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










Base de dados
Intervalo de ano de publicação
1.
Cell Biol Int ; 47(4): 787-795, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36626273

RESUMO

Tribbles homolog 2 (TRIB2) functions as an adapter protein that regulates signal transductions involved in a variety of cellular functions, including tumorigenesis. However, the role of TRIB2 in the proliferation of vascular smooth muscle cells (VSMCs) and the underlying expression mechanisms remain unclear. The present study investigated the role of TRIB2 in VSMC proliferation and revealed that TRIB2 expression increases following vascular injury and platelet-derived growth factor (PDGF)-BB-stimulated VSMCs. We found that pretreatment with diphenyleneiodonium (a nicotinamide adenine dinucleotide phosphate oxidase inhibitor), U0126 (an inhibitor of mitogen-activated protein kinase kinase 1 [MEK1]), or siRNA targeting the gene encoding early growth response 1 (EGR-1) significantly inhibits PDGF-BB-induced TRIB2 expression in VSMCs. Furthermore, TRIB2 knockdown significantly inhibits PDGF-BB-induced proliferation of VSMCs but does not affect the phosphorylation of AKT. However, phosphorylation of ERK1 and expression of proliferating cell nuclear antibody are significantly suppressed in VSMCs by PDGF-BB stimulation. Thus, PDGF-BB-induced TRIB2 expression is mediated by ROS/ERK/EGR-1 pathways and plays a critical role in VSMC proliferation via modulation of ERK activity. We propose TRIB2 as a promising therapeutic target for the prevention of neointima formation and vascular disease.


Assuntos
Proliferação de Células , Músculo Liso Vascular , Transdução de Sinais , Animais , Ratos , Becaplermina/metabolismo , Becaplermina/farmacologia , Movimento Celular , Proliferação de Células/genética , Células Cultivadas , Sistema de Sinalização das MAP Quinases , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo
2.
Front Vet Sci ; 8: 647189, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33644160

RESUMO

Feline infectious peritonitis (FIP) is a life-threatening infectious disease of cats caused by virulent feline coronavirus (FIP virus: FIPV). For the treatment of FIP, several effective antivirals were recently reported, but many of these are not available for practical use. 5-amino levulinic acid (5-ALA) is a low-molecular-weight amino acid synthesized in plant and animal cells. 5-ALA can be synthesized in a large amount, and it is widely applied in the medical and agricultural fields. We hypothesized that 5-ALA inhibits FIPV infection. Therefore, we evaluated its antiviral activity against FIPV in felis catus whole fetus-4 cells and feline primary macrophages. FIPV infection was significantly inhibited by 250 µM 5-ALA. Our study suggested that 5-ALA is applicable for the treatment and prevention of FIPV infection.

3.
Viruses ; 12(5)2020 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-32456286

RESUMO

Feline infectious peritonitis (FIP) is a viral disease with a high morbidity and mortality by the FIP virus (FIPV, virulent feline coronavirus). Several antiviral drugs for FIP have been identified, but many of these are expensive and not available in veterinary medicine. Hydroxychloroquine (HCQ) is a drug approved by several countries to treat malaria and immune-mediated diseases in humans, and its antiviral effects on other viral infections (e.g., SARS-CoV-2, dengue virus) have been confirmed. We investigated whether HCQ in association with interferon-ω (IFN-ω) is effective for FIPV in vitro. A total of 100 µM of HCQ significantly inhibited the replication of types I and II FIPV. Interestingly, the combination of 100 µM of HCQ and 104 U/mL of recombinant feline IFN-ω (rfIFN-ω, veterinary registered drug) increased its antiviral activity against type I FIPV infection. Our study suggested that HCQ and rfIFN-ω are applicable for treatment of FIP. Further clinical studies are needed to verify the combination of HCQ and rIFN-ω will be effective and safe treatment for cats with FIP.


Assuntos
Antivirais/farmacologia , Coronavirus Felino/efeitos dos fármacos , Hidroxicloroquina/farmacologia , Interferon Tipo I/farmacologia , Análise de Variância , Animais , Antivirais/uso terapêutico , Antivirais/toxicidade , Gatos , Linhagem Celular/efeitos dos fármacos , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/virologia , Coronavirus Felino/patogenicidade , Combinação de Medicamentos , Peritonite Infecciosa Felina/tratamento farmacológico , Peritonite Infecciosa Felina/virologia , Imunofluorescência/veterinária , Hidroxicloroquina/uso terapêutico , Hidroxicloroquina/toxicidade , Interferon Tipo I/uso terapêutico , Interferon Tipo I/toxicidade , Virulência
4.
Eur J Pharmacol ; 872: 172924, 2020 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-31958455

RESUMO

Brain and muscle Arnt-like protein-1 (BMAL1), a component of the molecular clock, is implicated in the development of cardiovascular diseases, including atherosclerosis and abdominal aortic aneurysms. However, the role of BMAL1 in vascular proliferation associated with vascular remodeling is unknown. In the present study, we investigated the mechanisms underlying BMAL1 expression in vascular smooth muscle cells (VSMCs) and the role of BMAL1 in VSMC proliferation. BMAL1 expression significantly increased in injured carotid arteries in C57BL/6J mice and platelet-derived growth factor (PDGF)-BB-stimulated VSMC cultures. Pretreatment with diphenyleneiodonium (an NADPH oxidase inhibitor) and U0126 or PD98059 (MEK Inhibitors) inhibited PDGF-BB-induced BMAL1 expression in a dose-dependent manner in VSMCs. In addition, the knockdown of early growth factor protein-1 (Egr-1) significantly inhibited PDGF-BB-induced BMAL1 mRNA or protein expression in VSMCs, and the knockdown of BMAL1 significantly decreased PDGF-BB-induced cell proliferation and extracellular signal-regulated kinase (ERK) phosphorylation but not Akt phosphorylation in VSMCs. The results demonstrate that PDGF-BB up-regulates BMAL1 expression through reactive oxygen species/ERK/Egr-1 pathways and that BMAL1 is involved in PDGF-BB-induced cell proliferation partially through ERK in VSMCs. Thus, BMAL1 may be a novel therapeutic target for the treatment of atherosclerosis including vascular remodeling.


Assuntos
Fatores de Transcrição ARNTL/metabolismo , Lesões das Artérias Carótidas/patologia , Proliferação de Células/fisiologia , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Fatores de Transcrição ARNTL/antagonistas & inibidores , Fatores de Transcrição ARNTL/genética , Animais , Aterosclerose/tratamento farmacológico , Aterosclerose/patologia , Becaplermina/farmacologia , Artérias Carótidas/patologia , Lesões das Artérias Carótidas/fisiopatologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ritmo Circadiano/fisiologia , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Músculo Liso Vascular/citologia , Cultura Primária de Células , Ratos , Espécies Reativas de Oxigênio/metabolismo , Remodelação Vascular/efeitos dos fármacos , Remodelação Vascular/fisiologia
5.
Cell Biol Int ; 44(3): 873-881, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31833156

RESUMO

Cardiac cell death is one of the major events implicated in doxorubicin-induced cardiotoxicity, which leads to heart failure. We recently reported that Yes-associated protein 1 (YAP1) regulates cell survival and apoptosis. However, it is unclear whether YAP1 regulates doxorubicin-induced cell death in cardiomyocytes. We investigated whether YAP1 is involved in doxorubicin-induced cell death using H9c2 cardiac cells and mouse heart. In an in vivo study, YAP1 protein expression was significantly decreased in hearts of doxorubicin-treated mice with increased caspase-3 activation. Doxorubicin also caused cell death by increasing caspase-3 activation in H9c2 cells. Doxorubicin reduced YAP1 protein expression and messenger RNA expression accompanied by increased phosphorylation of YAP1 at Ser127. Doxorubicin further increased cell death with increased caspase-3/7 activation in the absence of YAP1 when compared with doxorubicin or siYAP1 treatment alone. Overexpression of constitutively active YAP1 (YAP1-5SA) using an adenovirus gene transfer technique significantly reversed doxorubicin-induced cell death by decreasing caspase-3/7 activation in H9c2 cells. Akt, a potential prosurvival factor, decreased in doxorubicin- and YAP1 short interfering RNA (siRNA)-treated cells. Doxorubicin further significantly decreased Akt protein expression when YAP1 was silenced. Overexpression of YAP1 canceled decreased Akt protein expression induced by doxorubicin treatment in H9c2 cells. In conclusion, these results suggest that doxorubicin-induced cardiac cell death is mediated in part by down-regulation of YAP1 and YAP1-targeted gene, Akt. Modulating YAP1 and its related Hippo pathway on local cardiomyocytes may be a promising therapeutic approach for doxorubicin-induced cardiotoxicity.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas de Ciclo Celular/fisiologia , Morte Celular/efeitos dos fármacos , Doxorrubicina/toxicidade , Miócitos Cardíacos/efeitos dos fármacos , Animais , Cardiotoxicidade , Caspase 3/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/citologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Proteínas de Sinalização YAP
6.
Eur J Pharmacol ; 815: 470-477, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-28951205

RESUMO

Apoptosis of vascular smooth muscle cells (VSMCs) has been implicated in the progression of atherosclerosis, especially in vascular remodelling and plaque rupture. Although it is known that Yes-associated protein 1 (YAP1) is a critical molecule that regulates cell proliferation, differentiation and apoptosis, the role of YAP1 in VSMCs apoptosis remains unknown. In this study, we investigated whether YAP1 modulates VSMC apoptosis induced by endoplasmic reticulum (ER) stress. In cultured VSMC, tunicamycin caused cell death accompanied by an increase in caspase-3 processing and C/EBP homologous protein (CHOP) expression. YAP1 protein expression was downregulated by tunicamycin and the phosphorylation of YAP1 at the Ser127 site was significantly increased by tunicamycin. Tunicamycin further decreased cell viability followed by an increase in caspase-3 processing in the absence of YAP1 when compared with treatment only with tunicamycin or siYAP1. On the other hand, overexpression of a constitutively active YAP1 (YAP1-5SA), which lacks five serine phosphorylation sites, significantly prevented the caspase-3 processing and restored the decrease in cell viability induced by tunicamycin. Overexpression of YAP1-5SA significantly inhibited tunicamycin-induced caspase-8 processing without affecting phosphorylation of p-38 and Akt. Furthermore, the overexpression of YAP1-5SA significantly restored the decrease in ANKRD1 expression induced by tunicamycin. The inhibition of tunicamycin-induced caspase-3 cleavage by YAP1-5SA was markedly attenuated in ANKRD1-knockdown cells. These results demonstrate that ER stress can alter intracellular YAP1 protein expression in VSMCs and that YAP1 is protective against VSMC apoptosis induced by ER stress through inhibiting caspase8/3 activation mediated in part by upregulation of ANKRD1.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Estresse do Retículo Endoplasmático , Músculo Liso Vascular/citologia , Animais , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Caspase 3/metabolismo , Morte Celular/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Masculino , Proteínas Musculares/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Repressoras/metabolismo , Tunicamicina/farmacologia , Proteínas de Sinalização YAP , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
Cell Biol Int ; 40(11): 1204-1211, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27569904

RESUMO

Impaired insulin signaling in skeletal muscle cells causes insulin resistance associated with the onset of type 2 diabetes. Although interleukin (IL)-1ß has been considered to be implicated in the pathogenesis of type 2 diabetes, the action of prolonged stimulation with IL-1ß on the insulin signaling pathway in skeletal muscle cells remains poorly understood. In the current study, we investigated the effect of IL-1ß stimulation on insulin signal transduction from the insulin receptor (IR), resulting in glucose transporter 4 (GLUT4) translocation in skeletal muscle cells. In L6-GLUT4myc cells, stimulation with IL-1ß for 24 h promoted GLUT4 translocation to the plasma membrane and increased glucose uptake in a concentration-dependent manner, whereas short-term stimulation with IL-1 for up to 6 h did not affect that. In addition, stimulation with IL-1ß for 24 h further increased insulin-stimulated GLUT4 translocation. Interestingly, stimulation with IL-1ß for 24 h did not cause any change in the phosphorylation of insulin signal molecules IR, insulin receptor substrate (IRS)-1, Akt, and p21-activated kinase (PAK1). Stimulation with IL-1ß for 24 h significantly increased AMP-activated protein kinase (AMPK) phosphorylation and GLUT4 protein expression. Small interfering RNA (siRNA) targeting AMPK1/2 significantly inhibited IL-1ß-stimulated GLUT4 translocation. These results suggest that prolonged stimulation with IL-1ß positively regulates GLUT4 translocation in skeletal muscle cells. IL-1ß may have a beneficial effect on maintaining glucose homeostasis in skeletal muscle cells in patients with type 2 diabetes. .


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Interleucina-1beta/farmacologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Glucose/metabolismo , Insulina/metabolismo , Músculo Esquelético/enzimologia , Fosforilação , Transporte Proteico , RNA Interferente Pequeno , Ratos , Receptor de Insulina/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos
8.
Cell Biol Int ; 40(3): 269-76, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26514426

RESUMO

Increased A disintegrin and metalloprotease 17 (ADAM17) expression in vascular smooth muscle cells (VSMC) is implicated in the development of cardiovascular diseases including atherosclerosis and hypertension. Although cilostazol, type III phosphodiesterase (PDE III) inhibitor, has recently been found to inhibit VSMC proliferation, the mechanisms remain largely unclear. Here, we hypothesized that cilostazol regulates the ADAM17 expression in VSMC. In cultured VSMC, interleukin (IL)-1α and IL-1ß significantly increased ADAM17 expression. MEK inhibitor U0126, NF-κB inhibitor BAY-11-7085, and siRNA targeting p65/RelA significantly inhibited IL-1α or IL-ß-induced ADAM17 expression. Cilostazol significantly inhibited IL-1α or IL-1ß-induced extracellular signal-regulated kinase (ERK) phosphorylation and ADAM17 expression. Unexpectedly, cilostamide, dibutryl cAMP, and forskolin did not affect IL-1-induced ADAM17 expression. Our results clearly demonstrated that IL-1 induces ADAM17 expression through ERK/NF-κB activation in VSMCs. Moreover, the inhibitory effects of cilostazol on IL-1-induced ADAM17 expression may be independent of the cAMP signaling pathway in VSMC. These novel findings may provide important clues to understanding the expression mechanisms of ADAM17 and the inhibitory mechanisms of cilostazol in VSMC proliferation.


Assuntos
Proteína ADAM17/metabolismo , AMP Cíclico/metabolismo , Interleucina-1/farmacologia , Transdução de Sinais/efeitos dos fármacos , Tetrazóis/farmacologia , Animais , Células Cultivadas , Cilostazol , MAP Quinases Reguladas por Sinal Extracelular , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Fosforilação/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo
9.
J Vet Med Sci ; 78(2): 341-5, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26460314

RESUMO

We collected rectal swabs from dogs in Japan during 2011 to 2014, and canine coronavirus (CCoV) nucleocapsid gene was detected by RT-PCR. The relationship between CCoV infection and the manifestation of diarrhea symptoms was investigated, and a correlation was noted (df=1, χ(2)=8.90, P<0.005). The types of CCoV detected in samples from CCoV-infected dogs were CCoV-I in 88.9% and CCoV-II in 7.4%, respectively. We retrospectively investigated the seroprevalence of CCoV-I in dogs in Japan during 1998 to 2006. The sera were tested with a neutralizing antibody test. In the absence of CCoV-I laboratory strain, we used feline coronavirus (FCoV)-I that shares high sequence homology in the S protein with CCoV-I. 77.7% of the sera were positive for neutralizing anti-FCoV-I antibodies.


Assuntos
Infecções por Coronavirus/veterinária , Coronavirus Canino , Doenças do Cão/virologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Infecções por Coronavirus/sangue , Infecções por Coronavirus/epidemiologia , Proteínas do Nucleocapsídeo de Coronavírus , Coronavirus Canino/genética , Doenças do Cão/sangue , Doenças do Cão/epidemiologia , Cães , Feminino , Japão/epidemiologia , Masculino , Proteínas do Nucleocapsídeo/genética , RNA Viral , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Estudos Retrospectivos
10.
Cell Biol Int ; 39(9): 1053-60, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25858022

RESUMO

Doxorubicin is an anthracycline antibiotic widely used in cancer treatment. Although its antitumor efficacy appears to be dose dependent, its clinical use is greatly restricted by the development of cardiotoxicity associated with apoptosis. Although caveolin-1, the major structural protein in caveolae, can positively or negatively regulate apoptosis depending on the stimulus or cell types, the contribution of caveolin-1 to doxorubicin-induced apoptosis remains unknown. This study was performed to identify the regulatory role of caveolin-1 on doxorubicin-induced apoptosis in H9c2 cardiac cells using a genetic approach. Caveolin-1 knockdown with a short hairpin (sh) RNA adenovirus, but not overexpression of wild-type caveolin-1, resulted in a marked inhibition of doxorubicin-induced caspase-3 cleavage. However, caveolin-1 knockdown tended to protect against doxorubicin-induced decrease in cell viability, but it did not significantly reverse cell death induced by doxorubicin. Doxorubicin stimulated the phosphorylation of p38 and extracellular signal regulated kinase (ERK). Doxorubicin-induced caspase-3 cleavage was inhibited by U0126, a MEK inhibitor or SB203580, a p38 inhibitor. Caveolin-1 knockdown markedly inhibited doxorubicin-induced p-38 phosphorylation but not ERK-mediated p-53 phosphorylation in H9c2 cardiac cells. Our results suggest that reduced caveolin-1 expression plays an anti-apoptotic role in doxorubicin-induced apoptosis but that it is insufficient to prevent such an apoptosis in H9c2 cardiac cells.


Assuntos
Apoptose/efeitos dos fármacos , Caveolina 1/biossíntese , Doxorrubicina/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Animais , Antibióticos Antineoplásicos/farmacologia , Caspase 3/metabolismo , Cavéolas/metabolismo , Caveolina 1/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
J Pharmacol Sci ; 122(2): 93-102, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23698110

RESUMO

AMP-activated protein kinase (AMPK) contributes to the acceleration of insulin signaling. However, the mechanism by which AMPK regulates insulin signaling remains unclear. Serine phosphorylation of insulin receptor substrate (IRS)-1 negatively regulates insulin signaling. Here we investigated the role of AMPK in serine phosphorylation of IRS-1 at 636/639 and 307, which is induced by tumor necrosis factor (TNF)-α in 3T3L1 adipocytes. We demonstrated that the AMPK activator 5-aminoimidazole-4-carboxamide-1-d-ribofuranoside (AICAR) significantly inhibited the TNF-α-induced serine phosphorylation of IRS-1 at 636/639 and 307 by suppression of extracellular signal-regulated kinase (ERK) phosphorylation but not c-Jun-NH2-terminal kinase (JNK) phosphorylation. In addition, AICAR stimulation resulted in enhanced interaction between ERK and MAP kinase phosphatase-4 (DUSP9/MKP-4) without affecting DUSP9/MPK4 mRNA synthesis. Moreover, intraperitoneal administration (0.25 g/kg) of AICAR to db/db mice improved blood glucose levels and inhibited the phosphorylation of ERK in adipose tissue. In conclusion, we propose a new mechanism in which AICAR suppresses TNF-α-induced serine phosphorylation of IRS-1 at 636/639 and 307 by enhancing the interaction between ERK and DUSP9/MKP-4. Taken together, these findings provide evidence that AMPK plays a crucial role in improving of type 2 diabetes.


Assuntos
Células 3T3-L1/metabolismo , Proteínas Quinases Ativadas por AMP/fisiologia , Aminoimidazol Carboxamida/análogos & derivados , Hipoglicemiantes/farmacologia , Proteínas Substratos do Receptor de Insulina/metabolismo , Ribonucleotídeos/farmacologia , Serina/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Aminoimidazol Carboxamida/administração & dosagem , Aminoimidazol Carboxamida/farmacologia , Animais , Células Cultivadas , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/etiologia , Fosfatases de Especificidade Dupla/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hipoglicemiantes/administração & dosagem , Injeções Intraperitoneais , Resistência à Insulina/fisiologia , Camundongos , Camundongos Endogâmicos , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , Fosforilação/efeitos dos fármacos , Ribonucleotídeos/administração & dosagem , Transdução de Sinais , Estimulação Química , Fator de Necrose Tumoral alfa/antagonistas & inibidores
12.
J Pharmacol Sci ; 116(2): 181-7, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21597238

RESUMO

Effects of azelnidipine were examined and compared with those of amlodipine on stunned myocardium in dogs. The left anterior descending (LAD) coronary artery was ligated for 20 min and subsequently released for 60 min. A vehicle, azelnidipine (0.3 mg/kg), or amlodipine (0.3 or 1 mg/kg) was injected intravenously 20 min before LAD ligation. The heart rate increased after a depressor response in the presence of amlodipine, while it decreased despite a decrease in arterial pressures in the presence of azelnidipine. After reperfusion, the coronary flow (CF) significantly increased in the presence of azelnidipine, but did not change with amlodipine after reperfusion. A positive inotropic effect was observed after treatment with both calcium antagonists. Ischemia significantly decreased the percentage of segment shortening (%SS) in all groups. Treatment with both calcium antagonists significantly increased %SS after reperfusion, although high-energy phosphate levels did not improve in the presence of calcium antagonists 60 min after reperfusion. Mortality with azelnidipine was significantly lower than that with 0.3 mg/kg amlodipine immediately after reperfusion. In conclusion, improvement in myocardial stunning after pretreatment with azelnidipine is associated with an increase in CF after reperfusion. The negative chronotropic action may have contributed to decreased mortality due to reperfusion arrhythmias. Azelnidipine is more beneficial than amlodipine and may provide an additional advantage to patients with angina and hypertension.


Assuntos
Anlodipino/farmacologia , Ácido Azetidinocarboxílico/análogos & derivados , Bloqueadores dos Canais de Cálcio/farmacologia , Di-Hidropiridinas/farmacologia , Doenças do Cão/prevenção & controle , Traumatismo por Reperfusão/prevenção & controle , Animais , Ácido Azetidinocarboxílico/farmacologia , Doenças do Cão/mortalidade , Cães , Metabolismo Energético , Feminino , Masculino , Miocárdio/metabolismo , Traumatismo por Reperfusão/mortalidade
13.
J Pharmacol Sci ; 109(1): 94-101, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19129682

RESUMO

3-Hydroxy-3-methylglutaryl CoA reductase inhibitors (statins) are safe and well-tolerated therapeutic drugs. However, they occasionally induce myotoxicity such as myopathy and rhabdomyolysis. Here, we investigated the mechanism of statin-induced myotoxicity in L6 fibroblasts and in rats in vivo. L6 fibroblasts were differentiated and then treated with pravastatin, simvastatin, or fluvastatin for 72 h. Hydrophobic simvastatin and fluvastatin decreased cell viability in a dose-dependent manner via apoptosis characterized by typical nuclear fragmentation and condensation and caspase-3 activation. Both hydrophobic statins transferred RhoA localization from the cell membrane to the cytosol. These changes induced by both hydrophobic statins were completely abolished by the co-application of geranylgeranylpyrophosphate (GGPP). Y27632, a Rho-kinase inhibitor, mimicked the hydrophobic statin-induced apoptosis. Hydrophilic pravastatin did not affect the viability of the cells. Fluvastatin was continuously infused (2.08 mg/kg at an infusion rate of 0.5 mL/h) into the right internal jugular vein of the rats in vivo for 72 h. Fluvastatin infusion significantly elevated the plasma CPK level and transferred RhoA localization in the skeletal muscle from the cell membrane to the cytosol. In conclusion, RhoA dysfunction due to loss of lipid modification with GGPP is involved in the mechanisms of statin-induced skeletal muscle toxicity.


Assuntos
Fibroblastos/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/toxicidade , Músculo Esquelético/efeitos dos fármacos , Amidas/farmacologia , Androstadienos/administração & dosagem , Androstadienos/farmacologia , Animais , Caspase 3/metabolismo , Técnicas de Cultura de Células/métodos , Sobrevivência Celular/efeitos dos fármacos , Creatina Quinase/antagonistas & inibidores , Creatina Quinase/metabolismo , Relação Dose-Resposta a Droga , Ácidos Graxos Monoinsaturados/administração & dosagem , Ácidos Graxos Monoinsaturados/toxicidade , Fibroblastos/citologia , Fibroblastos/metabolismo , Fluvastatina , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Indóis/administração & dosagem , Indóis/toxicidade , Infusões Intravenosas , Células L , Camundongos , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Pravastatina/administração & dosagem , Pravastatina/toxicidade , Piridinas/farmacologia , Ratos , Sinvastatina/administração & dosagem , Sinvastatina/toxicidade , Frações Subcelulares/química , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Fatores de Tempo , Wortmanina , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
14.
Yakugaku Zasshi ; 129(1): 155-61, 2009 Jan.
Artigo em Japonês | MEDLINE | ID: mdl-19122443

RESUMO

Mevalotin containing pravastatin, a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, is the brand medicine and well known to be effective for patients with dyslipidemia. Now, more than 20 generic pravastatins are available for clinical therapy. We compared pharmaceutical property of Mevan,a generic pravastatin, with that of Mevalotin.According to the definition of the Japanese Pharmacopoeia, Mevalotin 10 mg tablets were uniform in pravastatin content, whereas 5 mg tablets were rather variable. Variation in pravastatin content of Mevan 5 mg tablets was the same as Mevalotin 5 mg, whereas that of 10 mg tablets was very variable. The plasma concentration of pravastatin in the normal rabbits continuously increased until 180 min after oral administration of 30 mg Mevan, whereas it increased in a biphasic pattern after 30 mg Mevalotin.All rabbits were fed 0.2% cholesterol diet throughout the experiment. After 8 weeks, oral administration of either Mevalotin or Mevan was started at the dose of 30 mg pravastatin/day for 16 weeks. After a transient increase for a few weeks, the plasma levels of total- and LDL-cholesterol gradually decreased in Mevalotingroup, whereas these levels did not significantly changed in Mevan group within 16 weeks. The level of HDL-cholesterol in Mevan group tended to increase but not in Mevalotin group. The triglyceride level in Mevan group changed as well as that in Mevalotin group until 10 weeks after administration, and then gradually increased. The present results suggest that pharmaceutical properties of Mevan are not always identical with those of Mevalotin.


Assuntos
Colesterol na Dieta/administração & dosagem , LDL-Colesterol/sangue , Dieta Aterogênica , Medicamentos Genéricos , Inibidores de Hidroximetilglutaril-CoA Redutases/sangue , Pravastatina/sangue , Administração Oral , Animais , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Masculino , Pravastatina/administração & dosagem , Pravastatina/química , Pravastatina/farmacologia , Coelhos , Comprimidos , Fatores de Tempo
15.
J Pharmacol Sci ; 107(1): 80-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18469500

RESUMO

A number of patients with hyperlipidemia are prescribed 3-hydroxy-3-methylglutaryl-CoA reductase inhibitors that are concomitantly used along with the treatment of diabetes mellitus. The effects of atorvastatin and pravastatin on insulin-induced glucose uptake and the related signal transduction in 3T3L1 adipocytes were studied. 3T3L1 fibroblasts were differentiated into adipocytes, pretreated with atorvastatin or pravastatin, and then exposed to insulin. Glucose uptake and the amount of insulin signal proteins were measured. Atorvastatin significantly decreased insulin-stimulated 2-deoxyglucose uptake in 3T3L1 adipocytes associated with the prevention of translocation of GLUT4 into the plasma membrane. The amounts of Rab4 and RhoA that required lipid modification with farnesyl or geranylgeranyl pyrophosphate, in the membrane fraction were decreased by atorvastatin. Insulin-induced tyrosine phosphorylation of IRS-1 and serine/threonine phosphorylation of Akt were reduced by atorvastatin. Pravastatin did not modify these insulin-induced changes in the signal transduction. Inhibitors of the RhoA/Rho kinase system, C3 and Y27632, as well as atorvastatin reduced insulin-induced changes in signal transduction. Atorvastatin and pravastatin did not affect messenger RNA expression, protein level, and tyrosine phosphorylation of insulin receptors. In conclusion, hydrophobic atorvastatin decreases the glucose uptake by 3T3L1 adipocytes since it can enter the cell and prevents lipid modification of some proteins that are involved in the insulin signal transduction process.


Assuntos
Adipócitos/efeitos dos fármacos , Glucose/metabolismo , Ácidos Heptanoicos/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Insulina/metabolismo , Pravastatina/farmacologia , Pirróis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Células 3T3-L1 , ADP Ribose Transferases/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adipócitos/enzimologia , Adipócitos/metabolismo , Amidas/farmacologia , Animais , Atorvastatina , Toxinas Botulínicas/farmacologia , Relação Dose-Resposta a Droga , Transportador de Glucose Tipo 4/metabolismo , Proteínas Substratos do Receptor de Insulina , Camundongos , Fosforilação , Fosfatos de Poli-Isoprenil/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Prenilação de Proteína , Transporte Proteico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridinas/farmacologia , RNA Mensageiro/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Proteínas rab4 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/antagonistas & inibidores , Proteínas rho de Ligação ao GTP/metabolismo , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP
16.
J Pharmacol Sci ; 106(4): 593-9, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18403900

RESUMO

Incomplete recovery of myocardial contraction after reperfusion following brief ischemia is called the "stunning phenomenon" in an animal experiment. A hydrophilic 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor (statin) does not affect this phenomenon, but lipophilic statins further reduce the contraction during reperfusion. The effects of novel hydrophilic rosuvastatin and lipophilic pitavastatin on myocardial stunning in dogs were examined. In a preliminary experiment in vitro, pitavastatin reduced L6 cell viability at 10(-6) M and higher, whereas rosuvastatin and pravastatin up to 10(-5) M did not show such effects. An empty capsule or a capsule filled with rosuvastatin (2 mg/kg per day) or pitavastatin (0.4 mg/kg per day) was orally administered to dogs. After 3 weeks, both statins lowered the serum cholesterol level to the same extent. Under pentobarbital anesthesia, dogs were subjected to 15-min ischemia followed by 120-min reperfusion. Ischemia arrested the myocardial contraction in the ischemic area, and reperfusion recovered it but incompletely, showing the stunning phenomenon. Rosuvastatin did not modify the stunning phenomenon, while pitavastatin further deteriorated the myocardial contraction during reperfusion.


Assuntos
Fluorbenzenos/toxicidade , Inibidores de Hidroximetilglutaril-CoA Redutases/toxicidade , Contração Miocárdica/efeitos dos fármacos , Isquemia Miocárdica/fisiopatologia , Miocárdio Atordoado/etiologia , Miocárdio/metabolismo , Pirimidinas/toxicidade , Quinolinas/toxicidade , Sulfonamidas/toxicidade , Administração Oral , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Colesterol/sangue , Modelos Animais de Doenças , Cães , Relação Dose-Resposta a Droga , Metabolismo Energético/efeitos dos fármacos , Feminino , Fluorbenzenos/administração & dosagem , Hemodinâmica/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Masculino , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Isquemia Miocárdica/complicações , Isquemia Miocárdica/metabolismo , Miocárdio Atordoado/induzido quimicamente , Miocárdio Atordoado/metabolismo , Miocárdio Atordoado/fisiopatologia , Pirimidinas/administração & dosagem , Quinolinas/administração & dosagem , Ratos , Rosuvastatina Cálcica , Sulfonamidas/administração & dosagem , Fatores de Tempo
17.
Pharmacology ; 82(1): 22-9, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18434761

RESUMO

Olmesartan is a selective angiotensin II type 1 receptor (AT1) antagonist. In pentobarbital-anesthetized open-chest dogs, ischemia/reperfusion was induced by ligating the left anterior descending coronary artery for 20 min and releasing it for 60 min, respectively. The myocardial contraction in the ischemic area decreased and returned towards its pre-ischemic level during reperfusion but incompletely. Olmesartan improved the recovery of myocardial contraction during reperfusion associated with restoration of myocardial ATP. Angiotensin II repelled by AT1 receptors occupied by olmesartan can reach and stimulate the angiotensin II type 2 (AT2) receptors, resulting in some beneficial effects on the ischemic myocardium. In fact, AT2 receptor mRNA was found in the adult dog myocardium. In addition, the plasma level of angiotensin II was significantly increased by olmesartan. PD123319, a selective AT2 receptor antagonist, however, did not modify the effect of olmesartan on the cardiac contraction. The hypertensive response to exogenous angiotensin II was completely inhibited by olmesartan, whereas PD123319 did not abolish the effect of olmesartan. In conclusion, olmesartan protects the ischemic/reperfused heart against ischemic injury through inhibition of AT1 receptors but not indirect activation of AT2 receptors.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Imidazóis/farmacologia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Miocárdio Atordoado/tratamento farmacológico , Receptor Tipo 2 de Angiotensina/fisiologia , Tetrazóis/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Bloqueadores do Receptor Tipo 2 de Angiotensina II , Animais , Cães , Feminino , Hemodinâmica/efeitos dos fármacos , Imidazóis/uso terapêutico , Masculino , Contração Miocárdica/efeitos dos fármacos , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miocárdio Atordoado/fisiopatologia , Miocárdio/metabolismo , Piridinas/farmacologia , Tetrazóis/uso terapêutico
18.
J Gen Virol ; 89(Pt 4): 1025-1029, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18343845

RESUMO

Infection of the monocyte/macrophage lineage with feline infectious peritonitis virus (FIPV) is enhanced in the presence of anti-FIPV antibodies (antibody-dependent enhancement or ADE). We investigated the following unclear points concerning ADE of FIPV infection: (i) involvement of the virus receptor, feline aminopeptidase N (fAPN), in ADE activity in FIPV infection; (ii) necessity of acidification of the endosome in cellular invasion of FIPV. Virus receptor-blocking experiments using anti-fAPN antibodies at 4 or 37 degrees C and experiments using fAPN-negative U937 cells revealed that fAPN is not involved in ADE of FIPV infection. Experiments using lysosomotropic agents clarified that acidification of the endosome is necessary for cellular invasion by FIPV, regardless of the presence or absence of antibodies. These findings may be very important for understanding the mechanism of ADE of FIPV infection.


Assuntos
Anticorpos Antivirais/farmacologia , Antígenos CD13/fisiologia , Coronavirus Felino/fisiologia , Endossomos/metabolismo , Peritonite Infecciosa Felina/imunologia , Peritonite Infecciosa Felina/virologia , Receptores Virais/fisiologia , Cloreto de Amônio/farmacologia , Animais , Anticorpos Facilitadores , Gatos , Relação Dose-Resposta a Droga , Endossomos/efeitos dos fármacos , Macrolídeos/farmacologia , Macrófagos Alveolares/citologia , Macrófagos Alveolares/virologia , Replicação Viral/efeitos dos fármacos
19.
J Vet Med Sci ; 68(1): 21-5, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16462112

RESUMO

Canine coronavirus (CCoV) has been reported to cause acute diarrhea mainly in young pups. CCoV and feline coronavirus are classified as group 1 coronaviruses. However, it has recently been reported in the United Kingdom that the group 2 coronavirus gene, which is more closely related to the bovine coronavirus (BCoV) and human coronavirus strain OC43, has been detected in respiratory tract tissue samples from dogs with respiratory disease. In this study, we examined the prevalence of antibodies to group 2 coronaviruses in domestic dogs and cats in Japan by a neutralization test using BCoV. All 104 feline serum samples were negative (<1:5) for anti-BCoV antibodies. In contrast, of the 898 canine serum samples, 160 (17.8%) were positive for anti-BCoV antibodies, and the antibody titers ranged from 1:5 to more than 1:640, with 1:160 being the most frequent. No correlation was found between the titers of the anti-BCoV and anti-CCoV antibodies in the 198 serum samples of dogs with a known history of CCoV vaccination. We amplified, by RT-PCR, group 2 coronavirus-specific hemagglutination/esterase genes in the oral swabs of a total of 10 young pups presenting with or having recovered from respiratory signs, or having anti-BCoV antibodies, with the result that 2 pups were positive for the hemagglutination/esterase genes. These results strongly suggest that an unknown group 2 coronavirus as well as the known enteritis-causing CCoV (group 1 coronavirus) is prevalent among domestic dogs in Japan.


Assuntos
Infecções por Coronavirus/veterinária , Coronavirus/imunologia , Doenças do Cão/epidemiologia , Doenças do Cão/virologia , Animais , Anticorpos Antivirais/sangue , Coronavirus/genética , Infecções por Coronavirus/epidemiologia , Primers do DNA , Cães , Genes Virais/genética , Japão/epidemiologia , Testes de Neutralização , Prevalência , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Biol Pharm Bull ; 28(11): 2092-5, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16272695

RESUMO

We examined whether 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (statins) improve glucose intolerance in spontaneously diabetic Goto-Kakizaki (GK) rats or not. The fasting blood glucose, plasma insulin, and serum cholesterol levels were significantly higher in GK rats than those in age-matched Wistar rats. All rats were given orally once a day 0.5% carboxymethylcellulose, pravastatin 8 mg/kg, simvastatin 8 mg/kg, or atorvastatin 8 mg/kg. An oral glucose tolerance test (OGTT) was performed before and 3, 6 and 12 weeks after statin treatments. The hyperglycemic response to OGTT in GK rats significantly exceeded that in Wistar rats. The plasma insulin level in GK rats increased with age until 14-week-old (treated for 6 weeks), and then decreased. Glucose intake significantly increased the plasma insulin in almost all rats. The increment of plasma insulin due to OGTT in GK rats appeared to be less than that in Wistar rats, because the basal level was already high in GK rats. Pravastatin, simvastatin, and atorvastatin did not modify changes in blood glucose and plasma insulin induced by glucose intake. In conclusion, long-term treatments of GK rats with statins did not improve glucose intolerance observed during OGTT.


Assuntos
Diabetes Mellitus/genética , Intolerância à Glucose/tratamento farmacológico , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Animais , Atorvastatina , Glicemia/metabolismo , Peso Corporal/efeitos dos fármacos , Colesterol/sangue , Teste de Tolerância a Glucose , Ácidos Heptanoicos/uso terapêutico , Insulina/sangue , Resistência à Insulina/fisiologia , Masculino , Pravastatina/uso terapêutico , Pirróis/uso terapêutico , Ratos , Ratos Wistar , Sinvastatina/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...