1
|
Zhou RP, Liang HY, Hu WR, Ding J, Li SF, Chen Y, Zhao YJ, Lu C, Chen FH, Hu W. Modulators of ASIC1a and its potential as a therapeutic target for age-related diseases. Ageing Res Rev 2023; 83:101785. [PMID: 36371015 DOI: 10.1016/j.arr.2022.101785] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 10/30/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022]
Abstract
Age-related diseases have become more common with the advancing age of the worldwide population. Such diseases involve multiple organs, with tissue degeneration and cellular apoptosis. To date, there is a general lack of effective drugs for treatment of most age-related diseases and there is therefore an urgent need to identify novel drug targets for improved treatment. Acid-sensing ion channel 1a (ASIC1a) is a degenerin/epithelial sodium channel family member, which is activated in an acidic environment to regulate pathophysiological processes such as acidosis, inflammation, hypoxia, and ischemia. A large body of evidence suggests that ASIC1a plays an important role in the development of age-related diseases (e.g., stroke, rheumatoid arthritis, Huntington's disease, and Parkinson's disease.). Herein we present: 1) a review of ASIC1a channel properties, distribution, and physiological function; 2) a summary of the pharmacological properties of ASIC1a; 3) and a consideration of ASIC1a as a potential therapeutic target for treatment of age-related disease.
Collapse
Affiliation(s)
- Ren-Peng Zhou
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Hong-Yu Liang
- The Second School of Clinical Medicine, Anhui Medical University, Hefei 230032, China
| | - Wei-Rong Hu
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Jie Ding
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Shu-Fang Li
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Yong Chen
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Ying-Jie Zhao
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Chao Lu
- First Affiliated Hospital, Anhui University of Science & Technology, Huainan 232001, China
| | - Fei-Hu Chen
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
2
|
Talukdar A, Maddhesiya P, Namsa ND, Doley R. Snake venom toxins targeting the central nervous system. TOXIN REV 2022. [DOI: 10.1080/15569543.2022.2084418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Amit Talukdar
- Department of Molecular Biology and Biotechnology, Tezpur University, Assam, India
| | - Priya Maddhesiya
- Cell Biology and Anatomy, Ludwig Maximilian University (LMU), Munich, Germany
| | - Nima Dondu Namsa
- Department of Molecular Biology and Biotechnology, Tezpur University, Assam, India
| | - Robin Doley
- Department of Molecular Biology and Biotechnology, Tezpur University, Assam, India
| |
Collapse
|
3
|
Okada Y, Sato-Numata K, Sabirov RZ, Numata T. Cell Death Induction and Protection by Activation of Ubiquitously Expressed Anion/Cation Channels. Part 2: Functional and Molecular Properties of ASOR/PAC Channels and Their Roles in Cell Volume Dysregulation and Acidotoxic Cell Death. Front Cell Dev Biol 2021; 9:702317. [PMID: 34307382 PMCID: PMC8299559 DOI: 10.3389/fcell.2021.702317] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/18/2021] [Indexed: 12/18/2022] Open
Abstract
For survival and functions of animal cells, cell volume regulation (CVR) is essential. Major hallmarks of necrotic and apoptotic cell death are persistent cell swelling and shrinkage, and thus they are termed the necrotic volume increase (NVI) and the apoptotic volume decrease (AVD), respectively. A number of ubiquitously expressed anion and cation channels play essential roles not only in CVR but also in cell death induction. This series of review articles address the question how cell death is induced or protected with using ubiquitously expressed ion channels such as swelling-activated anion channels, acid-activated anion channels, and several types of TRP cation channels including TRPM2 and TRPM7. In the Part 1, we described the roles of swelling-activated VSOR/VRAC anion channels. Here, the Part 2 focuses on the roles of the acid-sensitive outwardly rectifying (ASOR) anion channel, also called the proton-activated chloride (PAC) anion channel, which is activated by extracellular protons in a manner sharply dependent on ambient temperature. First, we summarize phenotypical properties, the molecular identity, and the three-dimensional structure of ASOR/PAC. Second, we highlight the unique roles of ASOR/PAC in CVR dysfunction and in the induction of or protection from acidotoxic cell death under acidosis and ischemic conditions.
Collapse
Affiliation(s)
- Yasunobu Okada
- National Institute for Physiological Sciences (NIPS), Okazaki, Japan.,Department of Physiology, School of Medicine, Aichi Medical University, Nagakute, Japan.,Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kaori Sato-Numata
- Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan.,Japan Society for the Promotion of Science, Tokyo, Japan
| | - Ravshan Z Sabirov
- Laboratory of Molecular Physiology, Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Tomohiro Numata
- Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
4
|
Uchitel OD, González Inchauspe C, Weissmann C. Synaptic signals mediated by protons and acid-sensing ion channels. Synapse 2019; 73:e22120. [PMID: 31180161 DOI: 10.1002/syn.22120] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/05/2019] [Accepted: 06/05/2019] [Indexed: 01/04/2023]
Abstract
Extracellular pH changes may constitute significant signals for neuronal communication. During synaptic transmission, changes in pH in the synaptic cleft take place. Its role in the regulation of presynaptic Ca2+ currents through multivesicular release in ribbon-type synapses is a proven phenomenon. In recent years, protons have been recognized as neurotransmitters that participate in neuronal communication in synapses of several regions of the CNS such as amygdala, nucleus accumbens, and brainstem. Protons are released by nerve stimulation and activate postsynaptic acid-sensing ion channels (ASICs). Several types of ASIC channels are expressed in the peripheral and central nervous system. The influx of Ca2+ through some subtypes of ASICs, as a result of synaptic transmission, agrees with the participation of ASICs in synaptic plasticity. Pharmacological and genetical inhibition of ASIC1a results in alterations in learning, memory, and phenomena like fear and cocaine-seeking behavior. The recognition of endogenous molecules, such as arachidonic acid, cytokines, histamine, spermine, lactate, and neuropeptides, capable of inhibiting or potentiating ASICs suggests the existence of mechanisms of synaptic modulation that have not yet been fully identified and that could be tuned by new emerging pharmacological compounds with potential therapeutic benefits.
Collapse
Affiliation(s)
- Osvaldo D Uchitel
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado", Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología molecular y Neurociencias (IFIBYNE) CONICET, Universidad de Buenos Aires, Ciudad Universitaria, (C1428EGA), Ciudad Autónoma de Buenos Aires, Argentina
| | - Carlota González Inchauspe
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado", Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología molecular y Neurociencias (IFIBYNE) CONICET, Universidad de Buenos Aires, Ciudad Universitaria, (C1428EGA), Ciudad Autónoma de Buenos Aires, Argentina
| | - Carina Weissmann
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado", Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología molecular y Neurociencias (IFIBYNE) CONICET, Universidad de Buenos Aires, Ciudad Universitaria, (C1428EGA), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
5
|
Soto E, Ortega-Ramírez A, Vega R. Protons as Messengers of Intercellular Communication in the Nervous System. Front Cell Neurosci 2018; 12:342. [PMID: 30364044 PMCID: PMC6191491 DOI: 10.3389/fncel.2018.00342] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/14/2018] [Indexed: 12/18/2022] Open
Abstract
In this review, evidence demonstrating that protons (H+) constitute a complex, regulated intercellular signaling mechanisms are presented. Given that pH is a strictly regulated variable in multicellular organisms, localized extracellular pH changes may constitute significant signals of cellular processes that occur in a cell or a group of cells. Several studies have demonstrated that the low pH of synaptic vesicles implies that neurotransmitter release is always accompanied by the co-release of H+ into the synaptic cleft, leading to transient extracellular pH shifts. Also, evidence has accumulated indicating that extracellular H+ concentration regulation is complex and implies a source of protons in a network of transporters, ion exchangers, and buffer capacity of the media that may finally establish the extracellular proton concentration. The activation of membrane transporters, increased production of CO2 and of metabolites, such as lactate, produce significant extracellular pH shifts in nano- and micro-domains in the central nervous system (CNS), constituting a reliable signal for intercellular communication. The acid sensing ion channels (ASIC) function as specific signal sensors of proton signaling mechanism, detecting subtle variations of extracellular H+ in a range varying from pH 5 to 8. The main question in relation to this signaling system is whether it is only synaptically restricted, or a volume modulator of neuron excitability. This signaling system may have evolved from a metabolic activity detection mechanism to a highly localized extracellular proton dependent communication mechanism. In this study, evidence showing the mechanisms of regulation of extracellular pH shifts and of the ASICs and its function in modulating the excitability in various systems is reviewed, including data and its role in synaptic neurotransmission, volume transmission and even segregated neurotransmission, leading to a reliable extracellular signaling mechanism.
Collapse
Affiliation(s)
- Enrique Soto
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | | | - Rosario Vega
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| |
Collapse
|
6
|
More SV, Choi DK. Emerging preclinical pharmacological targets for Parkinson's disease. Oncotarget 2018; 7:29835-63. [PMID: 26988916 PMCID: PMC5045437 DOI: 10.18632/oncotarget.8104] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/08/2016] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurological condition caused by the degeneration of dopaminergic neurons in the basal ganglia. It is the most prevalent form of Parkinsonism, categorized by cardinal features such as bradykinesia, rigidity, tremors, and postural instability. Due to the multicentric pathology of PD involving inflammation, oxidative stress, excitotoxicity, apoptosis, and protein aggregation, it has become difficult to pin-point a single therapeutic target and evaluate its potential application. Currently available drugs for treating PD provide only symptomatic relief and do not decrease or avert disease progression resulting in poor patient satisfaction and compliance. Significant amount of understanding concerning the pathophysiology of PD has offered a range of potential targets for PD. Several emerging targets including AAV-hAADC gene therapy, phosphodiesterase-4, potassium channels, myeloperoxidase, acetylcholinesterase, MAO-B, dopamine, A2A, mGlu5, and 5-HT-1A/1B receptors are in different stages of clinical development. Additionally, alternative interventions such as deep brain stimulation, thalamotomy, transcranial magnetic stimulation, and gamma knife surgery, are also being developed for patients with advanced PD. As much as these therapeutic targets hold potential to delay the onset and reverse the disease, more targets and alternative interventions need to be examined in different stages of PD. In this review, we discuss various emerging preclinical pharmacological targets that may serve as a new promising neuroprotective strategy that could actually help alleviate PD and its symptoms.
Collapse
Affiliation(s)
- Sandeep Vasant More
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, South Korea
| | - Dong-Kug Choi
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, South Korea
| |
Collapse
|
7
|
Neuroprotective Effects of Paeoniflorin on 6-OHDA-Lesioned Rat Model of Parkinson’s Disease. Neurochem Res 2016; 41:2923-2936. [DOI: 10.1007/s11064-016-2011-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 07/14/2016] [Accepted: 07/16/2016] [Indexed: 02/01/2023]
|
8
|
Zhou RP, Wu XS, Wang ZS, Xie YY, Ge JF, Chen FH. Novel Insights into Acid-Sensing Ion Channels: Implications for Degenerative Diseases. Aging Dis 2015; 7:491-501. [PMID: 27493834 DOI: 10.14336/ad.2015.1213] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/13/2015] [Indexed: 12/16/2022] Open
Abstract
Degenerative diseases often strike older adults and are characterized by progressive deterioration of cells, eventually leading to tissue and organ degeneration for which limited effective treatment options are currently available. Acid-sensing ion channels (ASICs), a family of extracellular H(+)-activated ligand-gated ion channels, play critical roles in physiological and pathological conditions. Aberrant activation of ASICs is reported to regulate cell apoptosis, differentiation and autophagy. Accumulating evidence has highlighted a dramatic increase and activation of ASICs in degenerative disorders, including multiple sclerosis, Parkinson's disease, Huntington's disease, intervertebral disc degeneration and arthritis. In this review, we have comprehensively discussed the critical roles of ASICs and their potential utility as therapeutic targets in degenerative diseases.
Collapse
Affiliation(s)
- Ren-Peng Zhou
- 1Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China; 2The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Xiao-Shan Wu
- 1Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China; 2The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Zhi-Sen Wang
- 1Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China; 2The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Ya-Ya Xie
- 1Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China; 2The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Jin-Fang Ge
- 1Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China; 2The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Fei-Hu Chen
- 1Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China; 2The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| |
Collapse
|
9
|
Liu S, Cheng XY, Wang F, Liu CF. Acid-sensing ion channels: potential therapeutic targets for neurologic diseases. Transl Neurodegener 2015; 4:10. [PMID: 26029363 PMCID: PMC4449961 DOI: 10.1186/s40035-015-0031-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 05/14/2015] [Indexed: 01/14/2023] Open
Abstract
Maintaining the physiological pH of interstitial fluid is crucial for normal cellular functions. In disease states, tissue acidosis is a common pathologic change causing abnormal activation of acid-sensing ion channels (ASICs), which according to cumulative evidence, significantly contributes to inflammation, mitochondrial dysfunction, and other pathologic mechanisms (i.e., pain, stroke, and psychiatric conditions). Thus, it has become increasingly clear that ASICs are critical in the progression of neurologic diseases. This review is focused on the importance of ASICs as potential therapeutic targets in combating neurologic diseases.
Collapse
Affiliation(s)
- Sha Liu
- />Department of Neurology, the Second Affiliated Hospital of Soochow University, Soochow University, 1055 Sanxiang Road, Suzhou, 215004 China
| | - Xiao-Yu Cheng
- />Department of Neurology, the Second Affiliated Hospital of Soochow University, Soochow University, 1055 Sanxiang Road, Suzhou, 215004 China
| | - Fen Wang
- />Department of Neurology, the Second Affiliated Hospital of Soochow University, Soochow University, 1055 Sanxiang Road, Suzhou, 215004 China
- />Institute of Neuroscience, Soochow University, Suzhou, 215123 China
| | - Chun-Feng Liu
- />Department of Neurology, the Second Affiliated Hospital of Soochow University, Soochow University, 1055 Sanxiang Road, Suzhou, 215004 China
- />Institute of Neuroscience, Soochow University, Suzhou, 215123 China
- />Beijing Key Laboratory for Parkinson’s Disease, Beijing, 100053 China
| |
Collapse
|
10
|
de Ceglia R, Chaabane L, Biffi E, Bergamaschi A, Ferrigno G, Amadio S, Del Carro U, Mazzocchi N, Comi G, Bianchi V, Taverna S, Forti L, D'Adamo P, Martino G, Menegon A, Muzio L. Down-sizing of neuronal network activity and density of presynaptic terminals by pathological acidosis are efficiently prevented by Diminazene Aceturate. Brain Behav Immun 2015; 45:263-76. [PMID: 25499583 DOI: 10.1016/j.bbi.2014.12.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 12/02/2014] [Accepted: 12/02/2014] [Indexed: 11/26/2022] Open
Abstract
Local acidosis is associated with neuro-inflammation and can have significant effects in several neurological disorders, including multiple sclerosis, brain ischemia, spinal cord injury and epilepsy. Despite local acidosis has been implicated in numerous pathological functions, very little is known about the modulatory effects of pathological acidosis on the activity of neuronal networks and on synaptic structural properties. Using non-invasive MRI spectroscopy we revealed protracted extracellular acidosis in the CNS of Experimental Autoimmune Encephalomyelitis (EAE) affected mice. By multi-unit recording in cortical neurons, we established that acidosis affects network activity, down-sizing firing and bursting behaviors as well as amplitudes. Furthermore, a protracted acidosis reduced the number of presynaptic terminals, while it did not affect the postsynaptic compartment. Application of the diarylamidine Diminazene Aceturate (DA) during acidosis significantly reverted both the loss of neuronal firing and bursting and the reduction of presynaptic terminals. Finally, in vivo DA delivery ameliorated the clinical disease course of EAE mice, reducing demyelination and axonal damage. DA is known to block acid-sensing ion channels (ASICs), which are proton-gated, voltage-insensitive, Na(+) permeable channels principally expressed by peripheral and central nervous system neurons. Our data suggest that ASICs activation during acidosis modulates network electrical activity and exacerbates neuro-degeneration in EAE mice. Therefore pharmacological modulation of ASICs in neuroinflammatory diseases could represent a new promising strategy for future therapies aimed at neuro-protection.
Collapse
Affiliation(s)
- Roberta de Ceglia
- Neuroimmunolgy Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, San Raffaele Scientific Institute, Italy
| | - Linda Chaabane
- Neuroimmunolgy Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, San Raffaele Scientific Institute, Italy; Department of Neurology, Institute of Experimental Neurology (INSPE), Vita Salute San Raffaele University, Milan, Italy
| | - Emilia Biffi
- Neuroengineering and Medical Robotics Laboratory, Department of Electronics, Information and Bioengineering, Politecnico di Milano, Italy; Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Andrea Bergamaschi
- Neuroimmunolgy Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, San Raffaele Scientific Institute, Italy
| | - Giancarlo Ferrigno
- Neuroengineering and Medical Robotics Laboratory, Department of Electronics, Information and Bioengineering, Politecnico di Milano, Italy
| | - Stefano Amadio
- Neurophysiology Unit, Division of Neuroscience, San Raffaele Scientific Institute, Italy
| | - Ubaldo Del Carro
- Neurophysiology Unit, Division of Neuroscience, San Raffaele Scientific Institute, Italy
| | - Nausicaa Mazzocchi
- Advanced Light and Electron Microscopy Bio-Imaging Centre, Experimental Imaging Centre, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giancarlo Comi
- Department of Neurology, Institute of Experimental Neurology (INSPE), Vita Salute San Raffaele University, Milan, Italy
| | - Veronica Bianchi
- Dulbecco Telethon Institute at San Raffaele Scientific Institute, Division of Neuroscience, 20132 Milan, Italy
| | - Stefano Taverna
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Lia Forti
- Center for Neuroscience and Dept. of Theoretical and Applied Sciences, Biomedical Division, University of Insubria, 21052 Busto Arsizio, Italy
| | - Patrizia D'Adamo
- Dulbecco Telethon Institute at San Raffaele Scientific Institute, Division of Neuroscience, 20132 Milan, Italy
| | - Gianvito Martino
- Neuroimmunolgy Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, San Raffaele Scientific Institute, Italy.
| | - Andrea Menegon
- Advanced Light and Electron Microscopy Bio-Imaging Centre, Experimental Imaging Centre, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Luca Muzio
- Neuroimmunolgy Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, San Raffaele Scientific Institute, Italy.
| |
Collapse
|
11
|
Du J, Reznikov LR, Welsh MJ. Expression and activity of acid-sensing ion channels in the mouse anterior pituitary. PLoS One 2014; 9:e115310. [PMID: 25506946 PMCID: PMC4266673 DOI: 10.1371/journal.pone.0115310] [Citation(s) in RCA: 11] [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: 07/25/2014] [Accepted: 11/21/2014] [Indexed: 12/20/2022] Open
Abstract
Acid sensing ion channels (ASICs) are proton-gated cation channels that are expressed in the nervous system and play an important role in fear learning and memory. The function of ASICs in the pituitary, an endocrine gland that contributes to emotions, is unknown. We sought to investigate which ASIC subunits were present in the pituitary and found mRNA expression for all ASIC isoforms, including ASIC1a, ASIC1b, ASIC2a, ASIC2b, ASIC3 and ASIC4. We also observed acid-evoked ASIC-like currents in isolated anterior pituitary cells that were absent in mice lacking ASIC1a. The biophysical properties and the responses to PcTx1, amiloride, Ca2+ and Zn2+ suggested that ASIC currents were mediated predominantly by heteromultimeric channels that contained ASIC1a and ASIC2a or ASIC2b. ASIC currents were also sensitive to FMRFamide (Phe-Met-Arg-Phe amide), suggesting that FMRFamide-like compounds might endogenously regulate pituitary ASICs. To determine whether ASICs might regulate pituitary cell function, we applied low pH and found that it increased the intracellular Ca2+ concentration. These data suggest that ASIC channels are present and functionally active in anterior pituitary cells and may therefore influence their function.
Collapse
Affiliation(s)
- Jianyang Du
- Howard Hughes Medical Institute, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
- Department of Internal Medicine, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Leah R. Reznikov
- Department of Internal Medicine, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Michael J. Welsh
- Howard Hughes Medical Institute, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
- Department of Internal Medicine, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
- Department of Molecular Physiology and Biophysics, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|
12
|
Boillat A, Alijevic O, Kellenberger S. Calcium entry via TRPV1 but not ASICs induces neuropeptide release from sensory neurons. Mol Cell Neurosci 2014; 61:13-22. [DOI: 10.1016/j.mcn.2014.04.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 03/14/2014] [Accepted: 04/23/2014] [Indexed: 11/25/2022] Open
|
13
|
Li MH, Liu SQ, Inoue K, Lan J, Simon RP, Xiong ZG. Acid-sensing ion channels in mouse olfactory bulb M/T neurons. ACTA ACUST UNITED AC 2014; 143:719-31. [PMID: 24821964 PMCID: PMC4035746 DOI: 10.1085/jgp.201310990] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Functional acid-sensing ion channels are present in olfactory bulb neurons and may contribute to normal olfaction. The olfactory bulb contains the first synaptic relay in the olfactory pathway, the sensory system in which odorants are detected enabling these chemical stimuli to be transformed into electrical signals and, ultimately, the perception of odor. Acid-sensing ion channels (ASICs), a family of proton-gated cation channels, are widely expressed in neurons of the central nervous system. However, no direct electrophysiological and pharmacological characterizations of ASICs in olfactory bulb neurons have been described. Using a combination of whole-cell patch-clamp recordings and biochemical and molecular biological analyses, we demonstrated that functional ASICs exist in mouse olfactory bulb mitral/tufted (M/T) neurons and mainly consist of homomeric ASIC1a and heteromeric ASIC1a/2a channels. ASIC activation depolarized cultured M/T neurons and increased their intracellular calcium concentration. Thus, ASIC activation may play an important role in normal olfactory function.
Collapse
Affiliation(s)
- Ming-Hua Li
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR 97239
| | | | - Koichi Inoue
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA 30310
| | | | - Roger P Simon
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA 30310
| | - Zhi-Gang Xiong
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA 30310
| |
Collapse
|
14
|
Chu XP, Grasing KA, Wang JQ. Acid-sensing ion channels contribute to neurotoxicity. Transl Stroke Res 2013; 5:69-78. [PMID: 24323724 DOI: 10.1007/s12975-013-0305-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 10/06/2013] [Accepted: 11/04/2013] [Indexed: 12/13/2022]
Abstract
Acidosis that occurs under pathological conditions not only affects intracellular signaling molecules, but also directly activates a unique family of ligand-gated ion channels: acid-sensing ion channels (ASICs). ASICs are widely expressed throughout the central and peripheral nervous systems and play roles in pain sensation, learning and memory, and fear conditioning. Overactivation of ASICs contributes to neurodegenerative diseases such as ischemic brain/spinal cord injury, multiple sclerosis, Parkinson's disease, and Huntington's disease. Thus, targeting ASICs might be a potential therapeutic strategy for these conditions. This mini-review focuses on the electrophysiology and pharmacology of ASICs and roles of ASICs in neuronal toxicity.
Collapse
Affiliation(s)
- Xiang-Ping Chu
- Departments of Basic Medical Science and Anesthesiology, School of Medicine, University of Missouri-Kansas City, 2411 Holmes Street, Room M3-417, Kansas City, MO, 64108-2792, USA,
| | | | | |
Collapse
|
15
|
Bhattacharya P, Pandey AK, Paul S, Patnaik R, Yavagal DR. Aquaporin-4 inhibition mediates piroxicam-induced neuroprotection against focal cerebral ischemia/reperfusion injury in rodents. PLoS One 2013; 8:e73481. [PMID: 24023878 PMCID: PMC3762750 DOI: 10.1371/journal.pone.0073481] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 07/22/2013] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND AND PURPOSE Aquaporin-4(AQP4) is an abundant water channel protein in brain that regulates water transport to maintain homeostasis. Cerebral edema resulting from AQP4 over expression is considered to be one of the major determinants for progressive neuronal insult during cerebral ischemia. Although, both upregulation and downregulation of AQP4 expression is associated with brain pathology, over expression of AQP4 is one of the chief contributors of water imbalance in brain during ischemic pathology. We have found that Piroxicam binds to AQP4 with optimal binding energy value. Thus, we hypothesized that Piroxicam is neuroprotective in the rodent cerebral ischemic model by mitigating cerebral edema via AQP4 regulation. METHODS Rats were treated with Piroxicam OR placebo at 30 min prior, 2 h post and 4 h post 60 minutes of MCAO followed by 24 hour reperfusion. Rats were evaluated for neurological deficits and motor function just before sacrifice. Brains were harvested for infarct size estimation, water content measurement, biochemical analysis, RT-PCR and western blot experiments. RESULTS Piroxicam pretreatment thirty minutes prior to ischemia and four hour post reperfusion afforded neuroprotection as evident through significant reduction in cerebral infarct volume, improvement in motor behavior, neurological deficit and reduction in brain edema. Furthermore, ischemia induced surge in levels of nitrite and malondialdehyde were also found to be significantly reduced in ischemic brain regions in treated animals. This neuroprotection was found to be associated with inhibition of acid mediated rise in intracellular calcium levels and also downregulated AQP4 expression. CONCLUSIONS Findings of the present study provide significant evidence that Piroxicam acts as a potent AQP4 regulator and renders neuroprotection in focal cerebral ischemia. Piroxicam could be clinically exploited for the treatment of brain stroke along with other anti-stroke therapeutics in future.
Collapse
Affiliation(s)
- Pallab Bhattacharya
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, (U.P.), India
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Anand Kumar Pandey
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, (U.P.), India
| | - Sudip Paul
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, (U.P.), India
- Department of Biomedical Engineering, North Eastern Hill University (NEHU), Shillong, Meghalaya, India
| | - Ranjana Patnaik
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, (U.P.), India
| | - Dileep R. Yavagal
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| |
Collapse
|
16
|
Bae YJ, Yoo JC, Park N, Kang D, Han J, Hwang E, Park JY, Hong SG. Acute Hypoxia Activates an ENaC-like Channel in Rat Pheochromocytoma (PC12) Cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2013; 17:57-64. [PMID: 23440317 PMCID: PMC3579106 DOI: 10.4196/kjpp.2013.17.1.57] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 11/28/2012] [Accepted: 12/04/2012] [Indexed: 12/17/2022]
Abstract
Cells can resist and even recover from stress induced by acute hypoxia, whereas chronic hypoxia often leads to irreversible damage and eventually death. Although little is known about the response(s) to acute hypoxia in neuronal cells, alterations in ion channel activity could be preferential. This study aimed to elucidate which channel type is involved in the response to acute hypoxia in rat pheochromocytomal (PC12) cells as a neuronal cell model. Using perfusing solution saturated with 95% N(2) and 5% CO(2), induction of cell hypoxia was confirmed based on increased intracellular Ca(2+) with diminished oxygen content in the perfusate. During acute hypoxia, one channel type with a conductance of about 30 pS (2.5 pA at -80 mV) was activated within the first 2~3 min following onset of hypoxia and was long-lived for more than 300 ms with high open probability (P(o), up to 0.8). This channel was permeable to Na(+) ions, but not to K(+), Ca(+), and Cl(-) ions, and was sensitively blocked by amiloride (200 nM). These characteristics and behaviors were quite similar to those of epithelial sodium channel (ENaC). RT-PCR and Western blot analyses confirmed that ENaC channel was endogenously expressed in PC12 cells. Taken together, a 30-pS ENaC-like channel was activated in response to acute hypoxia in PC12 cells. This is the first evidence of an acute hypoxia-activated Na(+) channel that can contribute to depolarization of the cell.
Collapse
Affiliation(s)
- Yeon Ju Bae
- Department of Physiology, Institute of Health Sciences and Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju 660-751, Korea
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Takahashi K, Ohta T. Low pH enhances 2-aminoethoxydiphenyl borate-induced cell death of PC12 cells. Toxicol Lett 2012; 215:161-6. [DOI: 10.1016/j.toxlet.2012.10.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 10/17/2012] [Accepted: 10/18/2012] [Indexed: 11/15/2022]
|
18
|
Combination therapy of Ifenprodil with Piroxicam may be an effective therapeutic intervention in cerebral stroke: A hypothesis. Med Hypotheses 2012; 79:516-8. [DOI: 10.1016/j.mehy.2012.07.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 07/08/2012] [Indexed: 11/19/2022]
|
19
|
Bhattacharya P, Pandey AK, Paul S, Patnaik R. Neuroprotective potential of Piroxicam in cerebral ischemia: An in silico evaluation of the hypothesis to explore its therapeutic efficacy by inhibition of aquaporin-4 and acid sensing ion channel1a. Med Hypotheses 2012; 79:352-7. [DOI: 10.1016/j.mehy.2012.05.033] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 05/02/2012] [Accepted: 05/21/2012] [Indexed: 11/17/2022]
|
20
|
Abstract
Acid-sensing ion channels (ASICs) are present in neurons and may contribute to chemoreception. Among six subunits of ASICs, ASIC1 is mainly expressed in the central nervous system. Recently, multiple sites in the brain including the lateral hypothalamus (LH) have been found to be sensitive to extracellular acidification. Since LH contains orexin neurons and innervates the medulla respiratory center, we hypothesize that ASIC1 is expressed on the orexin neuron and contributes to acid-induced increase in respiratory drive. To test this hypothesis, we used double immunofluorescence to determine whether ASIC1 is expressed on orexin neurons in the LH, and assessed integrated phrenic nerve discharge (iPND) in intact rats in response to acidification of the LH. We found that ASIC1 was co-localized with orexinA in the LH. Microinjection of acidified artificial cerebrospinal fluid increased the amplitude of iPND by 70% (pH 7.4 v.s. pH 6.5:1.05±0.12 v.s. 1.70±0.10, n = 6, P<0.001) and increased the respiratory drive (peak amplitude of iPND/inspiratory time, PA/Ti) by 40% (1.10±0.23 v.s. 1.50±0.38, P<0.05). This stimulatory effect was abolished by blocking ASIC1 with a nonselective inhibitor (amiloride 10 mM), a selective inhibitor (PcTX1, 10 nM) or by damaging orexin neurons in the LH. Current results support our hypothesis that the orexin neuron in the LH can exert an excitation on respiration via ASIC1 during local acidosis. Since central acidification is involved in breathing dysfunction in a variety of pulmonary diseases, understanding its underlying mechanism may improve patient management.
Collapse
|
21
|
Song N, Zhang G, Geng W, Liu Z, Jin W, Li L, Cao Y, Zhu D, Yu J, Shen L. Acid sensing ion channel 1 in lateral hypothalamus contributes to breathing control. PLoS One 2012; 7:e39982. [PMID: 22792205 PMCID: PMC3391217 DOI: 10.1371/journal.pone.0039982] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 05/30/2012] [Indexed: 12/20/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are present in neurons and may contribute to chemoreception. Among six subunits of ASICs, ASIC1 is mainly expressed in the central nervous system. Recently, multiple sites in the brain including the lateral hypothalamus (LH) have been found to be sensitive to extracellular acidification. Since LH contains orexin neurons and innervates the medulla respiratory center, we hypothesize that ASIC1 is expressed on the orexin neuron and contributes to acid-induced increase in respiratory drive. To test this hypothesis, we used double immunofluorescence to determine whether ASIC1 is expressed on orexin neurons in the LH, and assessed integrated phrenic nerve discharge (iPND) in intact rats in response to acidification of the LH. We found that ASIC1 was co-localized with orexinA in the LH. Microinjection of acidified artificial cerebrospinal fluid increased the amplitude of iPND by 70% (pH 7.4 v.s. pH 6.5:1.05±0.12 v.s. 1.70±0.10, n = 6, P<0.001) and increased the respiratory drive (peak amplitude of iPND/inspiratory time, PA/Ti) by 40% (1.10±0.23 v.s. 1.50±0.38, P<0.05). This stimulatory effect was abolished by blocking ASIC1 with a nonselective inhibitor (amiloride 10 mM), a selective inhibitor (PcTX1, 10 nM) or by damaging orexin neurons in the LH. Current results support our hypothesis that the orexin neuron in the LH can exert an excitation on respiration via ASIC1 during local acidosis. Since central acidification is involved in breathing dysfunction in a variety of pulmonary diseases, understanding its underlying mechanism may improve patient management.
Collapse
Affiliation(s)
- Nana Song
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guihong Zhang
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenye Geng
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zibing Liu
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weizhong Jin
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Li Li
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yinxiang Cao
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Danian Zhu
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jerry Yu
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pulmonary Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Linlin Shen
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
- * E-mail:
| |
Collapse
|
22
|
Cognitive effects of NSAIDs in cerebral ischemia: a hypothesis exploring mechanical action mediated pharmacotherapy. Med Hypotheses 2012; 79:393-5. [PMID: 22771072 DOI: 10.1016/j.mehy.2012.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 05/26/2012] [Accepted: 06/03/2012] [Indexed: 12/19/2022]
Abstract
Cerebral ischemia is associated with altered neuronal mechanics leading to dynamic reshaping of neuronal structures, giving rise to a cascade of biological pathways leading to many deleterious consequences and cognitive deficits. Memory and learning specifically are mediated by neurotransmitter release from vesicles clustered at the synapse. Mechanical tension is an important factor governing the amount of vesicular neurotransmitter release in response to an action potential. Neuroinflammation in cerebral ischemia leads to altered mechanical/physical forces on neurons which gives rise to abnormal mechanical tension along the neuron resulting in neurotransmitter imbalance leading to cognitive dysfunction. We consider the possibility that modulation of mechanical forces on neurons may be a therapeutic strategy to help prevent cognitive deficit in cerebral ischemia. Here we show how NSAIDs may act as candidate pharmacological molecules which have the ability to inhibit neuroinflammation and which can alter neuronal mechanics by their COX-2 inhibiting property.
Collapse
|
23
|
An in-silico strategy to explore neuroprotection by quercetin in cerebral ischemia: a novel hypothesis based on inhibition of matrix metalloproteinase (MMPs) and acid sensing ion channel 1a (ASIC1a). Med Hypotheses 2012; 79:76-81. [PMID: 22543073 DOI: 10.1016/j.mehy.2012.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 01/10/2012] [Accepted: 04/01/2012] [Indexed: 12/17/2022]
Abstract
Cerebral ischemia are caused by acute interruption of the brain arterial blood supply, typically by a thrombus or embolus, leading to neuronal insult and the remainder damage are caused by blood vessel rupture, leading to hemorrhage. Acidosis and matrix metalloproteinase activation are the central and prominent metabolic feature of ischemic brain. The combined inhibition of MMPs and ASIC1a channels can offer a new therapeutic approach in cerebral stroke management. Moreover, the combined inhibition of MMPs and ASIC1a with flavonoids remains unknown against neuroprotection in animal models of cerebral ischemia. Flavonoids are believed to act as health-promoting substances and some of them have antioxidant and anti-inflammatory properties. Therefore, the target of the present study was in-silico evaluation of the neuroprotective efficacy of quercetin in rat model of focal cerebral ischemia/reperfusion (I/R) injury and efforts were made to analyze its inhibitory effects on MMPs activation and ASIC1a channels mediated downstream survival/damage mechanisms. Thus on the basis of our in-silico studies we hypothesize that quercetin can be a neuroprotective agent in rat model of focal cerebral ischemia/reperfusion (I/R) injury due to its inhibitory effects on MMPs activation and ASIC1a channels mediated downstream survival/damage mechanisms.
Collapse
|
24
|
Bhattacharya P, Pandey AK, Paul S. Minocycline and magnesium in combination may be a good therapeutic intervention for cerebral ischemia. Med Hypotheses 2011; 77:1129-31. [PMID: 21985758 DOI: 10.1016/j.mehy.2011.09.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Accepted: 09/13/2011] [Indexed: 11/17/2022]
Abstract
A neuroprotective strategy through a combination therapy is always being superior to any other singular therapeutic interventions, as these acts through a multifauceted approach within the brain during cerebral ischemia. Therefore, the development of a potential new combination of drug is necessitated which can bring about desirable improved neuroprotection targeting different pathways against ischemic stroke. Numerous past studies have enumerated the neuroprotective roles of minocycline and magnesium administered in single against cerebral ischemia in animal model hence we hypothesized that by using magnesium with minocycline in combination would provide additive neuroprotection than either of the agents used alone. In this article, we discuss our hypothesis regarding the possibility of minocycline and magnesium as a potent combination which may have a positive therapeutic role in treatment of cerebral ischemia through its anti-inflammatory, anti-apoptotic and anti-oxidative characteristics with magnesium contributing as a regulator of increased calcium influx.
Collapse
Affiliation(s)
- Pallab Bhattacharya
- School of Biomedical Engineering, Institute of Technology, Banaras Hindu University, Varanasi 221005, UP, India.
| | | | | |
Collapse
|
25
|
Sun X, Cao YB, Hu LF, Yang YP, Li J, Wang F, Liu CF. ASICs mediate the modulatory effect by paeoniflorin on alpha-synuclein autophagic degradation. Brain Res 2011; 1396:77-87. [DOI: 10.1016/j.brainres.2011.04.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 04/02/2011] [Accepted: 04/05/2011] [Indexed: 11/28/2022]
|
26
|
Li MH, Inoue K, Si HF, Xiong ZG. Calcium-permeable ion channels involved in glutamate receptor-independent ischemic brain injury. Acta Pharmacol Sin 2011; 32:734-40. [PMID: 21552295 DOI: 10.1038/aps.2011.47] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Brain ischemia is a leading cause of death and long-term disabilities worldwide. Unfortunately, current treatment is limited to thrombolysis, which has limited success and a potential side effect of intracerebral hemorrhage. Searching for new cell injury mechanisms and therapeutic interventions has become a major challenge in the field. It has been recognized for many years that intracellular Ca(2+) overload in neurons is essential for neuronal injury associated with brain ischemia. However, the exact pathway(s) underlying the toxic Ca(2+) loading remained elusive. This review discusses the role of two Ca(2+)-permeable cation channels, TRPM7 and acid-sensing channels, in glutamate-independent Ca(2+) toxicity associated with brain ischemia.
Collapse
|
27
|
Pandey AK, Hazari PP, Patnaik R, Mishra AK. The role of ASIC1a in neuroprotection elicited by quercetin in focal cerebral ischemia. Brain Res 2011; 1383:289-99. [PMID: 21281608 DOI: 10.1016/j.brainres.2011.01.085] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 01/21/2011] [Accepted: 01/21/2011] [Indexed: 12/29/2022]
Abstract
One of the major instigators to neuronal cell death and brain damage following cerebral ischemia is calcium dysregulation. The intracellular calcium overload resulting from glutamate excitotoxicity is considered a major determinant for neuronal loss during cerebral ischemia. Moreover, ASIC1a activation due to acidosis also promotes intracellular calcium overload during ischemic insult. Interestingly, ASIC1a was found to be inhibited by some flavonoids which carry an anti-inflammatory property particularly quercetin, which could be exploited in hypoxic conditions like cerebral ischemia. This encourages us to investigate the neuroprotective effect of quercetin besides its possible downstream signaling mechanism in focal cerebral ischemia. The treatment of quercetin 30min before ischemia and 4h after reperfusion shows significant protection from ischemic injury as noticed by reduction in cerebral infarct volume and neurobehavioral deficit. In addition to earlier calcium dependent rise in the levels of nitrite and MDA exhibited marked reduction (P<0.01) in their levels when given quercetin pretreatment in ischemic brain regions. The quercetin treatment also reduced the spectrin break down products (SBDP) caused by ischemic activation of calcium dependent protease calpain. In ex-vivo study, it was also observed that quercetin inhibited the acid mediated intracellular calcium levels in rat brain synaptoneurosomes. These studies suggest the neuroprotective role of quercetin in focal cerebral ischemia by regulation of ASIC1a.
Collapse
Affiliation(s)
- Anand Kumar Pandey
- Division of Cyclotron & Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig SK Mazumdar Road, Delhi 110054, India
| | | | | | | |
Collapse
|
28
|
The amiloride derivative phenamil attenuates pulmonary vascular remodeling by activating NFAT and the bone morphogenetic protein signaling pathway. Mol Cell Biol 2010; 31:517-30. [PMID: 21135135 DOI: 10.1128/mcb.00884-10] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Pulmonary artery hypertension (PAH) is characterized by elevated pulmonary artery resistance and increased medial thickness due to deregulation of vascular remodeling. Inactivating mutations of the BMPRII gene, which encodes a receptor for bone morphogenetic proteins (BMPs), are identified in ∼60% of familial PAH (FPAH) and ∼30% of idiopathic PAH (IPAH) patients. It has been hypothesized that constitutive reduction in BMP signal by BMPRII mutations may cause abnormal vascular remodeling by promoting dedifferentiation of vascular smooth muscle cells (vSMCs). Here, we demonstrate that infusion of the amiloride analog phenamil during chronic-hypoxia treatment in rat attenuates development of PAH and vascular remodeling. Phenamil induces Tribbles homolog 3 (Trb3), a positive modulator of the BMP pathway that acts by stabilizing the Smad family signal transducers. Through induction of Trb3, phenamil promotes the differentiated, contractile vSMC phenotype characterized by elevated expression of contractile genes and reduced cell growth and migration. Phenamil activates the Trb3 gene transcription via activation of the calcium-calcineurin-nuclear factor of activated T cell (NFAT) pathway. These results indicate that constitutive elevation of Trb3 by phenamil is a potential therapy for IPAH and FPAH.
Collapse
|
29
|
Mishra V, Verma R, Raghubir R. Neuroprotective effect of flurbiprofen in focal cerebral ischemia: The possible role of ASIC1a. Neuropharmacology 2010; 59:582-8. [DOI: 10.1016/j.neuropharm.2010.08.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 08/08/2010] [Accepted: 08/16/2010] [Indexed: 01/09/2023]
|
30
|
Li M, Kratzer E, Inoue K, Simon RP, Xiong ZG. Developmental change in the electrophysiological and pharmacological properties of acid-sensing ion channels in CNS neurons. J Physiol 2010; 588:3883-900. [PMID: 20724366 DOI: 10.1113/jphysiol.2010.192922] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are proton-gated cation channels that play important roles in the CNS including synaptic plasticity and acidosis-mediated neuronal injury. ASIC1a and ASIC2a subunits are predominant in CNS neurons, where homomultimeric and heteromultimeric channel configurations co-exist. Since ASIC1a and ASIC2a have dramatic differences in pH sensitivity, Ca(2+) permeability and channel kinetics, any change in the level of individual subunits may have significant effects on the properties and functions of ASICs. Using patch-clamp recording, fluorescent Ca(2+) imaging and molecular biological techniques, we show dramatic developmental changes in the properties of ASICs in mouse cortical neurons. For example, the amplitude of ASIC currents increases whereas desensitization decreases with neuronal maturation. Decreased H(+) affinity and acid-evoked [Ca(2+)](i) but increased Zn(2+) potentiation were also recorded in mature neurons. RT-PCR revealed significant increases in the ratio of ASIC2/ASIC1 mRNA with neuronal maturation. Thus, contributions of ASIC1a and ASIC2a to overall ASIC-mediated responses undergo distinct developmental changes. These findings may help in understanding the precise role of ASICs in physiological and pathological conditions at different developmental stages.
Collapse
Affiliation(s)
- Minghua Li
- Robert S. Dow Neurobiology Laboratories, Legacy Clinical Research Center, 1225 NE 2nd Ave, Portland, OR 97232, USA
| | | | | | | | | |
Collapse
|
31
|
Cao BY, Yang YP, Luo WF, Mao CJ, Han R, Sun X, Cheng J, Liu CF. Paeoniflorin, a potent natural compound, protects PC12 cells from MPP+ and acidic damage via autophagic pathway. JOURNAL OF ETHNOPHARMACOLOGY 2010; 131:122-129. [PMID: 20558269 DOI: 10.1016/j.jep.2010.06.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Revised: 04/26/2010] [Accepted: 06/04/2010] [Indexed: 05/29/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Paeoniflorin (PF) is the principal bioactive component of Radix Paeoniae alba, which is widely used in Traditional Chinese Medicine for the treatment of neurodegenerative disorders such as Parkinson's disease (PD). AIM OF THE STUDY To evaluate the neuroprotective effects of PF on MPP(+)- or acid- (pH 5.0) induced injury in cultured PC12 cells and to investigate the activity of autophagy-lysosome pathway (ALP). Amiloride (Ami), a non-selective blocker of acid-sensing ion channels (ASICs), as a positive control drug, since it is neuroprotective in rodent models of PD. MATERIALS AND METHODS The cell viability was analyzed with MTT assay. The cell injury was assessed by lactate dehydrogenase (LDH) assay. Flow cytometry and Western blot analysis were used to study the apoptotic, calcium influx and autophagic mechanisms. RESULTS Ami (100 microM) and PF (50 microM) both protected PC12 cells against MPP(+)- or acid-induced injury as assessed by MTT assay, lactate dehydrogenase release, and apoptosis rate. The concentrations of cytosolic free Ca(2+) were raised after exposure to MPP(+) or acidosis, while Ami and PF both reduced the influx of Ca(2+). More importantly, we found that the mechanisms of neuroprotective effects of Ami and PF were closely associated with the upregulation of LC3-II protein, which is specifically associated with autophagic vacuole membranes. Furthermore, application of MPP(+) or acid induced the overexpression of LAMP2a, which is directly correlated with the activity of the chaperone-mediated autophagy pathway. However, Ami and PF inhibited the overexpression of LAMP2a. CONCLUSIONS Our data provide the first experimental evidence that PF modulates autophagy in models of neuron injury, as well as providing the first indication of a relationship between ASICs and ALP.
Collapse
Affiliation(s)
- Bi-Yin Cao
- Department of Neurology, Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Suman A, Mehta B, Guo ML, Chu XP, Fibuch EE, Mao LM, Wang JQ. Alterations in subcellular expression of acid-sensing ion channels in the rat forebrain following chronic amphetamine administration. Neurosci Res 2010; 68:1-8. [PMID: 20566346 DOI: 10.1016/j.neures.2010.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 05/27/2010] [Accepted: 06/01/2010] [Indexed: 12/20/2022]
Abstract
Acid-sensing ion channels (ASICs) are densely expressed in broad areas of mammalian brains and actively modulate synaptic transmission and a variety of neuronal activities. To explore whether ASICs are linked to addictive properties of drugs of abuse, we investigated the effect of the psychostimulant amphetamine on subcellular ASIC expression in the rat forebrain in vivo. Repeated administration of amphetamine (once daily for 7 days, 1.25 mg/kg for days 1/7, 4 mg/kg for days 2-6) induced typical behavioral sensitization. At a 14-day withdrawal period, ASIC1 protein levels were increased in the defined surface and intracellular compartments in the striatum (both caudate putamen and nucleus accumbens) in amphetamine-treated rats relative to saline-treated rats as detected by a surface protein cross-linking assay. ASIC2 proteins, however, remained stable in the striatum. In the medial prefrontal cortex, repeated amphetamine administration had no effect on ASIC1 expression in either the surface or the intracellular pool. However, amphetamine selectively reduced the surface expression of ASIC2 in this region. These data identify ASICs as a sensitive target to repeated stimulant exposure. The region- and compartment-specific regulation of ASIC1 and ASIC2 expression may constitute a key synaptic adaptation in reward circuits critical for psychomotor plasticity.
Collapse
Affiliation(s)
- Ajay Suman
- Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Acidosis is a common feature of the human brain during ischemic stroke and is known to cause neuronal injury. However, the mechanism underlying acidosis-mediated injury of the human brain remains elusive. We show that a decrease in the extracellular pH evoked inward currents characteristic of acid-sensing ion channels (ASICs) and increased intracellular Ca(2+) in cultured human cortical neurons. Acid-sensing ion channels in human cortical neurons show electrophysiological and pharmacological properties distinct from those in neurons of the rodent brain. Reverse transcriptase-PCR and western blot detected a high level of the ASIC1a subunit with little or no expression of other ASIC subunits. Treatment of human cortical neurons with acidic solution induced substantial cell injury, which was attenuated by the ASIC1a blockade. Thus, functional homomeric ASIC1a channels are predominantly expressed in neurons from the human brain. Activation of these channels has an important role in acidosis-mediated injury of human brain neurons.
Collapse
|
34
|
Acid-evoked Ca2+ signalling in rat sensory neurones: effects of anoxia and aglycaemia. Pflugers Arch 2010; 459:159-81. [PMID: 19806360 PMCID: PMC2765625 DOI: 10.1007/s00424-009-0715-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2008] [Revised: 07/30/2009] [Accepted: 08/14/2009] [Indexed: 12/11/2022]
Abstract
Ischaemia excites sensory neurones (generating pain) and promotes calcitonin gene-related peptide release from nerve endings. Acidosis is thought to play a key role in mediating excitation via the activation of proton-sensitive cation channels. In this study, we investigated the effects of acidosis upon Ca2+ signalling in sensory neurones from rat dorsal root ganglia. Both hypercapnic (pHo 6.8) and metabolic–hypercapnic (pHo 6.2) acidosis caused a biphasic increase in cytosolic calcium concentration ([Ca2+]i). This comprised a brief Ca2+ transient (half-time approximately 30 s) caused by Ca2+ influx followed by a sustained rise in [Ca2+]i due to Ca2+ release from caffeine and cyclopiazonic acid-sensitive internal stores. Acid-evoked Ca2+ influx was unaffected by voltage-gated Ca2+-channel inhibition with nickel and acid sensing ion channel (ASIC) inhibition with amiloride but was blocked by inhibition of transient receptor potential vanilloid receptors (TRPV1) with (E)-3-(4-t-butylphenyl)-N-(2,3-dihydrobenzo[b][1,4] dioxin-6-yl)acrylamide (AMG 9810; 1 μM) and N-(4-tertiarybutylphenyl)-4-(3-cholorphyridin-2-yl) tetrahydropryazine-1(2H)-carbox-amide (BCTC; 1 μM). Combining acidosis with anoxia and aglycaemia increased the amplitude of both phases of Ca2+ elevation and prolonged the Ca2+ transient. The Ca2+ transient evoked by combined acidosis, aglycaemia and anoxia was also substantially blocked by AMG 9810 and BCTC and, to a lesser extent, by amiloride. In summary, the principle mechanisms mediating increase in [Ca2+]i in response to acidosis are a brief Ca2+ influx through TRPV1 followed by sustained Ca2+ release from internal stores. These effects are potentiated by anoxia and aglycaemia, conditions also prevalent in ischaemia. The effects of anoxia and aglycaemia are suggested to be largely due to the inhibition of Ca2+-clearance mechanisms and possible increase in the role of ASICs.
Collapse
|
35
|
Saminathan R, Pachiappan A, Feng L, Rowan EG, Gopalakrishnakone P. Transcriptome profiling of neuronal model cell PC12 from rat pheochromocytoma. Cell Mol Neurobiol 2009; 29:533-48. [PMID: 19194798 PMCID: PMC11505829 DOI: 10.1007/s10571-009-9345-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Accepted: 01/05/2009] [Indexed: 10/21/2022]
Abstract
GeneChip microarray is a cutting-edge technology being used to study the expression patterns of genes with in a particular cell type. In this study, the Affymetrix RAE230A platform was used to profile stably expressed mRNA transcripts from PC12 cells at passage 5 and 15. The whole-cell PC12 transcriptome revealed that a total of 7,531 stable transcripts (P < 0.05), corresponding to 6,785 genes, were found to be consistently expressed between passage 5 and 15. The data analysis revealed 3,080 functional proteins, belonging to 13 families, which indicate that about 65% of the proteins expressed in PC12 cells are uncharacterized. By using our custom-built rat neuronal reference genome database, we mapped endogenously expressed stable neuronal transcripts from PC12 cells comprising about 765 genes responsible for neuronal function and disease. These neuronal transcripts were further analyzed to provide a genetic blueprint that can be used by neurobiologist to unravel the complex cellular and molecular mechanisms underlying biological functions and their associated signalling networks for diseases affecting the nervous system.
Collapse
Affiliation(s)
- Ramasamy Saminathan
- Venom and Toxin Research Programme, Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | | | | | | | | |
Collapse
|
36
|
Zhang GC, Mao LM, Wang JQ, Chu XP. Upregulation of acid-sensing ion channel 1 protein expression by chronic administration of cocaine in the mouse striatum in vivo. Neurosci Lett 2009; 459:119-22. [PMID: 19427358 DOI: 10.1016/j.neulet.2009.05.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Revised: 05/04/2009] [Accepted: 05/05/2009] [Indexed: 10/20/2022]
Abstract
Acid-sensing ion channels (ASICs) are ligand-gated cation channels activated by a drop in extracellular pH. They are enriched in the mammalian brain with a high synaptic density. Accumulating evidence suggests that ASIC1 contributes to synaptic activity related to learning/memory and fear conditioning, and also plays critical roles in neurodegenerative diseases. In this study, we explored the effect of the psychostimulant, cocaine, on protein expression of ASICs in the mouse forebrain in vivo. We found that chronic systemic injection of cocaine (20mg/kg, once daily for 5 consecutive days; 14 days of withdrawal) increased ASIC1, but not ASIC2, protein levels in the striatum, including the dorsal (caudate putamen) and the ventral (nucleus accumbens) striatum. No significant changes in ASIC1 or 2 protein levels in the median prefrontal cortex and the hippocampus were observed following the chronic cocaine administration. These data demonstrate that chronic cocaine exposure can upregulate ASIC expression in the striatum in a subunit-selective manner.
Collapse
Affiliation(s)
- Guo-Chi Zhang
- Department of Basic Medical Science, University of Missouri-Kansas City School of Medicine, 2411 Holmes Street, Kansas City, MO 64108, USA
| | | | | | | |
Collapse
|
37
|
Jiang Q, Li MH, Papasian CJ, Branigan D, Xiong ZG, Wang JQ, Chu XP. Characterization of acid-sensing ion channels in medium spiny neurons of mouse striatum. Neuroscience 2009; 162:55-66. [PMID: 19376200 DOI: 10.1016/j.neuroscience.2009.04.029] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 04/10/2009] [Accepted: 04/13/2009] [Indexed: 11/17/2022]
Abstract
Acid-sensing ion channels (ASICs) regulate synaptic activities and play important roles in neurodegenerative diseases. They are highly expressed in the striatum, where medium spiny neurons (MSNs) are a major population. Given that the properties of ASICs in MSNs are unknown, in this study, we characterized ASICs in MSNs of the mouse striatum. A rapid drop in extracellular pH induced transient inward currents in all MSNs. The pH value for half-maximal activation was 6.25, close to that obtained in homomeric ASIC1a channels. Based on psalmotoxin 1 and zinc sensitivity, ASIC1a (70.5% of neurons) and heteromeric ASIC1a-2 channels (29.5% of neurons) appeared responsible for the acid-induced currents in MSNs. ASIC currents were diminished in MSNs from ASIC1, but not ASIC2, null mice. Furthermore, a drop in pH induced calcium influx by activating homomeric ASIC1a channels. Activation of ASICs increased the membrane excitability of MSNs and lowering extracellular Ca2+ potentiated ASIC currents. Our data suggest that the homomeric ASIC1a channel represents a majority of the ASIC isoform in MSNs. The potential function of ASICs in the striatum requires further investigation.
Collapse
Affiliation(s)
- Q Jiang
- Department of Basic Medical Science, University of Missouri-Kansas City School of Medicine, 2411 Holmes Street, Kansas City, MO 64108, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Functional characterization of acid-sensing ion channels in cultured neurons of rat inferior colliculus. Neuroscience 2008; 154:461-72. [DOI: 10.1016/j.neuroscience.2008.03.040] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Revised: 03/12/2008] [Accepted: 03/13/2008] [Indexed: 11/22/2022]
|
39
|
Weng XC, Zheng JQ, Jin QE, Ma XY. Inhibition of acid-induced apoptosis by targeting ASIC1a mRNA with short hairpin RNA. Acta Pharmacol Sin 2007; 28:1621-7. [PMID: 17883949 DOI: 10.1111/j.1745-7254.2007.00627.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIM To study the role of acid-sensing ion channel (ASIC) 1a in the cell death and apoptosis induced by extracellular acid in C6 glioma cells. METHODS The stable ASIC1a-silenced C6 cell line, built with RNA interference technology, were confirmed by RT-PCR and Western blot analysis. The cell viability following acid exposure was analyzed with lactate dehydrogenase (LDH) and 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay. The apoptotic cells dyed with Annexin-V and propidium iodide were measured with a flow cytometer, while the changes of cell cycle were also assayed. RESULTS The downregulation of ASIC1a proteins by stable transfection of short hairpin RNA decreased the cell death percentage and increased cell viability following acid exposure with LDH and the MTT assay. The rate of apoptosis was lower in the ASIC1a-silenced cell line than that in the wild-type C6 cell line. The percentage of sub-G0 cells was lower in the ASIC1a-silenced C6 cells than that in the wild-type cells. CONCLUSION Extracellular acid induced cell death and apoptosis via ASIC1a mechanisms in the C6 glioma cells.
Collapse
Affiliation(s)
- Xie-Chuan Weng
- Beijing Institute of Pharmacology and Toxicology, the Academy of Military Medical Sciences, Beijing 100850, China.
| | | | | | | |
Collapse
|
40
|
Smith ESJ, Zhang X, Cadiou H, McNaughton PA. Proton binding sites involved in the activation of acid-sensing ion channel ASIC2a. Neurosci Lett 2007; 426:12-7. [PMID: 17881127 DOI: 10.1016/j.neulet.2007.07.047] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2007] [Revised: 06/21/2007] [Accepted: 07/13/2007] [Indexed: 10/23/2022]
Abstract
Most acid-sensing ion channel (ASIC) subunits are activated by protons, but ASIC2b (a splice variant of ASIC2a) is acid-insensitive. Differences in protonatable residues between the extracellular loop regions of ASIC2a and ASIC2b may explain this difference. Site-directed mutagenesis, combined with immunocytochemistry and whole-cell patch clamp, demonstrated that mutating any one of five ASIC2a sites produces channels that traffic normally to the cell surface membrane but are insensitive to protons. One of the mutants forms functional heteromers with ASIC1a and ASIC2a, demonstrating that ion transport is intact in this mutant. These five sites may be involved in the activation of ASIC2a by protons.
Collapse
Affiliation(s)
- Ewan St J Smith
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| | | | | | | |
Collapse
|
41
|
Meltzer RH, Kapoor N, Qadri YJ, Anderson SJ, Fuller CM, Benos DJ. Heteromeric Assembly of Acid-sensitive Ion Channel and Epithelial Sodium Channel Subunits. J Biol Chem 2007; 282:25548-59. [PMID: 17613525 DOI: 10.1074/jbc.m703825200] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Amiloride-sensitive ion channels are formed from homo- or heteromeric combinations of subunits from the epithelial Na+ channel (ENaC)/degenerin superfamily, which also includes the acid-sensitive ion channel (ASIC) family. These channel subunits share sequence homology and topology. In this study, we have demonstrated, using confocal fluorescence resonance energy transfer microscopy and co-immunoprecipitation, that ASIC and ENaC subunits are capable of forming cross-clade intermolecular interactions. We have also shown that combinations of ASIC1 with ENaC subunits exhibit novel electrophysiological characteristics compared with ASIC1 alone. The results of this study suggest that heteromeric complexes of ASIC and ENaC subunits may underlie the diversity of amiloride-sensitive cation conductances observed in a wide variety of tissues and cell types where co-expression of ASIC and ENaC subunits has been observed.
Collapse
Affiliation(s)
- Robert H Meltzer
- Department of Physiology and Biophysics, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | | | | | |
Collapse
|
42
|
Chai S, Li M, Lan J, Xiong ZG, Saugstad JA, Simon RP. A kinase-anchoring protein 150 and calcineurin are involved in regulation of acid-sensing ion channels ASIC1a and ASIC2a. J Biol Chem 2007; 282:22668-77. [PMID: 17548344 PMCID: PMC3799797 DOI: 10.1074/jbc.m703624200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Acid-sensing ion channel (ASIC) 1a and ASIC2a are acid-sensing ion channels in central and peripheral neurons. ASIC1a has been implicated in long-term potentiation of synaptic transmission and ischemic brain injury, whereas ASIC2a is involved in mechanosensation. Although the biological role and distribution of ASIC1a and ASIC2a subunits in brain have been well characterized, little is known about the intracellular regulation of these ion channels that modulates their function. Using pulldown assays and mass spectrometry, we have identified A kinase-anchoring protein (AKAP)150 and the protein phosphatase calcineurin as binding proteins to ASIC2a. Extended pulldown and co-immunoprecipitation assays showed that these regulatory proteins also interact with ASIC1a. Transfection of rat cortical neurons with constructs encoding green fluorescent protein- or hemagglutinin-tagged channels showed expression of ASIC1a and ASIC2a in punctate and clustering patterns in dendrites that co-localized with AKAP150. Inhibition of protein kinase A binding to AKAPs by Ht-31 peptide reduces ASIC currents in cortical neurons and Chinese hamster ovary cells, suggesting a role of AKAP150 in association with protein kinase A in ASIC function. We also demonstrated a regulatory function of calcineurin in ASIC1a and ASIC2a activity. Cyclosporin A, an inhibitor of calcineurin, increased ASIC currents in Chinese hamster ovary cells and in cortical neurons, suggesting that activity of ASICs is inhibited by calcineurin-dependent dephosphorylation. These data imply that ASIC down-regulation by calcineurin could play an important role under pathological conditions accompanying intracellular Ca(2+) overload and tissue acidosis to circumvent harmful activities mediated by these channels.
Collapse
Affiliation(s)
- Sunghee Chai
- Robert S. Dow Neurobiology Laboratories, Legacy Research, Portland, Oregon 97232, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Cho JH, Askwith CC. Potentiation of acid-sensing ion channels by sulfhydryl compounds. Am J Physiol Cell Physiol 2007; 292:C2161-74. [PMID: 17392378 DOI: 10.1152/ajpcell.00598.2006] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The acid-sensing ion channels (ASICs) are voltage-independent ion channels activated by acidic extracellular pH. ASICs play a role in sensory transduction, behavior, and acidotoxic neuronal death, which occurs during stroke and ischemia. During these conditions, the extracellular concentration of sulfhydryl reducing agents increases. We used perforated patch-clamp technique to analyze the impact of sulfhydryls on H(+)-gated currents from Chinese hamster ovary (CHO) cells expressing human ASIC1a (hASIC1a). We found that hASIC1a currents activated by pH 6.5 were increased almost twofold by the sulfhydryl-containing reducing agents dithiothreitol (DTT) and glutathione. DTT shifted the pH-dose response of hASIC1a toward a more neutral pH (pH(0.5) from 6.54 to 6.69) and slowed channel desensitization. The effect of reducing agents on native mouse hippocampal neurons and transfected mouse ASIC1a was similar. We found that the effect of DTT on hASIC1a was mimicked by the metal chelator TPEN, and mutant hASIC1a channels with reduced TPEN potentiation showed reduced DTT potentiation. Furthermore, the addition of DTT in the presence of TPEN did not result in further increases in current amplitude. These results suggest that the effect of DTT on hASIC1a is due to relief of tonic inhibition by transition metal ions. We found that all ASICs examined remained potentiated following the removal of DTT. This effect was reversed by the oxidizing agent DTNB in hASIC1a, supporting the hypothesis that DTT also impacts ASICs via a redox-sensitive site. Thus sulfhydryl compounds potentiate H(+)-gated currents via two mechanisms, metal chelation and redox modulation of target amino acids.
Collapse
Affiliation(s)
- Jun-Hyeong Cho
- Dept. of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | | |
Collapse
|
44
|
Abstract
Within the first 2 min of global brain ischemia, extracellular [K+] ([K+]o) increases above 60 mM and [Na+](o) drops to about 50 mM, indicating a massive K+ efflux and Na+ influx, a phenomenon known as anoxic depolarization (AD). Similar ionic shifts take place during repetitive peri-infarct depolarizations (PID) in the area penumbra in focal brain ischemia. The size of ischemic infarct is determined by the duration of AD and PID. However, the mechanism of cytosolic [Ca2+] ([Ca2+]c) elevation during AD or PID is poorly understood. Our data show that the exposure of cultured rat hippocampal CA1 neurons to AD-like conditions promptly elevates [Ca2+]c to about 30 microM. These high [Ca2+]c elevations depend on external Ca2+ and can be prevented by removing Na+ or by simultaneously inhibiting NMDA and AMPA/kainate receptors. These data indicate that [Ca2+]c elevations during AD result from Na+ influx via either NMDA or AMPA/kainate channels. The mechanism of the Na-dependent [Ca2+]c elevations may involve a reversal of plasmalemmal Na+/Ca2+ (NCX) and/or Na+/Ca2+ + K+ (NCKX) exchangers. KB-R7943, an NCX inhibitor, suppresses a fraction of the Na-dependent Ca2+ influx during AD. Therefore, Ca2+ influx via NCX and a KB-R7943-resistant pathway (possibly NCKX) is involved. Inhibition of the Na-dependent Ca2+ influx is likely to decrease ischemic brain damage. No drugs are known that are able to inhibit the KB-R7943-resistant component of Na-dependent Ca2+ influx during AD. The present data encourage development of such agents as potential therapeutic means to limit ischemic brain damage after stroke or heart attack.
Collapse
Affiliation(s)
- Lech Kiedrowski
- Department of Psychiatry, The Psychiatric Institute, 1601 W. Taylor St., Room 334W, Chicago, IL 60612, USA.
| |
Collapse
|
45
|
Abstract
Intracellular calcium toxicity remains the central feature in the pathophysiology of ischaemic cell death in brain. Glutamate-gated channels have been thought to be the major sites of ischaemia-induced toxic calcium entry, but the failure of glutamate antagonists in clinical trials has suggested that glutamate-independent mechanisms of calcium entry during ischaemia must exist and may prove central to ischaemic injury. We have shown that ASICs (acid-sensing ion channels) in brain are glutamate-independent vehicles of calcium flux and transport calcium in greater measure in the setting of the two major neurochemical components of ischaemia: acidosis and substrate depletion. Pharmacological blockade of ASICs markedly attenuates stroke injury with a robust therapeutic time window of 5 h following stroke onset. Here, we describe this new mechanism of calcium toxicity in brain ischaemia and offer a potential new therapy for stroke.
Collapse
Affiliation(s)
- R Simon
- Legacy Clinical Research and Technology Center, 1225 NE 2nd Avenue, Portland, OR 97232, USA.
| | | |
Collapse
|
46
|
Kiedrowski L. Critical role of sodium in cytosolic [Ca2+] elevations in cultured hippocampal CA1 neurons during anoxic depolarization. J Neurochem 2007; 100:915-23. [PMID: 17241128 DOI: 10.1111/j.1471-4159.2006.04308.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Although the extent of ischemic brain damage is directly proportional to the duration of anoxic depolarization (AD), the mechanism of cytosolic [Ca(2+)] ([Ca(2+)](c)) elevation during AD is poorly understood. To address the mechanism in this study, [Ca(2+)](c) was monitored in cultured rat hippocampal CA1 neurons loaded with a Ca-sensitive dye, fura-2FF, and exposed to an AD-simulating medium containing (in mmol/L): K(+) 65, Na(+) 50, Ca(2+) 0.13, glutamate 0.1, and pH reduced to 6.6. Application of this medium promptly elevated [Ca(2+)](c) to about 30 micromol/L, but only if oxygen was removed, the respiratory chain was inhibited, or if the mitochondria were uncoupled. These high [Ca(2+)](c) elevations depended on external Ca(2+) and could not be prevented by inhibiting NMDA or alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA)/kainate receptors, or gadolinium-sensitive channels. However, they could be prevented by removing external Na(+) or simultaneously inhibiting NMDA and AMPA/kainate receptors; 2-[2-[4-(4-nitrobenzyloxy)phenyl]ethyl]isothiourea methanesulfonate (KB-R7943), an inhibitor of plasmalemmal Na(+)/Ca(2+) exchanger, partly suppressed them. The data indicate that the [Ca(2+)](c) elevations to 30 micromol/L during AD result from Na(+) influx. Activation of either NMDA or AMPA/kainate channels provides adequate Na(+) influx to induce these [Ca(2+)](c) elevations, which are mediated by KB-R7943-sensitive and KB-R7943-resistant mechanisms.
Collapse
Affiliation(s)
- Lech Kiedrowski
- The Psychiatric Institute, Departments of Psychiatry and Pharmacology, The University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
47
|
Dynamics of desensitization and recovery of proton-activated ion channels in pheochromocytoma cells. NEUROPHYSIOLOGY+ 2007. [DOI: 10.1007/s11062-007-0002-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
48
|
Chu XP, Close N, Saugstad JA, Xiong ZG. ASIC1a-specific modulation of acid-sensing ion channels in mouse cortical neurons by redox reagents. J Neurosci 2006; 26:5329-39. [PMID: 16707785 PMCID: PMC3799800 DOI: 10.1523/jneurosci.0938-06.2006] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2005] [Revised: 04/09/2006] [Accepted: 04/10/2006] [Indexed: 11/21/2022] Open
Abstract
Acid-sensing ion channel (ASIC)-1a, the major ASIC subunit with Ca2+ permeability, is highly expressed in the neurons of CNS. Activation of these channels with resultant intracellular Ca2+ accumulation plays a critical role in normal synaptic plasticity, learning/memory, and in acidosis-mediated glutamate receptor-independent neuronal injury. Here we demonstrate that the activities of ASICs in CNS neurons are tightly regulated by the redox state of the channels and that the modulation is ASIC1a subunit dependent. In cultured mouse cortical neurons, application of the reducing agents dramatically potentiated, whereas the oxidizing agents inhibited the ASIC currents. However, in neurons from the ASIC1 knock-out mice, neither oxidizing agents nor reducing reagents had any effect on the acid-activated current. In Chinese Hamster Ovary cells, redox-modifying agents only affected the current mediated by homomeric ASIC1a, but not homomeric ASIC1b, ASIC2a, or ASIC3. In current-clamp recordings and Ca(2+)-imaging experiments, the reducing agents increased but the oxidizing agents decreased acid-induced membrane depolarization and the intracellular Ca2+ accumulation. Site-directed mutagenesis studies identified involvement of cysteine 61 and lysine 133, located in the extracellular domain of the ASIC1a subunit, in the modulation of ASICs by oxidizing and reducing agents, respectively. Our results suggest that redox state of the ASIC1a subunit is an important factor in determining the overall physiological function and the pathological role of ASICs in the CNS.
Collapse
|
49
|
Lingueglia E, Deval E, Lazdunski M. FMRFamide-gated sodium channel and ASIC channels: a new class of ionotropic receptors for FMRFamide and related peptides. Peptides 2006; 27:1138-52. [PMID: 16516345 DOI: 10.1016/j.peptides.2005.06.037] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2005] [Accepted: 06/22/2005] [Indexed: 12/13/2022]
Abstract
FMRFamide and related peptides typically exert their action through G-protein coupled receptors. However, two ionotropic receptors for these peptides have recently been identified. They are both members of the epithelial amiloride-sensitive Na+ channel and degenerin (ENaC/DEG) family of ion channels. The invertebrate FMRFamide-gated Na+ channel (FaNaC) is a neuronal Na+-selective channel which is directly gated by micromolar concentrations of FMRFamide and related tetrapeptides. Its response is fast and partially desensitizing, and FaNaC has been proposed to participate in peptidergic neurotransmission. On the other hand, mammalian acid-sensing ion channels (ASICs) are not gated but are directly modulated by FMRFamide and related mammalian peptides like NPFF and NPSF. ASICs are activated by external protons and are therefore extracellular pH sensors. They are expressed both in the central and peripheral nervous system and appear to be involved in many physiological and pathophysiological processes such as hippocampal long-term potentiation and defects in learning and memory, acquired fear-related behavior, retinal function, brain ischemia, pain sensation in ischemia and inflammation, taste perception, hearing functions, and mechanoperception. The potentiation of ASIC activity by endogenous RFamide neuropeptides probably participates in the response to noxious acidosis in sensory and central neurons. Available data also raises the possibility of the existence of still unknown FMRFamide related endogenous peptides acting as direct agonists for ASICs.
Collapse
Affiliation(s)
- Eric Lingueglia
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-Université de Nice-Sophia Antipolis UMR 6097, 660 route des Lucioles, Sophia Antipolis, 06560 Valbonne, France.
| | | | | |
Collapse
|
50
|
Xiong ZG, Chu XP, Simon RP. Ca2+-Permeable Acid-sensing Ion Channels and Ischemic Brain Injury. J Membr Biol 2006; 209:59-68. [PMID: 16685601 DOI: 10.1007/s00232-005-0840-x] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2005] [Indexed: 01/01/2023]
Abstract
Acidosis is a common feature of brain in acute neurological injury, particularly in ischemia where low pH has been assumed to play an important role in the pathological process. However, the cellular and molecular mechanisms underlying acidosis-induced injury remain unclear. Recent studies have demonstrated that activation of Ca(2+)-permeable acid-sensing ion channels (ASIC1a) is largely responsible for acidosis-mediated, glutamate receptor-independent, neuronal injury. In cultured mouse cortical neurons, lowering extracellular pH to the level commonly seen in ischemic brain activates amiloride-sensitive ASIC currents. In the majority of these neurons, ASICs are permeable to Ca(2+), and an activation of these channels induces increases in the concentration of intracellular Ca(2+) ([Ca(2+)](i)). Activation of ASICs with resultant [Ca(2+)](i) loading induces time-dependent neuronal injury occurring in the presence of the blockers for voltage-gated Ca(2+) channels and the glutamate receptors. This acid-induced injury is, however, inhibited by the blockers of ASICs, and by reducing [Ca(2+)](o). In focal ischemia, intracerebroventricular administration of ASIC1a blockers, or knockout of the ASIC1a gene protects brain from injury and does so more potently than glutamate antagonism. Furthermore, pharmacological blockade of ASICs has up to a 5 h therapeutic time window, far beyond that of glutamate antagonists. Thus, targeting the Ca(2+)-permeable acid-sensing ion channels may prove to be a novel neuroprotective strategy for stroke patients.
Collapse
Affiliation(s)
- Z-G Xiong
- Robert S. Dow Neurobiology Laboratories, Legacy Clinical Research Center, 1225 NE 2nd Ave, Portland, OR 97232, USA.
| | | | | |
Collapse
|