1
|
Hu Y, Tao W. Microenvironmental Variations After Blood-Brain Barrier Breakdown in Traumatic Brain Injury. Front Mol Neurosci 2021; 14:750810. [PMID: 34899180 PMCID: PMC8662751 DOI: 10.3389/fnmol.2021.750810] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is linked to several pathologies. The blood-brain barrier (BBB) breakdown is considered to be one of the initial changes. Further, the microenvironmental alteration following TBI-induced BBB breakdown can be multi-scaled, constant, and dramatic. The microenvironmental variations after disruption of BBB includes several pathological changes, such as cerebral blood flow (CBF) alteration, brain edema, cerebral metabolism imbalances, and accumulation of inflammatory molecules. The modulation of the microenvironment presents attractive targets for TBI recovery, such as reducing toxic substances, inhibiting inflammation, and promoting neurogenesis. Herein, we briefly review the pathological alterations of the microenvironmental changes following BBB breakdown and outline potential interventions for TBI recovery based on microenvironmental modulation.
Collapse
Affiliation(s)
- Yue Hu
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weiwei Tao
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
2
|
King P, Wan J, Guo AA, Guo S, Jiang Y, Liu M. Regulation of gliomagenesis and stemness through acid sensor ASIC1a. Int J Oncol 2021; 59:82. [PMID: 34515325 PMCID: PMC8448544 DOI: 10.3892/ijo.2021.5262] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/28/2021] [Indexed: 01/29/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most prevalent and aggressive type of adult gliomas. Despite intensive therapy including surgery, radiation, and chemotherapy, invariable tumor recurrence occurs, which suggests that glioblastoma stem cells (GSCs) render these tumors persistent. Recently, the induction of GSC differentiation has emerged as an alternative method to treat GBM, and most of the current studies aim to convert GSCs to neurons by a combination of transcriptional factors. As the tumor microenvironment is typically acidic due to increased glycolysis and consequently leads to an increased production of lactic acid in tumor cells, in the present study, the role of acid‑sensing ion channel 1a (ASIC1a), an acid sensor, was explored as a tumor suppressor in gliomagenesis and stemness. The bioinformatics data from The Cancer Genome Atlas revealed that ASIC1 expression levels in GBM tumor tissues were lower than those in normal brain, and glioma patients with high ASIC1 expression had longer survival than those with low ASIC1 expression. Our immunohistochemistry data from tissue microarray revealed that ASIC1a expression was negatively associated with glioma grading. Functional studies revealed that the downregulation of ASIC1a promoted glioma cell proliferation and invasion, while upregulation of ASIC1a inhibited their proliferation and invasion. Furthermore, ASIC1a suppressed growth and proliferation of glioma cells through G1/S arrest and apoptosis induction. Mechanistically, ASIC1a negatively modulated glioma stemness via inhibition of the Notch signaling pathway and GSC markers CD133 and aldehyde dehydrogenase 1. ASIC1a is a tumor suppressor in gliomagenesis and stemness and may serve as a promising prognostic biomarker and target for GBM patients.
Collapse
Affiliation(s)
- Pendelton King
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Jingwei Wan
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Alyssa Aihui Guo
- Department of Biomedical Sciences, School of Medicine Greenville, University of South Carolina, Greenville, SC 29605, USA
| | - Shanchun Guo
- Department of Chemistry, Xavier University, New Orleans, LA 70125, USA
| | - Yugang Jiang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Mingli Liu
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| |
Collapse
|
3
|
Cheng S, Mao X, Lin X, Wehn A, Hu S, Mamrak U, Khalin I, Wostrack M, Ringel F, Plesnila N, Terpolilli NA. Acid-Ion Sensing Channel 1a Deletion Reduces Chronic Brain Damage and Neurological Deficits after Experimental Traumatic Brain Injury. J Neurotrauma 2021; 38:1572-1584. [PMID: 33779289 DOI: 10.1089/neu.2020.7568] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) causes long-lasting neurodegeneration and cognitive impairments; however, the underlying mechanisms of these processes are not fully understood. Acid-sensing ion channels 1a (ASIC1a) are voltage-gated Na+- and Ca2+-channels shown to be involved in neuronal cell death; however, their role for chronic post-traumatic brain damage is largely unknown. To address this issue, we used ASIC1a-deficient mice and investigated their outcome up to 6 months after TBI. ASIC1a-deficient mice and their wild-type (WT) littermates were subjected to controlled cortical impact (CCI) or sham surgery. Brain water content was analyzed 24 h and behavioral outcome up to 6 months after CCI. Lesion volume was assessed longitudinally by magnetic resonance imaging and 6 months after injury by histology. Brain water content was significantly reduced in ASIC1a-/- animals compared to WT controls. Over time, ASIC1a-/- mice showed significantly reduced lesion volume and reduced hippocampal damage. This translated into improved cognitive function and reduced depression-like behavior. Microglial activation was significantly reduced in ASIC1a-/- mice. In conclusion, ASIC1a deficiency resulted in reduced edema formation acutely after TBI and less brain damage, functional impairments, and neuroinflammation up to 6 months after injury. Hence, ASIC1a seems to be involved in chronic neurodegeneration after TBI.
Collapse
Affiliation(s)
- Shiqi Cheng
- Institute for Stroke and Dementia Research, Munich University Hospital, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Xiang Mao
- Institute for Stroke and Dementia Research, Munich University Hospital, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Xiangjiang Lin
- Institute for Stroke and Dementia Research, Munich University Hospital, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Antonia Wehn
- Institute for Stroke and Dementia Research, Munich University Hospital, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Senbin Hu
- Institute for Stroke and Dementia Research, Munich University Hospital, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Uta Mamrak
- Institute for Stroke and Dementia Research, Munich University Hospital, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Igor Khalin
- Institute for Stroke and Dementia Research, Munich University Hospital, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Maria Wostrack
- Department of Neurosurgery, Technical University Munich, Munich, Germany
| | - Florian Ringel
- Department of Neurosurgery, University Medical Center Mainz, Mainz, Germany
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research, Munich University Hospital, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nicole A Terpolilli
- Institute for Stroke and Dementia Research, Munich University Hospital, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Department of Neurosurgery, Munich University Hospital, Munich, Germany
| |
Collapse
|
4
|
Ruan N, Tribble J, Peterson AM, Jiang Q, Wang JQ, Chu XP. Acid-Sensing Ion Channels and Mechanosensation. Int J Mol Sci 2021; 22:ijms22094810. [PMID: 34062742 PMCID: PMC8125064 DOI: 10.3390/ijms22094810] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 12/16/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are mainly proton-gated cation channels that are activated by pH drops and nonproton ligands. They are part of the degenerin/epithelial sodium channel superfamily due to their sodium permeability. Predominantly expressed in the central nervous system, ASICs are involved in synaptic plasticity, learning/memory, and fear conditioning. These channels have also been implicated in multiple disease conditions, including ischemic brain injury, multiple sclerosis, Alzheimer’s disease, and drug addiction. Recent research has illustrated the involvement of ASICs in mechanosensation. Mechanosensation is a form of signal transduction in which mechanical forces are converted into neuronal signals. Specific mechanosensitive functions have been elucidated in functional ASIC1a, ASIC1b, ASIC2a, and ASIC3. The implications of mechanosensation in ASICs indicate their subsequent involvement in functions such as maintaining blood pressure, modulating the gastrointestinal function, and bladder micturition, and contributing to nociception. The underlying mechanism of ASIC mechanosensation is the tether-gate model, which uses a gating-spring mechanism to activate ASIC responses. Further understanding of the mechanism of ASICs will help in treatments for ASIC-related pathologies. Along with the well-known chemosensitive functions of ASICs, emerging evidence has revealed that mechanosensitive functions of ASICs are important for maintaining homeostasis and contribute to various disease conditions.
Collapse
|
5
|
Song XL, Liu DS, Qiang M, Li Q, Liu MG, Li WG, Qi X, Xu NJ, Yang G, Zhu MX, Xu TL. Postsynaptic Targeting and Mobility of Membrane Surface-Localized hASIC1a. Neurosci Bull 2021; 37:145-165. [PMID: 32996060 PMCID: PMC7870742 DOI: 10.1007/s12264-020-00581-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/14/2020] [Indexed: 01/19/2023] Open
Abstract
Acid-sensing ion channels (ASICs), the main H+ receptors in the central nervous system, sense extracellular pH fluctuations and mediate cation influx. ASIC1a, the major subunit responsible for acid-activated current, is widely expressed in brain neurons, where it plays pivotal roles in diverse functions including synaptic transmission and plasticity. However, the underlying molecular mechanisms for these functions remain mysterious. Using extracellular epitope tagging and a novel antibody recognizing the hASIC1a ectodomain, we examined the membrane targeting and dynamic trafficking of hASIC1a in cultured cortical neurons. Surface hASIC1a was distributed throughout somata and dendrites, clustered in spine heads, and co-localized with postsynaptic markers. By extracellular pHluorin tagging and fluorescence recovery after photobleaching, we detected movement of hASIC1a in synaptic spine heads. Single-particle tracking along with use of the anti-hASIC1a ectodomain antibody revealed long-distance migration and local movement of surface hASIC1a puncta on dendrites. Importantly, enhancing synaptic activity with brain-derived neurotrophic factor accelerated the trafficking and lateral mobility of hASIC1a. With this newly-developed toolbox, our data demonstrate the synaptic location and high dynamics of functionally-relevant hASIC1a on the surface of excitatory synapses, supporting its involvement in synaptic functions.
Collapse
Affiliation(s)
- Xing-Lei Song
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Di-Shi Liu
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Min Qiang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Qian Li
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China
| | - Ming-Gang Liu
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China
| | - Wei-Guang Li
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China
| | - Xin Qi
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Nan-Jie Xu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China
| | - Guang Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Michael Xi Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Tian-Le Xu
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China.
| |
Collapse
|
6
|
Ritzel RM, He J, Li Y, Cao T, Khan N, Shim B, Sabirzhanov B, Aubrecht T, Stoica BA, Faden AI, Wu LJ, Wu J. Proton extrusion during oxidative burst in microglia exacerbates pathological acidosis following traumatic brain injury. Glia 2020; 69:746-764. [PMID: 33090575 PMCID: PMC7819364 DOI: 10.1002/glia.23926] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/01/2020] [Accepted: 10/07/2020] [Indexed: 01/02/2023]
Abstract
Acidosis is among the least studied secondary injury mechanisms associated with neurotrauma. Acute decreases in brain pH correlate with poor long‐term outcome in patients with traumatic brain injury (TBI), however, the temporal dynamics and underlying mechanisms are unclear. As key drivers of neuroinflammation, we hypothesized that microglia directly regulate acidosis after TBI, and thereby, worsen neurological outcomes. Using a controlled cortical impact model in adult male mice we demonstrate that intracellular pH in microglia and extracellular pH surrounding the lesion site are significantly reduced for weeks after injury. Microglia proliferation and production of reactive oxygen species (ROS) were also increased during the first week, mirroring the increase in extracellular ROS levels seen around the lesion site. Microglia depletion by a colony stimulating factor 1 receptor (CSF1R) inhibitor, PLX5622, markedly decreased extracellular acidosis, ROS production, and inflammation in the brain after injury. Mechanistically, we identified that the voltage‐gated proton channel Hv1 promotes oxidative burst activity and acid extrusion in microglia. Compared to wildtype controls, microglia lacking Hv1 showed reduced ability to generate ROS and extrude protons. Importantly, Hv1‐deficient mice exhibited reduced pathological acidosis and inflammation after TBI, leading to long‐term neuroprotection and functional recovery. Our data therefore establish the microglial Hv1 proton channel as an important link that integrates inflammation and acidosis within the injury microenvironment during head injury.
Collapse
Affiliation(s)
- Rodney M Ritzel
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Junyun He
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yun Li
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Tuoxin Cao
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Niaz Khan
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Bosung Shim
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Boris Sabirzhanov
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Taryn Aubrecht
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Bogdan A Stoica
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Alan I Faden
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland, USA.,University of Maryland Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Junfang Wu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland, USA.,University of Maryland Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Wang T, Zhou G, He M, Xu Y, Rusyniak WG, Xu Y, Ji Y, Simon RP, Xiong ZG, Zha XM. GPR68 Is a Neuroprotective Proton Receptor in Brain Ischemia. Stroke 2020; 51:3690-3700. [PMID: 33059544 PMCID: PMC7678672 DOI: 10.1161/strokeaha.120.031479] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Supplemental Digital Content is available in the text. Brain acidosis is prevalent in stroke and other neurological diseases. Acidosis can have paradoxical injurious and protective effects. The purpose of this study is to determine whether a proton receptor exists in neurons to counteract acidosis-induced injury.
Collapse
Affiliation(s)
- Tao Wang
- Department of Physiology and Cell Biology (T.W., G.Z., M.H., Yuanyuan Xu, X.-m.Z.), University of South Alabama College of Medicine, Mobile
| | - Guokun Zhou
- Department of Physiology and Cell Biology (T.W., G.Z., M.H., Yuanyuan Xu, X.-m.Z.), University of South Alabama College of Medicine, Mobile.,Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, China (G.Z., Y.J.)
| | - Mindi He
- Department of Physiology and Cell Biology (T.W., G.Z., M.H., Yuanyuan Xu, X.-m.Z.), University of South Alabama College of Medicine, Mobile
| | - Yuanyuan Xu
- Department of Physiology and Cell Biology (T.W., G.Z., M.H., Yuanyuan Xu, X.-m.Z.), University of South Alabama College of Medicine, Mobile
| | - W G Rusyniak
- Department of Neurosurgery (W.G.R.), University of South Alabama College of Medicine, Mobile
| | - Yan Xu
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis (Yan Xu)
| | - Yonghua Ji
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, China (G.Z., Y.J.)
| | - Roger P Simon
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA (R.P.S., Z.-G.X.)
| | - Zhi-Gang Xiong
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA (R.P.S., Z.-G.X.)
| | - Xiang-Ming Zha
- Department of Physiology and Cell Biology (T.W., G.Z., M.H., Yuanyuan Xu, X.-m.Z.), University of South Alabama College of Medicine, Mobile
| |
Collapse
|
8
|
Abstract
Cerebral edema is a pathological hallmark of various central nervous system (CNS) insults, including traumatic brain injury (TBI) and excitotoxic injury such as stroke. Due to the rigidity of the skull, edema-induced increase of intracranial fluid significantly complicates severe CNS injuries by raising intracranial pressure and compromising perfusion. Mortality due to cerebral edema is high. With mortality rates up to 80% in severe cases of stroke, it is the leading cause of death within the first week. Similarly, cerebral edema is devastating for patients of TBI, accounting for up to 50% mortality. Currently, the available treatments for cerebral edema include hypothermia, osmotherapy, and surgery. However, these treatments only address the symptoms and often elicit adverse side effects, potentially in part due to non-specificity. There is an urgent need to identify effective pharmacological treatments for cerebral edema. Currently, ion channels represent the third-largest target class for drug development, but their roles in cerebral edema remain ill-defined. The present review aims to provide an overview of the proposed roles of ion channels and transporters (including aquaporins, SUR1-TRPM4, chloride channels, glucose transporters, and proton-sensitive channels) in mediating cerebral edema in acute ischemic stroke and TBI. We also focus on the pharmacological inhibitors for each target and potential therapeutic strategies that may be further pursued for the treatment of cerebral edema.
Collapse
|
9
|
Histidine Residues Are Responsible for Bidirectional Effects of Zinc on Acid-Sensing Ion Channel 1a/3 Heteromeric Channels. Biomolecules 2020; 10:biom10091264. [PMID: 32887365 PMCID: PMC7565092 DOI: 10.3390/biom10091264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 12/16/2022] Open
Abstract
Acid-sensing ion channel (ASIC) subunits 1a and 3 are highly expressed in central and peripheral sensory neurons, respectively. Endogenous biomolecule zinc plays a critical role in physiological and pathophysiological conditions. Here, we found that currents recorded from heterologously expressed ASIC1a/3 channels using the whole-cell patch-clamp technique were regulated by zinc with dual effects. Co-application of zinc dose-dependently potentiated both peak amplitude and the sustained component of heteromeric ASIC1a/3 currents; pretreatment with zinc between 3 to 100 µM exerted the same potentiation as co-application. However, pretreatment with zinc induced a significant inhibition of heteromeric ASIC1a/3 channels when zinc concentrations were over 250 µM. The potentiation of heteromeric ASIC1a/3 channels by zinc was pH dependent, as zinc shifted the pH dependence of ASIC1a/3 currents from a pH50 of 6.54 to 6.77; whereas the inhibition of ASIC1a/3 currents by zinc was also pH dependent. Furthermore, we systematically mutated histidine residues in the extracellular domain of ASIC1a or ASIC3 and found that histidine residues 72 and 73 in both ASIC1a and ASIC3, and histidine residue 83 in the ASIC3 were responsible for bidirectional effects on heteromeric ASIC1a/3 channels by zinc. These findings suggest that histidine residues in the extracellular domain of heteromeric ASIC1a/3 channels are critical for zinc-mediated effects.
Collapse
|
10
|
Yoder N, Gouaux E. The His-Gly motif of acid-sensing ion channels resides in a reentrant 'loop' implicated in gating and ion selectivity. eLife 2020; 9:e56527. [PMID: 32496192 PMCID: PMC7308080 DOI: 10.7554/elife.56527] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/03/2020] [Indexed: 12/18/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are proton-gated members of the epithelial sodium channel/degenerin (ENaC/DEG) superfamily of ion channels and are expressed throughout the central and peripheral nervous systems. The homotrimeric splice variant ASIC1a has been implicated in nociception, fear memory, mood disorders and ischemia. Here, we extract full-length chicken ASIC1 (cASIC1) from cell membranes using styrene maleic acid (SMA) copolymer, elucidating structures of ASIC1 channels in both high pH resting and low pH desensitized conformations by single-particle cryo-electron microscopy (cryo-EM). The structures of resting and desensitized channels reveal a reentrant loop at the amino terminus of ASIC1 that includes the highly conserved 'His-Gly' (HG) motif. The reentrant loop lines the lower ion permeation pathway and buttresses the 'Gly-Ala-Ser' (GAS) constriction, thus providing a structural explanation for the role of the His-Gly dipeptide in the structure and function of ASICs.
Collapse
Affiliation(s)
- Nate Yoder
- Vollum Institute, Oregon Health & Science UniversityPortlandUnited States
| | - Eric Gouaux
- Vollum Institute, Oregon Health & Science UniversityPortlandUnited States
- Howard Hughes Medical Institute, Oregon Health & Science UniversityPortlandUnited States
| |
Collapse
|
11
|
Osmakov DI, Koshelev SG, Lyukmanova EN, Shulepko MA, Andreev YA, Illes P, Kozlov SA. Multiple Modulation of Acid-Sensing Ion Channel 1a by the Alkaloid Daurisoline. Biomolecules 2019; 9:biom9080336. [PMID: 31382492 PMCID: PMC6722837 DOI: 10.3390/biom9080336] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 01/10/2023] Open
Abstract
Acid-sensing ion channels (ASICs) are proton-gated sodium-selective channels that are expressed in the peripheral and central nervous systems. ASIC1a is one of the most intensively studied isoforms due to its importance and wide representation in organisms, but it is still largely unexplored as a target for therapy. In this study, we demonstrated response of the ASIC1a to acidification in the presence of the daurisoline (DAU) ligand. DAU alone did not activate the channel, but in combination with protons, it produced the second peak component of the ASIC1a current. This second peak differs from the sustained component (which is induced by RF-amide peptides), as the second (DAU-induced) peak is completely desensitized, with the same kinetics as the main peak. The co-application of DAU and mambalgin-2 indicated that their binding sites do not overlap. Additionally, we found an asymmetry in the pH activation curve of the channel, which was well-described by a mathematical model based on the multiplied probabilities of protons binding with a pool of high-cooperative sites and a single proton binding with a non-cooperative site. In this model, DAU targeted the pool of high-cooperative sites and, when applied with protons, acted as an inhibitor of ASIC1a activation. Moreover, DAU's occupation of the same binding site most probably reverses the channel from steady-state desensitization in the pH 6.9-7.3 range. DAU features disclose new opportunities in studies of ASIC structure and function.
Collapse
Affiliation(s)
- Dmitry I Osmakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia.
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya str. 8, bld. 2, 119991 Moscow, Russia.
| | - Sergey G Koshelev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Ekaterina N Lyukmanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Mikhail A Shulepko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Yaroslav A Andreev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya str. 8, bld. 2, 119991 Moscow, Russia
| | - Peter Illes
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, University of Leipzig, 04107 Leipzig, Germany
| | - Sergey A Kozlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| |
Collapse
|
12
|
Zhou RP, Leng TD, Yang T, Chen FH, Xiong ZG. Acute Ethanol Exposure Promotes Autophagy-Lysosome Pathway-Dependent ASIC1a Protein Degradation and Protects Against Acidosis-Induced Neurotoxicity. Mol Neurobiol 2018; 56:3326-3340. [PMID: 30120732 DOI: 10.1007/s12035-018-1289-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/31/2018] [Indexed: 12/12/2022]
Abstract
Tissue acidosis is a common feature of brain ischemia which causes neuronal injury. Activation of acid-sensing ion channel 1a (ASIC1a) plays an important role in acidosis-mediated neurotoxicity. Acute ethanol administration has been shown to provide neuroprotective effects during ischemic stroke, but the precise mechanisms have yet to be determined. In this study, we investigated the effect of ethanol on the activity/expression of ASIC1a channels and acidosis-induced neurotoxicity. We showed that acute treatment of neuronal cells with ethanol for more than 3 h could reduce ASIC1a protein expression, ASIC currents, and acid-induced [Ca2+]i elevation. We further demonstrated that ethanol-induced reduction of ASIC1a expression is mediated by autophagy-lysosome pathway (ALP)-dependent protein degradation. Finally, we showed that ethanol protected neuronal cells against acidosis-induced cytotoxicity, which effect was mimicked by autophagy activator rapamycin and abolished by autophagy inhibitor CQ. Together, these results indicate that moderate acute ethanol exposure can promote autophagy-lysosome pathway-dependent ASIC1a protein degradation and protect against acidosis-induced neurotoxicity.
Collapse
Affiliation(s)
- Ren-Peng Zhou
- Department of Pharmacology, The Second Hospital of Anhui Medical University, Hefei, 230601, China
- Neurobiology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, 30310, USA
| | - Tian-Dong Leng
- Neurobiology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, 30310, USA
| | - Tao Yang
- Neurobiology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, 30310, USA
| | - Fei-Hu Chen
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, 230032, China.
| | - Zhi-Gang Xiong
- Neurobiology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, 30310, USA.
| |
Collapse
|
13
|
Pathophysiology and treatment of cerebral edema in traumatic brain injury. Neuropharmacology 2018; 145:230-246. [PMID: 30086289 DOI: 10.1016/j.neuropharm.2018.08.004] [Citation(s) in RCA: 287] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/24/2018] [Accepted: 08/03/2018] [Indexed: 12/30/2022]
Abstract
Cerebral edema (CE) and resultant intracranial hypertension are associated with unfavorable prognosis in traumatic brain injury (TBI). CE is a leading cause of in-hospital mortality, occurring in >60% of patients with mass lesions, and ∼15% of those with normal initial computed tomography scans. After treatment of mass lesions in severe TBI, an important focus of acute neurocritical care is evaluating and managing the secondary injury process of CE and resultant intracranial hypertension. This review focuses on a contemporary understanding of various pathophysiologic pathways contributing to CE, with a subsequent description of potential targeted therapies. There is a discussion of identified cellular/cytotoxic contributors to CE, as well as mechanisms that influence blood-brain-barrier (BBB) disruption/vasogenic edema, with the caveat that this distinction may be somewhat artificial since molecular processes contributing to these pathways are interrelated. While an exhaustive discussion of all pathways with putative contributions to CE is beyond the scope of this review, the roles of some key contributors are highlighted, and references are provided for further details. Potential future molecular targets for treating CE are presented based on pathophysiologic mechanisms. We thus aim to provide a translational synopsis of present and future strategies targeting CE after TBI in the context of a paradigm shift towards precision medicine. This article is part of the Special Issue entitled "Novel Treatments for Traumatic Brain Injury".
Collapse
|
14
|
Cheng YR, Jiang BY, Chen CC. Acid-sensing ion channels: dual function proteins for chemo-sensing and mechano-sensing. J Biomed Sci 2018; 25:46. [PMID: 29793480 PMCID: PMC5966886 DOI: 10.1186/s12929-018-0448-y] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/15/2018] [Indexed: 12/22/2022] Open
Abstract
Background Acid-sensing ion channels (ASICs) are a group of amiloride-sensitive ligand-gated ion channels belonging to the family of degenerin/epithelial sodium channels. ASICs are predominantly expressed in both the peripheral and central nervous system and have been characterized as potent proton sensors to detect extracellular acidification in the periphery and brain. Main body Here we review the recent studies focusing on the physiological roles of ASICs in the nervous system. As the major acid-sensing membrane proteins in the nervous system, ASICs detect tissue acidosis occurring at tissue injury, inflammation, ischemia, stroke, and tumors as well as fatiguing muscle to activate pain-sensing nerves in the periphery and transmit pain signals to the brain. Arachidonic acid and lysophosphocholine have been identified as endogenous non-proton ligands activating ASICs in a neutral pH environment. On the other hand, ASICs are found involved in the tether model mechanotransduction, in which the extracellular matrix and cytoplasmic cytoskeletons act like a gating-spring to tether the mechanically activated ion channels and thus transmit the stimulus force to the channels. Accordingly, accumulating evidence has shown ASICs play important roles in mechanotransduction of proprioceptors, mechanoreceptors and nociceptors to monitor the homoeostatic status of muscle contraction, blood volume, and blood pressure as well as pain stimuli. Conclusion Together, ASICs are dual-function proteins for both chemosensation and mechanosensation involved in monitoring physiological homoeostasis and pathological signals.
Collapse
Affiliation(s)
- Yuan-Ren Cheng
- Department of Life Science, National Taiwan University, Taipei, 106, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, 128, Academia Rd. Sec. 2, Taipei, 115, Taiwan
| | - Bo-Yang Jiang
- Department of Life Science, National Taiwan University, Taipei, 106, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, 128, Academia Rd. Sec. 2, Taipei, 115, Taiwan
| | - Chih-Cheng Chen
- Department of Life Science, National Taiwan University, Taipei, 106, Taiwan. .,Institute of Biomedical Sciences, Academia Sinica, 128, Academia Rd. Sec. 2, Taipei, 115, Taiwan. .,Taiwan Mouse Clinic - National Comprehensive Mouse Phenotyping and Drug Testing Center, Academia Sinica, Taipei, 115, Taiwan.
| |
Collapse
|
15
|
Abstract
Acid-sensing ion channels (ASICs) are a family of ion channels, consisting of four members; ASIC1 to 4. These channels are sensitive to changes in pH and are expressed throughout the central and peripheral nervous systems-including brain, spinal cord, and sensory ganglia. They have been implicated in a number of neurological conditions such as stroke and cerebral ischemia, traumatic brain injury, and epilepsy, and more recently in migraine. Their expression within areas of interest in the brain in migraine, such as the hypothalamus and PAG, their demonstrated involvement in preclinical models of meningeal afferent signaling, and their role in cortical spreading depression (the electrophysiological correlate of migraine aura), has enhanced research interest into these channels as potential therapeutic targets in migraine. Migraine is a disorder with a paucity of both acute and preventive therapies available, in which at best 50% of patients respond to available medications, and these medications often have intolerable side effects. There is therefore a great need for therapeutic development for this disabling condition. This review will summarize the understanding of the structure and CNS expression of ASICs, the mechanisms for their potential role in nociception, recent work in migraine, and areas for future research and drug development.
Collapse
Affiliation(s)
- Nazia Karsan
- Headache Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, Denmark Hill, London, SE5 9PJ, UK
| | - Eric B Gonzales
- TCU and UNTHSC School of Medicine (applicant for LCME accreditation), Department of Medical Education, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA
| | - Gregory Dussor
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, BSB-14, Richardson, TX, 75080, USA.
| |
Collapse
|
16
|
Xu Y, Jiang YQ, Li C, He M, Rusyniak WG, Annamdevula N, Ochoa J, Leavesley SJ, Xu J, Rich TC, Lin MT, Zha XM. Human ASIC1a mediates stronger acid-induced responses as compared with mouse ASIC1a. FASEB J 2018; 32:3832-3843. [PMID: 29447005 DOI: 10.1096/fj.201701367r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Acid-sensing ion channels (ASICs) are the major proton receptor in the brain and a key mediator of acidosis-induced neuronal injuries in disease. Most of published data on ASIC function came from studies performed in mice, and relatively little is known about potential differences between human and mouse ASICs (hASIC and mASIC, respectively). This information is critical for us to better interpret the functional importance of ASICs in human disease. Here, we examined the expression of ASICs in acutely resected human cortical tissue. Compared with mouse cortex, human cortical tissue showed a similar ratio of ASIC1a:ASIC2a expression, had reduced ASIC2b level, and exhibited a higher membrane:total ratio of ASIC1a. We further investigated the mechanism for higher surface trafficking of hASIC1a in heterologous cells. A single amino acid at position 285 was critical for increased N-glycosylation and surface expression of hASIC1a. Consistent with the changes in trafficking and current, cells expressing hASIC1a or mASIC1a S285P mutant had a higher acid-activated calcium increase and exhibited worsened acidotoxicity. These data suggest that ASICs are likely to have a larger impact on acidosis-induced neuronal injuries in humans than mice, and this effect is, at least in part, a result of more efficient trafficking of hASIC1a.-Xu, Y., Jiang, Y.-Q., Li, C., He, M., Rusyniak, W. G., Annamdevula, N., Ochoa, J., Leavesley, S. J., Xu, J., Rich, T. C., Lin, M. T., Zha, X.-M. Human ASIC1a mediates stronger acid-induced responses as compared with mouse ASIC1a.
Collapse
Affiliation(s)
- Yuanyuan Xu
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.,Department of Neuropharmacology and Drug Discovery, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Yu-Qing Jiang
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.,The Third Hospital of Hebei Medical University, ShiJiaZhuang, China
| | - Ce Li
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.,Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Mindi He
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - W George Rusyniak
- Department of Neurosurgery, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Naga Annamdevula
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Juan Ochoa
- Department of Neurology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Silas J Leavesley
- Chemical and Biomolecular Engineering, College of Engineering, University of South Alabama, Mobile, Alabama, USA
| | - Jiangping Xu
- Department of Neuropharmacology and Drug Discovery, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Thomas C Rich
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Mike T Lin
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Xiang-Ming Zha
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| |
Collapse
|
17
|
Hu W, Tung YC, Zhang Y, Liu F, Iqbal K. Involvement of Activation of Asparaginyl Endopeptidase in Tau Hyperphosphorylation in Repetitive Mild Traumatic Brain Injury. J Alzheimers Dis 2018; 64:709-722. [PMID: 29889065 PMCID: PMC6087458 DOI: 10.3233/jad-180177] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2018] [Indexed: 12/13/2022]
Abstract
Traumatic brain injury (TBI) is an established risk factor for the development of neurodegeneration and dementia late in life. Repetitive mild TBI (r-mTBI) is directly associated with chronic traumatic encephalopathy (CTE), a progressive neurodegenerative disorder characterized by focal perivascular to widespread Alzheimer-type neurofibrillary pathology of hyperphosphorylated tau. Studies in animal models have shown hyperphosphorylation of tau after TBI. However, the molecular mechanisms by which TBI leads to tau pathology are not understood. In this study, we employed western blots and immunohistochemistry to test, in triple-transgenic mouse model of Alzheimer's disease (3xTg-AD), the effect of r-mTBI on tau hyperphosphorylation and activation of asparaginyl endopeptidase (AEP), a cysteine proteinase which is known to be involved in tau hyperphosphorylation. We found that the level of active AEP was increased and correlated with the level of tau hyperphosphorylation following r-mTBI, and that fimbria showed increased immunoreactivity to phospho-tau. In addition, inhibitor 2 of protein phosphatase 2A (I2PP2A) was translocated from neuronal nucleus to the cytoplasm and colocalized with hyperphosphorylated tau. These data suggest the involvement of AEP-I2PP2A-PP2A-ptau pathway in tau pathology in TBI.
Collapse
Affiliation(s)
- Wen Hu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
- Key Laboratory for Neuroregeneration of Ministry of Education and Co-innovation Center for Neuroregeneration of Jiangsu Province, Nantong University, Nantong, Jiangsu, P.R. China
| | - Yunn Chyn Tung
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Yanchong Zhang
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
- Key Laboratory for Neuroregeneration of Ministry of Education and Co-innovation Center for Neuroregeneration of Jiangsu Province, Nantong University, Nantong, Jiangsu, P.R. China
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
- Key Laboratory for Neuroregeneration of Ministry of Education and Co-innovation Center for Neuroregeneration of Jiangsu Province, Nantong University, Nantong, Jiangsu, P.R. China
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| |
Collapse
|
18
|
Jiang YQ, Zha XM. miR-149 reduces while let-7 elevates ASIC1a expression in vitro. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2017; 9:147-152. [PMID: 29209451 PMCID: PMC5698691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 10/26/2017] [Indexed: 06/07/2023]
Abstract
Acid-sensing ion channel 1a (ASIC1a) is the key subunit that determines acid-activated currents in neurons. ASIC1a is important for neural plasticity, learning, and for multiple neurological diseases, including stroke, multiple sclerosis, and traumatic injuries. These findings underline the importance for better defining mechanisms that regulate ASIC1a expression. During the past decade, microRNA has emerged as one important group of regulatory molecules in controlling protein expression. However, little is known about whether microRNA regulates ASIC1a. Here, we assessed several microRNAs that have predicted targeting sequences in the 3' untranslated region (UTR) of mouse ASIC1a. Our results indicated that miR-144 and -149 reduced ASIC1a expression while Let-7 increased ASIC1a protein levels. miR-30c, -98, -125, -182* had no significant effect. Since a reduction in ASIC1a expression may have translational potentials in treating neuronal injury, we further asked whether the effect of miR-144 and miR-149, both reduced ASIC1a expression, was through specific targeting of the predicted sites on ASIC1a. We mutated the targeting sequence of miR-144 and miR-149 in ASIC1a UTR. The effect of miR-149 was abolished in the corresponding mutation. In contrast, miR-144 still reduced ASIC1a level when its predicted target sequence was mutated. This result indicates that miR-149 targets the 3'UTR of ASIC1a and reduces its expression.
Collapse
Affiliation(s)
- Yu-Qing Jiang
- Department of Physiology and Cell Biology, University of South Alabama College of MedicineMobile, AL 36688, USA
- The Third Hospital of Hebei Medical University139 Ziqiang Road, Shijiazhuang 050051, Hebei Province, China
| | - Xiang-Ming Zha
- Department of Physiology and Cell Biology, University of South Alabama College of MedicineMobile, AL 36688, USA
| |
Collapse
|
19
|
Ievglevskyi O, Isaev D, Netsyk O, Romanov A, Fedoriuk M, Maximyuk O, Isaeva E, Akaike N, Krishtal O. Acid-sensing ion channels regulate spontaneous inhibitory activity in the hippocampus: possible implications for epilepsy. Philos Trans R Soc Lond B Biol Sci 2017; 371:rstb.2015.0431. [PMID: 27377725 DOI: 10.1098/rstb.2015.0431] [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] [Accepted: 04/18/2016] [Indexed: 12/12/2022] Open
Abstract
Acid-sensing ion channels (ASICs) play an important role in numerous functions in the central and peripheral nervous systems ranging from memory and emotions to pain. The data correspond to a recent notion that each neuron and many glial cells of the mammalian brain express at least one member of the ASIC family. However, the mechanisms underlying the involvement of ASICs in neuronal activity are poorly understood. However, there are two exceptions, namely, the straightforward role of ASICs in proton-based synaptic transmission in certain brain areas and the role of the Ca(2+)-permeable ASIC1a subtype in ischaemic cell death. Using a novel orthosteric ASIC antagonist, we have found that ASICs specifically control the frequency of spontaneous inhibitory synaptic activity in the hippocampus. Inhibition of ASICs leads to a strong increase in the frequency of spontaneous inhibitory postsynaptic currents. This effect is presynaptic because it is fully reproducible in single synaptic boutons attached to isolated hippocampal neurons. In concert with this observation, inhibition of the ASIC current diminishes epileptic discharges in a low Mg(2+) model of epilepsy in hippocampal slices and significantly reduces kainate-induced discharges in the hippocampus in vivo Our results reveal a significant novel role for ASICs.This article is part of the themed issue 'Evolution brings Ca(2+) and ATP together to control life and death'.
Collapse
Affiliation(s)
- O Ievglevskyi
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, 01024 Kyiv, Ukraine
| | - D Isaev
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, 01024 Kyiv, Ukraine
| | - O Netsyk
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, 01024 Kyiv, Ukraine
| | - A Romanov
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, 01024 Kyiv, Ukraine
| | - M Fedoriuk
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, 01024 Kyiv, Ukraine
| | - O Maximyuk
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, 01024 Kyiv, Ukraine
| | - E Isaeva
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, 01024 Kyiv, Ukraine
| | - N Akaike
- Research Division for Clinical Pharmacology, Medical Corporation, JyuryoGroup, Kumamoto Kinoh Hospital, 6-8-1 Yamamuro, Kitaku, Kumamoto 860-8518, Japan
| | - O Krishtal
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, 01024 Kyiv, Ukraine
| |
Collapse
|
20
|
Chiacchiaretta M, Latifi S, Bramini M, Fadda M, Fassio A, Benfenati F, Cesca F. Neuronal hyperactivity causes Na +/H + exchanger-induced extracellular acidification at active synapses. J Cell Sci 2017; 130:1435-1449. [PMID: 28254883 DOI: 10.1242/jcs.198564] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/28/2017] [Indexed: 12/12/2022] Open
Abstract
Extracellular pH impacts on neuronal activity, which is in turn an important determinant of extracellular H+ concentration. The aim of this study was to describe the spatio-temporal dynamics of extracellular pH at synaptic sites during neuronal hyperexcitability. To address this issue we created ex.E2GFP, a membrane-targeted extracellular ratiometric pH indicator that is exquisitely sensitive to acidic shifts. By monitoring ex.E2GFP fluorescence in real time in primary cortical neurons, we were able to quantify pH fluctuations during network hyperexcitability induced by convulsant drugs or high-frequency electrical stimulation. Sustained hyperactivity caused a pH decrease that was reversible upon silencing of neuronal activity and located at active synapses. This acidic shift was not attributable to the outflow of synaptic vesicle H+ into the cleft nor to the activity of membrane-exposed H+ V-ATPase, but rather to the activity of the Na+/H+-exchanger. Our data demonstrate that extracellular synaptic pH shifts take place during epileptic-like activity of neural cultures, emphasizing the strict links existing between synaptic activity and synaptic pH. This evidence may contribute to the understanding of the physio-pathological mechanisms associated with hyperexcitability in the epileptic brain.
Collapse
Affiliation(s)
- Martina Chiacchiaretta
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova 16132, Italy.,Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova 16132, Italy
| | - Shahrzad Latifi
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova 16132, Italy
| | - Mattia Bramini
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova 16132, Italy
| | - Manuela Fadda
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova 16132, Italy
| | - Anna Fassio
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova 16132, Italy.,Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova 16132, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova 16132, Italy.,Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova 16132, Italy
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova 16132, Italy
| |
Collapse
|
21
|
Jiang N, Wu J, Leng T, Yang T, Zhou Y, Jiang Q, Wang B, Hu Y, Ji YH, Simon RP, Chu XP, Xiong ZG, Zha XM. Region specific contribution of ASIC2 to acidosis-and ischemia-induced neuronal injury. J Cereb Blood Flow Metab 2017; 37:528-540. [PMID: 26861816 PMCID: PMC5381448 DOI: 10.1177/0271678x16630558] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Acidosis in the brain plays a critical role in neuronal injury in neurological diseases, including brain ischemia. One key mediator of acidosis-induced neuronal injury is the acid-sensing ion channels (ASICs). Current literature has focused on ASIC1a when studying acid signaling. The importance of ASIC2, which is also widely expressed in the brain, has not been appreciated. We found here a region-specific effect of ASIC2 on acid-mediated responses. Deleting ASIC2 reduced acid-activated current in cortical and striatal neurons, but had no significant effect in cerebellar granule neurons. In addition, we demonstrated that ASIC2 was important for ASIC1a expression, and that ASIC2a but not 2b facilitated ASIC1a surface trafficking in the brain. Further, we showed that ASIC2 deletion attenuated acidosis/ischemia-induced neuronal injury in organotypic hippocampal slices but had no effect in organotypic cerebellar slices. Consistent with an injurious role of ASIC2, we showed that ASIC2 deletion significantly protected the mouse brain from ischemic damage in vivo. These data suggest a critical region-specific contribution of ASIC2 to neuronal injury and reveal an important functional difference between ASIC2a and 2b in the brain.
Collapse
Affiliation(s)
- Nan Jiang
- 1 Department of Physiology and Cell Biology, University of South Alabama, Mobile, USA.,2 School of Life Sciences, Shanghai University, Shanghai, China
| | - Junjun Wu
- 1 Department of Physiology and Cell Biology, University of South Alabama, Mobile, USA.,3 China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Tiandong Leng
- 4 Department of Neurobiology, Morehouse School of Medicine, Atlanta, USA
| | - Tao Yang
- 4 Department of Neurobiology, Morehouse School of Medicine, Atlanta, USA
| | - Yufan Zhou
- 1 Department of Physiology and Cell Biology, University of South Alabama, Mobile, USA
| | - Qian Jiang
- 5 Department of Basic Medical Science, University of Missouri-Kansas City, Kansas City, USA
| | - Bin Wang
- 6 Department of Mathematics and Statistics, University of South Alabama, Mobile, USA
| | - Youjia Hu
- 3 China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Yong-Hua Ji
- 2 School of Life Sciences, Shanghai University, Shanghai, China
| | - Roger P Simon
- 4 Department of Neurobiology, Morehouse School of Medicine, Atlanta, USA
| | - Xiang-Ping Chu
- 5 Department of Basic Medical Science, University of Missouri-Kansas City, Kansas City, USA
| | - Zhi-Gang Xiong
- 4 Department of Neurobiology, Morehouse School of Medicine, Atlanta, USA
| | - Xiang-Ming Zha
- 1 Department of Physiology and Cell Biology, University of South Alabama, Mobile, USA
| |
Collapse
|
22
|
Koehn LM, Noor NM, Dong Q, Er SY, Rash LD, King GF, Dziegielewska KM, Saunders NR, Habgood MD. Selective inhibition of ASIC1a confers functional and morphological neuroprotection following traumatic spinal cord injury. F1000Res 2016; 5:1822. [PMID: 28105306 PMCID: PMC5200949 DOI: 10.12688/f1000research.9094.2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2016] [Indexed: 12/31/2022] Open
Abstract
Tissue loss after spinal trauma is biphasic, with initial mechanical/haemorrhagic damage at the time of impact being followed by gradual secondary expansion into adjacent, previously unaffected tissue. Limiting the extent of this secondary expansion of tissue damage has the potential to preserve greater residual spinal cord function in patients. The acute tissue hypoxia resulting from spinal cord injury (SCI) activates acid-sensing ion channel 1a (ASIC1a). We surmised that antagonism of this channel should provide neuroprotection and functional preservation after SCI. We show that systemic administration of the spider-venom peptide PcTx1, a selective inhibitor of ASIC1a, improves locomotor function in adult Sprague Dawley rats after thoracic SCI. The degree of functional improvement correlated with the degree of tissue preservation in descending white matter tracts involved in hind limb locomotor function. Transcriptomic analysis suggests that PcTx1-induced preservation of spinal cord tissue does not result from a reduction in apoptosis, with no evidence of down-regulation of key genes involved in either the intrinsic or extrinsic apoptotic pathways. We also demonstrate that trauma-induced disruption of blood-spinal cord barrier function persists for at least 4 days post-injury for compounds up to 10 kDa in size, whereas barrier function is restored for larger molecules within a few hours. This temporary loss of barrier function provides a “
treatment window” through which systemically administered drugs have unrestricted access to spinal tissue in and around the sites of trauma. Taken together, our data provide evidence to support the use of ASIC1a inhibitors as a therapeutic treatment for SCI. This study also emphasizes the importance of objectively grading the functional severity of initial injuries (even when using standardized impacts) and we describe a simple scoring system based on hind limb function that could be adopted in future studies.
Collapse
Affiliation(s)
- Liam M Koehn
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Natassya M Noor
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Qing Dong
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Sing-Yan Er
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Australia
| | - Lachlan D Rash
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Australia; School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - Glenn F King
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Australia
| | | | - Norman R Saunders
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Mark D Habgood
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| |
Collapse
|
23
|
Koehn LM, Noor NM, Dong Q, Er SY, Rash LD, King GF, Dziegielewska KM, Saunders NR, Habgood MD. Selective inhibition of ASIC1a confers functional and morphological neuroprotection following traumatic spinal cord injury. F1000Res 2016; 5:1822. [PMID: 28105306 PMCID: PMC5200949 DOI: 10.12688/f1000research.9094.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2016] [Indexed: 11/10/2023] Open
Abstract
Tissue loss after spinal trauma is biphasic, with initial mechanical/haemorrhagic damage at the time of impact being followed by gradual secondary expansion into adjacent, previously unaffected tissue. Limiting the extent of this secondary expansion of tissue damage has the potential to preserve greater residual spinal cord function in patients. The acute tissue hypoxia resulting from spinal cord injury (SCI) activates acid-sensing ion channel 1a (ASIC1a). We surmised that antagonism of this channel should provide neuroprotection and functional preservation after SCI. We show that systemic administration of the spider-venom peptide PcTx1, a selective inhibitor of ASIC1a, improves locomotor function in adult Sprague Dawley rats after thoracic SCI. The degree of functional improvement correlated with the degree of tissue preservation in descending white matter tracts involved in hind limb locomotor function. Transcriptomic analysis suggests that PcTx1-induced preservation of spinal cord tissue does not result from a reduction in apoptosis, with no evidence of down-regulation of key genes involved in either the intrinsic or extrinsic apoptotic pathways. We also demonstrate that trauma-induced disruption of blood-spinal cord barrier function persists for at least 4 days post-injury for compounds up to 10 kDa in size, whereas barrier function is restored for larger molecules within a few hours. This temporary loss of barrier function provides a " treatment window" through which systemically administered drugs have unrestricted access to spinal tissue in and around the sites of trauma. Taken together, our data provide evidence to support the use of ASIC1a inhibitors as a therapeutic treatment for SCI. This study also emphasizes the importance of objectively grading the functional severity of initial injuries (even when using standardized impacts) and we describe a simple scoring system based on hind limb function that could be adopted in future studies.
Collapse
Affiliation(s)
- Liam M Koehn
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Natassya M Noor
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Qing Dong
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Sing-Yan Er
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Australia
| | - Lachlan D Rash
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Australia; School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - Glenn F King
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Australia
| | | | - Norman R Saunders
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Mark D Habgood
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| |
Collapse
|
24
|
Wang YZ, Wang JJ, Huang Y, Liu F, Zeng WZ, Li Y, Xiong ZG, Zhu MX, Xu TL. Tissue acidosis induces neuronal necroptosis via ASIC1a channel independent of its ionic conduction. eLife 2015; 4. [PMID: 26523449 PMCID: PMC4629285 DOI: 10.7554/elife.05682] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 09/23/2015] [Indexed: 12/15/2022] Open
Abstract
Acidotoxicity is common among neurological disorders, such as ischemic stroke. Traditionally, Ca2+ influx via homomeric acid-sensing ion channel 1a (ASIC1a) was considered to be the leading cause of ischemic acidotoxicity. Here we show that extracellular protons trigger a novel form of neuronal necroptosis via ASIC1a, but independent of its ion-conducting function. We identified serine/threonine kinase receptor interaction protein 1 (RIP1) as a critical component of this form of neuronal necroptosis. Acid stimulation recruits RIP1 to the ASIC1a C-terminus, causing RIP1 phosphorylation and subsequent neuronal death. In a mouse model of focal ischemia, middle cerebral artery occlusion causes ASIC1a-RIP1 association and RIP1 phosphorylation in affected brain areas. Deletion of the Asic1a gene significantly prevents RIP1 phosphorylation and brain damage, suggesting ASIC1a-mediated RIP1 activation has an important role in ischemic neuronal injury. Our findings indicate that extracellular protons function as a novel endogenous ligand that triggers neuronal necroptosis during ischemia via ASIC1a independent of its channel function. DOI:http://dx.doi.org/10.7554/eLife.05682.001 What happens in the minutes and hours after a stroke can determine how much brain damage occurs. In some types of stroke, a blood clot cuts off the blood supply to part of the brain, depriving the brain cells of oxygen and other nutrients, including glucose. One of the consequences is that the blood-starved brain becomes more acidic, which triggers cell death. Protecting brain cells from acidity-induced death could therefore reduce the damage caused by a stroke, and may also be an effective treatment for other brain disorders that involve increased brain acidity, like multiple sclerosis and Huntington's disease. To create such treatments, researchers must first understand how increased acidity in the brain triggers cell death. A protein called the acid-sensing ion channel 1a (ASIC1a) is thought to contribute to acid-induced cell death by allowing calcium to flow into cells. However, this increased flow of calcium occurs only briefly (for seconds) in response to increased acidity, which cannot explain why the severity of cell death strongly depends on the length of increased brain acidity that lasts for hours during stroke. Wang, Wang et al. now show that while ASIC1a is essential for acid-induced brain cell death, this is not because it allows calcium to enter cells. Instead, when acid levels increase, a protein called RIP1 comes to bind to one end of the ASIC1a protein. This causes the addition of a phosphate tag to RIP1, an important cellular process well known to cause the cell to die. Wang, Wang et al. found that in mice genetically engineered to lack ASIC1a, the phosphate tag is not added to RIP1, and the brain cells survive the increased acidity caused by stroke. This suggests that preventing ASIC1a and RIP1 from interacting could be a new way to protect brain cells from the increased acidity caused by brain diseases. DOI:http://dx.doi.org/10.7554/eLife.05682.002
Collapse
Affiliation(s)
- Yi-Zhi Wang
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing-Jing Wang
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Huang
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fan Liu
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Zheng Zeng
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Li
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-Gang Xiong
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, United States
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, United States
| | - Tian-Le Xu
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
25
|
Buta A, Maximyuk O, Kovalskyy D, Sukach V, Vovk M, Ievglevskyi O, Isaeva E, Isaev D, Savotchenko A, Krishtal O. Novel Potent Orthosteric Antagonist of ASIC1a Prevents NMDAR-Dependent LTP Induction. J Med Chem 2015; 58:4449-61. [PMID: 25974655 DOI: 10.1021/jm5017329] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Acid sensing ion channels 1a (ASIC1a) are of crucial importance in numerous physiological and pathological processes in the brain. Here we demonstrate that novel 2-oxo-2H-chromene-3-carboxamidine derivative 5b, designed with molecular modeling approach, inhibits ASIC1a currents with an apparent IC50 of 27 nM when measured at pH 6.7. Acidification to 5.0 decreases the inhibition efficacy by up to 3 orders of magnitude. The 5b molecule not only shifts pH dependence of ASIC1a activation but also inhibits its maximal evoked response. These findings suggest that compound 5b binds to pH sensor of ASIC1a acting as orthosteric noncompetitive antagonist. At 100 nM, compound 5b completely inhibits induction of long-term potentiation (LTP) in CA3-CA1 but not in MF-CA3 synapses. These findings support the knockout data indicating the crucial modulatory role of ASIC1a channels in the NMDAR-dependent LTP and introduce a novel type of ASIC1a antagonists.
Collapse
Affiliation(s)
- Andriy Buta
- †Bogomoletz Institute of Physiology of NAS Ukraine, 4 Bogomoletz Str., 01024 Kyiv, Ukraine.,§State Key Laboratory for Molecular and Cellular Biology, 4 Bogomoletz Str., 01024 Kyiv, Ukraine
| | - Oleksandr Maximyuk
- †Bogomoletz Institute of Physiology of NAS Ukraine, 4 Bogomoletz Str., 01024 Kyiv, Ukraine.,§State Key Laboratory for Molecular and Cellular Biology, 4 Bogomoletz Str., 01024 Kyiv, Ukraine
| | - Dmytro Kovalskyy
- ∥ChemBio Center, Taras Shevchenko University of Kyiv, 67 Chervonotkatska Str., 02094 Kyiv, Ukraine
| | - Volodymyr Sukach
- ‡Institute of Organic Chemistry of NAS Ukraine, 5 Murmanska Str., 02660 Kyiv, Ukraine
| | - Mykhailo Vovk
- ‡Institute of Organic Chemistry of NAS Ukraine, 5 Murmanska Str., 02660 Kyiv, Ukraine
| | - Oleksandr Ievglevskyi
- †Bogomoletz Institute of Physiology of NAS Ukraine, 4 Bogomoletz Str., 01024 Kyiv, Ukraine
| | - Elena Isaeva
- †Bogomoletz Institute of Physiology of NAS Ukraine, 4 Bogomoletz Str., 01024 Kyiv, Ukraine
| | - Dmytro Isaev
- †Bogomoletz Institute of Physiology of NAS Ukraine, 4 Bogomoletz Str., 01024 Kyiv, Ukraine
| | - Alina Savotchenko
- †Bogomoletz Institute of Physiology of NAS Ukraine, 4 Bogomoletz Str., 01024 Kyiv, Ukraine
| | - Oleg Krishtal
- †Bogomoletz Institute of Physiology of NAS Ukraine, 4 Bogomoletz Str., 01024 Kyiv, Ukraine.,§State Key Laboratory for Molecular and Cellular Biology, 4 Bogomoletz Str., 01024 Kyiv, Ukraine
| |
Collapse
|
26
|
Vick JS, Askwith CC. ASICs and neuropeptides. Neuropharmacology 2015; 94:36-41. [PMID: 25592215 DOI: 10.1016/j.neuropharm.2014.12.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 12/04/2014] [Accepted: 12/08/2014] [Indexed: 11/30/2022]
Abstract
The acid sensing ion channels (ASICs) are proton-gated cation channels expressed throughout the nervous system. ASICs are activated during acidic pH fluctuations, and recent work suggests that they are involved in excitatory synaptic transmission. ASICs can also induce neuronal degeneration and death during pathological extracellular acidosis caused by ischemia, autoimmune inflammation, and traumatic injury. Many endogenous neuromodulators target ASICs to affect their biophysical characteristics and contributions to neuronal activity. One of the most unconventional types of modulation occurs with the interaction of ASICs and neuropeptides. Collectively, FMRFamide-related peptides and dynorphins potentiate ASIC activity by decreasing the proton-sensitivity of steady state desensitization independent of G protein-coupled receptor activation. By decreasing the proton-sensitivity of steady state desensitization, the FMRFamide-related peptides and dynorphins permit ASICs to remain active at more acidic basal pH. Unlike the dynorphins, some FMRFamide-related peptides also potentiate ASIC activity by slowing inactivation and increasing the sustained current. Through mechanistic studies, the modulation of ASICs by FMRFamide-related peptides and dynorphins appears to be through distinct interactions with the extracellular domain of ASICs. Dynorphins are expressed throughout the nervous system and can increase neuronal death during prolonged extracellular acidosis, suggesting that the interaction between dynorphins and ASICs may have important consequences for the prevention of neurological injury. The overlap in expression of FMRFamide-related peptides with ASICs in the dorsal horn of the spinal cord suggests that their interaction may have important consequences for the treatment of pain during injury and inflammation. This article is part of the Special Issue entitled 'Acid-Sensing Ion Channels in the Nervous System'.
Collapse
Affiliation(s)
- Jonathan S Vick
- The Department of Neuroscience, The Ohio State University Wexner Medical Center, United States
| | - Candice C Askwith
- The Department of Neuroscience, The Ohio State University Wexner Medical Center, United States.
| |
Collapse
|
27
|
Huang Y, Jiang N, Li J, Ji YH, Xiong ZG, Zha XM. Two aspects of ASIC function: Synaptic plasticity and neuronal injury. Neuropharmacology 2015; 94:42-8. [PMID: 25582290 DOI: 10.1016/j.neuropharm.2014.12.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 12/06/2014] [Accepted: 12/09/2014] [Indexed: 12/17/2022]
Abstract
Extracellular brain pH fluctuates in both physiological and disease conditions. The main postsynaptic proton receptor is the acid-sensing ion channels (ASICs). During the past decade, much progress has been made on protons, ASICs, and neurological disease. This review summarizes the recent progress on synaptic role of protons and our current understanding of how ASICs contribute to various types of neuronal injury in the brain. This article is part of the Special Issue entitled 'Acid-Sensing Ion Channels in the Nervous System'.
Collapse
Affiliation(s)
- Yan Huang
- School of Pharmacy, Anhui Medical University, Hefei, China; Neuroscience Institute, Morehouse School of Medicine, Atlanta GA 30310, USA
| | - Nan Jiang
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA; Lab of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai, China
| | - Jun Li
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Yong-Hua Ji
- Lab of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai, China
| | - Zhi-Gang Xiong
- Neuroscience Institute, Morehouse School of Medicine, Atlanta GA 30310, USA.
| | - Xiang-ming Zha
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA.
| |
Collapse
|
28
|
Lin SH, Sun WH, Chen CC. Genetic exploration of the role of acid-sensing ion channels. Neuropharmacology 2015; 94:99-118. [PMID: 25582292 DOI: 10.1016/j.neuropharm.2014.12.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 12/03/2014] [Accepted: 12/04/2014] [Indexed: 12/27/2022]
Abstract
Advanced gene targeting technology and related tools in mice have been incorporated into studies of acid-sensing ion channels (ASICs). A single ASIC subtype can be knocked out specifically and screened thoroughly for expression in the nervous system at the cellular level. Mapping studies have further shed light on the initiation and identification of related behavioral phenotypes. Here we review studies involving genetically engineered mouse models used to investigate the physiological function of individual ASIC subtypes: ASIC1 (and ASIC1a), ASIC2, ASIC3 and ASIC4. We discuss the detailed expression studies and significant phenotypes revealed with gene knockout for most known Asic subtypes. Each strategy designed to manipulate mouse genetics has advantages and disadvantages. We discuss the limitations of these Asic-knockout models and propose future directions to solve the genetic issues. This article is part of the Special Issue entitled 'Acid-Sensing Ion Channels in the Nervous System'.
Collapse
Affiliation(s)
- Shing-Hong Lin
- Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan.
| | - Wei-Hsin Sun
- Department of Life Sciences, National Central University, Jhongli 32054, Taiwan.
| | - Chih-Cheng Chen
- Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan; Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Academia Sinica, Taipei 115, Taiwan.
| |
Collapse
|
29
|
Protons are a neurotransmitter that regulates synaptic plasticity in the lateral amygdala. Proc Natl Acad Sci U S A 2014; 111:8961-6. [PMID: 24889629 DOI: 10.1073/pnas.1407018111] [Citation(s) in RCA: 208] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Stimulating presynaptic terminals can increase the proton concentration in synapses. Potential receptors for protons are acid-sensing ion channels (ASICs), Na(+)- and Ca(2+)-permeable channels that are activated by extracellular acidosis. Those observations suggest that protons might be a neurotransmitter. We found that presynaptic stimulation transiently reduced extracellular pH in the amygdala. The protons activated ASICs in lateral amygdala pyramidal neurons, generating excitatory postsynaptic currents. Moreover, both protons and ASICs were required for synaptic plasticity in lateral amygdala neurons. The results identify protons as a neurotransmitter, and they establish ASICs as the postsynaptic receptor. They also indicate that protons and ASICs are a neurotransmitter/receptor pair critical for amygdala-dependent learning and memory.
Collapse
|
30
|
The electrogenic sodium bicarbonate cotransporter NBCe1 is a high-affinity bicarbonate carrier in cortical astrocytes. J Neurosci 2014; 34:1148-57. [PMID: 24453308 DOI: 10.1523/jneurosci.2377-13.2014] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The electrogenic sodium bicarbonate cotransporter NBCe1 (SLC4A4) is a robust regulator of intracellular H(+) and a significant base carrier in many cell types. Using wild-type (WT) and NBCe1-deficient (NBC-KO) mice, we have studied the role of NBCe1 in cortical astrocytes in culture and in situ by monitoring intracellular H(+) using the H(+)-sensitive dye BCECF [2',7'-bis-(carboxyethyl)-5-(and-6)-carboxyfluorescein] in wide-field and confocal microscopy. Adding 0.1-3 mm HCO3(-) to an O2-gassed, HEPES-buffered saline solution lowered the intracellular H(+) concentration with a Km of 0.65 mm HCO3(-) in WT astrocytes, but slowly raised [H(+)]i in NBCe1-KO astrocytes. Human NBCe1 heterologously expressed in Xenopus oocytes could be activated by adding 1-3 mm HCO3(-), and even by residual HCO3(-) in a nominally CO2/HCO3(-)-free saline solution. Our results demonstrate a surprisingly high apparent bicarbonate sensitivity mediated by NBCe1 in cortical astrocytes, suggesting that NBCe1 may operate over a wide bicarbonate concentration in these cells.
Collapse
|
31
|
Proton-sensitive cation channels and ion exchangers in ischemic brain injury: new therapeutic targets for stroke? Prog Neurobiol 2014; 115:189-209. [PMID: 24467911 DOI: 10.1016/j.pneurobio.2013.12.008] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 11/28/2013] [Accepted: 12/24/2013] [Indexed: 12/13/2022]
Abstract
Ischemic brain injury results from complicated cellular mechanisms. The present therapy for acute ischemic stroke is limited to thrombolysis with the recombinant tissue plasminogen activator (rtPA) and mechanical recanalization. Therefore, a better understanding of ischemic brain injury is needed for the development of more effective therapies. Disruption of ionic homeostasis plays an important role in cell death following cerebral ischemia. Glutamate receptor-mediated ionic imbalance and neurotoxicity have been well established in cerebral ischemia after stroke. However, non-NMDA receptor-dependent mechanisms, involving acid-sensing ion channel 1a (ASIC1a), transient receptor potential melastatin 7 (TRPM7), and Na(+)/H(+) exchanger isoform 1 (NHE1), have recently emerged as important players in the dysregulation of ionic homeostasis in the CNS under ischemic conditions. These H(+)-sensitive channels and/or exchangers are expressed in the majority of cell types of the neurovascular unit. Sustained activation of these proteins causes excessive influx of cations, such as Ca(2+), Na(+), and Zn(2+), and leads to ischemic reperfusion brain injury. In this review, we summarize recent pre-clinical experimental research findings on how these channels/exchangers are regulated in both in vitro and in vivo models of cerebral ischemia. The blockade or transgenic knockdown of these proteins was shown to be neuroprotective in these ischemia models. Taken together, these non-NMDA receptor-dependent mechanisms may serve as novel therapeutic targets for stroke intervention.
Collapse
|