1
|
Shi X, Liu R, Wang Y, Yu T, Zhang K, Zhang C, Gu Y, Zhang L, Wu J, Wang Q, Zhu F. Inhibiting acid-sensing ion channel exerts neuroprotective effects in experimental epilepsy via suppressing ferroptosis. CNS Neurosci Ther 2024; 30:e14596. [PMID: 38357854 PMCID: PMC10867794 DOI: 10.1111/cns.14596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 12/16/2023] [Accepted: 12/21/2023] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Epilepsy is a chronic neurological disease characterized by repeated and unprovoked epileptic seizures. Developing disease-modifying therapies (DMTs) has become important in epilepsy studies. Notably, focusing on iron metabolism and ferroptosis might be a strategy of DMTs for epilepsy. Blocking the acid-sensing ion channel 1a (ASIC1a) has been reported to protect the brain from ischemic injury by reducing the toxicity of [Ca2+ ]i . However, whether inhibiting ASIC1a could exert neuroprotective effects and become a novel target for DMTs, such as rescuing the ferroptosis following epilepsy, remains unknown. METHODS In our study, we explored the changes in ferroptosis-related indices, including glutathione peroxidase (GPx) enzyme activity and levels of glutathione (GSH), iron accumulation, lipid degradation products-malonaldehyde (MDA) and 4-hydroxynonenal (4-HNE) by collecting peripheral blood samples from adult patients with epilepsy. Meanwhile, we observed alterations in ASIC1a protein expression and mitochondrial microstructure in the epileptogenic foci of patients with drug-resistant epilepsy. Next, we accessed the expression and function changes of ASIC1a and measured the ferroptosis-related indices in the in vitro 0-Mg2+ model of epilepsy with primary cultured neurons. Subsequently, we examined whether blocking ASIC1a could play a neuroprotective role by inhibiting ferroptosis in epileptic neurons. RESULTS Our study first reported significant changes in ferroptosis-related indices, including reduced GPx enzyme activity, decreased levels of GSH, iron accumulation, elevated MDA and 4-HNE, and representative mitochondrial crinkling in adult patients with epilepsy, especially in epileptogenic foci. Furthermore, we found that inhibiting ASIC1a could produce an inhibitory effect similar to ferroptosis inhibitor Fer-1, alleviate oxidative stress response, and decrease [Ca2+ ]i overload by inhibiting the overexpressed ASIC1a in the in vitro epilepsy model induced by 0-Mg2+ . CONCLUSION Inhibiting ASIC1a has potent neuroprotective effects via alleviating [Ca2+ ]i overload and regulating ferroptosis on the models of epilepsy and may act as a promising intervention in DMTs.
Collapse
Affiliation(s)
- Xiaorui Shi
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Ru Liu
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Advanced Innovation Center for Human Brain ProtectionCapital Medical UniversityBeijingChina
| | - Yingting Wang
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Tingting Yu
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Kai Zhang
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Chao Zhang
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Yuyu Gu
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Limin Zhang
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Jianping Wu
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Advanced Innovation Center for Human Brain ProtectionCapital Medical UniversityBeijingChina
| | - Qun Wang
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Center of Epilepsy, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Fei Zhu
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| |
Collapse
|
2
|
Castellanos LCS, Gatto RG, Malnati GOM, Montes MM, Uchitel OD, Weissmann C. Redistribution of ASIC1a channels triggered by IL-6: Potential role of ASIC1a in neuroinflammation. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166927. [PMID: 37907140 DOI: 10.1016/j.bbadis.2023.166927] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 11/02/2023]
Abstract
Cytokines, particularly IL-6, play a crucial role in modulating immune responses in the central nervous system (CNS). Elevated IL-6 levels have been observed in neuroinflammatory conditions, as well as in the sera and brains of patients with neurodegenerative diseases such as Parkinson's, Huntington's, Multiple Sclerosis, and Alzheimer's. Additionally, alterations in regional brain pH have been noted in these conditions. Acid-sensing ion channels (ASICs), including ASIC1a, activated by low pH levels, are highly abundant in the CNS and have recently been associated with various neurological disorders. Our study examined the impact of IL-6 on ASIC1a channels in cell cultures, demonstrating IL-6-induced the redistribution of cytosolic ASIC1a channels to the cell membrane. This redistribution was accompanied by increased ASIC1a current amplitude upon activation, as well as elevated levels of phosphorylated CaMKII and ERK kinases. Additionally, we observed posttranslational modifications on the ASIC1a channel itself. These findings provide insight into a potential link between inflammatory processes and neurodegenerative mechanisms, highlighting ASIC1a channels as promising therapeutic targets in these conditions.
Collapse
Affiliation(s)
| | - Rodolfo Gabriel Gatto
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
| | | | - Mayra Micaela Montes
- Instituto de Fisiología Biología Molecular y Neurociencias-IFIBYNE-UBA-CONICET, LFBM, Argentina
| | - Osvaldo Daniel Uchitel
- Instituto de Fisiología Biología Molecular y Neurociencias-IFIBYNE-UBA-CONICET, LFBM, Argentina
| | - Carina Weissmann
- Instituto de Fisiología Biología Molecular y Neurociencias-IFIBYNE-UBA-CONICET, LFBM, Argentina.
| |
Collapse
|
3
|
Foster VS, Saez N, King GF, Rank MM. Acute inhibition of acid sensing ion channel 1a after spinal cord injury selectively affects excitatory synaptic transmission, but not intrinsic membrane properties, in deep dorsal horn interneurons. PLoS One 2023; 18:e0289053. [PMID: 37939057 PMCID: PMC10631665 DOI: 10.1371/journal.pone.0289053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/10/2023] [Indexed: 11/10/2023] Open
Abstract
Following a spinal cord injury (SCI), secondary damage mechanisms are triggered that cause inflammation and cell death. A key component of this secondary damage is a reduction in local blood flow that initiates a well-characterised ischemic cascade. Downstream hypoxia and acidosis activate acid sensing ion channel 1a (ASIC1a) to trigger cell death. We recently showed that administration of a potent venom-derived inhibitor of ASIC1a, Hi1a, leads to tissue sparing and improved functional recovery when delivered up to 8 h after ischemic stroke. Here, we use whole-cell patch-clamp electrophysiology in a spinal cord slice preparation to assess the effect of acute ASIC1a inhibition, via a single dose of Hi1a, on intrinsic membrane properties and excitatory synaptic transmission long-term after a spinal cord hemisection injury. We focus on a population of interneurons (INs) in the deep dorsal horn (DDH) that play a key role in relaying sensory information to downstream motoneurons. DDH INs in mice treated with Hi1a 1 h after a spinal cord hemisection showed no change in active or passive intrinsic membrane properties measured 4 weeks after SCI. DDH INs, however, exhibit significant changes in the kinetics of spontaneous excitatory postsynaptic currents after a single dose of Hi1a, when compared to naive animals (unlike SCI mice). Our data suggest that acute ASIC1a inhibition exerts selective effects on excitatory synaptic transmission in DDH INs after SCI via specific ligand-gated receptor channels, and has no effect on other voltage-activated channels long-term after SCI.
Collapse
Affiliation(s)
- Victoria S. Foster
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
- St George’s, University of London, Medical School, London, England
| | - Natalie Saez
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St Lucia, Queensland, Australia
| | - Glenn F. King
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St Lucia, Queensland, Australia
| | - Michelle M. Rank
- Department of Anatomy and Physiology, School of Biomedical Science, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
4
|
Liu H, Lu WL, Hong HQ, Li MJ, Ye MP, Rao QF, Kong JL, Luan SH, Huang Y, Hu QH, Wu FR. CaM/CaMKII mediates activation and proliferation of hepatic stellate cells regulated by ASIC1a. Front Pharmacol 2022; 13:996667. [PMID: 36588718 PMCID: PMC9797583 DOI: 10.3389/fphar.2022.996667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
The activation of hepatic stellate cells (HSCs) is closely related to hepatic fibrosis and plays a key role in its occurrence and development. In the damaged liver, inhibition of the activation, proliferation, and clearance of HSCs is an important therapeutic strategy. However, the mechanism underlying the activation of HSCs is not completely clear. Acid-sensitive ion channel 1a (ASIC1a) is a cation channel activated by extracellular acid, which is responsible for the transport of Ca2+ and Na+ and participates in the activation of HSCs and the occurrence and development of many inflammatory diseases, suggesting that ASIC1a plays an important role in liver fibrosis. A previous study by the project team found that when the membrane channel protein ASIC1a was opened, intracellular Ca2+ levels increased, the expression of CaM/CaMKII in HSCs was high, and HSC was activated and proliferated. Therefore, we established an SD rat model of hepatic fibrosis and induced HSC-T6 activation by stimulating ASIC1a with acid in vitro. In vivo, CCl4 was used to induce liver fibrosis in rats, and different doses of KN93 (0.5, 1, and 2 mg/kg/d) and colchicine (0.1 mg/kg/d) were administered. Eight weeks later, the activities of ALT and AST in serum were measured and hematoxylin-eosin and Masson staining in liver tissue, and immunohistochemistry analysis were performed in SD rats. The expressions of ASIC1a, α-SMA, Collagen-1, CaM, and CaMKII were detected. In vitro, we activated HSC-T6 cells by stimulating ASIC1a with acid. The results showed that inhibition of ASIC1a could improve acid-induced HSCs activation. In addition, CaM/CaMKII was expressed in HSC of rats with hepatic fibrosis regulated by ASIC1a. After blocking or silencing the expression of CaMKII, the fibrosis marker protein can be down-regulated. KN93 also reduced inflammation and improved the activation, proliferation and fibrosis of HSC. In summary, we concluded that CaM/CaMKII participates in ASIC1a regulation of the proliferation and activation of HSC and promotes the occurrence of liver fibrosis.
Collapse
Affiliation(s)
- Hui Liu
- Institute for Liver Diseases of Anhui Medical University, Hefei, China,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Wei-Li Lu
- Institute for Liver Diseases of Anhui Medical University, Hefei, China,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Hai-Qin Hong
- Institute for Liver Diseases of Anhui Medical University, Hefei, China,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Meng-Jun Li
- Institute for Liver Diseases of Anhui Medical University, Hefei, China,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Man-Ping Ye
- Institute for Liver Diseases of Anhui Medical University, Hefei, China,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Qiu-Fan Rao
- Institute for Liver Diseases of Anhui Medical University, Hefei, China,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Jin-Ling Kong
- Institute for Liver Diseases of Anhui Medical University, Hefei, China,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Shao-Hua Luan
- Institute for Liver Diseases of Anhui Medical University, Hefei, China,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Yan Huang
- Institute for Liver Diseases of Anhui Medical University, Hefei, China,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Qing-Hua Hu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China,*Correspondence: Qing-Hua Hu, ; Fan-Rong Wu,
| | - Fan-Rong Wu
- Institute for Liver Diseases of Anhui Medical University, Hefei, China,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China,*Correspondence: Qing-Hua Hu, ; Fan-Rong Wu,
| |
Collapse
|
5
|
Verkest C, Salinas M, Diochot S, Deval E, Lingueglia E, Baron A. Mechanisms of Action of the Peptide Toxins Targeting Human and Rodent Acid-Sensing Ion Channels and Relevance to Their In Vivo Analgesic Effects. Toxins (Basel) 2022; 14:toxins14100709. [PMID: 36287977 PMCID: PMC9612379 DOI: 10.3390/toxins14100709] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/30/2022] [Accepted: 10/02/2022] [Indexed: 11/16/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are voltage-independent H+-gated cation channels largely expressed in the nervous system of rodents and humans. At least six isoforms (ASIC1a, 1b, 2a, 2b, 3 and 4) associate into homotrimers or heterotrimers to form functional channels with highly pH-dependent gating properties. This review provides an update on the pharmacological profiles of animal peptide toxins targeting ASICs, including PcTx1 from tarantula and related spider toxins, APETx2 and APETx-like peptides from sea anemone, and mambalgin from snake, as well as the dimeric protein snake toxin MitTx that have all been instrumental to understanding the structure and the pH-dependent gating of rodent and human cloned ASICs and to study the physiological and pathological roles of native ASICs in vitro and in vivo. ASICs are expressed all along the pain pathways and the pharmacological data clearly support a role for these channels in pain. ASIC-targeting peptide toxins interfere with ASIC gating by complex and pH-dependent mechanisms sometimes leading to opposite effects. However, these dual pH-dependent effects of ASIC-inhibiting toxins (PcTx1, mambalgin and APETx2) are fully compatible with, and even support, their analgesic effects in vivo, both in the central and the peripheral nervous system, as well as potential effects in humans.
Collapse
Affiliation(s)
- Clément Verkest
- CNRS (Centre National de la Recherche Scientifique), IPMC (Institut de Pharmacologie Moléculaire et Cellulaire), LabEx ICST (Laboratory of Excellence in Ion Channel Science and Therapeutics), FHU InovPain (Fédération Hospitalo-Universitaire “Innovative Solutions in Refractory Chronic Pain”), Université Côte d’Azur, 660 Route des Lucioles, Sophia-Antipolis, 06560 Nice, France
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Miguel Salinas
- CNRS (Centre National de la Recherche Scientifique), IPMC (Institut de Pharmacologie Moléculaire et Cellulaire), LabEx ICST (Laboratory of Excellence in Ion Channel Science and Therapeutics), FHU InovPain (Fédération Hospitalo-Universitaire “Innovative Solutions in Refractory Chronic Pain”), Université Côte d’Azur, 660 Route des Lucioles, Sophia-Antipolis, 06560 Nice, France
| | - Sylvie Diochot
- CNRS (Centre National de la Recherche Scientifique), IPMC (Institut de Pharmacologie Moléculaire et Cellulaire), LabEx ICST (Laboratory of Excellence in Ion Channel Science and Therapeutics), FHU InovPain (Fédération Hospitalo-Universitaire “Innovative Solutions in Refractory Chronic Pain”), Université Côte d’Azur, 660 Route des Lucioles, Sophia-Antipolis, 06560 Nice, France
| | - Emmanuel Deval
- CNRS (Centre National de la Recherche Scientifique), IPMC (Institut de Pharmacologie Moléculaire et Cellulaire), LabEx ICST (Laboratory of Excellence in Ion Channel Science and Therapeutics), FHU InovPain (Fédération Hospitalo-Universitaire “Innovative Solutions in Refractory Chronic Pain”), Université Côte d’Azur, 660 Route des Lucioles, Sophia-Antipolis, 06560 Nice, France
| | - Eric Lingueglia
- CNRS (Centre National de la Recherche Scientifique), IPMC (Institut de Pharmacologie Moléculaire et Cellulaire), LabEx ICST (Laboratory of Excellence in Ion Channel Science and Therapeutics), FHU InovPain (Fédération Hospitalo-Universitaire “Innovative Solutions in Refractory Chronic Pain”), Université Côte d’Azur, 660 Route des Lucioles, Sophia-Antipolis, 06560 Nice, France
| | - Anne Baron
- CNRS (Centre National de la Recherche Scientifique), IPMC (Institut de Pharmacologie Moléculaire et Cellulaire), LabEx ICST (Laboratory of Excellence in Ion Channel Science and Therapeutics), FHU InovPain (Fédération Hospitalo-Universitaire “Innovative Solutions in Refractory Chronic Pain”), Université Côte d’Azur, 660 Route des Lucioles, Sophia-Antipolis, 06560 Nice, France
- Correspondence:
| |
Collapse
|
6
|
Retinoic Acid-Differentiated Neuroblastoma SH-SY5Y Is an Accessible In Vitro Model to Study Native Human Acid-Sensing Ion Channels 1a (ASIC1a). BIOLOGY 2022; 11:biology11020167. [PMID: 35205034 PMCID: PMC8868828 DOI: 10.3390/biology11020167] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 12/19/2022]
Abstract
Simple Summary Human neuroblastoma SH-SY5Y is used in neurobiology for studying various neuropathophysiological processes. In this study, we differentiated neuroblastoma cells into a neuronal-like phenotype with retinoic acid and studied if functional acid-sensing, transient receptor potential vanilloid-1 and ankyrin-1 ion channels were expressed in it. We found that homomeric acid-sensing ion channels 1a were expressed predominantly and yielded large ionic currents that can be modulated with different ligands. This channel plays important roles in synaptic plasticity, neurodegeneration, and pain perception. Thus, retinoic acid-treated neuroblastoma is a suitable model system for pharmacological testing on native human acid-sensing ion channels 1a. This approach can facilitate the development of new drugs for neuroprotection and pain management. Abstract Human neuroblastoma SH-SY5Y is a prominent neurobiological tool used for studying neuropathophysiological processes. We investigated acid-sensing (ASIC) and transient receptor potential vanilloid-1 (TRPV1) and ankyrin-1 (TRPA1) ion channels present in untreated and differentiated neuroblastoma SH-SY5Y to propose a new means for their study in neuronal-like cells. Using a quantitative real-time PCR and a whole-cell patch-clamp technique, ion channel expression profiles, functionality, and the pharmacological actions of their ligands were characterized. A low-level expression of ASIC1a and ASIC2 was detected in untreated cells. The treatment with 10 μM of retinoic acid (RA) for 6 days resulted in neuronal differentiation that was accompanied by a remarkable increase in ASIC1a expression, while ASIC2 expression remained almost unaltered. In response to acid stimuli, differentiated cells showed prominent ASIC-like currents. Detailed kinetic and pharmacological characterization suggests that homomeric ASIC1a is a dominant isoform among the present ASIC channels. RA-treatment also reduced the expression of TRPV1 and TRPA1, and minor electrophysiological responses to their agonists were found in untreated cells. Neuroblastoma SH-SY5Y treated with RA can serve as a model system to study the effects of different ligands on native human ASIC1a in neuronal-like cells. This approach can improve the characterization of modulators for the development of new neuroprotective and analgesic drugs.
Collapse
|
7
|
William M, Singh S, Chu XP. Commentary: Large Acid-Evoked Currents, Mediated by ASIC1a, Accompany Differentiation in Human Dopaminergic Neurons. Front Cell Neurosci 2021; 15:789354. [PMID: 34880731 PMCID: PMC8646021 DOI: 10.3389/fncel.2021.789354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 10/29/2021] [Indexed: 11/18/2022] Open
Affiliation(s)
- Matthew William
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Som Singh
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Xiang-Ping Chu
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| |
Collapse
|
8
|
Salinas Castellanos LC, Uchitel OD, Weissmann C. Signaling Pathways in Proton and Non-proton ASIC1a Activation. Front Cell Neurosci 2021; 15:735414. [PMID: 34675777 PMCID: PMC8523820 DOI: 10.3389/fncel.2021.735414] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/09/2021] [Indexed: 11/13/2022] Open
Abstract
Acid-sensing ion channels (ASICs) regulate synaptic activities and play important roles in neurodegenerative diseases as well as pain conditions. Classically, ASICs are described as transiently activated by a reduced pH, followed by desensitization; the activation allows sodium influx, and in the case of ASIC1a-composed channels, also calcium to some degree. Several factors are emerging and extensively analyzed as modulators, activating, inhibiting, and potentiating specific channel subunits. However, the signaling pathways triggered by channel activation are only starting to be revealed.The channel has been recently shown to be activated through a mechanism other than proton-mediated. Indeed, the large extracellular loop of these channels opens the possibility that other non-proton ligands might exist. One such molecule discovered was a toxin present in the Texas coral snake venom. The finding was associated with the activation of the channel at neutral pH via the toxin and causing intense and unremitting pain.By using different pharmacological tools, we analyzed the downstream signaling pathway triggered either by the proton and non-proton activation for human, mouse, and rat ASIC1a-composed channels in in vitro models. We show that for all species analyzed, the non-protonic mode of activation determines the activation of the ERK signaling cascade at a higher level and duration compared to the proton mode.This study adds to the growing evidence of the important role ASIC1a channels play in different physiological and pathological conditions and also hints at a possible pathological mechanism for a sustained effect.
Collapse
Affiliation(s)
| | | | - Carina Weissmann
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE—UBA CONICET), Facultad de Ciencias, Exactas y Naturales de la Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
9
|
Wu BM, Bargaineer J, Zhang L, Yang T, Xiong ZG, Leng TD. Upregulation of acid sensing ion channel 1a (ASIC1a) by hydrogen peroxide through the JNK pathway. Acta Pharmacol Sin 2021; 42:1248-1255. [PMID: 33184449 PMCID: PMC8285496 DOI: 10.1038/s41401-020-00559-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/18/2020] [Indexed: 02/02/2023] Open
Abstract
Oxidative stress is intimately tied to neurodegenerative diseases, including Parkinson's disease and amyotrophic lateral sclerosis, and acute injuries, such as ischemic stroke and traumatic brain injury. Acid sensing ion channel 1a (ASIC1a), a proton-gated ion channel, has been shown to be involved in the pathogenesis of these diseases. However, whether oxidative stress affects the expression of ASIC1a remains elusive. In the current study, we examined the effect of hydrogen peroxide (H2O2), a major reactive oxygen species (ROS), on ASIC1a protein expression and channel function in NS20Y cells and primary cultured mouse cortical neurons. We found that treatment of the cells with H2O2 (20 µM) for 6 h or longer increased ASIC1a protein expression and ASIC currents without causing significant cell injury. H2O2 incubation activated mitogen-activated protein kinases (MAPKs) pathways, including the extracellular signal-regulated kinase1/2 (ERK1/2), c-Jun N-terminal kinase (JNK), and p38 pathways. We found that neither inhibition of the MEK/ERK pathway by U0126 nor inhibition of the p38 pathway by SB203580 affected H2O2-induced ASIC1a expression, whereas inhibition of the JNK pathway by SP600125 potently decreased ASIC1a expression and abolished the H2O2-mediated increase in ASIC1a expression and ASIC currents. Furthermore, we found that H2O2 pretreatment increased the sensitivity of ASIC currents to the ASIC1a inhibitor PcTx1, providing additional evidence that H2O2 increases the expression of functional ASIC1a channels. Together, our data demonstrate that H2O2 increases ASIC1a expression/activation through the JNK signaling pathway, which may provide insight into the pathogenesis of neurological disorders that involve both ROS and activation of ASIC1a.
Collapse
Affiliation(s)
- Bao-Ming Wu
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Jaree Bargaineer
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Ling Zhang
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Tao Yang
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Zhi-Gang Xiong
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, 30310, USA.
| | - Tian-Dong Leng
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, 30310, USA.
| |
Collapse
|
10
|
Zhou G, Zha XM. GPR68 Contributes to Persistent Acidosis-Induced Activation of AGC Kinases and Tyrosine Phosphorylation in Organotypic Hippocampal Slices. Front Neurosci 2021; 15:692217. [PMID: 34113235 PMCID: PMC8185064 DOI: 10.3389/fnins.2021.692217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 04/29/2021] [Indexed: 12/28/2022] Open
Abstract
Persistent acidosis occurs in ischemia and multiple neurological diseases. In previous studies, acidic stimulation leads to rapid increase in intracellular calcium in neurons. However, it remains largely unclear how a prolonged acidosis alters neuronal signaling. In our previous study, we found that GPR68-mediated PKC activities are protective against acidosis-induced injury in cortical slices. Here, we first asked whether the same principle holds true in organotypic hippocampal slices. Our data showed that 1-h pH 6 induced PKC phosphorylation in a GPR68-dependent manner. Go6983, a PKC inhibitor worsened acidosis-induced neuronal injury in wild type (WT) but had no effect in GPR68−/− slices. Next, to gain greater insights into acid signaling in brain tissue, we treated organotypic hippocampal slices with pH 6 for 1-h and performed a kinome profiling analysis by Western blot. Acidosis had little effect on cyclin-dependent kinase (CDK) or casein kinase 2 activity, two members of the CMGC family, or Ataxia telangiectasia mutated (ATM)/ATM and RAD3-related (ATR) activity, but reduced the phosphorylation of MAPK/CDK substrates. In contrast, acidosis induced the activation of CaMKIIα, PKA, and Akt. Besides these serine/threonine kinases, acidosis also induced tyrosine phosphorylation. Since GPR68 is widely expressed in brain neurons, we asked whether GPR68 contributes to acidosis-induced signaling. Deleting GPR68 had no effect on acidosis-induced CaMKII phosphorylation, attenuated that of phospho-Akt and phospho-PKA substrates, while abolishing acidosis-induced tyrosine phosphorylation. These data demonstrate that prolonged acidosis activates a network of signaling cascades, mediated by AGC kinases, CaMKII, and tyrosine kinases. GPR68 is the primary mediator for acidosis-induced activation of PKC and tyrosine phosphorylation, while both GPR68-dependent and -independent mechanisms contribute to the activation of PKA and Akt.
Collapse
Affiliation(s)
- Guokun Zhou
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL, United States
| | - Xiang-Ming Zha
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL, United States
| |
Collapse
|
11
|
Neuhof A, Tian Y, Reska A, Falkenburger BH, Gründer S. Large Acid-Evoked Currents, Mediated by ASIC1a, Accompany Differentiation in Human Dopaminergic Neurons. Front Cell Neurosci 2021; 15:668008. [PMID: 33986647 PMCID: PMC8110905 DOI: 10.3389/fncel.2021.668008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/06/2021] [Indexed: 12/21/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are proton-gated Na+ channels. They contribute to synaptic transmission, neuronal differentiation and neurodegeneration. ASICs have been mainly characterized in neurons from mice or rats and our knowledge of their properties in human neurons is scarce. Here, we functionally characterized ASICs in differentiating LUHMES cells, a human mesencephalic cell line with characteristics of dopaminergic neurons. We find that LUHMES cells express functional ASICs, predominantly homomeric ASIC1a. Expression starts early during differentiation with a striking surge in current amplitude at days 4-6 of differentiation, a time point where-based on published data-LUHMES cells start expressing synaptic markers. Peak ASIC expression therefore coincides with a critical period of LUHMES cell differentiation. It was associated with increased excitability, but not paralleled by an increase in ASIC1 mRNA or protein. In differentiating as well as in terminally differentiated LUHMES cells, ASIC activation by slight acidification elicited large currents, action potentials and a rise in cytosolic Ca2+. Applying the ASIC pore blocker diminazene during differentiation reduced the length of neurites, consistent with the hypothesis that ASICs play a critical role in LUHMES cell differentiation. In summary, our study establishes LUHMES cells as a valuable model to study the role of ASICs for neuronal differentiation and potentially also cell death in a human cell line.
Collapse
Affiliation(s)
- Andreas Neuhof
- Department of Neurology, Institute of Physiology, RWTH Aachen University, Aachen, Germany.,Department of Neurology, RWTH Aachen University, Aachen, Germany
| | - Yuemin Tian
- Department of Neurology, Institute of Physiology, RWTH Aachen University, Aachen, Germany
| | - Anna Reska
- Department of Neurology, Institute of Physiology, RWTH Aachen University, Aachen, Germany
| | | | - Stefan Gründer
- Department of Neurology, Institute of Physiology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
12
|
Zhou G, Wang T, Zha XM. RNA-Seq analysis of knocking out the neuroprotective proton-sensitive GPR68 on basal and acute ischemia-induced transcriptome changes and signaling in mouse brain. FASEB J 2021; 35:e21461. [PMID: 33724568 PMCID: PMC7970445 DOI: 10.1096/fj.202002511r] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/14/2021] [Accepted: 02/04/2021] [Indexed: 12/17/2022]
Abstract
Brain acid signaling plays important roles in both physiological and disease conditions. One key neuronal metabotropic proton receptor in the brain is GPR68, which contributes to hippocampal long-term potentiation (LTP) and mediates neuroprotection in acidotic and ischemic conditions. Here, to gain greater understanding of GPR68 function in the brain, we performed mRNA-Seq analysis in mice. First, we studied sham-operated animals to determine baseline expression. Compared to wild type (WT), GPR68-/- (KO) brain downregulated genes that are enriched in Gene Ontology (GO) terms of misfolding protein binding, response to organic cyclic compounds, and endoplasmic reticulum chaperone complex. Next, we examined the expression profile following transient middle cerebral artery occlusion (tMCAO). tMCAO-upregulated genes cluster to cytokine/chemokine-related functions and immune responses, while tMCAO-downregulated genes cluster to channel activities and synaptic signaling. For proton-sensitive receptors, tMCAO downregulated ASIC1a and upregulated GPR4 and GPR65, but had no effect on ASIC2, PAC, or GPR68. GPR68 deletion did not alter the expression of these proton receptors, either at baseline or after ischemia. Lastly, we performed GeneVenn analysis of differential genes at baseline and post-tMCAO. Ischemia upregulated the expression of three hemoglobin genes, along with H2-Aa, Ppbp, Siglece, and Tagln, in WT but not in KO. Immunostaining showed that tMCAO-induced hemoglobin localized to neurons. Western blot analysis further showed that hemoglobin induction is GPR68-dependent. Together, these data suggest that GPR68 deletion at baseline disrupts chaperone functions and cellular signaling responses and imply a contribution of hemoglobin-mediated antioxidant mechanism to GPR68-dependent neuroprotection in ischemia.
Collapse
Affiliation(s)
- Guokun Zhou
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL, USA
| | - Tao Wang
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL, USA
| | - Xiang-Ming Zha
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL, USA
| |
Collapse
|
13
|
Song XL, Liu DS, Qiang M, Li Q, Liu MG, Li WG, Qi X, Xu NJ, Yang G, Zhu MX, Xu TL. Postsynaptic Targeting and Mobility of Membrane Surface-Localized hASIC1a. Neurosci Bull 2021; 37:145-165. [PMID: 32996060 PMCID: PMC7870742 DOI: 10.1007/s12264-020-00581-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/14/2020] [Indexed: 01/19/2023] Open
Abstract
Acid-sensing ion channels (ASICs), the main H+ receptors in the central nervous system, sense extracellular pH fluctuations and mediate cation influx. ASIC1a, the major subunit responsible for acid-activated current, is widely expressed in brain neurons, where it plays pivotal roles in diverse functions including synaptic transmission and plasticity. However, the underlying molecular mechanisms for these functions remain mysterious. Using extracellular epitope tagging and a novel antibody recognizing the hASIC1a ectodomain, we examined the membrane targeting and dynamic trafficking of hASIC1a in cultured cortical neurons. Surface hASIC1a was distributed throughout somata and dendrites, clustered in spine heads, and co-localized with postsynaptic markers. By extracellular pHluorin tagging and fluorescence recovery after photobleaching, we detected movement of hASIC1a in synaptic spine heads. Single-particle tracking along with use of the anti-hASIC1a ectodomain antibody revealed long-distance migration and local movement of surface hASIC1a puncta on dendrites. Importantly, enhancing synaptic activity with brain-derived neurotrophic factor accelerated the trafficking and lateral mobility of hASIC1a. With this newly-developed toolbox, our data demonstrate the synaptic location and high dynamics of functionally-relevant hASIC1a on the surface of excitatory synapses, supporting its involvement in synaptic functions.
Collapse
Affiliation(s)
- Xing-Lei Song
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Di-Shi Liu
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Min Qiang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Qian Li
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China
| | - Ming-Gang Liu
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China
| | - Wei-Guang Li
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China
| | - Xin Qi
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Nan-Jie Xu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China
| | - Guang Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Michael Xi Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Tian-Le Xu
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China.
| |
Collapse
|
14
|
Xu Y, Chen F. Acid-Sensing Ion Channel-1a in Articular Chondrocytes and Synovial Fibroblasts: A Novel Therapeutic Target for Rheumatoid Arthritis. Front Immunol 2021; 11:580936. [PMID: 33584647 PMCID: PMC7876322 DOI: 10.3389/fimmu.2020.580936] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 12/14/2020] [Indexed: 12/19/2022] Open
Abstract
Acid-sensing ion channel 1a (ASIC1a) is a member of the extracellular H+-activated cation channel family. Emerging evidence has suggested that ASIC1a plays a crucial role in the pathogenesis of rheumatoid arthritis (RA). Specifically, ASIC1a could promote inflammation, synovial hyperplasia, articular cartilage, and bone destruction; these lead to the progression of RA, a chronic autoimmune disease characterized by chronic synovial inflammation and extra-articular lesions. In this review, we provided a brief overview of the molecular properties of ASIC1a, including the basic biological characteristics, tissue and cell distribution, channel blocker, and factors influencing the expression and function, and focused on the potential therapeutic targets of ASIC1a in RA and possible mechanisms of blocking ASIC1a to improve RA symptoms, such as regulation of apoptosis, autophagy, pyroptosis, and necroptosis of articular cartilage, and synovial inflammation and invasion of fibroblast-like cells in synovial tissue.
Collapse
Affiliation(s)
- Yayun Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Feihu Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| |
Collapse
|
15
|
Wang T, He M, Zha XM. Time-dependent progression of hemorrhagic transformation after transient ischemia and its association with GPR68-dependent protection. BRAIN HEMORRHAGES 2020; 1:185-191. [PMID: 33575546 DOI: 10.1016/j.hest.2020.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hemorrhagic transformation (HT) following ischemia is one complication which worsens stroke outcome. During and after ischemia-reperfusion, persistent reduction of brain pH occurs. In a recent study, we found that GPR68 functions as a neuronal proton receptor and mediates a protective pathway in brain ischemia. Here, we asked whether GPR68 contributes HT after ischemia. At 24 hr after transient middle cerebral artery occlusion (tMCAO), 58% of the wild-type (WT) mice exhibited some degrees of mild HT. At 72 hr, 95% of the WT showed HT with 42% exhibited large "parenchymal" type hemorrhage. In the GPR68-/- mice, there was a trend of increase in both the incidence and severity of HT at both time points. Mice with severe hemorrhage exhibited significantly larger infarct than those with no to mild hemorrhage. Next, we compared % infarct of GPR68-/- vs WT based on their HT categories. GPR68 deletion increased % infarct when the HT severity is mild. In contrast, for mice exhibiting large area HT, the two genotypes had no difference in % infarct. These data showed that GPR68-dependent signaling leads to protection when HT is mild.
Collapse
Affiliation(s)
- Tao Wang
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL, USA
| | - Mindi He
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL, USA
| | - Xiang-Ming Zha
- Department of Physiology and Cell Biology, University of South Alabama College of Medicine, Mobile, AL, USA
| |
Collapse
|
16
|
Wang T, Zhou G, He M, Xu Y, Rusyniak WG, Xu Y, Ji Y, Simon RP, Xiong ZG, Zha XM. GPR68 Is a Neuroprotective Proton Receptor in Brain Ischemia. Stroke 2020; 51:3690-3700. [PMID: 33059544 PMCID: PMC7678672 DOI: 10.1161/strokeaha.120.031479] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Supplemental Digital Content is available in the text. Brain acidosis is prevalent in stroke and other neurological diseases. Acidosis can have paradoxical injurious and protective effects. The purpose of this study is to determine whether a proton receptor exists in neurons to counteract acidosis-induced injury.
Collapse
Affiliation(s)
- Tao Wang
- Department of Physiology and Cell Biology (T.W., G.Z., M.H., Yuanyuan Xu, X.-m.Z.), University of South Alabama College of Medicine, Mobile
| | - Guokun Zhou
- Department of Physiology and Cell Biology (T.W., G.Z., M.H., Yuanyuan Xu, X.-m.Z.), University of South Alabama College of Medicine, Mobile.,Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, China (G.Z., Y.J.)
| | - Mindi He
- Department of Physiology and Cell Biology (T.W., G.Z., M.H., Yuanyuan Xu, X.-m.Z.), University of South Alabama College of Medicine, Mobile
| | - Yuanyuan Xu
- Department of Physiology and Cell Biology (T.W., G.Z., M.H., Yuanyuan Xu, X.-m.Z.), University of South Alabama College of Medicine, Mobile
| | - W G Rusyniak
- Department of Neurosurgery (W.G.R.), University of South Alabama College of Medicine, Mobile
| | - Yan Xu
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis (Yan Xu)
| | - Yonghua Ji
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, China (G.Z., Y.J.)
| | - Roger P Simon
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA (R.P.S., Z.-G.X.)
| | - Zhi-Gang Xiong
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA (R.P.S., Z.-G.X.)
| | - Xiang-Ming Zha
- Department of Physiology and Cell Biology (T.W., G.Z., M.H., Yuanyuan Xu, X.-m.Z.), University of South Alabama College of Medicine, Mobile
| |
Collapse
|
17
|
Zhang Y, Qian X, Yang X, Niu R, Song S, Zhu F, Zhu C, Peng X, Chen F. ASIC1a induces synovial inflammation via the Ca 2+/NFATc3/ RANTES pathway. Theranostics 2020; 10:247-264. [PMID: 31903118 PMCID: PMC6929608 DOI: 10.7150/thno.37200] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 09/09/2019] [Indexed: 12/11/2022] Open
Abstract
Rationale: Synovial inflammation is one of the main pathological features of rheumatoid arthritis (RA) and is a key factor leading to the progression of RA. Understanding the regulatory mechanism of synovial inflammation is crucial for the treatment of RA. Acid-sensing ion channel 1a (ASIC1a) is an H+-gated cation channel that promotes the progression of RA, but the role of ASIC1a in synovial inflammation is unclear. This study aimed to investigate whether ASIC1a is involved in the synovial inflammation and explore the underlying mechanisms in vitro and in vivo. Methods: The expression of ASIC1a and nuclear factor of activated T cells (NFATs) were analyzed by Western blotting, immunofluorescence, and immunohistochemistry both in vitro and in vivo. The Ca2+ influx mediated by ASIC1a was detected by calcium imaging and flow cytometry. The role of ASIC1a in inflammation was studied in rats with adjuvant-induced arthritis (AA). Inflammatory cytokine profile was analyzed by protein chip in RA synovial fibroblasts (RASF) and verified by a magnetic multi-cytokine assay and ELISA. The NFATc3-regulated RANTES (Regulated upon activation, normal T cell expressed and secreted) gene transcription was investigated by ChIP-qPCR and dual-luciferase reporter assay. Results: The expression of ASIC1a was significantly increased in human RA synovial tissues and primary human RASF as well as in ankle synovium of AA rats. Activated ASIC1a mediated Ca2+ influx to increase [Ca2+]i in RASF. The activation/overexpression of ASIC1a in RASF up-regulated the expression of inflammatory cytokines RANTES, sTNF RI, MIP-1a, IL-8, sTNF RII, and ICAM-1 among which RANTES was increased most remarkably. In vivo, ASIC1a promoted inflammation, synovial hyperplasia, articular cartilage, and bone destruction, leading to the progression of AA. Furthermore, activation of ASIC1a upregulated the nuclear translocation of NFATc3, which bound to RANTES promoter and directly regulated gene transcription to enhance RANTES expression. Conclusion: ASIC1a induces synovial inflammation, which leads to the progression of RA. Our study reveals a novel RA inflammation regulatory mechanism and indicates that ASIC1a might be a potential therapeutic target for RA.
Collapse
Affiliation(s)
- Yihao Zhang
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Xuewen Qian
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Xiaojuan Yang
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Ruowen Niu
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Sujing Song
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Fei Zhu
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Chuanjun Zhu
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Xiaoqing Peng
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Feihu Chen
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| |
Collapse
|
18
|
Blockade of Acid-Sensing Ion Channels Attenuates Recurrent Hypoglycemia-Induced Potentiation of Ischemic Brain Damage in Treated Diabetic Rats. Neuromolecular Med 2019; 21:454-466. [PMID: 31134484 DOI: 10.1007/s12017-019-08546-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/17/2019] [Indexed: 12/18/2022]
Abstract
Diabetes is a chronic metabolic disease and cerebral ischemia is a serious complication of diabetes. Anti-diabetic therapy mitigates this complication but increases the risk of exposure to recurrent hypoglycemia (RH). We showed previously that RH exposure increases ischemic brain damage in insulin-treated diabetic (ITD) rats. The present study evaluated the hypothesis that increased intra-ischemic acidosis in RH-exposed ITD rats leads to pronounced post-ischemic hypoperfusion via activation of acid-sensing (proton-gated) ion channels (ASICs). Streptozotocin-diabetic rats treated with insulin were considered ITD rats. ITD rats were exposed to RH for 5 days and were randomized into Psalmotoxin1 (PcTx1, ASIC1a inhibitor), APETx2 (ASIC3 inhibitor), or vehicle groups. Transient global cerebral ischemia was induced overnight after RH. Cerebral blood flow was measured using laser Doppler flowmetry. Ischemic brain injury in hippocampus was evaluated using histopathology. Post-ischemic hypoperfusion in RH-exposed rats was of greater extent than that in control rats. Inhibition of ASICs prevented RH-induced increase in the extent of post-ischemic hypoperfusion and ischemic brain injury. Since ASIC activation-induced store-operated calcium entry (SOCE) plays a role in vascular tone, next we tested if acidosis activates SOCE via activating ASICs in vascular smooth muscle cells (VSMCs). We observed that SOCE in VSMCs at lower pH is ASIC3 dependent. The results show the role of ASIC in post-ischemic hypoperfusion and increased ischemic damage in RH-exposed ITD rats. Understanding the pathways mediating exacerbated ischemic brain injury in RH-exposed ITD rats may help lower diabetic aggravation of ischemic brain damage.
Collapse
|
19
|
Battaglia M, Rossignol O, Bachand K, D'Amato FR, De Koninck Y. Amiloride modulation of carbon dioxide hypersensitivity and thermal nociceptive hypersensitivity induced by interference with early maternal environment. J Psychopharmacol 2019; 33:101-108. [PMID: 29968500 DOI: 10.1177/0269881118784872] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Early life adversities are risk factors for anxiety disorders and for pain syndromes, which are, in turn, highly comorbid with anxiety disorders. Repeated cross-fostering mouse pups to adoptive lactating females induces epigenetic modification and heightened mRNA-expression of the acid-sensing-ion-channel-1 gene, altered nociception, and hypersensitivity to 6% carbon dioxide air mixtures, a trait marker of specific human anxiety disorders such as, most clearly and prominently, panic disorder. AIMS We hypothesized that the acid-sensing ion channel inhibitor amiloride can modulate repeated cross-fostering animals' exaggerated responses to carbon dioxide and nociceptive thermal stimulation. METHODS Respiratory carbon dioxide sensitivity was assessed by plethysmography during 6% carbon dioxide air mixture challenges, and nociception was assessed by latency of paw withdrawal to thermal stimulation, in repeated cross-fostering and control animals. To circumvent the blood-brain barrier, prior to testing, amiloride was nebulized in a plethysmograph. Data were analyzed by general linear models. RESULTS Analyses of tidal volume responses to 6% carbon dioxide of animals pre-treated with nebulized amiloride/saline in a randomized crossover design showed significant modulatory effect of amiloride, and amiloride×repeated cross-fostering interaction. In contrast, repeated cross-fostering animals' responses to 6% carbon dioxide after intraperitoneal amiloride, saline, or no treatment, were no different. Analyses of responses to thermal stimuli showed a significant modulatory effect of nebulized amiloride, and repeated cross-fostering×amiloride interaction. CONCLUSIONS Single-dose nebulized amiloride decreased repeated cross-fostering animals' carbon dioxide sensitivity and nociception indices to levels that were no different from those of control animals. Inasmuch as these results pertain to human anxiety and/or pain hypersensitivity, our findings provide a rationale for studying inhaled amiloride in some anxiety disorders and/or pain syndromes.
Collapse
Affiliation(s)
- Marco Battaglia
- Child Youth and Emerging Adult Programme, Centre for Addiction & Mental Health, Toronto, ON, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Québec, QC, Canada
| | - Orlane Rossignol
- CERVO Brain Research Centre, Québec Mental Health Institute, Québec, QC, Canada
| | - Karine Bachand
- CERVO Brain Research Centre, Québec Mental Health Institute, Québec, QC, Canada
| | - Francesca R D'Amato
- Institute of Cell Biology and Neurobiology, National Research Council, Rome, Italy
| | - Yves De Koninck
- CERVO Brain Research Centre, Québec Mental Health Institute, Québec, QC, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Québec, QC, Canada
| |
Collapse
|
20
|
Qiang M, Dong X, Zha Z, Zuo XK, Song XL, Zhao L, Yuan C, Huang C, Tao P, Hu Q, Li WG, Hu W, Li J, Nie Y, Buratto D, Zonta F, Ma P, Yu Z, Liu L, Zhang Y, Yang B, Xie J, Xu TL, Qu Z, Yang G, Lerner RA. Selection of an ASIC1a-blocking combinatorial antibody that protects cells from ischemic death. Proc Natl Acad Sci U S A 2018; 115:E7469-E7477. [PMID: 30042215 PMCID: PMC6094137 DOI: 10.1073/pnas.1807233115] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Acid-sensing ion channels (ASICs) have emerged as important, albeit challenging therapeutic targets for pain, stroke, etc. One approach to developing therapeutic agents could involve the generation of functional antibodies against these channels. To select such antibodies, we used channels assembled in nanodiscs, such that the target ASIC1a has a configuration as close as possible to its natural state in the plasma membrane. This methodology allowed selection of functional antibodies that inhibit acid-induced opening of the channel in a dose-dependent way. In addition to regulation of pH, these antibodies block the transport of cations, including calcium, thereby preventing acid-induced cell death in vitro and in vivo. As proof of concept for the use of these antibodies to modulate ion channels in vivo, we showed that they potently protect brain cells from death after an ischemic stroke. Thus, the methodology described here should be general, thereby allowing selection of antibodies to other important ASICs, such as those involved in pain, neurodegeneration, and other conditions.
Collapse
Affiliation(s)
- Min Qiang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210 Shanghai, China
| | - Xue Dong
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210 Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, 201210 Shanghai, China
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031 Shanghai, China
- University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Zhao Zha
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210 Shanghai, China
| | - Xiao-Kun Zuo
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya Medical College of Central South University, 570100 Haikou, China
| | - Xing-Lei Song
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Lixia Zhao
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210 Shanghai, China
| | - Chao Yuan
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210 Shanghai, China
| | - Chen Huang
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Pingdong Tao
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210 Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, 201210 Shanghai, China
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031 Shanghai, China
- University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Qin Hu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Wei-Guang Li
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Wanhui Hu
- iHuman Institute, ShanghaiTech University, 201210 Shanghai, China
| | - Jie Li
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210 Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, 201210 Shanghai, China
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031 Shanghai, China
- University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Yan Nie
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210 Shanghai, China
| | - Damiano Buratto
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210 Shanghai, China
| | - Francesco Zonta
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210 Shanghai, China
| | - Peixiang Ma
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210 Shanghai, China
| | - Zheng Yu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210 Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, 201210 Shanghai, China
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031 Shanghai, China
- University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Lili Liu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210 Shanghai, China
| | - Yi Zhang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210 Shanghai, China
| | - Bei Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210 Shanghai, China
| | - Jia Xie
- Department of Chemistry, Scripps Research Institute, La Jolla, CA 92037
| | - Tian-Le Xu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Zhihu Qu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210 Shanghai, China;
| | - Guang Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210 Shanghai, China;
| | - Richard A Lerner
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210 Shanghai, China;
- Department of Chemistry, Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|