Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Molecules ; 23(10)2018 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-30249030

RESUMEN

Xanthine-based KMUP-1 was shown to inhibit phosphodiesterases (PDEs) and modulate G-protein coupled receptors (GPCRs) to lower hyperlipidemia and body weight. This study further investigated whether KMUP-1 affects adipogenesis and lipolysis in 3T3-L1 preadipocytes. KMUP-1 (1⁻40 µM) concentration-dependently attenuated Oil Red O (ORO) staining and decreased triglyceride (TG) accumulation, indicating adipogenesis inhibition in 3T3-L1 cells. In contrast, the ß-agonist ractopamine increased ORO staining and TG accumulation and adipogenesis. KMUP-1 (1⁻40 µM) also reduced MAPKs/Akt/PPARγ expression, PPARγ1/PPARγ2 mRNA, and p-ERK immunoreactivity at the adipogenesis stage, but enhanced hormone sensitive lipase (HSL) immunoreactivity at the lipolysis stage. Addition of protein kinase A (PKA) or protein kinase G (PKG) antagonist (KT5720 or KT5728) to adipocytes did not affect HSL immunoreactivity. However, KMUP-1 did increase HSL immunoreactivity and the effect was reduced by PKA or PKG antagonist. Simvastatin, theophylline, caffeine, and sildenafil, like KMUP-1, also enhanced HSL immunoreactivity. Phosphorylated HSL (p-HSL) was enhanced by KMUP-1, indicating increased lipolysis in mature 3T3-L1 adipocytes. Decreases of MAPKs/Akt/PPARγ during adipogenesis contributed to inhibition of adipocyte differentiation, and increases of PKA/PKG at lipolysis contributed to HSL activation and TG hydrolysis. Taken together, the data suggest that KMUP-1 can inhibit hyperadiposity in 3T3-L1 adipocytes.


Asunto(s)
Adipocitos/citología , Adipogénesis/efectos de los fármacos , Piperidinas/farmacología , Transducción de Señal/efectos de los fármacos , Triglicéridos/metabolismo , Xantinas/farmacología , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Lipólisis/efectos de los fármacos , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , PPAR gamma/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Esterol Esterasa/metabolismo
2.
Mol Med Rep ; 15(6): 4199-4206, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28440482

RESUMEN

We have previously demonstrated that KMUP-1, a xanthine-based nitric oxide enhancer, attenuates diabetic glomerulosclerosis, while increasing renal endothelial nitric oxide synthase expression in rats. However, the anti­fibrotic mechanisms of KMUP­1 treatment in diabetic nephropathy in terms of cell biology and transforming growth factor-ß1 (TGF­ß1) remain unclear. Therefore, the present study involved investigating the effects of KMUP­1 on high glucose (HG) or TGF­ß1­induced pro­fibrotic proteins in mouse mesangial (MES13) cells, and the effects of KMUP­1 on streptozotocin (STZ)­induced diabetic rats. It was identified that KMUP­1 (10 µM) attenuated HG (30 mM)­induced cell hypertrophy while attenuating TGF­ß1 gene transcription and bioactivity in MES13 cells. In addition, KMUP­1 attenuated TGF­ß1 (5 ng/ml)­induced Smad2/3 phosphorylation while attenuating HG or TGF­ß1­induced collagen IV and fibronectin protein expression. Furthermore, KMUP­1 attenuated HG­decreased Suv39h1 and H3K9me3 levels. Finally, KMUP­1 attenuated diabetes-induced collagen IV and fibronectin protein expression in STZ­diabetic rats at 8 weeks. In conclusion, KMUP­1 attenuates HG and TGF­ß1­induced pro­fibrotic proteins in mesangial cells and attenuation of TGF­ß1­induced signaling and attenuation of HG­decreased Suv39h1 expression may be two of the anti-fibrotic mechanisms of KMUP­1.


Asunto(s)
Fibrosis/metabolismo , Glucosa/metabolismo , Células Mesangiales/metabolismo , Piperidinas/metabolismo , Xantinas/metabolismo , Animales , Línea Celular , Colágeno Tipo IV/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Fibronectinas/metabolismo , Fibrosis/patología , Histonas/metabolismo , Células Mesangiales/patología , Metiltransferasas/metabolismo , Ratones , Transducción de Señal/fisiología , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Estreptozocina/farmacología , Factor de Crecimiento Transformador beta1/metabolismo
3.
Pediatr Pulmonol ; 52(4): 443-457, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28029743

RESUMEN

BACKGROUND: Pulmonary hypertension (PH) is a devastating disorder, for which no therapy is curative. It has been reported that pulmonary vascular remodeling, associated with increasing mean pulmonary arterial pressure and upregulated expression of endothelial nitric oxide synthase (eNOS), endothelin-1 (ET-1), RhoA/RhoH-kinase results in the development of PH. Oxidative stress and the RhoA/Rho-kinase pathway are also thought to be involved in the pathophysiology of PH. Statins are 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (HMG-CoA reductase inhibitors) with pleiotropic effects and are potential agents for the treatment of PH. In this study, we investigated the beneficial effects of simvastatin on the development of PH secondary to left ventricular dysfunction. METHODS: A PH secondary to left ventricular dysfunction model was established in 6-week-old aortic-banded rats. The pulmonary expression of Rho kinase, ET-1, eNOS, p-eNOS, nitrite/nitrate (NOx), cGMP, p47Phox , and p67Phox were investigated in the early-treatment group, to which was administered simvastatin (30 mg/kg/day) from days 1 to 42 or the late-treatment group, to which was administered simvastatin (30 mg/kg/day) from days 29 to 42. RESULTS: Simvastatin attenuated the mean pulmonary artery pressure, pulmonary arteriolar remodeling, plasma brain natriuretic peptide, ET-1, reactive oxygen species, and the NADPH oxidase 2 regulatory subunits, p47Phox and p67Phox , and upregulated pulmonary p-eNOS, NOx, and cGMP in both the early- and late-treated groups. CONCLUSIONS: Inhibiting HMG-CoA reductase may have therapeutic potential for preventing and attenuating the development of PH in left ventricular dysfunction through the Rho-kinase pathway and NADPH oxidase. A translational study in humans is needed to substantiate these findings. Pediatr Pulmonol. 2017;52:443-457. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hipertensión Pulmonar/tratamiento farmacológico , NADPH Oxidasas/metabolismo , Simvastatina/uso terapéutico , Disfunción Ventricular Izquierda/tratamiento farmacológico , Quinasas Asociadas a rho/metabolismo , Animales , Modelos Animales de Enfermedad , Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/metabolismo , Masculino , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Disfunción Ventricular Izquierda/complicaciones , Disfunción Ventricular Izquierda/metabolismo
4.
Kaohsiung J Med Sci ; 32(12): 607-613, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27914611

RESUMEN

We have demonstrated that KMUP-1 (7-[2-[4-(2-chlorobenzene)piperazinyl]ethyl]-1,3-dimethylxanthine) blunts monocrotaline-induced pulmonary arterial hypertension by altering Ca2+ sensitivity, K+-channel function, endothelial nitric oxide synthase activity, and RhoA/Rho kinase (ROCK) expression. This study further investigated whether KMUP-1 impedes uridine 5'-triphosphate (UTP)-inhibited delayed rectifying K+ (KDR) current in rat pulmonary arteries involved the RhoA/ROCK signaling. Pulmonary artery smooth muscle cells (PASMCs) were enzymatically dissociated from rat pulmonary arteries. KMUP-1 (30µM) attenuated UTP (30µM)-mediated membrane depolarization and abolished UTP-enhanced cytosolic Ca2+ concentration. Whole-cell patch-clamp electrophysiology was used to monitor KDR currents. A voltage-dependent KDR current was isolated and shown to consist of a 4-aminopyridine (5mM)-sensitive component and an insensitive component. The 4-aminopyridine sensitive KDR current was suppressed by UTP (30µM). The ROCK inhibitor Y27632 (30µM) abolished the ability of UTP to inhibit the KDR current. Like Y27632, KMUP-1 (30µM) similarly abolished UTP-inhibited KDR currents. Superfused protein kinase A and protein kinase G inhibitors (KT5720, 300nM and KT5823, 300nM) did not affect UTP-inhibited KDR currents, but the currents were restored by adding KMUP-1 (30µM) to the superfusate. KMUP-1 reversal of KDR current inhibition by UTP predominantly involves the ROCK inhibition. The results indicate that the RhoA/ROCK signaling pathway plays a key role in eliciting PASMCs depolarization caused by UTP, which would result in pulmonary artery constriction. KMUP-1 blocks UTP-mediated PASMCs depolarization, suggesting that it would prevent abnormal pulmonary vasoconstriction.


Asunto(s)
Canales de Potasio de Tipo Rectificador Tardío/metabolismo , Miocitos del Músculo Liso/metabolismo , Óxido Nítrico/metabolismo , Piperidinas/farmacología , Arteria Pulmonar/citología , Uridina Trifosfato/farmacología , Xantinas/farmacología , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , 4-Aminopiridina/farmacología , Amidas/farmacología , Animales , Calcio/metabolismo , Citosol/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Femenino , Activación del Canal Iónico/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Piridinas/farmacología , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
5.
Int J Mol Sci ; 17(8)2016 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-27548140

RESUMEN

KMUP-1 (7-[2-[4-(2-chlorobenzene)piperazinyl]ethyl]-1,3-dimethylxanthine) has been reported to cause hepatic fat loss. However, the action mechanisms of KMUP-1 in obesity-induced steatohepatitis remains unclear. This study elucidated the steatohepatitis via matrix metallopeptidase 9 (MMP-9) and tumor necrosis factor α (TNFα), and related lipolysis via hormone sensitive lipase (HSL) and adipose triglyceride lipase (ATGL) by KMUP-1. KMUP-1 on steatohepatitis-associated HSL/p-HSL/ATGL/MMP-9/TNFα/interleukin-10 (IL-10) and infiltration of M1/M2 macrophages in obese mice were examined. KMUP-1 was administered by oral gavage from weeks 1-14 in high-fat diet (HFD)-supplemented C57BL/6J male mice (protection group) and from weeks 8-14, for 6 weeks, in HFD-induced obese mice (treatment group). Immunohistochemistry (IHC) and hematoxylin and eosin (H&E) staining of tissues, oil globules number and size, infiltration and switching of M1/M2 macrophages were measured to determine the effects on livers. IL-10 and MMP-9 proteins were explored to determine the effects of KMUP-1 on M1/M2 macrophage polarization in HFD-induced steatohepatitis. Long-term administration of KMUP-1 reversed HFD-fed mice increased in body weight, sGOT/sGPT, triglyceride (TG) and glucose. Additionally, KMUP-1 decreased MMP-9 and reactive oxygen species (ROS), and increased HSL/p-HSL and IL-10 in HFD mice livers. In conclusion, KMUP-1, a phosphodiesterase inhibitor (PDEI), was shown to reduce lipid accumulation in liver tissues, suggesting that it could be able to prevent or treat steatohepatitis induced by HFD.


Asunto(s)
Hígado Graso/tratamiento farmacológico , Interleucina-10/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Piperidinas/química , Piperidinas/uso terapéutico , Esterol Esterasa/metabolismo , Teofilina/química , Xantinas/química , Xantinas/uso terapéutico , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Glucemia/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Hígado/efectos de los fármacos , Hígado/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Obesos , Especies Reactivas de Oxígeno/metabolismo , Triglicéridos/sangre , Factor de Necrosis Tumoral alfa/metabolismo
6.
J Pharm Pharmacol ; 68(6): 810-8, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27109251

RESUMEN

OBJECTIVES: Endothelial damage is strongly associated with cardiovascular diseases such as atherosclerosis, thrombosis and hypertension. Endothelial progenitor cells (EPCs) are primitive bone marrow (BM) cells that possess the capacity to mature into endothelial cells and play a role in neovascularization and vascular remodelling. This study aimed to investigate whether KMUP-1, a synthetic xanthine-based derivative, atorvastatin and simvastatin, can prevent endothelial dysfunction and apoptosis induced by hypoxia and to elucidate the underlying mechanisms. METHODS: Mononuclear cells were separated and were induced to differentiate into EPCs. KMUP-1, atorvastatin or simvastatin were administered prior to hypoxia. KEY FINDINGS: We found that EPCs exposed to hypoxia increased apoptosis as well as diminished proliferation. Pretreatment with KMUP-1, atorvastatin and simvastatin significantly prevented hypoxia-induced EPCs death and apoptosis, with associated increased of the Bcl-2/Bax ratio, and reduced caspase-3 and caspase-9 expression. We also assessed the nitrite production and Ser(1177)-phospho-eNOS expression and found that KMUP-1, atorvastatin and simvastatin not only increased the secretion of NO compared with the hypoxia group but also upregulated the eNOS activation. CONCLUSIONS: KMUP-1 inhibited hypoxia-induced dysfunction and apoptosis in EPCs, which may be mediated through suppressing oxidative stress, upregulating eNOS and downregulating the caspase-3 signalling pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Progenitoras Endoteliales/efectos de los fármacos , Activadores de Enzimas/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Piperidinas/farmacología , Xantinas/farmacología , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Atorvastatina/farmacología , Hipoxia de la Célula , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citoprotección , Células Progenitoras Endoteliales/enzimología , Células Progenitoras Endoteliales/patología , Activación Enzimática , Óxido Nítrico/metabolismo , Nitritos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Simvastatina/farmacología
7.
Kaohsiung J Med Sci ; 32(2): 55-67, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26944323

RESUMEN

KMUP-3 (7-{2-[4-(4-nitrobenzene) piperazinyl]ethyl}-1, 3-dimethylxanthine) displays cardioprotection and increases cardiac output, and is suggested to increase cardiac performance and improve myocardial infarction. To determine whether KMUP-3 improves outcomes in hypoperfused myocardium by inducing Ca(2+) sensitization to oppose protein kinase (PK)G-mediated Ca(2+) blockade, we measured left ventricular systolic blood pressure, maximal rates of pressure development, mean arterial pressure and heart rate in rats, and measured contractility and expression of PKs/RhoA/Rho kinase (ROCK)II in beating guinea pig left atria. Hemodynamic changes induced by KMUP-3 (0.5-3.0 mg/kg, intravenously) were inhibited by Y27632 [(R)-(+)-trans-4-1-aminoethyl)-N-(4-Pyridyl) cyclohexane carboxamide] and ketanserin (1 mg/kg, intravenously). In electrically stimulated left guinea pig atria, positive inotropy induced by KMUP-3 (0.1-100µM) was inhibited by the endothelial NO synthase (eNOS) inhibitors N-nitro-l-arginine methyl ester (L-NAME) and 7-nitroindazole, cyclic AMP antagonist SQ22536 [9-(terahydro-2-furanyl)-9H-purin-6-amine], soluble guanylyl cyclase (sGC) antagonist ODQ (1H-[1,2,4] oxadiazolo[4,3-a] quinoxalin-1-one), RhoA inhibitor C3 exoenzyme, ß-blocker propranolol, 5-hydroxytryptamine 2A antagonist ketanserin, ROCK inhibitor Y27632 and KMUP-1 (7-{2-[4-(2-chlorobenzene) piperazinyl]ethyl}-1, 3-dimethylxanthine) at 10µM. Western blotting assays indicated that KMUP-3 (0.1-10µM) increased PKA, RhoA/ROCKII, and PKC translocation and CIP-17 (an endogenous 17-kDa inhibitory protein) activation. In spontaneous right atria, KMUP-3 induced negative chronotropy that was blunted by 7-nitroindazole and atropine. In neonatal myocytes, L-NAME inhibited KMUP-3-induced eNOS phosphorylation and RhoA/ROCK activation. In H9c2 cells, Y-27632 (50µM) and PKG antagonist KT5823 [2,3,9,10,11,12-hexahydro-10R- methoxy-2,9-dimethyl-1-oxo-9S,12R-epoxy-1H-diindolo(1,2,3-fg:3',2',1'-kl) pyrrolo(3,4-i)(1,6)benzodiazocine-10-carboxylic acid, methyl ester] (3µM) reversed KMUP-3 (1-100µM)-induced Ca(2+)-entry blockade. GPCR agonist activity of KMUP-3 appeared opposed to KMUP-1, and increased cardiac output via Ca(2+) sensitization, and displayed cardioprotection via cyclic GMP/PKG-mediated myocardial preconditioning in animal studies.


Asunto(s)
Gasto Cardíaco/efectos de los fármacos , Cardiotónicos/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Piperidinas/farmacología , Xantinas/farmacología , Animales , Presión Sanguínea , Señalización del Calcio , Carbazoles/farmacología , Línea Celular , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Evaluación Preclínica de Medicamentos , Femenino , Cobayas , Atrios Cardíacos/fisiopatología , Masculino , Transporte de Proteínas , Ratas Wistar , Receptores Acoplados a Proteínas G/metabolismo , Presión Ventricular
8.
J Lipid Res ; 56(11): 2070-84, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26351364

RESUMEN

The phosphodiesterase inhibitor (PDEI)/eNOS enhancer KMUP-1, targeting G-protein coupled receptors (GPCRs), improves dyslipidemia. We compared its lipid-lowering effects with simvastatin and explored hormone-sensitive lipase (HSL) translocation in hepatic fat loss. KMUP-1 HCl (1, 2.5, and 5 mg/kg/day) and simvastatin (5 mg/kg/day) were administered in C57BL/6J male mice fed a high-fat diet (HFD) by gavage for 8 weeks. KMUP-1 inhibited HFD-induced plasma/liver TG, total cholesterol, and LDL; increased HDL/3-hydroxy-3-methylglutaryl-CoA reductase (HMGR)/Rho kinase II (ROCK II)/PPARγ/ABCA1; and decreased liver and body weight. KMUP-1 HCl in drinking water (2.5 mg/200 ml tap water) for 1-14 or 8-14 weeks decreased HFD-induced liver and body weight and scavenger receptor class B type I expression and increased protein kinase A (PKA)/PKG/LDLRs/HSL expression and immunoreactivity. In HepG2 cells incubated with serum or exogenous mevalonate, KMUP-1 (10(-7)∼10(-5) M) reversed HMGR expression by feedback regulation, colocalized expression of ABCA1/apolipoprotein A-I/LXRα/PPARγ, and reduced exogenous geranylgeranyl pyrophosphate/farnesyl pyrophosphate (FPP)-induced RhoA/ROCK II expression. A guanosine 3',5'-cyclic monophosphate (cGMP) antagonist reversed KMUP-1-induced ROCK II reduction, indicating cGMP/eNOS involvement. KMUP-1 inceased PKG and LDLRs surrounded by LDL and restored oxidized LDL-induced PKA expresion. Unlike simvastatin, KMUP-1 could not inhibit (14)C mevalonate formation. KMUP-1 could, but simvastatin could not, decrease ROCK II expression by exogenous FPP/CGPP. KMUP-1 improves HDL via PPARγ/LXRα/ABCA1/Apo-I expression and increases LDLRs/PKA/PKG/HSL expression and immunoreactivity, leading to TG hydrolysis to lower hepatic fat and body weight.


Asunto(s)
Hiperlipoproteinemias/tratamiento farmacológico , Hipolipemiantes/farmacología , Piperidinas/farmacología , Xantinas/farmacología , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Dieta Alta en Grasa/efectos adversos , Evaluación Preclínica de Medicamentos , Células Hep G2 , Humanos , Hidroximetilglutaril-CoA Reductasas/metabolismo , Hiperlipoproteinemias/etiología , Hipolipemiantes/uso terapéutico , Grasa Intraabdominal/efectos de los fármacos , Grasa Intraabdominal/fisiología , Lipoproteínas HDL/sangre , Lipoproteínas LDL/metabolismo , Hígado/patología , Masculino , Ácido Mevalónico/metabolismo , Ratones Endogámicos C57BL , PPAR gamma/metabolismo , Piperidinas/uso terapéutico , Receptores de LDL/metabolismo , Receptores Depuradores de Clase B/metabolismo , Sistemas de Mensajero Secundario , Esterol Esterasa/metabolismo , Xantinas/uso terapéutico
9.
Molecules ; 20(6): 10435-49, 2015 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-26056815

RESUMEN

The signaling cascades of the mitogen activated protein kinase (MAPK) family, calcineurin/NFATc4, and PI3K/Akt/GSK3, are believed to participate in endothelin-1 (ET-1)-induced cardiac hypertrophy. The aim of this study was to investigate whether KMUP-1, a synthetic xanthine-based derivative, prevents cardiomyocyte hypertrophy induced by ET-1 and to elucidate the underlying mechanisms. We found that in H9c2 cardiomyocytes, stimulation with ET-1 (100 nM) for 4 days induced cell hypertrophy and enhanced expressions of hypertrophic markers, including atrial natriuretic peptide and brain natriuretic peptide, which were all inhibited by KMUP-1 in a dose-dependent manner. In addition, KMUP-1 prevented ET-1-induced intracellular reactive oxygen species generation determined by the DCFH-DA assay in cardiomyocytes. KMUP-1 also attenuated phosphorylation of ERK1/2 and Akt/GSK-3ß, and activation of calcineurin/NFATc4 and RhoA/ROCK pathways induced by ET-1. Furthermore, we found that the expression of heme oxygenase-1 (HO-1), a stress-response enzyme implicated in cardio-protection, was up-regulated by KMUP-1. Finally, KMUP-1 attenuated ET-1-stimulated activator protein-1 DNA binding activity. In conclusion, KMUP-1 attenuates cardiomyocyte hypertrophy induced by ET-1 through inhibiting ERK1/2, calcineurin/NFATc4 and RhoA/ROCK pathways, with associated cardioprotective effects via HO-1 activation. Therefore, KMUP-1 may have a role in pharmacological therapy of cardiac hypertrophy.


Asunto(s)
Calcineurina/metabolismo , Endotelina-1/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Hemo-Oxigenasa 1/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Factores de Transcripción NFATC/metabolismo , Piperidinas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Xantinas/farmacología , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Activación Enzimática , Glucógeno Sintasa Quinasa 3 beta , Hipertrofia , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Biológicos , Miocitos Cardíacos/patología , Unión Proteica , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1
10.
Int J Biol Sci ; 11(6): 633-42, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25999786

RESUMEN

Serotonin (5-hydroxytryptamine, 5-HT) is a potent pulmonary vasoconstrictor that promotes pulmonary artery smooth muscle cell (PASMC) proliferation. 5-HT-induced K(+) channel inhibition increases [Ca(2+)]i in PASMCs, which is a major trigger for pulmonary vasoconstriction and development of pulmonary arterial hypertension (PAH). This study investigated whether KMUP-1 reduces pulmonary vasoconstriction in isolated pulmonary arteries (PAs) and attenuates 5-HT-inhibited K(+) channel activities in PASMCs. In endothelium-denuded PA rings, KMUP-1 (1 µM) dose-dependently reduced 5-HT (100 µM) mediated contractile responses. Responses to KMUP-1 were reversed by K(+) channel inhibitors (TEA, 10 mM, 4-aminopyridine, 5 mM, and paxilline, 10 µM). In primary PASMCs, KMUP-1 also dose-dependently restored 5-HT-inhibited voltage-gated K(+)-channel (Kv1.5 and Kv2.1) and large-conductance Ca(2+)-activated K(+)-channel (BKCa) proteins, as confirmed by immunofluorescent staining. Furthermore, 5-HT (10 µM)-inhibited Kv1.5 protein was unaffected by the PKA inhibitor KT5720 (1 µM) and the PKC activator PMA (1 µM), but these effects were reversed by KMUP-1 (1 µM), 8-Br-cAMP (100 µM), chelerythrine (1 µM), and KMUP-1 combined with a PKA/PKC activator or inhibitor. Notably, KMUP-1 reversed 5-HT-inhibited Kv1.5 protein and this response was significantly attenuated by co-incubation with the PKC activator PMA, suggesting that 5-HT-mediated PKC signaling can be modulated by KMUP-1. In conclusion, KMUP-1 ameliorates 5-HT-induced vasoconstriction and K(+)-channel inhibition through the PKC pathway, which could be valuable to prevent the development of PAH.


Asunto(s)
Piperidinas/farmacología , Canales de Potasio/efectos de los fármacos , Proteína Quinasa C/fisiología , Arteria Pulmonar/metabolismo , Serotonina/fisiología , Vasoconstricción/efectos de los fármacos , Xantinas/farmacología , Animales , Técnicas In Vitro , Masculino , Contracción Muscular/efectos de los fármacos , Miocitos del Músculo Liso/química , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Canales de Potasio/fisiología , Proteína Quinasa C/metabolismo , Arteria Pulmonar/química , Ratas Sprague-Dawley , Serotonina/metabolismo
11.
Kaohsiung J Med Sci ; 31(5): 241-54, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25910559

RESUMEN

Ascorbic acid bound to KMUP-1 and sildenafil were examined for their antioxidant effects on vascular endothelium growth factor (VEGF) and endothelium nitric oxide synthase (eNOS) in hypoxic pulmonary artery (PA). Inhaled KMUP-1 and oral sildenafil released NO from eNOS. The effect of buffered l-ascorbic acid, alone and bound to KMUP-1 or sildenafil, for treating pulmonary arterial hypertension (PAH) is unclear. In this study, the antioxidant capacity of ascorbic acid increased the beneficial effects of KMUP-1 on PAH. KMUP-1A and sildenafil-A (5 mg/kg/d) were administered to hypoxic PAH rats. Pulmonary artery blood pressure, and VEGF, Rho kinase II (ROCK II), eNOS, soluble guanylate cyclase (sGC-α), and protein kinase G expression in lung tissues were measured to link PAH and right ventricular hypertrophy. Hypoxic rats had higher pulmonary artery blood pressure, greater PA medial wall thickness and cardiac weight, and a higher right ventricle/left ventricle + septum [RV/(LV+S)] ratio than normoxic rats. Oral KMUP-1A or sildenafil-A for 21 days in hypoxia prevented the rarefaction of eNOS in immunohistochemistry (IHC), reduced the IHC of VEGF in PAs, restored eNOS/protein kinase G/phosphodiesterase 5A; unaffected sGC-α and inactivated ROCK II expression were also found in lung tissues. In normoxic PA, KMUP-1A/Y27632 (10µM) increased eNOS and reduced ROCK II. ROCK II/reactive oxidative species was increased and eNOS was reduced after long-term hypoxia for 21 days. KMUP-1A or Y27632 blunted ROCK II in short-term hypoxic PA at 24 hours. l-Ascorbic acid + l-sodium ascorbate (40, 80µM) buffer alone directly inhibited the IHC of VEGF in hypoxic PA. Finally, KMUP-1A or sildenafil-A reduced PAH and associated right ventricular hypertrophy.


Asunto(s)
Ácido Ascórbico/uso terapéutico , Óxido Nítrico Sintasa de Tipo III/metabolismo , Piperidinas/uso terapéutico , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Citrato de Sildenafil/uso terapéutico , Xantinas/uso terapéutico , Amidas/farmacología , Animales , Ácido Ascórbico/química , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/metabolismo , Hipoxia/tratamiento farmacológico , Hipoxia/metabolismo , Masculino , Piperidinas/química , Piridinas/farmacología , Ratas , Citrato de Sildenafil/química , Factores de Crecimiento Endotelial Vascular/metabolismo , Xantinas/química
12.
J Cell Physiol ; 230(9): 2038-48, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25536014

RESUMEN

Phosphodiesterase (PDE) inhibitors have been suggested as a possible candidate for the treatment of osteopenia, including osteoporosis. KMUP-1 is a novel xanthine derivative with inhibitory activities on the PDE 3, 4, and 5 iso-enzymes to suppress the degradation of cAMP and cGMP. This study aimed to investigate the effect of KMUP-1 on osteoblast differentiation and the underlying cellular and molecular mechanisms. Primary osteoblasts and osteoblastic MC3T3-E1 cells were examined. KMUP-1 enhanced alkaline phosphatase (ALP) activity and mineralization compared to untreated controls in primary osteoblasts and MC3T3-E1 cells. KMUP-1 also increased the mRNA expression of the osteoblastic differentiation markers, including collagen type 1a, ALP, osteocalcin, osteoprotegerin, BMP-2, and Runx2, a key transcription regulator for osteoblastic differentiation. The osteogenic effect of KMUP-1 was abolished by BMP signaling inhibitor, noggin. Furthermore, we found that KMUP-1 upregulated Smad1/5/8 phosphorylations with subsequent BRE-Luc activation confirmed by transient transfection assay. In addition, KMUP-1 inactivated glycogen synthase kinase-3ß (GSK-3ß), with associated nuclear translocation of ß-catenin. Co-treatment with H89 and KT5823, cAMP and cGMP pathway inhibitors, respectively, reversed the KMUP-1-induced activations of Smad1/5/8, ß-catenin, and Runx2. The findings demonstrate for the first time that KMUP-1 can promote osteoblast maturation and differentiation in vitro via BMP-2/Smad1/5/8 and Wnt/ß-catenin pathways. These effects are mediated, in part, by the cAMP and cGMP signaling. Thus, KMUP-1 may be a novel osteoblast activator and a potential new therapy for osteoporosis.


Asunto(s)
Proteína Morfogenética Ósea 2/biosíntesis , Diferenciación Celular/efectos de los fármacos , Piperidinas/administración & dosificación , Proteína Smad1/biosíntesis , Proteína Smad5/biosíntesis , Proteína Smad8/biosíntesis , Xantinas/administración & dosificación , Animales , Proteína Morfogenética Ósea 2/genética , Calcificación Fisiológica , Línea Celular , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Ratones , Osteoblastos/efectos de los fármacos , ARN Mensajero/biosíntesis , Proteína Smad1/genética , Proteína Smad5/genética , Proteína Smad8/genética , Vía de Señalización Wnt
13.
Kaohsiung J Med Sci ; 30(6): 267-78, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24835346

RESUMEN

This study investigates whether endothelin-1 (ET-1) mediates monocrotaline (MCT)-induced pulmonary artery hypertension (PAH) and right ventricular hypertrophy (RVH), and if so, whether the G-protein coupled receptor antagonist KMUP-1 (7-{2-[4-(2-chlorobenzene)piperazinyl]ethyl}-1,3-dimethylxanthine) inhibits ET-1-mediated PA constriction and the aforementioned pathological changes. In a chronic rat model, intraperitoneal MCT (60 mg/kg) induced PAH and increased PA medial wall thickening and RV/left ventricle + septum weight ratio on Day 21 after MCT injection. Treatment with sublingual KMUP-1 (2.5 mg/kg/day) for 21 days prevented these changes and restored vascular endothelial nitric oxide synthase (eNOS) immunohistochemical staining of lung tissues. Western blotting analysis demonstrated that KMUP-1 enhanced eNOS, soluble guanylate cyclase, and protein kinase G levels, and reduced ET-1 expression and inactivated Rho kinase II (ROCKII) in MCT-treated lung tissue over long-term administration. In MCT-treated rats, KMUP-1 decreased plasma ET-1 on Day 21. KMUP-1 (3.6 mg/kg) maximally appeared at 0.25 hours in the plasma and declined to basal levels within 24 hours after sublingual administration. In isolated PA of MCT-treated rats, compared with control and pretreatment with l-NG-nitroarginine methyl ester (100 µM), KMUP-1 (0.1-100 µM) inhibited ET-1 (0.01 µM)-induced vasoconstriction. Endothelium-denuded PA sustained higher contractility in the presence of KMUP-1. In a 24-hour culture of smooth muscle cells (i.e., PA smooth muscle cells or PASMCs), KMUP-1 (0.1-10 µM) inhibited RhoA- and ET-1-induced RhoA activation. KMUP-1 prevented MCT-induced PAH, PA wall thickening, and RVH by enhancing eNOS and suppressing ET-1/ROCKII expression. In vitro, KMUP-1 inhibited ET-1-induced PA constriction and ET-1-dependent/independent RhoA activation of PASMCs. In summary, KMUP-1 attenuates ET-1-induced/ET-1-mediated PA constriction, and could thus aid in the treatment of PAH caused by MCT.


Asunto(s)
Endotelina-1/metabolismo , Hipertensión Pulmonar/enzimología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Piperidinas/farmacología , Arteria Pulmonar/patología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Xantinas/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Peso Corporal , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Endotelina-1/sangre , Guanilato Ciclasa/metabolismo , Frecuencia Cardíaca/efectos de los fármacos , Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/complicaciones , Hipertrofia Ventricular Derecha/enzimología , Hipertrofia Ventricular Derecha/patología , Hipertrofia Ventricular Derecha/fisiopatología , Técnicas In Vitro , Masculino , Monocrotalina , Piperazinas/farmacología , Piperidinas/sangre , Piperidinas/uso terapéutico , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/fisiopatología , Purinas/farmacología , Ratas Wistar , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Citrato de Sildenafil , Guanilil Ciclasa Soluble , Sulfonamidas/farmacología , Vasoconstricción/efectos de los fármacos , Xantinas/sangre , Xantinas/uso terapéutico , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
14.
Mol Pharm ; 11(5): 1621-31, 2014 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-24669856

RESUMEN

Neuropathic pain is characterized by spontaneous pain, hyperalgesia, and allodynia. The aim of this study was to investigate whether KMUP-1 (7-[2-[4-(2-chlorobenzene)piperazinyl]ethyl]-1,3-dimethylxanthine) could improve pain hypersensitivity and reduce inflammatory mediators, and also explore possible mechanisms in the rat sciatic nerve using bilateral chronic constriction injury (CCI) to induce neuropathic pain. Sprague-Dawley rats were randomly divided into four groups: Sham, Sham+KMUP-1, CCI, and CCI+KMUP-1. KMUP-1 (5 mg/kg/day) was injected intraperitoneally starting at day 1 after surgery. Mechanical and thermal responses were assessed before surgery and at days 3, 7, and 14 after CCI. Sciatic nerves around the injury site were isolated for Western blots and enzyme-linked immunosorbent assay to analyze protein and cytokine levels. The results show that thermal hyperalgesia and mechanical allodynia were reduced in the KMUP-1 treated group as compared to that in the CCI group. Inflammatory proteins (COX2, iNOS, and nNOS) and proinflammatory cytokines (TNF-α and IL-1ß) induced by CCI were decreased in the KMUP-1 treated group at day 7 after surgery. KMUP-1 also inhibited neuropathic pain-related mechanisms, including p38 and ERK activation, but not JNK. Furthermore, KMUP-1 blocked IκB phosphorylation (p-IκB) and phospho-nuclear factor κB (p-NF-κB) translocation to nuclei. Double immunofluorescent staining further demonstrated that p-IκB (an indicator of activated NFκB) and p-NFκB proteins were almost abolished by KMUP-1 in peripheral macrophages and spinal microglia cells at day 7 after surgery. On the basis of these findings, we concluded that KMUP-1 has antiinflammatory and antihyperalgesia properties in CCI-induced neuropathic pain via decreases in MAPKs and NF-κB activation.


Asunto(s)
Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Piperidinas/química , Piperidinas/uso terapéutico , Xantina/química , Xantinas/química , Xantinas/uso terapéutico , Animales , Western Blotting , Hiperalgesia , Inmunohistoquímica , Masculino , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Ratas , Ratas Sprague-Dawley
15.
Kaohsiung J Med Sci ; 30(3): 116-24, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24581211

RESUMEN

Eugenosedin-A (Eu-A) effects on vascular endothelial dysfunction and oxidative stress in a hyperlipidemic rat model were investigated. Rats were randomly divided into four groups: two control groups and two treatment groups. The control rats received a regular diet or high fat diet (HFD); the treatment rats fed received an HFD with 5 mg/kg Eu-A or atorvastatin for 10 weeks. No changes in serotonin levels were observed in the four groups; norepinephrine levels were enhanced in the HFD group which was attenuated by Eu-A and atorvastatin. In the HFD group, the vascular reactivity was increased by vasoconstrictors (5-nonyloxytryptamine, 5-HT, and phenylephrine) and decreased by an endothelium-dependent vasorelaxant, carbachol. Protein levels of α1-adrenergic receptors (not 5-HT1B/2A), reactive oxygen species (ROS) p47(phox), p67(phox), and gp91(phox), and oxidative damage markers 3-nitrotyrosine (3-NT) and 4-hydroxy-2-nonenal (4-HNE) were increased, but endothelial nitric oxide synthase (eNOS), P-eNOS and vasodilator-stimulated phosphoprotein phosphorylation (P-VASP) were decreased. Catalase and superoxide dismutase (SOD-1 and SOD-2) proteins were increased, but glutathione peroxidase (GPx) was decreased in the aorta. Eu-A and atorvastatin reduced vasoconstrictor-induced aortic contractions that might be related to 5-HT1B/2A and α1-adrenergic receptors inhibitory activities. Eu-A and atorvastatin improved eNOS/P-eNOS, P-VASP, GPx, and malondialdehyde (MDA) levels, and decreased ROS and oxidative damage markers. Taken together, we suggest that Eu-A can ameliorate hyperlipidemia-induced vascular endothelial dysfunction and oxidative dysregulation.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Hiperlipidemias/complicaciones , NADPH Oxidasas/metabolismo , Piperazinas/farmacología , Receptores Adrenérgicos/metabolismo , Serotonina/metabolismo , Animales , Aorta Torácica , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Moléculas de Adhesión Celular/metabolismo , Masculino , Malondialdehído/metabolismo , Proteínas de Microfilamentos/metabolismo , Norepinefrina/sangre , Estrés Oxidativo/efectos de los fármacos , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Serotonina/sangre , Superóxido Dismutasa/metabolismo
16.
Int J Urol ; 21(1): 87-92, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23692571

RESUMEN

OBJECTIVES: To investigate whether 7-[2-[4-(2-chlorophenyl) piperazinyl] ethyl]-1,3-di-methylxanthine (KMUP-1) inhibits the effects of testosterone on the development of benign prostatic hyperplasia and sensitizes prostate contraction. METHODS: A benign prostatic hyperplasia animal model was established by subcutaneous injections of testosterone (3 mg/kg/day, s.c.) for 4 weeks in adult male Sprague-Dawley rats. Animals were divided into six groups: control, testosterone, testosterone with KMUP-1 (2.5, 5 mg/kg/day), sildenafil (5 mg/kg/day) or doxazosin (5 mg/kg/day). After 4 weeks, the animals were killed, and prostate tissues were prepared for isometric tension measurement and western blotting analysis. KMUP-1, Y27632, zaprinast, doxazosin or tamsulosin were used at various concentrations to determine the contractility sensitized by phenylephrine (10 µmol/L). RESULTS: KMUP-1 inhibited testosterone-induced phosphorylation of extracellular signal-regulated phosphorylated protein kinase and mitogen-activated protein kinase kinase and Rho kinase-II activation. Sildenafil and doxazosin significantly decreased benign prostatic hyperplasia-induced mitogen-activated protein kinase kinase and Rho kinase-II activation. The decreased expressions of soluble guanylate cyclase α1 was reversed by KMUP-1, doxazosin and sildenafil. Soluble guanylate cyclase ß1 and protein kinase G were increased by KMUP-1, doxazosin, and sildenafil in the testosterone-treated benign prostatic hyperplasia group. Phosphodiesterase-5A was increased by testosterone and inhibited by KMUP-1 (5 mg/kg/day) or sildenafil (5 mg/kg/day). KMUP-1 inhibited phenylephrine-sensitized prostate contraction of rats treated with testosterone. CONCLUSIONS: Mitogen-activated protein kinase 1/extracellular regulated protein kinases kinase, soluble guanylate cyclase/cyclic guanosine monophosphate, protein kinase/protein kinase G and Rho kinase-II are related to prostate smooth muscle tone and proliferation induced by testosterone. KMUP-1 inhibits testosterone-induced prostate hyper-contractility and mitogen-activated protein kinase 1/extracellular regulated protein kinases kinase-phosphorylation, and it inactivates Rho kinase-II by cyclic guanosine monophosphate, protein kinase and α1A-adenergic blockade. Thus, KMUP-1 might be a beneficial pharmacotherapy for benign prostatic hyperplasia.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/fisiología , GMP Cíclico/fisiología , Guanilato Ciclasa/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Piperidinas/farmacología , Hiperplasia Prostática/prevención & control , Receptores Citoplasmáticos y Nucleares/fisiología , Xantinas/farmacología , Quinasas Asociadas a rho/fisiología , Animales , Proteínas Quinasas Dependientes de GMP Cíclico/efectos de los fármacos , Modelos Animales de Enfermedad , Guanilato Ciclasa/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Guanilil Ciclasa Soluble , Quinasas Asociadas a rho/efectos de los fármacos
17.
Pediatr Pulmonol ; 49(8): 734-44, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24167111

RESUMEN

Pulmonary vascular remodeling, characterized by disordered proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs), is a pathognomonic feature of pulmonary arterial hypertension. Thus, pharmacologic strategy targeting on anti-proliferation and anti-migration of PASMCs may have therapeutic implications for PAH. Here we investigated the effects and underlying mechanisms of B-type natriuretic peptide (BNP) on angiotensin II (Ang II)-induced proliferation and migration of PASMCs. Proliferation and migration of PASMCs cultured from Wistar rats were induced by Ang II, with or without BNP treatment. In addition, potential underlying mechanisms including cell cycle progression, Ca(2+) overload, reactive oxygen species (ROS) production, signal transduction of MAPK and Akt, and the cGMP/PKG pathway were examined. We found that BNP inhibited Ang II-induced PASMCs proliferation and migration dose dependently. BNP could also arrest the cell cycle progression in the G0/G1-phase. In addition, BNP attenuated intracellular calcium overload caused by Ang II. Moreover, Ang II-induced ROS production was mitigated by BNP, with associated down-regulation of NAD(P)H oxidase 1 (Nox1) and reduced mitochondrial ROS production. Finally, Ang II-activated MAPKs and Akt were also counteracted by BNP. Of note, all these effects of BNP were abolished by a PKG inhibitor (Rp-8-Br-PET-cGMPS). In conclusion, BNP inhibits Ang II-induced PASMCs proliferation and migration. These effects are potentially mediated by decreased calcium influx, reduced ROS production by Nox1 and mitochondria, and down-regulation of MAPK and Akt signal transduction, through the cGMP/PKG pathway. Therefore, this study implicates that BNP may have a therapeutic role in pulmonary vascular remodeling.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Natriuréticos/farmacología , Péptido Natriurético Encefálico/farmacología , Arteria Pulmonar/citología , Angiotensina II/farmacología , Animales , Calcio/metabolismo , Células Cultivadas , Regulación hacia Abajo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , NADH NADPH Oxidorreductasas/efectos de los fármacos , NADH NADPH Oxidorreductasas/metabolismo , NADPH Oxidasa 1 , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Arteria Pulmonar/efectos de los fármacos , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno , Vasoconstrictores/farmacología
18.
PLoS One ; 8(7): e69468, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936022

RESUMEN

BACKGROUND: KMUP-1 is a xanthine derivative with inhibitory activities on the phosphodiesterase (PDE) 3,4 and 5 isoenzymes to suppress the degradation of cyclic AMP and cyclic GMP. However, the effects of KMUP-1 on osteoclast differentiation are still unclear. In this study, we investigated whether KMUP-1 inhibits osteoclastogenesis induced by RANKL in RAW 264.7 cells and bone loss induced by ovariectomy in mice, and the underlying mechanisms. PRINCIPAL FINDINGS: In vitro, KMUP-1 inhibited RANKL-induced TRAP activity, the formation of multinucleated osteoclasts and resorption-pit formation. It also inhibited key mediators of osteoclastogenesis including IL-1ß, IL-6, TNF-α and HMGB1. In addition, KMUP-1 inhibited RANKL-induced activation of signaling molecules (Akt, MAPKs, calcium and NF-κB), mRNA expression of osteoclastogensis-associated genes (TRAP, MMP-9, Fra-1, and cathepsin K) and transcription factors (c-Fos and NFATc1). Furthermore, most inhibitory effects of KMUP-1 on RANKL-mediated signal activations were reversed by a protein kinase A inhibitor (H89) and a protein kinase G inhibitor (KT5823). In vivo, KMUP-1 prevented loss of bone mineral content, preserved serum alkaline phosphate and reduced serum osteocalcin in ovariectomized mice. CONCLUSIONS: KMUP-1 inhibits RANKL-induced osteoclastogenesis in vitro and protects against ovariectomy-induced bone loss in vivo. These effects are mediated, at least in part, by cAMP and cGMP pathways. Therefore, KMUP-1 may have a role in pharmacologic therapy of osteoporosis.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Calcio/metabolismo , Osteoclastos/efectos de los fármacos , Osteoporosis/prevención & control , Ovariectomía , Piperidinas/farmacología , Ligando RANK/genética , Xantinas/farmacología , Animales , Densidad Ósea , Calcineurina/genética , Calcineurina/metabolismo , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos BALB C , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Osteoclastos/metabolismo , Osteoclastos/patología , Osteoporosis/genética , Osteoporosis/metabolismo , Osteoporosis/patología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ligando RANK/metabolismo , Transducción de Señal
20.
J Pharm Pharmacol ; 65(2): 300-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23278698

RESUMEN

OBJECTIVES: Previous studies have shown eugenosedin-A, a 5-HT(1B/2A) and α(1)/α(2)/ß(1)-adrenergic blocker, is able to decrease cholesterol levels, hyperglycaemia and inflammation in hyperlipidaemic mice induced by high-fat diet (HFD). The aim of this study is to examine the effects of eugenosedin-A on the inhibition of adhesion molecules of platelets, the aorta and acyl-coenzymeA:cholesterol acyltransferase-1 (ACAT-1) of macrophages in a hyperlipidaemic rat model. METHODS: Six-week-old Sprague-Dawley rats were randomly divided into two control and treatment groups. The control rats received either a regular diet or HFD and the treatment groups were fed HFD with either 5 mg/kg eugenosedin-A or atorvastatin for a 10-week period. KEY FINDINGS: Compared with the two control groups, the HFD group had lower levels of high-density lipoprotein, higher concentrations of triglycerides, total cholesterol, low-density lipoprotein and insulin. The expression of adhesion molecules in platelets, aorta and monocyte-macrophage were enhanced by HFD. HFD also increased upstream proteins and their phosphorylated form in the aorta. In treatment groups, eugenosedin-A and atorvastatin improved HFD-induced hyperlipidaemia and levels of insulin. Eugenosedin-A reduced the upregulation of P-selectin, ICAM-1, ICAM-2, ICAM-3, VCAM, PECAM in platelets and inhibited E-selectin, ICAM-1, ICAM-2, ICAM-3, VCAM and PECAM protein levels in the aorta. Eugenosedin-A reduced the ACAT-1 protein expression of monocyte-macrophages. The expression of PKCα, MAPKs, IKKα and p65 and their phosphorylated form were reduced in treatment groups. CONCLUSIONS: Taken together, hyperlipidaemia enhances the expression of adhesion molecules and ACAT-1 protein, and eugenosedin-A ameliorates those increases. Through inhibition of MAPK- and p-65-mediated NF-κB pathway, eugenosedin-A decreases the quantity of adhesion molecules.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Dieta Alta en Grasa/efectos adversos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , FN-kappa B/antagonistas & inhibidores , Piperazinas/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/antagonistas & inhibidores , Acetil-CoA C-Acetiltransferasa/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Peso Corporal/efectos de los fármacos , Modelos Animales de Enfermedad , Selectina E/metabolismo , Hiperlipidemias/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Factor de Transcripción ReIA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA