Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
Stroke ; 55(6): 1660-1671, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38660789

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-3ß,5α,6ß-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 ASIC1ß 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.


Subject(s)
Acid Sensing Ion Channels , Acidosis , Cricetulus , Mice, Knockout , Animals , Acid Sensing Ion Channels/metabolism , Acid Sensing Ion Channels/genetics , Mice , CHO Cells , Acidosis/metabolism , Acidosis/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/drug therapy , Neurons/drug effects , Neurons/metabolism , Cricetinae , Neuroprotective Agents/pharmacology , Cholestanols/pharmacology , Mice, Inbred C57BL , Acid Sensing Ion Channel Blockers/pharmacology , Male , Cells, Cultured
2.
eNeuro ; 11(2)2024 Feb.
Article in English | MEDLINE | ID: mdl-38233146

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.


Subject(s)
Brain Injuries , Ischemic Stroke , Stroke , Animals , Mice , Acid Sensing Ion Channels/genetics , Acid Sensing Ion Channels/metabolism , Brain/metabolism , Stroke/drug therapy
3.
Biomolecules ; 13(3)2023 03 10.
Article in English | MEDLINE | ID: mdl-36979442

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.


Subject(s)
Acid Sensing Ion Channels , Sodium-Calcium Exchanger , Cricetinae , Mice , Animals , Cricetulus , Sodium-Calcium Exchanger/genetics , CHO Cells
4.
Biomolecules ; 12(9)2022 09 01.
Article in English | MEDLINE | ID: mdl-36139053

ABSTRACT

In this study, we characterize biophysical changes in NMDA receptor function in response to brief non-injurious ischemic stress (ischemic preconditioning). Electrophysiological studies show NMDA receptor function is reduced following preconditioning in cultured rat cortical neurons. This functional change is not due to changes in the reversal potential of the receptor, but an increase in desensitization. We performed concentration-response analysis of NMDA-evoked currents, and demonstrate that preconditioned neurons show a reduced potency of NMDA to evoke currents, an increase in Mg2+ sensitivity, but no change in glycine sensitivity. Antagonists studies show a reduced inhibition of GluN2B antagonists that have an allosteric mode of action (ifenprodil and R-25-6981), but competitive antagonists at the GluR2A and 2B receptor (NVP-AMM077 and conantokin-G) appear to have similar potency to block currents. Biochemical studies show a reduction in membrane surface GluN2B subunits, and an increased co-immunoprecipitation of GluN2A with GluN2B subunits, suggestive of tri-heteromeric receptor formation. Finally, we show that blocking actin remodeling with jasplakinolide, a mechanism of rapid ischemic tolerance, prevents NMDA receptor functional changes and co-immunoprecipitation of GluN2A and 2B subunits. Together, this study shows that alterations in NMDA receptor function following preconditioning ischemia are associated with neuroprotection in rapid ischemic tolerance.


Subject(s)
N-Methylaspartate , Receptors, N-Methyl-D-Aspartate , Actins , Animals , Glycine/pharmacology , Ischemia , Rats
5.
J Cereb Blood Flow Metab ; 42(8): 1349-1363, 2022 08.
Article in English | MEDLINE | ID: mdl-35301897

ABSTRACT

Extracellular proton concentration is at 40 nM when pH is 7.4. In disease conditions such as brain ischemia, proton concentration can reach µM range. To respond to this increase in extracellular proton concentration, the mammalian brain expresses at least three classes of proton receptors. Acid-sensing ion channels (ASICs) are the main neuronal cationic proton receptor. The proton-activated chloride channel (PAC), which is also known as (aka) acid-sensitive outwardly rectifying anion channel (ASOR; TMEM206), mediates acid-induced chloride currents. Besides proton-activated channels, GPR4, GPR65 (aka TDAG8, T-cell death-associated gene 8), and GPR68 (aka OGR1, ovarian cancer G protein-coupled receptor 1) function as proton-sensitive G protein-coupled receptors (GPCRs). Though earlier studies on these GPCRs mainly focus on peripheral cells, we and others have recently provided evidence for their functional importance in brain injury. Specifically, GPR4 shows strong expression in brain endothelium, GPR65 is present in a fraction of microglia, while GPR68 exhibits predominant expression in brain neurons. Here, to get a better view of brain acid signaling and its contribution to ischemic injury, we will review the recent findings regarding the differential contribution of proton-sensitive GPCRs to cerebrovascular function, neuroinflammation, and neuronal injury following acidosis and brain ischemia.


Subject(s)
Brain Ischemia , Protons , Acid Sensing Ion Channels/metabolism , Animals , Hydrogen-Ion Concentration , Mammals/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
6.
Acta Pharmacol Sin ; 42(8): 1248-1255, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33184449

ABSTRACT

Oxidative stress is intimately tied to neurodegenerative diseases, including Parkinson's disease and amyotrophic lateral sclerosis, and acute injuries, such as ischemic stroke and traumatic brain injury. Acid sensing ion channel 1a (ASIC1a), a proton-gated ion channel, has been shown to be involved in the pathogenesis of these diseases. However, whether oxidative stress affects the expression of ASIC1a remains elusive. In the current study, we examined the effect of hydrogen peroxide (H2O2), a major reactive oxygen species (ROS), on ASIC1a protein expression and channel function in NS20Y cells and primary cultured mouse cortical neurons. We found that treatment of the cells with H2O2 (20 µM) for 6 h or longer increased ASIC1a protein expression and ASIC currents without causing significant cell injury. H2O2 incubation activated mitogen-activated protein kinases (MAPKs) pathways, including the extracellular signal-regulated kinase1/2 (ERK1/2), c-Jun N-terminal kinase (JNK), and p38 pathways. We found that neither inhibition of the MEK/ERK pathway by U0126 nor inhibition of the p38 pathway by SB203580 affected H2O2-induced ASIC1a expression, whereas inhibition of the JNK pathway by SP600125 potently decreased ASIC1a expression and abolished the H2O2-mediated increase in ASIC1a expression and ASIC currents. Furthermore, we found that H2O2 pretreatment increased the sensitivity of ASIC currents to the ASIC1a inhibitor PcTx1, providing additional evidence that H2O2 increases the expression of functional ASIC1a channels. Together, our data demonstrate that H2O2 increases ASIC1a expression/activation through the JNK signaling pathway, which may provide insight into the pathogenesis of neurological disorders that involve both ROS and activation of ASIC1a.


Subject(s)
Acid Sensing Ion Channels/metabolism , Hydrogen Peroxide/pharmacology , MAP Kinase Signaling System/drug effects , Animals , Butadienes/pharmacology , Cell Line, Tumor , Imidazoles/pharmacology , Mice , Neurons/drug effects , Neurons/metabolism , Nitriles/pharmacology , Oxidative Stress/drug effects , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Up-Regulation/drug effects
7.
Theranostics ; 10(26): 11976-11997, 2020.
Article in English | MEDLINE | ID: mdl-33204324

ABSTRACT

Rapid increase in aging populations is an urgent problem because older adults are more likely to suffer from disabilities and age-related diseases (ARDs), burdening healthcare systems and society in general. ARDs are characterized by the progressive deterioration of tissues and organs over time, eventually leading to tissue and organ failure. To date, there are no effective interventions to prevent the progression of ARDs. Hence, there is an urgent need for new treatment strategies. Ferroptosis, an iron-dependent cell death, is linked to normal development and homeostasis. Accumulating evidence, however, has highlighted crucial roles for ferroptosis in ARDs, including neurodegenerative and cardiovascular diseases. In this review, we a) summarize initiation, regulatory mechanisms, and molecular signaling pathways involved in ferroptosis, b) discuss the direct and indirect involvement of the activation and/or inhibition of ferroptosis in the pathogenesis of some important diseases, and c) highlight therapeutic targets relevant for ARDs.


Subject(s)
Aging/pathology , Cardiovascular Diseases/drug therapy , Diabetes Mellitus/drug therapy , Ferroptosis/drug effects , Neurodegenerative Diseases/drug therapy , Pulmonary Disease, Chronic Obstructive/drug therapy , Aged , Aged, 80 and over , Aging/drug effects , Animals , Cardiovascular Diseases/pathology , Cell Line, Tumor , Diabetes Mellitus/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Ferroptosis/physiology , Humans , Iron/metabolism , Neurodegenerative Diseases/pathology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Pulmonary Disease, Chronic Obstructive/pathology , Signal Transduction/drug effects , Signal Transduction/physiology
8.
Stroke ; 51(12): 3690-3700, 2020 12.
Article in English | MEDLINE | ID: mdl-33059544

ABSTRACT

BACKGROUND AND PURPOSE: Brain acidosis is prevalent in stroke and other neurological diseases. Acidosis can have paradoxical injurious and protective effects. The purpose of this study is to determine whether a proton receptor exists in neurons to counteract acidosis-induced injury. METHODS: We analyzed the expression of proton-sensitive GPCRs (G protein-coupled receptors) in the brain, examined acidosis-induced signaling in vitro, and studied neuronal injury using in vitro and in vivo mouse models. RESULTS: GPR68, a proton-sensitive GPCR, was present in both mouse and human brain, and elicited neuroprotection in acidotic and ischemic conditions. GPR68 exhibited wide expression in brain neurons and mediated acidosis-induced PKC (protein kinase C) activation. PKC inhibition exacerbated pH 6-induced neuronal injury in a GPR68-dependent manner. Consistent with its neuroprotective function, GPR68 overexpression alleviated middle cerebral artery occlusion-induced brain injury. CONCLUSIONS: These data expand our knowledge on neuronal acid signaling to include a neuroprotective metabotropic dimension and offer GPR68 as a novel therapeutic target to alleviate neuronal injuries in ischemia and multiple other neurological diseases.


Subject(s)
Acidosis/metabolism , Brain/metabolism , Infarction, Middle Cerebral Artery/metabolism , Neurons/metabolism , Neuroprotection/genetics , Receptors, G-Protein-Coupled/metabolism , Animals , Humans , Ischemic Stroke/metabolism , Mice , Mice, Knockout , Neuroprotection/physiology , Protein Kinase C/metabolism , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction
9.
Mol Neurobiol ; 57(11): 4754-4766, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32783140

ABSTRACT

Tissue acidosis is a common feature in many pathological conditions. Activation of acid-sensing ion channel 1a (ASIC1a) plays a key role in acidosis-mediated neurotoxicity. Protein kinase C (PKC) activity has been proved to be associated with many physiological processes and pathological conditions; however, whether PKC activation regulates ASIC1a protein expression and channel function remains ill defined. In this study, we demonstrated that treatment with phorbol 12-myristate 13-acetate (PMA, a PKC activator) for 6 h significantly increased ASIC1a protein expression and ASIC currents in NS20Y cells, a neuronal cell line, and in primary cultured mouse cortical neurons. In contrast, treatment with Calphostin C (a nonselective PKC inhibitor) for 6 h or longer decreased ASIC1a protein expression and ASIC currents. Similar to Calphostin C, PKC α and ßI inhibitor Go6976 exposure also reduced ASIC1a protein expression. The reduction in ASIC1a protein expression by PKC inhibition involves a change in ASIC1a protein degradation, which is mediated by ubiquitin-proteasome system (UPS)-dependent degradation pathway. In addition, we showed that PKC regulation of ASIC1a protein expression involves NF-κB signaling pathway. Consistent with their effects on ASIC1a protein expression and channel function, PKC inhibition protected NS20Y cells against acidosis-induced cytotoxicity, while PKC activation potentiated acidosis-induced cells injury. Together, these results indicate that ASIC1a protein expression and channel function are closely regulated by the activity of protein kinase C and its downstream signaling pathway(s).


Subject(s)
Acid Sensing Ion Channels/metabolism , NF-kappa B/metabolism , Protein Kinase C/metabolism , Signal Transduction , Animals , Carbazoles , Cell Line , Cerebral Cortex/cytology , Mice , Naphthalenes/pharmacology , Neurons/drug effects , Neurons/metabolism , Neurotoxicity Syndromes/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Biosynthesis/drug effects , Proteolysis/drug effects , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Ubiquitin/metabolism
10.
Curr Protein Pept Sci ; 21(10): 985-992, 2020.
Article in English | MEDLINE | ID: mdl-32503408

ABSTRACT

Transient receptor potential melastatin 7 (TRPM7), along with the closely related TRPM6, are unique channels that have dual operations: cation permeability and kinase activity. In contrast to the limited tissue distribution of TRPM6, TRPM7 is widely expressed among tissues and is therefore implicated in a variety of cellular functions physiologically and pathophysiologically. The discovery of TRPM7's unique structure imparting dual ion channel and kinase activities shed light onto novel and peculiar biological functions, such as Mg2+ homeostasis, cellular Ca2+ flickering, and even intranuclear transcriptional regulation by a cleaved kinase domain translocated to nuclei. Interestingly, at a higher level, TRPM7 participates in several biological processes in the nervous and cardiovascular systems, in which excitatory responses in neurons and cardiomyocytes are critical for their function. Here, we review the roles of TRPM7 in cells involved in the nervous and cardiovascular systems and discuss its potential as a future therapeutic target.


Subject(s)
Cardiovascular System/metabolism , Myocytes, Cardiac/metabolism , Nervous System/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/genetics , Stroke/metabolism , TRPM Cation Channels/genetics , Calcium/metabolism , Cardiotonic Agents/therapeutic use , Cardiovascular System/drug effects , Cardiovascular System/pathology , Cations, Divalent , Gene Expression , Homeostasis/drug effects , Homeostasis/genetics , Humans , Ion Transport/drug effects , Magnesium/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Nervous System/drug effects , Nervous System/pathology , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/therapeutic use , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Stroke/genetics , Stroke/pathology , Stroke/prevention & control , Synaptic Transmission , TRPM Cation Channels/chemistry , TRPM Cation Channels/metabolism
11.
Hippocampus ; 30(7): 715-723, 2020 07.
Article in English | MEDLINE | ID: mdl-32057164

ABSTRACT

Prohormone convertase 2 (PC2) is essential for the biosynthesis of many neuropeptides, including several of them in hippocampus. In mouse brain, lacking an enzymatically active PC2 (PC2-null) causes accumulation of many neuropeptides in their precursor or intermediate forms. Little is known about how a PC2-null state may affect the function of the hippocampus. In this study, adult PC2-null mice and their wildtype (WT) littermates were subjected to three analyses to determine possible changes associated with PC2-null at physiological, behavioral, and molecular levels, respectively, under normal and stressed conditions. Electrophysiological recordings of hippocampal slices were performed to measure evoked field-excitatory postsynaptic potentials (EPSP), long-term potentiation (LTP), and paired-pulse facilitation (PPF). Morris water maze (MWM) testing was conducted to examine behavioral changes that are indicative of hippocampal integrity. Quantitative mass spectrometry analysis was used to determine changes in the hippocampal proteome in response to a focal cerebral ischemic insult. We found that there were no significant differences in the threshold of evoked EPSPs between PC2-null and WT animals. However, an increase in LTP in both triggering rate and amplitude was observed in PC2-null mice, suggesting that PC2 may be involved in regulating synaptic strength. The PPF, on the other hand, showed a decrease in PC2-null mice, suggesting a presynaptic mechanism. Consistent with changes in LTP, PC2-null mice displayed decreased latencies in finding the escape platform in the MWM test. Further, after distal focal cerebral ischemia, the hippocampal proteomes incurred changes in both WT and PC2-null mice, with a prominent change in proteins associated with neurotransmission, exocytosis, and transport processes seen in the PC2-null but not WT mice. Taken together, our results suggest that PC2 is involved in regulating hippocampal synaptic plasticity, learning, and memory behaviors, as well as the hippocampal response to stresses originating in other regions of the brain.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Hippocampus/enzymology , Maze Learning/physiology , Proprotein Convertase 2/deficiency , Animals , Brain Ischemia/enzymology , Brain Ischemia/genetics , Female , Male , Mice , Mice, Knockout , Organ Culture Techniques , Proprotein Convertase 2/genetics
12.
Stroke ; 50(10): 2902-2911, 2019 10.
Article in English | MEDLINE | ID: mdl-31412757

ABSTRACT

Background and Purpose- Sex differences in the incidence and outcome of stroke have been well documented. The severity of stroke in women is, in general, significantly lower than that in men, which is mediated, at least in part, by the protective effects of ß-estradiol. However, the detailed mechanisms underlying the neuroprotection by ß-estradiol are still elusive. Recent studies have demonstrated that activation of ASIC1a (acid-sensing ion channel 1a) by tissue acidosis, a common feature of brain ischemia, plays an important role in ischemic brain injury. In the present study, we assessed the effects of ß-estradiol on acidosis-mediated and ischemic neuronal injury both in vitro and in vivo and explored the involvement of ASIC1a and underlying mechanism. Methods- Cultured neurons and NS20Y cells were subjected to acidosis-mediated injury in vitro. Cell viability and cytotoxicity were measured by methylthiazolyldiphenyl-tetrazolium bromide and lactate dehydrogenase assays, respectively. Transient (60 minutes) focal ischemia in mice was induced by suture occlusion of the middle cerebral artery in vivo. ASIC currents were recorded using whole-cell patch-clamp technique while intracellular Ca2+ concentration was measured with fluorescence imaging using Fura-2. ASIC1a expression was detected by Western blotting and quantitative real-time polymerase chain reaction. Results- Treatment of neuronal cells with ß-estradiol decreased acidosis-induced cytotoxicity. ASIC currents and acid-induced elevation of intracellular Ca2+ were all attenuated by ß-estradiol treatment. In addition, we showed that ß-estradiol treatment reduced ASIC1a protein expression, which was mediated by increased protein degradation, and that estrogen receptor α was involved. Finally, we showed that the level of ASIC1a protein expression in brain tissues and the degree of neuroprotection by ASIC1a blockade were lower in female mice, which could be attenuated by ovariectomy. Conclusions- ß-estradiol can protect neurons against acidosis-mediated neurotoxicity and ischemic brain injury by suppressing ASIC1a protein expression and channel function. Visual Overview- An online visual overview is available for this article.


Subject(s)
Acid Sensing Ion Channels/metabolism , Estradiol/pharmacology , Neurons/drug effects , Stroke/metabolism , Acidosis/complications , Animals , Brain Ischemia/complications , Brain Ischemia/metabolism , Brain Ischemia/pathology , Female , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Stroke/etiology , Stroke/pathology
13.
Brain Circ ; 5(1): 8-11, 2019.
Article in English | MEDLINE | ID: mdl-31001594

ABSTRACT

Stroke is a leading cause of death and long-term disabilities. Despite decades of extensive efforts in search of brain injury mechanisms and therapeutic interventions, pharmacological treatment is limited to the use of thrombolytic agent tissue plasminogen activator, which has limited therapeutic time window and potential side effect of intracranial hemorrhage. Over the past few years, endovascular thrombectomy with stent-retriever devices combined with advanced imaging modalities has transformed the standard of stroke care, offering an opportunity to improve the outcome in selected patients as late as 24 h after the onset of stroke. This mini-review summarizes the advancement in the treatment of ischemic stroke, from thrombolysis to thrombectomy and remaining challenges in the field.

14.
Mol Neurobiol ; 56(5): 3326-3340, 2019 May.
Article in English | MEDLINE | ID: mdl-30120732

ABSTRACT

Tissue acidosis is a common feature of brain ischemia which causes neuronal injury. Activation of acid-sensing ion channel 1a (ASIC1a) plays an important role in acidosis-mediated neurotoxicity. Acute ethanol administration has been shown to provide neuroprotective effects during ischemic stroke, but the precise mechanisms have yet to be determined. In this study, we investigated the effect of ethanol on the activity/expression of ASIC1a channels and acidosis-induced neurotoxicity. We showed that acute treatment of neuronal cells with ethanol for more than 3 h could reduce ASIC1a protein expression, ASIC currents, and acid-induced [Ca2+]i elevation. We further demonstrated that ethanol-induced reduction of ASIC1a expression is mediated by autophagy-lysosome pathway (ALP)-dependent protein degradation. Finally, we showed that ethanol protected neuronal cells against acidosis-induced cytotoxicity, which effect was mimicked by autophagy activator rapamycin and abolished by autophagy inhibitor CQ. Together, these results indicate that moderate acute ethanol exposure can promote autophagy-lysosome pathway-dependent ASIC1a protein degradation and protect against acidosis-induced neurotoxicity.


Subject(s)
Acid Sensing Ion Channels/metabolism , Acidosis/complications , Autophagy , Ethanol/adverse effects , Lysosomes/metabolism , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , Proteolysis , Animals , Apoptosis/drug effects , Autophagy/drug effects , Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/drug effects , Ion Channel Gating/drug effects , Lysosomes/drug effects , MAP Kinase Signaling System/drug effects , Mice , Neurons/drug effects , Neurons/metabolism , Proteolysis/drug effects , Sodium Channels/metabolism
15.
J Cell Physiol ; 234(3): 2194-2203, 2019 03.
Article in English | MEDLINE | ID: mdl-30229908

ABSTRACT

Noncoding RNAs (ncRNAs) were initially thought to be transcriptional byproducts. However, recent advances of ncRNAs research have increased our understanding of the importance of ncRNA in gene regulation and disease pathogenesis. Consistent with these developments, liver fibrosis research is also experiencing rapid growth in the investigation of links between ncRNAs and the pathology of this disease. The initial focus was on studying the function and regulation mechanisms of microRNAs (miRNAs). However, recently, elucidation of the mechanisms of long noncoding RNAs (lncRNAs) and lncRNA-mediated liver fibrosis has just commenced. In this review, we emphasize on abnormal expression of lncRNAs in liver fibrosis. Furthermore, we also discuss that the interaction of lncRNAs with miRNAs is involved in the regulation of the expression of protein-coding genes in liver fibrosis. Recent advances in understanding dysregulated lncRNAs expression and the lncRNAs-miRNAs interaction in liver fibrosis will help for developing new therapeutic targets and biomarkers of liver fibrosis.


Subject(s)
Liver Cirrhosis/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Biomarkers/metabolism , Gene Expression Regulation/genetics , Humans , Liver Cirrhosis/pathology
16.
J Biol Chem ; 293(37): 14393-14406, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30076216

ABSTRACT

High-glucose (HG) levels and hyperglycemia associated with diabetes are known to cause neuronal damage. The detailed molecular mechanisms, however, remain to be elucidated. Here, we investigated the role of transient receptor potential melastatin 7 (TRPM7) channels in HG-mediated endoplasmic reticulum stress (ERS) and injury of NS20Y neuronal cells. The cells were incubated in the absence or presence of HG for 48 h. We found that mRNA and protein levels of TRPM7 and of ERS-associated proteins, such as C/EBP homologous protein (CHOP), 78-kDa glucose-regulated protein (GRP78), and inducible nitric-oxide synthase (iNOS), increased in HG-treated cells, along with significantly increased TRPM7-associated currents in these cells. Similar results were obtained in cerebral cortical tissue from an insulin-deficiency model of diabetic mice. Moreover, HG treatment of cells activated ERS-associated proapoptotic caspase activity and induced cellular injury. Interestingly, a NOS inhibitor, l-NAME, suppressed the HG-induced increase of TRPM7 expression and cellular injury. siRNA-mediated TRPM7 knockdown or chemical inhibition of TRPM7 activity also suppressed HG-induced ERS and decreased cleaved caspase-12/caspase-3 levels and cell injury. Of note, TRPM7 overexpression increased ERS and cell injury independently of its kinase activity. Taken together, our findings suggest that TRPM7 channel activities play a key role in HG-associated ERS and cytotoxicity through an apoptosis-inducing signaling cascade involving HG, iNOS, TRPM7, ERS proteins, and caspases.


Subject(s)
Apoptosis/physiology , Endoplasmic Reticulum Stress/physiology , Glucose/metabolism , Neurons/cytology , TRPM Cation Channels/physiology , Animals , Brain/metabolism , Caspases/metabolism , Diabetes Mellitus, Experimental/metabolism , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism
17.
CNS Neurosci Ther ; 24(9): 828-836, 2018 09.
Article in English | MEDLINE | ID: mdl-29962085

ABSTRACT

AIMS: It is unclear whether the impaired BRS plays a key role in the incidence of cardiovascular diseases. The molecular mechanism of impaired BRS remains to be fully elucidated. We hypothesized that selection of rats based on deficient and normal intrinsic BRS would yield models that reflect cardiovascular diseases risk. METHODS AND RESULTS: Twenty generations of selection produced arterial baroreflex low rats and normal rats that differed in BRS by about 2.5-fold change. Metabolic syndrome (including hypertension, overweight, hyperlipemia, and hyperglycemia) emerged in ABR-DRs. Although ABR-DRs consumed less food, they gained significantly more body weight. CONCLUSION: Our study demonstrated that intrinsic low BRS induced hypertension and metabolic disorder. Restoration of impaired BRS might be a potent target of therapeutic intervention in metabolic syndrome.


Subject(s)
Animals, Genetically Modified/genetics , Baroreflex/physiology , Blood Pressure/physiology , Heart Rate/physiology , Metabolic Syndrome/genetics , Selective Breeding/genetics , Animals , Female , Hypertension/genetics , Hypertension/physiopathology , Male , Metabolic Syndrome/physiopathology , Rats , Rats, Sprague-Dawley
18.
Behav Brain Res ; 337: 246-251, 2018 Jan 30.
Article in English | MEDLINE | ID: mdl-28912013

ABSTRACT

Acid-sensing ion channels (ASICs) are cation channels activated by protons. ASIC1a, a primary ASIC subunit in the brain, was recently characterized in the olfactory bulb. The present study tested the hypothesis that ASIC1a is essential for normal olfactory function. Olfactory behavior of wild-type (WT) and ASIC1-/- mice was evaluated by using three standard olfactory tests: (1) the buried food test, (2) the olfactory habituation test, and (3) the olfactory preference test. In buried food test, ASIC1-/- mice had significantly longer latency to uncover buried food than WT mice. In olfactory habituation test, ASIC1-/- mice had increased sniffing time with acidic odorants. In olfactory preference test, ASIC1-/- mice did not exhibit normal avoidance behavior for 2, 5- dihydro-2, 4, 5-trimethylthiazoline (TMT). Consistent with ASIC1 knockout, ASIC1 inhibition by nasal administration of PcTX1 increased the latency for WT mice to uncover the buried food. Together, these findings suggest a key role for ASIC1a in normal olfactory function.


Subject(s)
Acid Sensing Ion Channels/metabolism , Smell/genetics , Acid Sensing Ion Channels/genetics , Administration, Intranasal , Animals , Anti-Inflammatory Agents/pharmacology , Feeding Behavior/drug effects , Feeding Behavior/physiology , Gluconates/pharmacology , Habituation, Psychophysiologic/drug effects , Habituation, Psychophysiologic/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptides/pharmacology , Reaction Time/drug effects , Reaction Time/genetics , Smell/drug effects , Spider Venoms/pharmacology , Thiazoles/administration & dosage
19.
J Mol Neurosci ; 62(1): 79-87, 2017 May.
Article in English | MEDLINE | ID: mdl-28386642

ABSTRACT

Carboxypeptidase E (CPE), first discovered as a prohormone processing enzyme, has also now been shown to be a secreted neurotrophic factor (neurotrophic factor-α1, NF-α1) that acts extracellularly as a signaling molecule to mediate neuroprotection, cortical stem cell differentiation, and antidepressive-like behavior in mice. Since brain-derived neurotrophic factor (BDNF) has very similar trophic functions, and its processing from pro-BDNF involves intracellular sorting of pro-BDNF to the regulated secretory pathway by CPE acting as a sorting receptor, we investigated whether the lack of CPE/NF-α1 would affect BDNF-TrkB signaling in mice. Previous studies have shown that CPE/NF-α1 knock-out (KO) mice exhibited severe neurodegeneration of the hippocampal CA3 region which raises the question of why other neurotrophic factors such as BDNF could not compensate for the deficiency of CPE. Here, we show that the expressions of pro-BDNF mRNA and protein in hippocampus of CPE-KO mice were similar to WT mice, but mature BDNF was ∼40% less in the CPE-KO mice, suggesting decreased intracellular processing of pro-BDNF. Furthermore, TrkB receptor levels were similar in both genotypes, but there was significantly decreased phosphorylation of TrkB receptor in the CPE-KO mice. Electrophysiological studies showed lack of formation of long-term potentiation in hippocampal slices of CPE-KO mice compared to WT mice, which was not rescued by application of BDNF, indicating dysfunction of the BDNF-TrkB signaling system. The CPE-KO mice showed normal postsynaptic AMPA response to kainate application in hippocampal slices and dissociated neurons. Our findings indicate that CPE/NF-α1 is essential for normal BDNF-TrkB signaling function in mouse hippocampus.


Subject(s)
CA3 Region, Hippocampal/metabolism , Carboxypeptidase H/genetics , Receptor, trkB/metabolism , Signal Transduction , Animals , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , CA3 Region, Hippocampal/cytology , Carboxypeptidase H/metabolism , Cells, Cultured , Excitatory Amino Acid Agonists/pharmacology , Kainic Acid/pharmacology , Long-Term Potentiation , Mice , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Receptors, AMPA/agonists
20.
J Cereb Blood Flow Metab ; 37(2): 528-540, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26861816

ABSTRACT

Acidosis in the brain plays a critical role in neuronal injury in neurological diseases, including brain ischemia. One key mediator of acidosis-induced neuronal injury is the acid-sensing ion channels (ASICs). Current literature has focused on ASIC1a when studying acid signaling. The importance of ASIC2, which is also widely expressed in the brain, has not been appreciated. We found here a region-specific effect of ASIC2 on acid-mediated responses. Deleting ASIC2 reduced acid-activated current in cortical and striatal neurons, but had no significant effect in cerebellar granule neurons. In addition, we demonstrated that ASIC2 was important for ASIC1a expression, and that ASIC2a but not 2b facilitated ASIC1a surface trafficking in the brain. Further, we showed that ASIC2 deletion attenuated acidosis/ischemia-induced neuronal injury in organotypic hippocampal slices but had no effect in organotypic cerebellar slices. Consistent with an injurious role of ASIC2, we showed that ASIC2 deletion significantly protected the mouse brain from ischemic damage in vivo. These data suggest a critical region-specific contribution of ASIC2 to neuronal injury and reveal an important functional difference between ASIC2a and 2b in the brain.


Subject(s)
Acid Sensing Ion Channels/metabolism , Brain Ischemia/pathology , Brain/pathology , Neurons/pathology , Acid Sensing Ion Channels/analysis , Acid Sensing Ion Channels/genetics , Acidosis , Animals , Brain/metabolism , Brain Ischemia/genetics , Brain Ischemia/metabolism , Gene Deletion , Male , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Neurons/metabolism , Neuroprotection
SELECTION OF CITATIONS
SEARCH DETAIL
...