Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Molecules ; 24(13)2019 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-31277363

RESUMEN

Berberine is reported to have multiple biological effects, including antimicrobial, anti-inflammatory, and antitumor activities, and 13-alkyl-substituted berberines show higher activity than berberine against certain bacterial species and human cancer cell lines. In particular, 13-ethylberberine (13-EBR) was reported to have anti-inflammatory effects in endotoxin-activated macrophage and septic mouse models. Thus, in this study, we aimed to examine the anticancer effects of 13-EBR and its mechanisms in radiotherapy-resistant (RT-R) MDA-MB-231 cells derived from the highly metastatic MDA-MB-231 cells. When we compared the gene expression between MDA-MB-231 and RT-R MDA-MB-231 cells with an RNA microarray, RT-R MDA-MB-231 showed higher levels of anti-apoptotic genes and lower levels of pro-apoptotic genes compared to MDA-MB-231 cells. Accordingly, we examined the effect of 13-EBR on the induction of apoptosis in RT-R MDA-MB-231 and MDA-MB-231 cells. The results showed that 13-EBR reduced the proliferation and colony-forming ability of both MDA-MB-231 and RT-R MDA-MB-231 cells. Moreover, 13-EBR induced apoptosis by promoting both intracellular and mitochondrial reactive oxygen species (ROS) and by regulating the apoptosis-related proteins involved in the intrinsic pathway, not in the extrinsic pathway. These results suggest that 13-EBR has pro-apoptotic effects in RT-R MDA-MB-231 and MDA-MB-231 cells by inducing mitochondrial ROS production and activating the mitochondrial apoptotic pathway, providing useful insights into new potential therapeutic strategies for RT-R breast cancer treatment.


Asunto(s)
Apoptosis/efectos de los fármacos , Berberina/farmacología , Neoplasias de la Mama/patología , Neoplasias de la Mama/radioterapia , Mitocondrias/metabolismo , Transducción de Señal , Berberina/química , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Mitocondrias/efectos de los fármacos , Modelos Biológicos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
2.
Biochem Biophys Res Commun ; 501(1): 73-79, 2018 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-29680657

RESUMEN

Excessive inflammation plays a detrimental role in endotoxemia. A recent study indicated that alarmins such as high mobility group box 1 (HMGB1) have drawn attention as therapeutic targets of sepsis. Post-translational modification (i.e., acetylation of lysine residues) of HMGB1 leads to the release of HMGB1 into the cellular space, operating as a warning signal that induces inflammation. Sirtuin 1 (SIRT1) has been shown to negatively regulate HMGB1 hyperacetylation and its extracellular release in sepsis. Therefore, we hypothesized that the S-nitrosylation (SNO) of SIRT1 may disrupt the ability of SIRT1 to negatively regulate the hyperacetylation of HMGB1. As long as the S-nitrosylation of SIRT1 occurs during septic conditions, it may worsen the situation. We found that the activity of SIRT1 decreased as the SNO-SIRT1 levels increased, resulting in HMGB1 release by LPS in RAW264.7 cells. Both the iNOS inhibitor (1400 W) and silencing iNOS significantly inhibited SNO-SIRT1, allowing increases in SIRT1 activity that decreased the HMGB1 release by LPS. SNAP, a NO donor, significantly increased both SNO-SIRT1 levels and the HMGB1 release that was accompanied by decreased sirt1 activity. However, sirtinol, a Sirt1 inhibitor, by itself decreased Sirt1 activity compared to that of the control, so that it did not affect already increased SNO-SIRT levels by SNAP. Most importantly, in lung tissues of LPS-endotoxic mice, significantly increased levels of SNO-SIRT were found, which was inhibited by 1400 W treatment. Plasma nitrite and HMGB1 levels were significantly higher than those in the sham controls, and the elevated levels were significantly lowered in the presence of 1400 W. We concluded that the S-nitrosylation of Sirt1 under endotoxic conditions may uninhibit the acetylation of HMGB1 and its extracellular release.


Asunto(s)
Endotoxemia/metabolismo , Proteína HMGB1/metabolismo , Sirtuina 2/metabolismo , Acetilación , Amidinas/farmacología , Animales , Bencilaminas/farmacología , Proteína HMGB1/sangre , Proteína HMGB1/química , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos BALB C , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/genética , Nitritos/sangre , Compuestos Nitrosos/química , Compuestos Nitrosos/metabolismo , Células RAW 264.7 , S-Nitroso-N-Acetilpenicilamina/farmacología , Sirtuina 2/química
3.
Inflamm Res ; 67(5): 445-453, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29497773

RESUMEN

OBJECTIVE: Although luteolin has shown to have anti-inflammatory action, no report is available whether luteolin inhibits HMGB1 and protects acute lung injury (ALI) in endotoxin rodents. We hypothesized that HO-1 induction by luteolin might play a crucial role for inhibition of pro-inflammatory mediators including HMGB1 through MAPK signaling in LPS-induced RAW264.7 cells, and it ameliorates ALI of endotoxin mice. METHODS: The effects of luteolin on the production of pro-inflammatory mediators in LPS-activated RAW264.7 cells and LPS-injected mice were evaluated. The mechanisms were investigated using various signal inhibitors. RESULTS: Luteolin significantly increased HO-1 expression through ERK1/2 signaling in a time- and concentration-dependent manner. Indeed, luteolin inhibited pro-inflammatory mediators (HMGB1, iNOS/NO, COX-2, and NF-κB activity) in LPS-activated RAW264.7 cells. In addition, PD98059, an ERK1/2 inhibitor, treatment failed to inhibit production of these pro-inflammatory mediators by luteolin. Interestingly, luteolin augmented HO-1 induction through Ca2+ influx in RAW264.7 cells. Administration of luteolin significantly inhibited plasma HMGB1 level, and iNOS expression in the lung that resulted in a significant reduction of ALI in endotoxin mice that was reversed by a HO-1 inhibitor, ZnPPIX. CONCLUSION: Therefore, we conclude that luteolin has a great potential for treatment of ALI and related diseases, where HMGB1 is a therapeutic target.


Asunto(s)
Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/prevención & control , Ciclooxigenasa 2/efectos de los fármacos , Proteína HMGB1/antagonistas & inhibidores , Hemo-Oxigenasa 1/biosíntesis , Lipopolisacáridos/antagonistas & inhibidores , Luteolina/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Animales , Señalización del Calcio/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclooxigenasa 2/biosíntesis , Endotoxinas , Proteína HMGB1/biosíntesis , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Células RAW 264.7
4.
Toxicol Appl Pharmacol ; 284(2): 113-24, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25737164

RESUMEN

Honokiol is a bioactive neolignan compound isolated from the species of Magnolia. This study was designed to elucidate the cellular mechanism by which honokiol alleviates the development of non-alcoholic steatosis. HepG2 cells were treated with honokiol for 1h, and then exposed to 1mM free fatty acid (FFA) for 24h to simulate non-alcoholic steatosis in vitro. C57BL/6 mice were fed with a high-fat diet for 28days, and honokiol (10mg/kg/day) was daily treated. Honokiol concentration-dependently attenuated intracellular fat overloading and triglyceride (TG) accumulation in FFA-exposed HepG2 cells. These effects were blocked by pretreatment with an AMP-activated protein kinase (AMPK) inhibitor. Honokiol significantly inhibited sterol regulatory element-binding protein-1c (SREBP-1c) maturation and the induction of lipogenic proteins, stearoyl-CoA desaturase-1 (SCD-1) and fatty acid synthase (FAS) in FFA-exposed HepG2 cells, but these effects were blocked by pretreatment of an AMPK inhibitor. Honokiol induced AMPK phosphorylation and subsequent acetyl-CoA carboxylase (ACC) phosphorylation, which were inhibited by genetic deletion of liver kinase B1 (LKB1). Honokiol stimulated LKB1 phosphorylation, and genetic deletion of LKB1 blocked the effect of honokiol on SREBP-1c maturation and the induction of SCD-1 and FAS proteins in FFA-exposed HepG2 cells. Honokiol attenuated the increases in hepatic TG and lipogenic protein levels and fat accumulation in the mice fed with high-fat diet, while significantly induced LKB1 and AMPK phosphorylation. Taken together, our findings suggest that honokiol has an anti-lipogenic effect in hepatocytes, and this effect may be mediated by the LKB1-AMPK signaling pathway, which induces ACC phosphorylation and inhibits SREBP-1c maturation in hepatocytes.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Compuestos de Bifenilo/farmacología , Hepatocitos/efectos de los fármacos , Lignanos/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Animales , Línea Celular Tumoral , Dieta Alta en Grasa/métodos , Ácido Graso Sintasas/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Lipogénesis/efectos de los fármacos , Hígado/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Estearoil-CoA Desaturasa/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Triglicéridos/metabolismo
5.
J Cell Mol Med ; 18(10): 2031-43, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25109475

RESUMEN

Patients suffering from diabetes mellitus (DM) are at a severe risk of atherothrombosis. Early growth response (Egr)-1 is well characterized as a central mediator in vascular pathophysiology. We tested whether valsartan independent of Ang II type 1 receptor (AT1R) can reduce tissue factor (TF) and toll-like receptor (TLR)-2 and -4 by regulating Egr-1 in THP-1 cells and aorta in streptozotocin-induced diabetic mice. High glucose (HG, 15 mM) increased expressions of Egr-1, TF, TLR-2 and -4 which were significantly reduced by valsartan. HG increased Egr-1 expression by activation of PKC and ERK1/2 in THP-1 cells. Valsartan increased AMPK phosphorylation in a concentration and time-dependent manner via activation of LKB1. Valsartan inhibited Egr-1 without activation of PKC or ERK1/2. The reduced expression of Egr-1 by valsartan was reversed by either silencing Egr-1, or compound C, or DN-AMPK-transfected cells. Valsartan inhibited binding of NF-κB and Egr-1 to TF promoter in HG condition. Furthermore, valsartan reduced inflammatory cytokine (TNF-α, IL-6 and IL-1ß) production and NF-κB activity in HG-activated THP-1 cells. Interestingly, these effects of valsartan were not affected by either silencing AT1R in THP-1 cells or CHO cells, which were devoid of AT1R. Importantly, administration of valsartan (20 mg/kg, i.p) for 8 weeks significantly reduced plasma TF activity, expression of Egr-1, TLR-2, -4 and TF in thoracic aorta and improved glucose tolerance of streptozotocin-induced diabetic mice. Taken together, we concluded that valsartan may reduce atherothrombosis in diabetic conditions through AMPK/Egr-1 regulation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Tetrazoles/farmacología , Tromboplastina/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Valina/análogos & derivados , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Western Blotting , Células Cultivadas , Inmunoprecipitación de Cromatina , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Proteína 1 de la Respuesta de Crecimiento Precoz/antagonistas & inhibidores , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/citología , Monocitos/metabolismo , ARN Interferente Pequeño/genética , Receptor de Angiotensina Tipo 1/química , Valina/farmacología , Valsartán
6.
Breast Cancer Res ; 16(5): R77, 2014 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-25156554

RESUMEN

INTRODUCTION: Extracellular nucleotides are released and detectable in a high concentration within the tumor microenvironment. G protein-coupled P2Y2 nucleotide receptor (P2Y2R) is activated equipotently by adenosine triphosphate (ATP) and uridine 5'-triphosphate (UTP), which mediate proinflammatory responses such as cell migration and proliferation. However, the role of P2Y2R in the process of cancer metastasis remains unclear. This study aimed to determine the role of P2Y2R in the proliferation, migration and invasion of highly metastatic MDA-MB-231 breast cancer cells through crosstalk with endothelial cells (ECs). METHODS: ATP release and P2Y2R activity between high metastatic breast cancer cell MDA-MB-231 and low metastatic breast cancer cell MCF-7 were compared. Then, the role of P2Y2R on tumor growth and invasion via crosstalk with ECs was examined in vitro, using MDA-MB-231 cells and ECs transfected with control- or P2Y2R-siRNA, and in vivo, using an animal model injected with control-shRNA- or P2Y2R-shRNA-transfected MDA-MB-231 cells. RESULTS: We found that this highly metastatic breast cancer cell line released higher levels of ATP and showed a higher P2Y2R activity in comparison to a low metastatic breast cancer cell line, MCF-7. In MDA-MB-231 cells, P2Y2R activation by ATP or UTP increased proliferation at 24 or 72 hours, which was abolished by P2Y2R knock-down. In addition, the adhesion of MDA-MB-231 cells to ECs and cell migration were both significantly increased by ATP or UTP through the expression of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) in MDA-MB-231 or ECs but not in cells where P2Y2R was knocked down. Furthermore, ATP- or UTP-mediated activation of P2Y2R induced MDA-MB-231 invasion through ECs, increased matrix metalloproteinase-9 (MMP-9) activity and vascular endothelial growth factor (VEGF) production in MDA-MB-231 and induced the phosphorylation of vascular endothelial (VE)-cadherin in ECs. Tumor growth and metastasis to other tissues were dramatically reduced, and body weight was increased in mice injected with P2Y2R-shRNA-transfected MDA-MB-231 cells compared to mice injected with control shRNA-transfected MDA-MB-231 cells. CONCLUSION: This study suggests that P2Y2R may play an important role in cancer metastasis via modulation of the crosstalk between cancer cells and ECs.


Asunto(s)
Adenosina Trifosfato/fisiología , Neoplasias de la Mama/patología , Células Endoteliales/metabolismo , Neoplasias Pulmonares/secundario , Receptores Purinérgicos P2Y2/metabolismo , Animales , Antígenos CD/metabolismo , Neoplasias de la Mama/metabolismo , Cadherinas/metabolismo , Comunicación Celular , Movimiento Celular , Proliferación Celular , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Células MCF-7 , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Desnudos , Trasplante de Neoplasias , Fosforilación , Procesamiento Proteico-Postraduccional , Uridina Trifosfato/fisiología , Factor A de Crecimiento Endotelial Vascular/biosíntesis
7.
Blood ; 119(11): 2523-32, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22262759

RESUMEN

The circulating peptide hormone hepcidin maintains systemic iron homeostasis. Hepcidin production increases during inflammation and as a result of endoplasmic reticulum (ER) stress. Elevated hepcidin levels decrease dietary iron absorption and promote iron sequestration in reticuloendothelial macrophages. Furthermore, increased plasma hepcidin levels cause hypoferremia and the anemia associated with chronic diseases. The signal transduction pathways that regulate hepcidin during inflammation and ER stress include the IL-6-dependent STAT-3 pathway and the unfolded protein response-associated cyclic AMP response element-binding protein-H (CREBH) pathway, respectively. We show that carbon monoxide (CO) suppresses hepcidin expression elicited by IL-6- and ER-stress agents by inhibiting STAT-3 phosphorylation and CREBH maturation, respectively. The inhibitory effect of CO on IL-6-inducible hepcidin expression is dependent on the suppressor of cytokine signaling-3 (SOCS-3) protein. Induction of ER stress in mice resulted in increased hepatic and serum hepcidin. CO administration inhibited ER-stress-induced hepcidin expression in vivo. Furthermore, ER stress caused iron accumulation in splenic macrophages, which could be prevented by CO. Our findings suggest novel anti-inflammatory therapeutic applications for CO, as well as therapeutic targets for the amelioration of anemia in the hypoferremic condition associated with chronic inflammatory and metabolic diseases.


Asunto(s)
Antimetabolitos/farmacología , Péptidos Catiónicos Antimicrobianos/antagonistas & inhibidores , Monóxido de Carbono/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/antagonistas & inhibidores , Estrés del Retículo Endoplásmico/efectos de los fármacos , Inflamación/patología , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/metabolismo , Western Blotting , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Hepcidinas , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Hierro/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
8.
Exp Dermatol ; 23(7): 480-5, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24816122

RESUMEN

P2Y2 R has been shown to be upregulated in a variety of tissues in response to stress or injury and to mediate tissue regeneration through its ability to activate multiple signalling pathways. This study aimed to investigate the role of P2Y2 R in the wound-healing process and the mechanisms by which P2Y2 R activation promotes wound healing in fibroblasts. The role of P2Y2 R in skin wound healing was examined using a full-thickness skin wound model in wildtype (WT) and P2Y2 R(-/-) mice and an in vitro scratch wound model in control or P2Y2 R siRNA-transfected fibroblasts. WT mice showed significantly decreased wound size compared with P2Y2 R(-/-) mice at day 14 post-wounding, and immunohistochemical analysis showed that a proliferation marker Ki67 and extracellular matrix (ECM)-related proteins VEGF, collagen I, fibronectin and α-SMA were overexpressed in WT mice, which were reduced in P2Y2 R(-/-) mice. Scratch-wounded fibroblasts increased ATP release, which peaked at 5 min. In addition, scratch wounding increased the level of P2Y2 R mRNA. Activation of P2Y2 R by ATP or UTP enhanced proliferation and migration of fibroblasts in in vitro scratch wound assays and were blocked by P2Y2 R siRNA. Finally, ATP or UTP also increased the levels of ECM-related proteins through the activation of P2Y2 R in fibroblasts. This study suggests that P2Y2 R may be a potential therapeutic target to promote wound healing in chronic wound diseases.


Asunto(s)
Adenosina Trifosfato/metabolismo , Receptores Purinérgicos P2Y2/metabolismo , Uridina Trifosfato/metabolismo , Cicatrización de Heridas , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Silenciador del Gen , Inmunohistoquímica , Antígeno Ki-67/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Interferente Pequeño/metabolismo , Receptores Purinérgicos P2Y2/genética , Piel/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
J Biol Chem ; 286(28): 24735-42, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21606497

RESUMEN

Nicotine inhibits the release of TNF-α from macrophage through activation of STAT3. Tristetraprolin (TTP) is known to destabilize pro-inflammatory transcripts containing AU-rich elements (ARE) in 3'-untranslated region (3'-UTR). Here we show that in LPS-stimulated human macrophages the anti-inflammatory action of nicotine is mediated by TTP. Nicotine induced activation of STAT3 enhanced STAT3 binding to the TTP promoter, increased TTP promoter activity, and increased TTP expression resulting in the suppression of LPS-stimulated TNF-α production. Overexpression of a dominant negative mutant of STAT3 (R382W) or down-regulation of STAT3 by siRNA abolished nicotine-induced TTP expression and suppression of LPS-stimulated TNF-α production. Nicotine enhanced the decay of TNF-α mRNA and decreased luciferase expression of a TNF-α 3'-UTR reporter plasmid in U937 cells. However, siRNA to TTP abrogated these effects of nicotine. In this experiment, we are reporting for the first time the involvement of TTP in the cholinergic anti-inflammatory cascade consisting of nicotine-STAT3-TTP-dampening inflammation.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Lipopolisacáridos/toxicidad , Macrófagos/metabolismo , Nicotina/farmacología , Regiones Promotoras Genéticas , Tristetraprolina/metabolismo , Regiones no Traducidas 3'/genética , Sustitución de Aminoácidos , Humanos , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Mutación Missense , Agonistas Nicotínicos/farmacología , Unión Proteica , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Tristetraprolina/genética , Factor de Necrosis Tumoral alfa/biosíntesis , Células U937
10.
Apoptosis ; 17(5): 463-74, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22183510

RESUMEN

Growing lines of evidence suggests that high mobility group box-1 (HMGB1) plays an important role for promoting inflammation and apoptosis in brain ischemia. Previously, we demonstrated that inducers of heme oxygenase-1 (HO-1) significantly reduce HMGB1 release in inflammatory conditions in vitro and in vivo. Thus, we tested our hypothesis that higenamine protects brain injury by inhibition of middle cerebral artery occlusion (MCAO)-mediated HMGB1 release in vivo, and glucose/glucose oxidase (GOX)-induced apoptosis in C6 cells in vitro due to HO-1 induction. Higenamine increased HO-1 expression in C6 cells in both hypoxia and normoxia, in which the former was much more significant than the latter. Higenamine increased Nrf-2 luciferase activity, translocated Nrf-2 to nucleus, and increased phosphorylation of Akt in C6 cells. Consistent with this, LY 294002, a PI3K inhibitor, inhibited HO-1 induction by higenamine and apoptosis induced by glucose/GOX in C6 cells was prevented by higenamine, which effect was reversed by LY 294002. Importantly, administration of higenamine (i.p) significantly reduced brain infarct size, mortality rate, MPO activity and tissue expression of HMGB1 in MCAO rats. In addition, recombinant high mobility group box 1 induced apoptosis in C6 cells by increasing ratio of Bax/bcl-2 and cleaved caspase c, which was inhibited by higenamine, and all of these effects were reversed by co-treatment with ZnPPIX. Therefore, we conclude that higenamine, at least in part, protects brain cells against hypoxic damages by up-regulation of HO-1. Thus, higenamine may be beneficial for the use of ischemic injuries such as stroke.


Asunto(s)
Alcaloides/farmacología , Inducción Enzimática/efectos de los fármacos , Proteína HMGB1/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Transducción de Señal , Tetrahidroisoquinolinas/farmacología , Alcaloides/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Hipoxia de la Célula , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cromonas/farmacología , Proteína HMGB1/farmacología , Hipoxia-Isquemia Encefálica/enzimología , Hipoxia-Isquemia Encefálica/patología , Infarto de la Arteria Cerebral Media , Masculino , Morfolinas/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Peroxidasa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Tetrahidroisoquinolinas/uso terapéutico
11.
J Pharmacol Exp Ther ; 341(3): 850-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22442118

RESUMEN

Salvia miltiorrhiza (Danshen), a traditional Chinese herbal medicine, is commonly used for the prevention and treatment of cardiovascular disorders including atherosclerosis. However, the mechanisms responsible for the vasoprotective effects of Danshen remain largely unknown. Salvianolic acid B (Sal B) represents one of the most bioactive compounds that can be extracted from the water-soluble fraction of Danshen. We investigated the effects of Danshen and Sal B on the inflammatory response in murine macrophages. Danshen and Sal B both induced the expression of heme oxygenase-1 (HO-1) and inhibited nitric oxide (NO) production and inducible NO synthase (iNOS) expression in lipopolysaccharide (LPS)-activated RAW 264.7 cells. Inhibition of HO activity using Sn-protoporphyrin-IX (SnPP) abolished the inhibitory effect of Sal B on NO production and iNOS expression. Sal B increased macrophage arginase activity in a dose-dependent manner and diminished LPS-inducible tumor necrosis factor-α production. These effects were also reversed by SnPP. These data suggest that HO-1 expression plays an intermediary role in the anti-inflammatory effects of Sal B. In contrast to the observations in macrophages, Sal B dose-dependently inhibited arginase activity in murine liver, kidney, and vascular tissue. Furthermore, Sal B increased NO production in isolated mouse aortas through the inhibition of arginase activity and reduction of reactive oxygen species production. We conclude that Sal B improves vascular function by inhibiting inflammatory responses and promoting endothelium-dependent vasodilation. Taken together, we suggest that Sal B may represent a potent candidate therapeutic for the treatment of cardiovascular diseases associated with endothelial dysfunction.


Asunto(s)
Benzofuranos/farmacología , Medicamentos Herbarios Chinos/farmacología , Hemo-Oxigenasa 1/metabolismo , Macrófagos/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico/metabolismo , Animales , Arginasa/metabolismo , Western Blotting , Ensayo de Cambio de Movilidad Electroforética , Fibrinolíticos/farmacología , Humanos , Lipopolisacáridos/farmacología , Macrófagos/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Fenantrolinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Salvia miltiorrhiza/química , Factor de Necrosis Tumoral alfa/metabolismo , Enfermedades Vasculares/prevención & control
12.
Can J Physiol Pharmacol ; 90(7): 863-72, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22702717

RESUMEN

Mepivacaine is an aminoamide-linked local anesthetic with an intermediate duration that intrinsically produces vasoconstriction both in vivo and in vitro. The aims of this in-vitro study were to examine the direct effect of mepivacaine in isolated rat aortic rings and to determine the associated cellular mechanism with a particular focus on endothelium-derived vasodilators, which modulate vascular tone. In the aortic rings with or without endothelium, cumulative mepivacaine concentration-response curves were generated in the presence or absence of the following antagonists: N(ω)-nitro-L-arginine methyl ester [L-NAME], indomethacin, fluconazole, methylene blue, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one [ODQ], verapamil, and calcium-free Krebs solution. Mepivacaine produced vasoconstriction at low concentrations (1 × 10(-3) and 3 × 10(-3) mol/L) followed by vasodilation at a high concentration (1 × 10(-2) mol/L). The mepivacaine-induced contraction was higher in endothelium-denuded aortae than in endothelium-intact aortae. Pretreatment with L-NAME, ODQ, and methylene blue enhanced mepivacaine-induced contraction in the endothelium-intact rings, whereas fluconazole had no effect. Indomethacin slightly attenuated mepivacaine-induced contraction, whereas verapamil and calcium-free Krebs solution more strongly attenuated this contraction. The vasoconstriction induced by mepivacaine is attenuated mainly by the endothelial nitric oxide - cyclic guanosine monophosphate pathway. In addition, mepivacaine-induced contraction involves cyclooxygenase pathway activation and extracellular calcium influx via voltage-operated calcium channels.


Asunto(s)
Anestésicos Locales/farmacología , Aorta/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Mepivacaína/farmacología , Músculo Liso Vascular/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Animales , Aorta/metabolismo , Calcio/metabolismo , Canales de Calcio/metabolismo , Células Cultivadas , GMP Cíclico/metabolismo , Endotelio Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Óxido Nítrico/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Ratas , Ratas Sprague-Dawley , Vasodilatación/efectos de los fármacos
13.
Phytother Res ; 26(4): 605-12, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21971692

RESUMEN

The ethanol extract of the flower of P. vulgaris var. lilacina (EEPV) has been used traditionally as an antiinflammatory agent in many countries. Inducers of heme oxygenase-1 (HO-1) reduce high mobility group box 1 (HMGB1), a late phase cytokine, in sepsis. Although EEPV has been used as an antiinflammatory agent, no report is available as to whether it modifies HMGB1 in sepsis due to HO-1 induction. It was found that EEPV increased HO-1 protein expression in RAW 264.7 cells, which was significantly inhibited by LY294002, but not PD98059, SB203580 or SP600125. In addition, EEPV activated NF-E2-related factor (Nrf2) to move from the cytosol to the nucleus, and EEPV-induced HO-1 and activation of ARE-luciferase activity were significantly reduced by siNrf2 transfection and LY294002 but not SB203508. EEPV also significantly inhibited NF-κB luciferase activity, and decreased both iNOS/NO and COX-2/PGE(2) production in lipopolysaccharide (LPS)-stimulated macrophages which was reversed by siHO-1 RNA transfection. Importantly, EEPV inhibited HMGB1 release in LPS-activated macrophages in a PI3K-sensitive manner and reduced serum HMGB1 level and lung HMGB1 expression in cecal ligation and puncture (CLP)-induced septic mice. It is concluded that EEPV induces HO-1 expression through PI3K/Nrf2 signal pathways, which may be beneficial for the treatment of sepsis due to a reduction of HMGB1 release.


Asunto(s)
Ciego/lesiones , Proteína HMGB1/antagonistas & inhibidores , Hemo-Oxigenasa 1/metabolismo , Proteínas de la Membrana/metabolismo , Extractos Vegetales/farmacología , Prunella/química , Animales , Antracenos/farmacología , Ciego/patología , Línea Celular , Supervivencia Celular , Cromonas/farmacología , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Activación Enzimática , Inducción Enzimática , Etanol , Flavonoides/farmacología , Flores/química , Vectores Genéticos , Proteína HMGB1/sangre , Hemo-Oxigenasa 1/antagonistas & inhibidores , Imidazoles/farmacología , Lipopolisacáridos/efectos adversos , Pulmón/patología , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , Ratones Endogámicos BALB C , Morfolinas/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/metabolismo , Transporte de Proteínas , Piridinas/farmacología , Sepsis/patología , Transducción de Señal , Transfección
14.
Microsurgery ; 32(7): 563-70, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22821773

RESUMEN

Ischemia-reperfusion injury is a phenomenon that occurs when tissues are subjected to ischemia for a variable period of time, and then reperfused. Inflammatory reaction has been implicated as one of the most important mechanism of ischemia-reperfusion injury. The purpose of this study was to evaluate the anti-inflammatory effects of anthocyanins from black soybean seed coat on keratinocytes in vitro and ischemia-reperfusion injury in vivo. We investigated the inhibition, by anthocyanins, of the expression of various inflammatory genes associated with ischemia-reperfusion injury in the tumor necrosis factor-alpha-treated (TNF-α) immortalized epidermal keratinocyte cell line (HaCaT). We also investigated the effects of anthocyanins on the survival of skin flaps after ischemia-reperfusion injury in the rats. According to Western blot analysis and a luciferase activity assay, anthocyanins inhibited TNF-α-induced intercellular adhesion molecule-1 and cyclooxygenase-2 (COX-2) levels through the NF-κB-dependent pathway. Administration of anthocyanins (50 and 100 mg/kg) significantly improved the flap area survival in the 10-hour ischemic model from 62% to 74.5% and 83%, respectively (P = 0.001). The related cytokines in skin flap also changed as the same pattern as in vitro. Our results indicate that anthocyanins from black soybean seed coat had anti-inflammatory effects on the HaCaT cell line and increase the survival of skin flaps through anti-inflammatory properties against ischemia-reperfusion injury.


Asunto(s)
Antocianinas/farmacología , Glycine max , Queratinocitos/efectos de los fármacos , Fitoterapia , Extractos Vegetales/farmacología , Daño por Reperfusión/prevención & control , Colgajos Quirúrgicos/irrigación sanguínea , Animales , Antocianinas/uso terapéutico , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Biomarcadores/metabolismo , Western Blotting , Línea Celular , Citocinas/metabolismo , Supervivencia de Injerto , Queratinocitos/metabolismo , Masculino , Extractos Vegetales/uso terapéutico , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Semillas
15.
Biochem Biophys Res Commun ; 406(4): 564-9, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21352808

RESUMEN

This study evaluated peroxisome proliferator-activated receptor (PPAR) δ as a potential target for therapeutic intervention in Ang II-induced senescence in human vascular smooth muscle cells (hVSMCs). Activation of PPARδ by GW501516, a specific agonist of PPARδ, significantly inhibited the Ang II-induced premature senescence of hVSMCs. Agonist-activated PPARδ suppressed the generation of Ang II-triggered reactive oxygen species (ROS) with a concomitant reduction in DNA damage. Notably, GW501516 up-regulated the expression of antioxidant genes, such as glutathione peroxidase 1, thioredoxin 1, manganese superoxide dismutase and heme oxygenase 1. siRNA-mediated down-regulation of these antioxidant genes almost completely abolished the effects of GW501516 on ROS production and premature senescence in hVSMCs treated with Ang II. Taken together, the enhanced transcription of antioxidant genes is responsible for the PPARδ-mediated inhibition of premature senescence through sequestration of ROS in hVSMCs treated with Ang II.


Asunto(s)
Senescencia Celular/genética , Regulación de la Expresión Génica , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/fisiología , PPAR delta/fisiología , Angiotensina II/farmacología , Angiotensina II/fisiología , Senescencia Celular/efectos de los fármacos , Daño del ADN/genética , Glutatión Peroxidasa/genética , Hemo-Oxigenasa 1/genética , Humanos , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , PPAR delta/agonistas , PPAR delta/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/genética , Tiazoles/farmacología , Tiorredoxinas/genética , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba , Glutatión Peroxidasa GPX1
16.
Circ Res ; 105(1): 16-24, 2009 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-19461048

RESUMEN

Homeostasis of the extracellular matrix and apoptosis of vascular smooth muscle cells (VSMCs) are key components in the regulation of the stability of atherosclerotic plaques. Here, we demonstrate that peroxisome proliferator-activated receptor (PPAR)delta regulates extracellular matrix synthesis and degradation through transforming growth factor-beta1 and its effector, Smad3. Activation of PPARdelta strongly amplified the expression of types I and III collagen, fibronectin, elastin, and TIMP-3 (tissue inhibitor of metalloproteinases 3), but not of TIMP-1, matrix metalloproteinase-2 or -9. The effect of PPARdelta on the expression of type III collagen was dually regulated by the direct binding of PPARdelta and Smad3 to a direct repeat-1 site and a Smad-binding element, respectively, in the type III collagen gene promoter. The activation of PPARdelta attenuated apoptotic cell death in VSMCs induced by oxidized low-density lipoprotein, and similar antiapoptotic effects were observed on treatment of cells with exogenous type I and/or III collagen. Administration of a PPARdelta ligand GW501516 to mice also suppressed elastase-induced cell death of aortic VSMCs. These results suggest that PPARdelta-induced upregulation of extracellular matrix proteins exerts an antiapoptotic effect, thereby maintaining the stability of atherosclerotic plaques. Specific ligands of PPARdelta may aid in the therapeutic intervention of atherosclerosis by improving plaque stability and patient prognosis.


Asunto(s)
Apoptosis , Matriz Extracelular/metabolismo , Miocitos del Músculo Liso/metabolismo , PPAR delta/fisiología , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Aterosclerosis/patología , Células Cultivadas , Proteínas de la Matriz Extracelular/biosíntesis , Homeostasis , Humanos , Ligandos , Ratones , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , PPAR delta/metabolismo
17.
Can J Physiol Pharmacol ; 89(9): 681-9, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21861649

RESUMEN

Dexmedetomidine, a full agonist of the α2B-adrenoceptor that is mainly involved in vascular smooth muscle contraction, is primarily used for analgesia and sedation in intensive care units. High-dose dexmedetomidine produces hypertension in children and adults. The goal of this in vitro study was to investigate the role of the calcium (Ca(2+)) sensitization mechanism involving Rho-kinase, protein kinase C (PKC), and phosphoinositide 3-kinase (PI3-K) in mediating contraction of isolated rat aortic smooth muscle in response to dexmedetomidine. The effect of dexmedetomidine on the intracellular Ca(2+) level ([Ca(2+)]i) and tension was measured simultaneously. Dexmedetomidine concentration-response curves were generated in the presence or absence of the following antagonists: rauwolscine, Y 27632, LY 294002, GF 109203X, and verapamil. Dexmedetomidine-induced phosphorylation of PKC and membrane translocation of Rho-kinase were detected with Western blotting. Rauwolscine, Y 27632, GF 109203X, LY 294002, and verapamil attenuated dexmedetomidine-induced contraction. The slope of the [Ca(2+)]i-tension curve for dexmedetomidine was higher than that for KCl. Dexmedetomidine induced phosphorylation of PKC and membrane translocation of Rho-kinase. These results suggest that dexmedetomidine-induced contraction involves a Ca(2+) sensitization mechanism mediated by Rho-kinase, PKC, and PI3-K that is secondary to α2-adrenoceptor stimulation in rat aortic smooth muscle.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/metabolismo , Aorta/efectos de los fármacos , Calcio/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Dexmedetomidina/metabolismo , Dexmedetomidina/farmacología , Animales , Aorta/metabolismo , Calcio/farmacocinética , Dexmedetomidina/agonistas , Técnicas In Vitro , Masculino , Músculo Liso Vascular/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Cloruro de Potasio/farmacología , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos/metabolismo , Quinasas Asociadas a rho/metabolismo
18.
J Cell Mol Med ; 14(6B): 1747-59, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19538467

RESUMEN

Although the peroxisome proliferator-activated receptor (PPAR) delta has been implicated in the wound healing process, its exact role and mechanism of action have not been fully elucidated. Our previous findings showed that PPARdelta induces the expression of the transforming growth factor (TGF)-beta1, which has been implicated in the deposit of extracellular matrix proteins. Here, we demonstrate that administration of GW501516, a specific PPARdelta ligand, significantly promoted wound closure in the experimental mouse and had a profound effect on the expression of collagen types I and III, alpha-smooth muscle actin, pSmad3 and TGF-beta1, which play a pivotal role in wound healing processes. Activation of PPARdelta increased migration of human epidermal keratinocytes and dermal fibroblasts in in vitro scrape-wounding assays. Addition of a specific ALK5 receptor inhibitor SB431542 significantly suppressed GW501516-induced migration of human keratinocytes and fibroblasts. In these cells, activated PPARdelta also induced the expression of collagen types I and III and fibronectin in a TGF-beta1-dependent or -independent manner. The effect of PPARdelta on the expression of type III collagen was dually regulated by the direct binding of PPARdelta and Smad3 to a direct repeat-1 site and a Smad-binding element, respectively, of the type III gene promoter. Taken together, these results demonstrated that PPARdelta plays an important role in skin wound healing in vivo and that it functions by accelerating extracellular matrix-mediated cellular interactions in a process mediated by the TGF-beta1/Smad3 signaling-dependent or - independent pathway.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , PPAR delta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba/genética , Cicatrización de Heridas/genética , Animales , Movimiento Celular , Células Cultivadas , Colágeno Tipo III/genética , Colágeno Tipo III/metabolismo , Dermis/citología , Células Epidérmicas , Proteínas de la Matriz Extracelular/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Ligandos , Ratones , PPAR alfa/metabolismo , PPAR gamma/metabolismo , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Elementos de Respuesta/genética , Transducción de Señal , Proteína smad3/metabolismo , Transcripción Genética , Factor de Crecimiento Transformador beta1/genética
19.
Nitric Oxide ; 23(4): 251-7, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20713168

RESUMEN

Beyond its vasodilator role, vascular nitric oxide (NO), which is synthesized by endothelial NO synthase (eNOS) via its activation, has been shown to play a number of other beneficial roles in the vascular system; it inhibits proliferation of vascular smooth muscle cells, prevents platelet aggregation, and regulates endothelial apoptosis. Such beneficial roles have been shown to be implicated in the regulation of endothelial functions. A loss of NO bioavailability that may result either from decreased eNOS expression and activity or from increased NO degradation is associated with endothelial dysfunction, a key factor in the development of vascular diseases. Heme oxygenase-1 (HO-1), an inducible enzyme, catalyzes the oxidative degradation of heme to free iron, carbon monoxide, and biliverdin, the latter being subsequently converted into bilirubin. In the vascular system, HO-1 and heme degradation products perform important physiological functions, which are ultimately linked to the protection of vascular cells. Studies have shown that HO-1 and heme degradation products exert vasodilatory, antioxidant, anti-inflammatory, antiproliferative and anti-apoptotic effects on vascular cells. Interestingly, these effects of HO-1 and its by-products are similar, at least in part, to those of eNOS-derived NO; this similarity may prompt investigators to study a possible relationship between eNOS-derived NO and HO-1 pathways. Many studies have been reported, and accumulating evidence suggests that HO-1 and heme degradation products can improve vascular function, at least in part, by compensating for the loss of NO bioavailability. This paper will provide the possible pathway explaining how HO-1 and heme degradation products can preserve vascular NO.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Músculo Liso Vascular/metabolismo , Óxido Nítrico/metabolismo , Animales , Hemo-Oxigenasa 1/genética , Humanos , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteínas Quinasas/metabolismo , Factores de Transcripción/metabolismo
20.
Circ Res ; 102(2): 193-200, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18007025

RESUMEN

The peroxisome proliferator-activated receptor (PPAR)delta has been implicated in the pathogenesis of atherogenic disorders. However, its physiological roles and functions in vascular smooth muscle cells (VSMCs) remain relatively unclear. In the present study, we show that the gene encoding transforming growth factor (TGF)-beta1 is a PPARdelta target in VSMCs. The PPARdelta activator GW501516 upregulates TGF-beta1 expression in a dose- and time-dependent manner. This induction is attenuated significantly by the presence of small interfering RNA against PPARdelta or GW9662, an inhibitor of PPARdelta. Furthermore, activated PPARdelta induces TGF-beta1 promoter activity by binding to the direct repeat-1 response element TGF-beta1-direct repeat-1. Mutations in the 5' or 3' half-sites of the response element totally abrogate transcriptional activation and PPARdelta binding, which suggests that this site is a novel type of PPARdelta response element. In addition, ligand-activated PPARdelta attenuated the promoter activity and expression of monocyte chemoattractant protein-1 induced by interleukin-1beta. These effects were significantly reduced in the presence of small interfering RNA against PPARdelta, anti-TGF-beta1 antibody, or a TGF-beta type I receptor inhibitor. Decreased monocyte chemoattractant protein-1 expression induced by PPARdelta was mediated by the effector of TGF-beta1, Smad3. Finally, administration of GW501516 to mice upregulated TGF-beta1, whereas the expression of proinflammatory genes including monocyte chemoattractant protein-1 was significantly attenuated in the thoracic aorta. Taken together, these results demonstrate the presence of a novel TGF-beta1-mediated pathway in the antiinflammatory activities of PPARdelta.


Asunto(s)
PPAR delta/metabolismo , Factor de Crecimiento Transformador beta1/genética , Animales , Aterosclerosis/patología , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación , Ligandos , Ratones , Músculo Liso Vascular/citología , PPAR delta/fisiología , ARN Mensajero , Ratas , Tiazoles/administración & dosificación , Tiazoles/farmacología , Factor de Crecimiento Transformador beta1/análisis , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA