Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Cereb Blood Flow Metab ; 42(8): 1349-1363, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35301897

RESUMO

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.


Assuntos
Isquemia Encefálica , Prótons , Canais Iônicos Sensíveis a Ácido/metabolismo , Animais , Concentração de Íons de Hidrogênio , Mamíferos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
2.
Acta Pharmacol Sin ; 42(8): 1248-1255, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33184449

RESUMO

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.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Peróxido de Hidrogênio/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Animais , Butadienos/farmacologia , Linhagem Celular Tumoral , Imidazóis/farmacologia , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Nitrilas/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Regulação para Cima/efeitos dos fármacos
3.
Theranostics ; 10(26): 11976-11997, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33204324

RESUMO

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.


Assuntos
Envelhecimento/patologia , Doenças Cardiovasculares/tratamento farmacológico , Diabetes Mellitus/tratamento farmacológico , Ferroptose/efeitos dos fármacos , Doenças Neurodegenerativas/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/efeitos dos fármacos , Animais , Doenças Cardiovasculares/patologia , Linhagem Celular Tumoral , Diabetes Mellitus/patologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Ferroptose/fisiologia , Humanos , Ferro/metabolismo , Doenças Neurodegenerativas/patologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Doença Pulmonar Obstrutiva Crônica/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
4.
Stroke ; 50(10): 2902-2911, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31412757

RESUMO

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.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Estradiol/farmacologia , Neurônios/efeitos dos fármacos , Acidente Vascular Cerebral/metabolismo , Acidose/complicações , Animais , Isquemia Encefálica/complicações , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/patologia
5.
Mol Neurobiol ; 56(5): 3326-3340, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30120732

RESUMO

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.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Acidose/complicações , Autofagia , Etanol/efeitos adversos , Lisossomos/metabolismo , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle , Proteólise , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Cálcio/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação do Canal Iônico/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteólise/efeitos dos fármacos , Canais de Sódio/metabolismo
6.
Behav Brain Res ; 337: 246-251, 2018 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-28912013

RESUMO

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.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Olfato/genética , Canais Iônicos Sensíveis a Ácido/genética , Administração Intranasal , Animais , Anti-Inflamatórios/farmacologia , Comportamento Alimentar/efeitos dos fármacos , Comportamento Alimentar/fisiologia , Gluconatos/farmacologia , Habituação Psicofisiológica/efeitos dos fármacos , Habituação Psicofisiológica/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Peptídeos/farmacologia , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/genética , Olfato/efeitos dos fármacos , Venenos de Aranha/farmacologia , Tiazóis/administração & dosagem
7.
Biochim Biophys Acta Mol Basis Dis ; 1863(3): 674-686, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27979710

RESUMO

Long non-coding RNAs (lncRNAs) are increasingly recognized as major players in regulating various biological processes. LncRNA HOX transcript antisense RNA (Hotair) has been extensively studied in cancer. However, the role of Hotair in liver fibrosis remains unknown. Here we observed that Hotair expression was significantly increased in CCl4-induced mouse liver fibrosis models, human fibrotic livers and activated hepatic stellate cells (HSCs) by TGF-ß1 stimulation. Enforced expression of Hotair in LX-2 cells promoted cell proliferation and activation while inhibition of its expression had an opposite effect. Furthermore, we found that Hotair may act as an endogenous 'sponge' of miR-148b, which regulates expression of the DNMT1/MEG3/p53 pathways in HSCs. Intriguingly, Hotair enhanced polycomb repressive complex 2 (PRC2) occupancy and histone H3K27me3 repressive marks, specifically at the MEG3 promoter region. Finally, we found that Hotair forms an RNA/DNA hybrid and recruits PRC2 to MEG3 promoter. These data suggest that Hotair inhibition may represent a promising therapeutic option for suppressing liver fibrosis.


Assuntos
Células Estreladas do Fígado/metabolismo , Cirrose Hepática/genética , RNA Longo não Codificante/genética , Regulação para Cima , Animais , Linhagem Celular , DNA (Citosina-5-)-Metiltransferase 1/genética , Epigênese Genética , Regulação da Expressão Gênica , Células Estreladas do Fígado/patologia , Humanos , Cirrose Hepática/patologia , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/genética
8.
Oncotarget ; 8(5): 8283-8293, 2017 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-28030818

RESUMO

In neurons, up-regulation of Notch activity either inhibits neurite extension or causes retraction of neurites. Conversely, inhibition of Notch1 facilitates neurite extension. Acid-sensing ion channels (ASICs) are a family of proton-gated cation channels, which play critical roles in synaptic plasticity, learning and memory and spine morphogenesis. Our pilot proteomics data from ASIC1a knock out mice implicated that ASIC1a may play a role in regulating Notch signaling, therefore, we explored whether or not ASIC1a regulates neurite growth during neuronal development through Notch signaling. In this study, we determined the effects of ASIC1a on neurite growth in a mouse neuroblastoma cell line, NS20Y cells, by modulating ASIC1a expression. We also determined the relationship between ASIC1a and Notch signaling on neuronal differentiation. Our results showed that down-regulation of ASIC1a in NS20Y cells inhibits CPT-cAMP induced neurite growth, while over expression of ASIC1a promotes its growth. In addition, down-regulation of ASIC1a increased the expression of Notch1 and its target gene Survivin while inhibitor of Notch significantly prevented the neurite extension induced by ASIC1a in NS20Y cells. These data indicate that Notch1 signaling may be required for ASIC1a-mediated neurite growth and neuronal differentiation.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Neuritos/metabolismo , Neuroblastoma/metabolismo , Crescimento Neuronal , Receptor Notch1/metabolismo , Transdução de Sinais , Canais Iônicos Sensíveis a Ácido/genética , Animais , Linhagem Celular Tumoral , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Dipeptídeos , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Camundongos , Neuritos/efeitos dos fármacos , Neuritos/patologia , Neuroblastoma/genética , Neuroblastoma/patologia , Crescimento Neuronal/efeitos dos fármacos , Interferência de RNA , Receptor Notch1/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção
9.
CNS Neurosci Ther ; 23(3): 216-221, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27917616

RESUMO

BACKGROUND: The algal protein Channelrhodopsin-2 (ChR2) has been widely used in recent years in optogenetic technique to investigate the functions of complex neuronal networks through minimally invasive and temporally precise photostimulation of genetically defined neurons. However, as with any other new technique, current optogentic approaches have various limitations. In addition, how ChR2 may behave in response to complex biochemical changes associated with various physiological/pathological conditions is largely unknown. AIM: In this study, we investigated whether a change in redox state of the cell affects the activity of ChR2 channels. METHODS: Whole-cell patch-clamp recordings were used to examine the effect of reducing and oxidizing agents on ChR2 currents activated by blue light. RESULTS: We show that the reducing agent dithiothreitol (DTT) dramatically potentiates the ChR2 currents in a reversible and concentration-dependent manner. Glutathione, an endogenous reducing agent, shows a similar effect on ChR2 currents. The oxidizing agent 5,5'-dithio-bis-(2-nitrobenzoic acid) (DTNB) has no effect on ChR2 currents by itself; however, it completely reverses the potentiating effect of DTT. DTT also causes a shift in the current-voltage relationship by 23 ± 4.31 mV, suggesting a change in ion selectivity. CONCLUSION: Taken together, these data suggest that redox modification of ChR2 plays an important role in its sensitivity to the light stimulation. Our findings not only help for a better understanding of how ChR2 may behave in physiological/pathological conditions where changes in redox state are common, but also provide a new direction for further optimization of this important opsin.


Assuntos
Ditiotreitol/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Substâncias Redutoras/farmacologia , Animais , Biofísica , Células CHO , Channelrhodopsins , Cricetulus , Ácido Ditionitrobenzoico/farmacologia , Relação Dose-Resposta a Droga , Estimulação Elétrica , Glutationa/farmacologia , Luz , Oxirredução/efeitos dos fármacos , Técnicas de Patch-Clamp , Transfecção
10.
Mol Brain ; 9(1): 68, 2016 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-27342076

RESUMO

Cultured neuronal cell lines can express properties of mature neurons if properly differentiated. Although the precise mechanisms underlying neuronal differentiation are not fully understood, the expression and activation of ion channels, particularly those of Ca(2+)-permeable channels, have been suggested to play a role. In this study, we explored the presence and characterized the properties of acid-sensing ion channels (ASICs) in NS20Y cells, a neuronal cell line previously used for the study of neuronal differentiation. In addition, the potential role of ASICs in cell differentiation was explored. Reverse Transcription Polymerase Chain Reaction and Western blot revealed the presence of ASIC1 subunits in these cells. Fast drops of extracellular pH activated transient inward currents which were blocked, in a dose dependent manner, by amiloride, a non-selective ASIC blocker, and by Psalmotoxin-1 (PcTX1), a specific inhibitor for homomeric ASIC1a and heteromeric ASIC1a/2b channels. Incubation of cells with PcTX1 significantly reduced the differentiation of NS20Y cells induced by cpt-cAMP, as evidenced by decreased neurite length, dendritic complexity, decreased expression of functional voltage gated Na(+) channels. Consistent with ASIC1a inhibition, ASIC1a knockdown with small interference RNA significantly attenuates cpt-cAMP-induced increase of neurite outgrowth. In summary, we described the presence of functional ASICs in NS20Y cells and demonstrate that ASIC1a plays a role in the differentiation of these cells.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Diferenciação Celular , Neurônios/citologia , Neurônios/metabolismo , Canais Iônicos Sensíveis a Ácido/genética , Amilorida/farmacologia , Animais , Células CHO , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cricetinae , Cricetulus , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Ativação do Canal Iônico/efeitos dos fármacos , Camundongos , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Neurogênese/efeitos dos fármacos , Neurogênese/genética , Neurônios/efeitos dos fármacos , Peptídeos/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Canais de Sódio/metabolismo , Venenos de Aranha/farmacologia , Tionucleotídeos/farmacologia
11.
J Neurochem ; 138(2): 354-61, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27123541

RESUMO

Increased expression of serum- and glucocorticoid-inducible kinase 1 (SGK1) can be induced by stress and growth factors in mammals, and plays an important role in cancer, diabetes, and hypertension. A recent work suggested that SGK1 activity restores damage in a stroke model. To further investigate the role of SGKs in ischemic brain injury, we examined how SGK inhibitors influence stroke outcome in vivo and neurotoxicity in vitro. Infarct volumes were compared in adult mice with middle cerebral artery occlusion, followed by 24 h reperfusion, in the absence or presence of SGK inhibitors. Neurotoxicity assay, electrophysiological recording, and fluorescence Ca(2+) imaging were carried out using cultured cortical neurons to evaluate the underlying mechanisms. Contrary to our expectation, infarct volume by stroke decreased significantly when SGK inhibitor, gsk650394, or EMD638683, was administrated 30 min before middle cerebral artery occlusion under normal and diabetic conditions. SGK inhibitors reduced neurotoxicity mediated by N-methyl-D-aspartate (NMDA) receptors, a leading factor responsible for cell death in stroke. SGK inhibitors also ameliorated Ca(2+) increase and peak amplitude of NMDA current in cultured neurons. In addition, SGK inhibitor gsk650394 decreased phosphorylation of Nedd4-2 and inhibited voltage-gated sodium currents. These observations suggest that SGK activity exacerbates stroke damage and that SGK inhibitors may be useful candidates for therapeutic intervention. To investigate the role of serum- and glucocorticoid-inducible kinases (SGKs) in ischemic brain injury, we examined how SGK inhibitors influence stroke outcome. Infarct volumes induced by middle cerebral artery occlusion were decreased significantly by SGK inhibitors. The inhibitors also reduced glutamate toxicity, at least partly, by attenuation of NMDA and voltage-gated sodium currents. Thus, SGK inhibition attenuates stroke damage.


Assuntos
Glucocorticoides/farmacologia , Proteínas Imediatamente Precoces/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Benzamidas/farmacologia , Hidrazinas/farmacologia , Proteínas Imediatamente Precoces/antagonistas & inibidores , Proteínas Imediatamente Precoces/sangue , Infarto da Artéria Cerebral Média/tratamento farmacológico , Masculino , Camundongos Endogâmicos C57BL , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico , Ubiquitina-Proteína Ligases/metabolismo
12.
CNS Neurosci Ther ; 21(3): 252-61, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25438992

RESUMO

BACKGROUND: Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor with a dismal prognosis. Despite intensive study on tumor biology, the underlying mechanisms of the unlimited proliferation and progressive local invasion are still poorly understood, and no effective treatment has been developed for GBM patients. AIMS: We determine the role of TRPM7 channels in the growth, migration, and infiltration of malignant glioma cells. METHODS: Using a combination of RT-PCR, Western blot, and patch-clamp techniques, we demonstrated the expression of functional TRPM7 channels of A172 cells, a human glioma cell line, as well as in human glioma tissues. Furthermore, we evaluated the role of TRPM7 in growth, migration, and infiltration of A172 cells with MTT and transwell migration and invasion assays. RESULTS: We showed the expression of functional TRPM7 channels in both A172 cells and human glioma tissues. Suppression of TRPM7 expression with TRPM7-siRNA dramatically reduced the proliferation, migration, and invasion of A172 cells. Pharmacological inhibition of TRPM7 channel with 2-aminoethoxydiphenyl borate (2-APB) showed a similar effect as TRPM7-siRNA. CONCLUSION: We demonstrate that human glioma cells express functional TRPM7 channel and that activation of this channel plays an important role in the proliferation, migration, and invasion of malignant glioma cells. TRPM7 channel may represent a novel and promising target for therapeutic intervention of malignant glioma.


Assuntos
Neoplasias Encefálicas/fisiopatologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Glioblastoma/fisiopatologia , Invasividade Neoplásica/fisiopatologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Canais de Cátion TRPM/antagonistas & inibidores , Western Blotting , Compostos de Boro/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Glioblastoma/tratamento farmacológico , Humanos , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/metabolismo , RNA Interferente Pequeno , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo
13.
Cell Signal ; 26(12): 2773-81, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25192910

RESUMO

Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor in adults with median survival time of 14.6 months. A small fraction of cancer stem cells (CSC) initiate and maintain tumors thus driving glioma tumorigenesis and being responsible for resistance to classical chemo- and radio-therapies. It is desirable to identify signaling pathways related to CSC to develop novel therapies to selectively target them. Transient receptor potential cation channel, subfamily M, member 7, also known as TRPM7 is a ubiquitous, Ca(2+) and Mg(2+) permeable ion channels that are special in being both an ion channel and a serine/threonine kinase. In studies of glioma cells silenced for TRPM7, we demonstrated that Notch (Notch1, JAG1, Hey2, and Survivin) and STAT3 pathways are down regulated in glioma cells grown in monolayer. Furthermore, phospho-STAT3, Notch target genes and CSC markers (ALDH1 and CD133) were significantly higher in spheroid glioma CSCs when compared with monolayer cultures. The results further show that tyrosine-phosphorylated STAT3 binds and activates the ALDH1 promoters in glioma cells. We found that TRMP7-induced upregulation of ALDH1 expression is associated with increases in ALDH1 activity and is detectable in stem-like cells when expanded as spheroid CSCs. Finally, TRPM7 promotes proliferation, migration and invasion of glioma cells. These demonstrate that TRPM7 activates JAK2/STAT3 and/or Notch signaling pathways and leads to increased cell proliferation and migration. These findings for the first time demonstrates that TRPM7 (1) activates a previously unrecognized STAT3→ALDH1 pathway, and (2) promotes the induction of ALDH1 activity in glioma cells.


Assuntos
Glioma/metabolismo , Glioma/patologia , Células-Tronco Neoplásicas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Notch/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Canais de Cátion TRPM/metabolismo , Família Aldeído Desidrogenase 1 , Sequência de Bases , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular , Imunofluorescência , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Glioma/enzimologia , Glioma/genética , Humanos , Isoenzimas/genética , Modelos Biológicos , Dados de Sequência Molecular , Invasividade Neoplásica , Células-Tronco Neoplásicas/enzimologia , Células-Tronco Neoplásicas/patologia , Fosfotirosina/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Retinal Desidrogenase/genética , Frações Subcelulares/metabolismo , Regulação para Cima
14.
Curr Drug Targets ; 13(2): 263-71, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22204324

RESUMO

Protons are important signals for neuronal function. In the central nervous system (CNS), proton concentrations change locally when synaptic vesicles release their acidic contents into the synaptic cleft, and globally in ischemia, seizures, traumatic brain injury, and other neurological disorders due to lactic acid accumulation. The finding that protons gate a distinct family of ion channels, the acid-sensing ion channels (ASICs), has shed new light on the mechanism of acid signaling and acidosis-associated neuronal injury. Accumulating evidence has suggested that ASICs play important roles in physiological processes such as synaptic plasticity, learning/memory, fear conditioning, and retinal integrity, and in pathological conditions such as brain ischemia, multiple sclerosis, epileptic seizures, and malignant glioma. Thus, targeting these channels may lead to novel therapeutic interventions for neurological disorders. The goal of this review is to provide an update on recent advances in our understanding of the functions of ASICs in the CNS.


Assuntos
Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Canais de Sódio/metabolismo , Canais Iônicos Sensíveis a Ácido , Animais , Humanos , Prótons
15.
Artigo em Inglês | MEDLINE | ID: mdl-21760970

RESUMO

Tumor necrosis factor-α (TNFα) is a pleiotropic cytokine that can regulate cell survival, inflammation or, under certain circumstances, trigger cell death. Previous work in rat seizure models and analysis of temporal lobe samples from epilepsy patients has suggested seizures activate TNF receptor 1 (TNFR1). Here we explored the activation and functional significance of TNFR1 signaling in the mouse hippocampus using in vitro and in vivo models of seizure-induced neuronal injury. Focal-onset status epilepticus in mice upregulated TNFR1 levels and led to formation of TNFR1-TNFR-associated death domain (TRADD) and TRADD-Fas-associated death domain (FADD) binding. Seizure-like injury modeled in vitro by removal of chronic excitatory blockade in mouse hippocampal neurons also activated this TNFR1 signaling pathway. Prior exposure of hippocampal neurons to a non-harmful seizure episode, via NMDA receptor blockade, 24 h prior to injurious seizures significantly reduced cell death and modeled epileptic tolerance in vitro. TNFR1 complex formation with TRADD and TRADD-FADD binding were reduced in tolerant cells. Finally, TNFR1 signaling and cell death were reduced by PKF-242-484, a dual matrix metaloproteinase/TNFα converting enzyme inhibitor. The present study shows that TNFR1 signaling is activated in mouse seizure models and may contribute to neuropathology in vitro and in vivo while suppression of this pathway may underlie neuroprotection in epileptic tolerance.

16.
Artigo em Inglês | MEDLINE | ID: mdl-21760973

RESUMO

Cancer is a leading cause of death in the world. Conventional treatments have severe side effects and low survival rate. It is important to discover new targets and therapeutic strategies to improve the clinical outcomes of cancer patients. Ion channels are specialized membrane proteins that play important roles in various physiological processes. Recent studies have shown that abnormal expression and/or activity of a number of ion channels e.g. voltage-gated K(+), Na(+), Ca(2+) channels, TRP channels, and epithelial Na(+)/degenerin family of ion channels, are involved in the growth/proliferation, migration and/or invasion of cancer cells. In this review, we summarize the present knowledge about the roles of different ion channels in the development of cancer.

17.
Sci Signal ; 3(111): ra15, 2010 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-20197544

RESUMO

Exposing the brain to sublethal ischemia affects the response to a subsequent, otherwise injurious ischemia, resulting in transcriptional suppression and neuroprotection, a response called ischemic tolerance. Here, we show that the proteomic signature of the ischemic-tolerant brain is characterized by increased abundance of transcriptional repressors, particularly polycomb group (PcG) proteins. Knocking down PcG proteins precluded the induction of ischemic tolerance, whereas in an in vitro model, overexpressing the PcG proteins SCMH1 or BMI1 induced tolerance to ischemia without preconditioning. We found that PcG proteins are associated with the promoter regions of genes encoding two potassium channel proteins that show decreased abundance in ischemic-tolerant brains. Furthermore, PcG proteins decreased potassium currents in cultured neuronal cells, and knocking down potassium channels elicited tolerance without preconditioning. These findings reveal a previously unknown mechanism of neuroprotection that involves gene repressors of the PcG family.


Assuntos
Isquemia Encefálica/fisiopatologia , Proteínas Repressoras/genética , Proteínas Repressoras/fisiologia , Animais , Sequência de Bases , Isquemia Encefálica/genética , Epigênese Genética , Técnicas In Vitro , Canal de Potássio Kv1.5/genética , Canal de Potássio Kv1.5/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/metabolismo , Proteínas Nucleares/genética , Complexo Repressor Polycomb 1 , Proteínas do Grupo Polycomb , Regiões Promotoras Genéticas , Proteômica , Proteínas Proto-Oncogênicas/genética , RNA Interferente Pequeno/genética , Proteínas Repressoras/antagonistas & inibidores , Superfamília Shaker de Canais de Potássio/genética , Superfamília Shaker de Canais de Potássio/metabolismo , Transdução de Sinais
18.
Rhinology ; 47(2): 187-91, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19593977

RESUMO

OBJECTIVE: To study the immune function of adult Chinese patients with chronic rhinosinusitis (CRS) to elucidate its potential role in the pathogenesis of CRS. METHODS: A prospective three-arm case-control study. The study population comprised 72 CRS patients without nasal polyps (NPs), 95 CRS patients with NPs, and 110 healthy controls. The concentrations of serum immunoglobulin A (IgA), M (IgM), G (IgG), IgG subclasses (IgG1-4), complement component 3 (C3), and complement component 4 (C4) were measured by nephelometry. Serum mannose-binding lectin (MBL) levels were analyzed by enzyme-linked immunosorbent assay. All CRS patients had a complete blood count with differential, atopic status evaluation, coronal computed tomographic (CT) scan of the sinuses, and nasal endoscopy. RESULTS: Frequency of immunoglobulin, C3, C4, or MBL deficiency showed no difference among groups. The prevalence of coexistence of MBL and immunoglobulin or complement component deficiency did not differ significantly among groups either. However, compared with controls, decreased IgG3 levels were found in CRS patients without NPs, and increased C3 and MBL levels was found in both CRS patients with and without NPs. Moreover, MBL levels were significantly higher in CRS patients with NPs than in CRS patients without NPs, which positively correlated with extent of disease seen on CT scan and endoscopy, and peripheral eosinophil count. CONCLUSIONS: Immunoglobulin, C3, C4, and MBL deficiency is not the main cause of CRS in adult Chinese patients. However, on the contrary, increased C3 and MBL levels in serum might play a modulatory role in CRS development.


Assuntos
Complemento C3/metabolismo , Lectina de Ligação a Manose/sangue , Rinite/sangue , Sinusite/sangue , Estudos de Casos e Controles , Distribuição de Qui-Quadrado , China , Doença Crônica , Ensaio de Imunoadsorção Enzimática , Humanos , Pólipos Nasais/sangue , Estudos Prospectivos , Rinite/imunologia , Sinusite/imunologia , Estatísticas não Paramétricas
19.
Artigo em Chinês | MEDLINE | ID: mdl-18826101

RESUMO

OBJECTIVE: To detect the presence of ion channel protein and its role in cell growth and proliferation in human head and neck squamous carcinoma cells (SCC). METHODS: Human head and neck squamous carcinoma SCC-25 cell line was tested with transient receptor potential melastatin 7 (TRPM7) antibody using the method of immunocytochemistry. The role of TRPM7 in cell growth and proliferation was evaluated through its blockade by ion channel blockers and specific siRNA using lactate dehydrogenase (LDH) assay technique. RESULTS: Clear immunoreactivity against TRPM7 was detected in almost all SCC-25 cells tested, whereas no immunoreactivity was observed in negative control. The inhibitory effect of Gd3+, a non-specific ion channel blocker, on cell growth and proliferation was potent. Addition of 10 micromol/L Gd3+ (n = 16) and 100 micromol/L Gd3+ (n = 16) in the culture medium significantly inhibited the growth of SCC-25 cells, as compared with control cells growing in normal medium (t was 4.1414 and 6.2661, P was 0.0256 and 0.0082 respectively). However, the effect of 2-APB was striking. Cell proliferation was almost totally suppressed in the presence of 100 micromol/L 2-APB (t = 13.4493, P = 0.0008, n = 16) compared with cells growing in normal medium. Suppression of TRPM7 expression by siRNA also significantly inhibited the growth and proliferation of these cells (t = 4.3446, P = 0.0002, n = 32, compared with nontransfected cells),whereas cells transfected with negative control siRNA showed no difference in cell proliferation compared with nontransfected cells. CONCLUSIONS: All of those results strongly suggest the existence of TRPM7 channel in human head and neck squamous carcinoma cells. Ion channel blockers serve as a potent inhibitor of SCC-25 cell proliferation. The striking inhibitory effect of 2-APB on cell growth and proliferation may promise clinical workers an inspiring remedy for fighting against carcinoma.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Canais de Cátion TRPM/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Proteínas Serina-Treonina Quinases
20.
Am J Rhinol ; 22(4): 376-80, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18702901

RESUMO

BACKGROUND: Clara cell 10-kDa protein (CC10) is an anti-inflammatory molecule and has been implicated in the involvement of the pathogenesis of asthma and chronic rhinosinusitis (CRS). A single nucleotide polymorphism (SNP) in CC10 gene (A + 38G) was previously shown to be associated with asthma and plasma CC10 levels. The purpose of this study is to examine whether there is an association between the CC10 A + 38G SNP, plasma CC10 levels, and CRS in a central Chinese population of Han nationality. METHODS: The CC10 A + 38G SNP was analyzed by means of polymerase chain reaction with restriction fragment length polymorphism and plasma CC10 levels were measured using enzyme-linked immunosorbent assay in 220 patients with CRS (90 patients with nasal polyps [NPs] and 130 patients without NPs) and 180 healthy control subjects. Among 220 patients with CRS, 108 patients were atopic subjects. Severity of disease was determined by coronal computed tomography (CT) scan in CRS patients, which was graded according to Lund and Mackay. RESULTS: The frequency of the A allele was 0.394, which was not significantly higher than the frequencies of other reported ethnic groups except for German. No association between the CC10 A + 38G SNP and CRS, any subgroup of CRS, or CRS severity could be found. Although subjects carrying the AA genotype had a significantly lower plasma CC10 concentration than those carrying the GG and GA genotypes in both CRS and control groups (p = 0.00 for all), no association was found between the plasma CC10 levels and CRS phenotype. CONCLUSION: The CC10 A + 38G SNP may not exert a substantial influence on the development of CRS in the Chinese Han population.


Assuntos
DNA/genética , Polimorfismo de Nucleotídeo Único , Rinite/genética , Sinusite/genética , Uteroglobina/genética , Adulto , China/epidemiologia , Doença Crônica , Inibidores Enzimáticos , Ensaio de Imunoadsorção Enzimática , Feminino , Frequência do Gene , Predisposição Genética para Doença , Genótipo , Humanos , Masculino , Prevalência , Rinite/epidemiologia , Rinite/metabolismo , Sinusite/epidemiologia , Sinusite/metabolismo , Uteroglobina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA