Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
JCI Insight ; 7(4)2022 02 22.
Article in English | MEDLINE | ID: mdl-35025766

ABSTRACT

Recent research on altering threat memory has focused on a reconsolidation window. During reconsolidation, threat memories are retrieved and become labile. Reconsolidation of distinct threat memories is synapse dependent, whereas the underlying regulatory mechanism of the specificity of reconsolidation is poorly understood. We designed a unique behavioral paradigm in which a distinct threat memory can be retrieved through the associated conditioned stimulus. In addition, we proposed a regulatory mechanism by which the activation of acid-sensing ion channels (ASICs) strengthens the distinct memory trace associated with the memory reconsolidation to determine its specificity. The activation of ASICs by CO2 inhalation, when paired with memory retrieval, triggers the reactivation of the distinct memory trace, resulting in greater memory lability. ASICs potentiate the memory trace by altering the amygdala-dependent synaptic transmission and plasticity at selectively targeted synapses. Our results suggest that inhaling CO2 during the retrieval event increases the lability of a threat memory through a synapse-specific reconsolidation process.


Subject(s)
Acid Sensing Ion Channels/genetics , Behavior, Animal , Conditioning, Classical/physiology , Gene Expression Regulation , Memory/physiology , RNA/genetics , Acid Sensing Ion Channels/biosynthesis , Acoustic Stimulation , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal
2.
FASEB J ; 36(1): e22083, 2022 01.
Article in English | MEDLINE | ID: mdl-34918385

ABSTRACT

Proton pump inhibitors (PPIs) are the mainstay of therapy for gastroesophageal reflux disease (GERD) but up to 60% of patients have inadequate response to therapy. Acid sensing ion channels (ASICs) play important roles in nociception. This study aimed to investigate whether the increased expression of ASICs results in neuronal hyperexcitability in GERD. Esophageal biopsies were taken from GERD patients and healthy subjects to compare expression of ASIC1 and 3. Next, gene and protein expression of ASIC1 and 3 from esophageal mucosa and dorsal root ganglia (DRG) neurons were measured by qPCR, Western-blot and immunofluorescence in rodent models of reflux esophagitis (RE), non-erosive reflux disease (NERD), and sham operated groups. Excitability of DRG neurons in the GERD and sham groups were also tested by whole-cell patch-clamp recordings. We demonstrated that ASIC1 and 3 expression were significantly increased in patients with RE compared with healthy controls. This correlated positively with symptom severity of heartburn and regurgitation (p < .001). Next, ASIC1 and 3 gene and protein expression in rodent models of RE and NERD were similarly increased in esophageal mucosa as well as T3-T5 DRG neurons compared with sham operation. DRG neurons from RE animals showed hyperexcitability compared with sham group. However, intrathecal injection of ASIC specific inhibitors, PcTx1 and APTEx-2, as well as silencing ASIC1 and 3 genes with specific siRNAs prevented visceral hypersensitivity. Overall, upregulation of ASIC1 and 3 may lead to visceral hypersensitivity in RE and NERD and may be a potential therapeutic target for PPI non-responsive patients.


Subject(s)
Acid Sensing Ion Channels/biosynthesis , Esophagus/metabolism , Gastroesophageal Reflux/metabolism , Heartburn/metabolism , Up-Regulation , Acid Sensing Ion Channels/genetics , Animals , Gastroesophageal Reflux/genetics , Heartburn/genetics , Humans , Male , Rats , Rats, Sprague-Dawley
3.
Circulation ; 144(12): 947-960, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34264749

ABSTRACT

BACKGROUND: Ischemia-reperfusion injury (IRI) is one of the major risk factors implicated in morbidity and mortality associated with cardiovascular disease. During cardiac ischemia, the buildup of acidic metabolites results in decreased intracellular and extracellular pH, which can reach as low as 6.0 to 6.5. The resulting tissue acidosis exacerbates ischemic injury and significantly affects cardiac function. METHODS: We used genetic and pharmacologic methods to investigate the role of acid-sensing ion channel 1a (ASIC1a) in cardiac IRI at the cellular and whole-organ level. Human induced pluripotent stem cell-derived cardiomyocytes as well as ex vivo and in vivo models of IRI were used to test the efficacy of ASIC1a inhibitors as pre- and postconditioning therapeutic agents. RESULTS: Analysis of human complex trait genetics indicates that variants in the ASIC1 genetic locus are significantly associated with cardiac and cerebrovascular ischemic injuries. Using human induced pluripotent stem cell-derived cardiomyocytes in vitro and murine ex vivo heart models, we demonstrate that genetic ablation of ASIC1a improves cardiomyocyte viability after acute IRI. Therapeutic blockade of ASIC1a using specific and potent pharmacologic inhibitors recapitulates this cardioprotective effect. We used an in vivo model of myocardial infarction and 2 models of ex vivo donor heart procurement and storage as clinical models to show that ASIC1a inhibition improves post-IRI cardiac viability. Use of ASIC1a inhibitors as preconditioning or postconditioning agents provided equivalent cardioprotection to benchmark drugs, including the sodium-hydrogen exchange inhibitor zoniporide. At the cellular and whole organ level, we show that acute exposure to ASIC1a inhibitors has no effect on cardiac ion channels regulating baseline electromechanical coupling and physiologic performance. CONCLUSIONS: Our data provide compelling evidence for a novel pharmacologic strategy involving ASIC1a blockade as a cardioprotective therapy to improve the viability of hearts subjected to IRI.


Subject(s)
Acid Sensing Ion Channels/biosynthesis , Acid Sensing Ion Channels/genetics , Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Animals , Cells, Cultured , Female , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Isolated Heart Preparation/methods , Male , Mice , Mice, Knockout , Myocardial Ischemia/therapy , Myocardial Reperfusion Injury/therapy , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Polymorphism, Single Nucleotide/physiology , Recovery of Function/drug effects , Recovery of Function/physiology , Spider Venoms/pharmacology
4.
CNS Neurosci Ther ; 27(2): 244-255, 2021 02.
Article in English | MEDLINE | ID: mdl-33314662

ABSTRACT

AIMS: Irritable bowel syndrome (IBS) is a common functional gastrointestinal disease characterized by abdominal pain. Our recent study has shown that the acid-sensitive ion channel 1 (ASIC1) in dorsal root ganglion (DRG) is involved in stomachache of adult offspring rats subjected with prenatal maternal stress (PMS). MiR-485 is predicted to target the expression of ASIC1. The aim of the present study was designed to determine whether miR-485/ASIC1 signaling participates in enterodynia in the spinal dorsal horn of adult offspring rats with PMS. METHODS: Enterodynia was measured by colorectal distension (CRD). Western blotting, qPCR, and in situ hybridization were performed to detect the expression of ASICs and related miRNAs. Spinal synaptic transmission was also recorded by patch clamping. RESULTS: PMS offspring rats showed that spinal ASIC1 protein expression and synaptic transmission were significantly enhanced. Administration of ASICs antagonist amiloride suppressed the synaptic transmission and enterodynia. Besides, PMS induced a significant reduction in the expression of miR-485. Upregulating the expression markedly attenuated enterodynia, reversed the increase in ASIC1 protein and synaptic transmission. Furthermore, ASIC1 and miR-485 were co-expressed in NeuN-positive spinal dorsal horn neurons. CONCLUSIONS: Overall, these data suggested that miR-485 participated in enterodynia in PMS offspring, which is likely mediated by the enhanced ASIC1 activities.


Subject(s)
Abdominal Pain/metabolism , Acid Sensing Ion Channels/biosynthesis , MicroRNAs/biosynthesis , Prenatal Exposure Delayed Effects/metabolism , Spinal Cord/metabolism , Stress, Psychological/metabolism , Abdominal Pain/etiology , Abdominal Pain/genetics , Acid Sensing Ion Channels/genetics , Age Factors , Animals , Female , Male , MicroRNAs/genetics , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Rats , Rats, Sprague-Dawley , Stress, Psychological/complications , Stress, Psychological/genetics , Up-Regulation/physiology
5.
Neurochem Int ; 140: 104824, 2020 11.
Article in English | MEDLINE | ID: mdl-32841711

ABSTRACT

Neuropathic pain is one of the key features of the classical phenotype of Fabry disease (FD). Acid sensing ion channels (ASICs) are H+-gated cation channels, which belong to the epithelial sodium channel/DeGenerin superfamily, sensitive to the diuretic drug Amiloride. Molecular cloning has identified several distinct ASIC subunits. In particular the ASIC1a subunit has been associated to pain and its upregulation has been documented in animal models of pain. We analyzed the expression of ASIC1a channels in cellular models that mimic the accumulation of glycosphingolipids in FD (FD-GLs) like Gb3, and LysoGb3. We used mouse primary neurons from brain cortex and hippocampus -supraspinal structures that accumulate FD-GLs-, as well as HEK293 cells. Incubation with Gb3, lysoGb3 and the inhibitor (1-deoxy-galactonojirymicin, DJG) of the enzyme α-galactosidase A (Gla) lead to the upregulation of ASIC1a channels. In addition, activation of ASIC1a results in the activation of the MAPK ERK pathway, a signaling pathway associated with pain. Moreover, accumulation of glycosphingolipids results in activation of ERK, an effect that was prevented by blocking ASIC1a channels with the specific blocker Psalmotoxin. Our results suggest that FD-GLs accumulation and triggering of the ERK pathway via ASIC channels might be involved in the mechanism responsible for pain in FD, thus providing a new therapeutic target for pain relief treatment.


Subject(s)
Acid Sensing Ion Channels/biosynthesis , Fabry Disease/metabolism , Up-Regulation/physiology , Acid Sensing Ion Channels/genetics , Animals , Cells, Cultured , Fabry Disease/genetics , Fabry Disease/pathology , HEK293 Cells , Hippocampus/metabolism , Hippocampus/pathology , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Peptides/toxicity , Spider Venoms/toxicity , Up-Regulation/drug effects
6.
Neurosci Lett ; 721: 134631, 2020 03 16.
Article in English | MEDLINE | ID: mdl-31734291

ABSTRACT

BACKGROUND: Remifentanil induces hyperalgesia, but the underlying mechanisms are not fully understood. Acid-sensing ion channel 3 (ASIC3) plays a regulatory role in the pain pathway. This study aimed to explore the effect of remifentanil administration on postoperative pain and on ASIC3 expression at the prespinal and supraspinal levels in a rat model. METHODS: Rats were randomly allocated to the control, incision, remifentanil, and remifentanil + incision groups. Remifentanil was given by a 1-h intravenous infusion prior to plantar incision. Paw withdrawal mechanical threshold (PWMT) and paw withdrawal thermal latency (PWTL) were measured at different time points before and after incision to evaluate mechanical and thermal hyperalgesia, respectively. The dorsal root ganglion (DRG), hippocampus, and hypothalamus were obtained after sacrifice at 48 h post-incision for determination of the protein expression of ASIC3 using western blot. RESULTS: Remifentanil administration significantly induced mechanical and thermal hyperalgesia from 2 to 48 h after incision. In addition, remifentanil exposure remarkably stimulated ASIC3 protein expression in DRG, hippocampus, and hypothalamus of rats at 48 h after incision. CONCLUSION: Remifentanil-induced hyperalgesia is accompanied by increased ASIC3 expression at the DRG and supraspinal levels, implying a possible involvement of ASIC3 in remifentanil-induced hyperalgesia.


Subject(s)
Acid Sensing Ion Channels/biosynthesis , Ganglia, Spinal/metabolism , Hippocampus/metabolism , Hyperalgesia/metabolism , Hypothalamus/metabolism , Remifentanil/toxicity , Acid Sensing Ion Channels/genetics , Analgesics, Opioid/toxicity , Animals , Ganglia, Spinal/drug effects , Gene Expression , Hippocampus/drug effects , Hyperalgesia/chemically induced , Hyperalgesia/genetics , Hypothalamus/drug effects , Male , Rats , Rats, Sprague-Dawley
7.
J Neurosci ; 39(29): 5773-5793, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31101759

ABSTRACT

Chronic pain is a serious debilitating disease for which effective treatment is still lacking. Acid-sensing ion channel 1a (ASIC1a) has been implicated in nociceptive processing at both peripheral and spinal neurons. However, whether ASIC1a also contributes to pain perception at the supraspinal level remains elusive. Here, we report that ASIC1a in ACC is required for thermal and mechanical hypersensitivity associated with chronic pain. ACC-specific genetic deletion or pharmacological blockade of ASIC1a reduced the probability of cortical LTP induction and attenuated inflammatory thermal hyperalgesia and mechanical allodynia in male mice. Using cell type-specific manipulations, we demonstrate that ASIC1a in excitatory neurons of ACC is a major player in cortical LTP and pain behavior. Mechanistically, we show that ASIC1a tuned pain-related cortical plasticity through protein kinase C λ-mediated increase of membrane trafficking of AMPAR subunit GluA1 in ACC. Importantly, postapplication of ASIC1a inhibitors in ACC reversed previously established nociceptive hypersensitivity in both chronic inflammatory pain and neuropathic pain models. These results suggest that ASIC1a critically contributes to a higher level of pain processing through synaptic potentiation in ACC, which may serve as a promising analgesic target for treatment of chronic pain.SIGNIFICANCE STATEMENT Chronic pain is a debilitating disease that still lacks effective therapy. Ion channels are good candidates for developing new analgesics. Here, we provide several lines of evidence to support an important role of cortically located ASIC1a channel in pain hypersensitivity through promoting long-term synaptic potentiation in the ACC. Our results indicate a promising translational potential of targeting ASIC1a to treat chronic pain.


Subject(s)
Acid Sensing Ion Channels/biosynthesis , Gyrus Cinguli/metabolism , Isoenzymes/deficiency , Neuralgia/metabolism , Neuronal Plasticity/physiology , Pain Measurement/methods , Protein Kinase C/deficiency , 6-Cyano-7-nitroquinoxaline-2,3-dione/administration & dosage , Acid Sensing Ion Channels/genetics , Animals , Cells, Cultured , Gyrus Cinguli/drug effects , Isoenzymes/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microinjections/methods , Neuralgia/genetics , Neuralgia/prevention & control , Neuronal Plasticity/drug effects , Organ Culture Techniques , Pain Measurement/drug effects , Protein Kinase C/genetics
8.
Stem Cells Dev ; 28(14): 920-932, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31119982

ABSTRACT

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are potential sources for cardiac regeneration and drug development. hiPSC-CMs express all the cardiac ion channels and the unique cardiac Ca2+-signaling phenotype. In this study, we tested for expression of acid sensing ion channels (ASICs) in spontaneously beating cardiomyocytes derived from three different hiPSC lines (IMR-90, iPSC-K3, and Ukki011-A). Rapid application of solutions buffered at pH 6.7, 6.0, or 5.0 triggered rapidly activating and slowly inactivating voltage-independent inward current that reversed at voltages positive to ENa, was suppressed by 5 µM amiloride and withdrawal of [Na+]o, like neuronal ASIC currents. ASIC currents were expressed at much lower percentages and densities in undifferentiated hiPSC and in dermal fibroblasts. ASIC1 mRNA and protein were measured in first 60 days but decreased in 100 days postdifferentiation hiPSC cultures. Hyperacidification (pH 5 and 6) also triggered large Ca2+ transients in intact hiPSC-CMs that were neither ruthenium red nor amiloride-sensitive, but were absent in whole cell-clamped hiPSC-CMs. Neither ASIC1 current nor its protein was detected in rat adult cardiomyocytes, but hyperacidification did activate smaller and slowly activating currents with drug sensitivity similar to TRPV channels. Considering ASIC expression in developing but not adult myocardium, a role in heart development is likely.


Subject(s)
Acid Sensing Ion Channels/biosynthesis , Cell Differentiation , Gene Expression Regulation , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Cell Line, Tumor , Dermis/cytology , Dermis/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology
9.
Exp Mol Pathol ; 106: 27-33, 2019 02.
Article in English | MEDLINE | ID: mdl-30485799

ABSTRACT

Costal cartilage is much understudied compared to the load bearing cartilages. Abnormally grown costal cartilages are associated with the inherited chest wall deformities pectus excavatum and pectus carinatum resulting in sunken or pigeon chest respectively. A lack of understanding of the ultrastructural and molecular biology properties of costal cartilage is a major confounder in predicting causes and outcomes of these disorders. Due to the avascular nature of cartilage, chondrocytes metabolize glycolytically, producing an acidic environment. During physical activity hydrogen ions move within cartilage driven by compressive forces, thus at any one time, chondrocytes experience transient changes in pH. A variety of ion channels on chondrocytes plasma membrane equip them to function in the rapidly changing conditions they experience. In this paper we describe reduced expression of the ASIC2 gene encoding the acid sensing ion channel isoform 2 (previously referred to as ACCN1 or ACCN) in patients with chest wall deformities. We hypothesized that chondrocytes from these patients cannot respond normally to changes in pH that are an integral part of the biology of this tissue. Activation of ASICs indirectly creates a cascade ultimately dependent on intracellular calcium transients. The objective of this paper was to compare internal calcium signaling in response to external pH changes in costal chondrocytes from patients with chest wall deformities and healthy individuals. Although the molecular mechanism through which chondrocytes are regulated by acidosis remains unknown, we observed reduced amplitudes of calcium rise in patient chondrocytes exposed to low pH that become further impaired upon repeat exposure.


Subject(s)
Acid Sensing Ion Channels/biosynthesis , Acidosis/pathology , Chondrocytes/drug effects , Costal Cartilage/drug effects , Funnel Chest/pathology , Pectus Carinatum/pathology , Acid Sensing Ion Channels/genetics , Adolescent , Calcium Signaling/drug effects , Cells, Cultured , Chondrocytes/metabolism , Costal Cartilage/pathology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Hydrogen-Ion Concentration , Male , RNA, Messenger/biosynthesis , Young Adult
10.
Neurosci Lett ; 684: 200-204, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30114476

ABSTRACT

Sex differences occur in nociceptive pain, and estrogens are involved in the sex differences. Our previous study shows sex differences exist in acidosis-induced nociception in rats, with females being more sensitive than males to acetic acid. However, the mechanisms underlying the sex differences remain unclear. We report here17ß-estradiol (E2) up-regulates expression of acid-sensing ion channel 3 (ASIC3), which can mediate the acidosis-induced events. The recombinant plasmid of pCDNA3.1-ASIC3-GFP and pCDNA3.1-estrogen receptor α (ERα) were cotransfected to 293 T cells by lipid transfection method. And western blot assays showed expression of ASIC3. We found that E2 markedly increases ASIC3 protein expression in a dose- and time- dependent manner in 293 T cells expressing ASIC3 and ERα. The up-regulating effect of E2 on ASIC3 protein expression is almost completely blocked by the addition of MPP, a specific ERα antagonist. We also observed that sex differences occur in ASIC3 expression in rat dorsal root ganglia (DRG) and in acetic acid-induced nociceptive responses. ASIC3 protein expression in female rat DRG is higher than those in male rat DRG. And female rats are more sensitive to acetic acid-induced nociception than males. ASIC3 protein expression in DRG decreases significantly after ovariectomy, but not after orchiectomy. These results suggest that E2 up-regulates ASIC3 expression through ERα, which may contribute to sex differences in acetic acid-induced nociception.


Subject(s)
Acid Sensing Ion Channels/biosynthesis , Estradiol/pharmacology , Estrogens/pharmacology , Sex Characteristics , Up-Regulation/physiology , Acid Sensing Ion Channels/genetics , Animals , Dose-Response Relationship, Drug , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Gene Expression , HEK293 Cells , Humans , Male , Rats, Sprague-Dawley , Up-Regulation/drug effects
11.
Tumour Biol ; 39(6): 1010428317705750, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28618956

ABSTRACT

Acid-sensing ion channels, a proton-gated cation channel, can be activated by low extracellular pH and involved in pathogenesis of some tumors such as glioma and breast cancer. However, the role of acid-sensing ion channels in the growth of lung cancer cell is unclear. In this study, we investigated the expression of acid-sensing ion channels in human lung cancer cell line A549 and their possible role in proliferation and migration of A549 cells. The results show that acid-sensing ion channel 1, acid-sensing ion channel 2, and acid-sensing ion channel 3 are expressed in A549 cells at the messenger RNA and protein levels, and acid-sensing ion channel-like currents were elicited by extracellular acid stimuli. Moreover, we found that acidic extracellular medium or overexpressing acid-sensing ion channel 1a promotes proliferation and migration of A549 cells. In addition psalmotoxin 1, a specific acid-sensing ion channel 1a inhibitor, or acid-sensing ion channel 1a knockdown can abolish the effect of acid stimuli on A549 cells. In addition, acid-sensing ion channels mediate increase of [Ca2+]i induced by low extracellular pH in A549 cells. All these results indicate that acid-sensing ion channel-calcium signal mediate lung cancer cell proliferation and migration induced by extracellular acidosis, and acid-sensing ion channels may serve as a prognostic marker and a therapeutic target for lung cancer.


Subject(s)
Acid Sensing Ion Channels/biosynthesis , Biomarkers, Tumor/biosynthesis , Lung Neoplasms/genetics , A549 Cells , Acid Sensing Ion Channels/genetics , Acidosis/genetics , Acidosis/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lung Neoplasms/pathology , Prognosis
12.
J Musculoskelet Neuronal Interact ; 16(2): 145-51, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27282458

ABSTRACT

OBJECTIVES: Nociceptors are expressed at peripheral terminals of neurons. Recent studies have shown that TRPV1, a nociceptor, is expressed in bone tissue and regulates bone metabolism. We have demonstrated that a TRPV1 antagonist improved pain-like behavior in ovariectomized (OVX) mice. The aim of this study was to determine whether nociceptors, including TRPV1, acid-sensing ion channel (ASIC) and P2X2/3 are expressed in bone cells, and to examine the effects of nociceptor antagonists on bone metabolism. METHODS: The expression of nociceptors in femoral bone tissue and cultured bone marrow cells in OVX and sham-operated mice were examined. The effects of nociceptor antagonists on the up-regulated expression of bone metabolic markers, Runx2, Osterix, osteocalcin and RANKL, were also examined. RESULTS: TRPV1, ASIC 2 and 3, and P2X2 and 3, were expressed in bone tissue and bone marrow cells, and the expression levels of ASIC1 and 2, and P2X2 were significantly increased in OVX mice in comparison with those in sham mice. Treatment with nociceptor antagonists significantly inhibited the expression of bone metabolic markers in OVX mice. CONCLUSION: An array of nociceptors, TRPV1, ASICs and P2X2/3, could simultaneously regulate not only increases in skeletal pain but also bone turnover in OVX mice.


Subject(s)
Acid Sensing Ion Channels/biosynthesis , Bone and Bones/metabolism , Receptors, Purinergic P2X2/biosynthesis , Receptors, Purinergic P2X3/biosynthesis , TRPV Cation Channels/biosynthesis , Animals , Female , Humans , Mice , Mice, Inbred C57BL , Osteoporosis, Postmenopausal/metabolism , Ovariectomy , Polymerase Chain Reaction
13.
EMBO J ; 35(4): 414-28, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26772186

ABSTRACT

Extracellular pH variations are seen as the principal endogenous signal that triggers activation of Acid-Sensing Ion Channels (ASICs), which are basically considered as proton sensors, and are involved in various processes associated with tissue acidification. Here, we show that human painful inflammatory exudates, displaying non-acidic pH, induce a slow constitutive activation of human ASIC3 channels. This effect is largely driven by lipids, and we identify lysophosphatidylcholine (LPC) and arachidonic acid (AA) as endogenous activators of ASIC3 in the absence of any extracellular acidification. The combination of LPC and AA evokes robust depolarizing current in DRG neurons at physiological pH 7.4, increases nociceptive C-fiber firing, and induces pain behavior in rats, effects that are all prevented by ASIC3 blockers. Lipid-induced pain is also significantly reduced in ASIC3 knockout mice. These findings open new perspectives on the roles of ASIC3 in the absence of tissue pH variation, as well as on the contribution of those channels to lipid-mediated signaling.


Subject(s)
Acid Sensing Ion Channels/biosynthesis , Arachidonic Acid/metabolism , Lysophosphatidylcholines/metabolism , Nociceptors/physiology , Animals , Cell Line , Ganglia, Spinal/cytology , Humans , Mice, Knockout , Pain , Rats
14.
Mol Neurobiol ; 53(1): 485-498, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25476599

ABSTRACT

Recent studies have indicated that acid-sensing ion channels may play a significant role in the termination of epilepsy. In particular, acid-sensing ion channel 3 (ASIC3) is expressed in the central nervous system and is most sensitive to extracellular pH. However, whether ASIC3 plays a role in epilepsy is unknown. In this study, qRT-PCR, Western blot, immunohistochemistry, double immunofluorescence labeling, and slice recordings were used. We first detected elevated ASIC3 expression patterns in the brains of temporal lobe epilepsy patients and epileptic rats. ASIC3 was expressed in neurons and glia in both humans and in an experimental model of epilepsy, and ASIC3 was colocalized with inhibitory GABAergic interneurons. By blocking ASIC3 with its antagonist APETx2, we observed that injected APETx2 shortened the latency to seizure and increased the incidence of generalized tonic clonic seizure compared to the control group in models of both pilocarpine- and pentylenetetrazole (PTZ)-induced seizures. Additionally, blocking ASIC3 significantly decreased the frequency of action potential (AP) firing in interneurons. Moreover, APETx2 significantly reduced the amplitudes and frequencies of miniature inhibitory postsynaptic currents (mIPSCs) while showed no differences with the APETx2 + bicuculline group and the bicuculline group. These findings suggest that elevated levels of ASIC3 may serve as an anti-epileptic mechanism via postsynaptic mechanisms in interneurons. It could represent a novel therapeutic strategy for epilepsy treatment.


Subject(s)
Acid Sensing Ion Channels/biosynthesis , Epilepsy, Temporal Lobe/metabolism , Interneurons/metabolism , Adolescent , Adult , Animals , Child , Cnidarian Venoms/pharmacology , Epilepsy, Temporal Lobe/pathology , Female , Gene Expression Regulation , Humans , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Interneurons/drug effects , Interneurons/pathology , Male , Rabbits , Rats, Sprague-Dawley , Temporal Lobe/drug effects , Temporal Lobe/metabolism , Temporal Lobe/pathology , Young Adult
15.
Int Immunopharmacol ; 29(2): 748-760, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26359543

ABSTRACT

The inflammatory cytokine interleukin-6 (IL-6) is a causative agent of rheumatoid arthritis (RA), a chronic inflammatory disease complicated with degenerative arthritic cartilage. However, the precise mechanism of IL-6 on chondrocyte apoptosis is largely unclear. Acid-sensing ion channels (ASICs), a family of extracellular H(+)-activated cation channels, can be transiently activated by extracellular acid and play a pivotal role in acid-induced cell injury. In the present study, to investigate the role of IL-6 in regulating acid-induced articular chondrocyte apoptosis, primary rat articular chondrocytes were subjected to different treatments with or without IL-6 in the presence of acid. The results showed that the mRNA and protein expressions of ASIC1a were significantly increased in articular cartilage and chondrocytes of adjuvant arthritis (AA) rats. IL-6 could dramatically upregulate the level of ASIC1a in a time- and dose-dependent manner, and induce the activation of JAK2, STAT3, ERK, JNK and NF-κB in articular chondrocytes. Moreover, both the respective inhibitors of these signaling pathways and the specific antibody against IL-6 receptor (tocilizumab) could partially abrogate the ASIC1a upregulation induced by IL-6. Furthermore, IL-6 inhibited the cell viability and enhanced LDH release, [Ca(2+)]i elevation, and apoptosis in acid-induced articular chondrocytes, and these changes could be reversed by using psalmotoxin 1(PcTX1), which is the specific antagonist of ASIC1a. In addition, pretreatment with PcTX1 could inhibit the downregulated expression of Bcl-2 and the upregulated expression of Bax induced by IL-6 in acid-induced articular chondrocytes. Taken together, these results indicated that IL-6 could enhance acid-induced articular chondrocyte apoptosis, the mechanism of which might partially be involved with its ability of regulating the activation of ASIC1a-dependent JAK2/STAT3 and MAPK/NF-κB signaling pathways.


Subject(s)
Acid Sensing Ion Channels/biosynthesis , Apoptosis/drug effects , Cartilage, Articular/cytology , Cartilage, Articular/drug effects , Chondrocytes/drug effects , Interleukin-6/pharmacology , Acid Sensing Ion Channels/drug effects , Acids , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/biosynthesis , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Interleukin-6/antagonists & inhibitors , L-Lactate Dehydrogenase/metabolism , Male , Peptides/pharmacology , Rats , Rats, Sprague-Dawley , Spider Venoms/pharmacology , Up-Regulation/drug effects
16.
Bone Joint J ; 97-B(6): 824-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26033064

ABSTRACT

The purpose of this study was to evaluate the expression of acid-sensing ion channels (ASICs) in the capsule and synovial fluid of patients with frozen shoulder. Capsular tissue and synovial fluid were obtained from 18 patients with idiopathic frozen shoulder (FS group) and 18 patients with instability of the shoulder (control group). The expressions of ASIC1, ASIC2, and ASIC3 in the capsule were determined using the reverse transcriptase-polymerase chain reaction, immunoblot analysis, and immunohistochemistry (IHC). The concentrations in synovial fluid were evaluated using an enzyme-linked immunosorbent assay. The mRNA expression of ASIC1, ASIC2 and ASIC3 in the capsule were significantly increased in the FS group compared with the control group. The protein levels of these three ASICs were also increased. The increased expressions were confirmed by IHC. Of the ASICs, ASIC3 showed the greatest increase in both mRNA and levels of expression compared with the control group. The levels of ASIC1 and ASIC3 in synovial fluid were significantly increased in the FS group. This study suggests that ASICs may play a role as mediators of inflammatory pain and be involved in the pathogenesis of frozen shoulder.


Subject(s)
Acid Sensing Ion Channels/metabolism , Bursitis/metabolism , Joint Capsule/metabolism , Up-Regulation/physiology , Acid Sensing Ion Channels/biosynthesis , Bursitis/physiopathology , Humans , Immunohistochemistry , RNA, Messenger/metabolism , Synovial Fluid/metabolism
17.
Sci Rep ; 5: 10143, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25988357

ABSTRACT

Genetic variants in the human ortholog of acid-sensing ion channel-1a subunit (ASIC1a) gene are associated with panic disorder and amygdala dysfunction. Both fear learning and activity-induced long-term potentiation (LTP) of cortico-basolateral amygdala (BLA) synapses are impaired in ASIC1a-null mice, suggesting a critical role of ASICs in fear memory formation. In this study, we found that ASICs were differentially expressed within the amygdala neuronal population, and the extent of LTP at various glutamatergic synapses correlated with the level of ASIC expression in postsynaptic neurons. Importantly, selective deletion of ASIC1a in GABAergic cells, including amygdala output neurons, eliminated LTP in these cells and reduced fear learning to the same extent as that found when ASIC1a was selectively abolished in BLA glutamatergic neurons. Thus, fear learning requires ASIC-dependent LTP at multiple amygdala synapses, including both cortico-BLA input synapses and intra-amygdala synapses on output neurons.


Subject(s)
Acid Sensing Ion Channels/genetics , Amygdala/metabolism , Excitatory Postsynaptic Potentials/physiology , Fear/physiology , Long-Term Potentiation/physiology , Acid Sensing Ion Channels/biosynthesis , Animals , Fear/psychology , Female , GABAergic Neurons/physiology , Learning , Male , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/physiology , Synapses/metabolism
18.
Cell Tissue Res ; 360(2): 223-31, 2015 May.
Article in English | MEDLINE | ID: mdl-25585988

ABSTRACT

Acid-sensing ion channels (ASICs) are H(+)-gated, voltage-insensitive cation channels involved in synaptic transmission, mechanosensation and nociception. Different ASICs have been detected in the retina of mammals but it is not known whether they are expressed in adult zebrafish, a commonly used animal model to study the retina in both normal and pathological conditions. We study the expression and distribution of ASIC2 and ASIC4 in the retina of adult zebrafish and its regulation by light using PCR, in situ hybridization, western blot and immunohistochemistry. We detected mRNA encoding zASIC2 and zASIC4.2 but not zASIC4.1. ASIC2, at the mRNA or protein level, was detected in the outer nuclear layer, the outer plexiform layer, the inner plexiform layer, the retinal ganglion cell layer and the optic nerve. ASIC4 was expressed in the photoreceptors layer and to a lesser extent in the retinal ganglion cell layer. Furthermore, the expression of both ASIC2 and ASIC4.2 was down-regulated by light and darkness. These results are the first demonstration that ASIC2 and ASIC4 are expressed in the adult zebrafish retina and suggest that zebrafish could be used as a model organism for studying retinal pathologies involving ASICs.


Subject(s)
Acid Sensing Ion Channels/biosynthesis , Eye Proteins/biosynthesis , Gene Expression Regulation/physiology , Retina/metabolism , Zebrafish Proteins/biosynthesis , Zebrafish/metabolism , Animals , Retina/cytology
19.
Tumour Biol ; 36(6): 4309-17, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25613068

ABSTRACT

An acidic microenvironment promotes carcinoma cell proliferation and migration. Acid-sensing ion channels (ASICs) are H(+), Ca(2+), and Na(+)-gated cation channels that are activated by changes in the extracellular pH, and ASIC1α may be associated with tumor proliferation and migration. Here, we investigated the role of ASIC1α in hepatocellular carcinoma (HCC) migration and invasion. The expression of ASIC1α was examined in 15 paired HCC and adjacent non-tumor tissues by immunohistochemistry. Reverse transcription (RT)-PCR and Western blotting were used to assess ASIC1α messenger RNA (mRNA) and protein expression in the HCC cell line SMMC-7721 cultured in different pH media or transfected with short hairpin RNA (shRNA) against ASIC1α. Cell migration ability was detected by wound healing and Transwell assays. ASIC1α expression was significantly higher in tumor tissues than in non-tumor tissues, and it was higher in HCC with postoperative metastasis than in that without metastasis. ASIC1α mRNA and protein expression was significantly higher in SMMC-7721 cells cultured at pH 6.5 than in those cultured at pH 7.4 and 6.0. shRNA-mediated silencing of ASIC1α significantly downregulated ASIC1α mRNA and protein expression compared with negative control or untransfected cells and inhibited HCC cell migration and invasion. ASIC1α is overexpressed in HCC tissues and associated with advanced clinical stage. A moderately acidic extracellular environment promoted ASIC1α expression, and silencing of ASIC1α expression inhibited the migration and invasion of HCC cells. Suppression of ASIC1α expression by RNAi attenuated the malignant phenotype of HCC cells, suggesting a novel approach for anticancer gene therapy.


Subject(s)
Acid Sensing Ion Channels/genetics , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Acid Sensing Ion Channels/biosynthesis , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/pathology , Male , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , Neoplasm Staging , RNA, Small Interfering
20.
J Chem Neuroanat ; 61-62: 161-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25462386

ABSTRACT

Extracellular acidification activates a family of proteins known as acid-sensing ion channels (ASICs). One ASIC subtype, ASIC type 1 (ASIC1), may play an important role in synaptic plasticity, memory, fear conditioning and ischemic brain injury. ASIC1 is found primarily in neurons, but one report showed its expression in isolated mouse cerebrovascular cells. In this study, we sought to determine if ASIC1 is present in intact rat and human major cerebral arteries. A potential physiological significance of such a finding is suggested by studies showing that nitric oxide (NO), which acts as a powerful vasodilator, may modulate proton-gated currents in cultured cells expressing ASIC1s. Because both constitutive NO synthesizing enzymes, neuronal nitric oxide synthase (nNOS) and endothelial NOS (eNOS), are expressed in cerebral arteries we also studied the anatomical relationship between ASIC1 and nNOS or eNOS in both rat and human cerebral arteries. Western blot analysis demonstrated ASIC1 in cerebral arteries from both species. Immunofluorescent histochemistry and confocal microscopy also showed that ASIC1-immunoreactivity (IR), colocalized with the smooth muscle marker alpha-smooth muscle actin (SMA), was present in the anterior cerebral artery (ACA), middle cerebral artery (MCA), posterior cerebral artery (PCA) and basilar artery (BA) of rat and human. Expression of ASIC1 in cerebral arteries is consistent with a role for ASIC1 in modulating cerebrovascular tone both in rat and human. Potential interactions between smooth muscle ASIC1 and nNOS or eNOS were supported by the presence of nNOS-IR in the neighboring adventitial layer and the presence of nNOS-IR and eNOS-IR in the adjacent endothelial layer of the cerebral arteries.


Subject(s)
Acid Sensing Ion Channels/biosynthesis , Cerebral Arteries/enzymology , Nitric Oxide Synthase/biosynthesis , Acid Sensing Ion Channels/analysis , Adolescent , Aged , Aged, 80 and over , Animals , Blotting, Western , Cadaver , Fluorescent Antibody Technique , Humans , Male , Microscopy, Confocal , Middle Aged , Muscle, Smooth, Vascular/metabolism , Nitric Oxide Synthase/analysis , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...