Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pharmacol Res ; 196: 106947, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37797660

RESUMO

Hydrogen sulfide (H2S) has been extensively studied as a signal molecule in the body for the past 30 years. Researchers have conducted studies using both natural and synthetic sources of H2S, known as H2S donors, which have different characteristics in terms of how they release H2S. These donors can be inorganic salts or have various organic structures. In recent years, certain types of sulfur compounds found naturally in foods have been characterized as H2S donors and explored for their potential health benefits. These compounds are referred to as "sulfanutraceuticals," a term that combines "nutrition" and "pharmaceutical". It is used to describe products derived from food sources that offer additional health advantages. By introducing the terms "sulfaceuticals" and "sulfanutraceuticals," we categorize sulfur-containing substances based on their origin and their use in both preclinical and clinical research, as well as in dietary supplements.


Assuntos
Sulfeto de Hidrogênio , Compostos de Enxofre/uso terapêutico , Enxofre
2.
Biochem Pharmacol ; 215: 115728, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37524208

RESUMO

BACKGROUND: Hydrogen sulfide (H2S) is a gasotransmitter deeply involved in cardiovascular homeostasis and implicated in the myocardial protection against ischemia/reperfusion. The post-translational persulfidation of cysteine residues has been identified as the mechanism through which H2S regulates a plethora of biological targets. Erucin (ERU) is an isothiocyanate produced upon hydrolysis of the glucosinolate glucoerucin, presents in edible plants of Brassicaceae family, such as Eruca sativa Mill., and it has emerged as a slow and long-lasting H2S-donor. AIM: In this study the cardioprotective profile of ERU has been investigated and the action mechanism explored, focusing on the possible role of the recently identified mitochondrial Kv7.4 (mitoKv7.4) potassium channels. RESULTS: Interestingly, ERU showed to release H2S and concentration-dependently protected H9c2 cells against H2O2-induced oxidative damage. Moreover, in in vivo model of myocardial infarct ERU showed protective effects, reducing the extension of ischemic area, the levels of troponin I and increasing the amount of total AnxA1, as well as co-related inflammatory outcomes. Conversely, the pre-treatment with XE991, a blocker of Kv7.4 channels, abolished them. In isolated cardiac mitochondria ERU exhibited the typical profile of a mitochondrial potassium channels opener, in particular, this isothiocyanate produced a mild depolarization of mitochondrial membrane potential, a reduction of calcium accumulation into the matrix and finally a flow of potassium ions. Finally, mitoKv7.4 channels were persulfidated in ERU-treated mitochondria. CONCLUSIONS: ERU modulates the cardiac mitoKv7.4 channels and this mechanism may be relevant for cardioprotective effects.


Assuntos
Sulfeto de Hidrogênio , Traumatismo por Reperfusão Miocárdica , Humanos , Peróxido de Hidrogênio/farmacologia , Sulfeto de Hidrogênio/farmacologia , Sulfeto de Hidrogênio/metabolismo , Isotiocianatos/farmacologia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Canais de Potássio , Mitocôndrias Cardíacas
3.
J Biol Inorg Chem ; 22(4): 519-526, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28074300

RESUMO

The X-ray structure of human apo-S100Z has been solved and compared with that of the zebrafish calcium-bound S100Z, which is the closest in sequence. Human apo-S100A12, which shows only 43% sequence identity to human S100Z, has been used as template model to solve the crystallographic phase problem. Although a significant buried surface area between the two physiological dimers is present in the asymmetric unit of human apo-S100Z, the protein does not form the superhelical arrangement in the crystal as observed for the zebrafish calcium-bound S100Z and human calcium-bound S100A4. These findings further demonstrate that calcium plays a fundamental role in triggering quaternary structure formation in several S100s. Solving the X-ray structure of human apo-S100Z by standard molecular replacement procedures turned out to be a challenge and required trying different models and different software tools among which only one was successful. The model that allowed structure solution was that with one of the lowest sequence identity with the target protein among the S100 family in the apo state. Based on the previously solved zebrafish holo-S100Z, a putative human holo-S100Z structure has been then calculated through homology modeling; the differences between the experimental human apo and calculated holo structure have been compared to those existing for other members of the family.


Assuntos
Proteínas S100/química , Animais , Cálcio/química , Cristalografia por Raios X , Humanos , Modelos Moleculares , Proteínas S100/genética , Proteínas S100/isolamento & purificação , Solubilidade , Soluções , Peixe-Zebra
4.
Nitric Oxide ; 47: 25-33, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25795591

RESUMO

Hydrogen sulfide (H2S) and nitric oxide (NO) play pivotal roles in the cardiovascular system. Conflicting results have been reported about their cross-talk. This study investigated their interplays in coronary bed of normotensive (NTRs) and spontaneously hypertensive rats (SHRs). The effects of H2S- (NaHS) and NO-donors (sodium nitroprusside, SNP) on coronary flow (CF) were measured in Langendorff-perfused hearts of NTRs and SHRs, in the absence or in the presence of propargylglycine (PAG, inhibitor of H2S biosynthesis), L-NAME (inhibitor of NO biosynthesis), ODQ (inhibitor of guanylate cyclase), L-Cysteine (substrate for H2S biosynthesis) or L-Arginine (substrate for NO biosynthesis). In NTRs, NaHS and SNP increased CF; their effects were particularly evident in Angiotensin II (AngII)-contracted coronary arteries. The dilatory effects of NaHS were abolished by L-NAME and ODQ; conversely, PAG abolished the effects of SNP. In SHRs, high levels of myocardial ROS production were observed. NaHS and SNP did not reduce the oxidative stress, but produced clear increases of the basal CF. In contrast, in AngII-contracted coronary arteries of SHRs, significant hyporeactivity to NaHS and SNP was observed. In SHRs, the vasodilatory effects of NaHS were only modestly affected by L-NAME and ODQ; PAG poorly influenced the effects of SNP. Then, in NTRs, the vascular actions of H2S required NO and vice versa. By contrast, in SHRs, the H2S-induced actions scarcely depend on NO release; as well, the NO effects are largely H2S-independent. These results represent the first step for understanding pathophysiological mechanisms of NO/H2S interplays under both normotensive and hypertensive conditions.


Assuntos
Vasos Coronários/metabolismo , Sulfeto de Hidrogênio/metabolismo , Hipertensão , Óxido Nítrico/metabolismo , Animais , Vasos Coronários/efeitos dos fármacos , Sulfeto de Hidrogênio/farmacologia , Masculino , Nitroprussiato/farmacologia , Ratos , Ratos Endogâmicos SHR , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
5.
Med Res Rev ; 35(3): 520-53, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25346462

RESUMO

Brief periods of ischemia are known to confer to the myocardium an increased resistance to the injury due to a later and more prolonged ischemic episode. This phenomenon, known as ischemic preconditioning (IPreC), is ensured by different biological mechanisms. Although an exhaustive comprehension of them has not been reached yet, it is widely accepted that mitochondria are pivotally involved in controlling cell life and death, and thus in IPreC. Among the several signaling pathways involved, as triggers and/or end effectors, in the mitochondrial mechanisms of cardioprotection, an important role is played by the activation of potassium channels located in the mitochondrial inner membrane (mitoK) of cardiomyocytes. Presently, different types of mitoK channels have been recognized in the heart, such as ATP-sensitive (mitoKATP) and calcium-activated (mitoBK(Ca) and mitoSK(Ca)) potassium channels. Consistently, drugs modulating mitoK, on one hand, have been employed as useful experimental tools for early basic studies on IPreC. On the other hand, activators of mitoK are promising and innovative therapeutic agents for limiting the myocardial injury due to ischemic episodes. In this review, we report the experimental evidence supporting the role of mitoK in signaling pathways in the mechanisms of cardioprotection and an overview on the most important molecules acting as modulators of these channels, with their profiles of selectivity. Some innovative pharmaceutical strategies for mitochondriotropic drugs have been also reported. Finally, an appendix describing the main experimental approaches usually employed to study mitoK in isolated mitochondria or in intact cells has been added.


Assuntos
Cardiotônicos/química , Mitocôndrias/metabolismo , Canais de Potássio/metabolismo , Animais , Cálcio/química , Morte Celular , Humanos , Pós-Condicionamento Isquêmico , Precondicionamento Isquêmico , Camundongos , Mitocôndrias/patologia , Mitocôndrias Cardíacas/patologia , Miocárdio/patologia , Traumatismo por Reperfusão , Transdução de Sinais
6.
Pharmacol Res ; 78: 1-9, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24083950

RESUMO

Selective cyclooxygenase 2 (COX2) inhibitors (COXIBs) are effective anti-inflammatory and analgesic drugs with improved gastrointestinal (GI) safety compared to nonselective nonsteroidal anti-inflammatory drugs known as traditional (tNSAIDs). However, their use is associated with a cardiovascular (CV) hazard (i.e. increased incidence of thrombotic events and hypertension) due to the inhibition of COX2-dependent vascular prostacyclin. Aiming to design COX2-selective inhibitors with improved CV safety, new NO-releasing COXIBs (NO-COXIBs) have been developed. In these hybrid drugs, the NO-mediated CV effects are expected to compensate for the COXIB-mediated inhibition of prostacyclin. This study evaluates the potential CV beneficial effects of VA694, a promising NO-COXIB, the anti-inflammatory effects of which have been previously characterized in several in vitro and in vivo experimental models. When incubated in hepatic homogenate, VA694 acted as a slow NO-donor. Moreover, it caused NO-mediated relaxant effects in the vascular smooth muscle. The chronic oral administration of VA694 to young spontaneously hypertensive rats (SHRs) significantly slowed down the age-related development of hypertension and was associated with increased plasma levels of nitrates, stable end-metabolites of NO. Furthermore, a significant improvement of coronary flow and a significant reduction of endothelial dysfunction were observed in SHRs submitted to chronic administration of VA694. In conclusion, VA694 is a promising COX2-inhibiting hybrid drug, showing NO releasing properties which may mitigate the CV deleterious effects associated with the COX2-inhibition.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Inibidores de Ciclo-Oxigenase 2/farmacologia , Fatores Relaxantes Dependentes do Endotélio/administração & dosagem , Endotélio/efeitos dos fármacos , Hipertensão/tratamento farmacológico , Nitratos/farmacologia , Óxido Nítrico/administração & dosagem , Pirróis/farmacologia , Animais , Anti-Inflamatórios não Esteroides/química , Pressão Sanguínea/efeitos dos fármacos , Vasos Coronários/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase 2/química , Endotélio/patologia , Fatores Relaxantes Dependentes do Endotélio/farmacologia , Hipertensão/sangue , Masculino , Nitratos/sangue , Nitratos/química , Óxido Nítrico/farmacologia , Nitritos/sangue , Pirróis/química , Ratos , Ratos Endogâmicos SHR , Ratos Wistar , Fluxo Sanguíneo Regional/efeitos dos fármacos
7.
Bioorg Med Chem ; 21(14): 4186-91, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23707646

RESUMO

Large conductance calcium activated potassium channels (BKCa) are fundamental in the control of cellular excitability. Thus, compounds that activate BKCa channels could provide potential therapies in the treatment of pathologies of the cardiovascular and central nervous system. A series of novel N-arylbenzamide compounds, and the reference compound NS1619, were evaluated for BKCa channel opener properties in Human Embryonic Kidney (HEK293) cells expressing the human BKCa channel α-subunit alone or α+ß1-subunit complex. Channel activity was determined using a non-radioactive Rb(+) efflux assay to construct concentration effect curves for each compound. All N-arylbenzamide compounds and NS1619 evoked significant (p <0.05) concentration related increases in Rb(+) efflux both in cells expressing α-subunit alone or α+ß1-subunits. Co-expression of the ß1-subunit modified the Rb(+) efflux responses, relative to that obtained in cells expressing the α-subunit alone, for most of the N-arylbenzamide compounds, in contrast to NS1619. The EC40 values of NS1619, BKMe1 and BKOEt1 were not significantly affected by the co-expression of the BKCa channel α+ß1-subunits. In contrast, 5 other N-arylbenzamides (BKPr2, BKPr3, BKPr4, BKH1 and BKVV) showed a significant (p <0.05) 2- to 10-fold increase in EC40 values when tested on the BKCa α+ß1-subunit expressing cells compared to BKCa α-subunit expressing cells. Further, the Emax values for BKPr4, BKVV and BKH1 were lower in the BKCa channel α+ß1-subunit expressing cells. In conclusion, the N-arylbenzamides studied, like NS1619, were able to activate BKCa channels formed of the α-subunit only. The co-expression of the ß1-subunit, however, modified the ability of certain compounds to active the channel leading to differentiated pharmacodynamic profiles.


Assuntos
Benzamidas/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/efeitos dos fármacos , Benzimidazóis/farmacologia , Células HEK293 , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Estrutura Molecular , Subunidades Proteicas/metabolismo
8.
Biochem Pharmacol ; 85(11): 1634-43, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23567997

RESUMO

Naringenin (NAR), flavonoid abundant in the genus Citrus, has been reported to interact with the large-conductance calcium-activated potassium channels (BK). Since activators of BK channels expressed in cardiac mitochondria trigger protective effects in several models of myocardial ischemia/reperfusion (I/R), this work aimed to evaluate the potential cardioprotective effects of NAR and the involvement of mitochondrial BK channels. In an in vivo model of acute infarct in rats, NAR (100mg/kg i.p.) significantly reduced the heart injury induced by I/R. This effect was antagonized by the selective BK-blocker paxilline (PAX). The cardioprotective dose of NAR did not cause significant effects on the blood pressure. In Largendorff-perfused rat hearts submitted to ischemia/reperfusion, NAR improved the post-ischemic functional parameters (left ventricle developed pressure and dP/dt) with lower extension of myocardial injury. On isolated rat cardiac mitochondria, NAR caused a concentration-dependent depolarization of mitochondrial membrane and caused a trans-membrane flow of thallium (potassium-mimetic cation). Both these effects were antagonized by selective blockers of BK channels. Furthermore, NAR half-reduced the calcium accumulation into the matrix of cardiac mitochondria exposed to high calcium concentrations. In conclusion, NAR exerts anti-ischemic effects through a "pharmacological preconditioning" that it is likely to be mediated, at least in part, by the activation of mitochondrial BK channels.


Assuntos
Cardiotônicos/farmacologia , Flavanonas/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Animais , Pressão Sanguínea , Cálcio/metabolismo , Transporte de Íons , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Potássio/metabolismo , Ratos , Ratos Wistar
10.
Pharmacol Res ; 70(1): 27-34, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23287425

RESUMO

Hydrogen sulphide (H2S) has been recently hypothesized to be an endogenous adipocyte-derived relaxing factor, evoking vasorelaxation of conductance and resistance vessels. Although the activation of ATP-sensitive potassium channels is known to play a central role in H2S-induced vasorelaxation, activation of vascular Kv7 voltage-gated potassium channels has also been suggested. To investigate this possibility, the ability of selective activators and blockers of distinct classes of potassium channels to affect vasodilation induced by the H2S-donor NaHS, as well as NaHS-induced Rb(+) efflux in endothelium-denuded rat aortic rings, was investigated. NaHS-induced changes of membrane potential were fluorimetrically assessed on human vascular smooth muscle (VSM) cells. Modulation of Kv7.4 channels by NaHS was assessed by electrophysiological studies, upon their heterologous expression in CHO cells. In isolated aortic rings, NaHS evoked vasorelaxing responses associated with an increase of Rb(+)-efflux. NaHS promoted membrane hyperpolarization of human VSM cells. These effects were antagonized by selective blockers of Kv7 channels. The H2S-donor caused a left-shift of current activation threshold of Kv7.4 channels expressed in CHO cells. Altogether, these results suggest that the activation of Kv7.4 channels is a key mechanism in the vascular effects of H2S. Given the relevant roles played by Kv7.4 channels in VSM contractility and by H2S in circulatory homeostasis regulation, these findings provide interesting insights to improve our understanding of H2S pathophysiology and to focus on Kv7.4 channels as novel targets for therapeutic approaches via the "H2S-system".


Assuntos
Aorta/efeitos dos fármacos , Sulfeto de Hidrogênio/farmacologia , Canais de Potássio KCNQ/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Sulfetos/farmacologia , Vasodilatação/efeitos dos fármacos , Animais , Aorta/metabolismo , Linhagem Celular , Interpretação Estatística de Dados , Relação Dose-Resposta a Droga , Endotélio Vascular/fisiologia , Humanos , Técnicas In Vitro , Canais de Potássio KCNQ/biossíntese , Masculino , Potenciais da Membrana/efeitos dos fármacos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Ratos , Ratos Wistar , Vasoconstritores/farmacologia
11.
Curr Med Chem ; 19(20): 3325-36, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22680638

RESUMO

Hydrogen sulphide (H(2)S) is now viewed as an important endogenous gasotransmitter, which exhibits many beneficial effects on the cardiovascular system. H(2)S is biosynthesized in mammalian tissues by both non-enzymatic processes and several enzymatic pathways ensured by cystathionine-ß-synthase and cystathionine-γ-lyase. H(2)S is endowed with the antioxidant properties of inorganic and organic sulphites, being a scavenger of reactive oxygen species. Furthermore, H(2)S triggers other important effects and the activation of ATP-sensitive potassium channels (KATP) accounts for its vasorelaxing and cardioprotective effects. H(2)S also inhibits smooth muscle proliferation and platelet aggregation. Conversely, the impairment of H(2)S contributes to the pathogenesis of hypertension and is involved in cardiovascular complications associated with diabetes mellitus. There is also evidence of a link between H(2)S and endothelial nitric oxide (NO). Recent observations indicate a possible pathogenic link between deficiencies of H(2)S activity and the progress of endothelial dysfunction. These biological aspects of endogenous H(2)S led to consider this mediator as "the new NO" and to evaluate new attractive opportunities to develop innovative classes of drugs. In this review, the main roles played by H(2)S in the cardiovascular system and the first examples of H(2)S-donor drugs are discussed. Some hybrid drugs are also addressed in this review. In such compounds opportune H(2)S-releasing moieties are conjugated to well-known drugs to improve their pharmacodynamic profile or to reduce the potential for adverse effects.


Assuntos
Sistema Cardiovascular/metabolismo , Sulfeto de Hidrogênio/metabolismo , Animais , Anti-Inflamatórios não Esteroides/metabolismo , Anti-Inflamatórios não Esteroides/farmacologia , Circulação Sanguínea/efeitos dos fármacos , Sistema Cardiovascular/efeitos dos fármacos , Linhagem Celular , Humanos
12.
J Med Chem ; 52(15): 4838-43, 2009 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-19601647

RESUMO

The inhibition of a subgroup of human serine/threonine protein phosphatases is responsible for the cytotoxicity of cantharidin and norcantharidin against tumor cells. It is shown that the anhydride rings of cantharidin and norcantharidin are hydrolyzed when bound to the catalytic domain of the human serine/threonine protein phosphatases 5 (PP5c), and the high-resolution crystal structures of PP5c complexed with the corresponding dicarboxylic acid derivatives of the two molecules are reported. Norcantharidin shows a unique binding conformation with the catalytically active Mn2PP5c, while cantharidin is characterized by a double conformation in its binding mode to the protein. Different binding modes of norcantharidin are observed depending of whether the starting ligand is in the anhydride or in the dicarboxylic acid form. All these structures will provide the basis for the rational design of new cantharidin-based drugs.


Assuntos
Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Cantaridina/farmacologia , Inibidores Enzimáticos/farmacologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Sítios de Ligação , Compostos Bicíclicos Heterocíclicos com Pontes/química , Cantaridina/química , Interações Hidrofóbicas e Hidrofílicas , Fosfoproteínas Fosfatases/química , Conformação Proteica , Relação Estrutura-Atividade
13.
Cardiovasc Hematol Agents Med Chem ; 6(1): 1-19, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18220718

RESUMO

One of the main approaches to the treatment of cardiovascular diseases is to block pathways and enzymes within the Renin-Angiotensin System (RAS) involved in the modulation of Angiotensin II. Besides this complex system, many other alternative strategies may represent interesting targets for new and more effective cardiovascular therapies. Many different approaches have led medicinal chemists to develop new molecules with the aim of improving current antihypertensive therapies. The development of these new compounds is based on different strategies which include the synthesis of new hybrid compounds in which two or more pharmacophore groups are combined together to give a new entity with better pharmacodynamic properties and fewer side effects, and the development of new molecules with targets such as renin, angiotensin (1-7) and urotensin-II. The aim of this review is to present various approaches used to improve antihypertensive therapy, developing both original molecules with new mechanisms of action (such as renin inhibitors, or Mas-agonists) and new hybrid cardiovascular drugs targeting multiple factors involved in hypertensive disease (NO-ACE inhibitors, NO-sartans, AT1/ETA antagonists).


Assuntos
Anti-Hipertensivos/química , Anti-Hipertensivos/uso terapêutico , Desenho de Fármacos , Hipertensão/tratamento farmacológico , Pressão Sanguínea/efeitos dos fármacos , Humanos , Hipertensão/metabolismo , Hipertensão/fisiopatologia
14.
J Appl Toxicol ; 27(3): 270-5, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17265420

RESUMO

Experimental approaches on anaesthetised guinea pigs have been shown recently to be satisfactorily predictive of the torsadogenic risk of drugs. This work aimed at obtaining additional data, for a further understanding of the reliability and/or the limits of this model. Clonidine (non-torsadogenic in humans) induced a lengthening of the ECG parameter of RR in anaesthetised guinea pigs, without any corresponding increase of QT (corrected by the algorithms of Bazett and Fridericia). Thus, 'QT correct' prolonging effects produced by drugs torsadogenic in humans, on the guinea pig model are primarily due to inhibition of cardiac repolarisation. The corresponding RR prolongation is a consequence (not the cause) of this primary effect. Astemizole, haloperidol and terfenadine, torsadogenic in humans, produced in Langendorff perfused guinea pig hearts a prolongation of the QT interval. Chlorprotixene (non-torsadogenic) did not produce any significant effect on QT. These results are fully consistent with previous observations in anaesthetised guinea pigs. In Langendorff perfused hearts, pentobarbital does not affect cardiac repolarisation and does not potentiate the QT-prolonging effect of astemizole. Together with the findings reported by many authors, these data suggest that ECG recording in anaesthetised guinea pigs is a reliable model for cardiac safety studies evaluating the influence of drugs on the repolarisation process.


Assuntos
Coração/efeitos dos fármacos , Síndrome do QT Longo/induzido quimicamente , Torsades de Pointes/induzido quimicamente , Animais , Anti-Hipertensivos/administração & dosagem , Anti-Hipertensivos/toxicidade , Astemizol/administração & dosagem , Astemizol/toxicidade , Clonidina/administração & dosagem , Clonidina/toxicidade , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/induzido quimicamente , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/fisiopatologia , Eletrocardiografia/efeitos dos fármacos , Cobaias , Haloperidol/administração & dosagem , Haloperidol/toxicidade , Coração/fisiopatologia , Técnicas In Vitro , Injeções Intravenosas , Síndrome do QT Longo/fisiopatologia , Perfusão/métodos , Terfenadina/administração & dosagem , Terfenadina/toxicidade , Torsades de Pointes/fisiopatologia
15.
Cardiovasc Hematol Agents Med Chem ; 5(1): 79-90, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17266550

RESUMO

Brief periods of ischaemia induce in the myocardium an increased resistance to the injury due to a subsequent, more prolonged ischaemic episode. This phenomenon, known as ischaemic pre-conditioning (IPC), articulated in two distinct phases (an early and a delayed one), is ensured by different biological mechanisms. Although an exhaustive comprehension of these mechanisms has not yet been reached, it is widely accepted that among the various signals involved as triggers and/or end-effectors, an important role is undoubtedly played by the activation of cardiac ATP-sensitive potassium channels (K(ATP)). In the myocardial cells, K(ATP) channels have been identified both in the sarcolemmal membrane (sarc-K(ATP)) and in the mitochondrial inner membrane (mito-K(ATP)). Although many experimental findings suggest that a role of sarc-K(ATP) channel activation in IPC cannot be excluded, in the last few years, many authors have indicated that this phenomenon could be attributed to the exclusive (or at least prevalent) activation of the mito-K(ATP) channels. Conversely, drugs modulating the K(ATP) channels (as activators or blockers), on one hand, have been employed as useful experimental tools for basic studies on IPC. On the other hand, K(ATP)-openers have been viewed as promising possible therapeutic agents for limiting the myocardial injury due to ischaemic episodes. In particular, those molecules exhibiting a good degree of selectivity towards the mito-K(ATP) channels have been indicated as potential anti-ischaemic cardio-protective pharmacological tools, devoid of other biological effects (such as negative inotropic activity, hypotension or hyperglycaemia) linked to the activation of cardiac and non-cardiac sarcK(ATP) channels. In this paper, we wish to report the experimental evidence supporting the role of sarc- and mito-K(ATP) channels in IPC, the relative signalling pathways potentially involved in the mechanisms of cardio-protection and, finally, an overview of the most important molecules acting as activators or blockers of K(ATP) channels, with their selectivity profiles.


Assuntos
Trifosfato de Adenosina/farmacologia , Isquemia Miocárdica/tratamento farmacológico , Canais de Potássio/efeitos dos fármacos , Animais , Cálcio/metabolismo , Humanos , Precondicionamento Isquêmico Miocárdico , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/fisiologia , Transdução de Sinais
16.
Br J Pharmacol ; 149(8): 1013-21, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17088866

RESUMO

UNLABELLED: BACKGROUND AND PURPOSE. The aim of this study was to investigate, in vascular smooth muscle cells, the mechanical and electrophysiological effects of (+/-)-naringenin. EXPERIMENTAL APPROACH: Aorta ring preparations and single tail artery myocytes were employed for functional and patch-clamp experiments, respectively. KEY RESULTS: (+/-)-Naringenin induced concentration-dependent relaxation in endothelium-denuded rat aortic rings pre-contracted with either 20 mM KCl or noradrenaline (pIC(50) values of 4.74 and 4.68, respectively). Tetraethylammonium, iberiotoxin, 4-aminopyridine and 60 mM KCl antagonised (+/-)-naringenin-induced vasorelaxation, while glibenclamide did not produce any significant antagonism. Naringin [(+/-)-naringenin 7-beta-neohesperidoside] caused a concentration-dependent relaxation of rings pre-contracted with 20 mM KCl, although its potency and efficacy were significantly lower than those of (+/-)-naringenin. In rat tail artery myocytes, (+/-)-naringenin increased large conductance Ca(2+)-activated K(+) (BK(Ca)) currents in a concentration-dependent manner; this stimulation was iberiotoxin-sensitive and fully reversible upon drug wash-out. (+/-)-Naringenin accelerated the activation kinetics of BK(Ca) current, shifted, by 22 mV, the voltage dependence of the activation curve to more negative potentials, and decreased the slope of activation. (+/-)-Naringenin-induced stimulation of BK(Ca) current was insensitive either to changes in the intracellular Ca(2+) concentration or to the presence, in the pipette solution, of the fast Ca(2+) chelator BAPTA. However, such stimulation was diminished when the K(+) gradient across the membrane was reduced. CONCLUSIONS AND IMPLICATIONS: The vasorelaxant effect of the naturally-occurring flavonoid (+/-)-naringenin on endothelium-denuded vessels was due to the activation of BK(Ca) channels in myocytes.


Assuntos
Flavanonas/farmacologia , Músculo Liso Vascular/metabolismo , Canais de Potássio Cálcio-Ativados/agonistas , Animais , Artérias/citologia , Artérias/efeitos dos fármacos , Artérias/metabolismo , Cálcio/metabolismo , Separação Celular , Quelantes/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Eletrofisiologia , Técnicas In Vitro , Cinética , Masculino , Potenciais da Membrana/efeitos dos fármacos , Relaxamento Muscular/efeitos dos fármacos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Vasodilatação/efeitos dos fármacos
17.
Curr Med Chem ; 13(6): 609-25, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16529554

RESUMO

Nitric oxide (NO) is an endogenous compound, which plays a fundamental role in the modulation of the function of the cardiovascular system, where it induces vasorelaxing and antiplatelet responses, mainly through the stimulation of guanylate cyclase and the increase of cGMP. Many drugs of common, time-honoured clinical use (for example, glycerol trinitrate and all the vasodilator nitrites and nitrates) act via the release of exogenous NO, thus mimicking the effects of the endogenous factor. In the last few years, a revision of the "one-compound-one-target" paradigm has led pharmacologists and pharmaceutical chemists to develop new classes of molecules which combine different pharmacodynamic properties. This innovative pharmacological/pharmaceutical strategy has produced hybrid drugs, with a dual mechanism of action: a) the slow release of nitric oxide and b) another fundamental pharmacodynamic profile. These drugs have been obtained by inserting appropriate NO-donor chemical groups (i.e. nitrate esters, nitrosothiols, etc.), linked to a known drug, by means of a variable spacer moiety. These new pharmacodynamic hybrids present the advantage of combining a basic mechanism of action (for example, cyclooxygenase inhibition, beta-antagonism or ACE inhibition) with a slow release of NO, which may be useful either to reduce adverse side effects (for example, the gastrotoxicity of NSAIDs), or to improve the effectiveness of the drug (for example, conferring direct vasorelaxing and antiplatelet effects on an ACE-inhibitor). The aim of this review is to present the chemical features of NO-releasing hybrids of cardiovascular drugs, and to explain the pharmacological improvements obtained by the addition of the NO-donor properties.


Assuntos
Fármacos Cardiovasculares/farmacologia , Sistema Cardiovascular/efeitos dos fármacos , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico/metabolismo , Fármacos Cardiovasculares/química , Fármacos Cardiovasculares/metabolismo , Humanos , Conformação Molecular , Doadores de Óxido Nítrico/química , Doadores de Óxido Nítrico/metabolismo , Relação Estrutura-Atividade
18.
Curr Med Chem ; 11(20): 2691-706, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15544470

RESUMO

Many non-cardiovascular drugs of common clinical use cause, as an unwanted accessory property, the prolongation of the cardiac repolarisation process, due to the block of the HERG (Human Ether-a-go-go Related Gene) potassium channel, responsible for the repolarising I(Kr) current. This delayed cardiac repolarisation process can be often unmasked by a prolongation of the QT interval of the ECG. In these conditions, premature action potentials can generate morphologically anomalous after-polarisations, and trigger a dangerous kind of polymorphic ventricular tachyarrhythmia, known as torsade de pointes, which can evolve in ventricular fibrillation and death. The risk associated with the torsadogenic cardiotoxicity of drugs, which prolong the QT interval has been the topic of documents produced by many health authorities, giving important issues about the preclinical and clinical evaluation of cardiac safety. Besides, public and private research laboratories developed several experimental in vitro or in vivo strategies, aimed to an early recognition of the influence of a drug (or of a drug-candidate) on the HERG channel and/or on the cardiac repolarisation process. Also the identification of a possible pharmacophore model, common in all or at least in numerous torsadogenic drugs, could represent a first step for the development of useful in silico approaches, allowing a preliminary indication about the potential torsadogenic property of a given molecule. In this work, we described the electrophysiological basis of torsade de pointes and listed several pharmacological classes of torsadogenic drugs. Among them, we focused our attention on antipsychotics, with an accurate overview on the experimental and clinical reports about their torsadogenic properties. Moreover, a common structural feature exhibited by these drugs, despite of their remarkable chemical differences, is evidenced by a computational approach and is indicated as a possible "facilitating" requirement for their torsadogenic properties. Together with other remarks, coming from different computational studies, the individuation of a satisfactory "toxicophore" model could be greatly useful, for the theoretical prediction of torsadogenic properties of a given chemical moiety and for the design of new drugs devoid of such an undesired and potentially lethal side-effect.


Assuntos
Antipsicóticos/efeitos adversos , Antipsicóticos/química , Proteínas de Transporte de Cátions/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Torsades de Pointes/induzido quimicamente , Animais , Antipsicóticos/metabolismo , Proteínas de Transporte de Cátions/fisiologia , Canais de Potássio Éter-A-Go-Go , Humanos , Síndrome do QT Longo/induzido quimicamente , Síndrome do QT Longo/fisiopatologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Torsades de Pointes/fisiopatologia
19.
J Appl Toxicol ; 24(3): 217-22, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15211616

RESUMO

Many non-cardiovascular drugs can prolong the QT interval of the electrocardiogram (ECG); this is an accessory property not necessary for their pharmacological action and generally linked to the block of the potassium HERG channels and delayed cardiac repolarization. The QT prolongation can lead to a dangerous tachyarrhythmia, called torsade de pointes, and potentially to fatal ventricular fibrillation. The experimental approaches, aimed at an early identification of this undesidered property, often require sophisticated and expensive equipment or the use of superior animal species (dog, primates) that cannot be employed easily for ethical and/or economic reasons. This work aimed to study drug-induced QT prolongation in anaesthetized guinea-pigs and to evaluate the reliability of such an experimental approach to obtain a satisfying predictive parameter of the torsadogenicity of drugs in humans. Seven drugs that were torsadogenic in humans (astemizole, cisapride, haloperidol, quinidine, sotalol, terfenadine and thioridazine) and two that were non-torsadogenic (chlorprotixene and diazepam) were administered i.v. to guinea-pigs under pentobarbital anaesthesia. The ECGs were recorded by four electrodes inserted in the subcutaneous layer of the limbs. Both RR and QT intervals were measured in Leads II and III and then the correct QT values were calculated by Bazett and Fridericia algorithms (QTcB and QTcF, respectively). All the drugs, with the exception of chlorprotixene and diazepam, produced a dose-dependent prolongation of the QT and RR intervals and a significant increase of QTcB and QTcF values. It can be concluded that this method represents a rapid and low-cost procedure to evaluate the cardiac safety pro fi le in the preliminary screening of a high number of drugs or drug candidates.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Eletrocardiografia/efeitos dos fármacos , Torsades de Pointes/induzido quimicamente , Animais , Avaliação Pré-Clínica de Medicamentos/métodos , Cobaias , Masculino , Valor Preditivo dos Testes
20.
J Mol Biol ; 336(3): 707-16, 2004 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-15095982

RESUMO

The catalytic domain of matrix metalloproteinase-10 (MMP-10) has been expressed in Escherichia coli and its crystal structure solved at 2.1 A resolution. The availability of this structure allowed us to critically examine the small differences existing between the catalytic domains of MMP-3 and MMP-10, which show the highest sequence identity among all MMPs. Furthermore, the binding mode of N-isobutyl-N-[4-methoxyphenylsulfonyl]glycyl hydroxamic acid (NNGH), which is one of the most known commercial inhibitors of MMPs, is described for the first time.


Assuntos
Metaloendopeptidases/química , Estrutura Terciária de Proteína , Sequência de Aminoácidos , Domínio Catalítico , Cristalografia por Raios X , Humanos , Metaloproteinase 10 da Matriz , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...