Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Neuropharmacology ; 203: 108884, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34785163

RESUMO

Epilepsy, one of the most common and most disabling neurological disorders, is characterized by spontaneous recurrent seizures, often associated with structural brain alterations and cognitive and psychiatric comorbidities. In about 30% of patients, the seizures are resistant to current treatments; so more effective treatments are urgently needed. Among the ∼30 clinically approved antiseizure drugs, retigabine (ezogabine) is the only drug that acts as a positive allosteric modulator (or opener) of voltage-gated Kv7 potassium channels, which is particularly interesting for some genetic forms of epilepsy. Here we describe a novel dual-mode-of-action compound, GRT-X (N-[(3-fluorophenyl)-methyl]-1-(2-methoxyethyl)-4-methyl-2-oxo-(7-trifluoromethyl)-1H-quinoline-3-carboxylic acid amide) that activates both Kv7 potassium channels and the mitochondrial translocator protein 18 kDa (TSPO), leading to increased synthesis of brain neurosteroids. TSPO activators are known to exert anti-inflammatory, neuroprotective, anxiolytic, and antidepressive effects, which, together with an antiseizure effect (mediated by Kv7 channels), would be highly relevant for the treatment of epilepsy. This prompted us to compare the antiseizure efficacy of retigabine and GRT-X in six mouse and rat models of epileptic seizures, including the 6-Hz model of difficult-to-treat focal seizures. Furthermore, the tolerability of the two compounds was compared in mice and rats. Potency comparisons were based on both doses and peak plasma concentrations. Overall, GRT-X was more effective than retigabine in three of the six seizure models used here, the most important difference being the high efficacy in the 6-Hz (32 mA) seizure model in mice. Based on drug plasma levels, GRT-X was at least 30 times more potent than retigabine in the latter model. These data indicate that GRT-X is a highly interesting novel anti-seizure drug with a unique (first-in-class) dual-mode mechanism of action.


Assuntos
Anticonvulsivantes/uso terapêutico , Carbamatos/uso terapêutico , Fenilenodiaminas/uso terapêutico , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Receptores de GABA/metabolismo , Convulsões/tratamento farmacológico , Convulsões/metabolismo , Animais , Anticonvulsivantes/química , Anticonvulsivantes/farmacologia , Células CHO , Carbamatos/farmacologia , Cricetulus , Relação Dose-Resposta a Droga , Eletrochoque/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Fenilenodiaminas/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Convulsões/etiologia , Resultado do Tratamento
2.
Eur J Pharmacol ; 905: 174190, 2021 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-34015322

RESUMO

Narirutin is one of the most common flavanones found in citrus fruits. The vascular effects of its analogues naringenin and naringin have been reported but its effects on the cardiovascular system are largely unknown. In this study, relaxation effect of narirutin and its mechanisms of action were investigated by measuring isometric tension in rat mesenteric arteries. Patch-clamping was also used to study the effect of narirutin on potassium channels in vascular smooth muscle cells. Moreover, its effects on phosphorylation of endothelial nitric oxide synthase, cAMP level and phosphodiesterase activity in rat mesenteric arteries were studied by Western blot and biochemical assays. The results showed that pre-incubation of rat mesenteric arteries with narirutin had no influence on acetylcholine-induced endothelial-dependent relaxation. However, narirutin caused a direct concentration-dependent relaxation in rat mesenteric arteries. This relaxation effect was comparable to that of narirutin's structural analogue naringenin. Narirutin-induced relaxation was reduced by the removal of endothelium, NG-nitro-L-arginine methyl ester (a nitric oxide synthase inhibitor), and 4-aminopyridine (a voltage-gated potassium channel blocker). In addition, narirutin increased the phosphorylation of endothelial nitric oxide synthase and increased the voltage-dependent potassium current in mesenteric arterial smooth muscle cells. These effects were abolished by protein kinase A inhibitor. Furthermore, narirutin could increase cAMP level and inhibit phosphodiesterase activity in rat mesenteric arteries. In conclusion, narirutin has vasorelaxing effect and the mechanism involves the inhibition of phosphodiesterase, which increases intracellular cAMP, thereby stimulating the endothelial nitric oxide synthase and activating the voltage-gated potassium channels in vascular smooth muscle cells.


Assuntos
Dissacarídeos/farmacologia , Flavanonas/farmacologia , Artérias Mesentéricas/efeitos dos fármacos , Relaxamento Muscular/efeitos dos fármacos , Óxido Nítrico/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , Vasodilatadores/farmacologia , Animais , AMP Cíclico/metabolismo , Endotélio Vascular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Óxido Nítrico Sintase Tipo III/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Ratos Sprague-Dawley
3.
Eur J Pharmacol ; 898: 173991, 2021 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-33684451

RESUMO

In the present study, we investigated the vasorelaxant effects of alogliptin, an oral antidiabetic drug in the dipeptidyl peptidase-4 (DPP-4) inhibitor class, using phenylephrine (Phe)-induced pre-contracted aortic rings. Alogliptin induced vasorelaxation in a dose-dependent manner. Pre-treatment with the voltage-dependent K+ (Kv) channel inhibitor 4-aminopyridine (4-AP) significantly decreased the vasorelaxant effect of alogliptin, whereas pre-treatment with the inwardly rectifying K+ (Kir) channel inhibitor Ba2+, ATP-sensitive K+ (KATP) channel inhibitor glibenclamide, and large-conductance Ca2+-activated K+ (BKCa) channel inhibitor paxilline did not alter the effects of alogliptin. Although pre-treatment with the Ca2+ channel inhibitor nifedipine did not affect the vasorelaxant effect of alogliptin, pre-treatment with the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) pump inhibitors thapsigargin and cyclopiazonic acid effectively attenuated the vasorelaxant response of alogliptin. Neither cGMP/protein kinase G (PKG)-related signaling pathway inhibitors (guanylyl cyclase inhibitor ODQ and PKG inhibitor KT 5823) nor cAMP/protein kinase A (PKA)-related signaling pathway inhibitors (adenylyl cyclase inhibitor SQ 22536 and PKA inhibitor KT 5720) reduced the vasorelaxant effect of alogliptin. Similarly, the vasorelaxant effect of alogliptin was not changed by endothelium removal or pre-treatment with the nitric oxide (NO) synthase inhibitor L-NAME or the small- and intermediate-conductance Ca2+-activated K+ (SKCa and IKCa) channel inhibitors apamin and TRAM-34. Based on these results, we suggest that alogliptin induced vasorelaxation in rabbit aortic smooth muscle by activating Kv channels and the SERCA pump independent of other K+ channels, cGMP/PKG-related or cAMP/PKA-related signaling pathways, and the endothelium.


Assuntos
Músculo Liso Vascular/efeitos dos fármacos , Piperidinas/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Uracila/análogos & derivados , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/enzimologia , Ativação Enzimática , Masculino , Músculo Liso Vascular/enzimologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Coelhos , Transdução de Sinais , Uracila/farmacologia
4.
ChemMedChem ; 15(12): 1078-1088, 2020 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-32338831

RESUMO

The slow delayed rectifier potassium current (IKs ) is formed by the KCNQ1 (Kv 7.1) channel, an ion channel of four α-subunits that modulates KCNE1 ß-subunits. IKs is central to the repolarization of the cardiac action potential. Loss of function mutation reducing ventricular cardiac IKs cause the long-QT syndrome (LQTS), a disorder that predisposes patients to arrhythmia and sudden death. Current therapy for LQTS is inadequate. Rottlerin, a natural product of the kamala tree, activates IKs and has the potential to provide a new strategy for rational drug therapy. In this study, we show that simple modifications such as penta-acetylation or penta-methylation of rottlerin blunts activation activity. Total synthesis was used to prepare side-chain-modified derivatives that slowed down KCNQ1/KCNE1 channel deactivation to different degrees. A binding hypothesis of rottlerin is provided that opens the way to improved IKs activators as novel therapeutics for the treatment of LQTS.


Assuntos
Acetofenonas/farmacologia , Benzopiranos/farmacologia , Canal de Potássio KCNQ1/agonistas , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , Proteínas de Xenopus/agonistas , Acetofenonas/síntese química , Acetofenonas/metabolismo , Animais , Benzopiranos/síntese química , Benzopiranos/metabolismo , Sítios de Ligação , Humanos , Canal de Potássio KCNQ1/metabolismo , Simulação de Acoplamento Molecular , Oócitos/efeitos dos fármacos , Ligação Proteica , Xenopus laevis
5.
Acta Physiol (Oxf) ; 229(4): e13471, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32223014

RESUMO

AIM: We aimed to assess the ability of natural and modified polyunsaturated fatty acids (PUFAs) to shorten QT interval in ex-vivo and in-vivo guinea pig hearts. METHODS: The effect of one natural (docosahexaenoic acid [DHA]) and three modified (linoleoyl glycine [Lin-GLY], docosahexaenoyl glycine [DHA-GLY], N-arachidonoyl taurine [N-AT]) PUFAs on ventricular action potential duration (APD) and QT interval was studied in a E4031 drug-induced long QT2 model of ex-vivo guinea pig hearts. The effect of DHA-GLY on QT interval was also studied in in-vivo guinea pig hearts upon intravenous administration. The effect of modified PUFAs on IKs was studied using Xenopus laevis oocytes expressing human KCNQ1 and KCNE1. RESULTS: All tested PUFAs shortened ADP and QT interval in ex-vivo guinea pig hearts, however, with different ability in restoring baseline APD/QT interval with specific modified PUFAs being most efficacious. Despite comparable ability in activating the human KCNQ1/KCNE1 channel, Lin-GLY was not as effective in shortening APD/QT interval as DHA-GLY in ex-vivo hearts. By constructing a guinea pig-like KCNE1, we found Lin-GLY to induce less activating effect compared with DHA-GLY on human KCNQ1 co-expressed with guinea pig-like KCNE1. Docosahexaenoyl glycine was studied in more detail and was found to shorten QT interval in in-vivo guinea pig hearts. CONCLUSION: Our results show that specific PUFAs shorten QT interval in guinea pig hearts. The tendency of modified PUFAs with pronounced IKs channel activating effect to better restore QT interval suggests that modifying PUFAs to target the IKs channel is a means to improve the QT-shortening effect.


Assuntos
Ácidos Graxos Insaturados/farmacologia , Coração/efeitos dos fármacos , Canal de Potássio KCNQ1/agonistas , Potenciais de Ação , Animais , Cobaias , Ventrículos do Coração , Técnicas In Vitro , Síndrome do QT Longo , Oócitos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , Xenopus laevis
6.
Mol Pharmacol ; 97(2): 132-144, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31722973

RESUMO

The pairing of KCNQ1 and KCNE1 subunits together mediates the cardiac slow delayed rectifier current (I Ks ), which is partly responsible for cardiomyocyte repolarization and physiologic shortening of the cardiac action potential. Mefenamic acid, a nonsteroidal anti-inflammatory drug, has been identified as an I Ks activator. Here, we provide a biophysical and pharmacological characterization of mefenamic acid's effect on I Ks Using whole-cell patch clamp, we show that mefenamic acid enhances I Ks activity in both a dose- and stoichiometry-dependent fashion by changing the slowly activating and deactivating I Ks current into an almost linear current with instantaneous onset and slowed tail current decay, sensitive to the I Ks blocker (3R,4S)-(+)-N-[3-hydroxy-2,2-dimethyl-6-(4,4,4-trifluorobutoxy) chroman-4-yl]-N-methylmethanesulfonamide (HMR1556). Both single channels, which reveal no change in the maximum conductance, and whole-cell studies, which reveal a dramatically altered conductance-voltage relationship despite increasingly longer interpulse intervals, suggest mefenamic acid decreases the voltage sensitivity of the I Ks channel and shifts channel gating kinetics toward more negative potentials. Modeling studies revealed that changes in voltage sensor activation kinetics are sufficient to reproduce the dose and frequency dependence of mefenamic acid action on I Ks channels. Mutational analysis showed that mefenamic acid's effect on I Ks required residue K41 and potentially other surrounding residues on the extracellular surface of KCNE1, and explains why the KCNQ1 channel alone is insensitive to up to 1 mM mefenamic acid. Given that mefenamic acid can enhance all I Ks channel complexes containing different ratios of KCNQ1 to KCNE1, it may provide a promising therapeutic approach to treating life-threatening cardiac arrhythmia syndromes. SIGNIFICANCE STATEMENT: The channels which generate the cardiac slow delayed rectifier K+ current (I Ks ) are composed of KCNQ1 and KCNE1 subunits. Due to the critical role played by I Ks in heartbeat regulation, enhancing I Ks current has been identified as a promising therapeutic strategy to treat various heart rhythm diseases. Most I Ks activators, unfortunately, only work on KCNQ1 alone and not the physiologically relevant I Ks channel. We have demonstrated that mefenamic acid can enhance I Ks in a dose- and stoichiometry-dependent fashion, regulated by its interactions with KCNE1.


Assuntos
Antiarrítmicos/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Ácido Mefenâmico/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Potássio/metabolismo , Animais , Antiarrítmicos/uso terapêutico , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/fisiopatologia , Relação Dose-Resposta a Droga , Fibroblastos , Células HEK293 , Frequência Cardíaca/fisiologia , Humanos , Canal de Potássio KCNQ1/química , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Mutagênese Sítio-Dirigida , Mutação , Miocárdio/metabolismo , Técnicas de Patch-Clamp , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais/efeitos dos fármacos
7.
Eur J Pharmacol ; 870: 172873, 2020 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-31866408

RESUMO

Hydroxy-safflor yellow A (HSYA) can exert a variety of effects upon the vascular system. However, the underlying mechanisms are not clear. The present study is to investigate its vasodilating effect and the mechanisms. Wistar-Kyoto (WKY) rats and spontaneously hypertensive rats (SHR) were enrolled for studying effects of HSYA on blood pressure, vasodilation, intracellular Ca2+ transient and membrane ion channels. Vasodilation and intracellular Ca2+ transient were measured by using vasomotor assay and fluorescence imaging system, respectively. The effect of HSYA on the large conductance Ca2+ activated and voltage-gated potassium channel (BKCa channel) currents in rat mesentery artery and on L-type calcium channel (Ca-L) currents in HEK293cells expressed with Ca-L were investigated using patch clamp techniques. Blood pressure of SHR and WKY rats were concentration dependently reduced by HSYA with a larger effect of HSYA in SHR than that in WKY rats. The tension of mesenteric arteries induced by 3 µM phenylephrine was attenuated by HSYA (IC50 = 90.8 µΜ). Patch clamp study showed that HSYA could activate BKCa channels and suppress Ca-L channels in a concentration dependent manner. The results of calcium signaling assays indicated that HSYA could reduce the intracellular free Ca2+ level. These findings demonstrate that HSYA could activate BKCa channels and inhibit Ca-L channels and reduce intracellular free Ca2+ level, which are probably important for its vasodilatory effect.


Assuntos
Agonistas dos Canais de Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/metabolismo , Chalcona/análogos & derivados , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , Quinonas/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Agonistas dos Canais de Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/metabolismo , Sinalização do Cálcio , Chalcona/metabolismo , Chalcona/farmacologia , Células HEK293 , Humanos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Artérias Mesentéricas/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Técnicas de Patch-Clamp , Fenilefrina/metabolismo , Quinonas/metabolismo , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Vasodilatação/efeitos dos fármacos
8.
J Cardiovasc Pharmacol ; 74(3): 218-224, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31356552

RESUMO

Relaxation and changes in the transmembrane potential of vascular smooth muscle induced by ORM-3819, a novel inodilating compound, were investigated in isolated porcine coronary arteries. Isometric tone was studied on arterial rings precontracted by KCl (30 mM), and resting membrane potential was investigated by a conventional microelectrode technique. ORM-3819 in the concentration range 0.38-230.6 µM evoked concentration-dependent relaxation with a maximum value of 58.1% and an effective concentration of the relaxing substance that caused 50% of maximum relaxation of 72.2 µM. The maximum hyperpolarization produced by ORM-3819 at a concentration of 120 µM (-2.6 ± 0.81 mV, N = 10) did not differ significantly from that induced by C-type natriuretic peptide (CNP), an endogenous hyperpolarizing mediator, at a concentration of 1.4 µM (-3.6 ± 0.38 mV, N = 17). The same effect elicited by the known inodilator levosimendan was less pronounced at a concentration of 3.7 µM: -1.82 ± 0.44 mV, N = 22 (P < 0.05 vs. CNP). The voltage-gated potassium channel inhibitor 4-aminopyridine, at a concentration of 5 mM, attenuated the relaxation induced by ORM-3819 at concentrations of 41.6 or 117.2 µM. These results suggest that ORM-3819 is a potent vasodilating agent able to relieve coronary artery vasospasm by causing hyperpolarization of vascular smooth muscle cells through processes involving activation of voltage-gated potassium channels.


Assuntos
Vasos Coronários/efeitos dos fármacos , Hidrazonas/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , Piridazinas/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Vasos Coronários/metabolismo , Técnicas In Vitro , Potenciais da Membrana , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Peptídeo Natriurético Tipo C/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Transdução de Sinais , Simendana/farmacologia , Sus scrofa
9.
Microvasc Res ; 123: 42-49, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30367840

RESUMO

Tetrahydroxystilbene glucoside (TSG) is the main water-soluble component in Polygonum multiflorum Thunb, and it has many cardioprotective effects. Although TSG is able to relax blood vessels, its relaxation of rat superior mesenteric arteries and the underlying mechanism of this process are not clearly understood. The aim of the present study was to use in vivo and in vitro models to investigate the arterial relaxation effect of TSG on rat superior mesenteric arteries and the mechanisms involved. We found that TSG concentration-dependently relaxed the superior mesenteric artery with or without endothelium. The vasorelaxation induced by TSG is not related to the vasodilator derived factor NO but is rather by the inhibition of COX-2 activity and decreased TXA2. We also found that the vasorelaxation induced by TSG was attenuated by 4­AP. Moreover, TSG also inhibited the contraction induced by an increase in external calcium concentration in Ca2+-free medium plus KCl (60 mM). These results suggest that TSG induces relaxation in mesenteric arterial rings through an endothelium-dependent pathway that involves the inhibition of COX-2 activity and decreased in TXA2 and through an endothelium-independent pathway via opening of a voltage-dependent K+ channel, blockade of Ca2+ influx and release of intracellular Ca2+.


Assuntos
Inibidores de Ciclo-Oxigenase 2/farmacologia , Endotélio Vascular/efeitos dos fármacos , Glucosídeos/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Estilbenos/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Ciclo-Oxigenase 2/metabolismo , Relação Dose-Resposta a Droga , Endotélio Vascular/metabolismo , Masculino , Artéria Mesentérica Superior/efeitos dos fármacos , Artéria Mesentérica Superior/metabolismo , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Ratos Sprague-Dawley , Tromboxano A2/metabolismo
10.
Acta Physiol (Oxf) ; 225(2): e13186, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30184322

RESUMO

AIM: The IKs channel is important for termination of the cardiac action potential. Hundreds of loss-of-function mutations in the IKs channel reduce the K+ current and, thereby, delay the repolarization of the action potential, causing Long QT Syndrome. Long QT predisposes individuals to Torsades de Pointes which can lead to ventricular fibrillation and sudden death. Polyunsaturated fatty acids (PUFAs) are potential therapeutics for Long QT Syndrome, as they affect IKs channels. However, it is unclear which properties of PUFAs are essential for their effects on IKs channels. METHODS: To understand how PUFAs influence IKs channel activity, we measured effects on IKs current by two-electrode voltage clamp while changing different properties of the hydrocarbon tail. RESULTS: There was no, or weak, correlation between the tail length or number of double bonds in the tail and the effects on or apparent binding affinity for IKs channels. However, we found a strong correlation between the positions of the double bonds relative to the head group and effects on IKs channels. CONCLUSION: Polyunsaturated fatty acids with double bonds closer to the head group had higher apparent affinity for IKs channels and increased IKs current more; shifting the bonds further away from the head group reduced apparent binding affinity for and effects on the IKs current. Interestingly, we found that ω-6 and ω-9 PUFAs, with the first double bond closer to the head group, left-shifted the voltage dependence of activation the most. These results allow for informed design of new therapeutics targeting IKs channels in Long QT Syndrome.


Assuntos
Ácidos Graxos Ômega-6/farmacologia , Ácidos Graxos Insaturados/farmacologia , Canal de Potássio KCNQ1/agonistas , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , Potenciais de Ação , Animais , Células Cultivadas , Ácidos Graxos Ômega-6/química , Ácidos Graxos Insaturados/química , Humanos , Canal de Potássio KCNQ1/metabolismo , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Xenopus laevis
11.
Cardiovasc Toxicol ; 19(3): 244-254, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30519910

RESUMO

This study investigated vildagliptin-induced vasodilation and its related mechanisms using phenylephrine induced precontracted rabbit aortic rings. Vildagliptin induced vasodilation in a concentration-dependent manner. Pretreatment with the large-conductance Ca2+-activated K+ channel blocker paxilline, ATP-sensitive K+ channel blocker glibenclamide, and inwardly rectifying K+ channel blocker Ba2+ did not affect the vasodilatory effects of vildagliptin. However, application of the voltage-dependent K+ (Kv) channel inhibitor 4-aminopyridine significantly reduced the vasodilatory effects of vildagliptin. In addition, application of either of two sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA) inhibitors, thapsigargin or cyclopiazonic acid, effectively inhibited the vasodilatory effects of vildagliptin. These vasodilatory effects were not affected by pretreatment with adenylyl cyclase, protein kinase A (PKA), guanylyl cyclase, or protein kinase G (PKG) inhibitors, or by removal of the endothelium. From these results, we concluded that vildagliptin induced vasodilation via activation of Kv channels and the SERCA pump. However, other K+ channels, PKA/PKG-related signaling cascades associated with vascular dilation, and the endothelium were not involved in vildagliptin-induced vasodilation.


Assuntos
Inibidores da Dipeptidil Peptidase IV/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Vildagliptina/farmacologia , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/enzimologia , Ativação Enzimática , Masculino , Músculo Liso Vascular/enzimologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Coelhos , Transdução de Sinais
12.
Methods Enzymol ; 602: 339-368, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29588038

RESUMO

Voltage-gated ion channels (VGICs) of excitable tissues are emerging as targets likely involved in both the therapeutic and toxic effects of inhaled and intravenous general anesthetics. Whereas sevoflurane and propofol inhibit voltage-gated Na+ channels (Navs), sevoflurane potentiates certain voltage-gated K+ channels (Kvs). The combination of these effects would dampen neural excitability and, therefore, might contribute to the clinical endpoints of general anesthesia. As the body of work regarding the interaction of general anesthetics with VGICs continues to grow, a multidisciplinary approach involving functional, biochemical, structural, and computational techniques, many of which are detailed in other chapters, has increasingly become necessary to solve the molecular mechanism of general anesthetic action on VGICs. Here, we focus on electrophysiological and modeling approaches and methodologies to describe how our work has elucidated the biophysical basis of the inhibition Navs by propofol and the potentiation of Kvs by sevoflurane.


Assuntos
Anestésicos Gerais/farmacologia , Eletrofisiologia/métodos , Ativação do Canal Iônico/efeitos dos fármacos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Canais de Sódio Disparados por Voltagem/metabolismo , Animais , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Simulação por Computador , Eletrodos , Eletrofisiologia/instrumentação , Células HEK293 , Humanos , Ativação do Canal Iônico/fisiologia , Modelos Biológicos , Oócitos , Técnicas de Patch-Clamp/instrumentação , Técnicas de Patch-Clamp/métodos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , Propofol/farmacologia , Proteínas Recombinantes/metabolismo , Sevoflurano/farmacologia , Transfecção/instrumentação , Transfecção/métodos , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Xenopus laevis
13.
Cardiovasc Ther ; 36(1)2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28834298

RESUMO

AIMS: We investigated the vasorelaxant effect of nateglinide and its related mechanisms using phenylephrine (Phe)-induced precontracted aortic rings. METHODS: Arterial tone measurement was performed in aortic smooth muscle. RESULTS: The application of nateglinide induced vasorelaxation in a concentration-dependent manner. Pretreatment with the large-conductance Ca2+ -activated K+ (BKCa ) channel inhibitor paxilline, the inwardly rectifying K+ (Kir) channel inhibitor Ba2+ , and ATP-sensitive K+ (KATP ) channel inhibitor glibenclamide did not affect the vasorelaxant effect of nateglinide. However, pretreatment with the voltage-dependent K+ (Kv) channel inhibitor 4-aminopyridine (4-AP) effectively reduced the vasorelaxant effect of nateglinide. Pretreatment with the Ca2+ inhibitor nifedipine and the sarcoplasmic/endoplasmic reticulum Ca2+ -ATPase inhibitor thapsigargin did not change the vasorelaxant effect of nateglinide. Additionally, the vasorelaxant effect of nateglinide was not altered in the presence of an adenylyl cyclase, a protein kinase A, a guanylyl cyclase, or a protein kinase G inhibitor. The vasorelaxant effect of nateglinide was not affected by the elimination of the endothelium. In addition, pretreatment with a nitric oxide synthase inhibitor, L-NAME, and a small-conductance Ca2+ -activated K+ (SKCa ) channel inhibitor, apamin, did not change the vasorelaxant effect of nateglinide. CONCLUSION: Nateglinide induced vasorelaxation via the activation of the Kv channel independent of other K+ channels, Ca2+ channels, intracellular Ca2+ ([Ca2+ ]i ), and the endothelium.


Assuntos
Cicloexanos/farmacologia , Hipoglicemiantes/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Fenilalanina/análogos & derivados , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/metabolismo , Relação Dose-Resposta a Droga , Técnicas In Vitro , Masculino , Músculo Liso Vascular/metabolismo , Nateglinida , Fenilalanina/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Coelhos , Transdução de Sinais/efeitos dos fármacos
14.
Life Sci ; 188: 1-9, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28855109

RESUMO

AIMS: The vasorelaxant effects of the anti-diabetic drug, mitiglinide in phenylephrine (Phe)-pre-contracted aortic rings were examined. MATERIALS AND METHODS: Arterial tone measurement was performed in aortic smooth muscle cells. KEY FINDINGS: Mitiglinide dose-dependently induced vasorelaxation. Application of the large-conductance Ca2+-activated K+ (BKCa) channel blocker paxilline, inwardly rectifying K+ (Kir) channel blocker Ba2+, and ATP-sensitive K+ (KATP) channel blocker glibenclamide did not affect the vasorelaxant effect of mitiglinide. However, application of the voltage-dependent K+ (Kv) channel blocker 4-AP, effectively inhibited mitiglinide-induced vasorelaxation. Although pretreatment with the Ca2+ channel blocker nifedipine did not alter the mitiglinide-induced vasorelaxation, pretreatment with the sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA) pump inhibitor thapsigargin and cyclopiazonic acid reduced the vasorelaxant effect of mitiglinide. In addition, the vasorelaxant effect of mitiglinide was not affected by the inhibitors of adenylyl cyclase, protein kinase A, guanylyl cyclase, or protein kinase G. Elimination of the endothelium and inhibition of endothelium-dependent vasorelaxant mechanisms also did not change the vasorelaxant effect of mitiglinide. SIGNIFICANCE: We proposed that mitiglinide induces vasorelaxation via activation of Kv channels and SERCA pump. However, the vasorelaxant effects of mitiglinide did not involve other K+ channels, Ca2+ channels, PKA/PKG signaling pathways, or the endothelium.


Assuntos
Aorta Torácica/fisiologia , Isoindóis/farmacologia , Músculo Liso/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/fisiologia , Vasodilatadores/farmacologia , 4-Aminopiridina/farmacologia , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Aorta Torácica/efeitos dos fármacos , Bário/farmacologia , Carbazóis/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Endotélio Vascular/efeitos dos fármacos , Glibureto/farmacologia , Indóis/farmacologia , Isoindóis/antagonistas & inibidores , Masculino , Músculo Liso/efeitos dos fármacos , Nifedipino/farmacologia , Oxidiazóis/farmacologia , Fenilefrina/farmacologia , Pirróis/farmacologia , Quinoxalinas/farmacologia , Coelhos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Tapsigargina/farmacologia , Vasodilatadores/antagonistas & inibidores
15.
Nat Rev Neurol ; 13(5): 290-301, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28418022

RESUMO

The discovery, in 2010, of autoantibodies against the extracellular proteins LGI1 and Caspr2 facilitated a change of view regarding the clinical importance of voltage-gated potassium channel (VGKC) antibodies. Currently, these antibodies are all classified as VGKC-complex antibodies, and are commonly considered to have a similar clinical value. However, studies from the past few years show that the immune responses mediated by these antibodies have differing clinical relevance. Here, we review the clinical importance of these immune responses in three settings: patients with anti-LGI1 antibodies, patients with anti-Caspr2 antibodies, and patients with antibodies against the VGKC complex that lack LGI1 and Caspr2 specificity. Antibodies against LGI1 and Caspr2 are associated with different but well-defined syndromes, whereas the clinical importance of VGKC-complex antibodies without LGI1 and Caspr2 specificity is questionable. We describe each of these syndromes, discuss the function of the target antigens and review the limited paediatric literature on the topic. The findings emphasize the importance of defining these disorders according to the molecular identity of the targets (LGI1 or Caspr2), and caution against the use of VGKC-complex antibodies for the diagnosis and treatment of patients without further definition of the antigen.


Assuntos
Autoanticorpos/imunologia , Doenças Autoimunes do Sistema Nervoso/imunologia , Encefalite/imunologia , Proteínas de Membrana/imunologia , Proteínas do Tecido Nervoso/imunologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/imunologia , Proteínas/imunologia , Animais , Doenças Autoimunes do Sistema Nervoso/diagnóstico , Doenças Autoimunes do Sistema Nervoso/fisiopatologia , Encefalite/diagnóstico , Encefalite/fisiopatologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas
16.
Neurourol Urodyn ; 36(2): 280-285, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-26536146

RESUMO

AIMS: Kv7 voltage-gated potassium channels have been suggested to modulate mechano-afferent transduction and nociception in the bladder. We investigated the effects of retigabine, a Kv7 channel activator, on rhythmic bladder contractions (RBCs), and single-unit afferent activities (SAAs) of the primary bladder mechanosensitive afferent nerve fibers in urethane-anesthetized rats. In addition, the effects of pretreatment with retigabine on the nociceptive behaviors provoked by an intravesical instillation of resiniferatoxin (RTX) were evaluated in the conscious condition. METHODS: Female Sprague-Dawley rats were used. Under urethane anesthesia, saline was instilled into the bladder until RBCs were induced reproducibly. Then, the effects of intravenous, cumulative administrations of retigabine (0.1-3 mg/kg) or vehicle (saline) on RBCs were assessed. In separate animals, SAAs of Aδ- and C-fibers were identified by electrical stimulation of the pelvic nerve and by bladder distention with saline. After baseline recording, vehicle or retigabine (0.01-1 mg/kg) was administered intravenously and further recordings were performed. Under pretreatment with vehicle or retigabine (3 mg/kg intraperitoneally), the frequencies of lower abdominal licking and freezing were counted and scored as the bladder nociceptive behaviors induced by intravesical RTX instillation (3 µM, 0.3 ml). RESULTS: Retigabine dose-dependently decreased both the frequency and the amplitude of RBCs and SAAs of both Aδ- and C-fibers. The effect on RBCs was more potent on the frequency than the amplitude. Retigabine inhibited the RTX-induced abdominal licking, but not freezing. CONCLUSION: Kv7 channels are likely to be implicated in inhibition of bladder mechano- and nociceptive sensory transduction. Neurourol. Urodynam. 36:280-285, 2017. © 2015 Wiley Periodicals, Inc.


Assuntos
Comportamento Animal/efeitos dos fármacos , Carbamatos/farmacologia , Mecanotransdução Celular/efeitos dos fármacos , Moduladores de Transporte de Membrana/farmacologia , Neurônios Aferentes/efeitos dos fármacos , Nociceptividade/efeitos dos fármacos , Fenilenodiaminas/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , Bexiga Urinária/inervação , Animais , Feminino , Ratos , Ratos Sprague-Dawley
17.
Br J Pharmacol ; 173(24): 3480-3491, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27677924

RESUMO

BACKGROUND AND PURPOSE: Kv 7 (Kv 7.1-7.5) channels play an important role in the regulation of neuronal excitability and the cardiac action potential. Growing evidence suggests Kv 7.4/Kv 7.5 channels play a crucial role in regulating vascular smooth muscle contractility. Most of the reported Kv 7 openers have shown poor selectivity across these five subtypes. In this study, fasudil - a drug used for cerebral vasospasm - has been found to be a selective opener of Kv 7.4/Kv 7.5 channels. EXPERIMENTAL APPROACH: A perforated whole-cell patch technique was used to record the currents and membrane potential. Homology modelling and a docking technique were used to investigate the interaction between fasudil and the Kv 7.4 channel. An isometric tension recording technique was used to assess the vascular tension. KEY RESULTS: Fasudil selectively and potently enhanced Kv 7.4 and Kv 7.4/Kv 7.5 currents expressed in HEK293 cells, and shifted the voltage-dependent activation curve in a more negative direction. Fasudil did not affect either Kv 7.2 and Kv 7.2/Kv 7.3 currents expressed in HEK293 cells, the native neuronal M-type K+ currents, or the resting membrane potential in small rat dorsal root ganglia neurons. The Val248 in S5 and Ile308 in S6 segment of Kv 7.4 were critical for this activating effect of fasudil. Fasudil relaxed precontracted rat small arteries in a concentration-dependent fashion; this effect was antagonized by the Kv 7 channel blocker XE991. CONCLUSIONS AND IMPLICATIONS: These results suggest that fasudil is a selective Kv 7.4/Kv 7.5 channel opener and provide a new dimension for developing selective Kv 7 modulators and a new prospective for the use, action and mechanism of fasudil.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/antagonistas & inibidores , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/química , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Animais , Antracenos/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Masculino , Estrutura Molecular , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Vasodilatadores/antagonistas & inibidores , Vasodilatadores/química
18.
J Mol Cell Cardiol ; 72: 126-37, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24631769

RESUMO

Accurate diagnosis of predisposition to long QT syndrome is crucial for reducing the risk of cardiac arrhythmias. In recent years, drug-induced provocative tests have proved useful to unmask some latent mutations linked to cardiac arrhythmias. In this study we expanded this concept by developing a prototype for a computational provocative screening test to reveal genetic predisposition to acquired long-QT syndrome (aLQTS). We developed a computational approach to reveal the pharmacological properties of IKr blocking drugs that are most likely to cause aLQTS in the setting of subtle alterations in IKr channel gating that would be expected to result from benign genetic variants. We used the model to predict the most potentially lethal combinations of kinetic anomalies and drug properties. In doing so, we also implicitly predicted ideal inverse therapeutic properties of K channel openers that would be expected to remedy a specific defect. We systematically performed "in silico mutagenesis" by altering discrete kinetic transition rates of the Fink et al. Markov model of human IKr channels, corresponding to activation, inactivation, deactivation and recovery from inactivation of IKr channels. We then screened and identified the properties of IKr blockers that caused acquired long QT and therefore unmasked mutant phenotypes for mild, moderate and severe variants. Mutant IKr channels were incorporated into the O'Hara et al. human ventricular action potential (AP) model and subjected to simulated application of a wide variety of IKr-drug interactions in order to identify the characteristics that selectively exacerbate the AP duration (APD) differences between wild-type and IKr mutated cells. Our results show that drugs with disparate affinities to conformation states of the IKr channel are key to amplify variants underlying susceptibility to acquired long QT syndrome, an effect that is especially pronounced at slow frequencies. Finally, we developed a mathematical formulation of the M54T MiRP1 latent mutation and simulated a provocative test. In this setting, application of dofetilide dramatically amplified the predicted QT interval duration in the M54T hMiRP1 mutation compared to wild-type.


Assuntos
Antiarrítmicos/efeitos adversos , Ventrículos do Coração/metabolismo , Síndrome do QT Longo/metabolismo , Modelos Estatísticos , Bloqueadores dos Canais de Potássio/efeitos adversos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , Potenciais de Ação/efeitos dos fármacos , Astemizol/efeitos adversos , Cisaprida/efeitos adversos , Simulação por Computador , Expressão Gênica , Predisposição Genética para Doença , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/patologia , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Cinética , Síndrome do QT Longo/induzido quimicamente , Síndrome do QT Longo/genética , Síndrome do QT Longo/patologia , Mutação , Fenetilaminas/efeitos adversos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Conformação Proteica , Índice de Gravidade de Doença , Sotalol/efeitos adversos , Sulfonamidas/efeitos adversos , Terfenadina/efeitos adversos
19.
Fiziol Zh (1994) ; 59(3): 18-24, 2013.
Artigo em Russo | MEDLINE | ID: mdl-23957160

RESUMO

Carbon monoxide (CO) is one of a family of gas transmitters. In this article we present the results of mechanographic investigations of the mechanisms of CO action on a rat thoracic aorta segments. We found that relaxing effect of CO donor CORM-2 on vascular smooth muscles is mediated mainly by opening of voltage-dependent potassium channels in smooth muscle cells: 4-aminopyridine, blocking these channels, almost completely eliminated the CO-induced vasorelaxation of the segments precontracted by depolarization of the smooth muscle cells membranes with high potassium (30 mM KCl) solution or by phenylephrine (10 microM). For the first time we documented that CORM-2 reduces the nicardipine-sensitive input of 45Ca2+ in freshly isolated aorta cells. There are reasons to suggest that the L-type voltage-dependent calcium channels of vascular smooth muscle cells are another target for CO, which is implemented in the relaxing effect of this gas transmitter. Additional research is needed to determine the influence of ruthenium complexes (Ru(II)) on phenomenology of carbon monoxide effects.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Cálcio/metabolismo , Monóxido de Carbono/farmacologia , Contração Muscular/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Animais , Aorta/efeitos dos fármacos , Aorta/fisiologia , Radioisótopos de Cálcio , Monóxido de Carbono/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Masculino , Contração Muscular/fisiologia , Músculo Liso Vascular/fisiologia , Compostos Organometálicos/administração & dosagem , Compostos Organometálicos/metabolismo , Fenilefrina/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Cloreto de Potássio/farmacologia , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Técnicas de Cultura de Tecidos , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia
20.
Contraception ; 88(4): 485-91, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23845210

RESUMO

There is a global need for an ideal method of male contraception. However, the development of male contraceptives has not been well successful. Research on sperm-specific ion channels, especially the recent advance obtained from electrophysiological studies, has emphasized the conception that those channels are targets with the most potential to develop non-hormonal male contraceptives. While summarizing the general options for male contraception, this review focuses on the properties and functions of sperm ion channels together with the attempts of utilizing these channels to develop male contraceptives. We believe that a deeper insight into the signaling and molecular mechanisms by which ion channels regulate sperm functions will pave the way for developing novel male-based contraceptives.


Assuntos
Anticoncepcionais Masculinos/farmacologia , Drogas em Investigação/farmacologia , Canais Iônicos/antagonistas & inibidores , Espermatozoides/efeitos dos fármacos , Anoctamina-1 , Canais de Cálcio/química , Canais de Cálcio/metabolismo , Agonistas dos Canais de Cloreto , Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/metabolismo , Humanos , Canais Iônicos/agonistas , Canais Iônicos/metabolismo , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta , Canais de Potássio Ativados por Cálcio de Condutância Alta , Masculino , Proteínas de Membrana/agonistas , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Especificidade de Órgãos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Subunidades Proteicas/agonistas , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Espermatozoides/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA