• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 9
  • 6
  • 4
  • 1
  • Tagged with
  • 25
  • 25
  • 17
  • 14
  • 12
  • 10
  • 8
  • 8
  • 8
  • 8
  • 6
  • 6
  • 6
  • 6
  • 5
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
11

KATP Channel Action in Vascular Tone Regulation During Septic Shock: Beyond Physiology

Shi, Weiwei 23 March 2009 (has links)
Septic shock is a major cause of deaths resulting from uncontrolled inflammation and circulatory failure. Recent studies suggest that the vascular isoform of ATP-sensitive K+ (KATP) channels is an important contributor to septic susceptibility. To understand the molecular mechanisms for channel regulation during sepsis, we performed studies in isolated endothelium-denuded mesenteric rings. Lipopolysaccharides (LPS) induced vascular relaxation and hyporeactivity to phenylephrine. The LPS-treated aortic smooth muscle cells displayed hyperpolarization and augmentation of KATP channel activity. Both were due to an up-regulation of Kir6.1 and SUR2B surface expression. The up-regulation relied on transcriptional and translational mechanisms, in which nuclear factor-¦ÊB (NF-¦ÊB) and Protein kinase A (PKA) played a critical role. Oxidative stress occurs during sepsis and may act as another regulatory mechanism affecting KATP channel activity and vascular contractility. We found that micromolar concentrations of H2O2 impaired the pinacidil-induced vasodilation. The effect attributed to the suppression of KATP channel activity, which can be fully produced by reactivity oxidants. Unlike the Kir6.1/SUR2B channel, the Kir6.2/SUR2B channel was insensitive to 1mM H2O2, indicating that the modulation sites are located in Kir6.1. Site-directed mutational analysis showed that three cysteine residues located in N-terminus and the core region of Kir6.1 were likely to mediate the redox-dependent channel modulation. Arginine vasopressin (AVP) is a vasoconstrictor that is successfully applied to manage sepsis. However, the downstream target of AVP is uncertain. Our studies show that AVP-induced vasoconstriction depended on V1a receptor, Protein kinase C (PKC) and KATP channel. Additionally, AVP decreased Kir6.1/SUR2B channel activity through V1a receptor. The inhibitory effect was caused by a suppression of the channel open state probability. The channel inhibition was mediated by phosphorylation of the channel protein by PKC. The widespread involvement of the vascular KATP channel in vascular responses to endotoxemia strongly suggests that the temporospatial control of channel activity may constitute an important intervention to vascular tone, blood pressure and organ-tissue perfusion in septic shock. Such a control appears feasible by targeting several modulatory mechanisms of intracellular signaling, Kir6.1/SUR2B expression, redox state and channel protein phosphorylation as demonstrated in this dissertation.
12

Etudes moléculaires du canal potassique sensible a l'ATP : "gating", pathologie et optogénétique / Molecular studies of ATP-sensitive potassium channels : gating, pathology, and optogenetics

Reyes Mejia, Gina Catalina 23 September 2016 (has links)
Les canaux potassiques sensibles à l’ATP (KATP) sont des canaux omniprésents liant excitabilité et énergie cellulaire. Ils fonctionnent en captant le niveau relatif des nucléotides ATP et ADP à l’intérieur des cellules: Les premiers bloquant le canal et les derniers l’activant. De plus le phospholipide phosphatidylinositol4,5-bisphosphate (PIP2) est connu pour être un puissant régulateur des canaux KATP. Ceux-ci sont présents dans la plupart des tissus excitables et sont impliqués dans un grand nombre de fonctions physiologiques. L’objectif de ma thèse consiste à désigner un bloc dépendant de la lumière au niveau de ces KATP, afin de contrôler son activité optiquement tout en gardant ses propriétés natives. Cela a été accompli par la mutation de différents résidus en cystéine. Ce canal KATP complètement dépendant de la lumière, pourrait être utilisé pour réguler les actions de potentiels via la lumière afin de piloter différents aspects d’électrophysiologie cellulaire mais aussi de développer des applications de photo-traitements.J’ai également réalisé la cartographie fonctionnelle des résidus impliqués dans le gating du canal Kir6.2 sous le contrôle de protéines membranaires interagissant avec le domaine N-terminal. Cela a été réalisé par le design d’un canal artificiel Kir6.2 formé par la fusion du C-terminal d’un RCPG avec le N-terminal du canal. Des structures cristallographiques et des caractérisations fonctionnelles des canaux potassiques ont permis de mettre en évidence la présence de deux portes dans les domaines transmembranaires : le filtre de sélectivité et le « gate A » à l’interface cytoplasmique, et le troisième « gate » dans le domaine cytoplasmique du canal Kir connu sous le nom de « G loop gate ». Enfin j’ai caractérisé de mutations dans le gène ABCC9 codant pour SUR2A et associé au syndrome de Cantu (CS). Ces mutations sont localisées dans le domaine transmembranaire 0 (TMD0) de SUR2A, un domaine essentiel dans l’interaction entre Kir6.2 et SUR dans le complexe KATP. Les résultats suggèrent que les deux mutations cause une hyperactivité du canal via 2 mécanismes distincts : (1) Une diminution de la sensibilité de l’ATP affectant la modulation du PIP2, mais qui n’affecte pas l’activation par le Mg-ADP ou (2) aucun effets en réponse à l’ATP ou Mg-ADP, mais une sensibilité accrue au PIP2. Ces découvertes soulignent le rôle essentiel du TMD0 dans la modulation du « gating » de Kir6.2. En particulier, cela démontre qu’il y a un contrôle de la réponse du canal par des effecteurs intracellulaires qui se fixent sur Kir6.2, impliquant des interactions très liées entre Kir6.2 et la région TMD0. / ATP-sensitive K+ (KATP) channels are ubiquitous channels designed to couple excitability to cellular energy. They perform this function by sensing the relative levels of the intracellular nucleotides ATP and ADP; with ATP blocking the channel and ADP activating it. Additionally, the phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2) is known to be a strong regulator of KATP channels. These channels are present in many excitable tissues and involved in many physiological functions. The aim of this thesis is to design a light dependent block of the KATP channel, in order to control its activity and have it under optical control while at the same time retaining its native properties. This was accomplished by mutating specific residues to cysteines. This light dependent blocked KATP channel, could be used to regulate action potentials with light to tune diverse aspects of cellular electrophysiology and potentially photo-pharmacology treatment. We also performed a functional mapping of the Kir6.2 channel gate(s) under the control of membrane proteins interacting with the N-terminal domain. This was performed by using a unique artificial gate Kir6.2 channel formed by fusing a GPCR C-terminus to the Kir6.2 N terminus. Crystallographic structures and functional characterizations of potassium channels demonstrated the presence of two gates in the transmembrane domains: the selectivity filter and the "A" gate at the cytoplasmic interface, and a third gate in the cytoplasmic domain of Kir channels known as the G loop gate. Unexpectedly, our results demonstrated that several gates could be involved suggesting a concerted mechanism. Finally, we characterized two single-point mutations in the ABCC9 gene encoding SUR2, that are associated with Cantu syndrome (CS). These mutations are localized in transmembrane domain 0 (TMD0) of SUR2A, an essential domain which mediates the interaction between Kir6.2 and SUR within the K-ATP channel complex. Results suggest that the two mutations cause KATP channel hyperactivity through two divergent mechanisms: (1) a decreased sensitivity to ATP inhibition and affecting the modulation by PIP2, and that does not affect activation by Mg-ADP or (2) any effect on the response to ATP and Mg-ADP, but more sensitive to activation by PIP2. These discoveries underline the essential role of TMD0 in the gating modulation of Kir6.2. They demonstrate in particular that it can control the response of the channel to intracellular effectors that bind to Kir6.2, implying tight interactions between Kir6.2 and the TMD0 region.
13

Efeitos redox e protetores do pré-condicionamento isquêmico e da abertura do canal mitocondrial de potássio sensível a ATP contra morte celular por isquemia e reperfusão cardíaca / Redox and Protective Effects of Ischemic Preconditioning and Mitochondrial ATP-Sensitive K+ Channels Against Cardiac Cell Death Promoted by Ischemia and Reperfusion

Facundo, Héberty di Tarso Fernandes 22 March 2007 (has links)
Eventos isquêmicos seguidos por reperfusão levam ao dano celular e mitocondrial devido à abertura do poro de transição de permeabilidade mitocondrial (TPM). Todavia, o pré-condicionamento evita o dano celular por isquemia e reperfusão. Esse efeito protetor é semelhante ao obtido pela abertura do canal mitocondrial de potássio sensível a ATP (mitoKATP). Aqui, nós mostramos os mecanismos de sinalização que ativam o mitoKATP durante o pré-condicionamento, o papel redox destes canais e seu conseqüente mecanismo protetor. Usando células cardíacas HL-1, nós demonstramos que aumentos em espécies reativas de oxigênio (EROs) observadas durante o pré-condicionamento não foram revertidos por antagonistas do mitoKATP, que significativamente evitaram a proteção pelo pré-condicionamento. Isso sugere que essas espécies são formadas anteriormente à abertura do canal. Consistente com essa hipótese, a adição de catalase a corações perfundidos de rato e a células HL-1 promove reversão dos efeitos benéficos do pré-condicionamento, mas não do diazóxido (um agonista do mitoKATP). Por outro lado, 2-mercaptopropionil glicina preveniu a cardioproteção em ambos os casos, sugerindo que este composto deve apresentar outros efeitos além de antioxidante. De fato, verificamos que agentes redutores tiólicos interferem na ativação do mitoKATP mediada pelo diazóxido em mitocôndrias isoladas de coração de rato. Examinando como o mitoKATP pode ser ativado durante o pré-condicionamento, constatamos que EROs endógenas e exógenas fortemente ativaram o mitoKATP, sugerindo que o moderado aumento nas EROs durante o pré-condicionamento pode ativar esse canal. Uma vez ativado, o canal preveniu as condições (captação de Ca2+ e formação de EROs) que favorecem a ocorrência de TPM em situação de isquemia. A atividade deste canal também leva à diminuição de EROs gerados fisiologicamente ou durante períodos de isquemia e reperfusão, evitando o dano celular conseqüente. Este fato não envolveu nenhum aumento nos sistemas de remoção de oxidantes. Por outro lado, a inibição da TPM, usando ciclosporina A, preveniu o estresse oxidativo somente durante a reperfusão, mas protegeu as células de maneira indistinguível da abertura do mitoKATP. Juntos, nossos resultados sugerem que o mitoKATP age como um sensor para as EROs que diminui a sua geração em resposta a níveis aumentados de oxidantes. Em conseqüência, estes canais regulam o balanço redox em condições fisiológicas e previnem o estresse oxidativo em condições patológicas, inibindo com isso a ocorrência de TPM e morte celular isquêmica. / Ischemia followed by reperfusion results in impairment of cellular and mitochondrial functionality due to opening of mitochondrial permeability transition (MPT) pores. Nevertheless, preconditioning rescues cells from ischemic damage. Mitochondrial ATP-sensitive K+ channel (mitoKATP) opening also prevents cardiac ischemic cell death. Here we show the signaling mechanisms that activate mitoKATP during preconditioning, the redox role of these channels and consequent protective mechanisms. Using cardiac HL-1 cells, we found that increases in reactive oxygen species (ROS) observed during preconditioning were not inhibited by mitoKATP antagonists, although these drugs significantly avoided the protection afforded by preconditioning, suggesting their activation occurrs upstream of channel activity. Consistent with this, catalase addition to perfused rat hearts and HL-1 cells reversed the beneficial effects of preconditioning, but not of diazoxide (a mitoKATP agonist). On the other hand, 2-mercaptopropionylglycine prevented cardioprotection in both cases, suggesting this compound may present effects other than scavenging ROS. Indeed, thiol reducing agents impaired diazoxide-mediated activation of mitoKATP in isolated rat heart mitochondria. We found that endogenous or exogenous ROS strongly enhanced mitoKATP activity, suggesting that moderate increments in ROS release during preconditioning may activate mitoKATP. Furthermore, mitoKATP prevented conditions (Ca2+ uptake and ROS formation) that favor the opening of MPT pores under ischemic conditions. MitoKATP opening decreased ROS generation physiologically and during both ischemia and reperfusion, consequently avoiding cellular damage. This prevention does not involve an increase in oxidant removal systems. On the other hand, the inhibition of MPT, using cyclosporin A, prevented oxidative stress only during simulated reperfusion, but protected cells in a manner indistinguishable from mitoKATP opening. Collectively, our results suggest that mitoKATP acts as a ROS sensor that decreases mitochondrial ROS generation in response to enhanced local levels of oxidants. As a result, these channels regulate mitochondrial redox state under physiological conditions and prevent oxidative stress under pathological conditions, inhibiting MPT opening and ischemic cardiac damage.
14

Propriedades redox de canais de potássio mitocondriais ATP-sensíveis em cérebro de seu efeito neuroprotetor em excitotoxicidade / Redox Properties of Brain Mitochondrial ATP-Sensitive Potassium Channels and Neuroprotective Effects in Excitotoxicity

Maynara Fornazari 29 August 2008 (has links)
Muitos estudos demonstram que a abertura de canais de K+ mitocondriais sensíveis à ATP (mitoKATP) previnem contra danos promovidos por isquemia/reperfusão em coração. Em geral, esta proteção envolve mudanças no estado redox mitocondrial. Em cérebro, sabe-se que agonistas farmacológicos de mitoKATP também protegem em modelo de isquemia/reperfusão. Entretanto, os mecanismos envolvidos na prevenção de danos em cérebro ainda não estão claros. O objetivo principal deste trabalho é compreender os efeitos de canais de K+ mitocondriais ATP-sensíveis em tecido cerebral e os mecanismos pelos quais a sua ativação pode proteger contra danos promovidos por excitotoxicidade, uma das principais conseqüências de um evento isquêmico em cérebro. Neste contexto, demonstramos a proteção pelo mitoKATP em modelo de excitotoxicidade induzida pela ativação direta de receptores NMDA, utilizando cultura de células granulosas de cerebelo. Paralelamente a essa proteção, verificamos que a ativação de mitoKATP reduz a geração de espécies reativas de oxigênio (ROS). Em mitocôndrias isoladas, observamos que ROS geradas pela mitocôndria ativam mitoKATP cerebral, resultando em um aumento da captação de K+ para a matriz, medida através da técnica de inchamento mitocondrial. Em condições de baixa geração de ROS, a adição de H2O2 exógeno ativa o inchamento mitocondrial em resposta à entrada de K+ de modo prevenido por catalase, assim, confirmando que a atividade desses canais é redox-sensível. A ativação de mitoKATP por agonistas farmacológicos, como diazóxido, também é maior na presença de alta geração de ROS, conforme indicado por uma leve diminuição no potencial de membrana mitocondrial. Interessantemente, a adição de um redutor tiólico, 2-mercaptopropionilglicina (MPG) previne a ativação de mitoKATP. A ativação de mitoKATP não alterou a capacidade de captar Ca2+ pela mitocôndria, demonstrando que este não é o mecanismo pelo qual esses canais previnem morte celular excitotóxica. Não foram observados efeitos desses canais em modelo de excitotoxicidade in vivo e em modelo de doença neurodegenerativa, acidose metilmalônica. Juntos, nossos resultados demonstram que mitoKATP cerebrais agem como sensores de ROS mitocondrial, que quando ativados reduzem a liberação de ROS por um leve desacoplamento, prevenindo morte neuronal por excitotoxicidade NMDA-induzida / Several studies have shown that mitochondrial ATP-sensitive K+ channel (mitoKATP) opening prevents ischemia/reperfusion injuries in heart, in a manner involving changes in redox state. In brain, mitoKATP agonists also protect against ischemia/reperfusion. However, the exactly mechanism that mitoKATP protects the brain is still unclear. The purpose of this work is to understand the effects of mitochondrial ATP-sensitive K+ channels in brain and how this channel can protect against excitotoxic cell death, the main consequence of a cerebral ischemia. In this context, we demonstrate that mitoKATP protects against excitotoxicity promoted by NMDA receptor activation in cultured cerebellar granule cells. In paralell, we verified that mitoKATP activation also decreases reactive oxygen species (ROS). In isolated mitochondria, we observed that mitochondrially-generated ROS can activate brain mitoKATP, resulting in enhanced K+ uptake into the matrix, measured as swelling of the organelle. Under conditions in which mitochondrial ROS release is low, exogenous H2O2 activated swelling secondary to K+ entrance, in a manner prevented by catalase, confirming that the activity of this channel is redox-sensitive. Activation of mitoKATP channels by the pharmacological agonist diazoxide was also improved when endogenous mitochondrial ROS release was enhanced, as indicated by mild decreases in mitochondrial membrane potentials. Interessantly, mitoKATP activation was preveted by the thiol reductant 2-mercaptopropionylglycine (MPG). Mitochondrial Ca2+ uptake was not modified by opening mitoKATP, suggesting that this is not the mechanism through which this channel prevents excitotoxic cell death. In an in vivo excitotoxicity model and also neurodegenerative disease model, methylmalonic acidemia, the effects of mitoKATP agonists were not observed. Together, our results demonstrate that brain mitoKATP acts as a mitochondrial ROS sensor, which, when activated, prevents ROS release by mildly uncoupling respiration from oxidative phosphorylation, decreasing excitotoxic cell death
15

Propriedades redox de canais de potássio mitocondriais ATP-sensíveis em cérebro de seu efeito neuroprotetor em excitotoxicidade / Redox Properties of Brain Mitochondrial ATP-Sensitive Potassium Channels and Neuroprotective Effects in Excitotoxicity

Fornazari, Maynara 29 August 2008 (has links)
Muitos estudos demonstram que a abertura de canais de K+ mitocondriais sensíveis à ATP (mitoKATP) previnem contra danos promovidos por isquemia/reperfusão em coração. Em geral, esta proteção envolve mudanças no estado redox mitocondrial. Em cérebro, sabe-se que agonistas farmacológicos de mitoKATP também protegem em modelo de isquemia/reperfusão. Entretanto, os mecanismos envolvidos na prevenção de danos em cérebro ainda não estão claros. O objetivo principal deste trabalho é compreender os efeitos de canais de K+ mitocondriais ATP-sensíveis em tecido cerebral e os mecanismos pelos quais a sua ativação pode proteger contra danos promovidos por excitotoxicidade, uma das principais conseqüências de um evento isquêmico em cérebro. Neste contexto, demonstramos a proteção pelo mitoKATP em modelo de excitotoxicidade induzida pela ativação direta de receptores NMDA, utilizando cultura de células granulosas de cerebelo. Paralelamente a essa proteção, verificamos que a ativação de mitoKATP reduz a geração de espécies reativas de oxigênio (ROS). Em mitocôndrias isoladas, observamos que ROS geradas pela mitocôndria ativam mitoKATP cerebral, resultando em um aumento da captação de K+ para a matriz, medida através da técnica de inchamento mitocondrial. Em condições de baixa geração de ROS, a adição de H2O2 exógeno ativa o inchamento mitocondrial em resposta à entrada de K+ de modo prevenido por catalase, assim, confirmando que a atividade desses canais é redox-sensível. A ativação de mitoKATP por agonistas farmacológicos, como diazóxido, também é maior na presença de alta geração de ROS, conforme indicado por uma leve diminuição no potencial de membrana mitocondrial. Interessantemente, a adição de um redutor tiólico, 2-mercaptopropionilglicina (MPG) previne a ativação de mitoKATP. A ativação de mitoKATP não alterou a capacidade de captar Ca2+ pela mitocôndria, demonstrando que este não é o mecanismo pelo qual esses canais previnem morte celular excitotóxica. Não foram observados efeitos desses canais em modelo de excitotoxicidade in vivo e em modelo de doença neurodegenerativa, acidose metilmalônica. Juntos, nossos resultados demonstram que mitoKATP cerebrais agem como sensores de ROS mitocondrial, que quando ativados reduzem a liberação de ROS por um leve desacoplamento, prevenindo morte neuronal por excitotoxicidade NMDA-induzida / Several studies have shown that mitochondrial ATP-sensitive K+ channel (mitoKATP) opening prevents ischemia/reperfusion injuries in heart, in a manner involving changes in redox state. In brain, mitoKATP agonists also protect against ischemia/reperfusion. However, the exactly mechanism that mitoKATP protects the brain is still unclear. The purpose of this work is to understand the effects of mitochondrial ATP-sensitive K+ channels in brain and how this channel can protect against excitotoxic cell death, the main consequence of a cerebral ischemia. In this context, we demonstrate that mitoKATP protects against excitotoxicity promoted by NMDA receptor activation in cultured cerebellar granule cells. In paralell, we verified that mitoKATP activation also decreases reactive oxygen species (ROS). In isolated mitochondria, we observed that mitochondrially-generated ROS can activate brain mitoKATP, resulting in enhanced K+ uptake into the matrix, measured as swelling of the organelle. Under conditions in which mitochondrial ROS release is low, exogenous H2O2 activated swelling secondary to K+ entrance, in a manner prevented by catalase, confirming that the activity of this channel is redox-sensitive. Activation of mitoKATP channels by the pharmacological agonist diazoxide was also improved when endogenous mitochondrial ROS release was enhanced, as indicated by mild decreases in mitochondrial membrane potentials. Interessantly, mitoKATP activation was preveted by the thiol reductant 2-mercaptopropionylglycine (MPG). Mitochondrial Ca2+ uptake was not modified by opening mitoKATP, suggesting that this is not the mechanism through which this channel prevents excitotoxic cell death. In an in vivo excitotoxicity model and also neurodegenerative disease model, methylmalonic acidemia, the effects of mitoKATP agonists were not observed. Together, our results demonstrate that brain mitoKATP acts as a mitochondrial ROS sensor, which, when activated, prevents ROS release by mildly uncoupling respiration from oxidative phosphorylation, decreasing excitotoxic cell death
16

Vascular KATP Channel Modulation by S-Glutathionylation: A Novel Mechanism for Cellular Response to Oxidative Stress

Yang, Yang 29 April 2011 (has links)
The KATP channels play an important role in the membrane excitability and vascular tone regulation. Previous studies indicate that the function of KATP channels is disrupted in oxidative stress seen in a variety of cardiovascular diseases, while the underlying mechanism remains unclear. Here, we demonstrate S-glutathionylation to be a modulation mechanism underlying the oxidant-mediated vascular KATP channel inhibition, the molecular basis for the channel inhibition and the alleviation of the channel inhibition by vasoactive intestinal peptide (VIP). We found that an exposure of isolated mesenteric rings to H2O2 impaired the KATP channel-mediated vascular dilation. In whole-cell recordings and inside-out patches, micromolar H2O2 or diamide caused a strong inhibition of the vascular KATP channel (Kir6.1/SUR2B) in the presence, but not in the absence, of glutathione (GSH), indicating S-glutathionylation. By co-expressions of Kir6.1 or Kir6.2 with SUR2B subunits, we found that the oxidant sensitivity of the KATP channel relied on the Kir6.1 subunit. Systematic mutational analysis revealed three cysteine residues (Cys43, Cys120 and Cys176) to be important. Among them, Cys176 was prominent, contributing to >80% oxidant sensitivity. Biochemical pull-down assay with biotinylated glutathione ethyl ester (BioGEE) showed that mutations of Cys176 impaired the oxidant-induced incorporation of GSH to the Kir6.1 subunit. Simulation modeling of Kir6.1 S-glutathionylation revealed that after incorporation to residue 176, the GSH moiety occupied a space between slide helix and two transmembrane helices. This prevented the necessary conformational change of the inner helix for channel gating, and retained the channel in its closed state. VIP is a potent vasodilator, and is shown to have protective role against oxidative stress. We found that the channel was strongly augmented by VIP and the channel activation relied on PKA phosphorylation. These results therefore indicate that 1) the vascular KATP channel is strongly inhibited in oxidative stress, 2) S-glutathionylation underlies the oxidant-mediated KATP channel inhibition, 3) Cys176 in the Kir6.1 subunit is the major site for S-glutathionylation, and 4) the Kir6.1/SUR2B channel is activated in a PKA-dependent manner by VIP that has been previously shown to alleviate oxidative stress.
17

Efeitos gastroprotetores do timol em úlceras agudas e crônicas em ratos : evidências do envolvimento das prostaglandinas, canais para potássio sensíveis a ATP e secreção do muco gástrico / Gastroprotective effects of thymol on acute and chronic ulcers in rats: evidence involment of prostaglandins, ATP-sensitive K+ channels, and gastric mucus secretion

Diniz, Polyana Borges França 16 December 2016 (has links)
Coordenação de Aperfeiçoamento de Pessoal de Nível Superior - CAPES / Thymol, a monoterpene phenol derivative of cymene, is found in abundance in the essential oils produced by numerous herbs and spices such as thyme (Thymus vulgaris L.), oregano (Origanum vulgaris L.) and Lippia alba (mill.). Several biological effects have been described for thymol such as antioxidant, anti-inflammatory, local anesthetics, antinoceptive, healing and antibacterial properties. It was described the application of thymol in food Science, as herbicidad and inseticides. Moreover, essential oils containing thymol have been used in folk medicine to treat various physiological disorders such as gastrites, indigestion and stomach pains. The present study investigated the gastroprotective actions of thymol (10, 30 and 100 mg/kg, p.o.) in the acute indomethacin and ethanol and chronic acetic acid-induced ulcer models in rats. Some of the mechanisms underlying to the gastroprotective effect of thymol were investigated in the ethanol-induced ulcer model. Gastric secretion parameters (volume, pH, and total acidity) were also evaluted by the pylorus ligature model, and the mucus in the gastric content was determined. The antimicrobial activity against Helicobacter pylori of thymol was performed using the agar-well diffusion method. Thymol produced a dose dependent reduction (p < 0.01) on the total lesion area in the ethanol-induced ulcer model. This gastroprotection was also evaluated microscopically showing that the thymol at all doses decreased the loss of epithelial cells. The gastroprotective response caused by thymol (30 mg/kg) was signigicantly attenuated (P < 0.001) by intraperitoneal treatment of rats with indomethacin (a non-selective inhibitior of cyclo-oxygenase, 10 mg/kg) and glibenclamide (ATP-sensitive K channel blocker), but not by DL-Propargylglycine (PAG, a cystathionine-γ-lyase inhibitor) and Nw-nitro-L-arginine methyl ester hydrochloride (L-NAME, a non-selective inhibitor of nitric oxide synthase). Thymol (30 and 100 mg/kg) also reduced the ulcer index (p < 0.05) and the total lesion area (p < 0.001) in the indomethacin and acetic-induced ulcer models, respectively. In the model of pylorus ligature, the treatment with thymol failed to significantly change the gastric secretion parameters. However, after treatment with thymol (30 and 100 mg/kg) there was a significant increase (p < 0.01) in mucus production. Thymol showed no antimicrobial activity against H. pilory in vitro. Collectively, the present results provide evidence that thymol displays gastroprotective actions on the acute and chronic ulcer models involving mechanisms as increased amount of mucus, prostaglandins, and ATP-sensitive K+ channels. / O timol, um monoterpeno fenólico derivado do cimeno encontrado abundantemente em óleos essenciais, é produzido por numerosas plantas aromáticas e especiarias tais como tomilho (Timus vulgaris L.), orégano (Origanum vulgaris L.) e Lippia alba (mill.). Diversos efeitos biológicos têm sido descritos para o timol tais como antioxidante, anti-inflamatório, anestésico local, antinoceptivo, cicatrizante e antibactericida. É descrita a aplicação do timol em ciências dos alimentos, herbicida e inseticidas. Além disso, óleos essenciais contendo timol tem sido usados pela medicina popular para tratar diversas desordens fisiológicas como gastrite, indigestão e dores estomacais. O presente estudo investigou ações gastroprotetoras do timol (10, 30 e 100 mg/kg, v.o.) em modelos de úlcera aguda induzida por indometacina e etanol, e crônica induzida por ácido acético em ratos. Alguns dos mecanismos subjacentes ao efeito gastroprotetor do timol foram investigados em modelo de úlcera induzida por etanol. Parâmetros da secreção gástrica (volume, pH; e acidez total) também foram avaliados pelo modelo de ligadura de piloro, e o conteúdo do muco gástrico foi determinado. A atividade contra Helicobacter pylori do timol foi realizada pelo método de difusão em ágar. O timol produziu efeito dose-dependente reduzindo (p < 0,01) a área total de lesão em modelo de úlcera induzida por etanol. Esta gastroproteção também foi avaliada microscopicamente mostrando que o timol, em todas as doses, diminuiu a perda das células epiteliais. A resposta gastroprotetora causada pelo timol (30 mg/kg) foi significativamente atenuada (p < 0,001) pelo tratamento intraperitoneal dos ratos com indometacina (um inibidor não-seletivo da ciclo-oxigenase, 10 mg/kg) e glibenclamida (bloqueador de canais para potássio sensíveis a ATP), mas não pela DL-Propargilglicina (PAG, inibidor da cistationina-γ-liase) ou Nw-nitro-L-arginina metil éster cloridrato (L-NAME, um inibidor não-seletivo da sintase do óxido nítrico). O timol (30 e 100 mg/kg) também reduziu o índice de úlcera (p < 0,05) e área total de lesão (p < 0,001) por indometacina e por ácido acético, respectivamente. Em modelo de ligadura de piloro, o tratamento com timol não alterou os parâmetros de secreção gástrica. No entanto, após o tratamento com o timol houve um aumento significativo (p < 0,01) na produção do muco. O timol não apresentou atividade contra H. pylori in vitro. Coletivamente, os resultados apresentados fornecem evidências de que o timol exibe ações gastroprotetoras sobre os modelos de úlceras agudas e crônicas envolvendo mecanismos como o aumento da quantidade de muco, prostaglandinas e canais para potássio sensíveis a ATP.
18

Efeitos redox e protetores do pré-condicionamento isquêmico e da abertura do canal mitocondrial de potássio sensível a ATP contra morte celular por isquemia e reperfusão cardíaca / Redox and Protective Effects of Ischemic Preconditioning and Mitochondrial ATP-Sensitive K+ Channels Against Cardiac Cell Death Promoted by Ischemia and Reperfusion

Héberty di Tarso Fernandes Facundo 22 March 2007 (has links)
Eventos isquêmicos seguidos por reperfusão levam ao dano celular e mitocondrial devido à abertura do poro de transição de permeabilidade mitocondrial (TPM). Todavia, o pré-condicionamento evita o dano celular por isquemia e reperfusão. Esse efeito protetor é semelhante ao obtido pela abertura do canal mitocondrial de potássio sensível a ATP (mitoKATP). Aqui, nós mostramos os mecanismos de sinalização que ativam o mitoKATP durante o pré-condicionamento, o papel redox destes canais e seu conseqüente mecanismo protetor. Usando células cardíacas HL-1, nós demonstramos que aumentos em espécies reativas de oxigênio (EROs) observadas durante o pré-condicionamento não foram revertidos por antagonistas do mitoKATP, que significativamente evitaram a proteção pelo pré-condicionamento. Isso sugere que essas espécies são formadas anteriormente à abertura do canal. Consistente com essa hipótese, a adição de catalase a corações perfundidos de rato e a células HL-1 promove reversão dos efeitos benéficos do pré-condicionamento, mas não do diazóxido (um agonista do mitoKATP). Por outro lado, 2-mercaptopropionil glicina preveniu a cardioproteção em ambos os casos, sugerindo que este composto deve apresentar outros efeitos além de antioxidante. De fato, verificamos que agentes redutores tiólicos interferem na ativação do mitoKATP mediada pelo diazóxido em mitocôndrias isoladas de coração de rato. Examinando como o mitoKATP pode ser ativado durante o pré-condicionamento, constatamos que EROs endógenas e exógenas fortemente ativaram o mitoKATP, sugerindo que o moderado aumento nas EROs durante o pré-condicionamento pode ativar esse canal. Uma vez ativado, o canal preveniu as condições (captação de Ca2+ e formação de EROs) que favorecem a ocorrência de TPM em situação de isquemia. A atividade deste canal também leva à diminuição de EROs gerados fisiologicamente ou durante períodos de isquemia e reperfusão, evitando o dano celular conseqüente. Este fato não envolveu nenhum aumento nos sistemas de remoção de oxidantes. Por outro lado, a inibição da TPM, usando ciclosporina A, preveniu o estresse oxidativo somente durante a reperfusão, mas protegeu as células de maneira indistinguível da abertura do mitoKATP. Juntos, nossos resultados sugerem que o mitoKATP age como um sensor para as EROs que diminui a sua geração em resposta a níveis aumentados de oxidantes. Em conseqüência, estes canais regulam o balanço redox em condições fisiológicas e previnem o estresse oxidativo em condições patológicas, inibindo com isso a ocorrência de TPM e morte celular isquêmica. / Ischemia followed by reperfusion results in impairment of cellular and mitochondrial functionality due to opening of mitochondrial permeability transition (MPT) pores. Nevertheless, preconditioning rescues cells from ischemic damage. Mitochondrial ATP-sensitive K+ channel (mitoKATP) opening also prevents cardiac ischemic cell death. Here we show the signaling mechanisms that activate mitoKATP during preconditioning, the redox role of these channels and consequent protective mechanisms. Using cardiac HL-1 cells, we found that increases in reactive oxygen species (ROS) observed during preconditioning were not inhibited by mitoKATP antagonists, although these drugs significantly avoided the protection afforded by preconditioning, suggesting their activation occurrs upstream of channel activity. Consistent with this, catalase addition to perfused rat hearts and HL-1 cells reversed the beneficial effects of preconditioning, but not of diazoxide (a mitoKATP agonist). On the other hand, 2-mercaptopropionylglycine prevented cardioprotection in both cases, suggesting this compound may present effects other than scavenging ROS. Indeed, thiol reducing agents impaired diazoxide-mediated activation of mitoKATP in isolated rat heart mitochondria. We found that endogenous or exogenous ROS strongly enhanced mitoKATP activity, suggesting that moderate increments in ROS release during preconditioning may activate mitoKATP. Furthermore, mitoKATP prevented conditions (Ca2+ uptake and ROS formation) that favor the opening of MPT pores under ischemic conditions. MitoKATP opening decreased ROS generation physiologically and during both ischemia and reperfusion, consequently avoiding cellular damage. This prevention does not involve an increase in oxidant removal systems. On the other hand, the inhibition of MPT, using cyclosporin A, prevented oxidative stress only during simulated reperfusion, but protected cells in a manner indistinguishable from mitoKATP opening. Collectively, our results suggest that mitoKATP acts as a ROS sensor that decreases mitochondrial ROS generation in response to enhanced local levels of oxidants. As a result, these channels regulate mitochondrial redox state under physiological conditions and prevent oxidative stress under pathological conditions, inhibiting MPT opening and ischemic cardiac damage.
19

Comparison of in Vitro Preconditioning Responses of Isolated Pig and Rabbit Cardiomyocytes: Effects of a Protein Phosphatase Inhibitor, Fostriecin

Armstrong, S. C., Kao, R., Gao, W., Shivell, L. C., Downey, J. M., Honkanen, R. E., Ganote, C. E. 01 January 1997 (has links)
Calcium tolerant pig and rabbit cardiomyocytes were isolated using retrograde aortic perfusion of nominally calcium-free collagenase. Preconditioning protocols used 1 or 3 x l0-min episodes of ischemic pelleting or pre-incubation with 100 μM adenosine, followed by a 15-min post-incubation and 180-240-min ischemic pelleting. Control cells were incubated and washed in parallel with the experimental groups. Injury was assessed by determination of cell morphology, trypan blue permeability following osmotic swelling, lactate and HPLC analysis of adenine nucleotides. Preconditioned pig cardiomyocytes had a reduced rate of ischemic contracture, but protection occurred without conservation of ATP. Preconditioned rabbit cardiomyocytes were protected without significant changes in rates of ischemic contracture or ATP depletion. Incubation of ischemic cells with the protein phosphatase inhibitor, fostriecin, at PP2A-selective concentrations (0.1-10 μM), mimicked preconditioning in both rabbit and pig cardiomyocytes. In rabbits, the K(ATP) channel blocker, 5-hydroxydecanoate (5-HD), did not block preconditioning or fostriecin protection. In the pig, 5-HD blocked both preconditioning and fostriecin protection, with return of the rates of ischemic contracture to control. However, 5-HD was an effective blocker of protection only in early ischemia. Fostriecin mimicked preconditioning in the rabbit and the early responses of the preconditioned pig. Preconditioning appears associated with protein phosphorylation in both the rabbit and the pig, but major pathways leading to protection may differ in the two species.
20

The Impact of ROS Scavenging on NMDA and AMPA Receptor Whole Cell Currents in Pyramidal Neurons of the Anoxia Tolerant Western Painted Turtle

Dukoff, David 22 November 2013 (has links)
Extended periods of oxygen deprivation cause brain death in mammals but the western painted turtle overwinters in anoxic mud for months without damage. Neural protection is achieved through decreases in the whole cell currents of N-methyl-D-aspartate and alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (NMDAR and AMPAR) that are dependent on a mild increase in intracellular calcium from the mitochondria. The goal of this research was to determine if natural anoxic decreases in reactive oxidative species (ROS) serve as the signal to bring about these changes. Reductions in cellular ROS levels were demonstrated to have no effect on AMPAR currents or intracellular calcium and produced massive increases in NMDAR currents, indicating that ROS depression does not directly mediate anoxic alterations. Interestingly, mammalian neural tissue also experiences a similar increase in NMDAR whole cell current in response to reducing agents suggesting a possible conserved mechanism for normoxic receptor control.

Page generated in 0.0656 seconds