Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Sci ; 115(7): 2461-2472, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38655663

RESUMEN

L-type amino acid transporter 1 (LAT1) is specifically expressed in many malignancies, contributes to the transport of essential amino acids, such as leucine, and regulates the mammalian target of rapamycin (mTOR) signaling pathway. We investigated the expression profile and functional role of LAT1 in prostate cancer using JPH203, a specific inhibitor of LAT1. LAT1 was highly expressed in castration-resistant prostate cancer (CRPC) cells, including C4-2 and PC-3 cells, but its expression level was low in castration-sensitive LNCaP cells. JPH203 significantly inhibited [14C] leucine uptake in CRPC cells but had no effect in LNCaP cells. JPH203 inhibited the proliferation, migration, and invasion of CRPC cells but not of LNCaP cells. In C4-2 cells, Cluster of differentiation (CD) 24 was identified by RNA sequencing as a novel downstream target of JPH203. CD24 was downregulated in a JPH203 concentration-dependent manner and suppressed activation of the Wnt/ß-catenin signaling pathway. Furthermore, an in vivo study showed that JPH203 inhibited the proliferation of C4-2 cells in a castration environment. The results of this study indicate that JPH203 may exert its antitumor effect in CRPC cells via mTOR and CD24.


Asunto(s)
Antígeno CD24 , Movimiento Celular , Proliferación Celular , Transportador de Aminoácidos Neutros Grandes 1 , Neoplasias de la Próstata Resistentes a la Castración , Masculino , Humanos , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Línea Celular Tumoral , Animales , Proliferación Celular/efectos de los fármacos , Antígeno CD24/metabolismo , Ratones , Movimiento Celular/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Serina-Treonina Quinasas TOR/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Benzoxazoles/farmacología , Leucina/farmacología , Leucina/análogos & derivados , Ratones Desnudos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Tirosina/análogos & derivados
2.
Brain Res Bull ; 204: 110788, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37844783

RESUMEN

Xenin is a 25-amino acid peptide identified in human gastric mucosa, which is widely expressed in peripheral and central tissues. It is known that the central or peripheral administration of xenin decreases food intake in rodents. Nesfatin-1/NUCB2 (nesfatin-1) has been identified as an anorexic neuropeptide, it is often found co-localized with many peptides in the central nervous system. After the intracerebroventricular administration of xenin on nesfain-1-like immunoreactivity (LI) neurons, we examined its effects on food intake and water intake in rats. As a result, Fos-LI neurons were observed in the organum vasculosum of the laminae terminalis (OVLT), the median preoptic nucleus (MnPO), the subfornical organ (SFO), the supraoptic nucleus (SON), the paraventricular nucleus (PVN), the arcuate nucleus (Arc), the lateral hypothalamic area (LHA), the central amygdaloid nucleus (CAN), the dorsal raphe nucleus (DR), the locus coeruleus (LC), the area postrema (AP) and the nucleus of the solitary tract (NTS). After the administration, the number of Fos-LI neurons was significantly increased in the LC and the OVLT, the MnPO, the SFO, the SON, the PVN, the Arc, the LHA, the CAN, the DR, the AP and the NTS, compared with the control group. After the administration of xenin, we conducted double immunohistochemistry for Fos and nesfatin-1, and found that the number of nesfatin-1-LI neurons expressing Fos were significantly increased in the SON, the PVN, the Arc, the LHA, the CAN, the DR, the AP and the NTS, compared with the control group. The pretreatment of nesfatin-1 antisense significantly attenuated this xenin-induced feeding suppression, while that of nesfatin-1 missense showed no improvement. These results indicate that central administered xenin may have anorexia effects associated with activated central nesfatin-1 neurons.


Asunto(s)
Proteínas de Unión al Calcio , Proteínas de Unión al ADN , Humanos , Ratas , Animales , Proteínas de Unión al ADN/metabolismo , Nucleobindinas/metabolismo , Nucleobindinas/farmacología , Proteínas de Unión al Calcio/metabolismo , Neuronas/metabolismo
3.
J Pharmacol Sci ; 150(4): 201-203, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36344041

RESUMEN

Currently, the emergence of drug resistance is an important issue in the treatment of hepatitis B virus (HBV). Recently, our collaborating group developed a novel long-acting anti-HBV drug, E-CFCP. However, until this study, the effects of E-CFCP in the kidney have remained unclarified. Using cell viability and uptake assays, we examined the effects of E-CFCP on the function of renal organic anion transporters (OATs). No cytotoxicity was shown related to the E-CFCP in the renal OATs in either assay. Thus, this study suggested that E-CFCP may be a novel, excellent candidate drug for the treatment of drug-resistant HBV.


Asunto(s)
Hepatitis B , Transportadores de Anión Orgánico , Humanos , Hepatitis B/tratamiento farmacológico , Virus de la Hepatitis B , Riñón , Proteínas de Transporte de Membrana , Antivirales/farmacología , Antivirales/uso terapéutico , Farmacorresistencia Viral
4.
J Pharmacol Sci ; 150(4): 251-258, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36344047

RESUMEN

Amino acid transporters are responsible for the uptake of amino acids, critical for cell proliferation. L-type amino acid transporters play a major role in the uptake of essential amino acids. L-type amino acid transporter 1 (LAT1) exerts its functional properties by forming a dimer with 4F2hc. Utilizing this cancer-specificity, research on diagnostic imaging and therapeutic agents for malignant tumors targeting LAT1 progresses in various fields. In hormone-sensitive prostate cancer, the up-regulation of L-type amino acid transporter 3 (LAT3) through the androgen receptor (AR) has been identified. On the other hand, in castration-resistant prostate cancer, the negative regulation of LAT1 through AR has been determined. Furthermore, 4F2hc: a binding partner of LAT1, was identified as the specific downstream target of Androgen Receptor Splice Variant 7: AR-V7. LAT1 has been suggested to contribute to acquiring castration resistance in prostate cancer, making LAT1 a completely different therapeutic target from anti-androgens and taxanes. Increased expression of LAT1 has also been found in renal and bladder cancers, suggesting a contribution to acquiring malignancy and progression. In Japan, clinical trials of LAT1 inhibitors for solid tumors are in progress, and clinical applications are now underway. This article will summarize the relationship between LAT1 and urological malignancies.


Asunto(s)
Transportador de Aminoácidos Neutros Grandes 1 , Neoplasias de la Próstata , Neoplasias Urológicas , Humanos , Masculino , Sistemas de Transporte de Aminoácidos , Transportador de Aminoácidos Neutros Grandes 1/genética , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Receptores Androgénicos/genética , Neoplasias Urológicas/tratamiento farmacológico , Neoplasias Urológicas/genética
5.
J Pharmacol Sci ; 146(2): 82-87, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33941324

RESUMEN

Islatravir (ISL; 4'-ethynyl-2-fluoro-2'-deoxyadenosine or EFdA) is a novel reverse transcriptase translocation inhibitor and has a unique structure and high antiviral activity against wild-type and multidrug resistant HIV strains. In this study, we investigated whether islatravir (ISL) can cause kidney damage compared to tenofovir disoproxil fumarate (TDF) and tenofovir (TFV). We also investigated interactions of these drugs with organic anion transporters (OATs). There is a large gap in ISL concentration between the pharmacological dose to proximal tubular cells and the clinical dose. ISL is unlikely to be taken up via OAT1 or OAT3; therefore, OAT1 and OAT3 may not be involved in the injury to tubular cells. Present data strongly suggests that ISL is not toxic to proximal tubules because blood levels of ISL are not high enough to cause kidney damage in the clinical setting.


Asunto(s)
Desoxiadenosinas/efectos adversos , Desoxiadenosinas/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Transportadores de Anión Orgánico/metabolismo , Inhibidores de la Transcriptasa Inversa/efectos adversos , Inhibidores de la Transcriptasa Inversa/metabolismo , Lesión Renal Aguda/etiología , Células Cultivadas , Desoxiadenosinas/sangre , Relación Dosis-Respuesta a Droga , Humanos
6.
J Physiol Sci ; 71(1): 11, 2021 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-33789576

RESUMEN

L-type amino acid transporter 2 (LAT2) is a Na+-independent neutral amino acid transporter, whose function regulation system remains unclarified. Since protein kinase C (PKC) is known to regulate the functions of various transporters, we investigated whether human LAT2 (hLAT2) function is regulated by PKC. In mouse proximal tubule S2 cells, hLAT2 transport activity was upregulated by PKC activation. However, we found that the mRNA and protein expression of hLAT2 was not affected by PKC activation and that the upregulation was independent of the three potential PKC consensus sites in the hLAT2 amino acid sequence. Moreover, we found that PKC activation upregulated the Vmax value for hLAT2-mediated alanine transport, which was not accompanied by the induction of hLAT2 membrane insertion. In conclusion, we showed that hLAT2 function is upregulated by PKC activation, which is not related to either the de novo synthesis, the phosphorylation or the membrane insertion of hLAT2.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/metabolismo , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/metabolismo , Proteína Quinasa C/metabolismo , Sistema de Transporte de Aminoácidos y+/genética , Animales , Línea Celular , Supervivencia Celular , Clonación Molecular , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Indoles/farmacología , Maleimidas/farmacología , Ratones , Mutagénesis Sitio-Dirigida , Regulación hacia Arriba
7.
J Pharmacol Sci ; 144(1): 16-22, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32653341

RESUMEN

JPH203 is a novel anti-cancer drug targeting L-type amino acid transporter 1 (LAT1), which plays a primary role in the uptake of essential amino acids in tumor cells. Although a co-incubation inhibitory effect of JPH203 has been shown in a conventional uptake assay, its preincubation inhibitory effects have remained undetermined. Therefore, we aimed to characterize the preincubation inhibitory effects of JPH203 on LAT1 function using leucine uptake assays in LAT1-positive human colon cancer HT-29 cells. Preincubation of the cells with JPH203 (0.3 µM for 120 min) decreased the activity level to 30% of that in dimethylsulfoxide-treated cells. Similarly, in time-dependency analysis, preincubation of HT-29 cells with 10 µM JPH203 for 30, 60, and 120 min decreased the leucine uptake activity (42%, 32%, and 28% of that in control cells, respectively). Furthermore, the IC50 value of the combination of preincubation and co-incubation effects was lower than that of co-incubation inhibition alone (34.2 ± 3.6 nM vs. 99.2 ± 11.0 nM). In conclusion, we revealed that JPH203 has the capability to inhibit LAT1 function through preincubation effects. Moreover, preincubation synergistically enhances the co-incubation inhibitory effects. These findings provide a novel insight into the anti-cancer effects of JPH203 in cancer therapy.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Antineoplásicos/farmacología , Benzoxazoles/farmacología , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Ensayos de Selección de Medicamentos Antitumorales/métodos , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Tirosina/análogos & derivados , Relación Dosis-Respuesta a Droga , Células HT29 , Humanos , Transportador de Aminoácidos Neutros Grandes 1/fisiología , Leucina/metabolismo , Factores de Tiempo , Tirosina/farmacología
8.
Sci Rep ; 9(1): 16776, 2019 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-31748583

RESUMEN

Large neutral amino acid transporter 1 (LAT1, SLC7A5) is abundantly expressed in various types of cancer, and it has been thought to assist cancer progression through its activity for uptake of neutral amino acids. However, the roles of LAT1 in renal cell carcinoma (RCC) prognosis and treatment remain uncharacterized. Therefore, we first retrospectively examined the LAT1 expression profile and its associations with clinical factors in RCC tissues (n = 92). The results of immunohistochemistry showed that most of the tissues examined (92%) had cancer-associated LAT1 expression. Furthermore, the overall survival (OS) and progression-free survival (PFS) were shorter in patients with high LAT1 expression levels than in those with low LAT1 expression levels (P = 0.018 and 0.014, respectively), and these associations were further strengthened by the results of univariate and multivariate analyses. Next, we tested the effects of JPH203, which is a selective LAT1 inhibitor, on RCC-derived Caki-1 and ACHN cells. It was found that JPH203 inhibited the growth of these cell types in a dose-dependent manner. Moreover, JPH203 clearly suppressed their migration and invasion activities. Thus, our results show that LAT1 has a great potential to become not only a prognosis biomarker but also a therapeutic target in RCC clinical settings.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Neoplasias Renales/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Benzoxazoles/farmacología , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/patología , Masculino , Persona de Mediana Edad , Pronóstico , Supervivencia sin Progresión , Estudios Retrospectivos , Tirosina/análogos & derivados , Tirosina/farmacología
9.
J Physiol Sci ; 69(4): 635-642, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31098988

RESUMEN

Sodium-coupled monocarboxylate transporter SMCT1 (SLC5A8) mediates monocarboxylate transport in the proximal tubule of the kidney. We have identified PDZK1 and PDZ domain-containing RING finger 3 (PDZRN3) as potent binding partners of SMCT1, which has a PDZ motif (Thr-Arg-Leu), by yeast two-hybrid screening and revealed that PDZK1 enhances the transport activity of SMCT1. In this study, we aimed to characterize the interaction between SMCT1 and PDZRN3 as well as to examine how PDZRN3 regulates SMCT1 function. An interaction between SMCT1 and PDZRN3 through the PDZ motif was observed in a co-immunoprecipitation assay and yeast two-hybrid assay. A transport assay showed that PDZRN3 abolished the enhancing effect of PDZK1 on nicotinate uptake via SMCT1. Our results suggest that SMCT1 interacts with PDZRN3 and that PDZRN3 may regulate SMCT1 function by interfering with the interaction between SMCT1 and PDZK1.


Asunto(s)
Proteínas Portadoras/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Transporte Biológico/fisiología , Línea Celular , Células HEK293 , Humanos , Dominios y Motivos de Interacción de Proteínas/fisiología , Sodio , Ubiquitina-Proteína Ligasas
10.
Anticancer Res ; 39(1): 159-165, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30591453

RESUMEN

BACKGROUND/AIM: L-type amino acid transporter 1 (LAT1) is a promising molecular target for cancer therapy. The present study aimed to characterize the anti-cancer effects of JPH203, an LAT1-selective inhibitor, on gastrointestinal cancer cells. MATERIALS AND METHODS: Three esophageal, two gastric, and two colon cancer cell lines were used. Cytotoxic effects of JPH203 were assessed by a WST-8 assay. LAT1 mRNA levels were determined by quantitative PCR. The inhibitory property of JPH203 against LAT1 function was examined by a transport assay. RESULTS: JPH203 treatment significantly reduced the viability of all gastric and colon cancer cells. While LAT1 expression levels and inhibitory potencies of JPH203 on LAT1 functions were comparable among the cells, all the esophageal cells were resistant to JPH203. CONCLUSION: JPH203 was effective in reducing gastric and colon cancer cells. To clarify its cell type-dependent efficacy, identification of the causal factors for JPH203 resistance will be needed.


Asunto(s)
Benzoxazoles/farmacología , Neoplasias del Colon/tratamiento farmacológico , Neoplasias Gastrointestinales/tratamiento farmacológico , Transportador de Aminoácidos Neutros Grandes 1/genética , Tirosina/análogos & derivados , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Tirosina/farmacología
11.
PLoS One ; 11(10): e0164785, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27764176

RESUMEN

Insulin secretagogues are used for treatment of type 2 diabetes. We attempted to discover novel small molecules to stimulate insulin secretion by using in silico similarity search using sulfonylureas as query, followed by measurement of insulin secretion. Among 38 compounds selected by in silico similarity search, we found three diphenylsemicarbazides and one quinolone that stimulate insulin secretion. We focused on compound 8 (C8), which had the strongest insulin-secreting effect. Based on the structure-activity relationship of C8-derivatives, we identified diphenylthiosemicarbazide (DSC) 108 as the most potent secretagogue. DSC108 increased the intracellular Ca2+ level in MIN6-K8 cells. Competitive inhibition experiment and electrophysiological analysis revealed sulfonylurea receptor 1 (SUR1) to be the target of DSC108 and that this diphenylthiosemicarbazide directly inhibits ATP-sensitive K+ (KATP) channels. Pharmacokinetic analysis showed that DSC108 has a short half-life in vivo. Oral administration of DSC108 significantly suppressed the rises in blood glucose levels after glucose load in wild-type mice and improved glucose tolerance in the Goto-Kakizaki (GK) rat, a model of type 2 diabetes with impaired insulin secretion. Our data indicate that DSC108 is a novel insulin secretagogue, and is a lead compound for development of a new anti-diabetic agent.


Asunto(s)
Aminoácidos Cíclicos/farmacología , Hipoglucemiantes/farmacología , Insulina/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Semicarbacidas/farmacología , Tiosemicarbazonas/farmacología , Administración Oral , Aminoácidos Cíclicos/química , Aminoácidos Cíclicos/metabolismo , Aminoácidos Cíclicos/uso terapéutico , Animales , Glucemia/análisis , Calcio/metabolismo , Línea Celular , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/patología , Prueba de Tolerancia a la Glucosa , Gliburida/farmacología , Gliburida/uso terapéutico , Semivida , Hipoglucemiantes/química , Hipoglucemiantes/metabolismo , Hipoglucemiantes/uso terapéutico , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Canales KATP/antagonistas & inhibidores , Canales KATP/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Ratas , Semicarbacidas/sangre , Semicarbacidas/química , Semicarbacidas/metabolismo , Relación Estructura-Actividad , Compuestos de Sulfonilurea/química , Compuestos de Sulfonilurea/metabolismo , Compuestos de Sulfonilurea/farmacología , Receptores de Sulfonilureas/antagonistas & inhibidores , Receptores de Sulfonilureas/metabolismo , Tiosemicarbazonas/química , Tiosemicarbazonas/metabolismo , Tiosemicarbazonas/uso terapéutico
12.
J Pharmacol Sci ; 127(3): 370-6, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25837936

RESUMEN

Although it has been reported that endotoxin-induced expression of Nox1 in the heart contributes to apoptosis in cardiomyocytes, functional role of Nox1 at the physiological expression level has not been elucidated. The aim of this study was to clarify the role of Nox1 under a hypoxic condition using wild-type (WT, Nox1(+/Y)) and Nox1-deficient (Nox1(-/Y)) mice. ECG recordings from anesthetized mice revealed that Nox1(-/Y) mice were more sensitive to hypoxia, resulting in bradycardia, compared to WT mice. Atrial and ventricular electrocardiograms recorded from Langendorff-perfused hearts revealed that hypoxic perfusion more rapidly decreased heart rate in Nox1(-/Y) hearts compared with WT hearts. Sinus node recovery times measured under a hypoxic condition were prolonged more markedly in the Nox1(-/Y) hearts. Sinoatrial node dysfunction of Nox1(-/Y) hearts during hypoxia was ameriolated by the pre-treatment with the Ca(2+) channel blocker nifedipine or the K(+) channel opener pinacidil. Spontaneous action potentials were recorded from enzymatically-isolated sinoatrial node (SAN) cells under a hypoxic condition. There was no significant difference in the elapsed times from the commencement of hypoxia to asystole between WT and Nox1(-/Y) SAN cells. These findings suggest that Nox1 may have a protective effect against hypoxia-induced SAN dysfunction.


Asunto(s)
Bradicardia/etiología , Bradicardia/prevención & control , Hipoxia/complicaciones , NADH NADPH Oxidorreductasas/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Bradicardia/fisiopatología , Bloqueadores de los Canales de Calcio/farmacología , Bloqueadores de los Canales de Calcio/uso terapéutico , Modelos Animales de Enfermedad , Electrocardiografía , Frecuencia Cardíaca , Hipoxia/fisiopatología , Técnicas In Vitro , Moduladores del Transporte de Membrana/farmacología , Moduladores del Transporte de Membrana/uso terapéutico , Ratones Endogámicos C57BL , Ratones Transgénicos , NADPH Oxidasa 1 , Nifedipino/farmacología , Nifedipino/uso terapéutico , Pinacidilo/farmacología , Pinacidilo/uso terapéutico , Bloqueo Sinoatrial/etiología , Bloqueo Sinoatrial/fisiopatología , Bloqueo Sinoatrial/prevención & control , Nodo Sinoatrial/citología , Nodo Sinoatrial/fisiopatología
13.
J Pharmacol Sci ; 125(4): 406-14, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25030741

RESUMEN

The acetylcholine receptor-operated K(+) (KACh) channel may be a novel target for atrial-specific antiarrhythmic therapy. Recently it has been demonstrated that tertiapin, a selective blocker of KACh channel, suppressed aconitine-induced atrial fibrillation (AF) in dogs. However, the precise mechanism by which the KACh-channel blocker inhibits the aconitine-induced AF remains unknown. This study was undertaken to determine the role of KACh channel in aconitine-induced AF in guinea pigs. Tertiapin terminated the aconitine-induced AF in anesthetized guinea pigs. The results of an in vitro electrophysiological experiment using atrial cells and atrial preparations suggest that aconitine might activate KACh channels in atrial cells, probably by intracellular Na(+) accumulation, and inhibition of KACh channels by tertiapin might suppress AF by producing conduction block, probably due to further decrease in the resting membrane potential. Since it has been reported that constitutively active KACh channels can be observed in atrial cells of patients with chronic AF, aconitine-induced AF may be used as an experimental model for evaluation of drug effect on chronic AF.


Asunto(s)
Aconitina/efectos adversos , Fibrilación Atrial/inducido químicamente , Fibrilación Atrial/tratamiento farmacológico , Venenos de Abeja/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Animales , Venenos de Abeja/uso terapéutico , Células Cultivadas , Enfermedad Crónica , Modelos Animales de Enfermedad , Perros , Electrocardiografía , Fenómenos Electrofisiológicos/efectos de los fármacos , Cobayas , Atrios Cardíacos/citología , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/metabolismo , Técnicas In Vitro , Potenciales de la Membrana/efectos de los fármacos , Terapia Molecular Dirigida , Bloqueadores de los Canales de Potasio/uso terapéutico , Canales de Potasio/metabolismo , Canales de Potasio/fisiología , Sodio/metabolismo
14.
J Pharmacol Sci ; 112(4): 415-23, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20308804

RESUMEN

In guinea-pig cardiomyocytes, a cAMP-dependent Cl(-) current (I(Cl,cAMP)) flows through a cardiac isoform of the cystic fibrosis transmembrane conductance regulator (CFTR), which belongs to a family of the ATP-binding cassette (ABC) proteins. Although several K(+)-channel openers and sulfonylurea ATP-sensitive K(+) (K(ATP))-channel blockers reportedly inhibit I(Cl,cAMP), effects of nicorandil on the Cl(-) current have not been evaluated. This study was conducted to examine the effects of nicorandil on I(Cl,cAMP) in isolated guinea-pig ventricular cells using patch clamp techniques. Nicorandil in concentrations higher than 300 microM enhanced the I(Cl,cAMP) preactivated by 0.1 microM isoproterenol. The isoproterenol-induced I(Cl,cAMP) was inhibited by 100 microM glibenclamide, but not by 100 microM pinacidil. SNAP (S-nitroso-N-acetyl-D,L-penicillamine, 10 microM), a nitric oxide (NO) donor, similarly enhanced the isoproterenol-induced I(Cl,cAMP). However, SG-86, a denitrated metabolite possessing K(+ )channel-opening action, failed to enhance the Cl(-) current. When the I(Cl,cAMP) was activated by 3-isobutyl-1-methylxanthine (IBMX, 30 microM), either nicorandil or SNAP failed to enhance the isoproterenol-induced I(Cl,cAMP). Thus, nicorandil enhances I(Cl,cAMP) in guinea-pig cardiomyocytes through an increase in intracellular cGMP, although direct modulation of I(Cl,cAMP) by NO cannot be completely excluded.


Asunto(s)
Antihipertensivos/farmacología , Canales de Cloruro/efectos de los fármacos , AMP Cíclico/metabolismo , Ventrículos Cardíacos/efectos de los fármacos , Nicorandil/farmacología , Antagonistas Adrenérgicos beta/farmacología , Animales , Canales de Cloruro/metabolismo , Gliburida/farmacología , Cobayas , Ventrículos Cardíacos/citología , Hipoglucemiantes/farmacología , Isoproterenol/farmacología , Donantes de Óxido Nítrico/farmacología , Técnicas de Placa-Clamp , Pinacidilo/farmacología , S-Nitroso-N-Acetilpenicilamina/farmacología
15.
J Pharmacol Sci ; 110(2): 150-9, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19498275

RESUMEN

After the report of the Cardiac Arrhythmia Suppression Trial, a tabular framework of the Sicilian Gambit has been proposed to display actions of antiarrhythmic drugs on ion channels and receptors and to provide more rational pharmacotherapy of arrhythmias. However, because effects of antiarrhythmic drugs on If have not been thoroughly examined, we used patch clamp techniques to determine the effects of various antiarrhythmic drugs on the HCN (hyperpolarization-activated cyclic nucleotide-gated) channel currents. HCN4 channels, a dominant isoform of HCN channels in the heart, were expressed in HEK293 cells. Amiodarone and bepridil potently inhibited the HCN4 channel current with IC50 values of 4.5 and 4.9 microM, respectively, which were close to their therapeutic concentrations. The inhibitory effects of quinidine, disopyramide, cibenzoline, lidocaine, mexiletine, aprindine, propafenone, flecainide, propranolol, and verapamil on the HCN4 channel current were weak in their therapeutic concentrations, with IC50 values of 78.3, 249, 46.8, 276, 309, 43.7, 14.3, 1700, 50.5, and 44.9 microM, respectively, suggesting that the inhibitory effects on If would be clinically small. D,L-Sotalol hardly affected the HCN4 channel current. Information about the HCN4-channel effects of many antiarrhythmic drugs may be useful for determining the appropriate drug for treatment of various arrhythmias while minimizing adverse effects.


Asunto(s)
Antiarrítmicos/farmacología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/efectos de los fármacos , Antiarrítmicos/administración & dosificación , Línea Celular , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Electrofisiología , Humanos , Concentración 50 Inhibidora , Riñón/citología , Riñón/efectos de los fármacos , Riñón/metabolismo , Técnicas de Placa-Clamp
16.
J Pharmacol Sci ; 105(3): 229-39, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17965539

RESUMEN

Effects of azimilide, a class III antiarrhythmic drug, on the acetylcholine (ACh) receptor-operated K+ current (I K.ACh) and the delayed rectifier K+ current (IK) were examined in guinea-pig atrial cells using patch-clamp techniques. Effects of azimilide on experimental atrial fibrillation (AF) were also examined in isolated guinea-pig hearts. In single atrial myocytes, azimilide inhibited both the rapid (IKr) and slow component of IK (IKs). Azimilide inhibited the I K.ACh induced by carbachol (CCh, 1 microM), adenosine (10 microM), and intracellular loading of GTPgammaS (100 microM) in a concentration-dependent manner. The IC50 values of azimilide for inhibiting the CCh-, adenosine-, and GTPgammaS-induced I K.ACh were 1.25, 29.1, and 20.9 microM, respectively, suggesting that azimilide inhibits I K.ACh mainly by blocking the muscarinic receptors. Azimilide concentration-dependently (0.3 - 10 microM) prolonged the action potential duration (APD) in the absence and presence of muscarinic stimulation. In isolated hearts, perfusion of CCh shortened the duration of the monophasic action potential (MAP) and effective refractory period (ERP) of the left atrium and lowered the atrial fibrillation threshold (AFT). Addition of azimilide inhibited the induction of AF by prolonging the duration of MAP and ERP. The I K.ACh inhibition by azimilide may at least in part contribute to the effectiveness to prevent parasympathetic-type AF.


Asunto(s)
Fibrilación Atrial/prevención & control , Canales de Potasio de Tipo Rectificador Tardío/antagonistas & inhibidores , Imidazolidinas/farmacología , Antagonistas Muscarínicos/farmacología , Piperazinas/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Carbacol/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Guanosina 5'-O-(3-Tiotrifosfato)/farmacología , Cobayas , Atrios Cardíacos/efectos de los fármacos , Hidantoínas , Imidazolidinas/uso terapéutico , Técnicas In Vitro , Técnicas de Placa-Clamp , Piperazinas/uso terapéutico , Periodo Refractario Electrofisiológico/efectos de los fármacos
17.
Circulation ; 106(5): 613-9, 2002 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-12147545

RESUMEN

BACKGROUND: The endothelin (ET) system is activated in failing hearts. Congestive heart failure frequently is associated with ventricular arrhythmias, which may result from electrical remodeling such as changes of ionic current density and heterogeneous action potential prolongation. We examined the effects of long-term ET(A) receptor blockade on the electrophysiological properties of ventricular cells, the surface ECG, and the survival in BIO 14.6 cardiomyopathic hamsters. METHODS AND RESULTS: Membrane currents and action potentials were recorded from left ventricular cells isolated from normal F1beta hamsters and cardiomyopathic BIO 14.6 hamsters untreated and chronically treated with TA-0201, an ET(A) receptor antagonist. In ventricular cells of untreated BIO 14.6 hamsters, the action potential duration was prolonged and the densities of the L-type Ca2+ current (I(Ca,L)), the transient outward current (I(to)), the delayed rectifier K+ current (I(K)), and the inward rectifier K+ current (I(K1)) were decreased compared with those of F1beta hamsters. Long-term treatment with the ET(A) receptor antagonist significantly attenuated action potential duration prolongation and reduction of I(to), I(K), and I(Ca,L) in BIO 14.6 ventricular cells. Long-term ET(A) receptor blockade prevented the QT prolongation and ventricular arrhythmias and improved the survival rate in the cardiomyopathic hamsters. CONCLUSIONS: Long-term treatment with an ET(A) antagonist inhibits electrical remodeling such as downregulation of K+ and Ca2+ currents, action potential prolongation, and the increased QT interval and thereby suppresses ventricular arrhythmias in cardiomyopathic hearts. ET(A) receptor blockade may provide a new strategy for the prevention of ventricular arrhythmias associated with heart failure.


Asunto(s)
Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/fisiopatología , Antagonistas de los Receptores de Endotelina , Sistema de Conducción Cardíaco/efectos de los fármacos , Pirimidinas/uso terapéutico , Sulfonamidas/uso terapéutico , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Calcio/metabolismo , Canales de Calcio Tipo L/metabolismo , Enfermedad Crónica , Cricetinae , Modelos Animales de Enfermedad , Electrocardiografía/efectos de los fármacos , Técnicas Electrofisiológicas Cardíacas , Sistema de Conducción Cardíaco/fisiopatología , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/fisiopatología , Masculino , Miocardio/citología , Técnicas de Placa-Clamp , Canales de Potasio de Rectificación Interna/metabolismo , Receptor de Endotelina A , Tasa de Supervivencia , Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...