1
|
Zhu Y, Cao C, Li Z, Xu Z, Qian S, Zhang J, Li M, Hu X, Zhang A, Du N, Pan X, Wang X, Sun Y, Wang J, Huang Y. ASIC1a regulates ferroptosis in hepatic stellate cells via the Hippo/Yap-1 pathway in liver fibrosis. Int Immunopharmacol 2024; 143:113226. [PMID: 39353388 DOI: 10.1016/j.intimp.2024.113226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/28/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Liver fibrosis is a sustained process of liver tissue damage and repair caused by various physiological and pathological factors, with the activation and proliferation of hepatic stellate cells being central. Therefore, understanding and clarifying the relevant mechanisms of hepatic stellate cell activation and death is of great clinical significance for the treatment of liver fibrosis diseases. METHODS In vivo, recombinant adeno-associated virus was used to infect the liver of experimental mice, overexpressing ASIC1a, and based on this, a liver fibrosis model treated with sorafenib was constructed. In vitro, using RNA plasmid technology to transfect HSC-T6 cells, ASIC1a was overexpressed or silenced in the cells, and on this basis, PDGF-BB and Sorafenib were used to stimulate HSC-T6 cells, causing activated HSC-T6 to undergo ferroptosis. RESULTS The ferroptosis inducers Sorafenib and erastin can induce ferroptosis in HSCs, effectively inhibiting or reversing the progression of liver fibrosis. We found that the expression level of ASIC1a was significantly reduced in the livers of mice with liver fibrosis treated with Sorafenib. After treatment with an adeno-associated virus overexpressing ASIC1a, the therapeutic effect of Sorafenib was inhibited, and the level of ferroptosis induced by Sorafenib was also inhibited. The induction of ferroptosis in hepatic stellate cells in vitro depends on the presence of ASIC1a. By further exploring the potential mechanism, we observed that the overexpression of ASIC1a can promote an increase in YAP nuclear translocation, thereby regulating the activity of Hippo/YAP pathway signaling. After treatment with Sorafenib, the influx of Ca2+ significantly increased when ASIC1a was overexpressed, and BAPTA-AM intervention eliminated the intracellular Ca2+ accumulation induced by ASIC1a overexpression. CONCLUSIONS This indicated that the activation of YAP depends on the calcium ion influx induced by ASIC1a, which regulates ferroptosis in hepatic stellate cells by regulating the calcium ion-dependent Hippo/YAP pathway.
Collapse
Affiliation(s)
- Yueqin Zhu
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Office of Drug Clinical Trial Institutions, Anhui Provincial Cancer Hospital, Hefei 230031, China
| | - Chun Cao
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Zihao Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Zhou Xu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Shishun Qian
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Jingrong Zhang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Mengxue Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Xiaojie Hu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Anqi Zhang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Na Du
- Shanghai Songjiang District Central Hospital, Shanghai 201600, China
| | - Xuesheng Pan
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xinchen Wang
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Office of Drug Clinical Trial Institutions, Anhui Provincial Cancer Hospital, Hefei 230031, China
| | - Yancai Sun
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Office of Drug Clinical Trial Institutions, Anhui Provincial Cancer Hospital, Hefei 230031, China
| | - Jiajia Wang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| | - Yan Huang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
2
|
Berger KD, MacLean DM. Mechanism of acid-sensing ion channel modulation by Hi1a. J Gen Physiol 2024; 156:e202313519. [PMID: 39446054 DOI: 10.1085/jgp.202313519] [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: 12/08/2023] [Revised: 09/01/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024] Open
Abstract
Acid-sensing ion channels (ASICs) are trimeric cation-selective channels activated by extracellular acidification. Amongst many pathological roles, ASICs are an important mediator of ischemic cell death and hence an attractive drug target for stroke treatment as well as other conditions. A peptide called Hi1a, isolated from Australian funnel web spider venom, inhibits ASIC1a and attenuates cell death in a stroke model up to 8 h after stroke induction. Here, we set out to understand the molecular basis for Hi1a's action. Hi1a is a bivalent toxin with two inhibitory cystine knot domains joined by a short linker. We found that both Hi1a domains modulate human ASIC1a gating with the N-terminal domain impairing channel activation while the C-terminal domain produces a "pro-open" phenotype even at submicromolar concentrations. Interestingly, both domains bind at the same site since a single point mutation, F352A, abolishes functional effects and reduces toxin affinity in surface plasmon resonance measurements. Therefore, the action of Hi1a at ASIC1a appears to arise through a mutually exclusive binding model where either the N or C domain of a single Hi1a binds one ASIC1a subunit. An ASIC1a trimer may bind several inhibitory N domains and one or more pro-open C domains at any one time, accounting for the incomplete inhibition of wild type Hi1a. We also found that the functional differences between these two domains are partially transferred by mutagenesis, affording new insight into the channel function and possible novel avenues of drug design.
Collapse
Affiliation(s)
- Kyle D Berger
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - David M MacLean
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
3
|
Amaral-Silva L, Santin J. Neural Processing without O 2 and Glucose Delivery: Lessons from the Pond to the Clinic. Physiology (Bethesda) 2024; 39:0. [PMID: 38624246 DOI: 10.1152/physiol.00030.2023] [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: 11/29/2023] [Revised: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 04/17/2024] Open
Abstract
Neuronal activity requires a large amount of ATP, leading to a rapid collapse of brain function when aerobic respiration fails. Here, we summarize how rhythmic motor circuits in the brain stem of adult frogs, which normally have high metabolic demands, transform to produce proper output during severe hypoxia associated with emergence from hibernation. We suggest that general principles underlying plasticity in brain bioenergetics may be uncovered by studying nonmammalian models that face extreme environments, yielding new insights to combat neurological disorders involving dysfunctional energy metabolism.
Collapse
Affiliation(s)
- Lara Amaral-Silva
- Department of Biology, Wake Forest University, Winston-Salem, North Carolina, United States
- Division of Biology, University of Missouri, Columbia, Missouri, United States
| | - Joseph Santin
- Division of Biology, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
4
|
Majumder D. Ischemic Stroke: Pathophysiology and Evolving Treatment Approaches. Neurosci Insights 2024; 19:26331055241292600. [PMID: 39444789 PMCID: PMC11497522 DOI: 10.1177/26331055241292600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024] Open
Abstract
Stroke remains a leading cause of mortality and disability, with ischemic stroke being the most common type. It occurs due to reduced cerebral blood flow, leading to a cascade of events initiated by oxygen and nutrient deprivation, triggering excitotoxicity, oxidative stress, and inflammation and finally culminating in neuronal injury and death. Key molecular players in ischemic stroke include glutamate receptors, acid-sensing ion channels, and purinergic receptors, exacerbating cellular damage through calcium influx, oxidative stress, and mitochondrial dysfunction. Understanding these mechanisms has shaped therapeutic strategies, such as neuroprotective agents and stem cell therapies. Current treatments such as tissue plasminogen activator (tPA) emphasize timely intervention, yet challenges persist in patient-specific variability and accessibility. This review provides an overview of ischemic stroke pathophysiology, emphasizing cellular responses to ischemia and current and future therapeutic approaches including stem cell therapies aimed at mitigating stroke-induced disabilities and improving long-term outcomes.
Collapse
|
5
|
Budusan E, Payne CD, Gonzalez TI, Obergrussberger A, Becker N, Clark RJ, Johan Rosengren K, Rash LD, Cristofori-Armstrong B. The funnel-web spider venom derived single knot peptide Hc3a modulates acid-sensing ion channel 1a desensitisation. Biochem Pharmacol 2024; 228:116175. [PMID: 38552850 DOI: 10.1016/j.bcp.2024.116175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/17/2024] [Accepted: 03/26/2024] [Indexed: 04/07/2024]
Abstract
Acid-sensing ion channel 1a (ASIC1a) is a proton-gated channel involved in synaptic transmission, pain signalling, and several ischemia-associated pathological conditions. The spider venom-derived peptides PcTx1 and Hi1a are two of the most potent ASIC1a inhibitors known and have been instrumental in furthering our understanding of the structure, function, and biological roles of ASICs. To date, homologous spider peptides with different pharmacological profiles at ASIC1a have yet to be discovered. Here we report the characterisation of Hc3a, a single inhibitor cystine knot peptide from the Australian funnel-web spider Hadronyche cerberea with sequence similarity to PcTx1. We show that Hc3a has complex pharmacology and binds different ASIC1a conformational states (closed, open, and desensitised) with different affinities, with the most prominent effect on desensitisation. Hc3a slows the desensitisation kinetics of proton-activated ASIC1a currents across multiple application pHs, and when bound directly to ASIC1a in the desensitised conformation promotes current inhibition. The solution structure of Hc3a was solved, and the peptide-channel interaction examined via mutagenesis studies to highlight how small differences in sequence between Hc3a and PcTx1 can lead to peptides with distinct pharmacology. The discovery of Hc3a expands the pharmacological diversity of spider venom peptides targeting ASIC1a and adds to the toolbox of compounds to study the intricacies of ASIC1 gating.
Collapse
Affiliation(s)
- Elena Budusan
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - Colton D Payne
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - Tye I Gonzalez
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | | | | | - Richard J Clark
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - K Johan Rosengren
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia.
| | - Lachlan D Rash
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia.
| | - Ben Cristofori-Armstrong
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia.
| |
Collapse
|
6
|
Sun H, Yang T, Simon RP, Xiong ZG, Leng T. Role of Cholesterol Metabolic Enzyme CYP46A1 and Its Metabolite 24S-Hydroxycholesterol in Ischemic Stroke. Stroke 2024; 55:2492-2501. [PMID: 39224978 PMCID: PMC11421972 DOI: 10.1161/strokeaha.124.047803] [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: 05/09/2024] [Revised: 07/17/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND For several decades, it has been recognized that overactivation of the glutamate-gated N-methyl-D-aspartate receptors (NMDARs) and subsequent Ca2+ toxicity play a critical role in ischemic brain injury. 24S-hydroxycholesterol (24S-HC) is a major cholesterol metabolite in the brain, which has been identified as a potent positive allosteric modulator of NMDAR in rat hippocampal neurons. We hypothesize that 24S-HC worsens ischemic brain injury via its potentiation of the NMDAR, and reducing the production of 24S-HC by targeting its synthetic enzyme CYP46A1 provides neuroprotection. METHODS We tested this hypothesis using electrophysiological, pharmacological, and transgenic approaches and in vitro and in vivo cerebral ischemia models. RESULTS Our data show that 24S-HC potentiates NMDAR activation in primary cultured mouse cortical neurons in a concentration-dependent manner. At 10 µmol/L, it dramatically increases the steady-state currents by 51% and slightly increases the peak currents by 20%. Furthermore, 24S-HC increases NMDA and oxygen-glucose deprivation-induced cortical neuronal injury. The increased neuronal injury is largely abolished by NMDAR channel blocker MK-801, suggesting an NMDAR-dependent mechanism. Pharmacological inhibition of CYP46A1 by voriconazole or gene knockout of Cyp46a1 dramatically reduces ischemic brain injury. CONCLUSIONS These results identify a new mechanism and signaling cascade that critically impacts stroke outcome: CYP46A1 → 24S-HC → NMDAR → ischemic brain injury. They offer proof of principle for further development of new strategies for stroke intervention by targeting CYP46A1 or its metabolite 24S-HC.
Collapse
Affiliation(s)
- Huawei Sun
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA
| | - Tao Yang
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA
| | - Roger P Simon
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA
| | - Zhi-Gang Xiong
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA
| | - Tiandong Leng
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA
| |
Collapse
|
7
|
Sun W, Tiwari V, Davis G, Zhou G, Jonchhe S, Zha X. Time-Dependent Potentiation of the PERK Branch of UPR by GPR68 Offers Protection in Brain Ischemia. Stroke 2024; 55:2510-2521. [PMID: 39224971 PMCID: PMC11419283 DOI: 10.1161/strokeaha.124.048163] [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: 06/12/2024] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND In ischemia, acidosis occurs in/around injured tissue and parallels disease progression. Therefore, targeting an acid-sensitive receptor offers unique advantages in achieving the spatial and temporal specificity required for therapeutic interventions. We previously demonstrated that increased expression of GPR68 (G protein-coupled receptor 68), a proton-sensitive G protein-coupled receptor, mitigates ischemic brain injury. Here, we investigated the mechanism underlying GPR68-dependent protection. METHODS We performed biochemical and molecular analyses to examine poststroke signaling. We used in vitro brain slice cultures and in vivo mouse transient middle cerebral artery occlusion (tMCAO) models to investigate ischemia-induced injuries. RESULTS GPR68 deletion reduced PERK (protein kinase R-like ER kinase) expression in mouse brain. Compared with the wild-type mice, the GPR68-/- (knockout) mice exhibited a faster decline in eIF2α (eukaryotic initiation factor-2α) phosphorylation after tMCAO. Ogerin, a positive modulator of GPR68, stimulated eIF2α phosphorylation at 3 to 6 hours after tMCAO, primarily in the ipsilateral brain tissue. Consistent with the changes in eIF2α phosphorylation, Ogerin enhanced tMCAO-induced reduction in protein synthesis in ipsilateral brain tissue. In organotypic cortical slices, Ogerin reduced pH 6 and oxygen-glucose deprivation-induced neurotoxicity. Following tMCAO, intravenous delivery of Ogerin reduced brain infarction in wild-type but not knockout mice. Coapplication of a PERK inhibitor abolished Ogerin-induced protection. Delayed Ogerin delivery at 5 hours after tMCAO remained protective, and Ogerin has a similar protective effect in females. Correlated with these findings, tMCAO induced GPR68 expression at 6 hours, and Ogerin alters post-tMCAO proinflammatory/anti-inflammatory cytokine/chemokine expression profile. CONCLUSIONS These data demonstrate that GPR68 potentiation leads to neuroprotection, at least in part, through enhancing PERK-eIF2α activation in ischemic tissue but has little impact on healthy tissue.
Collapse
Affiliation(s)
- Wenyan Sun
- Division of Pharmacology and Pharmaceutical Sciences, University of Missouri-Kansas City (W.S., V.T., G.D., S.J., X.Z.)
- Now with: Tulane University, New Orleans, LA (W.S., V.T., X.Z.)
| | - Virendra Tiwari
- Division of Pharmacology and Pharmaceutical Sciences, University of Missouri-Kansas City (W.S., V.T., G.D., S.J., X.Z.)
- Now with: Tulane University, New Orleans, LA (W.S., V.T., X.Z.)
| | - Grace Davis
- Division of Pharmacology and Pharmaceutical Sciences, University of Missouri-Kansas City (W.S., V.T., G.D., S.J., X.Z.)
| | - Guokun Zhou
- Department of Physiology and Neuroscience, University of South Alabama, Mobile (G.Z.)
- Nantong University, Nantong City, China (G.Z.)
| | - Sarun Jonchhe
- Division of Pharmacology and Pharmaceutical Sciences, University of Missouri-Kansas City (W.S., V.T., G.D., S.J., X.Z.)
| | - Xiangming Zha
- Division of Pharmacology and Pharmaceutical Sciences, University of Missouri-Kansas City (W.S., V.T., G.D., S.J., X.Z.)
- Now with: Tulane University, New Orleans, LA (W.S., V.T., X.Z.)
| |
Collapse
|
8
|
Freuville L, Matthys C, Quinton L, Gillet JP. Venom-derived peptides for breaking through the glass ceiling of drug development. Front Chem 2024; 12:1465459. [PMID: 39398192 PMCID: PMC11468230 DOI: 10.3389/fchem.2024.1465459] [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: 07/16/2024] [Accepted: 09/04/2024] [Indexed: 10/15/2024] Open
Abstract
Venoms are complex mixtures produced by animals and consist of hundreds of components including small molecules, peptides, and enzymes selected for effectiveness and efficacy over millions of years of evolution. With the development of venomics, which combines genomics, transcriptomics, and proteomics to study animal venoms and their effects deeply, researchers have identified molecules that selectively and effectively act against membrane targets, such as ion channels and G protein-coupled receptors. Due to their remarkable physico-chemical properties, these molecules represent a credible source of new lead compounds. Today, not less than 11 approved venom-derived drugs are on the market. In this review, we aimed to highlight the advances in the use of venom peptides in the treatment of diseases such as neurological disorders, cardiovascular diseases, or cancer. We report on the origin and activity of the peptides already approved and provide a comprehensive overview of those still in development.
Collapse
Affiliation(s)
- Lou Freuville
- Laboratory of Mass Spectrometry, MolSys Research Unit, University of Liège, Liège, Belgium
| | - Chloé Matthys
- Laboratory of Molecular Cancer Biology, URPhyM, NARILIS, University of Namur, Namur, Belgium
| | - Loïc Quinton
- Laboratory of Mass Spectrometry, MolSys Research Unit, University of Liège, Liège, Belgium
| | - Jean-Pierre Gillet
- Laboratory of Molecular Cancer Biology, URPhyM, NARILIS, University of Namur, Namur, Belgium
| |
Collapse
|
9
|
Pu J, Han J, Yang J, Yu L, Wan H. Anaerobic Glycolysis and Ischemic Stroke: From Mechanisms and Signaling Pathways to Natural Product Therapy. ACS Chem Neurosci 2024; 15:3090-3105. [PMID: 39140296 DOI: 10.1021/acschemneuro.4c00371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024] Open
Abstract
Ischemic stroke is a serious condition that results in high rates of illness and death. Anaerobic glycolysis becomes the primary means of providing energy to the brain during periods of low oxygen levels, such as in the aftermath of an ischemic stroke. This process is essential for maintaining vital brain functions and has significant implications for recovery following a stroke. Energy supply by anaerobic glycolysis and acidosis caused by lactic acid accumulation are important pathological processes after ischemic stroke. Numerous natural products regulate glucose and lactate, which in turn modulate anaerobic glycolysis. This article focuses on the relationship between anaerobic glycolysis and ischemic stroke, as well as the associated signaling pathways and natural products that play a therapeutic role. These natural products, which can regulate anaerobic glycolysis, will provide new avenues and perspectives for the treatment of ischemic stroke in the future.
Collapse
Affiliation(s)
- Jia Pu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jin Han
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiehong Yang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Li Yu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, Zhejiang 310053, China
| | - Haitong Wan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
10
|
Wu PY, Lien CC. Modulation of Neurotransmission by Acid-Sensing Ion Channels. JOURNAL OF PHYSIOLOGICAL INVESTIGATION 2024; 67:242-248. [PMID: 39287486 DOI: 10.4103/ejpi.ejpi-d-24-00062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 07/20/2024] [Indexed: 09/19/2024]
Abstract
ABSTRACT Interstitial pH fluctuations occur normally in the brain and significantly modulate neuronal functions. Acid-sensing ion channels (ASICs), which serve as neuronal acid chemosensors, play important roles in synaptic plasticity, learning, and memory. However, the specific mechanisms by which ASICs influence neurotransmission remain elusive. Here, we report that ASICs modulate transmitter release and axonal excitability at a glutamatergic synapse in the rat and mouse hippocampus. Blocking ASIC1a channels with the tarantula peptide psalmotoxin 1 down-regulates basal transmission and alters short-term plasticity. Notably, the effect of psalmotoxin 1 on ASIC-mediated modulation is age-dependent, occurring only during a limited postnatal period (postnatal weeks 2-6). This finding suggests that protons, through the activation of ASICs, may act as modulators in synapse formation and maturation during early development.
Collapse
Affiliation(s)
- Pu-Yeh Wu
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Cheng-Chang Lien
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
11
|
Yin S, Zhang Y, Du B, Cao S, Wang K, Wei Q. Effects of intracranial artery stenosis of anterior circulation on cognition-A CT perfusion-based study. Brain Behav 2024; 14:e3521. [PMID: 39236078 PMCID: PMC11376367 DOI: 10.1002/brb3.3521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 03/19/2024] [Accepted: 04/19/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Intracranial atherosclerotic stenosis (ICAS) is one of the most important independent risk factors for stroke that is closely related to the occurrence of cognitive impairment. The relationship between ICAS and vascular cognitive impairment (VCI) remains unclear. Cerebral hemodynamic changes are one of the main causes of cognitive impairment. Computed tomographic perfusion (CTP) imaging can quantitatively analyze cerebral blood perfusion and quantify cerebral hemodynamic changes. Previous research on the relationship between hypoperfusion induced by ICAS and cognitive impairment, as well as its underlying mechanisms, remains relatively insufficient. This study is dedicated to elucidating the characteristics and potential mechanisms of cognitive impairment in ICAS patients with abnormal perfusion, utilizing CTP imaging as our primary investigative tool. METHODS This study recruited 82 patients who suffer from non-disabling ischemic stroke (IS group) and 28 healthy controls. All participants underwent comprehensive neuropsychological assessments both collectively and individually, in addition to CTP imaging. Within the patient group, we further categorized individuals into two subgroups: the ischemic penumbra group (IP, N = 28) and the benign oligemia group (BO, N = 54), based on CTP parameters-Tmax. The correlations between cognitive function and abnormal perfusion were explored. RESULTS The cognitive function, including the overall cognitive, memory, attention, executive functions, and language, was significantly impaired in the IS group compared with that in the control group. Further, there are statistical differences in the stroop color-word test-dot (Stroop-D) and Montreal Cognitive Assessment (MoCA) sub-items (memory + language) between the BO and IP groups. In the BO group, the score of Stroop-D is lower, and the MoCA sub-items are higher than the IP group. There is no correlation between CTP parameters and cognitive function. CONCLUSION Cognitive function is significantly impaired in patients with ICAS, which is related to cerebral perfusion. Executive, memory, and language function were better preserved in ICAS patients without IP. Hence, this study posits that managing hypoperfusion induced by ICAS may play a pivotal role in the development of VCI.
Collapse
Affiliation(s)
- Shanshan Yin
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui, China
| | - Ying Zhang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui, China
| | - Baogen Du
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui, China
| | - Shanshan Cao
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui, China
| | - Kai Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- The School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Qiang Wei
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui, China
| |
Collapse
|
12
|
Zhou R, Fu W, Vasylyev D, Waxman SG, Liu CJ. Ion channels in osteoarthritis: emerging roles and potential targets. Nat Rev Rheumatol 2024; 20:545-564. [PMID: 39122910 DOI: 10.1038/s41584-024-01146-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2024] [Indexed: 08/12/2024]
Abstract
Osteoarthritis (OA) is a highly prevalent joint disease that causes substantial disability, yet effective approaches to disease prevention or to the delay of OA progression are lacking. Emerging evidence has pinpointed ion channels as pivotal mediators in OA pathogenesis and as promising targets for disease-modifying treatments. Preclinical studies have assessed the potential of a variety of ion channel modulators to modify disease pathways involved in cartilage degeneration, synovial inflammation, bone hyperplasia and pain, and to provide symptomatic relief in models of OA. Some of these modulators are currently being evaluated in clinical trials. This review explores the structures and functions of ion channels, including transient receptor potential channels, Piezo channels, voltage-gated sodium channels, voltage-dependent calcium channels, potassium channels, acid-sensing ion channels, chloride channels and the ATP-dependent P2XR channels in the osteoarthritic joint. The discussion spans channel-targeting drug discovery and potential clinical applications, emphasizing opportunities for further research, and underscoring the growing clinical impact of ion channel biology in OA.
Collapse
Affiliation(s)
- Renpeng Zhou
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Wenyu Fu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Dmytro Vasylyev
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Chuan-Ju Liu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
13
|
Dong K, Chen F, Wang L, Lin C, Ying M, Li B, Huang T, Wang S. iMSC exosome delivers hsa-mir-125b-5p and strengthens acidosis resilience through suppression of ASIC1 protein in cerebral ischemia-reperfusion. J Biol Chem 2024; 300:107568. [PMID: 39019215 PMCID: PMC11363484 DOI: 10.1016/j.jbc.2024.107568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 07/01/2024] [Accepted: 06/12/2024] [Indexed: 07/19/2024] Open
Abstract
Acid-sensing ion channel 1 (ASIC1) is critical in acidotoxicity and significantly contributes to neuronal death in cerebral stroke. Pharmacological inhibition of ASIC1 has been shown to reduce neuronal death. However, the potential of utilizing exosomes derived from pluripotent stem cells to achieve inhibition of Asic1 remains to be explored. Developing qualified exosome products with precise and potent active ingredients suitable for clinical application is also ongoing. Here, we adopt small RNA-seq to interrogate the miRNA contents in exosomes of pluripotent stem cell induced mesenchymal stem cell (iMSC). RNA-seq was used to compare the oxygen-glucose deprivation-damaged neurons before and after the delivery of exosomes. We used Western blot to quantify the Asic1 protein abundance in neurons before and after exosome treatment. An in vivo test on rats validated the neuroprotective effect of iMSC-derived exosome and its active potent miRNA hsa-mir-125b-5p. We demonstrate that pluripotent stem cell-derived iMSCs produce exosomes with consistent miRNA contents and sustained expression. These exosomes efficiently rescue injured neurons, alleviate the pathological burden, and restore neuron function in rats under oxygen-glucose deprivation stress. Furthermore, we identify hsa-mir-125b-5p as the active component responsible for inhibiting the Asic1a protein and protecting neurons. We validated a novel therapeutic strategy to enhance acidosis resilience in cerebral stroke by utilizing exosomes derived from pluripotent stem cells with specific miRNA content. This holds promise for cerebral stroke treatment with the potential to reduce neuronal damage and improve clinical patient outcomes.
Collapse
Affiliation(s)
- Kai Dong
- Department of Neurology, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Fangyan Chen
- Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Liang Wang
- Department of Quality Control, Guidon Pharmaceutics, Beijing, China
| | - Chengyu Lin
- Department of Quality Control, Guidon Pharmaceutics, Beijing, China
| | - Mingyao Ying
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Bingnan Li
- Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China; Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China.
| | - Tao Huang
- Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Shuyan Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
14
|
Lai K, Pritišanac I, Liu ZQ, Liu HW, Gong LN, Li MX, Lu JF, Qi X, Xu TL, Forman-Kay J, Shi HB, Wang LY, Yin SK. Glutamate acts on acid-sensing ion channels to worsen ischaemic brain injury. Nature 2024; 631:826-834. [PMID: 38987597 PMCID: PMC11269185 DOI: 10.1038/s41586-024-07684-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 06/06/2024] [Indexed: 07/12/2024]
Abstract
Glutamate is traditionally viewed as the first messenger to activate NMDAR (N-methyl-D-aspartate receptor)-dependent cell death pathways in stroke1,2, but unsuccessful clinical trials with NMDAR antagonists implicate the engagement of other mechanisms3-7. Here we show that glutamate and its structural analogues, including NMDAR antagonist L-AP5 (also known as APV), robustly potentiate currents mediated by acid-sensing ion channels (ASICs) associated with acidosis-induced neurotoxicity in stroke4. Glutamate increases the affinity of ASICs for protons and their open probability, aggravating ischaemic neurotoxicity in both in vitro and in vivo models. Site-directed mutagenesis, structure-based modelling and functional assays reveal a bona fide glutamate-binding cavity in the extracellular domain of ASIC1a. Computational drug screening identified a small molecule, LK-2, that binds to this cavity and abolishes glutamate-dependent potentiation of ASIC currents but spares NMDARs. LK-2 reduces the infarct volume and improves sensorimotor recovery in a mouse model of ischaemic stroke, reminiscent of that seen in mice with Asic1a knockout or knockout of other cation channels4-7. We conclude that glutamate functions as a positive allosteric modulator for ASICs to exacerbate neurotoxicity, and preferential targeting of the glutamate-binding site on ASICs over that on NMDARs may be strategized for developing stroke therapeutics lacking the psychotic side effects of NMDAR antagonists.
Collapse
Affiliation(s)
- Ke Lai
- Department of Otorhinolaryngology, Shanghai Sixth People's Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai, China
| | - Iva Pritišanac
- Program in Molecular Medicine, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, Canada
- Department of Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Zhen-Qi Liu
- Department of Otorhinolaryngology, Shanghai Sixth People's Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Han-Wei Liu
- Department of Otorhinolaryngology, Shanghai Sixth People's Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-Na Gong
- Department of Otorhinolaryngology, Shanghai Sixth People's Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming-Xian Li
- Department of Otorhinolaryngology, Shanghai Sixth People's Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Fei Lu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Qi
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tian-Le Xu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Julie Forman-Kay
- Program in Molecular Medicine, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Hai-Bo Shi
- Department of Otorhinolaryngology, Shanghai Sixth People's Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lu-Yang Wang
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada.
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada.
| | - Shan-Kai Yin
- Department of Otorhinolaryngology, Shanghai Sixth People's Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
15
|
Wang Q, Yang F, Duo K, Liu Y, Yu J, Wu Q, Cai Z. The Role of Necroptosis in Cerebral Ischemic Stroke. Mol Neurobiol 2024; 61:3882-3898. [PMID: 38038880 DOI: 10.1007/s12035-023-03728-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/18/2023] [Indexed: 12/02/2023]
Abstract
Cerebral ischemia, also known as ischemic stroke, accounts for nearly 85% of all strokes and is the leading cause of disability worldwide. Due to disrupted blood supply to the brain, cerebral ischemic injury is trigged by a series of complex pathophysiological events including excitotoxicity, oxidative stress, inflammation, and cell death. Currently, there are few treatments for cerebral ischemia owing to an incomplete understanding of the molecular and cellular mechanisms. Accumulated evidence indicates that various types of programmed cell death contribute to cerebral ischemic injury, including apoptosis, ferroptosis, pyroptosis and necroptosis. Among these, necroptosis is morphologically similar to necrosis and is mediated by receptor-interacting serine/threonine protein kinase-1 and -3 and mixed lineage kinase domain-like protein. Necroptosis inhibitors have been shown to exert inhibitory effects on cerebral ischemic injury and neuroinflammation. In this review, we will discuss the current research progress regarding necroptosis in cerebral ischemia as well as the application of necroptosis inhibitors for potential therapeutic intervention in ischemic stroke.
Collapse
Affiliation(s)
- Qingsong Wang
- College of Pharmacy, Ningxia Medical University, Hui Autonomous Region, Yinchuan, 750004, Ningxia, China
| | - Fan Yang
- College of Pharmacy, Ningxia Medical University, Hui Autonomous Region, Yinchuan, 750004, Ningxia, China
| | - Kun Duo
- College of Pharmacy, Ningxia Medical University, Hui Autonomous Region, Yinchuan, 750004, Ningxia, China
| | - Yue Liu
- College of Pharmacy, Ningxia Medical University, Hui Autonomous Region, Yinchuan, 750004, Ningxia, China
| | - Jianqiang Yu
- College of Pharmacy, Ningxia Medical University, Hui Autonomous Region, Yinchuan, 750004, Ningxia, China
| | - Qihui Wu
- Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhenyu Cai
- College of Pharmacy, Ningxia Medical University, Hui Autonomous Region, Yinchuan, 750004, Ningxia, China.
- Shanghai Tenth People's Hospital, School of MedicineTongji University Cancer Center, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
16
|
Sun H, Yang T, Simon R, Xiong ZG, Leng T. Cholestane-3β,5α,6β-Triol Inhibits Acid-Sensing Ion Channels and Reduces Acidosis-Mediated Ischemic Brain Injury. Stroke 2024; 55:1660-1671. [PMID: 38660789 PMCID: PMC11126354 DOI: 10.1161/strokeaha.124.046963] [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: 01/11/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Activation of the acid-sensing ion channels (ASICs) by tissue acidosis, a common feature of brain ischemia, contributes to ischemic brain injury, while blockade of ASICs results in protection. Cholestane-3β,5α,6β-triol (Triol), a major cholesterol metabolite, has been demonstrated as an endogenous neuroprotectant; however, the mechanism underlying its neuroprotective activity remains elusive. In this study, we tested the hypothesis that inhibition of ASICs is a potential mechanism. METHODS The whole-cell patch-clamp technique was used to examine the effect of Triol on ASICs heterogeneously expressed in Chinese hamster ovary cells and ASICs endogenously expressed in primary cultured mouse cortical neurons. Acid-induced injury of cultured mouse cortical neurons and middle cerebral artery occlusion-induced ischemic brain injury in wild-type and ASIC1 and ASIC2 knockout mice were studied to examine the protective effect of Triol. RESULTS Triol inhibits ASICs in a subunit-dependent manner. In Chinese hamster ovary cells, it inhibits homomeric ASIC1a and ASIC3 without affecting ASIC1β and ASIC2a. In cultured mouse cortical neurons, it inhibits homomeric ASIC1a and heteromeric ASIC1a-containing channels. The inhibition is use-dependent but voltage- and pH-independent. Structure-activity relationship analysis suggests that hydroxyls at the 5 and 6 positions of the A/B ring are critical functional groups. Triol alleviates acidosis-mediated injury of cultured mouse cortical neurons and protects against middle cerebral artery occlusion-induced brain injury in an ASIC1a-dependent manner. CONCLUSIONS Our study identifies Triol as a novel ASIC inhibitor, which may serve as a new pharmacological tool for studying ASICs and may also be developed as a potential drug for treating stroke.
Collapse
Affiliation(s)
- Huawei Sun
- Department of Neurobiology, Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA 30329, USA
| | - Tao Yang
- Department of Neurobiology, Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA 30329, USA
| | - Roger Simon
- Department of Neurobiology, Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA 30329, USA
| | - Zhi-gang Xiong
- Department of Neurobiology, Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA 30329, USA
| | - Tiandong Leng
- Department of Neurobiology, Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA 30329, USA
| |
Collapse
|
17
|
Foster VS, Saez NJ, Gillespie ER, Jogia T, Reid C, Maljevic S, Jung W, Lao HW, Ruitenberg MJ, King GF. Genetic or Pharmacological Ablation of Acid-Sensing Ion Channel 1a (ASIC1a) Is Not Neuroprotective in a Mouse Model of Spinal Cord Injury. J Neurotrauma 2024; 41:1007-1019. [PMID: 36924276 DOI: 10.1089/neu.2022.0295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023] Open
Abstract
Acid-sensing ion channel 1a (ASIC1a) is a proton-activated channel that is expressed ubiquitously throughout the central nervous system and in various types of immune cells. Its role in spinal cord injury (SCI) is controversial; inhibition of ASIC1a has been reported to improve SCI pathology in vivo, but conversely, gene ablation increased kainite-mediated excitotoxic cell death in vitro. Here, we re-examined the role of ASIC1a in a mouse model of SCI. First, we observed functional outcomes up to 42 days post-operation (DPO) in SCI mice with a selective genetic ablation of ASIC1a. Mice lacking ASIC1a had significantly worsened locomotor ability and increased lesion size compared with mice possessing the ASIC1a gene. Next, we explored pharmacological antagonism of this ion channel by administering the potent ASIC1a inhibitor, Hi1a. Consistent with a role for ASIC1a to attenuate excitotoxicity, accelerated neuronal cell loss was found at the lesion site in SCI mice treated with Hi1a, but there were no differences in locomotor recovery. Moreover, ASIC1a inhibition did not cause significant alterations to neutrophil migration, microglial density, or blood-spinal cord barrier integrity.
Collapse
Affiliation(s)
- Victoria S Foster
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - Natalie J Saez
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - Ellen R Gillespie
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Queensland, Australia
| | - Trisha Jogia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Queensland, Australia
| | - Chantelle Reid
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Queensland, Australia
| | - Snezana Maljevic
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Woncheol Jung
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Queensland, Australia
| | - Hong W Lao
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Queensland, Australia
| | - Marc J Ruitenberg
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Queensland, Australia
- Trauma, Critical Care, and Recovery, Brisbane Diamantina Health Partners, Brisbane, Queensland, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
- Australian Research Council Center of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
18
|
Knezic A, Budusan E, Saez NJ, Broughton BRS, Rash LD, King GF, Widdop RE, McCarthy CA. Hi1a Improves Sensorimotor Deficit following Endothelin-1-Induced Stroke in Rats but Does Not Improve Functional Outcomes following Filament-Induced Stroke in Mice. ACS Pharmacol Transl Sci 2024; 7:1043-1054. [PMID: 38638162 PMCID: PMC11022283 DOI: 10.1021/acsptsci.3c00328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/07/2024] [Accepted: 03/04/2024] [Indexed: 04/20/2024]
Abstract
Activation of acid-sensing ion channel 1a (ASIC1a) plays a major role in mediating acidosis-induced neuronal injury following a stroke. Therefore, the inhibition of ASIC1a is a potential therapeutic avenue for the treatment of stroke. Venom-peptide Hi1a, a selective and highly potent ASIC1a inhibitor, reduces the infarct size and functional deficits when injected into the brain after stroke in rodents. However, its efficacy when administered using a clinically relevant route of administration remains to be established. Therefore, the current investigation aims to examine the efficacy of systemically administered Hi1a, using two different models of stroke in different species. Mice were subjected to the filament model of middle cerebral artery occlusion (MCAO) and treated with Hi1a systemically using either a single- or multiple-dosing regimen. 24 h poststroke, mice underwent functional testing, and the brain infarct size was assessed. Rats were subjected to endothelin-1 (ET-1)-induced MCAO and treated with Hi1a intravenously 2 h poststroke. Rats underwent functional tests prior to and for 3 days poststroke, when infarct volume was assessed. Mice receiving Hi1a did not show any improvements in functional outcomes, despite a trend toward reduced infarct size. This trend for reduced infarct size in mice was consistent regardless of the dosing regimen. There was also a trend toward lower infarct size in rats treated with Hi1a. More specifically, Hi1a reduced the amount of damage occurring within the somatosensory cortex, which was associated with an improved sensorimotor function in Hi1a-treated rats. Thus, this study suggests that Hi1a or more brain-permeable ASIC1a inhibitors are a potential stroke treatment.
Collapse
Affiliation(s)
- Adriana Knezic
- Cardiovascular Disease Program, Monash Biomedicine
Discovery Institute (BDI), Department of Pharmacology, Monash
University, Clayton, VIC 3800, Australia
| | - Elena Budusan
- School of Biomedical Sciences, Faculty of Medicine,
The University of Queensland, St Lucia, QLD 4072,
Australia
| | - Natalie J. Saez
- Institute for Molecular Bioscience, The
University of Queensland, St Lucia, QLD 4072,
Australia
- Australian Research Council Centre of Excellence for
Innovations in Peptide and Protein Science, The University of
Queensland, St Lucia, QLD 4072, Australia
| | - Brad R. S. Broughton
- Cardiovascular Disease Program, Monash Biomedicine
Discovery Institute (BDI), Department of Pharmacology, Monash
University, Clayton, VIC 3800, Australia
| | - Lachlan D. Rash
- School of Biomedical Sciences, Faculty of Medicine,
The University of Queensland, St Lucia, QLD 4072,
Australia
| | - Glenn F. King
- Institute for Molecular Bioscience, The
University of Queensland, St Lucia, QLD 4072,
Australia
- Australian Research Council Centre of Excellence for
Innovations in Peptide and Protein Science, The University of
Queensland, St Lucia, QLD 4072, Australia
| | - Robert E. Widdop
- Cardiovascular Disease Program, Monash Biomedicine
Discovery Institute (BDI), Department of Pharmacology, Monash
University, Clayton, VIC 3800, Australia
| | - Claudia A. McCarthy
- Cardiovascular Disease Program, Monash Biomedicine
Discovery Institute (BDI), Department of Pharmacology, Monash
University, Clayton, VIC 3800, Australia
| |
Collapse
|
19
|
Gründer S, Vanek J, Pissas KP. Acid-sensing ion channels and downstream signalling in cancer cells: is there a mechanistic link? Pflugers Arch 2024; 476:659-672. [PMID: 38175291 PMCID: PMC11006730 DOI: 10.1007/s00424-023-02902-z] [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: 11/23/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2024]
Abstract
It is increasingly appreciated that the acidic microenvironment of a tumour contributes to its evolution and clinical outcomes. However, our understanding of the mechanisms by which tumour cells detect acidosis and the signalling cascades that it induces is still limited. Acid-sensing ion channels (ASICs) are sensitive receptors for protons; therefore, they are also candidates for proton sensors in tumour cells. Although in non-transformed tissue, their expression is mainly restricted to neurons, an increasing number of studies have reported ectopic expression of ASICs not only in brain cancer but also in different carcinomas, such as breast and pancreatic cancer. However, because ASICs are best known as desensitizing ionotropic receptors that mediate rapid but transient signalling, how they trigger intracellular signalling cascades is not well understood. In this review, we introduce the acidic microenvironment of tumours and the functional properties of ASICs, point out some conceptual problems, summarize reported roles of ASICs in different cancers, and highlight open questions on the mechanisms of their action in cancer cells. Finally, we propose guidelines to keep ASIC research in cancer on solid ground.
Collapse
Affiliation(s)
- Stefan Gründer
- Institute of Physiology, RWTH Aachen University, Pauwelsstraße 30, 52074, Aachen, Germany.
| | - Jakob Vanek
- Institute of Physiology, RWTH Aachen University, Pauwelsstraße 30, 52074, Aachen, Germany
| | | |
Collapse
|
20
|
Platonov M, Maximyuk O, Rayevsky A, Hurmach V, Iegorova O, Naumchyk V, Bulgakov E, Cherninskyi A, Ozheredov D, Ryabukhin SV, Krishtal O, Volochnyuk DM. 4-(Azolyl)-Benzamidines as a Novel Chemotype for ASIC1a Inhibitors. Int J Mol Sci 2024; 25:3584. [PMID: 38612396 PMCID: PMC11011685 DOI: 10.3390/ijms25073584] [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: 11/26/2023] [Revised: 03/03/2024] [Accepted: 03/13/2024] [Indexed: 04/14/2024] Open
Abstract
Acid-sensing ion channels (ASICs) play a key role in the perception and response to extracellular acidification changes. These proton-gated cation channels are critical for neuronal functions, like learning and memory, fear, mechanosensation and internal adjustments like synaptic plasticity. Moreover, they play a key role in neuronal degeneration, ischemic neuronal injury, seizure termination, pain-sensing, etc. Functional ASICs are homo or heterotrimers formed with (ASIC1-ASIC3) homologous subunits. ASIC1a, a major ASIC isoform in the central nervous system (CNS), possesses an acidic pocket in the extracellular region, which is a key regulator of channel gating. Growing data suggest that ASIC1a channels are a potential therapeutic target for treating a variety of neurological disorders, including stroke, epilepsy and pain. Many studies were aimed at identifying allosteric modulators of ASIC channels. However, the regulation of ASICs remains poorly understood. Using all available crystal structures, which correspond to different functional states of ASIC1, and a molecular dynamics simulation (MD) protocol, we analyzed the process of channel inactivation. Then we applied a molecular docking procedure to predict the protein conformation suitable for the amiloride binding. To confirm the effect of its sole active blocker against the ASIC1 state transition route we studied the complex with another MD simulation run. Further experiments evaluated various compounds in the Enamine library that emerge with a detectable ASIC inhibitory activity. We performed a detailed analysis of the structural basis of ASIC1a inhibition by amiloride, using a combination of in silico approaches to visualize its interaction with the ion pore in the open state. An artificial activation (otherwise, expansion of the central pore) causes a complex modification of the channel structure, namely its transmembrane domain. The output protein conformations were used as a set of docking models, suitable for a high-throughput virtual screening of the Enamine chemical library. The outcome of the virtual screening was confirmed by electrophysiological assays with the best results shown for three hit compounds.
Collapse
Affiliation(s)
- Maksym Platonov
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Zabolotnogo Str., 150, 03143 Kyiv, Ukraine; (M.P.); (V.H.)
- Enamine Ltd., 78 Winston Churchill Str., 02660 Kyiv, Ukraine; (V.N.); (E.B.); (D.M.V.)
| | - Oleksandr Maximyuk
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, 4 Bogomoletz Str., 01024 Kyiv, Ukraine; (O.M.); (O.I.); (A.C.); (O.K.)
| | - Alexey Rayevsky
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Zabolotnogo Str., 150, 03143 Kyiv, Ukraine; (M.P.); (V.H.)
- Enamine Ltd., 78 Winston Churchill Str., 02660 Kyiv, Ukraine; (V.N.); (E.B.); (D.M.V.)
- Institute of Food Biotechnology and Genomics, National Academy of Sciences of Ukraine, Osypovskoho Str., 2A, 04123 Kyiv, Ukraine;
| | - Vasyl Hurmach
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Zabolotnogo Str., 150, 03143 Kyiv, Ukraine; (M.P.); (V.H.)
- Enamine Ltd., 78 Winston Churchill Str., 02660 Kyiv, Ukraine; (V.N.); (E.B.); (D.M.V.)
| | - Olena Iegorova
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, 4 Bogomoletz Str., 01024 Kyiv, Ukraine; (O.M.); (O.I.); (A.C.); (O.K.)
| | - Vasyl Naumchyk
- Enamine Ltd., 78 Winston Churchill Str., 02660 Kyiv, Ukraine; (V.N.); (E.B.); (D.M.V.)
- Institute of High Technologies, Taras Shevchenko National University of Kyiv, Volodymyrska Street 60, 01601 Kyiv, Ukraine
| | - Elijah Bulgakov
- Enamine Ltd., 78 Winston Churchill Str., 02660 Kyiv, Ukraine; (V.N.); (E.B.); (D.M.V.)
- Institute of Food Biotechnology and Genomics, National Academy of Sciences of Ukraine, Osypovskoho Str., 2A, 04123 Kyiv, Ukraine;
| | - Andrii Cherninskyi
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, 4 Bogomoletz Str., 01024 Kyiv, Ukraine; (O.M.); (O.I.); (A.C.); (O.K.)
| | - Danil Ozheredov
- Institute of Food Biotechnology and Genomics, National Academy of Sciences of Ukraine, Osypovskoho Str., 2A, 04123 Kyiv, Ukraine;
| | - Serhiy V. Ryabukhin
- Enamine Ltd., 78 Winston Churchill Str., 02660 Kyiv, Ukraine; (V.N.); (E.B.); (D.M.V.)
- Institute of High Technologies, Taras Shevchenko National University of Kyiv, Volodymyrska Street 60, 01601 Kyiv, Ukraine
- Institute of Organic Chemistry, National Academy of Sciences of Ukraine, 5 Academik Kukhar Str., 02660 Kyiv, Ukraine
| | - Oleg Krishtal
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, 4 Bogomoletz Str., 01024 Kyiv, Ukraine; (O.M.); (O.I.); (A.C.); (O.K.)
| | - Dmytro M. Volochnyuk
- Enamine Ltd., 78 Winston Churchill Str., 02660 Kyiv, Ukraine; (V.N.); (E.B.); (D.M.V.)
- Institute of High Technologies, Taras Shevchenko National University of Kyiv, Volodymyrska Street 60, 01601 Kyiv, Ukraine
- Institute of Organic Chemistry, National Academy of Sciences of Ukraine, 5 Academik Kukhar Str., 02660 Kyiv, Ukraine
| |
Collapse
|
21
|
Kappel S, Melek K, Ross-Kaschitza D, Hauert B, Gerber CE, Lochner M, Peinelt C. CBA (4-chloro-2-(2-chlorophenoxy)acetamido) benzoic acid) inhibits TMEM206 mediated currents and TMEM206 does not contribute to acid-induced cell death in colorectal cancer cells. Front Pharmacol 2024; 15:1369513. [PMID: 38515848 PMCID: PMC10955468 DOI: 10.3389/fphar.2024.1369513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 02/08/2024] [Indexed: 03/23/2024] Open
Abstract
Introduction: Upon activation at low pH, TMEM206 conducts Cl- ions across plasma and vesicular membranes. In a (patho)physiological context, TMEM206 was reported to contribute to acid-induced cell death in neurons, kidney and cervical epithelial cells. We investigated the role of TMEM206 in acid-induced cell death in colorectal cancer cells. In addition, we studied CBA as a new small molecule inhibitor for TMEM206. Methods: The role of TMEM206 in acid-induced cell death was studied with CRISPR/Cas9-mediated knockout and FACS analysis. The pharmacology of TMEM206 was determined with the patch clamp technique. Results: In colorectal cancer cells, TMEM206 is not a critical mediator of acid-induced cell death. CBA is a small molecule inhibitor of TMEM206 (IC50 = 9.55 µM) at low pH, at pH 6.0 inhibition is limited. Conclusion: CBA demonstrates effective and specific inhibition of TMEM206; however, its inhibitory efficacy is limited at pH 6.0. Despite this limitation, CBA is a potent inhibitor for functional studies at pH 4.5 and may be a promising scaffold for the development of future TMEM206 inhibitors.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Christine Peinelt
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
22
|
Wang H, Ma W, Hu W, Li X, Shen N, Li Z, Kong X, Lin T, Gao J, Zhu T, Che F, Chen J, Wan Q. Cathodal bilateral transcranial direct-current stimulation regulates selenium to confer neuroprotection after rat cerebral ischaemia-reperfusion injury. J Physiol 2024; 602:1175-1197. [PMID: 38431908 DOI: 10.1113/jp285806] [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: 10/15/2023] [Accepted: 02/13/2024] [Indexed: 03/05/2024] Open
Abstract
Non-invasive transcranial direct-current stimulation (tDCS) is a safe ischaemic stroke therapy. Cathodal bilateral tDCS (BtDCS) is a modified tDCS approach established by us recently. Because selenium (Se) plays a crucial role in cerebral ischaemic injury, we investigated whether cathodal BtDCS conferred neuroprotection via regulating Se-dependent signalling in rat cerebral ischaemia-reperfusion (I/R) injury. We first showed that the levels of Se and its transport protein selenoprotein P (SEPP1) were reduced in the rat cortical penumbra following I/R, whereas cathodal BtDCS prevented the reduction of Se and SEPP1. Interestingly, direct-current stimulation (DCS) increased SEPP1 level in cultured astrocytes subjected to oxygen-glucose deprivation reoxygenation (OGD/R) but had no effect on SEPP1 level in OGD/R-insulted neurons, indicating that DCS may increase Se in ischaemic neurons by enhancing the synthesis and secretion of SEPP1 in astrocytes. We then revealed that DCS reduced the number of injured mitochondria in OGD/R-insulted neurons cocultured with astrocytes. DCS and BtDCS prevented the reduction of the mitochondrial quality-control signalling, vesicle-associated membrane protein 2 (VAMP2) and syntaxin-4 (STX4), in OGD/R-insulted neurons cocultured with astrocytes and the ischaemic brain respectively. Under the same experimental conditions, downregulation of SEPP1 blocked DCS- and BtDCS-induced upregulation of VAMP2 and STX4. Finally, we demonstrated that cathodal BtDCS increased Se to reduce infract volume following I/R. Together, the present study uncovered a molecular mechanism by which cathodal BtDCS confers neuroprotection through increasing SEPP1 in astrocytes and subsequent upregulation of SEPP1/VAMP2/STX4 signalling in ischaemic neurons after rat cerebral I/R injury. KEY POINTS: Cathodal bilateral transcranial direct-current stimulation (BtDCS) prevents the reduction of selenium (Se) and selenoprotein P in the ischaemic penumbra. Se plays a crucial role in cerebral ischaemia injury. Direct-current stimulation reduces mitochondria injury and blocks the reduction of vesicle-associated membrane protein 2 (VAMP2) and syntaxin-4 (STX4) in oxygen-glucose deprivation reoxygenation-insulted neurons following coculturing with astrocytes. Cathodal BtDCS regulates Se/VAMP2/STX4 signalling to confer neuroprotection after ischaemia.
Collapse
Affiliation(s)
- Hui Wang
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wenlong Ma
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wenjie Hu
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiaohua Li
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Na Shen
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Zhuo Li
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiangyi Kong
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Tao Lin
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jingchen Gao
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Ting Zhu
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Fengyuan Che
- Central Laboratory, Department of Neurology, Linyi People's Hospital, Qingdao University, Linyi, Shandong, China
| | - Juan Chen
- Department of Neurology, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Qi Wan
- Institute of Neuroregeneration & Neurorehabilitation, School of Basic Medicine, Qingdao University, Qingdao, China
- Qingdao Gui-Hong Intelligent Medical Technology Co. Ltd, Qingdao, China
| |
Collapse
|
23
|
Zhang X, Zhang Y, Su Q, Liu Y, Li Z, Yong VW, Xue M. Ion Channel Dysregulation Following Intracerebral Hemorrhage. Neurosci Bull 2024; 40:401-414. [PMID: 37755675 PMCID: PMC10912428 DOI: 10.1007/s12264-023-01118-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/14/2023] [Indexed: 09/28/2023] Open
Abstract
Injury to the brain after intracerebral hemorrhage (ICH) results from numerous complex cellular mechanisms. At present, effective therapy for ICH is limited and a better understanding of the mechanisms of brain injury is necessary to improve prognosis. There is increasing evidence that ion channel dysregulation occurs at multiple stages in primary and secondary brain injury following ICH. Ion channels such as TWIK-related K+ channel 1, sulfonylurea 1 transient receptor potential melastatin 4 and glutamate-gated channels affect ion homeostasis in ICH. They in turn participate in the formation of brain edema, disruption of the blood-brain barrier, and the generation of neurotoxicity. In this review, we summarize the interaction between ions and ion channels, the effects of ion channel dysregulation, and we discuss some therapeutics based on ion-channel modulation following ICH.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Yan Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Qiuyang Su
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Zhe Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, AB, T2N 1N4, Canada.
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
24
|
Armstrong A, Yang T, Leng T, Xiong ZG. Inhibition of ASIC1a Improves Behavioral Recovery after Stroke. eNeuro 2024; 11:ENEURO.0341-23.2023. [PMID: 38233146 PMCID: PMC10866329 DOI: 10.1523/eneuro.0341-23.2023] [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: 09/05/2023] [Revised: 10/19/2023] [Accepted: 10/26/2023] [Indexed: 01/19/2024] Open
Abstract
Stroke continues to be a leading cause of death and long-term disabilities worldwide, despite extensive research efforts. The failure of multiple clinical trials raises the need for continued study of brain injury mechanisms and novel therapeutic strategies for ischemic stroke. The contribution of acid-sensing ion channel 1a (ASIC1a) to neuronal injury during the acute phase of stroke has been well studied; however, the long-term impact of ASIC1a inhibition on stroke recovery has not been established. The present study sought to bridge part of the translational gap by focusing on long-term behavioral recovery after a 30 min stroke in mice that had ASIC1a knocked out or inhibited by PcTX1. The neurological consequences of stroke in mice were evaluated before and after the stroke using neurological deficit score, open field, and corner turn test over a 28 d period. ASIC1a knock-out and inhibited mice showed improved neurological scores more quickly than wild-type control and vehicle-injected mice after the stroke. ASIC1a knock-out mice also recovered from mobility deficits in the open field test more quickly than wild-type mice, while PcTX1-injected mice did not experience significant mobility deficits at all after the stroke. In contrast to vehicle-injected mice that showed clear-sidedness bias in the corner turn test after stroke, PcTX1-injected mice never experienced significant-sidedness bias at all. This study supports and extends previous work demonstrating ASIC1a as a potential therapeutic target for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Ariel Armstrong
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia 30310
| | - Tao Yang
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia 30310
| | - Tiandong Leng
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia 30310
| | - Zhi-Gang Xiong
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia 30310
| |
Collapse
|
25
|
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
|
26
|
Liu Y, Ma J, DesJarlais RL, Hagan R, Rech J, Liu C, Miller R, Schoellerman J, Luo J, Letavic M, Grasberger B, Maher MP. Molecular determinants of ASIC1 modulation by divalent cations. Sci Rep 2024; 14:2320. [PMID: 38282035 PMCID: PMC10822848 DOI: 10.1038/s41598-024-52845-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/24/2024] [Indexed: 01/30/2024] Open
Abstract
Acid-sensing ion channels (ASICs) are proton-gated cation channels widely expressed in the nervous system. ASIC gating is modulated by divalent cations as well as small molecules; however, the molecular determinants of gating modulation by divalent cations are not well understood. Previously, we identified two small molecules that bind to ASIC1a at a novel site in the acidic pocket and modulate ASIC1 gating in a manner broadly resembling divalent cations, raising the possibility that these small molecules may help to illuminate the molecular determinants of gating modulation by divalent cations. Here, we examined how these two groups of modulators might interact as well as mutational effects on ASIC1a gating and its modulation by divalent cations. Our results indicate that binding of divalent cations to an acidic pocket site plays a key role in gating modulation of the channel.
Collapse
Affiliation(s)
- Yi Liu
- Neuroscience Discovery, Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Jichun Ma
- Therapeutics Discovery, Janssen Research & Development, L.L.C., Welsh & McKean Roads, P.O. Box 776, Spring House, PA, 19477, USA
| | - Renee L DesJarlais
- Therapeutics Discovery, Janssen Research & Development, L.L.C., Welsh & McKean Roads, P.O. Box 776, Spring House, PA, 19477, USA
| | - Rebecca Hagan
- Neuroscience Discovery, Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Jason Rech
- Therapeutics Discovery, Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Changlu Liu
- Neuroscience Discovery, Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Robyn Miller
- Therapeutics Discovery, Janssen Research & Development, L.L.C., Welsh & McKean Roads, P.O. Box 776, Spring House, PA, 19477, USA
| | - Jeffrey Schoellerman
- Neuroscience Discovery, Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Jinquan Luo
- Therapeutics Discovery, Janssen Research & Development, L.L.C., Welsh & McKean Roads, P.O. Box 776, Spring House, PA, 19477, USA
| | - Michael Letavic
- Therapeutics Discovery, Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Bruce Grasberger
- Therapeutics Discovery, Janssen Research & Development, L.L.C., Welsh & McKean Roads, P.O. Box 776, Spring House, PA, 19477, USA
| | - Michael P Maher
- Neuroscience Discovery, Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, USA.
| |
Collapse
|
27
|
Park G, Ge Q, Jin Z, Du J. Acid-Sensing Ion Channel 1a Contributes to the Prefrontal Cortex Ischemia-Enhanced Neuronal Activities in the Amygdala. Brain Sci 2023; 13:1684. [PMID: 38137132 PMCID: PMC10741891 DOI: 10.3390/brainsci13121684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/29/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Following a stroke, the emergence of amygdala-related disorders poses a significant challenge, with severe implications for post-stroke mental health, including conditions such as anxiety and depression. These disorders not only hinder post-stroke recovery but also elevate mortality rates. Despite their profound impact, the precise origins of aberrant amygdala function after a stroke remain elusive. As a target of reduced brain pH in ischemia, acid-sensing ion channels (ASICs) have been implicated in synaptic transmission after ischemia, hinting at their potential role in reshaping neural circuits following a stroke. This study delves into the intriguing relationship between post-stroke alterations and ASICs, specifically focusing on postsynaptic ASIC1a enhancement in the amygdala following prefrontal cortex (PFC) ischemia induced by endothelin-1 (ET-1) injection. Our findings intriguingly illustrate that mPFC ischemia not only accentuates the PFC to the amygdala circuit but also implicates ASIC1a in fostering augmented synaptic plasticity after ischemia. In contrast, the absence of ASIC1a impairs the heightened induction of long-term potentiation (LTP) in the amygdala induced by ischemia. This pivotal research introduces a novel concept with the potential to inaugurate an entirely new avenue of inquiry, thereby significantly enhancing our comprehension of the intricate mechanisms underlying post-stroke neural circuit reconfiguration. Importantly, these revelations hold the promise of paving the way for groundbreaking therapeutic interventions.
Collapse
Affiliation(s)
- Gyeongah Park
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Qian Ge
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Zhen Jin
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jianyang Du
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
28
|
Cullinan MM, Klipp RC, Camenisch A, Bankston JR. Dynamic landscape of the intracellular termini of acid-sensing ion channel 1a. eLife 2023; 12:RP90755. [PMID: 38054969 PMCID: PMC10699805 DOI: 10.7554/elife.90755] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023] Open
Abstract
Acid-sensing ion channels (ASICs) are trimeric proton-gated sodium channels. Recent work has shown that these channels play a role in necroptosis following prolonged acidic exposure like occurs in stroke. The C-terminus of ASIC1a is thought to mediate necroptotic cell death through interaction with receptor interacting serine threonine kinase 1 (RIPK1). This interaction is hypothesized to be inhibited at rest via an interaction between the C- and N-termini which blocks the RIPK1 binding site. Here, we use two transition metal ion FRET methods to investigate the conformational dynamics of the termini at neutral and acidic pH. We do not find evidence that the termini are close enough to be bound while the channel is at rest and find that the termini may modestly move closer together during acidification. At rest, the N-terminus adopts a conformation parallel to the membrane about 10 Å away. The distal end of the C-terminus may also spend time close to the membrane at rest. After acidification, the proximal portion of the N-terminus moves marginally closer to the membrane whereas the distal portion of the C-terminus swings away from the membrane. Together these data suggest that a new hypothesis for RIPK1 binding during stroke is needed.
Collapse
Affiliation(s)
- Megan M Cullinan
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Robert C Klipp
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
| | | | - John R Bankston
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
| |
Collapse
|
29
|
Rahman S, Iram S, Rehman MT, Hussain A, Jan AT, Kim J. Study of Amiloride Binding to Human Serum Albumin: Insights from Thermodynamic, Spectroscopic, and Molecular Docking Investigations. Molecules 2023; 28:7688. [PMID: 38067419 PMCID: PMC10707572 DOI: 10.3390/molecules28237688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
This study was undertaken to investigate the interaction between the sodium channel blocker amiloride (AML) and human serum albumin (HSA). A combination of multi-spectroscopic techniques and computational methods were employed to identify the AML binding site on HSA and the forces responsible for the formation of the HSA-AML complex. Our findings revealed that AML specifically binds to Sudlow's site II, located in subdomain IIIA of HSA, and that the complex formed is stabilized using van der Waals hydrogen-bonding and hydrophobic interactions. FRET analysis showed that the distance between AML and Trp214 was optimal for efficient quenching. UV-Vis spectroscopy and circular dichroism indicated minor changes in the structure of HSA after AML binding, and molecular dynamics simulations (MDS) conducted over 100 ns provided additional evidence of stable HSA-AML-complex formation. This study enhances understanding of the interaction between AML and HSA and the mechanism responsible.
Collapse
Affiliation(s)
- Safikur Rahman
- Munshi Singh College, BR Ambedkar Bihar University, Muzaffarpur 845401, India;
| | - Sana Iram
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 712-749, Republic of Korea;
| | - Md Tabish Rehman
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.T.R.); (A.H.)
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.T.R.); (A.H.)
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, India;
| | - Jihoe Kim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 712-749, Republic of Korea;
| |
Collapse
|
30
|
Rook ML, McCullock TW, Couch T, Lueck JD, MacLean DM. Photomodulation of the ASIC1a acidic pocket destabilizes the open state. Protein Sci 2023; 32:e4800. [PMID: 37805833 PMCID: PMC10599103 DOI: 10.1002/pro.4800] [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: 10/29/2022] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 10/09/2023]
Abstract
Acid-sensing ion channels (ASICs) are important players in detecting extracellular acidification throughout the brain and body. ASICs have large extracellular domains containing two regions replete with acidic residues: the acidic pocket, and the palm domain. In the resting state, the acidic pocket is in an expanded conformation but collapses in low pH conditions as the acidic side chains are neutralized. Thus, extracellular acidification has been hypothesized to collapse the acidic pocket that, in turn, ultimately drives channel activation. However, several observations run counter to this idea. To explore how collapse or mobility of the acidic pocket is linked to channel gating, we employed two distinct tools. First, we incorporated the photocrosslinkable noncanonical amino acids (ncAAs) 4-azido-L-phenylalanine (AzF) or 4-benzoyl-L-phenylalanine (BzF) into several positions in the acidic pocket. At both E315 and Y318, AzF incorporation followed by UV irradiation led to right shifts in pH response curves and accelerations of desensitization and deactivation, consistent with restrictions of acidic pocket mobility destabilizing the open state. Second, we reasoned that because Cl- ions are found in the open and desensitized structures but absent in the resting state structures, Cl- substitution would provide insight into how stability of the pocket is linked to gating. Anion substitution resulted in faster deactivation and desensitization, consistent with the acidic pocket regulating the stability of the open state. Taken together, our data support a model where acidic pocket collapse is not essential for channel activation. Rather, collapse of the acidic pocket influences the stability of the open state of the pore.
Collapse
Affiliation(s)
- Matthew L. Rook
- Department of Pharmacology and Physiology, School of Medicine and DentistryUniversity of RochesterRochesterNew YorkUSA
| | - Tyler W. McCullock
- Department of Pharmacology and Physiology, School of Medicine and DentistryUniversity of RochesterRochesterNew YorkUSA
| | - Tyler Couch
- Department of Pharmacology and Physiology, School of Medicine and DentistryUniversity of RochesterRochesterNew YorkUSA
| | - John D. Lueck
- Department of Pharmacology and Physiology, School of Medicine and DentistryUniversity of RochesterRochesterNew YorkUSA
- Deparment of Neurology, School of Medicine and DentistryUniversity of RochesterRochesterNew YorkUSA
- Center for RNA BiologyUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | - David M. MacLean
- Department of Pharmacology and Physiology, School of Medicine and DentistryUniversity of RochesterRochesterNew YorkUSA
| |
Collapse
|
31
|
Zhao S, Chi L, Chen H. CEGA: a method for inferring natural selection by comparative population genomic analysis across species. Genome Biol 2023; 24:219. [PMID: 37789379 PMCID: PMC10548728 DOI: 10.1186/s13059-023-03068-8] [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: 07/26/2022] [Accepted: 09/20/2023] [Indexed: 10/05/2023] Open
Abstract
We developed maximum likelihood method for detecting positive selection or balancing selection using multilocus or genomic polymorphism and divergence data from two species. The method is especially useful for investigating natural selection in noncoding regions. Simulations demonstrate that the method outperforms existing methods in detecting both positive and balancing selection. We apply the method to population genomic data from human and chimpanzee. The list of genes identified under selection in the noncoding regions is prominently enriched in pathways related to the brain and nervous system. Therefore, our method will serve as a useful tool for comparative population genomic analysis.
Collapse
Affiliation(s)
- Shilei Zhao
- CAS Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
- China National Center for Bioinformation, Beijing, 100101, China
- School of Future Technology, College of Life Sciences and Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lianjiang Chi
- CAS Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
- China National Center for Bioinformation, Beijing, 100101, China
| | - Hua Chen
- CAS Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.
- China National Center for Bioinformation, Beijing, 100101, China.
- School of Future Technology, College of Life Sciences and Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China.
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
| |
Collapse
|
32
|
Arya S, Bahuguna D, Bajad G, Loharkar S, Devangan P, Khatri DK, Singh SB, Madan J. Colloidal therapeutics in the management of traumatic brain injury: Portray of biomarkers and drug-targets, preclinical and clinical pieces of evidence and future prospects. Colloids Surf B Biointerfaces 2023; 230:113509. [PMID: 37595379 DOI: 10.1016/j.colsurfb.2023.113509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/28/2023] [Accepted: 08/05/2023] [Indexed: 08/20/2023]
Abstract
Complexity associated with the aberrant physiology of traumatic brain injury (TBI) makes its therapeutic targeting vulnerable. The underlying mechanisms of pathophysiology of TBI are yet to be completely illustrated. Primary injury in TBI is associated with contusions and axonal shearing whereas excitotoxicity, mitochondrial dysfunction, free radicals generation, and neuroinflammation are considered under secondary injury. MicroRNAs, proinflammatory cytokines, and Glial fibrillary acidic protein (GFAP) recently emerged as biomarkers in TBI. In addition, several approved therapeutic entities have been explored to target existing and newly identified drug-targets in TBI. However, drug delivery in TBI is hampered due to disruption of blood-brain barrier (BBB) in secondary TBI, as well as inadequate drug-targeting and retention effect. Colloidal therapeutics appeared helpful in providing enhanced drug availability to the brain owing to definite targeting strategies. Moreover, immense efforts have been put together to achieve increased bioavailability of therapeutics to TBI by devising effective targeting strategies. The potential of colloidal therapeutics to efficiently deliver drugs at the site of injury and down-regulate the mediators of TBI are serving as novel policies in the management of TBI. Therefore, in present manuscript, we have illuminated a myriad of molecular-targets currently identified and recognized in TBI. Moreover, particular emphasis is given to frame armamentarium of repurpose drugs which could be utilized to block molecular targets in TBI in addition to drug delivery barriers. The critical role of colloidal therapeutics such as liposomes, nanoparticles, dendrimers, and exosomes in drug delivery to TBI through invasive and non-invasive routes has also been highlighted.
Collapse
Affiliation(s)
- Shristi Arya
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Deepankar Bahuguna
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Gopal Bajad
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Soham Loharkar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Pawan Devangan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Dharmendra Kumar Khatri
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Shashi Bala Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India.
| |
Collapse
|
33
|
Cullinan MM, Klipp RC, Camenisch A, Bankston JR. Dynamic landscape of the intracellular termini of acid-sensing ion channel 1a. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.05.547693. [PMID: 37461628 PMCID: PMC10350031 DOI: 10.1101/2023.07.05.547693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Acid-sensing ion channels (ASICs) are trimeric proton-gated sodium channels. Recently it has been shown that these channels play a role in necroptosis following prolonged acidic exposure like occurs in stroke. The C-terminus of the channel is thought to mediate necroptotic cell death through interaction with receptor interacting serine threonine kinase 1 (RIPK1). This interaction is hypothesized to be inhibited at rest via an interaction between the C-terminus and the N-terminus which blocks the RIPK1 binding site. Here, we use a combination of two transition metal ion FRET methods to investigate the conformational dynamics of the termini while the channel is closed and desensitized. We do not find evidence that the termini are close enough to be bound while the channel is at rest and find that the termini may modestly move closer together when desensitized. At rest, the N-terminus adopts a conformation parallel to the membrane about 10 Å away. The distal end of the C-terminus may also spend time close to the membrane at rest. After acidification, the proximal portion of the N-terminus moves marginally closer to the membrane whereas the distal portion of the C-terminus swings away from the membrane. Together these data suggest that a new hypothesis for RIPK1 binding during stroke is needed.
Collapse
Affiliation(s)
- Megan M Cullinan
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Robert C Klipp
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Abigail Camenisch
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - John R Bankston
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
34
|
Yoshikawa M, Okubo M, Shirose K, Kan T, Kawaguchi M. d-Serine Increases Release of Acetylcholine in Rat Submandibular Glands. BIOLOGY 2023; 12:1227. [PMID: 37759626 PMCID: PMC10526048 DOI: 10.3390/biology12091227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/10/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023]
Abstract
d-serine has been observed in submandibular gland tissue in rats, but its functions remain to be clarified. Oral administration of d-serine, but not l-serine, increased its concentrations in the submandibular gland and pilocarpine-induced salivary secretion. In vivo microdialysis was used to collect the d- and l-enantiomers of amino acids from local interstitial fluid in the rat submandibular gland. The proportion of the d-form of serine in interstitial fluid was higher than that in plasma or saliva. Perfusion of the rat submandibular gland with d-serine and l-glutamic acid via the submandibular gland artery resulted in a significant increase in salivary secretion after stimulation of muscarinic receptors with carbachol. In vivo microdialysis applied to the submandibular glands of rats showed that infusion of d-serine along with l-glutamate through the microdialysis probe significantly elevated acetylcholine levels in local interstitial fluids in the submandibular glands of anesthetized rats as compared to that with l-glutamate alone in an N-methyl-d-aspartate receptor glycine site antagonist-sensitive manner. These results indicate that d-serine augments salivary secretion by increasing acetylcholine release in the salivary glands.
Collapse
Affiliation(s)
- Masanobu Yoshikawa
- Department of Clinical Pharmacology, School of Medicine, Tokai University, Isehara 259-1193, Japan
| | - Migiwa Okubo
- Kawano Dental Clinic, Yachimata 289-1101, Japan;
| | - Kosuke Shirose
- Department of Anesthesiology, School of Medicine, Tokai University, Isehara 259-1193, Japan; (K.S.); (T.K.)
| | - Takugi Kan
- Department of Anesthesiology, School of Medicine, Tokai University, Isehara 259-1193, Japan; (K.S.); (T.K.)
| | - Mitsuru Kawaguchi
- Tokyo Dental College, Misaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan;
| |
Collapse
|
35
|
Park G, Jin Z, Ge Q, Pan Y, Du J. Neuronal acid-sensing ion channel 1a regulates neuron-to-glioma synapses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.31.555794. [PMID: 37693494 PMCID: PMC10491214 DOI: 10.1101/2023.08.31.555794] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Neuronal activity promotes high-grade glioma progression via secreted proteins and neuron-to-glioma synapses, and glioma cells boost neuronal activity to further reinforce the malignant cycle. Whereas strong evidence supports that the activity of neuron-to-glioma synapses accelerates tumor progression, the molecular mechanisms that modulate the formation and function of neuron-to-glioma synapses remain largely unknown. Our recent findings suggest that a proton (H + ) signaling pathway actively mediates neuron-to-glioma synaptic communications by activating neuronal acid-sensing ion channel 1a (Asic1a), a predominant H + receptor in the central nervous system (CNS). Supporting this idea, our preliminary data revealed that local acid puff on neurons in high-grade glioma-bearing brain slices induces postsynaptic currents of glioma cells. Stimulating Asic1a knockout (Asic1a -/- ) neurons results in lower AMPA receptor-dependent excitatory postsynaptic currents (EPSCs) in glioma cells than stimulating wild-type (WT) neurons. Moreover, glioma-bearing Asic1a -/- mice exhibited reduced tumor size and survived longer than the glioma-bearing WT mice. Finally, pharmacologically targeting brain Asic1a inhibited high-grade glioma progression. In conclusion, our findings suggest that the neuronal H + -Asic1a axis plays a key role in regulating the neuron-glioma synapse. The outcomes of this study will greatly expand our understanding of how this deadly tumor integrates into the neuronal microenvironment.
Collapse
|
36
|
Pissas KP, Schilling M, Tian Y, Gründer S. Functional characterization of acid-sensing ion channels in the cerebellum-originating medulloblastoma cell line DAOY and in cerebellar granule neurons. Pflugers Arch 2023; 475:1073-1087. [PMID: 37474775 PMCID: PMC10409673 DOI: 10.1007/s00424-023-02839-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/03/2023] [Accepted: 07/09/2023] [Indexed: 07/22/2023]
Abstract
Acid-sensing ion channels (ASICs) are Na+ channels that are almost ubiquitously expressed in neurons of the brain. Functional ASIC1a is also expressed in glioblastoma stem cells, where it might sense the acidic tumor microenvironment. Prolonged acidosis induces cell death in neurons and reduces tumor sphere formation in glioblastoma via activation of ASIC1a. It is currently unknown whether ASICs are expressed and involved in acid-induced cell death in other types of brain tumors. In this study, we investigated ASICs in medulloblastoma, using two established cell lines, DAOY and UW228, as in vitro models. In addition, we characterized ASICs in the most numerous neuron of the brain, the cerebellar granule cell, which shares the progenitor cell with some forms of medulloblastoma. We report compelling evidence using RT-qPCR, western blot and whole-cell patch clamp that DAOY and cerebellar granule cells, but not UW228 cells, functionally express homomeric ASIC1a. Additionally, Ca2+-imaging revealed that extracellular acidification elevated intracellular Ca2+-levels in DAOY cells independently of ASICs. Finally, we show that overexpression of RIPK3, a key component of the necroptosis pathway, renders DAOY cells susceptible to acid-induced cell death via activation of ASIC1a. Our data support the idea that ASIC1a is an important acid sensor in brain tumors and that its activation has potential to induce cell death in tumor cells.
Collapse
Affiliation(s)
| | - Maria Schilling
- Institute of Physiology, RWTH Aachen University, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Yuemin Tian
- Institute of Physiology, RWTH Aachen University, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Stefan Gründer
- Institute of Physiology, RWTH Aachen University, Pauwelsstraße 30, 52074, Aachen, Germany.
| |
Collapse
|
37
|
Park G, Ge Q, Jin Z, Du J. Acid-sensing ion channel 1a contributes to the prefrontal cortex ischemia-enhanced neuronal activities in the amygdala. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.30.555556. [PMID: 37693395 PMCID: PMC10491201 DOI: 10.1101/2023.08.30.555556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Following a stroke, the emergence of amygdala-related disorders poses a significant challenge, with severe implications for post-stroke mental health, including conditions such as anxiety and depression. These disorders not only hinder post-stroke recovery but also elevate mortality rates. Despite their profound impact, the precise origins of aberrant amygdala function after stroke remain elusive. As a target of reduced brain pH in ischemia, acid-sensing ion channels (ASICs) have been implicated in synaptic transmission after ischemia, hinting at their potential role in reshaping neural circuits following a stroke. This study delves into the intriguing relationship between post-stroke alterations and ASICs, specifically focusing on postsynaptic ASIC1a enhancement in the amygdala following prefrontal cortex (PFC) ischemia induced by endothelin-1 (ET-1) injection. Our findings intriguingly illustrate that mPFC ischemia not only accentuates the PFC to amygdala circuit but also implicates ASIC1a in fostering augmented synaptic plasticity after ischemia. In contrast, the absence of ASIC1a impairs the heightened induction of long-term potentiation (LTP) in the amygdala induced by ischemia. This pivotal research introduces a novel concept with the potential to inaugurate an entirely new avenue of inquiry, thereby significantly enhancing our comprehension of the intricate mechanisms underlying post-stroke neural circuit reconfiguration. Importantly, these revelations hold the promise of paving the way for groundbreaking therapeutic interventions.
Collapse
|
38
|
Peng Z, Ziros PG, Martini T, Liao XH, Stoop R, Refetoff S, Albrecht U, Sykiotis GP, Kellenberger S. ASIC1a affects hypothalamic signaling and regulates the daily rhythm of body temperature in mice. Commun Biol 2023; 6:857. [PMID: 37591947 PMCID: PMC10435469 DOI: 10.1038/s42003-023-05221-2] [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: 11/21/2022] [Accepted: 08/05/2023] [Indexed: 08/19/2023] Open
Abstract
The body temperature of mice is higher at night than during the day. We show here that global deletion of acid-sensing ion channel 1a (ASIC1a) results in lower body temperature during a part of the night. ASICs are pH sensors that modulate neuronal activity. The deletion of ASIC1a decreased the voluntary activity at night of mice that had access to a running wheel but did not affect their spontaneous activity. Daily rhythms of thyrotropin-releasing hormone mRNA in the hypothalamus and of thyroid-stimulating hormone β mRNA in the pituitary, and of prolactin mRNA in the hypothalamus and pituitary were suppressed in ASIC1a-/- mice. The serum thyroid hormone levels were however not significantly changed by ASIC1a deletion. Our findings indicate that ASIC1a regulates activity and signaling in the hypothalamus and pituitary. This likely leads to the observed changes in body temperature by affecting the metabolism or energy expenditure.
Collapse
Affiliation(s)
- Zhong Peng
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Panos G Ziros
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Tomaz Martini
- Department of Biology/Unit of Biochemistry, Faculty of Sciences, University of Fribourg, Fribourg, Switzerland
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Xiao-Hui Liao
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Ron Stoop
- Center for Psychiatric Neurosciences, Hôpital de Cery, Lausanne University Hospital, Lausanne, Switzerland
| | - Samuel Refetoff
- Department of Medicine, The University of Chicago, Chicago, IL, USA
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA
- Committee on Genetics, The University of Chicago, Chicago, IL, USA
| | - Urs Albrecht
- Department of Biology/Unit of Biochemistry, Faculty of Sciences, University of Fribourg, Fribourg, Switzerland
| | - Gerasimos P Sykiotis
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Stephan Kellenberger
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
39
|
Fischer L, Schmidt A, Dopychai A, Joussen S, Joeres N, Oslender-Bujotzek A, Schmalzing G, Gründer S. Physiologically relevant acid-sensing ion channel (ASIC) 2a/3 heteromers have a 1:2 stoichiometry. Commun Biol 2023; 6:701. [PMID: 37422581 PMCID: PMC10329638 DOI: 10.1038/s42003-023-05087-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 06/29/2023] [Indexed: 07/10/2023] Open
Abstract
Acid-sensing ion channels (ASICs) sense extracellular protons and are involved in synaptic transmission and pain sensation. ASIC1a and ASIC3 are the ASIC subunits with the highest proton sensitivity. ASIC2a in contrast has low proton sensitivity but increases the variability of ASICs by forming heteromers with ASIC1a or ASIC3. ASICs are trimers and for the ASIC1a/2a heteromer it has been shown that subunits randomly assemble with a flexible 1:2/2:1 stoichiometry. Both heteromers have almost identical proton sensitivity intermediate between ASIC1a and ASIC2a. Here, we investigated the stoichiometry of the ASIC2a/3 heteromer. Using electrophysiology, we extensively characterized, first, cells expressing ASIC2a and ASIC3 at different ratios, second, concatemeric channels with a fixed subunit stoichiometry, and, third, channels containing loss-of-functions mutations in specific subunits. Our results conclusively show that only ASIC2a/3 heteromers with a 1:2 stoichiometry had a proton-sensitivity intermediate between ASIC2a and ASIC3. In contrast, the proton sensitivity of ASIC2a/3 heteromers with a 2:1 stoichiometry was strongly acid-shifted by more than one pH unit, which suggests that they are not physiologically relevant. Together, our results reveal that the proton sensitivity of the two ASIC2a/3 heteromers is clearly different and that ASIC3 and ASIC1a make remarkably different contributions to heteromers with ASIC2a.
Collapse
Affiliation(s)
- Leon Fischer
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, D-52074, Aachen, Germany
- Department of Anesthesiology, Technical University Dresden, Dresden, Germany
| | - Axel Schmidt
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, D-52074, Aachen, Germany
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Anke Dopychai
- Institute of Clinical Pharmacology, RWTH Aachen University, Wendlingweg, D-52074, Aachen, Germany
| | - Sylvia Joussen
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, D-52074, Aachen, Germany
| | - Niko Joeres
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, D-52074, Aachen, Germany
- Department of Nephrology and Clinical Immunology, RWTH Aachen University, Pauwelsstrasse 30, D-52074, Aachen, Germany
| | | | - Günther Schmalzing
- Institute of Clinical Pharmacology, RWTH Aachen University, Wendlingweg, D-52074, Aachen, Germany
| | - Stefan Gründer
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, D-52074, Aachen, Germany.
| |
Collapse
|
40
|
Yang Y, Jin S, Zhang J, Chen W, Lu Y, Chen J, Yan Z, Shen B, Ning Y, Shi Y, Chen J, Wang J, Xu S, Jia P, Teng J, Fang Y, Song N, Ding X. Acid-sensing ion channel 1a exacerbates renal ischemia-reperfusion injury through the NF-κB/NLRP3 inflammasome pathway. J Mol Med (Berl) 2023; 101:877-890. [PMID: 37246982 PMCID: PMC10300185 DOI: 10.1007/s00109-023-02330-7] [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: 05/02/2022] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/30/2023]
Abstract
Ischemia-reperfusion injury (IRI) is the main cause of acute kidney injury (AKI), and there is no effective therapy. Microenvironmental acidification is generally observed in ischemic tissues. Acid-sensing ion channel 1a (ASIC1a) can be activated by a decrease in extracellular pH which mediates neuronal IRI. Our previous study demonstrated that, ASIC1a inhibition alleviates renal IRI. However, the underlying mechanisms have not been fully elucidated. In this study, we determined that renal tubule-specific deletion of ASIC1a in mice (ASIC1afl/fl/CDH16cre) attenuated renal IRI, and reduced the expression of NLRP3, ASC, cleaved-caspase-1, GSDMD-N, and IL-1β. Consistent with these in vivo results, inhibition of ASIC1a by the specific inhibitor PcTx-1 protected HK-2 cells from hypoxia/reoxygenation (H/R) injury, and suppressed H/R-induced NLRP3 inflammasome activation. Mechanistically, the activation of ASIC1a by either IRI or H/R induced the phosphorylation of NF-κB p65, which translocates to the nucleus and promotes the transcription of NLRP3 and pro-IL-1β. Blocking NF-κB by treatment with BAY 11-7082 validated the roles of H/R and acidosis in NLRP3 inflammasome activation. This further confirmed that ASIC1a promotes NLRP3 inflammasome activation, which requires the NF-κB pathway. In conclusion, our study suggests that ASIC1a contributes to renal IRI by affecting the NF-κB/NLRP3 inflammasome pathway. Therefore, ASIC1a may be a potential therapeutic target for AKI. KEY MESSAGES: Knockout of ASIC1a attenuated renal ischemia-reperfusion injury. ASIC1a promoted the NF-κB pathway and NLRP3 inflammasome activation. Inhibition of the NF-κB mitigated the NLRP3 inflammasome activation induced by ASIC1a.
Collapse
Affiliation(s)
- Yan Yang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Shi Jin
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Jian Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Weize Chen
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Yufei Lu
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Jun Chen
- Department of Pathology, Changzheng Hospital, Naval Military Medical University, Shanghai, China
| | - Zhixin Yan
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Bo Shen
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Yichun Ning
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Yiqin Shi
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Jing Chen
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Jialin Wang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Sujuan Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Ping Jia
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Jie Teng
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Yi Fang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China
| | - Nana Song
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China.
- Fudan Zhangjiang Institute, Shanghai, China.
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney; Shanghai Institute of Kidney and Dialysis; Shanghai Key Laboratory of Kidney and Blood Purification; Hemodialysis quality control center of Shanghai, Shanghai, 200032, China.
| |
Collapse
|
41
|
López-Ramírez O, González-Garrido A. The role of acid sensing ion channels in the cardiovascular function. Front Physiol 2023; 14:1194948. [PMID: 37389121 PMCID: PMC10300344 DOI: 10.3389/fphys.2023.1194948] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/05/2023] [Indexed: 07/01/2023] Open
Abstract
Acid Sensing Ion Channels (ASIC) are proton sensors involved in several physiological and pathophysiological functions including synaptic plasticity, sensory systems and nociception. ASIC channels have been ubiquitously localized in neurons and play a role in their excitability. Information about ASIC channels in cardiomyocyte function is limited. Evidence indicates that ASIC subunits are expressed in both, plasma membrane and intracellular compartments of mammalian cardiomyocytes, suggesting unrevealing functions in the cardiomyocyte physiology. ASIC channels are expressed in neurons of the peripheral nervous system including the nodose and dorsal root ganglia (DRG), both innervating the heart, where they play a dual role as mechanosensors and chemosensors. In baroreceptor neurons from nodose ganglia, mechanosensation is directly associated with ASIC2a channels for detection of changes in arterial pressure. ASIC channels expressed in DRG neurons have several roles in the cardiovascular function. First, ASIC2a/3 channel has been proposed as the molecular sensor of cardiac ischemic pain for its pH range activation, kinetics and the sustained current. Second, ASIC1a seems to have a critical role in ischemia-induced injury. And third, ASIC1a, 2 and 3 are part of the metabolic component of the exercise pressure reflex (EPR). This review consists of a summary of several reports about the role of ASIC channels in the cardiovascular system and its innervation.
Collapse
Affiliation(s)
- Omar López-Ramírez
- Instituto de Oftalmología Fundación de Asistencia Privada Conde de Valenciana, I.A.P., Mexico City, Mexico
| | - Antonia González-Garrido
- Laboratorio de Enfermedades Mendelianas, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| |
Collapse
|
42
|
Rajeev V, Chai YL, Poh L, Selvaraji S, Fann DY, Jo DG, De Silva TM, Drummond GR, Sobey CG, Arumugam TV, Chen CP, Lai MKP. Chronic cerebral hypoperfusion: a critical feature in unravelling the etiology of vascular cognitive impairment. Acta Neuropathol Commun 2023; 11:93. [PMID: 37309012 PMCID: PMC10259064 DOI: 10.1186/s40478-023-01590-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/14/2023] Open
Abstract
Vascular cognitive impairment (VCI) describes a wide spectrum of cognitive deficits related to cerebrovascular diseases. Although the loss of blood flow to cortical regions critically involved in cognitive processes must feature as the main driver of VCI, the underlying mechanisms and interactions with related disease processes remain to be fully elucidated. Recent clinical studies of cerebral blood flow measurements have supported the role of chronic cerebral hypoperfusion (CCH) as a major driver of the vascular pathology and clinical manifestations of VCI. Here we review the pathophysiological mechanisms as well as neuropathological changes of CCH. Potential interventional strategies for VCI are also reviewed. A deeper understanding of how CCH can lead to accumulation of VCI-associated pathology could potentially pave the way for early detection and development of disease-modifying therapies, thus allowing preventive interventions instead of symptomatic treatments.
Collapse
Affiliation(s)
- Vismitha Rajeev
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Yuek Ling Chai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Luting Poh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Sharmelee Selvaraji
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, Singapore
| | - David Y Fann
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - T Michael De Silva
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Grant R Drummond
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Thiruma V Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
- NUS Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore.
- NUS Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
43
|
Ge H, Zhou T, Zhang C, Cun Y, Chen W, Yang Y, Zhang Q, Li H, Zhong J, Zhang X, Feng H, Hu R. Targeting ASIC1a Promotes Neural Progenitor Cell Migration and Neurogenesis in Ischemic Stroke. RESEARCH (WASHINGTON, D.C.) 2023; 6:0105. [PMID: 37275123 PMCID: PMC10234266 DOI: 10.34133/research.0105] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/13/2023] [Indexed: 06/07/2023]
Abstract
Cell replacement therapy using neural progenitor cells (NPCs) has been shown to be an effective treatment for ischemic stroke. However, the therapeutic effect is unsatisfactory due to the imbalanced homeostasis of the local microenvironment after ischemia. Microenvironmental acidosis is a common imbalanced homeostasis in the penumbra and could activate acid-sensing ion channels 1a (ASIC1a), a subunit of proton-gated cation channels following ischemic stroke. However, the role of ASIC1a in NPCs post-ischemia remains elusive. Here, our results indicated that ASIC1a was expressed in NPCs with channel functionality, which could be activated by extracellular acidification. Further evidence revealed that ASIC1a activation inhibited NPC migration and neurogenesis through RhoA signaling-mediated reorganization of filopodia formation, which could be primarily reversed by pharmacological or genetic disruption of ASIC1a. In vivo data showed that the knockout of the ASIC1a gene facilitated NPC migration and neurogenesis in the penumbra to improve behavioral recovery after stroke. Subsequently, ASIC1a gain of function partially abrogated this effect. Moreover, the administration of ASIC1a antagonists (amiloride or Psalmotoxin 1) promoted functional recovery by enhancing NPC migration and neurogenesis. Together, these results demonstrate targeting ASIC1a is a novel strategy potentiating NPC migration toward penumbra to repair lesions following ischemic stroke and even for other neurological diseases with the presence of niche acidosis.
Collapse
Affiliation(s)
- Hongfei Ge
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
- Medical Research Center, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Tengyuan Zhou
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
- Medical Research Center, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Chao Zhang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
- Medical Research Center, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Yupeng Cun
- Pediatric Research Institute,
Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, 400014 Chongqing, China
| | - Weixiang Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Yang Yang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Qian Zhang
- Medical Research Center, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Huanhuan Li
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Jun Zhong
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Xuyang Zhang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Hua Feng
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Rong Hu
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
- Medical Research Center, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| |
Collapse
|
44
|
Cao Y, Redd MA, Fang C, Mizikovsky D, Li X, Macdonald PS, King GF, Palpant NJ. New Drug Targets and Preclinical Modelling Recommendations for Treating Acute Myocardial Infarction. Heart Lung Circ 2023:S1443-9506(23)00139-7. [PMID: 37230806 DOI: 10.1016/j.hlc.2022.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/28/2022] [Accepted: 12/15/2022] [Indexed: 05/27/2023]
Abstract
Acute myocardial infarction (AMI) is the leading cause of morbidity and mortality worldwide and the primary underlying risk factor for heart failure. Despite decades of research and clinical trials, there are no drugs currently available to prevent organ damage from acute ischaemic injuries of the heart. In order to address the increasing global burden of heart failure, drug, gene, and cell-based regeneration technologies are advancing into clinical testing. In this review we highlight the burden of disease associated with AMI and the therapeutic landscape based on market analyses. New studies revealing the role of acid-sensitive cardiac ion channels and other proton-gated ion channels in cardiac ischaemia are providing renewed interest in pre- and post-conditioning agents with novel mechanisms of action that may also have implications for gene- and cell-based therapeutics. Furthermore, we present guidelines that couple new cell technologies and data resources with traditional animal modelling pipelines to help de-risk drug candidates aimed at treating AMI. We propose that improved preclinical pipelines and increased investment in drug target identification for AMI is critical to stem the increasing global health burden of heart failure.
Collapse
Affiliation(s)
- Yuanzhao Cao
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Meredith A Redd
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Chen Fang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Dalia Mizikovsky
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Xichun Li
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Peter S Macdonald
- Cardiopulmonary Transplant Unit, St Vincent's Hospital, Sydney, NSW, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Qld, Australia
| | - Nathan J Palpant
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia.
| |
Collapse
|
45
|
Cherninskyi A, Storozhuk M, Maximyuk O, Kulyk V, Krishtal O. Triggering of Major Brain Disorders by Protons and ATP: The Role of ASICs and P2X Receptors. Neurosci Bull 2023; 39:845-862. [PMID: 36445556 PMCID: PMC9707125 DOI: 10.1007/s12264-022-00986-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 08/14/2022] [Indexed: 11/30/2022] Open
Abstract
Adenosine triphosphate (ATP) is well-known as a universal source of energy in living cells. Less known is that this molecule has a variety of important signaling functions: it activates a variety of specific metabotropic (P2Y) and ionotropic (P2X) receptors in neuronal and non-neuronal cell membranes. So, a wide variety of signaling functions well fits the ubiquitous presence of ATP in the tissues. Even more ubiquitous are protons. Apart from the unspecific interaction of protons with any protein, many physiological processes are affected by protons acting on specific ionotropic receptors-acid-sensing ion channels (ASICs). Both protons (acidification) and ATP are locally elevated in various pathological states. Using these fundamentally important molecules as agonists, ASICs and P2X receptors signal a variety of major brain pathologies. Here we briefly outline the physiological roles of ASICs and P2X receptors, focusing on the brain pathologies involving these receptors.
Collapse
Affiliation(s)
- Andrii Cherninskyi
- Bogomoletz Institute of Physiology of National Academy of Sciences of Ukraine, Kyiv, 01024, Ukraine.
| | - Maksim Storozhuk
- Bogomoletz Institute of Physiology of National Academy of Sciences of Ukraine, Kyiv, 01024, Ukraine
| | - Oleksandr Maximyuk
- Bogomoletz Institute of Physiology of National Academy of Sciences of Ukraine, Kyiv, 01024, Ukraine
| | - Vyacheslav Kulyk
- Bogomoletz Institute of Physiology of National Academy of Sciences of Ukraine, Kyiv, 01024, Ukraine
| | - Oleg Krishtal
- Bogomoletz Institute of Physiology of National Academy of Sciences of Ukraine, Kyiv, 01024, Ukraine
| |
Collapse
|
46
|
Hulme J. COVID-19 and Diarylamidines: The Parasitic Connection. Int J Mol Sci 2023; 24:6583. [PMID: 37047556 PMCID: PMC10094973 DOI: 10.3390/ijms24076583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
As emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants (Omicron) continue to outpace and negate combinatorial vaccines and monoclonal antibody therapies targeting the spike protein (S) receptor binding domain (RBD), the appetite for developing similar COVID-19 treatments has significantly diminished, with the attention of the scientific community switching to long COVID treatments. However, treatments that reduce the risk of "post-COVID-19 syndrome" and associated sequelae remain in their infancy, particularly as no established criteria for diagnosis currently exist. Thus, alternative therapies that reduce infection and prevent the broad range of symptoms associated with 'post-COVID-19 syndrome' require investigation. This review begins with an overview of the parasitic-diarylamidine connection, followed by the renin-angiotensin system (RAS) and associated angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSSR2) involved in SARS-CoV-2 infection. Subsequently, the ability of diarylamidines to inhibit S-protein binding and various membrane serine proteases associated with SARS-CoV-2 and parasitic infections are discussed. Finally, the roles of diarylamidines (primarily DIZE) in vaccine efficacy, epigenetics, and the potential amelioration of long COVID sequelae are highlighted.
Collapse
Affiliation(s)
- John Hulme
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Sungnam-daero, Sujung-gu, Seongnam-si 461-701, Republic of Korea
| |
Collapse
|
47
|
Animal toxins: As an alternative therapeutic target following ischemic stroke condition. Life Sci 2023; 317:121365. [PMID: 36640901 DOI: 10.1016/j.lfs.2022.121365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 11/29/2022] [Accepted: 12/31/2022] [Indexed: 01/13/2023]
Abstract
Globally, Ischemic stroke (IS) has become the second leading cause of mortality and chronic disability. The process of IS has triggered by the blockages of blood vessels to form clots in the brain which initiates multiple interactions with the key signaling pathways, counting excitotoxicity, acidosis, ionic imbalance, inflammation, oxidative stress, and neuronal dysfunction of cells, and ultimately cells going under apoptosis. Currently, FDA has approved only tissue plasminogen activator therapy, which is effective against IS with few limitations. However, the mechanism of excitotoxicity and acidosis has spurred the investigation of a potential candidate for IS therapy. Acid-sensing ion channels (ASICs) and Voltage-gated Ca2+ channels (VDCCs) get activated and disturb the brain's normal physiology. Animal toxins are novel inhibitors of ASICs and VDCCs channels and have provided neuroprotective insights into the pathophysiology of IS. This review will discuss the potential directions of translational ASICs and VDCCs inhibitors research for clinical therapies.
Collapse
|
48
|
Sun HW, Chu XP, Simon RP, Xiong ZG, Leng TD. Inhibition of Acid-Sensing Ion Channels by KB-R7943, a Reverse Na+/Ca2+ Exchanger Inhibitor. Biomolecules 2023; 13:biom13030507. [PMID: 36979442 PMCID: PMC10046550 DOI: 10.3390/biom13030507] [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: 01/05/2023] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
KB-R7943, an isothiourea derivative, is widely used as a pharmacological inhibitor of reverse sodium–calcium exchanger (NCX). It has been shown to have neuroprotective and analgesic effects in animal models; however, the detailed molecular mechanisms remain elusive. In the current study, we investigated whether KB-R7943 modulates acid-sensing ion channels (ASICs), a group of proton-gated cation channels implicated in the pathophysiology of various neurological disorders, using the whole-cell patch clamp techniques. Our data show that KB-R7943 irreversibly inhibits homomeric ASIC1a channels heterologously expressed in Chinese Hamster Ovary (CHO) cells in a use- and concentration-dependent manner. It also reversibly inhibits homomeric ASIC2a and ASIC3 channels in CHO cells. Both the transient and sustained current components of ASIC3 are inhibited. Furthermore, KB-R7943 inhibits ASICs in primary cultured peripheral and central neurons. It inhibits the ASIC-like currents in mouse dorsal root ganglion (DRG) neurons and the ASIC1a-like currents in mouse cortical neurons. The inhibition of the ASIC1a-like current is use-dependent and unrelated to its effect on NCX since neither of the other two well-characterized NCX inhibitors, including SEA0400 and SN-6, shows an effect on ASIC. Our data also suggest that the isothiourea group, which is lacking in other structurally related analogs that do not affect ASIC1a-like current, may serve as a critical functional group. In summary, we characterize KB-R7943 as a new ASIC inhibitor. It provides a novel pharmacological tool for the investigation of the functions of ASICs and could serve as a lead compound for developing small-molecule drugs for treating ASIC-related disorders.
Collapse
Affiliation(s)
- Hua-Wei Sun
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Xiang-Ping Chu
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Roger P. Simon
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Zhi-Gang Xiong
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Tian-Dong Leng
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
- Correspondence:
| |
Collapse
|
49
|
Garcia SM, Naik JS, Resta TC, Jernigan NL. Acid-sensing ion channel 1a activates IKCa/SKCa channels and contributes to endothelium-dependent dilation. J Gen Physiol 2023; 155:e202213173. [PMID: 36484717 PMCID: PMC9984545 DOI: 10.1085/jgp.202213173] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 10/21/2022] [Accepted: 11/17/2022] [Indexed: 12/13/2022] Open
Abstract
Acid-sensing ion channel 1a (ASIC1a) belongs to a novel family of proton-gated cation channels that are permeable to both Na+ and Ca2+. ASIC1a is expressed in vascular smooth muscle and endothelial cells in a variety of vascular beds, yet little is known regarding the potential impact of ASIC1a to regulate local vascular reactivity. Our previous studies in rat mesenteric arteries suggest ASIC1a does not contribute to agonist-induced vasoconstriction but may mediate a vasodilatory response. The objective of the current study is to determine the role of ASIC1a in systemic vasodilatory responses by testing the hypothesis that the activation of endothelial ASIC1a mediates vasodilation of mesenteric resistance arteries through an endothelium-dependent hyperpolarization (EDH)-related pathway. The selective ASIC1a antagonist psalmotoxin 1 (PcTX1) largely attenuated the sustained vasodilatory response to acetylcholine (ACh) in isolated, pressurized mesenteric resistance arteries and ACh-mediated Ca2+ influx in freshly isolated mesenteric endothelial tubes. Similarly, basal tone was enhanced and ACh-induced vasodilation blunted in mesenteric arteries from Asic1a knockout mice. ASIC1a colocalizes with intermediate- and small-conductance Ca2+-activated K+ channels (IKCa and SKCa, respectively), and the IKCa/SKCa-sensitive component of the ACh-mediated vasodilation was blocked by ASIC1a inhibition. To determine the role of ASIC1a to activate IKCa/SKCa channels, we measured whole-cell K+ currents using the perforated-patch clamp technique in freshly isolated mesenteric endothelial cells. Inhibition of ASIC1a prevented ACh-induced activation of IKCa/SKCa channels. The ASIC1 agonist, α/β-MitTx, activated IKCa/SKCa channels and induced an IKCa/SKCa-dependent vasodilation. Together, the present study demonstrates that ASIC1a couples to IKCa/SKCa channels in mesenteric resistance arteries to mediate endothelium-dependent vasodilation.
Collapse
Affiliation(s)
- Selina M. Garcia
- Department of Cell Biology and Physiology University of New Mexico School of Medicine, Albuquerque, NM
| | - Jay S. Naik
- Department of Cell Biology and Physiology University of New Mexico School of Medicine, Albuquerque, NM
| | - Thomas C. Resta
- Department of Cell Biology and Physiology University of New Mexico School of Medicine, Albuquerque, NM
| | - Nikki L. Jernigan
- Department of Cell Biology and Physiology University of New Mexico School of Medicine, Albuquerque, NM
| |
Collapse
|
50
|
The Role of Zinc in Modulating Acid-Sensing Ion Channel Function. Biomolecules 2023; 13:biom13020229. [PMID: 36830598 PMCID: PMC9953155 DOI: 10.3390/biom13020229] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
Acid-sensing ion channels (ASICs) are proton-gated, voltage-independent sodium channels widely expressed throughout the central and peripheral nervous systems. They are involved in synaptic plasticity, learning/memory, fear conditioning and pain. Zinc, an important trace metal in the body, contributes to numerous physiological functions, with neurotransmission being of note. Zinc has been implicated in the modulation of ASICs by binding to specific sites on these channels and exerting either stimulatory or inhibitory effects depending on the ASIC subtype. ASICs have been linked to several neurological and psychological disorders, such as Alzheimer's disease, Parkinson's disease, ischemic stroke, epilepsy and cocaine addiction. Different ASIC isoforms contribute to the persistence of each of these neurological and psychological disorders. It is critical to understand how various zinc concentrations can modulate specific ASIC subtypes and how zinc regulation of ASICs can contribute to neurological and psychological diseases. This review elucidates zinc's structural interactions with ASICs and discusses the potential therapeutic implications zinc may have on neurological and psychological diseases through targeting ASICs.
Collapse
|