Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Kidney Blood Press Res ; 49(1): 114-123, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38246148

RESUMO

INTRODUCTION: A comprehensive pathophysiological mechanism to explain the relationship between high-salt intake and hypertension remains undefined. Evidence suggests that chloride, as the accompanying anion of sodium in dietary salt, is necessary to develop hypertension. We evaluated whether reducing dietary Cl- while keeping a standard Na+ intake modified blood pressure, cardiac hypertrophy, renal function, and vascular contractility after angiotensin II (AngII) infusion. METHODS: C56BL/6J mice fed with standard Cl- diet or a low-Cl- diet (equimolar substitution of Cl- by a mixture of Na+ salts, both diets with standard Na+ content) received AngII (infusion of 1.5 mg/kg/day) or vehicle for 14 days. We measured systolic blood pressure (SBP), glomerular filtration rate (GFR), natriuretic response to acute saline load, and contractility of aortic rings from mice infused with vehicle and AngII, in standard and low-Cl- diet. RESULTS: The mice fed the standard diet presented increased SBP and cardiac hypertrophy after AngII infusion. In contrast, low-Cl- diet prevented the increase of SBP and cardiac hypertrophy. AngII-infused mice fed a standard diet presented hampered natriuretic response to saline load, meanwhile the low-Cl- diet preserved natriuretic response in AngII-infused mice, without change in GFR. Aortic rings from mice fed with standard diet or low-Cl- diet and infused with AngII presented a similar contractile response. CONCLUSION: We conclude that the reduction in dietary Cl- as the accompanying anion of sodium in salt is protective from AngII pro-hypertensive actions due to a beneficial effect on kidney function and preserved natriuresis.


Assuntos
Angiotensina II , Pressão Sanguínea , Hipertensão , Rim , Animais , Angiotensina II/farmacologia , Camundongos , Hipertensão/induzido quimicamente , Hipertensão/prevenção & controle , Rim/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Taxa de Filtração Glomerular/efeitos dos fármacos , Cardiomegalia/prevenção & controle , Cardiomegalia/induzido quimicamente , Masculino , Cloreto de Sódio na Dieta/efeitos adversos , Cloreto de Sódio na Dieta/administração & dosagem , Camundongos Endogâmicos C57BL , Cloretos/administração & dosagem , Cloretos/farmacologia
2.
PLoS One ; 17(1): e0262628, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35015786

RESUMO

[This corrects the article DOI: 10.1371/journal.pone.0015658.].

3.
Rev. chil. anest ; 49(1): 168-171, 2020. ilus, tab
Artigo em Espanhol | LILACS | ID: biblio-1510430

RESUMO

Systemic toxicity by local anesthetics (LAs) is a severe and feared complication in anesthetic practice that generally results from the administration of an inappropriately high dose of LAs or an injection at an inappropriate place, either intravascular or a site with high absorption[1]. However, it is known that the susceptibility to these drugs may vary within the population, which may occur due to genetic changes in the LA binding site, located in the potential-dependent Na+ channels (Nav), thus increasing or decreasing its affinity and, therefore, its clinical consequences. We present a case of a 61 years-old female patient with a medical history of increased sensitivity to LAs. In this scenario, a genetic study was performed to exclude a Nav channel dysfunction.


La toxicidad sistémica por anestésicos locales (ALs) es una grave y temida complicación en la práctica anestésica que generalmente resulta de la administración de una dosis inapropiadamente alta de ALs o a una inyección en un lugar inadecuado, llámese intravascular o un sitio al alta absorción[1]. A pesar de lo anterior, es conocido que la susceptibilidad al efecto de estos fármacos puede variar dentro de la población, lo cual puede ocurrir debido a cambios genéticos en el sitio de unión de los AL, localizado en los canales de Na+ dependientes de potencial (Nav), incrementando o disminuyendo así su afinidad y, por ende, sus consecuencias clínicas. Presentamos el caso de una paciente de 61 años con historia de sensibilidad aumentada a Als. En este escenario, se le ofreció un estudio genético para excluir una disfunción específica a nivel de canal Nav


Assuntos
Humanos , Feminino , Pessoa de Meia-Idade , Canais de Sódio/efeitos dos fármacos , Hipersensibilidade a Drogas/etiologia , Hipersensibilidade a Drogas/genética , Anestésicos Locais/efeitos adversos , Lidocaína/efeitos adversos , Canais de Sódio/genética , Anestésicos Locais/farmacologia , Lidocaína/farmacologia
4.
J Cell Physiol ; 234(3): 2037-2050, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30343491

RESUMO

Transient Receptor Potential Melastatin 4 (TRPM4) is a Ca2+ -activated and voltage-dependent monovalent cation channel, which depolarizes the plasma cell membrane, thereby modulating Ca2+ influx across Ca2+ -permeable pathways. TRPM4 is involved in different physiological processes such as T cell activation and the migration of endothelial and certain immune cells. Overexpression of this channel has been reported in various types of tumors including prostate cancer. In this study, a significant overexpression of TRPM4 was found only in samples from cancer with a Gleason score higher than 7, which are more likely to spread. To evaluate whether TRPM4 overexpression was related to the spreading capability of tumors, TRPM4 was knockdown by using shRNAs in PC3 prostate cancer cells and the effect on cellular migration and invasion was analyzed. PC3 cells with reduced levels of TRPM4 (shTRPM4) display a decrease of the migration/invasion capability. A reduction in the expression of Snail1, a canonical epithelial to mesenchymal transition (EMT) transcription factor, was also observed. Consistently, these cells showed a significant change in the expression of key EMT markers such as MMP9, E-cadherin/N-cadherin, and vimentin, indicating a partial reversion of the EMT process. Whereas, the overexpression of TRPM4 in LnCaP cells resulted in increased levels of Snail1, reduction in the expression of E-cadherin and increase in their migration potential. This study suggests a new and indirect mechanism of regulation of migration/invasion process by TRPM4 in prostate cancer cells, by inducing the expression of Snail1 gene and consequently, increasing the EMT.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Canais de Cátion TRPM/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Masculino , Modelos Biológicos , Gradação de Tumores , Invasividade Neoplásica , Células PC-3 , Neoplasias da Próstata/genética , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/genética , Regulação para Cima
5.
Mol Oncol ; 12(2): 151-165, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28614631

RESUMO

Increased expression of the TRPM4 channel has been reported to be associated with the progression of prostate cancer. However, the molecular mechanism underlying its effect remains unknown. This work found that decreasing TRPM4 levels leads to the reduced proliferation of PC3 cells. This effect was associated with a decrease in total ß-catenin protein levels and its nuclear localization, and a significant reduction in Tcf/Lef transcriptional activity. Moreover, TRPM4 silencing increases the Ser33/Ser37/Thr41 ß-catenin phosphorylated population and reduces the phosphorylation of GSK-3ß at Ser9, suggesting an increase in ß-catenin degradation as the underlying mechanism. Conversely, TRPM4 overexpression in LNCaP cells increases the Ser9 inhibitory phosphorylation of GSK-3ß and the total levels of ß-catenin and its nonphosphorylated form. Finally, PC3 cells with reduced levels of TRPM4 showed a decrease in basal and stimulated phosphoactivation of Akt1, which is likely responsible for the decrease in GSK-3ß activity in these cells. Our results also suggest that the effect of TRPM4 on Akt1 is probably mediated by an alteration in the calcium/calmodulin-EGFR axis, linking TRPM4 activity with the observed effects in ß-catenin-related signaling pathways. These results suggest a role for TRPM4 channels in ß-catenin oncogene signaling and underlying mechanisms, highlighting this ion channel as a new potential target for future therapies in prostate cancer.


Assuntos
Proliferação de Células/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Canais de Cátion TRPM/metabolismo , beta Catenina/metabolismo , Cálcio/metabolismo , Calmodulina/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Glicogênio Sintase Quinase 3 beta/genética , Células HEK293 , Humanos , Masculino , Células PC-3 , Fosforilação/genética , Neoplasias da Próstata/genética , Proteínas Proto-Oncogênicas c-akt/genética , Canais de Cátion TRPM/genética , beta Catenina/genética
6.
Channels (Austin) ; 11(6): 624-635, 2017 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-28876976

RESUMO

Cerebral ischemia-reperfusion injury triggers a deleterious process ending in neuronal death. This process has two components, a glutamate-dependent and a glutamate-independent mechanism. In the glutamate-independent mechanism, neurons undergo a slow depolarization eventually leading to neuronal death. However, little is known about the molecules that take part in this process. Here we show by using mice cortical neurons in culture and ischemia-reperfusion protocols that TRPM4 is fundamental for the glutamate-independent neuronal damage. Thus, by blocking excitotoxicity, we reveal a slow activating, glibenclamide- and 9-phenanthrol-sensitive current, which is activated within 5 min upon ischemia-reperfusion onset. TRPM4 shRNA-based silenced neurons show a reduced ischemia-reperfusion induced current and depolarization. Neurons were protected from neuronal death up to 3 hours after the ischemia-reperfusion challenge. The activation of TRPM4 during ischemia-reperfusion injury involves the increase in both, intracellular calcium and H2O2, which may act together to produce a sustained activation of the channel.


Assuntos
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Neurônios/metabolismo , Neurônios/patologia , Oxigênio/metabolismo , Traumatismo por Reperfusão/patologia , Canais de Cátion TRPM/metabolismo , Animais , Morte Celular , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão/metabolismo
7.
PLoS One ; 10(9): e0139243, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26418693

RESUMO

Formylated peptides are chemotactic agents generated by pathogens. The most relevant peptide is fMLF (formyl-Met-Leu-Phe) which participates in several immune functions, such as chemotaxis, phagocytosis, cytokine release and generation of reactive oxygen species. In macrophages fMLF-dependent responses are dependent on both, an increase in intracellular calcium concentration and on a hyperpolarization of the membrane potential. However, the molecular entity underlying this hyperpolarization remains unknown and it is not clear whether changes in membrane potential are linked to the increase in intracellular Ca2+. In this study, differentiated U937 cells, as a macrophage-like cell model, was used to characterize the fMLF response using electrophysiological and Ca2+ imaging techniques. We demonstrate by means of pharmacological and molecular biology tools that fMLF induces a Ca2+-dependent hyperpolarization via activation of the K+ channel KCa3.1 and thus, enhancing fMLF-induced intracellular Ca2+ increase through an amplification of the driving force for Ca2+ entry. Consequently, enhanced Ca2+ influx would in turn lengthen the hyperpolarization, operating as a positive feedback mechanism for fMLF-induced Ca2+ signaling.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Diferenciação Celular/efeitos dos fármacos , CMP Cíclico/análogos & derivados , CMP Cíclico/farmacologia , Retroalimentação Fisiológica/efeitos dos fármacos , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/genética , Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Monócitos/fisiologia , Técnicas de Patch-Clamp , Interferência de RNA , Células U937
8.
Pflugers Arch ; 467(8): 1723-1732, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25231975

RESUMO

Transient receptor potential melastatin-like 4 (TRPM4) is a Ca(2+)-activated non-selective cation channel expressed in a wide range of human tissues. TRPM4 participates in a variety of physiological processes such as T cell activation, myogenic vasoconstriction, and allergic reactions. TRPM4 Ca(2+) sensitivity is enhanced by calmodulin (CaM) and phosphathydilinositol 4, 5-bisphosphate (PI(4,5)P2) binding, as well as, under certain conditions, PKC activation. However, information as to the mechanisms of modulation of this channel remains unknown, including direct identification of phosphorylation sites on TRPM4 and their role in channel features. Here, we use mass-spectrometric-based proteomic approaches (immunoprecipitation and tandem mass spectrometry) to unambiguously identify S839 as a phosphorylation site present on human TRPM4 expressed in a human cell line. Site-directed mutagenesis employing a serine to alanine mutation to eliminate phosphorylation, and a phospho-mimetic aspartate mutation, as well as biochemical and immunocytochemical experiments, revealed a role for S839 phosphorylation in the basolateral expression of TRPM4 channels in epithelial cells. Moreover, we demonstrated that casein kinase 1 (CK1) phosphorylates S839 and is responsible for the basolateral localization of TRPM4.


Assuntos
Caseína Quinase I/metabolismo , Canais de Cátion TRPM/metabolismo , Sequência de Aminoácidos , Cromatografia Líquida de Alta Pressão , Células HEK293 , Humanos , Imunoprecipitação , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Fosforilação , Transporte Proteico , Proteômica/métodos , Serina , Canais de Cátion TRPM/química , Canais de Cátion TRPM/genética , Espectrometria de Massas em Tandem , Transfecção
9.
Biochem Biophys Res Commun ; 411(1): 82-7, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21708127

RESUMO

Brain cell metabolism is intimately associated with intracellular oxidation-reduction (redox) balance. Glutamatergic transmission is accompanied with changes in substrate preference in neurons. Therefore, we studied cytoplasmatic redox changes in hippocampal neurons in culture exposed to glutamate. Neurons were transfected with HyPer, a genetically encoded redox biosensor for hydrogen peroxide which allows real-time imaging of the redox state. The rate of fluorescence decay, corresponding to the reduction of the biosensor was found to be augmented by low doses of glutamate (10 µM) as well as by pharmacological stimulation of NMDA glutamate receptors. Acute chelation of extracellular Ca(2+) abolished the glutamate-induced effect observed on HyPer fluorescence. Additional experiments indicated that mitochondrial function and hence energetic substrate availability commands the redox state of neurons and is required for the glutamate effect observed on the biosensor signal. Furthermore, our results implicated astrocytic metabolism in the changes of neuronal redox state observed with glutamate.


Assuntos
Citoplasma/metabolismo , Ácido Glutâmico/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Animais , Cálcio/metabolismo , Células Cultivadas , Citoplasma/efeitos dos fármacos , Ácido Glutâmico/farmacologia , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Oxirredução , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo
10.
Channels (Austin) ; 5(3): 280-6, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21525790

RESUMO

Angiotensin II regulation of L-type calcium currents in cardiac muscle is controversial and the underlying signaling events are not completely understood. Moreover, the possible role of auxiliary subunit composition of the channels in Angiotensin II modulation of L-type calcium channels has not yet been explored. In this work we study the role of Ca(v)ß subunits and the intracellular signaling responsible for L-type calcium current modulation by Angiotensin II. In cardiomyocytes, Angiotensin II exposure induces rapid inhibition of L-type current with a magnitude that is correlated with the rate of current inactivation. Semi-quantitative PCR of cardiomyocytes at different days of culture reveals changes in the Ca(v)ß subunits expression pattern that are correlated with the rate of current inactivation and with Angiotensin II effect. Over-expression of individual b subunits in heterologous systems reveals that the magnitude of Angiotensin II inhibition is dependent on the Ca(v)ß subunit isoform, with Ca(v)ß(1b) containing channels being more strongly regulated. Ca(v)ß(2a) containing channels were insensitive to modulation and this effect was partially due to the N-terminal palmitoylation sites of this subunit. Moreover, PLC or diacylglycerol lipase inhibition prevents the Angiotensin II effect on L-type calcium channels, while PKC inhibition with chelerythrine does not, suggesting a role of arachidonic acid in this process. Finally, we show that in intact cardiomyocytes the magnitude of calcium transients on spontaneous beating cells is modulated by Angiotensin II in a Ca(v)ß subunit-dependent manner. These data demonstrate that Ca(v)ß subunits alter the magnitude of inhibition of L-type current by Angiotensin II.


Assuntos
Angiotensina II/metabolismo , Canais de Cálcio Tipo L/metabolismo , Regulação da Expressão Gênica/fisiologia , Potenciais da Membrana/fisiologia , Proteínas Musculares/metabolismo , Miócitos Cardíacos/metabolismo , Angiotensina II/farmacologia , Animais , Antibacterianos/farmacologia , Ácido Araquidônico/metabolismo , Benzofenantridinas/farmacologia , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Lipase Lipoproteica/farmacologia , Lipoilação/efeitos dos fármacos , Lipoilação/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/genética , Miócitos Cardíacos/citologia , Fosfoinositídeo Fosfolipase C/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Ratos , Ratos Sprague-Dawley , Vasoconstritores/metabolismo , Vasoconstritores/farmacologia
11.
J Cell Physiol ; 226(1): 103-9, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20625999

RESUMO

Altered expression of some members of the TRP ion channel superfamily has been associated with the development of pathologies like cancer. In particular, TRPM4 levels are reportedly elevated in diffuse large B-cell non-Hodgkin lymphoma, prostate, and cervical cancer. However, whether such changes in TRPM4 expression may be relevant to genesis or progression of cancer remains unknown. Here we show that reducing TRPM4 expression decreases proliferation of HeLa cells, a cervical cancer-derived cell line. In this cell line, constitutive TRPM4 silencing promoted GSK-3ß-dependent degradation of ß-catenin and reduced ß-catenin/Tcf/Lef-dependent transcription. Conversely, overexpression of TRPM4 in T-REx 293 cells (a HEK293-derived cell line) increased cell proliferation and ß-catenin levels. Our results identify TRPM4 as an important, unanticipated regulator of the ß-catenin pathway, where aberrant signaling is frequently associated with cancer.


Assuntos
Transdução de Sinais/fisiologia , Canais de Cátion TRPM/metabolismo , Regulação para Cima/fisiologia , beta Catenina/metabolismo , Linhagem Celular , Proliferação de Células , Humanos , Canais de Cátion TRPM/genética , Transcrição Gênica , beta Catenina/genética
12.
J Biol Chem ; 285(48): 37150-8, 2010 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-20884614

RESUMO

Necrosis is associated with an increase in plasma membrane permeability, cell swelling, and loss of membrane integrity with subsequent release of cytoplasmic constituents. Severe redox imbalance by overproduction of reactive oxygen species is one of the main causes of necrosis. Here we demonstrate that H(2)O(2) induces a sustained activity of TRPM4, a Ca(2+)-activated, Ca(2+)-impermeant nonselective cation channel resulting in an increased vulnerability to cell death. In HEK 293 cells overexpressing TRPM4, H(2)O(2) was found to eliminate in a dose-dependent manner TRPM4 desensitization. Site-directed mutagenesis experiments revealed that the Cys(1093) residue is crucial for the H(2)O(2)-mediated loss of desensitization. In HeLa cells, which endogenously express TRPM4, H(2)O(2) elicited necrosis as well as apoptosis. H(2)O(2)-mediated necrosis but not apoptosis was abolished by replacement of external Na(+) ions with sucrose or the non-permeant cation N-methyl-d-glucamine and by knocking down TRPM4 with a shRNA directed against TRPM4. Conversely, transient overexpression of TRPM4 in HeLa cells in which TRPM4 was previously silenced re-established vulnerability to H(2)O(2)-induced necrotic cell death. In addition, HeLa cells exposed to H(2)O(2) displayed an irreversible loss of membrane potential, which was prevented by TRPM4 knockdown.


Assuntos
Apoptose , Peróxido de Hidrogênio/metabolismo , Necrose/metabolismo , Canais de Cátion TRPM/metabolismo , Motivos de Aminoácidos , Membrana Celular/química , Membrana Celular/metabolismo , Células HEK293 , Células HeLa , Humanos , Potenciais da Membrana , Necrose/genética , Canais de Cátion TRPM/química , Canais de Cátion TRPM/genética
13.
J Biol Chem ; 285(10): 7566-74, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20056605

RESUMO

Volume-sensitive outwardly rectifying (VSOR) Cl(-) channels are critical for the regulatory volume decrease (RVD) response triggered upon cell swelling. Recent evidence indicates that H(2)O(2) plays an essential role in the activation of these channels and that H(2)O(2) per se activates the channels under isotonic isovolumic conditions. However, a significant difference in the time course for current onset between H(2)O(2)-induced and hypotonicity-mediated VSOR Cl(-) activation is observed. In several cell types, cell swelling induced by hypotonic challenges triggers the release of ATP to the extracellular medium, which in turn, activates purinergic receptors and modulates cell volume regulation. In this study, we have addressed the effect of purinergic receptor activation on H(2)O(2)-induced and hypotonicity-mediated VSOR Cl(-) current activation. Here we show that rat hepatoma cells (HTC) exposed to a 33% hypotonic solution responded by rapidly activating VSOR Cl(-) current and releasing ATP to the extracellular medium. In contrast, cells exposed to 200 microm H(2)O(2) VSOR Cl(-) current onset was significantly slower, and ATP release was not detected. In cells exposed to either 11% hypotonicity or 200 microm H(2)O(2), exogenous addition of ATP in the presence of extracellular Ca(2+) resulted in a decrease in the half-time for VSOR Cl(-) current onset. Conversely, in cells that overexpress a dominant-negative mutant of the ionotropic receptor P2X4 challenged with a 33% hypotonic solution, the half-time for VSOR Cl(-) current onset was significantly slowed down. Our results indicate that, at high hypotonic imbalances, swelling-induced ATP release activates the purinergic receptor P2X4, which in turn modulates the time course of VSOR Cl(-) current onset in a extracellular Ca(2+)-dependent manner.


Assuntos
Tamanho Celular , Canais de Cloreto/metabolismo , Receptores Purinérgicos P2/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cálcio/metabolismo , Carcinoma Hepatocelular , Linhagem Celular , Linhagem Celular Tumoral , Canais de Cloreto/genética , Soluções Hipotônicas , Neoplasias Hepáticas , Técnicas de Patch-Clamp , Ratos , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2X4
14.
PLoS One ; 5(12): e15658, 2010 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-21209925

RESUMO

Prion-related disorders (PrDs) are fatal neurodegenerative disorders characterized by progressive neuronal impairment as well as the accumulation of an abnormally folded and protease resistant form of the cellular prion protein, termed PrP(RES). Altered endoplasmic reticulum (ER) homeostasis is associated with the occurrence of neurodegeneration in sporadic, infectious and familial forms of PrDs. The ER operates as a major intracellular calcium store, playing a crucial role in pathological events related to neuronal dysfunction and death. Here we investigated the possible impact of PrP misfolding on ER calcium homeostasis in infectious and familial models of PrDs. Neuro2A cells chronically infected with scrapie prions showed decreased ER-calcium content that correlated with a stronger upregulation of UPR-inducible chaperones, and a higher sensitivity to ER stress-induced cell death. Overexpression of the calcium pump SERCA stimulated calcium release and increased the neurotoxicity observed after exposure of cells to brain-derived infectious PrP(RES). Furthermore, expression of PrP mutants that cause hereditary Creutzfeldt-Jakob disease or fatal familial insomnia led to accumulation of PrP(RES) and their partial retention at the ER, associated with a drastic decrease of ER calcium content and higher susceptibility to ER stress. Finally, similar results were observed when a transmembrane form of PrP was expressed, which is proposed as a neurotoxic intermediate. Our results suggest that alterations in calcium homeostasis and increased susceptibility to ER stress are common pathological features of both infectious and familial PrD models.


Assuntos
Cálcio/química , Retículo Endoplasmático/metabolismo , Príons/química , Animais , Membrana Celular/metabolismo , Síndrome de Creutzfeldt-Jakob/metabolismo , Homeostase , Humanos , Camundongos , Chaperonas Moleculares/metabolismo , Desnaturação Proteica , Dobramento de Proteína , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Scrapie/metabolismo , Regulação para Cima
15.
Endocrinology ; 149(9): 4680-7, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18499752

RESUMO

Chloride permeability pathways and progesterone (P4) secretion elicited by human chorionic gonadotropin (hCG) in human granulosa cells were studied by electrophysiological techniques and single-cell volume, membrane potential and Ca2+i measurements. Reduction in extracellular Cl(-) and equimolar substitution by the membrane-impermeant anions glutamate or gluconate significantly increased hCG-stimulated P4 accumulation. A similar result was achieved by exposing the cells to hCG in the presence of a hypotonic extracellular solution. Conversely, P4 accumulation was drastically reduced in cells challenged with hCG exposed to a hypertonic solution. Furthermore, conventional Cl(-) channel inhibitors abolished hCG-mediated P4 secretion. In contrast, 25-hydroxycholesterol-mediated P4 accumulation was unaffected by Cl(-) channel blockers. In human granulosa cells, hCG triggered the activation of a tamoxifen-sensitive outwardly rectifying Cl(-) current comparable to the volume-sensitive outwardly rectifying Cl(-) current. Exposure of human granulosa cells to hCG induced a rapid 4,4'-diisothiocyanatostilbene-2,2-disulphonic acid-sensitive cell membrane depolarization that was paralleled with an approximately 20% decrease in cell volume. Treatment with hCG evoked oscillatory and nonoscillatory intracellular Ca2+ signals in human granulosa cells. Extracellular Ca2+ removal and 4,4'-diisothiocyanatostilbene-2,2-disulphonic acid abolished the nonoscillatory component while leaving the Ca2+ oscillations unaffected. It is concluded that human granulosa cells express functional the volume-sensitive outwardly rectifying Cl(-) channels that are activated by hCG, which are critical for plasma membrane potential changes, Ca2+ influx, and P4 production.


Assuntos
Canais de Cloreto/metabolismo , Gonadotropina Coriônica/farmacologia , Células da Granulosa/efeitos dos fármacos , Luteinização/efeitos dos fármacos , Ovário/efeitos dos fármacos , Progesterona/biossíntese , Cálcio/metabolismo , Células Cultivadas , Canais de Cloreto/fisiologia , Cloretos/metabolismo , Eletrofisiologia , Feminino , Células da Granulosa/metabolismo , Células da Granulosa/fisiologia , Humanos , Potenciais da Membrana/efeitos dos fármacos , Ovário/metabolismo
16.
Curr Mol Med ; 8(3): 187-206, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18473819

RESUMO

Apoptosis is a programmed form of cell death with well-defined morphological traits that are often associated with activation of caspases. More recently evidence has become available demonstrating that upon caspase inhibition alternative programs of cell death are executed, including ones with features characteristic of necrosis. These findings have changed our view of necrosis as a passive and essentially accidental form of cell death to that of an active, regulated and controllable process. Also necrosis has now been observed in parallel with, rather than as an alternative pathway to, apoptosis. Thus, cell death responses are extremely flexible despite being programmed. In this review, some of the hallmarks of different programmed cell death modes have been highlighted before focusing the discussion on necrosis. Obligatory events associated with this form of cell death include uncompensated cell swelling and related changes at the plasma membrane. In this context, representatives of the transient receptor channel family and their regulation are discussed. Also mechanisms that lead to execution of the necrotic cell death program are highlighted. Emphasis is laid on summarizing our understanding of events that permit switching between cell death modes and how they connect to necrosis. Finally, potential implications for the treatment of some disease states are mentioned.


Assuntos
Necrose/metabolismo , Animais , Apoptose , Autofagia , Caspases/metabolismo , Ceramidas/metabolismo , Humanos , Membranas Intracelulares/metabolismo , Modelos Biológicos , Canais de Cátion TRPC/metabolismo
17.
Free Radic Biol Med ; 44(6): 1146-60, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18191646

RESUMO

In lymphocytes, Fas activation leads to both apoptosis and necrosis, whereby the latter form of cell death is linked to delayed production of endogenous ceramide and is mimicked by exogenous administration of long- and short-chain ceramides. Here molecular events associated with noncanonical necrotic cell death downstream of ceramide were investigated in A20 B lymphoma and Jurkat T cells. Cell-permeable, C6-ceramide (C6), but not dihydro-C6-ceramide (DH-C6), induced necrosis in a time- and dose-dependent fashion. Rapid formation of reactive oxygen species (ROS) within 30 min of C6 addition detected by a dihydrorhodamine fluorescence assay, as well as by electron spin resonance, was accompanied by loss of mitochondrial membrane potential. The presence of N-acetylcysteine or ROS scavengers like Tiron, but not Trolox, attenuated ceramide-induced necrosis. Alternatively, adenovirus-mediated expression of catalase in A20 cells also attenuated cell necrosis but not apoptosis. Necrotic cell death observed following C6 exposure was associated with a pronounced decrease in ATP levels and Tiron significantly delayed ATP depletion in both A20 and Jurkat cells. Thus, apoptotic and necrotic death induced by ceramide in lymphocytes occurs via distinct mechanisms. Furthermore, ceramide-induced necrotic cell death is linked here to loss of mitochondrial membrane potential, production of ROS, and intracellular ATP depletion.


Assuntos
Trifosfato de Adenosina/metabolismo , Ceramidas/metabolismo , Linfócitos/metabolismo , Necrose/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Apoptose , Espectroscopia de Ressonância de Spin Eletrônica , Citometria de Fluxo , Sequestradores de Radicais Livres/farmacologia , Glutationa/efeitos dos fármacos , Glutationa/metabolismo , Humanos , Células Jurkat , Linfócitos/efeitos dos fármacos , Linfócitos/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia
18.
Cell Physiol Biochem ; 20(6): 773-80, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17982259

RESUMO

Volume-sensitive outwardly rectifying (VSOR) Cl(-) channels participate in several physiological processes such as regulatory volume decrease, cell cycle regulation, proliferation and apoptosis. Recent evidence points to a significant role of hydrogen peroxide (H(2)O(2)) in VSOR Cl(-) channel activation. The aim of this study was to determine the signalling pathways responsible for H(2)O(2)-induced VSOR Cl(-) channel activation. In rat hepatoma (HTC) cells, H(2)O(2) elicited a transient increase in tyrosine phosphorylation of phospholipase Cgamma1 (PLCgamma1) that was blocked by PP2, a Src-family protein kinases inhibitor. Also, H(2)O(2) triggered an increase in cytosolic [Ca(2+)] that paralleled the time course of PLCgamma1 phosphorylation. The H(2)O(2)-induced [Ca(2+)](i) rise was prevented by the generic phospholipase C (PLC) inhibitor U73122 and the inositol 1,4,5-trisphosphate-receptor (IP(3)R) blocker 2-APB. In line with these results, manoeuvres that prevented PLCgamma1 activation and/or [Ca(2+)](i) rise, abolished H(2)O(2)-induced VSOR Cl(-) currents. Furthermore, in cells that overexpress a phosphorylation-defective dominant mutant of PLCgamma1, H(2)O(2) did not induce activation of VSOR Cl(-) currents. All these H(2)O(2)-induced effects were independent of extracellular Ca(2+). Our findings suggest that activation of PLCgamma1 and subsequent Ca(2+)(i) mobilisation mediate H(2)O(2)-induced VSOR Cl(-) currents, indicating that H(2)O(2) operates via redox-sensitive signalling pathways akin to those activated by osmotic challenges.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Canais de Cloreto/metabolismo , Peróxido de Hidrogênio/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Neoplasias Hepáticas Experimentais/enzimologia , Fosfolipase C gama/metabolismo , Animais , Compostos de Boro/farmacologia , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Estrenos/farmacologia , Genes Dominantes , Proteínas Mutantes/metabolismo , Fosforilação/efeitos dos fármacos , Pirimidinas/farmacologia , Pirrolidinonas/farmacologia , Ratos , Quinases da Família src/antagonistas & inibidores
19.
Am J Physiol Cell Physiol ; 287(4): C963-70, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15163619

RESUMO

In a previous work the involvement of a fenamate-sensitive Ca(2+)-activated nonselective cation channel (NSCC) in free radical-induced rat liver cell necrosis was demonstrated (5). Therefore, we studied the effect of radical oxygen species and oxidizing agents on the gating behavior of a NSCC in a liver-derived epithelial cell line (HTC). Single-channel currents were recorded in HTC cells by the excised inside-out configuration of the patch-clamp technique. In this cell line, we characterize a 19-pS Ca(2+)-activated, ATP- and fenamate-sensitive NSCC nearly equally permeable to monovalent cations. In the presence of Fe(2+), exposure of the intracellular side of NSCC to H(2)O(2) increased their open probability (P(o)) by approximately 40% without affecting the unitary conductance. Desferrioxamine as well as the hydroxyl radical (.OH) scavenger MCI-186 inhibited the effect of H(2)O(2), indicating that the increase in P(o) was mediated by.OH. Exposure of the patch membrane to the oxidizing agent 5,5'-dithio-bis-2-nitrobenzoic acid (DTNB) had a similar effect to.OH. The increase in P(o) induced by.OH or DTNB was not reverted by preventing formation or by DTNB washout, respectively. However, the reducing agent dithiothreitol completely reversed the effects on P(o) of both.OH and DTNB. A similar increase in P(o) was observed by applying the physiological oxidizing molecule GSSG. Moreover, GSSG-oxidized channels showed enhanced sensitivity to Ca(2+). The effect of GSSG was fully reversed by GSH. These results suggest an intracellular site(s) of action of oxidizing agents on cysteine targets on the fenamate-sensitive NSCC protein implicated in epithelial cell necrosis.


Assuntos
Cálcio/metabolismo , Células Epiteliais/metabolismo , Radical Hidroxila/metabolismo , Ativação do Canal Iônico/fisiologia , Canais Iônicos/metabolismo , Animais , Cátions/metabolismo , Células Cultivadas , Cisteína/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Dissulfeto de Glutationa/farmacologia , Peróxido de Hidrogênio/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Canais Iônicos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Necrose , Oxidantes/farmacologia , Oxirredução , Técnicas de Patch-Clamp , Ratos , ortoaminobenzoatos/farmacologia
20.
Neurochem Res ; 29(1): 169-76, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14992276

RESUMO

The role of Ca2+ in the signaling transduction pathway involved in osmosensitive taurine efflux in HeLa cells was studied using radiotracer efflux techniques. Taurine efflux induced by extracellular hypotonicity was decreased by 85% by removal of extracellular Ca2+ and simultaneous depletion of intracellular Ca2+ stores with thapsigargin. Extracellular Ca2+ removal, thapsigargin treatment, or addition of Gd3+ all decreased taurine efflux by approximately 50%. To explore the putative signal transduction pathways involved in swelling-induced taurine efflux, HeLa cells were exposed to PP1, an inhibitor of the Src family of tyrosine kinases, the phospholipase C inhibitor U73122, the IP3 receptor antagonist 2-APB, and the generic protein kinase C inhibitor chelerythrine. All of these treatments caused approximately 50% inhibition of taurine release in Ca2+-rich extracellular medium and approximately 85%-90% in Ca2+-free conditions. The inhibitors of the conventional protein kinase C isoforms BIM-1 and Gö6976 reduced taurine efflux to a lesser extent. Acute (10-min) exposure to the phorbol ester tetradecanoyl phorbol acetate (TPA) increased taurine efflux in 25%, whilst overnight exposure had an inhibitory effect decreasing efflux by 22%. A working model for activation of osmosensitive taurine efflux in HeLa cells involving different Ca2+ signaling pathways is presented.


Assuntos
Cálcio/fisiologia , Transporte Biológico , Inibidores Enzimáticos/farmacologia , Estrenos/farmacologia , Células HeLa , Humanos , Osmose , Proteína Quinase C/metabolismo , Pirrolidinonas/farmacologia , Transdução de Sinais , Taurina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...