disorder. Genome-wide association studies (GWAS) have identified multiple loci that influence the risk of developing schizophrenia, although the causal variants driving these associations and their impacts on specific genes are largely unknown. We identify a significant correlation between schizophrenia risk and expression at 89 genes in the dorsolateral prefrontal cortex (P ≤ 9.43 × 10-6), including 20 novel genes. Genes whose expression correlate with schizophrenia were enriched for those involved in abnormal CNS synaptic transmission (PFDR = 0.02) and antigen processing and presentation of peptide antigen via MHC class I (PFDR = 0.02). Within the CNS synaptic transmission set, we identify individual significant candidate genes to which we assign direction of expression changes in schizophrenia. The findings provide strong candidates for experimentally probing the molecular basis of synaptic pathology in schizophrenia.
Collapse
Affiliation(s)
- Lynsey S Hall
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Christopher W Medway
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Oliver Pain
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| | - Antonio F Pardiñas
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Elliott G Rees
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Valentina Escott-Price
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Andrew Pocklington
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Nicholas J Bray
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Peter A Holmans
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - James T R Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Michael J Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Michael C O’Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| |
Collapse
|
12
|
Xu ZZ, Chen QY, Deng SY, Zhang M, Tan CY, Yang Wang, Ma KT, Li L, Si JQ, Zhu LC. 17β-Estradiol Attenuates Neuropathic Pain Caused by Spared Nerve Injury by Upregulating CIC-3 in the Dorsal Root Ganglion of Ovariectomized Rats. Front Neurosci 2019; 13:1205. [PMID: 31787875 PMCID: PMC6856564 DOI: 10.3389/fnins.2019.01205] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/24/2019] [Indexed: 12/14/2022] Open
Abstract
17β-estradiol plays a role in pain sensitivity, analgesic drug efficacy, and neuropathic pain prevalence, but the underlying mechanisms remain unclear. Here, we investigated whether voltage-gated chloride channel-3 (ClC-3) impacts the effects of 17β-estradiol (E2) on spared nerve injury (SNI)-induced neuropathic pain in ovariectomized (OVX) female Sprague Dawley rats that were divided into OVX, OVX + SNI, OVX + SNI + E2, OVX + SNI + E2 + DMSO (vehicle, dimethyl sulfoxide), or OVX + SNI + E2+Cltx (ClC-3-blocker chlorotoxin) groups. Changes in ClC-3 protein expression were monitored by western blot analysis. Behavioral testing used the paw withdrawal threshold to acetone irritation and paw withdrawal thermal latency (PWTL) to thermal stimulation. Immunofluorescence indicated the localization and protein expression levels of ClC-3. OVX + SNI + E2 rats were subcutaneously injected with 17β-estradiol once daily for 7 days; a sheathed tube was implanted, and chlorotoxin was injected for 4 days. Intrathecal Cltx to OVX and OVX + SNI rats was administered for 4 consecutive days (days 7–10 after SNI) to further determine the contribution of ClC-3 to neuropathic pain. Patch clamp technology in current clamp mode was used to measure the current threshold (rheobase) dorsal root ganglion (DRG) neurons and the minimal current that evoked action potentials (APs) as excitability parameters. The mean number of APs at double-strength rheobase verified neuronal excitability. There was no difference in behaviors and ClC-3 expression after OVX. Compared with OVX + SNI rats, OVX + SNI + E2 rats showed a lower paw withdrawal threshold to the acetone stimulus, but the PWTL was not significantly different, indicating increased sensitivity to cold but not to thermal pain. Co-immunofluorescent data revealed that ClC-3 was mainly distributed in A- and C-type nociceptive neurons, especially in medium/small-sized neurons. 17β-estradiol administration was associated with increased expression of ClC-3. 17β-estradiol-induced increase in ClC-3 expression was blocked by co-administration of Cltx. Cltx causes hyperalgesia and decreased expression of ClC-3 in OVX rats. Patch clamp results suggested that 17β-estradiol attenuated the excitability of neurons induced by SNI by up-regulating the expression of ClC-3 in the DRG of OVX rats. 17β-estradiol administration significantly improved cold allodynia thresholds in OVX rats with SNI. The mechanism for this decreased sensitivity may be related to the upregulation of ClC-3 expression in the DRG.
Collapse
Affiliation(s)
- Zhen-Zhen Xu
- Department of Anesthesiology, First Affiliated Hospital of Shihezi University, Shihezi, China.,Department of Physiology, Shihezi University School of Medicine, Shihezi, China.,Key Laboratory of Xinjiang Endemic and Ethnic Disease, Shihezi University School of Medicine, Shihezi, China.,Department of Anesthesiology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, China
| | - Qin-Yi Chen
- Department of Anesthesiology, First Affiliated Hospital of Shihezi University, Shihezi, China.,Department of Physiology, Shihezi University School of Medicine, Shihezi, China.,Key Laboratory of Xinjiang Endemic and Ethnic Disease, Shihezi University School of Medicine, Shihezi, China.,Department of Anesthesiology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, China
| | - Shi-Yu Deng
- Department of Anesthesiology, First Affiliated Hospital of Shihezi University, Shihezi, China.,Department of Physiology, Shihezi University School of Medicine, Shihezi, China.,Key Laboratory of Xinjiang Endemic and Ethnic Disease, Shihezi University School of Medicine, Shihezi, China
| | - Meng Zhang
- Department of Anesthesiology, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu, China
| | - Chao-Yang Tan
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China.,Key Laboratory of Xinjiang Endemic and Ethnic Disease, Shihezi University School of Medicine, Shihezi, China
| | - Yang Wang
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China.,Key Laboratory of Xinjiang Endemic and Ethnic Disease, Shihezi University School of Medicine, Shihezi, China
| | - Ke-Tao Ma
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China.,Key Laboratory of Xinjiang Endemic and Ethnic Disease, Shihezi University School of Medicine, Shihezi, China
| | - Li Li
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China.,Key Laboratory of Xinjiang Endemic and Ethnic Disease, Shihezi University School of Medicine, Shihezi, China.,Department of Physiology, Medical College of Jiaxing University, Jiaxing, China
| | - Jun-Qiang Si
- Department of Anesthesiology, First Affiliated Hospital of Shihezi University, Shihezi, China.,Department of Physiology, Shihezi University School of Medicine, Shihezi, China.,Key Laboratory of Xinjiang Endemic and Ethnic Disease, Shihezi University School of Medicine, Shihezi, China.,Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Physiology, School of Basic Medical Sciences, Wuhan University School of Medicine, Wuhan, China
| | - Li-Cang Zhu
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China.,Key Laboratory of Xinjiang Endemic and Ethnic Disease, Shihezi University School of Medicine, Shihezi, China
| |
Collapse
|
13
|
He Y, Luo X, Zhou B, Hu T, Meng X, Audano PA, Kronenberg ZN, Eichler EE, Jin J, Guo Y, Yang Y, Qi X, Su B. Long-read assembly of the Chinese rhesus macaque genome and identification of ape-specific structural variants. Nat Commun 2019; 10:4233. [PMID: 31530812 PMCID: PMC6749001 DOI: 10.1038/s41467-019-12174-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 08/27/2019] [Indexed: 12/20/2022] Open
Abstract
We present a high-quality de novo genome assembly (rheMacS) of the Chinese rhesus macaque (Macaca mulatta) using long-read sequencing and multiplatform scaffolding approaches. Compared to the current Indian rhesus macaque reference genome (rheMac8), rheMacS increases sequence contiguity 75-fold, closing 21,940 of the remaining assembly gaps (60.8 Mbp). We improve gene annotation by generating more than two million full-length transcripts from ten different tissues by long-read RNA sequencing. We sequence resolve 53,916 structural variants (96% novel) and identify 17,000 ape-specific structural variants (ASSVs) based on comparison to ape genomes. Many ASSVs map within ChIP-seq predicted enhancer regions where apes and macaque show diverged enhancer activity and gene expression. We further characterize a subset that may contribute to ape- or great-ape-specific phenotypic traits, including taillessness, brain volume expansion, improved manual dexterity, and large body size. The rheMacS genome assembly serves as an ideal reference for future biomedical and evolutionary studies. Comparative genomic analysis of human and primate relatives can reveal important biological and evolutionary insights. Here, the authors present a long-read assembly of the Chinese rhesus macaque genome and identify ape-specific structural variants.
Collapse
Affiliation(s)
- Yaoxi He
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Xin Luo
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Bin Zhou
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Ting Hu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiaoyu Meng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Peter A Audano
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Zev N Kronenberg
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Evan E Eichler
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, 98195, USA.,Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA
| | - Jie Jin
- Nextomics Biosciences, Wuhan, 430000, China
| | - Yongbo Guo
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Yanan Yang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Xuebin Qi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
| | - Bing Su
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China. .,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China. .,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
| |
Collapse
|
14
|
Wu Z, Huo Q, Ren L, Dong F, Feng M, Wang Y, Bai Y, Lüscher B, Li ST, Wang GL, Long C, Wang Y, Wu G, Chen G. Gluconate suppresses seizure activity in developing brains by inhibiting CLC-3 chloride channels. Mol Brain 2019; 12:50. [PMID: 31088565 PMCID: PMC6518791 DOI: 10.1186/s13041-019-0465-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 04/17/2019] [Indexed: 12/03/2022] Open
Abstract
Neonatal seizures are different from adult seizures, and many antiepileptic drugs that are effective in adults often fail to treat neonates. Here, we report that gluconate inhibits neonatal seizure by inhibiting CLC-3 chloride channels. We detect a voltage-dependent outward rectifying Cl− current mediated by CLC-3 Cl− channels in early developing brains but not adult mouse brains. Blocking CLC-3 Cl− channels by gluconate inhibits seizure activity both in neonatal brain slices and in neonatal animals with in vivo EEG recordings. Consistently, neonatal neurons of CLC-3 knockout mice lack the outward rectifying Cl− current and show reduced epileptiform activity upon stimulation. Mechanistically, we demonstrate that activation of CLC-3 Cl− channels alters intracellular Cl− homeostasis and enhances GABA excitatory activity. Our studies suggest that gluconate can suppress neonatal seizure activities through inhibiting CLC-3 Cl− channels in developing brains.
Collapse
Affiliation(s)
- Zheng Wu
- Department of Biology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Qingwei Huo
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China.,South China Research Center for Acupuncture-Moxibustion, Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou Univ Chinese Med, Guangzhou, 510006, China
| | - Liang Ren
- Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Fengping Dong
- Department of Biology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Mengyang Feng
- Department of Biology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yue Wang
- Department of Biology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yuting Bai
- Department of Biology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Bernhard Lüscher
- Department of Biology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Sheng-Tian Li
- Bio-X Institutes, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Guan-Lei Wang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Cheng Long
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Yun Wang
- Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Gangyi Wu
- Department of Biology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA.,School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Gong Chen
- Department of Biology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
15
|
Expression of plasma membrane calcium ATPases confers Ca 2+/H + exchange in rodent synaptic vesicles. Sci Rep 2019; 9:4289. [PMID: 30862855 PMCID: PMC6414521 DOI: 10.1038/s41598-019-40557-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 02/19/2019] [Indexed: 02/07/2023] Open
Abstract
Ca2+ transport into synaptic vesicles (SVs) at the presynaptic terminals has been proposed to be an important process for regulating presynaptic [Ca2+] during stimulation as well as at rest. However, the molecular identity of the transport system remains elusive. Previous studies have demonstrated that isolated SVs exhibit two distinct Ca2+ transport systems depending on extra-vesicular (cytosolic) pH; one is mediated by a high affinity Ca2+ transporter which is active at neutral pH and the other is mediated by a low affinity Ca2+/H+ antiporter which is maximally active at alkaline pH of 8.5. In addition, synaptic vesicle glycoprotein 2 s (SV2s), a major SV component, have been proposed to contribute to Ca2+ clearance from the presynaptic cytoplasm. Here, we show that at physiological pH, the plasma membrane Ca2+ ATPases (PMCAs) are responsible for both the Ca2+/H+ exchange activity and Ca2+ uptake into SVs. The Ca2+/H+ exchange activity monitored by acidification assay exhibited high affinity for Ca2+ (Km ~ 400 nM) and characteristic divalent cation selectivity for the PMCAs. Both activities were remarkably reduced by PMCA blockers, but not by a blocker of the ATPase that transfers Ca2+ from the cytosol to the lumen of sarcoplasmic endoplasmic reticulum (SERCA) at physiological pH. Furthermore, we rule out the contribution of SV2s, putative Ca2+ transporters on SVs, since both Ca2+/H+ exchange activity and Ca2+ transport were unaffected in isolated vesicles derived from SV2-deficient brains. Finally, using a PMCA1-pHluorin construct that enabled us to monitor cellular distribution and recycling properties in living neurons, we demonstrated that PMCA1-pHluorin localized to intracellular acidic compartments and recycled at presynaptic terminals in an activity-dependent manner. Collectively, our results imply that vesicular PMCAs may play pivotal roles in both presynaptic Ca2+ homeostasis and the modulation of H+ gradient in SVs.
Collapse
|
16
|
Chen YF, Chen ZX, Wang RH, Shi YW, Xue L, Wang XG, Zhao H. Knockdown of CLC-3 in the hippocampal CA1 impairs contextual fear memory. Prog Neuropsychopharmacol Biol Psychiatry 2019; 89:132-145. [PMID: 30025794 DOI: 10.1016/j.pnpbp.2018.07.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 06/11/2018] [Accepted: 07/05/2018] [Indexed: 01/01/2023]
Abstract
Previous studies support a critical role of hippocampus in contextual fear memory. Structural and functional alterations of hippocampus occur frequently in posttraumatic stress disorders (PTSD). Recent reports reveal that knockout of CLC-3, a member of the CLC family of anion channels and transporters, leads to neuronal degeneration and loss of hippocampus. However, the role of CLC-3 in contextual fear memory remains unknown. Using adenovirus and adeno-associated virus gene transfer to knockdown CLC-3 in hippocampal CA1, we investigate the role of CLC-3 in contextual fear memory. CLC-3 expression is increased in hippocampal CA1 after formation of long-term contextual fear memory. Knockdown of CLC-3 by adenovirus infusion in hippocampal CA1 significantly attenuates the contextual fear memory, reduces spine density, induces defects of excitatory synaptic ultrastructure showed by the decreased PSD length, PSD thickness and active zone length, and impairs L-LTP induction and maintenance. Knockdown of CLC-3 also induces the synaptic NMDAR subunit composition to an increased GluN2A/GluN2B ratio pattern and reduces the activity of CaMKII-α. Furthermore, selectively knockdown of CLC-3 in excitatory neurons by adeno-associated virus driven from CaMKII-α promoter is sufficient to impair long-term contextual fear memory. These findings highlight that CLC-3 in hippocampal CA1 is necessary for contextual fear memory.
Collapse
Affiliation(s)
- Ye-Fei Chen
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, China; Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Zi-Xiang Chen
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Run-Hua Wang
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Yan-Wei Shi
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, China; Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Li Xue
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, China; Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Xiao-Guang Wang
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, China; Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, China.
| | - Hu Zhao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, China; Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, China.
| |
Collapse
|
17
|
Rohrbough J, Nguyen H, Lamb FS. Modulation of ClC-3 gating and proton/anion exchange by internal and external protons and the anion selectivity filter. J Physiol 2018; 596:4091-4119. [PMID: 29917234 PMCID: PMC6117567 DOI: 10.1113/jp276332] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/07/2018] [Indexed: 01/13/2023] Open
Abstract
KEY POINTS The ClC-3 2Cl- /1H+ exchanger modulates endosome pH and Cl- concentration. We investigated the relationships between ClC-3-mediated ion transport (steady-state transport current, ISS ), gating charge (Q) and cytoplasmic alkalization. ClC-3 transport is functionally unidirectional. ClC-5 and ClC-3 display indistinguishable exchange ratios, but ClC-3 cycling is less "efficient", as reflected by a large Q/ISS . An M531A mutation predicted to increase water-wire stability and cytoplasmic proton supply improves efficiency. Protonation (pH 5.0) of the outer glutamate gate (Gluext ; E224) reduces Q, inhibits transport, and weakens coupling. Removal of the central tyrosine anion gate (Y572S) greatly increases uncoupled anion current. Tyrosine -OH removal (Y572F) alters anion selectivity and impairs coupling. E224 and Y572 act as anion barriers, and contribute to gating. The Y572 side chain and -OH regulate Q movement kinetics and voltage dependence. E224 and Y572 interact to create a "closed" inner gate conformation that maintains coupling during cycling. ABSTRACT We utilized plasma membrane-localized ClC-3 to investigate relationships between steady-state transport current (ISS ), gating charge (Q) movement, and cytoplasmic alkalization rate. ClC-3 exhibited lower transport efficiency than ClC-5, as reflected by a larger Q/ISS ratio, but an indistinguishable Cl- /H+ coupling ratio. External SCN- reduced H+ transport rate and uncoupled anion/H+ exchange by 80-90%. Removal of the external gating glutamate ("Gluext ") (E224A mutation) reduced Q and abolished H+ transport. We hypothesized that Methionine 531 (M531) impedes "water wire" H+ transfer from the cytoplasm to E224. Accordingly, an M531A mutation decreased the Q/ISS ratio by 50% and enhanced H+ transport. External protons (pH 5.0) inhibited ISS and markedly reduced Q while shifting the Q-voltage (V) relationship positively. The Cl- /H+ coupling ratio at pH 5.0 was significantly increased, consistent with externally protonated Gluext adopting an outward/open position. Internal "anion gate" removal (Y572S) dramatically increased ISS and impaired coupling, without slowing H+ transport rate. Loss of both gates (Y572S/E224A) resulted in a large "open pore" conductance. Y572F (removing only the phenolic hydroxide) and Y572S shortened Q duration similarly, resulting in faster Q kinetics at all voltages. These data reveal a complex relationship between Q and ion transport. Q/ISS must be assessed together with coupling ratio to properly interpret efficiency. Coupling and transport rate are influenced by the anion, internal proton supply and external protons. Y572 regulates H+ coupling as well as anion selectivity, and interacts directly with E224. Disruption of this "closed gate" conformation by internal protons may represent a critical step in the ClC-3 transport cycle.
Collapse
Affiliation(s)
- Jeffrey Rohrbough
- Departments of Pediatrics and Molecular Physiology & BiophysicsMonroe Carell Children's Hospital at Vanderbilt UniversityNashvilleTNUSA
| | - Hong‐Ngan Nguyen
- Departments of Pediatrics and Molecular Physiology & BiophysicsMonroe Carell Children's Hospital at Vanderbilt UniversityNashvilleTNUSA
| | - Fred S. Lamb
- Departments of Pediatrics and Molecular Physiology & BiophysicsMonroe Carell Children's Hospital at Vanderbilt UniversityNashvilleTNUSA
| |
Collapse
|
18
|
Jentsch TJ, Pusch M. CLC Chloride Channels and Transporters: Structure, Function, Physiology, and Disease. Physiol Rev 2018; 98:1493-1590. [DOI: 10.1152/physrev.00047.2017] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
CLC anion transporters are found in all phyla and form a gene family of eight members in mammals. Two CLC proteins, each of which completely contains an ion translocation parthway, assemble to homo- or heteromeric dimers that sometimes require accessory β-subunits for function. CLC proteins come in two flavors: anion channels and anion/proton exchangers. Structures of these two CLC protein classes are surprisingly similar. Extensive structure-function analysis identified residues involved in ion permeation, anion-proton coupling and gating and led to attractive biophysical models. In mammals, ClC-1, -2, -Ka/-Kb are plasma membrane Cl−channels, whereas ClC-3 through ClC-7 are 2Cl−/H+-exchangers in endolysosomal membranes. Biological roles of CLCs were mostly studied in mammals, but also in plants and model organisms like yeast and Caenorhabditis elegans. CLC Cl−channels have roles in the control of electrical excitability, extra- and intracellular ion homeostasis, and transepithelial transport, whereas anion/proton exchangers influence vesicular ion composition and impinge on endocytosis and lysosomal function. The surprisingly diverse roles of CLCs are highlighted by human and mouse disorders elicited by mutations in their genes. These pathologies include neurodegeneration, leukodystrophy, mental retardation, deafness, blindness, myotonia, hyperaldosteronism, renal salt loss, proteinuria, kidney stones, male infertility, and osteopetrosis. In this review, emphasis is laid on biophysical structure-function analysis and on the cell biological and organismal roles of mammalian CLCs and their role in disease.
Collapse
Affiliation(s)
- Thomas J. Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany; and Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Genova, Italy
| | - Michael Pusch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany; and Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Genova, Italy
| |
Collapse
|
19
|
Peng J, Chen W, Chen J, Yuan Y, Zhang J, He Y. Overexpression of chloride channel-3 predicts unfavorable prognosis and promotes cellular invasion in gastric cancer. Cancer Manag Res 2018; 10:1163-1175. [PMID: 29795988 PMCID: PMC5958948 DOI: 10.2147/cmar.s159790] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Chloride channel-3 (CLC-3) has been reported to promote the proliferation and invasion in various tumors, yet little is known about its role in gastric cancer. In the present study, we investigated the clinical significance of CLC-3 and its biological role in gastric cancer. METHODS Bioinformatic analysis, immunohistochemical staining, quantitative real-time polymerase chain reaction and Western blot assay were used to assess the expression of CLC-3 and its clinical significance in gastric cancer. The biological role of CLC-3 and its underlying mechanism were detected through in vitro experiments. RESULTS CLC-3 was highly expressed in gastric cancer tissues and cell lines, and high levels of CLC-3 were significantly associated with adverse clinicopathological parameters and shorter overall survival time in patients with gastric cancer. Functional studies revealed that silencing of CLC-3 decreased, while overexpression promoted, the proliferation, migration and invasion of gastric cancer cells in vitro. Mechanistic studies suggested that canonical TGF-β/Smad signaling pathway is involved in CLC-3-induced gastric cancer cells proliferation, migration and invasion. CONCLUSION These findings indicate the vital role of CLC-3 in gastric cancer progression and its potential role of a therapeutic target for treatment.
Collapse
Affiliation(s)
- Jianjun Peng
- Gastrointestinal Surgical Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Chen
- Gastrointestinal Surgical Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jianhui Chen
- Gastrointestinal Surgical Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yujie Yuan
- Gastrointestinal Surgical Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jian Zhang
- Gastrointestinal Surgical Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yulong He
- Gastrointestinal Surgical Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
20
|
An interplay between plasticity and parental phenotype determines impacts of ocean acidification on a reef fish. Nat Ecol Evol 2017; 2:334-342. [PMID: 29255298 DOI: 10.1038/s41559-017-0428-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 11/22/2017] [Indexed: 11/09/2022]
Abstract
The impacts of ocean acidification will depend on the ability of marine organisms to tolerate, acclimate and eventually adapt to changes in ocean chemistry. Here, we use a unique transgenerational experiment to determine the molecular response of a coral reef fish to short-term, developmental and transgenerational exposure to elevated CO2, and to test how these responses are influenced by variations in tolerance to elevated CO2 exhibited by the parents. Within-generation responses in gene expression to end-of-century predicted CO2 levels indicate that a self-amplifying cycle in GABAergic neurotransmission is triggered, explaining previously reported neurological and behavioural impairments. Furthermore, epigenetic regulator genes exhibited a within-generation specific response, but with some divergence due to parental phenotype. Importantly, we find that altered gene expression for the majority of within-generation responses returns to baseline levels following parental exposure to elevated CO2 conditions. Our results show that both parental variation in tolerance and cross-generation exposure to elevated CO2 are crucial factors in determining the response of reef fish to changing ocean chemistry.
Collapse
|
21
|
Fullard JF, Giambartolomei C, Hauberg ME, Xu K, Voloudakis G, Shao Z, Bare C, Dudley JT, Mattheisen M, Robakis NK, Haroutunian V, Roussos P. Open chromatin profiling of human postmortem brain infers functional roles for non-coding schizophrenia loci. Hum Mol Genet 2017; 26:1942-1951. [PMID: 28335009 DOI: 10.1093/hmg/ddx103] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 03/10/2017] [Indexed: 01/03/2023] Open
Abstract
Open chromatin provides access to DNA-binding proteins for the correct spatiotemporal regulation of gene expression. Mapping chromatin accessibility has been widely used to identify the location of cis regulatory elements (CREs) including promoters and enhancers. CREs show tissue- and cell-type specificity and disease-associated variants are often enriched for CREs in the tissues and cells that pertain to a given disease. To better understand the role of CREs in neuropsychiatric disorders we applied the Assay for Transposase Accessible Chromatin followed by sequencing (ATAC-seq) to neuronal and non-neuronal nuclei isolated from frozen postmortem human brain by fluorescence-activated nuclear sorting (FANS). Most of the identified open chromatin regions (OCRs) are differentially accessible between neurons and non-neurons, and show enrichment with known cell type markers, promoters and enhancers. Relative to those of non-neurons, neuronal OCRs are more evolutionarily conserved and are enriched in distal regulatory elements. Transcription factor (TF) footprinting analysis identifies differences in the regulome between neuronal and non-neuronal cells and ascribes putative functional roles to a number of non-coding schizophrenia (SCZ) risk variants. Among the identified variants is a Single Nucleotide Polymorphism (SNP) proximal to the gene encoding SNX19. In vitro experiments reveal that this SNP leads to an increase in transcriptional activity. As elevated expression of SNX19 has been associated with SCZ, our data provide evidence that the identified SNP contributes to disease. These results represent the first analysis of OCRs and TF-binding sites in distinct populations of postmortem human brain cells and further our understanding of the regulome and the impact of neuropsychiatric disease-associated genetic risk variants.
Collapse
Affiliation(s)
- John F Fullard
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Claudia Giambartolomei
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mads E Hauberg
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Biomedicine.,Centre for Integrative Sequencing (iSEQ), Aarhus University, Aarhus, Denmark.,The Lundbeck Foundation Initiative of Integrative Psychiatric Research (iPSYCH), Denmark
| | - Ke Xu
- Department of Genetics and Genomic Science and Institute for Multiscale Biology
| | - Georgios Voloudakis
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zhiping Shao
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Neuroscience.,Center for Molecular Biology and Genetics of Neurodegeneration
| | - Christopher Bare
- Flow Cytometry Center of Research Excellence, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joel T Dudley
- Department of Genetics and Genomic Science and Institute for Multiscale Biology
| | - Manuel Mattheisen
- Department of Biomedicine.,Centre for Integrative Sequencing (iSEQ), Aarhus University, Aarhus, Denmark.,The Lundbeck Foundation Initiative of Integrative Psychiatric Research (iPSYCH), Denmark
| | - Nikolaos K Robakis
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Neuroscience.,Center for Molecular Biology and Genetics of Neurodegeneration
| | - Vahram Haroutunian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Neuroscience.,Mental Illness Research, Education, and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, NY, USA
| | - Panos Roussos
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Genetics and Genomic Science and Institute for Multiscale Biology.,Mental Illness Research, Education, and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, NY, USA
| |
Collapse
|
22
|
Lu X, Ding Y, Niu Q, Xuan S, Yang Y, Jin Y, Wang H. ClC-3 chloride channel mediates the role of parathyroid hormone [1-34] on osteogenic differentiation of osteoblasts. PLoS One 2017; 12:e0176196. [PMID: 28437476 PMCID: PMC5402952 DOI: 10.1371/journal.pone.0176196] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 04/06/2017] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Different concentrations of parathyroid hormone [1-34] (PTH [1-34]) can have totally opposite effects on osteoblasts. Intermittent stimulation with PTH can significantly increase bone mineral density in vitro, mainly through the protein kinase A (PKA) signaling pathway, which phosphorylates runt-related transcription factor 2 (Runx2). The ClC-3 chloride channel, an important anion channel, can also promote osteogenesis via the Runx2 pathway based on recent studies. The purpose of our study, therefore, is to research whether the ClC-3 chloride channel has an effect on PTH osteodifferentiation in MC3T3-E1 cells. METHODS AND RESULTS A cell counting kit (CCK-8) and real-time PCR were used to investigate the impact of different PTH stimulation modes on MC3T3-E1 cell proliferation and osteogenesis-related gene expression, respectively. We found that the minimum inhibitory concentration of PTH was 10-9 M, and the expression of alkaline phosphatase (Alpl) and Runx2 were at the highest levels when treated with 10-9 M PTH. Next, we used real-time PCR and immunofluorescence technique to detect changes in ClC-3 in MC3T3-E1 cells under PTH treatment. The results showed higher expression of the ClC-3 chloride channel at 10-9 M intermittent PTH administration than in the other groups. Finally, we used the ClC-3 siRNA technique to examine the role of the ClC-3 chloride channel in the effect of PTH on the osteogenesis of osteoblasts, and we found an obvious decrease in the expression of bone sialoprotein (Ibsp), osteocalcin (Bglap), osterix (Sp7), Alpl and Runx2, the formation of mineralization nodules as well. CONCLUSIONS From the above data, we conclude that the expression of ClC-3 chloride channels in osteoblasts helps them respond to PTH stimulation, which mediates osteogenic differentiation.
Collapse
Affiliation(s)
- Xiaolin Lu
- State Key Laboratory of Military Stomatology, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yin Ding
- State Key Laboratory of Military Stomatology, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Qiannan Niu
- State Key Laboratory of Military Stomatology, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Shijie Xuan
- State Key Laboratory of Military Stomatology, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yan Yang
- State Key Laboratory of Military Stomatology, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yulong Jin
- Department of Hematology, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Huan Wang
- State Key Laboratory of Military Stomatology, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi, China
- * E-mail:
| |
Collapse
|
23
|
Farsi Z, Jahn R, Woehler A. Proton electrochemical gradient: Driving and regulating neurotransmitter uptake. Bioessays 2017; 39. [PMID: 28383767 DOI: 10.1002/bies.201600240] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Accumulation of neurotransmitters in the lumen of synaptic vesicles (SVs) relies on the activity of the vacuolar-type H+ -ATPase. This pump drives protons into the lumen, generating a proton electrochemical gradient (ΔμH+ ) across the membrane. Recent work has demonstrated that the balance between the chemical (ΔpH) and electrical (ΔΨ) components of ΔμH+ is regulated differently by some distinct vesicle types. As different neurotransmitter transporters use ΔpH and ΔΨ with different relative efficiencies, regulation of this gradient balance has the potential to influence neurotransmitter uptake. Nevertheless, the underlying mechanisms responsible for this regulation remain poorly understood. In this review, we provide an overview of current neurotransmitter uptake models, with a particular emphasis on the distinct roles of the electrical and chemical gradients and current hypotheses for regulatory mechanisms.
Collapse
Affiliation(s)
- Zohreh Farsi
- Max-Delbrück Center for Molecular Medicine, Berlin Institute for Medical Systems Biology, Berlin, Germany
| | - Reinhard Jahn
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Andrew Woehler
- Max-Delbrück Center for Molecular Medicine, Berlin Institute for Medical Systems Biology, Berlin, Germany
| |
Collapse
|
24
|
Albers HE, Walton JC, Gamble KL, McNeill JK, Hummer DL. The dynamics of GABA signaling: Revelations from the circadian pacemaker in the suprachiasmatic nucleus. Front Neuroendocrinol 2017; 44:35-82. [PMID: 27894927 PMCID: PMC5225159 DOI: 10.1016/j.yfrne.2016.11.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/16/2016] [Accepted: 11/22/2016] [Indexed: 12/31/2022]
Abstract
Virtually every neuron within the suprachiasmatic nucleus (SCN) communicates via GABAergic signaling. The extracellular levels of GABA within the SCN are determined by a complex interaction of synthesis and transport, as well as synaptic and non-synaptic release. The response to GABA is mediated by GABAA receptors that respond to both phasic and tonic GABA release and that can produce excitatory as well as inhibitory cellular responses. GABA also influences circadian control through the exclusively inhibitory effects of GABAB receptors. Both GABA and neuropeptide signaling occur within the SCN, although the functional consequences of the interactions of these signals are not well understood. This review considers the role of GABA in the circadian pacemaker, in the mechanisms responsible for the generation of circadian rhythms, in the ability of non-photic stimuli to reset the phase of the pacemaker, and in the ability of the day-night cycle to entrain the pacemaker.
Collapse
Affiliation(s)
- H Elliott Albers
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Neuroscience Institute, Georgia State University, Atlanta, GA 30302, United States.
| | - James C Walton
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Neuroscience Institute, Georgia State University, Atlanta, GA 30302, United States
| | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - John K McNeill
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Neuroscience Institute, Georgia State University, Atlanta, GA 30302, United States
| | - Daniel L Hummer
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Department of Psychology, Morehouse College, Atlanta, GA 30314, United States
| |
Collapse
|
25
|
Pang RP, Xie MX, Yang J, Shen KF, Chen X, Su YX, Yang C, Tao J, Liang SJ, Zhou JG, Zhu HQ, Wei XH, Li YY, Qin ZH, Liu XG. Downregulation of ClC-3 in dorsal root ganglia neurons contributes to mechanical hypersensitivity following peripheral nerve injury. Neuropharmacology 2016; 110:181-189. [DOI: 10.1016/j.neuropharm.2016.07.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 06/28/2016] [Accepted: 07/19/2016] [Indexed: 01/18/2023]
|
26
|
Eriksen J, Chang R, McGregor M, Silm K, Suzuki T, Edwards RH. Protons Regulate Vesicular Glutamate Transporters through an Allosteric Mechanism. Neuron 2016; 90:768-80. [PMID: 27133463 DOI: 10.1016/j.neuron.2016.03.026] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 02/25/2016] [Accepted: 03/21/2016] [Indexed: 11/25/2022]
Abstract
The quantal nature of synaptic transmission requires a mechanism to transport neurotransmitter into synaptic vesicles without promoting non-vesicular efflux across the plasma membrane. Indeed, the vesicular transport of most classical transmitters involves a mechanism of H(+) exchange, which restricts flux to acidic membranes such as synaptic vesicles. However, vesicular transport of the principal excitatory transmitter glutamate depends primarily on membrane potential, which would drive non-vesicular efflux, and the role of protons is unclear. Adapting electrophysiology to record currents associated with the vesicular glutamate transporters (VGLUTs), we characterize a chloride conductance that is gated by lumenal protons and chloride and supports glutamate uptake. Rather than coupling stoichiometrically to glutamate flux, lumenal protons and chloride allosterically activate vesicular glutamate transport. Gating by protons serves to inhibit what would otherwise be substantial non-vesicular glutamate efflux at the plasma membrane, thereby restricting VGLUT activity to synaptic vesicles.
Collapse
Affiliation(s)
- Jacob Eriksen
- Department of Physiology, UCSF School of Medicine, San Francisco, CA 94143, USA; Department of Neurology, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Roger Chang
- Department of Physiology, UCSF School of Medicine, San Francisco, CA 94143, USA; Department of Neurology, UCSF School of Medicine, San Francisco, CA 94143, USA; Graduate Program in Biomedical Sciences, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Matt McGregor
- Department of Physiology, UCSF School of Medicine, San Francisco, CA 94143, USA; Department of Neurology, UCSF School of Medicine, San Francisco, CA 94143, USA; Graduate Program in Neuroscience, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Katlin Silm
- Department of Physiology, UCSF School of Medicine, San Francisco, CA 94143, USA; Department of Neurology, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Toshiharu Suzuki
- Faculty of Science and Engineering, Waseda University, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Robert H Edwards
- Department of Physiology, UCSF School of Medicine, San Francisco, CA 94143, USA; Department of Neurology, UCSF School of Medicine, San Francisco, CA 94143, USA; Graduate Program in Biomedical Sciences, UCSF School of Medicine, San Francisco, CA 94143, USA; Graduate Program in Neuroscience, UCSF School of Medicine, San Francisco, CA 94143, USA.
| |
Collapse
|
27
|
Farsi Z, Preobraschenski J, van den Bogaart G, Riedel D, Jahn R, Woehler A. Single-vesicle imaging reveals different transport mechanisms between glutamatergic and GABAergic vesicles. Science 2016; 351:981-4. [DOI: 10.1126/science.aad8142] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/14/2016] [Indexed: 12/13/2022]
|
28
|
Takamori S. Presynaptic Molecular Determinants of Quantal Size. Front Synaptic Neurosci 2016; 8:2. [PMID: 26903855 PMCID: PMC4744840 DOI: 10.3389/fnsyn.2016.00002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 01/25/2016] [Indexed: 01/22/2023] Open
Abstract
The quantal hypothesis for the release of neurotransmitters at the chemical synapse has gained wide acceptance since it was first worked out at the motor endplate in frog skeletal muscle in the 1950’s. Considering the morphological identification of synaptic vesicles (SVs) at the nerve terminals that appeared to be homogeneous in size, the hypothesis proposed that signal transduction at synapses is mediated by the release of neurotransmitters packed in SVs that are individually uniform in size; the amount of transmitter in a synaptic vesicle is called a quantum. Although quantal size—the amplitude of the postsynaptic response elicited by the release of neurotransmitters from a single vesicle—clearly depends on the number and sensitivity of the postsynaptic receptors, accumulating evidence has also indicated that the amount of neurotransmitters stored in SVs can be altered by various presynaptic factors. Here, I provide an overview of the concepts and underlying presynaptic molecular underpinnings that may regulate quantal size.
Collapse
Affiliation(s)
- Shigeo Takamori
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University Kyoto, Japan
| |
Collapse
|
29
|
The plastic neurotransmitter phenotype of the hippocampal granule cells and of the moss in their messy fibers. J Chem Neuroanat 2015; 73:9-20. [PMID: 26703784 DOI: 10.1016/j.jchemneu.2015.11.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 10/29/2015] [Accepted: 11/03/2015] [Indexed: 01/09/2023]
Abstract
The granule cells (GCs) and their axons, the mossy fibers (MFs), make synapses with interneurons in the hilus and CA3 area of the hippocampus and with pyramidal cells of CA3, each with distinct anatomical and functional characteristics. Many features of synaptic communication observed at the MF synapses are not usually observed in most cortical synapses, and thus have drawn the attention of many groups studying different aspects of the transmission of information. One particular aspect of the GCs, that makes their study unique, is that they express a dual glutamatergic-GABAergic phenotype and several groups have contributed to the understanding of how two neurotransmitters of opposing actions can act on a single target when simultaneously released. Indeed, the GCs somata and their mossy fibers express in a regulated manner glutamate and GABA, GAD, VGlut and VGAT, all markers of both phenotypes. Finally, their activation provokes both glutamate-R-mediated and GABA-R-mediated synaptic responses in the postsynaptic cell targets and even in the MFs themselves. The developmental and activity-dependent expression of these phenotypes seems to follow a "logical" way to maintain an excitation-inhibition balance of the dentate gyrus-to-CA3 communication.
Collapse
|
30
|
Guzman RE, Miranda-Laferte E, Franzen A, Fahlke C. Neuronal ClC-3 Splice Variants Differ in Subcellular Localizations, but Mediate Identical Transport Functions. J Biol Chem 2015; 290:25851-62. [PMID: 26342074 DOI: 10.1074/jbc.m115.668186] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Indexed: 01/06/2023] Open
Abstract
ClC-3 is a member of the CLC family of anion channels and transporters, for which multiple functional properties and subcellular localizations have been reported. Since alternative splicing often results in proteins with diverse properties, we investigated to what extent alternative splicing might influence subcellular targeting and function of ClC-3. We identified three alternatively spliced ClC-3 isoforms, ClC-3a, ClC-3b, and ClC-3c, in mouse brain, with ClC-3c being the predominant splice variant. Whereas ClC-3a and ClC-3b are present in late endosomes/lysosomes, ClC-3c is targeted to recycling endosomes via a novel N-terminal isoleucine-proline (IP) motif. Surface membrane insertion of a fraction of ClC-3c transporters permitted electrophysiological characterization of this splice variant through whole-cell patch clamping on transfected mammalian cells. In contrast, neutralization of the N-terminal dileucine-like motifs was required for functional analysis of ClC-3a and ClC-3b. Heterologous expression of ClC-3a or ClC-3b carrying mutations in N-terminal dileucine motifs as well as WTClC-3c in HEK293T cells resulted in outwardly rectifying Cl(-) currents with significant capacitive current components. We conclude that alternative splicing of Clcn3 results in proteins with different subcellular localizations, but leaves the transport function of the proteins unaffected.
Collapse
Affiliation(s)
- Raul E Guzman
- From the Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Erick Miranda-Laferte
- From the Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Arne Franzen
- From the Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Christoph Fahlke
- From the Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, 52425 Jülich, Germany
| |
Collapse
|
31
|
Jentsch TJ. Discovery of CLC transport proteins: cloning, structure, function and pathophysiology. J Physiol 2015; 593:4091-109. [PMID: 25590607 DOI: 10.1113/jp270043] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/11/2015] [Indexed: 02/06/2023] Open
Abstract
After providing a personal description of the convoluted path leading 25 years ago to the molecular identification of the Torpedo Cl(-) channel ClC-0 and the discovery of the CLC gene family, I succinctly describe the general structural and functional features of these ion transporters before giving a short overview of mammalian CLCs. These can be categorized into plasma membrane Cl(-) channels and vesicular Cl(-) /H(+) -exchangers. They are involved in the regulation of membrane excitability, transepithelial transport, extracellular ion homeostasis, endocytosis and lysosomal function. Diseases caused by CLC dysfunction include myotonia, neurodegeneration, deafness, blindness, leukodystrophy, male infertility, renal salt loss, kidney stones and osteopetrosis, revealing a surprisingly broad spectrum of biological roles for chloride transport that was unsuspected when I set out to clone the first voltage-gated chloride channel.
Collapse
Affiliation(s)
- Thomas J Jentsch
- Leibniz-Institut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| |
Collapse
|
32
|
Wolfart J, Laker D. Homeostasis or channelopathy? Acquired cell type-specific ion channel changes in temporal lobe epilepsy and their antiepileptic potential. Front Physiol 2015; 6:168. [PMID: 26124723 PMCID: PMC4467176 DOI: 10.3389/fphys.2015.00168] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/19/2015] [Indexed: 01/16/2023] Open
Abstract
Neurons continuously adapt the expression and functionality of their ion channels. For example, exposed to chronic excitotoxicity, neurons homeostatically downscale their intrinsic excitability. In contrast, the “acquired channelopathy” hypothesis suggests that proepileptic channel characteristics develop during epilepsy. We review cell type-specific channel alterations under different epileptic conditions and discuss the potential of channels that undergo homeostatic adaptations, as targets for antiepileptic drugs (AEDs). Most of the relevant studies have been performed on temporal lobe epilepsy (TLE), a widespread AED-refractory, focal epilepsy. The TLE patients, who undergo epilepsy surgery, frequently display hippocampal sclerosis (HS), which is associated with degeneration of cornu ammonis subfield 1 pyramidal cells (CA1 PCs). Although the resected human tissue offers insights, controlled data largely stem from animal models simulating different aspects of TLE and other epilepsies. Most of the cell type-specific information is available for CA1 PCs and dentate gyrus granule cells (DG GCs). Between these two cell types, a dichotomy can be observed: while DG GCs acquire properties decreasing the intrinsic excitability (in TLE models and patients with HS), CA1 PCs develop channel characteristics increasing intrinsic excitability (in TLE models without HS only). However, thorough examination of data on these and other cell types reveals the coexistence of protective and permissive intrinsic plasticity within neurons. These mechanisms appear differentially regulated, depending on the cell type and seizure condition. Interestingly, the same channel molecules that are upregulated in DG GCs during HS-related TLE, appear as promising targets for future AEDs and gene therapies. Hence, GCs provide an example of homeostatic ion channel adaptation which can serve as a primer when designing novel anti-epileptic strategies.
Collapse
Affiliation(s)
- Jakob Wolfart
- Oscar Langendorff Institute of Physiology, University of Rostock Rostock, Germany
| | - Debora Laker
- Oscar Langendorff Institute of Physiology, University of Rostock Rostock, Germany
| |
Collapse
|
33
|
Huang LY, He Q, Liang SJ, Su YX, Xiong LX, Wu QQ, Wu QY, Tao J, Wang JP, Tang YB, Lv XF, Liu J, Guan YY, Pang RP, Zhou JG. ClC-3 chloride channel/antiporter defect contributes to inflammatory bowel disease in humans and mice. Gut 2014; 63:1587-95. [PMID: 24440986 DOI: 10.1136/gutjnl-2013-305168] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND ClC-3 channel/antiporter plays a critical role in a variety of cellular activities. ClC-3 has been detected in the ileum and colon. OBJECTIVE To determine the functions of ClC-3 in the gastrointestinal tract. DESIGN After administration of dextran sulfate sodium (DSS) or 2,4,6-trinitrobenzenesulfonic acid (TNBS), intestines from ClC-3-/- and wild-type mice were examined by histological, cellular, molecular and biochemical approaches. ClC-3 expression was determined by western blot and immunostaining. RESULTS ClC-3 expression was reduced in intestinal tissues from patients with UC or Crohn's disease and from mice treated with DSS. Genetic deletion of ClC-3 increased the susceptibility of mice to DSS- or TNBS-induced experimental colitis and prevented intestinal recovery. ClC-3 deficiency promoted DSS-induced apoptosis of intestinal epithelial cells through the mitochondria pathway. ClC-3 interacts with voltage-dependent anion channel 1, a key player in regulation of mitochondria cytochrome c release, but DSS treatment decreased this interaction. In addition, lack of ClC-3 reduced the numbers of Paneth cells and impaired the expression of antimicrobial peptides. These alterations led to dysfunction of the epithelial barrier and invasion of commensal bacteria into the mucosa. CONCLUSIONS A defect in ClC-3 may contribute to the pathogenesis of IBD by promoting intestinal epithelial cell apoptosis and Paneth cell loss, suggesting that modulation of ClC-3 expression might be a new strategy for the treatment of IBD.
Collapse
Affiliation(s)
- Lin-Yan Huang
- Department of Pharmacology, Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, Guangdong, China School of Medical Technology, Xuzhou Medical College, Xuzhou, Jiagsu, China
| | - Qing He
- Gastrointestinal Institute, the 6th Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Si-Jia Liang
- Department of Pharmacology, Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Ying-Xue Su
- Department of Pharmacology, Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Li-Xiong Xiong
- Department of Pharmacology, Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Qian-Qian Wu
- Department of Pharmacology, Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Qin-Yan Wu
- Gastrointestinal Institute, the 6th Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jing Tao
- Department of Pharmacology, Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jian-Ping Wang
- Department of Colorectal Surgery, The 6th Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yong-Bo Tang
- Department of Pharmacology, Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiao-Fei Lv
- Department of Pharmacology, Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jie Liu
- Department of Pharmacology, Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yong-Yuan Guan
- Department of Pharmacology, Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Rui-Ping Pang
- Department of Physiology, Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jia-Guo Zhou
- Department of Pharmacology, Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
34
|
Cuddapah VA, Robel S, Watkins S, Sontheimer H. A neurocentric perspective on glioma invasion. Nat Rev Neurosci 2014; 15:455-65. [PMID: 24946761 PMCID: PMC5304245 DOI: 10.1038/nrn3765] [Citation(s) in RCA: 583] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Malignant gliomas are devastating tumours that frequently kill patients within 1 year of diagnosis. The major obstacle to a cure is diffuse invasion, which enables tumours to escape complete surgical resection and chemo- and radiation therapy. Gliomas use the same tortuous extracellular routes of migration that are travelled by immature neurons and stem cells, frequently using blood vessels as guides. They repurpose ion channels to dynamically adjust their cell volume to accommodate to narrow spaces and breach the blood-brain barrier through disruption of astrocytic endfeet, which envelop blood vessels. The unique biology of glioma invasion provides hitherto unexplored brain-specific therapeutic targets for this devastating disease.
Collapse
Affiliation(s)
- Vishnu Anand Cuddapah
- Department of Neurobiology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, 1719 6th Avenue South, CIRC 425, Birmingham, Alabama 35294, USA
| | - Stefanie Robel
- Department of Neurobiology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, 1719 6th Avenue South, CIRC 425, Birmingham, Alabama 35294, USA
| | - Stacey Watkins
- Department of Neurobiology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, 1719 6th Avenue South, CIRC 425, Birmingham, Alabama 35294, USA
| | - Harald Sontheimer
- Department of Neurobiology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, 1719 6th Avenue South, CIRC 425, Birmingham, Alabama 35294, USA
| |
Collapse
|
35
|
Swelling-activated Cl− currents and intracellular CLC-3 are involved in proliferation of human pulmonary artery smooth muscle cells. J Hypertens 2014; 32:318-30. [DOI: 10.1097/hjh.0000000000000013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
36
|
Tarasenko A, Krupko O, Himmelreich N. New insights into molecular mechanism(s) underlying the presynaptic action of nitric oxide on GABA release. Biochim Biophys Acta Gen Subj 2014; 1840:1923-32. [PMID: 24480299 DOI: 10.1016/j.bbagen.2014.01.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 01/17/2014] [Accepted: 01/21/2014] [Indexed: 12/31/2022]
Abstract
BACKGROUND Nitric oxide (NO) is an important presynaptic modulator of synaptic transmission. Here, we aimed to correlate the release of the major inhibitory neurotransmitter GABA with intracellular events occurring in rat brain axon terminals during their exposure to NO in the range of nanomolar-low micromolar concentrations. METHODS Using [(3)H]GABA and fluorescent dyes (Fluo 4-AM, acridine orange and rhodamine 6G), the following parameters were evaluated: vesicular and cytosolic GABA pools, intracellular calcium concentration, synaptic vesicle acidification, and mitochondrial membrane potential. Diethylamine NONOate (DEA/NO) and S-nitroso-N-acetylpenicillamine (SNAP) were used as NO donors. RESULTS DEA/NO and SNAP (in the presence of dithiothreitol (DTT)) stimulated external Ca(2+)-independent [(3)H]GABA release, which was not attributed to a rise in intracellular calcium concentration. [(3)H]GABA release coincided with increasing GABA level in cytosol and decreasing the vesicular GABA content available for exocytotic release. There was a strong temporal correlation between NO-induced increase in cytosolic [GABA] and dissipation of both synaptic vesicle proton gradient and mitochondrial membrane potential. Dissipation was reversible, and recovery of both parameters correlated in time with re-accumulation of [(3)H]GABA into synaptic vesicles. The molar ratio of DTT to SNAP determined the rate and duration of the recovery processes. CONCLUSIONS We suggest that NO can stimulate GABA release via GABA transporter reversal resulting from increased GABA levels in cytosol. The latter is reversible and appears to be due to S-nitrosylation of key proteins, which affect the energy status of the pre-synapse. GENERAL SIGNIFICANCE Our findings provide new insight into molecular mechanism(s) underlying the presynaptic action of nitric oxide on inhibitory neurotransmission.
Collapse
Affiliation(s)
- Alla Tarasenko
- Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Leontovich Str. 9, Kyiv 01601, Ukraine.
| | - Olga Krupko
- Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Leontovich Str. 9, Kyiv 01601, Ukraine.
| | - Nina Himmelreich
- Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Leontovich Str. 9, Kyiv 01601, Ukraine.
| |
Collapse
|
37
|
Stauber T, Weinert S, Jentsch TJ. Cell biology and physiology of CLC chloride channels and transporters. Compr Physiol 2013; 2:1701-44. [PMID: 23723021 DOI: 10.1002/cphy.c110038] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Proteins of the CLC gene family assemble to homo- or sometimes heterodimers and either function as Cl(-) channels or as Cl(-)/H(+)-exchangers. CLC proteins are present in all phyla. Detailed structural information is available from crystal structures of bacterial and algal CLCs. Mammals express nine CLC genes, four of which encode Cl(-) channels and five 2Cl(-)/H(+)-exchangers. Two accessory β-subunits are known: (1) barttin and (2) Ostm1. ClC-Ka and ClC-Kb Cl(-) channels need barttin, whereas Ostm1 is required for the function of the lysosomal ClC-7 2Cl(-)/H(+)-exchanger. ClC-1, -2, -Ka and -Kb Cl(-) channels reside in the plasma membrane and function in the control of electrical excitability of muscles or neurons, in extra- and intracellular ion homeostasis, and in transepithelial transport. The mainly endosomal/lysosomal Cl(-)/H(+)-exchangers ClC-3 to ClC-7 may facilitate vesicular acidification by shunting currents of proton pumps and increase vesicular Cl(-) concentration. ClC-3 is also present on synaptic vesicles, whereas ClC-4 and -5 can reach the plasma membrane to some extent. ClC-7/Ostm1 is coinserted with the vesicular H(+)-ATPase into the acid-secreting ruffled border membrane of osteoclasts. Mice or humans lacking ClC-7 or Ostm1 display osteopetrosis and lysosomal storage disease. Disruption of the endosomal ClC-5 Cl(-)/H(+)-exchanger leads to proteinuria and Dent's disease. Mouse models in which ClC-5 or ClC-7 is converted to uncoupled Cl(-) conductors suggest an important role of vesicular Cl(-) accumulation in these pathologies. The important functions of CLC Cl(-) channels were also revealed by human diseases and mouse models, with phenotypes including myotonia, renal loss of salt and water, deafness, blindness, leukodystrophy, and male infertility.
Collapse
Affiliation(s)
- Tobias Stauber
- Leibniz-Institut für Molekulare Pharmakologie FMP and Max-Delbrück-Centrum für Molekulare Medizin MDC, Berlin, Germany
| | | | | |
Collapse
|
38
|
Münster-Wandowski A, Gómez-Lira G, Gutiérrez R. Mixed neurotransmission in the hippocampal mossy fibers. Front Cell Neurosci 2013; 7:210. [PMID: 24319410 PMCID: PMC3837298 DOI: 10.3389/fncel.2013.00210] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/23/2013] [Indexed: 01/14/2023] Open
Abstract
The hippocampal mossy fibers (MFs), the axons of the granule cells (GCs) of the dentate gyrus, innervate mossy cells and interneurons in the hilus on their way to CA3 where they innervate interneurons and pyramidal cells. Synapses on each target cell have distinct anatomical and functional characteristics. In recent years, the paradigmatic view of the MF synapses being only glutamatergic and, thus, excitatory has been questioned. Several laboratories have provided data supporting the hypothesis that the MFs can transiently release GABA during development and, in the adult, after periods of enhanced excitability. This transient glutamate-GABA co-transmission coincides with the transient up-regulation of the machinery for the synthesis and release of GABA in the glutamatergic GCs. Although some investigators have deemed this evidence controversial, new data has appeared with direct evidence of co-release of glutamate and GABA from single, identified MF boutons. However, this must still be confirmed by other groups and with other methodologies. A second, intriguing observation is that MF activation produced fast spikelets followed by excitatory postsynaptic potentials in a number of pyramidal cells, which, unlike the spikelets, underwent frequency potentiation and were strongly depressed by activation of metabotropic glutamate receptors. The spikelets persisted during blockade of chemical transmission and were suppressed by the gap junction blocker carbenoxolone. These data are consistent with the hypothesis of mixed electrical-chemical synapses between MFs and some pyramidal cells. Dye coupling between these types of principal cells and ultrastructural studies showing the co-existence of AMPA receptors and connexin 36 in this synapse corroborate their presence. A deeper consideration of mixed neurotransmission taking place in this synapse may expand our search and understanding of communication channels between different regions of the mammalian CNS.
Collapse
|
39
|
Dong C, Hu A, Ni Y, Zuo Y, Li GH. Effects of midazolam, pentobarbital and ketamine on the mRNA expression of ion channels in a model organism Daphnia pulex. BMC Anesthesiol 2013; 13:32. [PMID: 24134334 PMCID: PMC3879215 DOI: 10.1186/1471-2253-13-32] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 10/07/2013] [Indexed: 02/05/2023] Open
Abstract
Background Over the last few decades intensive studies have been carried out on the molecular targets mediating general anesthesia as well as the effects of general anesthetics. The γ-aminobutyric acid type A receptor (GABAAR) has been indicated as the primary target of general anaesthetics such as propofol, etomidate and isoflurane, and sedating drugs including benzodiazepines and barbiturates. The GABAAR is also involved in drug tolerance and dependence. However, the involvement of other ion channels is possible. Methods Using reverse transcription and quantitative PCR techniques, we systematically investigated changes in the mRNA levels of ion channel genes in response to exposure to midazolam, pentobarbital and ketamine in a freshwater model animal, Daphnia pulex. To retrieve the sequences of Daphnia ion channel genes, Blast searches were performed based on known human or Drosophila ion channel genes. Retrieved sequences were clustered with the maximum-likelihood method. To quantify changes in gene expression after the drug treatments for 4 hours, total RNA was extracted and reverse transcribed into cDNA and then amplified using quantitative PCR. Results A total of 108 ion channel transcripts were examined, and 19, 11 and 11 of them are affected by midazolam (100 μM), pentobarbital (200 μM) and ketamine (100 μM), respectively, covering a wide variety of ion channel types. There is some degree of overlap with midazolam- and pentobarbital-induced changes in the mRNA expression profiles, but ketamine causes distinct changes in gene expression pattern. In addition, flumazenil (10 μM) eliminates the effect of midazolam on the mRNA expression of the GABAA receptor subunit Rdl, suggesting a direct interaction between midazolam and GABAA receptors. Conclusions Recent research using high throughput technology suggests that changes in mRNA expression correlate with delayed protein expression. Therefore, the mRNA profile changes in our study may reflect the molecular targets not only in drug actions, but also in chronic drug addiction. Our data also suggest the possibility that hypnotic/anesthetic drugs are capable of altering the functions of the nervous system, as well as those non-nerve tissues with abundant ion channel expressions.
Collapse
Affiliation(s)
| | | | | | | | - Guo Hua Li
- Laboratory of Anesthesiology and Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
40
|
Wang L, Tu P, Bonet L, Aubrey KR, Supplisson S. Cytosolic transmitter concentration regulates vesicle cycling at hippocampal GABAergic terminals. Neuron 2013; 80:143-58. [PMID: 24094108 DOI: 10.1016/j.neuron.2013.07.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2013] [Indexed: 10/26/2022]
Abstract
Sustained synaptic transmission requires vesicle recycling and refilling with transmitter, two processes considered to proceed independently. Contrary to this assumption, we show here that depletion of cytosolic transmitter at GABAergic synapses reversibly reduces the number of recycling vesicles. Using paired recordings in hippocampal cultures, we show that repetitive activity causes two phases of reduction of the postsynaptic response. The first involves the classical depletion of the readily releasable and recycling pools, while the second reflects impairment of vesicle filling as GABA is consumed, since it can only be reversed by uptake of GABA or its precursors, glutamate or glutamine. Surprisingly, this second phase is associated with reduced quantal release, a faster depression rate and lower FM5-95 labeling, suggesting that the size of the cycling vesicular pool is regulated by cytosolic transmitter availability. Regulation of vesicular cycling may represent a general mechanism of presynaptic plasticity, matching synaptic release to transmitter supply.
Collapse
Affiliation(s)
- Lu Wang
- INSERM, U1024, F-75005 Paris, France; CNRS, UMR 8197, F-75005 Paris, France; Ecole Normale Supérieure, Institut de Biologie de l'ENS, IBENS, F-75005 Paris, France; Shanghai Key Laboratory of Brain Functional Genomics, East China Normal University, 200062 Shanghai, China
| | | | | | | | | |
Collapse
|
41
|
Mendoza-Torreblanca JG, Vanoye-Carlo A, Phillips-Farfán BV, Carmona-Aparicio L, Gómez-Lira G. Synaptic vesicle protein 2A: basic facts and role in synaptic function. Eur J Neurosci 2013; 38:3529-39. [DOI: 10.1111/ejn.12360] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 08/09/2013] [Accepted: 08/17/2013] [Indexed: 10/26/2022]
Affiliation(s)
| | | | | | | | - Gisela Gómez-Lira
- Department of Pharmacobiology; Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional; Calzada de los Tenorios 235 Col. Granjas Coapa C.P. 14330 D. F., Mexico
| |
Collapse
|
42
|
Van Liefferinge J, Massie A, Portelli J, Di Giovanni G, Smolders I. Are vesicular neurotransmitter transporters potential treatment targets for temporal lobe epilepsy? Front Cell Neurosci 2013; 7:139. [PMID: 24009559 PMCID: PMC3757300 DOI: 10.3389/fncel.2013.00139] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 08/11/2013] [Indexed: 12/18/2022] Open
Abstract
The vesicular neurotransmitter transporters (VNTs) are small proteins responsible for packing synaptic vesicles with neurotransmitters thereby determining the amount of neurotransmitter released per vesicle through fusion in both neurons and glial cells. Each transporter subtype was classically seen as a specific neuronal marker of the respective nerve cells containing that particular neurotransmitter or structurally related neurotransmitters. More recently, however, it has become apparent that common neurotransmitters can also act as co-transmitters, adding complexity to neurotransmitter release and suggesting intriguing roles for VNTs therein. We will first describe the current knowledge on vesicular glutamate transporters (VGLUT1/2/3), the vesicular excitatory amino acid transporter (VEAT), the vesicular nucleotide transporter (VNUT), vesicular monoamine transporters (VMAT1/2), the vesicular acetylcholine transporter (VAChT) and the vesicular γ-aminobutyric acid (GABA) transporter (VGAT) in the brain. We will focus on evidence regarding transgenic mice with disruptions in VNTs in different models of seizures and epilepsy. We will also describe the known alterations and reorganizations in the expression levels of these VNTs in rodent models for temporal lobe epilepsy (TLE) and in human tissue resected for epilepsy surgery. Finally, we will discuss perspectives on opportunities and challenges for VNTs as targets for possible future epilepsy therapies.
Collapse
|
43
|
Guzman RE, Grieschat M, Fahlke C, Alekov AK. ClC-3 is an intracellular chloride/proton exchanger with large voltage-dependent nonlinear capacitance. ACS Chem Neurosci 2013; 4:994-1003. [PMID: 23509947 DOI: 10.1021/cn400032z] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The chloride/proton exchangers ClC-3, ClC-4 and ClC-5 are localized in distinct intracellular compartments and regulate their luminal acidity. We used electrophysiology combined with fluorescence pH measurements to compare the functions of these three transporters. Since the expression of WT ClC-3 in the surface membrane was negligible, we removed an N-terminal retention signal for standard electrophysiological characterization of this isoform. This construct (ClC-313-19A) mediated outwardly rectifying coupled Cl(-)/H(+) antiport resembling the properties of ClC-4 and ClC-5. In addition, ClC-3 exhibited large electric capacitance, exceeding the nonlinear capacitances of ClC-4 and ClC-5. Mutations of the proton glutamate, a conserved residue at the internal side of the protein, decreased ion transport but increased nonlinear capacitances in all three isoforms. This suggests that nonlinear capacitances in mammalian ClC transporters are regulated in a similar manner. However, the voltage dependence and the amplitudes of these capacitances differed strongly between the investigated isoforms. Our results indicate that ClC-3 is specialized in mainly performing incomplete capacitive nontransporting cycles, that ClC-4 is an effective coupled transporter, and that ClC-5 displays an intermediate phenotype. Mathematical modeling showed that such functional differences would allow differential regulation of luminal acidification and chloride concentration in intracellular compartments.
Collapse
Affiliation(s)
- Raul E. Guzman
- Institut für Neurophysiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
- Institute of Complex Systems, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Matthias Grieschat
- Institut für Neurophysiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Christoph Fahlke
- Institut für Neurophysiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
- Institute of Complex Systems, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Alexi K. Alekov
- Institut für Neurophysiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| |
Collapse
|
44
|
Chen TT, Klassen TL, Goldman AM, Marini C, Guerrini R, Noebels JL. Novel brain expression of ClC-1 chloride channels and enrichment of CLCN1 variants in epilepsy. Neurology 2013; 80:1078-85. [PMID: 23408874 DOI: 10.1212/wnl.0b013e31828868e7] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To explore the potential contribution of genetic variation in voltage-gated chloride channels to epilepsy, we analyzed CLCN family (CLCN1-7) gene variant profiles in individuals with complex idiopathic epilepsy syndromes and determined the expression of these channels in human and murine brain. METHODS We used parallel exomic sequencing of 237 ion channel subunit genes to screen individuals with a clinical diagnosis of idiopathic epilepsy and evaluate the distribution of missense variants in CLCN genes in cases and controls. We examined regional expression of CLCN1 in human and mouse brain using reverse transcriptase PCR, in situ hybridization, and Western immunoblotting. RESULTS We found that in 152 individuals with sporadic epilepsy of unknown origin, 96.7% had at least one missense variant in the CLCN genes compared with 28.2% of 139 controls. Nonsynonymous single nucleotide polymorphisms in the "skeletal" chloride channel gene CLCN1 and in CLCN2, a putative human epilepsy gene, were detected in threefold excess in cases relative to controls. Among these, we report a novel de novo CLCN1 truncation mutation in a patient with pharmacoresistant generalized seizures and a dystonic writer's cramp without evidence of variants in other channel genes linked to epilepsy. Molecular localization revealed the unexpectedly widespread presence of CLCN1 mRNA transcripts and the ClC-1 subunit protein in human and murine brain, previously believed absent in neurons. CONCLUSIONS Our findings support a possible comorbid contribution of the "skeletal" chloride channel ClC-1 to the regulation of brain excitability and the need for further elucidation of the roles of CLCN genes in neuronal network excitability disorders.
Collapse
Affiliation(s)
- Tim T Chen
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | | | | | | | | | | |
Collapse
|
45
|
Farmer LM, Le BN, Nelson DJ. CLC-3 chloride channels moderate long-term potentiation at Schaffer collateral-CA1 synapses. J Physiol 2012; 591:1001-15. [PMID: 23165767 DOI: 10.1113/jphysiol.2012.243485] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The chloride channel CLC-3 is expressed in the brain on synaptic vesicles and postsynaptic membranes. Although CLC-3 is broadly expressed throughout the brain, the CLC-3 knockout mouse shows complete, selective postnatal neurodegeneration of the hippocampus, suggesting a crucial role for the channel in maintaining normal brain function. CLC-3 channels are functionally linked to NMDA receptors in the hippocampus; NMDA receptor-dependent Ca(2+) entry, activation of Ca(2+)/calmodulin kinase II and subsequent gating of CLC-3 link the channels via a Ca(2+)-mediated feedback loop. We demonstrate that loss of CLC-3 at mature synapses increases long-term potentiation from 135 ± 4% in the wild-type slice preparation to 154 ± 7% above baseline (P < 0.001) in the knockout; therefore, the contribution of CLC-3 is to reduce synaptic potentiation by ∼40%. Using a decoy peptide representing the Ca(2+)/calmodulin kinase II phosphorylation site on CLC-3, we show that phosphorylation of CLC-3 is required for its regulatory function in long-term potentiation. CLC-3 is also expressed on synaptic vesicles; however, our data suggest functionally separable pre- and postsynaptic roles. Thus, CLC-3 confers Cl(-) sensitivity to excitatory synapses, controls the magnitude of long-term potentiation and may provide a protective limit on Ca(2+) influx.
Collapse
Affiliation(s)
- Laurel M Farmer
- Department of Neurobiology, Pharmacology and Physiology, The University of Chicago, Chicago, IL 60637, USA
| | | | | |
Collapse
|
46
|
Yang H, Huang LY, Zeng DY, Huang EW, Liang SJ, Tang YB, Su YX, Tao J, Shang F, Wu QQ, Xiong LX, Lv XF, Liu J, Guan YY, Zhou JG. Decrease of Intracellular Chloride Concentration Promotes Endothelial Cell Inflammation by Activating Nuclear Factor-κB Pathway. Hypertension 2012; 60:1287-93. [DOI: 10.1161/hypertensionaha.112.198648] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Hui Yang
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center (H.Y., L.-Y.H., D.-Y.Z., E-W.H., S.-J.L., Y.-B.T., Y.-X.S., J.T., F.S., Q.-Q.W., L.-X.X., X.-F.L., J.L., Y.-Y.G., J.-G.Z.), and Department of Forensic Pathology (E.-W.H.), Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Cardiovascular Institute of Guangdong Academy of Medical Sciences, Medical Research Center of Guangdong General Hospital, Guangzhou, China (H.Y.); Guangzhou Forensic Science
| | - Lin-Yan Huang
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center (H.Y., L.-Y.H., D.-Y.Z., E-W.H., S.-J.L., Y.-B.T., Y.-X.S., J.T., F.S., Q.-Q.W., L.-X.X., X.-F.L., J.L., Y.-Y.G., J.-G.Z.), and Department of Forensic Pathology (E.-W.H.), Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Cardiovascular Institute of Guangdong Academy of Medical Sciences, Medical Research Center of Guangdong General Hospital, Guangzhou, China (H.Y.); Guangzhou Forensic Science
| | - De-Yi Zeng
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center (H.Y., L.-Y.H., D.-Y.Z., E-W.H., S.-J.L., Y.-B.T., Y.-X.S., J.T., F.S., Q.-Q.W., L.-X.X., X.-F.L., J.L., Y.-Y.G., J.-G.Z.), and Department of Forensic Pathology (E.-W.H.), Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Cardiovascular Institute of Guangdong Academy of Medical Sciences, Medical Research Center of Guangdong General Hospital, Guangzhou, China (H.Y.); Guangzhou Forensic Science
| | - Er-Wen Huang
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center (H.Y., L.-Y.H., D.-Y.Z., E-W.H., S.-J.L., Y.-B.T., Y.-X.S., J.T., F.S., Q.-Q.W., L.-X.X., X.-F.L., J.L., Y.-Y.G., J.-G.Z.), and Department of Forensic Pathology (E.-W.H.), Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Cardiovascular Institute of Guangdong Academy of Medical Sciences, Medical Research Center of Guangdong General Hospital, Guangzhou, China (H.Y.); Guangzhou Forensic Science
| | - Si-Jia Liang
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center (H.Y., L.-Y.H., D.-Y.Z., E-W.H., S.-J.L., Y.-B.T., Y.-X.S., J.T., F.S., Q.-Q.W., L.-X.X., X.-F.L., J.L., Y.-Y.G., J.-G.Z.), and Department of Forensic Pathology (E.-W.H.), Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Cardiovascular Institute of Guangdong Academy of Medical Sciences, Medical Research Center of Guangdong General Hospital, Guangzhou, China (H.Y.); Guangzhou Forensic Science
| | - Yong-Bo Tang
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center (H.Y., L.-Y.H., D.-Y.Z., E-W.H., S.-J.L., Y.-B.T., Y.-X.S., J.T., F.S., Q.-Q.W., L.-X.X., X.-F.L., J.L., Y.-Y.G., J.-G.Z.), and Department of Forensic Pathology (E.-W.H.), Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Cardiovascular Institute of Guangdong Academy of Medical Sciences, Medical Research Center of Guangdong General Hospital, Guangzhou, China (H.Y.); Guangzhou Forensic Science
| | - Ying-Xue Su
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center (H.Y., L.-Y.H., D.-Y.Z., E-W.H., S.-J.L., Y.-B.T., Y.-X.S., J.T., F.S., Q.-Q.W., L.-X.X., X.-F.L., J.L., Y.-Y.G., J.-G.Z.), and Department of Forensic Pathology (E.-W.H.), Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Cardiovascular Institute of Guangdong Academy of Medical Sciences, Medical Research Center of Guangdong General Hospital, Guangzhou, China (H.Y.); Guangzhou Forensic Science
| | - Jing Tao
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center (H.Y., L.-Y.H., D.-Y.Z., E-W.H., S.-J.L., Y.-B.T., Y.-X.S., J.T., F.S., Q.-Q.W., L.-X.X., X.-F.L., J.L., Y.-Y.G., J.-G.Z.), and Department of Forensic Pathology (E.-W.H.), Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Cardiovascular Institute of Guangdong Academy of Medical Sciences, Medical Research Center of Guangdong General Hospital, Guangzhou, China (H.Y.); Guangzhou Forensic Science
| | - Fei Shang
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center (H.Y., L.-Y.H., D.-Y.Z., E-W.H., S.-J.L., Y.-B.T., Y.-X.S., J.T., F.S., Q.-Q.W., L.-X.X., X.-F.L., J.L., Y.-Y.G., J.-G.Z.), and Department of Forensic Pathology (E.-W.H.), Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Cardiovascular Institute of Guangdong Academy of Medical Sciences, Medical Research Center of Guangdong General Hospital, Guangzhou, China (H.Y.); Guangzhou Forensic Science
| | - Qian-Qian Wu
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center (H.Y., L.-Y.H., D.-Y.Z., E-W.H., S.-J.L., Y.-B.T., Y.-X.S., J.T., F.S., Q.-Q.W., L.-X.X., X.-F.L., J.L., Y.-Y.G., J.-G.Z.), and Department of Forensic Pathology (E.-W.H.), Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Cardiovascular Institute of Guangdong Academy of Medical Sciences, Medical Research Center of Guangdong General Hospital, Guangzhou, China (H.Y.); Guangzhou Forensic Science
| | - Li-Xiong Xiong
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center (H.Y., L.-Y.H., D.-Y.Z., E-W.H., S.-J.L., Y.-B.T., Y.-X.S., J.T., F.S., Q.-Q.W., L.-X.X., X.-F.L., J.L., Y.-Y.G., J.-G.Z.), and Department of Forensic Pathology (E.-W.H.), Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Cardiovascular Institute of Guangdong Academy of Medical Sciences, Medical Research Center of Guangdong General Hospital, Guangzhou, China (H.Y.); Guangzhou Forensic Science
| | - Xiao-Fei Lv
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center (H.Y., L.-Y.H., D.-Y.Z., E-W.H., S.-J.L., Y.-B.T., Y.-X.S., J.T., F.S., Q.-Q.W., L.-X.X., X.-F.L., J.L., Y.-Y.G., J.-G.Z.), and Department of Forensic Pathology (E.-W.H.), Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Cardiovascular Institute of Guangdong Academy of Medical Sciences, Medical Research Center of Guangdong General Hospital, Guangzhou, China (H.Y.); Guangzhou Forensic Science
| | - Jie Liu
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center (H.Y., L.-Y.H., D.-Y.Z., E-W.H., S.-J.L., Y.-B.T., Y.-X.S., J.T., F.S., Q.-Q.W., L.-X.X., X.-F.L., J.L., Y.-Y.G., J.-G.Z.), and Department of Forensic Pathology (E.-W.H.), Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Cardiovascular Institute of Guangdong Academy of Medical Sciences, Medical Research Center of Guangdong General Hospital, Guangzhou, China (H.Y.); Guangzhou Forensic Science
| | - Yong-Yuan Guan
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center (H.Y., L.-Y.H., D.-Y.Z., E-W.H., S.-J.L., Y.-B.T., Y.-X.S., J.T., F.S., Q.-Q.W., L.-X.X., X.-F.L., J.L., Y.-Y.G., J.-G.Z.), and Department of Forensic Pathology (E.-W.H.), Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Cardiovascular Institute of Guangdong Academy of Medical Sciences, Medical Research Center of Guangdong General Hospital, Guangzhou, China (H.Y.); Guangzhou Forensic Science
| | - Jia-Guo Zhou
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center (H.Y., L.-Y.H., D.-Y.Z., E-W.H., S.-J.L., Y.-B.T., Y.-X.S., J.T., F.S., Q.-Q.W., L.-X.X., X.-F.L., J.L., Y.-Y.G., J.-G.Z.), and Department of Forensic Pathology (E.-W.H.), Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Cardiovascular Institute of Guangdong Academy of Medical Sciences, Medical Research Center of Guangdong General Hospital, Guangzhou, China (H.Y.); Guangzhou Forensic Science
| |
Collapse
|
47
|
Abstract
Luminal acidification is of pivotal importance for the physiology of the secretory and endocytic pathways and its diverse trafficking events. Acidification by the proton-pumping V-ATPase requires charge compensation by counterion currents that are commonly attributed to chloride. The molecular identification of intracellular chloride transporters and the improvement of methodologies for measuring intraorganellar pH and chloride have facilitated the investigation of the physiology of vesicular chloride transport. New data question the requirement of chloride for pH regulation of various organelles and furthermore ascribe functions to chloride that are beyond merely electrically shunting the proton pump. This review surveys the currently established and proposed intracellular chloride transporters and gives an overview of membrane-trafficking steps that are affected by the perturbation of chloride transport. Finally, potential mechanisms of membrane-trafficking modulation by chloride are discussed and put into the context of organellar ion homeostasis in general.
Collapse
Affiliation(s)
- Tobias Stauber
- Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin, 13125 Berlin, Germany.
| | | |
Collapse
|
48
|
|
49
|
Burette AC, Weinberg RJ, Sassani P, Abuladze N, Kao L, Kurtz I. The sodium-driven chloride/bicarbonate exchanger in presynaptic terminals. J Comp Neurol 2012; 520:1481-92. [PMID: 22102085 DOI: 10.1002/cne.22806] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The sodium-driven chloride/bicarbonate exchanger (NDCBE), a member of the SLC4 family of bicarbonate transporters, was recently found to modulate excitatory neurotransmission in hippocampus. By using light and electron microscopic immunohistochemistry, we demonstrate here that NDCBE is expressed throughout the adult rat brain, and selectively concentrates in presynaptic terminals, where it is closely associated with synaptic vesicles. NDCBE is in most glutamatergic axon terminals, and is also present in the terminals of parvalbumin-positive γ-aminobutyric acid (GABA)ergic cells. These findings suggest that NDCBE can regulate glutamatergic transmission throughout the brain, and point to a role for NDCBE as a possible regulator of GABAergic neurotransmission.
Collapse
Affiliation(s)
- Alain C Burette
- Department of Cell and Developmental Biology, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
| | | | | | | | | | | |
Collapse
|
50
|
O’Dell CM, Das A, Wallace IV G, Ray SK, Banik NL. Understanding the basic mechanisms underlying seizures in mesial temporal lobe epilepsy and possible therapeutic targets: a review. J Neurosci Res 2012; 90:913-24. [PMID: 22315182 PMCID: PMC11877321 DOI: 10.1002/jnr.22829] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 10/05/2011] [Accepted: 10/09/2011] [Indexed: 12/13/2022]
Abstract
Despite years of research, epilepsy remains a poorly understood disorder. In the past several years, work has been conducted on a variety of projects with the goal of better understanding the pathogenesis and progression of mesial temporal lobe epilepsy (MTLE), in particular, and how to exploit those properties to generate innovative therapies for treatment of refractory epilepsies. This review seeks to give an overview of common morphological and biochemical changes associated with epilepsy and proposed treatments to address those changes. Furthering the understanding of ictogenesis and epileptogenesis remains an important goal for scientists seeking to find more effective treatments for MTLE.
Collapse
Affiliation(s)
- Casey M. O’Dell
- Division of Neurology, Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina
| | - Arabinda Das
- Division of Neurology, Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina
| | - Gerald Wallace IV
- Division of Neurology, Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina
| | - Swapan K. Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Naren L. Banik
- Division of Neurology, Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|