Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Molecules ; 29(7)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38611859

ABSTRACT

A novel Lycopodium alkaloid, lycocasine A (1), and seven known Lycopodium alkaloids (2-8), were isolated from Lycopodiastrum casuarinoides. Their structures were determined through NMR, HRESIMS, and X-ray diffraction analysis. Compound 1 features an unprecedented 5/6/6 tricyclic skeleton, highlighted by a 5-aza-tricyclic[6,3,1,02,6]dodecane motif. In bioactivity assays, compound 1 demonstrated weak inhibitory activity against acid-sensing ion channel 1a.


Subject(s)
Alkaloids , Lycopodiaceae , Lycopodium , Acid Sensing Ion Channels , Alkaloids/pharmacology , Azacitidine
2.
Heart Lung Circ ; 32(7): 852-869, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37230806

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.


Subject(s)
Heart Failure , Myocardial Infarction , Myocardial Reperfusion Injury , Animals , Myocardial Reperfusion Injury/prevention & control , Myocardial Infarction/drug therapy , Heart , Heart Failure/prevention & control
3.
J Mol Med (Berl) ; 101(7): 877-890, 2023 07.
Article in English | MEDLINE | ID: mdl-37246982

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-1ß. 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-1ß. 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.


Subject(s)
Acute Kidney Injury , Reperfusion Injury , Mice , Animals , Inflammasomes/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Acid Sensing Ion Channels/genetics , Acid Sensing Ion Channels/metabolism , Mice, Knockout , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism
4.
Neuropsychiatr Dis Treat ; 18: 2181-2198, 2022.
Article in English | MEDLINE | ID: mdl-36187562

ABSTRACT

Background: The ketogenic diet (KD) is a proven therapy for refractory epilepsy. Although the anti-seizure properties of this diet are understood to a certain extent, the exploration of its neuroprotective effects and underlying mechanisms is still in its infancy. Tissue acidosis is a common feature of epileptogenic foci. Interestingly, the activation of acid-sensing ion channel 1a (ASIC1a), which mediates Ca2+-dependent neuronal injury during acidosis, has been found to be inhibited by ketone bodies in vitro. This prompted us to investigate whether the neuroprotective effects induced by the KD occur via ASIC1a and interconnected downstream mechanisms in a rat model of temporal lobe epilepsy. Methods: Male Sprague-Dawley rats were fed either the KD or a normal diet for four weeks after undergoing pilocarpine-induced status epilepticus (SE). The effects of KD on epileptogenesis, cognitive impairment and hippocampal neuron injury in the epileptic rats were subsequently evaluated by video electroencephalogram, Morris water maze test and Nissl staining, respectively. The expression of ASIC1a and cleaved caspase-3 in the hippocampus were determined using Western blot analysis during the chronic period following SE. Moreover, the intracellular Ca2+ concentration, mitochondrial membrane potential (MMP), mitochondrial reactive oxygen species (mROS) and cell apoptosis of hippocampal cells were detected by flow cytometry. Results: We found that the KD treatment strongly attenuated the spontaneous recurrent seizures, ameliorated learning and memory impairments and prevented hippocampal neuronal injury and apoptosis. The KD was also shown to inhibit the upregulation of ASIC1a and the ensuing intracellular Ca2+ overload in the hippocampus of the epileptic rats. Furthermore, the seizure-induced structure disruption of neuronal mitochondria, loss of MMP and accumulation of mROS were reversed by the KD treatment, suggesting that it has protective effects on mitochondria. Finally, the activation of caspase-3 was also inhibited by the KD. Conclusion: These findings indicate that the KD suppresses mitochondria-mediated apoptosis possibly by regulating ASIC1a to exert neuroprotective effects. This may provide a mechanistic explanation of the therapeutic effects of KD.

5.
Int J Biol Macromol ; 211: 128-139, 2022 Jun 30.
Article in English | MEDLINE | ID: mdl-35561854

ABSTRACT

Activation of hepatic stellate cells (HSCs) is a key cause of liver fibrosis. However, the mechanisms leading to the activation of HSCs are not fully understood. In the pathological process, acid-sensing ion channel 1a (ASIC1a) is widely involved in the development of inflammatory diseases, suggesting that ASIC1a may play an important role in liver fibrosis. We found that in an acidic environment, ASIC1a leads to HSC-T6 cell activation. Meanwhile, exosomes produced by activated HSC-T6 cells (HSC-EXOs) can be reabsorbed by quiescent HSC-T6 cells to promote their activation. Exosomes mainly carry miRNAs involved in intercellular information exchange. We performed exosome miRNA whole transcriptome sequencing. The results indicated that the acidic environment could alter the miRNA expression profile in the exosomes of HSC-T6 cells. Further studies revealed that ASIC1a promotes the activation of HSCs by regulating miR-301a-3p targeting B-cell translocation gene 1 (BTG1). In conclusion, our study found that ASIC1a may affect HSC activation through the exosomal miR-301a-3p/BTG1 axis, and inhibiting ASIC1a may be a promising treatment strategy for liver fibrosis.


Subject(s)
Acid Sensing Ion Channels/metabolism , Hepatic Stellate Cells/metabolism , MicroRNAs , Acid Sensing Ion Channels/genetics , Animals , Cell Line , Exosomes/genetics , Exosomes/metabolism , Humans , Liver Cirrhosis/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Rats
6.
J Cell Mol Med ; 26(10): 2777-2792, 2022 05.
Article in English | MEDLINE | ID: mdl-35426224

ABSTRACT

Multidrug resistance is the main obstacle to curing hepatocellular carcinoma (HCC). Acid-sensing ion channel 1a (ASIC1a) has critical roles in all stages of cancer progression, especially invasion and metastasis, and in resistance to therapy. Epithelial to mesenchymal transition (EMT) transforms epithelial cells into mesenchymal cells after being stimulated by extracellular factors and is closely related to tumour infiltration and resistance. We used Western blotting, immunofluorescence, qRT-PCR, immunohistochemical staining, MTT, colony formation and scratch healing assay to determine ASIC1a levels and its relationship to cell proliferation, migration and invasion. ASIC1a is overexpressed in HCC tissues, and the amount increased in resistant HCC cells. EMT occurred more frequently in drug-resistant cells than in parental cells. Inactivation of ASIC1a inhibited cell migration and invasion and increased the chemosensitivity of cells through EMT. Overexpression of ASIC1a upregulated EMT and increased the cells' proliferation, migration and invasion and induced drug resistance; knocking down ASIC1a with shRNA had the opposite effects. ASIC1a increased cell migration and invasion through EMT by regulating α and ß-catenin, vimentin and fibronectin expression via the AKT/GSK-3ß/Snail pathway driven by TGFß/Smad signals. ASIC1a mediates drug resistance of HCC through EMT via the AKT/GSK-3ß/Snail pathway.


Subject(s)
Acid Sensing Ion Channels , Carcinoma, Hepatocellular , Epithelial-Mesenchymal Transition , Liver Neoplasms , Acid Sensing Ion Channels/genetics , Acid Sensing Ion Channels/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Liver Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
8.
Front Cell Dev Biol ; 8: 596304, 2020.
Article in English | MEDLINE | ID: mdl-33195276

ABSTRACT

Acid-sensing ion channels (ASICs) are members of the degenerin/epithelial sodium channel superfamily. They are extracellular pH sensors that are activated by protons. Among all ASICs, ASIC1a is one of the most intensively studied isoforms because of its unique ability to be permeable to Ca2+. In addition, it is considered to contribute to various pathophysiological conditions. As a membrane proton receptor, the number of ASIC1a present on the cell surface determines its physiological and pathological functions, and this number partially depends on protein synthesis, degradation, and membrane trafficking processes. Recently, several studies have shown that various factors affect these processes. Therefore, this review elucidated the major factors and underlying molecular mechanisms affecting ASIC1a protein expression and membrane trafficking.

9.
Cell Biol Int ; 44(10): 2140-2152, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32678496

ABSTRACT

The pyroptosis is a causative agent of rheumatoid arthritis, a systemic autoimmune disease merged with degenerative articular cartilage. Nevertheless, the precise mechanism of extracellular acidosis on chondrocyte pyroptosis is largely unclear. Acid-sensing ion channels (ASICs) belong to an extracellular H+ -activated cation channel family. Accumulating evidence has highlighted activation of ASICs induced by extracellular acidosis upregulate calpain and calcineurin expression in arthritis. In the present study, to investigate the expression and the role of acid-sensing ion channel 1a (ASIC1a), calpain, calcineurin, and NLRP3 inflammasome proteins in regulating acid-induced articular chondrocyte pyroptosis, primary rat articular chondrocytes were subjected to different pH, different time, and different treatments with or without ASIC1a, calpain-2, and calcineurin, respectively. Initially, the research results showed that extracellular acidosis-induced the protein expression of ASIC1a in a pH- and time-dependent manner, and the messenger RNA and protein expressions of calpain, calcineurin, NLRP3, apoptosis-associated speck-like protein, and caspase-1 were significantly increased in a time-dependent manner. Furthermore, the inhibition of ASIC1a, calpain-2, or calcineurin, respectively, could decrease the cell death accompanied with the decreased interleukin-1ß level, and the decreased expression of ASIC1a, calpain-2, calcineurin, and NLRP3 inflammasome proteins. Taken together, these results indicated the activation of ASIC1a induced by extracellular acidosis could trigger pyroptosis of rat articular chondrocytes, the mechanism of which might partly be involved with the activation of calpain-2/calcineurin pathway.


Subject(s)
Acid Sensing Ion Channels/physiology , Arthritis, Experimental , Calcineurin/metabolism , Calpain/metabolism , Chondrocytes , Pyroptosis , Animals , Arthritis, Experimental/mortality , Arthritis, Experimental/pathology , Cells, Cultured , Chondrocytes/metabolism , Chondrocytes/pathology , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Rats , Rats, Sprague-Dawley
10.
Transl Cancer Res ; 9(12): 7519-7530, 2020 Dec.
Article in English | MEDLINE | ID: mdl-35117352

ABSTRACT

BACKGROUND: The microenvironment of various tumor tissues is acidic. Acid-sensing ion channels (ASICs) are a class of ligand-gated ion channels which are sensitive to extracellular protons and are often highly expressed in tumor tissues. Breast cancer, whose extracellular microenvironment is thought to be acidic, is the most common cancer type among females in the world. METHODS: Thirty breast cancer tissues and adjacent normal tissues of patients were collected from 2009 to 2015 at the Xinhua hospital affiliated to Shanghai Jiao Tong University School of Medicine. The expression of acid-sensing ion channel 1a (ASIC1a), a subtype of ASICs family, was detected by immunohistochemistry in breast cancer tissues, and the effect of ASIC1a on the physiological activity of tumor cells was analyzed in vitro and in vivo experiments. RESULTS: In this study, it was found that ASIC1a is highly expressed in the tissues of breast cancer patients. In vitro experiments revealed that down-regulation of ASIC1a by its antagonist PcTx-1 or ASIC1a siRNA could significantly weaken the migration, proliferation and invasion of tumor cells. In vivo studies, down-regulation or inhibition of the ASIC1a could inhibit breast tumor growth. CONCLUSIONS: The high expression of ASIC1a might be related to the enhanced biological activity of breast cancer cells. Whether ASIC1a is a potential therapeutic target for some types of breast cancer deserves further study.

11.
Front Immunol ; 11: 580936, 2020.
Article in English | MEDLINE | ID: mdl-33584647

ABSTRACT

Acid-sensing ion channel 1a (ASIC1a) is a member of the extracellular H+-activated cation channel family. Emerging evidence has suggested that ASIC1a plays a crucial role in the pathogenesis of rheumatoid arthritis (RA). Specifically, ASIC1a could promote inflammation, synovial hyperplasia, articular cartilage, and bone destruction; these lead to the progression of RA, a chronic autoimmune disease characterized by chronic synovial inflammation and extra-articular lesions. In this review, we provided a brief overview of the molecular properties of ASIC1a, including the basic biological characteristics, tissue and cell distribution, channel blocker, and factors influencing the expression and function, and focused on the potential therapeutic targets of ASIC1a in RA and possible mechanisms of blocking ASIC1a to improve RA symptoms, such as regulation of apoptosis, autophagy, pyroptosis, and necroptosis of articular cartilage, and synovial inflammation and invasion of fibroblast-like cells in synovial tissue.


Subject(s)
Acid Sensing Ion Channel Blockers/pharmacology , Acid Sensing Ion Channels/metabolism , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Synovial Membrane/metabolism , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Cartilage, Articular/pathology , Chondrocytes/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Models, Biological , Rats , Regulated Cell Death/physiology , Signal Transduction , Synovial Membrane/pathology
12.
Biosci Rep ; 39(11)2019 11 29.
Article in English | MEDLINE | ID: mdl-31696219

ABSTRACT

PURPOSE: In the degenerated intervertebral disc (IVD), matrix acidity challenges transplanted bone marrow mesenchymal stem cells (BMSCs). The Ca2+-permeable acid-sensing ion channel 1a (ASIC1a) is responsible for acidosis-mediated tissue injury. The aim of our study was to confirm whether ASIC1a activation induces BMSC apoptosis under conditions that mimic the acidic microenvironment of the degenerated IVD. METHODS: ASIC1a expression in rat BMSCs was investigated by real time-PCR, Western blot (WB) and immunofluorescence. The proliferation and apoptosis of BMSCs under acidic conditions were analyzed by MTT and TUNEL assays. Ca2+-imaging was used to assess the acid-induced increase in the intracellular Ca2+ concentration ([Ca2+]i). The activation of calpain and calcineurin was analyzed using specific kits, and WB analysis was performed to detect apoptosis-related proteins. Ultrastructural changes in BMSCs were observed using transmission electron microscopy (TEM). RESULTS: Acid exposure led to the activation of ASIC1a and increased BMSC apoptosis. The Ca2+ imaging assay showed a significant increase in the [Ca2+]i in response to a solution at pH 6.0. However, BMSC apoptosis and [Ca2+]i elevation were alleviated in the presence of an ASIC1a inhibitor. Moreover, ASIC1a mediated the Ca2+ influx-induced activation of calpain and calcineurin in BMSCs. WB analysis and TEM revealed mitochondrial apoptosis, which was inhibited by an ASIC1a inhibitor, in BMSCs under acidic conditions. CONCLUSIONS: The mimical acidic microenvironment of the degenerated IVD can induce BMSC apoptosis by activating Ca2+-permeable ASIC1a. An acid-induced elevation of [Ca2+]i in BMSCs leads to the subsequent activation of calpain and calcineurin, further resulting in increased mitochondrial permeability and mitochondrial-mediated apoptosis.


Subject(s)
Acid Sensing Ion Channels/metabolism , Apoptosis/physiology , Calcium/metabolism , Intervertebral Disc Degeneration/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Bone Marrow Cells/metabolism , Calpain/metabolism , Cells, Cultured , Intervertebral Disc/metabolism , Ion Transport/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
13.
Neuroscience ; 406: 389-404, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30926548

ABSTRACT

The over-activation of N-methyl-D-aspartate receptors (NMDARs) is the main cause of neuronal death in brain ischemia. Both the NMDAR and the Acid-sensing ion channel 1a (ASIC1a) are present in the postsynaptic membrane of the central nervous system (CNS) and participate in physiological and pathological processes. However, the specific role played by ASIC1a in these processes remains elusive. We hypothesize that NMDARs are the primary mediators of normal synaptic transmission and excitatory neuronal death, while ASIC1a plays a modulatory role in facilitating NMDAR function. Using various experimental approaches including patch-clamp recordings on hippocampal slices and CHO cells, primary cultures of hippocampal neurons, calcium imaging, Western blot, cDNA transfection studies, and transient middle cerebral artery occlusion (tMCAO) mouse models, we demonstrate that stimulation of ASIC1a facilitates NMDAR function and inhibition of ASIC1a suppresses NMDAR over-activation. One of our key findings is that activation of ASIC1a selectively facilitates the NR1/NR2A/NR2B triheteromeric subtype of NMDAR currents. In accordance, inhibition of ASIC1a profoundly reduced the NMDAR-mediated EPSCs in older mouse brains, which are known to express much higher levels of triheteromeric NMDARs than younger brains. Furthermore, brain infarct sizes were reduced by a greater degree in older mice compared to younger ones when ASIC1a activity was suppressed. These data suggest that ASIC1a activity selectively enhances the function of triheteromeric NMDARs and exacerbates ischemic neuronal death especially in older animal brains. We propose ASIC1a as a novel therapeutic target for preventing and reducing the detrimental effect of brain ischemia in humans.


Subject(s)
Acid Sensing Ion Channel Blockers/administration & dosage , Acid Sensing Ion Channels/physiology , Excitatory Amino Acid Agonists/administration & dosage , Nerve Tissue Proteins/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Drug Delivery Systems/methods , Female , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/agonists , Organ Culture Techniques , Receptors, N-Methyl-D-Aspartate/agonists
14.
Biochem Biophys Res Commun ; 511(2): 274-279, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30770101

ABSTRACT

Panic disorder (PD) is a multifactorial neuropsychiatric disorder. Our previous study has demonstrated that the nitric oxide (NO) pathway and the acid-sensing ion channel 1a (ASIC1a) level in the dorsal midbrain periaqueductal gray (dPAG) are involved in the modulation of panic-like responses. In addition, the prefrontal cortex (PFC) and the hippocampus also play a role in panic-like responses. However, no studies have investigated the protein level of ASIC1a in the PFC and hippocampus in a mouse model of panic-like disorders after alteration of the NO pathway in the dPAG. We investigated the production of a panic attack with intra-dPAG injections of SNAP, an NO donor, and 7-NI, an nNOS inhibitor. Moreover, we measured ASIC1a protein levels in the PFC and hippocampus. The rat exposure test (RET) is frequently used as an animal model of panic. In our study, C57BL/6 mice received an intra-dPAG injection of SNAP or 7-NI before RET; neurobehavioral tests were then conducted, followed by mechanistic evaluation through western blot analysis in the PFC and hippocampus. An intra-dPAG infusion of SNAP significantly increased the panic-like effect, whereas treatment with 7-NI decreased fear behavior. Mice treated with SNAP/7-NI showed significantly increased/decreased ASIC1a expression in the PFC, and a decreasing/increasing trend in the hippocampus. The present study suggests that the NO pathway in the dPAG plays a key role in panic-like responses in mice confronted by a rat, further, NO intra-dPAG injection also modulates the ASIC1a expression levels in the PFC and hippocampus.


Subject(s)
Acid Sensing Ion Channels/metabolism , Nitric Oxide/metabolism , Panic/drug effects , Prefrontal Cortex/drug effects , S-Nitroso-N-Acetylpenicillamine/pharmacology , Acid Sensing Ion Channels/analysis , Animals , Hippocampus/drug effects , Hippocampus/physiology , Male , Mice, Inbred C57BL , Periaqueductal Gray/drug effects , Periaqueductal Gray/physiology , Prefrontal Cortex/physiology , Rats, Sprague-Dawley
15.
Behav Brain Res ; 353: 32-39, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29953907

ABSTRACT

Predators induce defensive responses and fear behaviours in prey. The rat exposure test (RET) is frequently used as an animal model of panic. Nitric oxide (NO) which has been reported to be activated by the NMDA receptor, in turn mediates calcium/calmodulin-dependent protein kinase II (CaMKII) signalling pathways in defensive responses. ACCN2, the orthologous human gene of acid-sensing ion channel 1a (ASIC1a), is also associated with panic disorder; however, few studies have focused on the role of ASIC1a in the modulation of panic and calcium/CaMKII signalling by NO. In the present study, NG-nitro-L-arginine-methyl-ester (L-NAME; non-selective NOS inhibitor), S-nitroso-N-acetyl-D,L-penicillamine (SNAP; NO donor), and psalmotoxin (PcTx-1; selective ASIC1a blocker) were administered to the dorsal periaqueductal grey (dPAG) before the predator stimulus, and the roles of NO in the expression of ASIC1a, phosphorylation of CaMKIIα (p-CaMKIIα) and expression of calmodulin (CaM) were investigated. The effects of ASIC1a, p-CaMKIIα and CaM regulation were also examined. Our results showed that intra-dPAG infusion of L-NAME weakened panic-like behaviour and decreased ASIC1a, p-CaMKIIα and CaM expression levels, whereas intra-dPAG infusion of SNAP enhanced panic-like behaviour and increased ASIC1a, p-CaMKIIα and CaM levels. Intra-dPAG infusion of PcTx-1 also weakened panic-like behaviour and decreased p-CaMKIIα expression level. Taken together, these results indicate that NO and ASIC1a are involved in the modulation of RET-induced panic-like behaviour in the dPAG. NO regulates the calcium/CaMKII signalling pathways, and ASIC1a participates in this regulation.


Subject(s)
Acid Sensing Ion Channels/metabolism , Nitric Oxide/metabolism , Panic/physiology , Periaqueductal Gray/metabolism , Animals , Behavior, Animal/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calmodulin/metabolism , Central Nervous System Agents/pharmacology , Male , Mice, Inbred C57BL , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Periaqueductal Gray/drug effects
16.
Biores Open Access ; 7(1): 2-9, 2018.
Article in English | MEDLINE | ID: mdl-29445584

ABSTRACT

Acid-sensing ion channel 1a (ASIC1a) participates in human intervertebral disc degeneration (IVDD) and regulates the destiny of nucleus pulposus cells (NPCs) in acid stimulus. However, the mechanism of ASIC1a activation and its downstream pathway remain unclear. Endoplasmic reticulum (ER) stress also participates in the acid-induced apoptosis of NPCs. The main purpose of this study was to investigate whether there is any connection between ASIC1a and ER stress in an acid-induced nucleus pulposus degeneration model. The IVDs of Sprague-Dawley rats were stained by immunohistochemical staining to evaluate the expression of ASIC1a in normal and degenerated rat nucleus pulposus. ASIC1a expression was also quantified by quantitative real-time-polymerase chain reaction and Western blotting analysis. NPCs were exposed to the culture media with acidity at pH 7.2 and 6.5 for 24 h, with or without 4-phenylbutyrate (4-PBA, a blocker of the ER stress pathway). Cell apoptosis was examined by Annexin V/Propidium Iodide (PI) staining and was quantified using flow cytometry analysis. ASIC1a-mediated intracellular calcium was determined by Ca2+ imaging using Fura-2-AM. Acidity-induced changes in ER stress markers were studied using Western blotting analysis. In vivo, ASIC1a expression was upregulated in natural degeneration. In vitro, acid stimulus increased intracellular calcium levels, but this effect was blocked by knockdown of ASIC1a, and this reversal was partly inhibited by 4-PBA. In addition, blockade of ASIC1a reduced expression of ER stress markers, especially the proapoptotic markers. ASIC1a partly regulates ER stress and promotes apoptosis of NPCs under acid stimulus and may be a novel therapeutic target in IVDD.

17.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-704103

ABSTRACT

Objective To explore the expression of acid-sensing ion channel 1a ( ASIC1a) in cen-tral nervous system of mice with panic like behavior. Methods 20 male C57BL/6 mice were randomly di-vided into two groups according to their weight( 10 mice in each group):the group experienced rat exposure test of panic-like behavior model ( RET group ) and the control group ( Ctr group ) . A panic-like behavior model was established by rat exposure stimuli. Ten minutes defensive and avoidance behaviors of mice were recorded with a horizontal video camera. The anxiety level of mice was evaluated by elevated plus maze ( EPM) test.Western blot was used to detect the ASIC 1a expression in different brain areas of prefrontal cor-tex,hippocampus and periaqueductal gray (PAG). Results Compared with Ctr group,mice in RET group spent significantly more time in freezing ((5.83±1.92)s) than that of Ctr group ((1.00±0.45)s) (P<0.01),had significantly higher frequency of risk assessment behavior (5.33±0.49) than that of Ctr group (0.60±0.40) (P<0.01),spent significantly less time to contact the wire mesh ((17.83±4.38)s) than that of Ctr group((50.00±6.90)s) (P<0.01),and significantly more time of staying in the protected area((431.00±33.13)s) than that of Ctr group((264.40±40.43)s) (P<0.01).At the same time,RET group showed sig-nificantly lower time percent ((8.28±1.12)%) than Ctr group ((16.81±2.13)%) in opened arm (P<0.05) and significantly higher time percent ((80.08±4.26)%) than Ctr group ((60.91±5.27)%) in the closed arm (P<0.05).Western blot suggested that the expression level of ASIC 1a in the prefrontal cortex (1.32± 0.05) and hippocampus (2.56±0.30) significantly increased than that of Ctr group((0.98±0.07),(1.56± 0.16)( P<0.05),while significantly decreased in the PAG (0.83± 0.02) than that of Ctr group(1.26±0.05) ( P<0.05) . Conclusion Rat exposure stimuli can induce panic-like behavior among mice,which increases the expression of ASIC 1a in the prefrontal cortex and hippocampus,but decreases the level of ASIC 1a in the PAG.

18.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-664489

ABSTRACT

Acid-sensing ion channels ( ASICs) are proton-gated channels expressed widely in the central nervous systems and pe-ripheral tissues, among which ASIC1a is a core part and plays an important role in many physiological and pathological proces-ses.As a key receptor for extracellular protons , ASIC1a is in-volved in a variety of pathophysiological processes involving tis-sue acidosis, such as pain, inflammation, seizures and multiple sclerosis.Autoimmune disease is a chronic inflammatory dis-ease , and the excessive activation of T , B cells leads to multiple tissue and organ damage when the body responds to autoantigen immune response . In recent years , studies have found that ASIC1a plays an important role in the development of various au-toimmune diseases.In this paper, the biological characteristics of ASIC1a are briefly reviewed , and the research progress of ASIC1a in the development and progression of autoimmune dis-eases is discussed .

19.
Osteoporos Int ; 28(7): 2221-2231, 2017 07.
Article in English | MEDLINE | ID: mdl-28462470

ABSTRACT

Activated acid-sensing ion channel 1a (ASIC1a) is involved in acid-induced osteoclastogenesis by regulating activation of the transcription factor NFATc1. These results indicated that ASIC1a activation by extracellular acid may cause osteoclast migration and adhesion through Ca2+-dependent integrin/Pyk2/Src signaling pathway. INTRODUCTION: Osteoclast adhesion and migration are responsible for osteoporotic bone loss. Acidic conditions promote osteoclastogenesis. ASIC1a in osteoclasts is associated with acid-induced osteoclastogenesis through modulating transcription factor NFATc1 activation. However, the influence and the detailed mechanism of ASIC1a in regulating osteoclast adhesion and migration, in response to extracellular acid, are not well characterized. METHODS: In this study, knockdown of ASIC1a was achieved in bone marrow macrophage cells using small interfering RNA (siRNA). The adhesion and migration abilities of osteoclast precursors and osteoclasts were determined by adhesion and migration assays, in vitro. Bone resorption was performed to measure osteoclast function. Cytoskeletal changes were assessed by F-actin ring formation. αvß3 integrin expression in osteoclasts was measured by flow cytometry. Western blotting and co-immunoprecipitation were performed to measure alterations in integrin/Pyk2/Src signaling pathway. RESULTS: Our results showed that blockade of ASIC1a using ASIC1a-siRNA inhibited acid-induced osteoclast precursor migration and adhesion, as well as osteoclast adhesion and bone resorption; we also demonstrated that inhibition of ASIC1a decreased the cell surface αvß3 integrin and ß3 protein expression. Moreover, blocking of ASIC1a inhibited acidosis-induced actin ring formation and reduced Pyk2 and Src phosphorylation in osteoclasts and also inhibited the acid-induced association of the αvß3 integrin/Src/Pyk2. CONCLUSION: Together, these results highlight a key functional role of ASIC1a/αvß3 integrin/Pyk2/Src signaling pathway in migration and adhesion of osteoclasts.


Subject(s)
Acid Sensing Ion Channels/physiology , Acidosis/metabolism , Osteoclasts/physiology , Acid Sensing Ion Channels/genetics , Acidosis/pathology , Animals , Bone Resorption/pathology , Bone Resorption/physiopathology , Cell Adhesion/physiology , Cell Movement/physiology , Cells, Cultured , Focal Adhesion Kinase 2/physiology , Gene Knockdown Techniques , Integrin alphaVbeta3/physiology , Male , Osteogenesis/physiology , RNA, Small Interfering/genetics , Rats, Sprague-Dawley , Signal Transduction/physiology , src-Family Kinases/physiology
20.
Proc Natl Acad Sci U S A ; 114(14): 3750-3755, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28320941

ABSTRACT

Stroke is the second-leading cause of death worldwide, yet there are no drugs available to protect the brain from stroke-induced neuronal injury. Acid-sensing ion channel 1a (ASIC1a) is the primary acid sensor in mammalian brain and a key mediator of acidosis-induced neuronal damage following cerebral ischemia. Genetic ablation and selective pharmacologic inhibition of ASIC1a reduces neuronal death following ischemic stroke in rodents. Here, we demonstrate that Hi1a, a disulfide-rich spider venom peptide, is highly neuroprotective in a focal model of ischemic stroke. Nuclear magnetic resonance structural studies reveal that Hi1a comprises two homologous inhibitor cystine knot domains separated by a short, structurally well-defined linker. In contrast with known ASIC1a inhibitors, Hi1a incompletely inhibits ASIC1a activation in a pH-independent and slowly reversible manner. Whole-cell, macropatch, and single-channel electrophysiological recordings indicate that Hi1a binds to and stabilizes the closed state of the channel, thereby impeding the transition into a conducting state. Intracerebroventricular administration to rats of a single small dose of Hi1a (2 ng/kg) up to 8 h after stroke induction by occlusion of the middle cerebral artery markedly reduced infarct size, and this correlated with improved neurological and motor function, as well as with preservation of neuronal architecture. Thus, Hi1a is a powerful pharmacological tool for probing the role of ASIC1a in acid-mediated neuronal injury and various neurological disorders, and a promising lead for the development of therapeutics to protect the brain from ischemic injury.


Subject(s)
Acid Sensing Ion Channel Blockers/administration & dosage , Acid Sensing Ion Channels/metabolism , Neuroprotective Agents/administration & dosage , Spider Venoms/administration & dosage , Stroke/drug therapy , Acid Sensing Ion Channel Blockers/chemistry , Acid Sensing Ion Channel Blockers/pharmacology , Animals , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Magnetic Resonance Spectroscopy , Male , Neuroprotective Agents/pharmacology , Rats , Spider Venoms/chemistry , Spider Venoms/pharmacology , Stroke/complications , Stroke/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...