Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 38(1): e23362, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38102979

RESUMEN

Endothelial dysfunction (ED) is commonly considered a crucial initiating step in the pathogenesis of numerous cardiovascular diseases. The coupling of endothelial nitric oxide synthase (eNOS) is important in maintaining normal endothelial functions. However, it still remains elusive whether and how eNOS SUMOylation affects the eNOS coupling. In the study, we investigate the roles and possible action mechanisms of protein inhibitor of activated STAT 1 (PIAS1) in ED. Human umbilical vein endothelial cells (HUVECs) treated with palmitate acid (PA) in vitro and ApoE-/- mice fed with high-fat diet (HFD) in vivo were constructed as the ED models. Our in vivo data show that PIAS1 alleviates the dysfunction of vascular endothelium by increasing nitric oxide (NO) level, reducing malondialdehyde (MDA) level, and activating the phosphatidylinositol 3-kinase-protein kinase B-endothelial nitric oxide synthase (PI3K-AKT-eNOS) signaling in ApoE-/- mice. Our in vitro data also show that PIAS1 can SUMOylate eNOS under endogenous conditions; moreover, it antagonizes the eNOS uncoupling induced by PA. The findings demonstrate that PIAS1 alleviates the dysfunction of vascular endothelium by promoting the SUMOylation and inhibiting the uncoupling of eNOS, suggesting that PIAS1 would become an early predictor of atherosclerosis and a new potential target of the hyperlipidemia-related cardiovascular diseases.


Asunto(s)
Homeostasis , Animales , Humanos , Ratones , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Enfermedades Cardiovasculares/metabolismo , Endotelio Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Inhibidoras de STAT Activados/genética , Proteínas Inhibidoras de STAT Activados/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Sumoilación
2.
Int J Obes (Lond) ; 48(7): 913-922, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38374247

RESUMEN

BACKGROUND: Asprosin (ASP) is a newly discovered adipokine secreted by white adipose tissue (WAT), which can regulate the homeostasis of glucose and lipid metabolism. However, it is not clear whether it can regulate the browning of WAT and mitophagy during the browning process. Accordingly, this study aims to investigate the effects and possible mechanisms of ASP on the browning of WAT and mitophagy in vivo and in vitro. METHODS: In in vivo experiments, some mouse models were used including adipose tissue ASP-specific deficiency (ASP-/-), high fat diet (HFD)-induced obesity and white adipose browning; in in vitro experiments, some cell models were also established and used, including ASP-deficient 3T3-L1 preadipocyte (ASP-/-) and CL-316243 (CL, 1 µM)-induced browning. Based on these models, the browning of WAT and mitophagy were evaluated by morphology, functionality and molecular markers. RESULTS: Our in vivo data show that adipose tissue-specific deletion of ASP contributes to weight loss in mice; supplementation of ASP inhibits the expressions of browning-related proteins including UCP1, PRDM16 and PGC1ɑ during the cold exposure-induced browning, and promotes the expressions of mitophagy-related proteins including PINK1 and Parkin under the conditions of whether normal diet (ND) or HFD. Similarly, our in vitro data also show that the deletion of ASP in 3T3-L1 cells significantly increases the expressions of the browning-related proteins and decreases the expressions of the mitophagy-related proteins. CONCLUSIONS: These data demonstrate that ASP deletion can facilitate the browning and inhibit mitophagy in WAT. The findings will lay an experimental foundation for the development of new drugs targeting ASP and the clinical treatment of metabolic diseases related to obesity.


Asunto(s)
Tejido Adiposo Pardo , Tejido Adiposo Blanco , Mitofagia , Obesidad , Animales , Ratones , Mitofagia/fisiología , Obesidad/metabolismo , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Pardo/metabolismo , Dieta Alta en Grasa , Masculino , Células 3T3-L1 , Ratones Endogámicos C57BL , Adipocitos/metabolismo , Modelos Animales de Enfermedad
3.
Drug Dev Res ; 85(4): e22213, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38798186

RESUMEN

Asprosin (ASP) is a newly-identified adipokine and plays important roles in energy metabolism homeostasis. However, there is no report on whether and how ASP is involved in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). Therefore, in the study, we investigated the protective effects of ASP-deficiency on the liver in the NAFLD model mice and the detrimental effects of ASP treatment on the human normal hepatocytes (LO2 cell line). More important, we explored the underlying mechanism from the perspective of lipid metabolism and inflammation. In the in vivo experiments, our data showed that the ASP-deficiency significantly alleviated the high-fat diet-induced inflammation and NAFLD, inhibited the hepatic fat deposition and downregulated the expressions of fat acid synthase (FASN), peroxisome proliferator-activated receptor γ (PPARγ) and forkhead box protein O1 (FOXO1); moreover, the ASP-deficiency attenuated the inflammatory state and inhibited the activation of the IKK/NF-κBp65 inflammation pathway. In the in vitro experiments, our results revealed that ASP treatment caused and even exacerbated the injury of LO2 cells induced by FFA; In contrast, the ASP treatment upregulated the expressions of PPARγ, FOXO1, FASN, ACC and acyl-CoA oxidase 1 (ACOX1) and elevated the reactive oxygen species (ROS) levels. Accordingly, these results demonstrate that ASP causes NAFLD through disrupting lipid metabolism and promoting the inflammation mediated by ROS.


Asunto(s)
Dieta Alta en Grasa , Fibrilina-1 , Inflamación , Metabolismo de los Lípidos , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico , Especies Reactivas de Oxígeno , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Humanos , Ratones , Inflamación/metabolismo , Masculino , Dieta Alta en Grasa/efectos adversos , Línea Celular , PPAR gamma/metabolismo , Hepatocitos/metabolismo , Hepatocitos/efectos de los fármacos , Modelos Animales de Enfermedad , Hígado/metabolismo , Hígado/patología , Adipoquinas
4.
Int J Mol Sci ; 23(24)2022 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-36555807

RESUMEN

Asprosin (ASP) is a recently identified adipokine secreted by white adipose tissue (WAT). It plays important roles in the maintenance of glucose homeostasis in the fasting state and in the occurrence and development of obesity. However, there is no report on whether and how ASP would inhibit angiogenesis and fat browning in the mouse adipose microenvironment. Therefore, the study sought to investigate the effects of ASP-knockout on angiogenesis and fat browning, and to identify the interaction between them in the ASP-knockout mouse adipose microenvironment. In the experiments in vivo, the ASP-knockout alleviated the obesity induced by a high fat diet (HFD) and increased the expressions of the browning-related proteins including uncoupling protein 1 (UCP1), PRD1-BF-1-RIZ1 homologus domain-containing protein-16 (PRDM16) and PPAR gamma coactivator 1 (PGC1-α) and the endothelial cell marker (CD31). In the experiments in vitro, treatment with the conditional medium (CM) from ASP-knockout adipocytes (ASP-/--CM) significantly promoted the proliferation, migration and angiogenesis of vascular endothelial cells, and increased the expressions of vascular endothelial growth factor (VEGF)/vascular endothelial growth factor receptor 2 (VEGFR2) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/endothelial nitric oxide synthase (eNOS) pathway proteins. In addition, the treatment with CM from endothelial cells (EC-CM) markedly reduced the accumulation of lipid droplets and increased the expressions of the browning-related proteins and the mitochondrial contents. Moreover, the treatment with EC-CM significantly improved the energy metabolism in 3T3-L1 adipocytes. These results highlight that ASP-knockout can promote the browning and angiogenesis of WAT, and the fat browning and angiogenesis can interact in the mouse adipose microenvironment, which contributes to weight loss in the mice with obesity.


Asunto(s)
Células Endoteliales , Fosfatidilinositol 3-Quinasas , Ratones , Animales , Células Endoteliales/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ratones Noqueados , Obesidad/genética , Obesidad/metabolismo , Tejido Adiposo Blanco/metabolismo , Pérdida de Peso , Tejido Adiposo Pardo/metabolismo , Dieta Alta en Grasa/efectos adversos , Ratones Endogámicos C57BL , Células 3T3-L1
5.
J Cell Mol Med ; 23(1): 216-226, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30398029

RESUMEN

Previous study has shown that thiazolidinediones (TZDs) improved endothelium insulin resistance (IR) induced by high glucose concentration (HG)/hyperglycaemia through a PPARγ-dependent-NFκB trans-repression mechanism. However, it is unclear, whether changes in PPARγ expression affect the endothelium IR and what the underlying mechanism is. In the present study, we aimed to address this issue. HG-treated human umbilical vascular endothelial cells (HUVEC) were transfected by either PPARγ-overexpressing (Ad-PPARγ) or PPARγ-shRNA-containing (Ad-PPARγ-shRNA) adenoviral vectors. Likewise, the rats fed by high-fat diet (HFD) were infected by intravenous administration of Ad-PPARγ or Ad-PPARγ-shRNA. The levels of nitric oxide (NO), endothelin-1 (ET-1) and cytokines (TNFα, IL-6, sICAM-1 and sVCAM-1) and the expression levels of PPARγ, eNOS, AKT, p-AKT, IKKα/ß and p-IKKα/ß and IκBα were examined; and the interaction between PPARγ and NFκB-P65 as well as vascular function were evaluated. Our present results showed that overexpression of PPARγ notably increased the levels of NO, eNOS, p-AKT and IκBα as well as the interaction of PPARγ and NFκB-P65, and decreased the levels of ET-1, p-IKKα/ß, TNFα, IL-6, sICAM-1 and sVCAM-1. In contrast, down-expression of PPARγ displayed the opposite effects. The results demonstrate that the overexpression of PPARγ improves while the down-expression worsens the endothelium IR via a PPARγ-mediated NFκB trans-repression dependent manner. The findings suggest PPARγ is a potential therapeutic target for diabetic vascular complications.


Asunto(s)
Endotelio Vascular/fisiología , Resistencia a la Insulina/fisiología , FN-kappa B/metabolismo , PPAR gamma/metabolismo , Células 3T3-L1 , Animales , Citocinas/metabolismo , Glucosa/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Óxido Nítrico Sintasa de Tipo III/metabolismo , PPAR gamma/genética , Ratas Sprague-Dawley , Vasodilatación/fisiología
6.
J Cell Physiol ; 234(11): 19663-19674, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30982983

RESUMEN

Sumoylation of peroxisome proliferator-activated receptor Î³ (PPARγ) affects its stabilization, sublocalization, and transcriptional activity. However, it remains largely unknown whether PPARγ sumoylation inhibits the transactivation effect, leading to endothelium insulin resistance (IR). To test this possibility, human umbilical vascular endothelial cells (HUVECs) with a 90% confluence were randomly allocated to two batches. One batch was first pretreated with or without vitamin E for 24 hr and the other infected with adenoviruses containing either PIAS1-shRNA (protein inhibitor of activated STAT1-short hairpin RNA) or scramble shRNA. Cells were suffered from high glucose and palmitic acid (PA) exposure for further 48 hr. The levels of PPARγ, p-IKK, IKK, and NcoR (nuclear corepressors) were measured by western blot analysis. The interaction of IKK and PIAS1, as well as the PPARγ sumoylation, were examined by coimmunoprecipitation. The results showed that the exposure of high glucose and PA induced reactive oxygen species (ROS) production and IKK activation in HUVECs, promoting the interaction of IKK and PIAS1 and the sumoylation of PPARγ. However, vitamin E and PIAS1-shRNA partially decreased ROS production and IKK activation induced by high glucose and PA exposure. These data indicate that ROS-IKK-PIAS1 pathway mediates PPARγ sumoylation, leading to endothelium IR via stabilizing PPARγ-NcoR complex. These findings benefit understanding of regulatory networks of insulin signaling, which might provide a potential target to prevent and cure IR-related diseases.


Asunto(s)
Resistencia a la Insulina/genética , Insulina/genética , Co-Represor 1 de Receptor Nuclear/genética , PPAR gamma/genética , Factor de Transcripción STAT1/genética , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Redes Reguladoras de Genes/efectos de los fármacos , Glucosa/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Insulina/metabolismo , Complejos Multiproteicos/genética , PPAR gamma/antagonistas & inhibidores , Ácido Palmítico/farmacología , Proteínas Inhibidoras de STAT Activados/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Sumoilación/genética , Vitamina E/farmacología
7.
Horm Metab Res ; 51(11): 741-748, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31295749

RESUMEN

Osteopontin (OPN), a secreted glycoprotein, is involved in various pathophysiological processes including immune response, inflammation, tumor formation, and metabolism. OPN exists in 2 forms, secreted-OPN (sOPN) and intracellular-OPN (iOPN). While they might have different biological activities, it remains largely unknown whether sOPN and iOPN induce the differentiation of brown adipocytes. To test this possibility, 3T3-L1 cells were induced by DMI induction with or without recombinant human OPN (rhOPN, 10, 50, 100, 200 µM), respectively. Meanwhile, another batch of 3T3-L1 cells were infected with Ad-GFP-ap2-OPN and followed by DMI differentiation. Subsequently, the infected cells were treated with either anti-CD44 antibody or immunoglobulin G (Ig G). Accumulation of lipid droplets was visualized by Oil red O staining and protein levels were assayed by western blotting analysis. The results showed that sOPN and not rhOPN, notably increased the accumulation of lipid droplets and the expression of brown adipocyte-related genes. Moreover, neutralization of CD44 partially abrogated the effects induced by sOPN. These data demonstrate that sOPN and not rhOPN has the capacity to induce the differentiation of white preadipocytes into brown adipocytes through a CD44-dependent mechanism. The findings might provide a potential target for sOPN to combat obesity.


Asunto(s)
Adipogénesis , Tejido Adiposo Pardo/citología , Diferenciación Celular , Receptores de Hialuranos/metabolismo , Osteopontina/metabolismo , Células 3T3-L1 , Tejido Adiposo Pardo/metabolismo , Animales , Técnicas In Vitro , Ratones
8.
Can J Physiol Pharmacol ; 95(12): 1480-1487, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28787583

RESUMEN

PIO, a synthetic ligand for PPARγ, is used clinically to treat T2DM. However, little is known about its protective effects on endothelium and the underlying mechanisms. In this study, we sought to investigate the protective effects of PIO on endothelium and its probable mechanisms: 95% confluent wild type (WT) HUVECs and PPARγLow-HUVECs that we first injured with HG (33 mmol·L-1) were first pretreated with 10 µmol·L-1 of GW9662 for 30 min, and then treated the cells with different concentrations of PIO (5, 10, or 20 µmol·L-1) for 24 h. Finally, we measured the levels of NO, ET1, TNFα, and IL6 in the cell culture supernatant. These cells were then used to determine cell viability, caspase3 activity, the levels of IKKα/ß mRNA, IKKα/ß, and NFκB-p65. Severe dysfunction and activation of IKKα/ß-NFκB signaling occurred after we exposed HUVECs to HG. Conversely, treatment with PIO significantly attenuated the dysfunction and the activation of IKKα/ß-NFκB signaling induced by HG in a dose-dependent manner. Moreover, the protective effects of PIO were completely abrogated by GW9662 or down-regulation of PPARγ. Taken together, the results indicate that PIO protects HUVECs against the HG-induced dysfunction through the inhibition of IKKα/ß-NFκB signaling mediated by PPARγ.


Asunto(s)
Citoprotección/efectos de los fármacos , Glucosa/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Quinasa I-kappa B/metabolismo , PPAR gamma/metabolismo , Tiazolidinedionas/farmacología , Factor de Transcripción ReIA/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Caspasa 3/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Quinasa I-kappa B/genética , Pioglitazona , Transducción de Señal/efectos de los fármacos
9.
J Cardiovasc Pharmacol ; 67(6): 519-25, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26859198

RESUMEN

Adhesion of monocytes to the vascular endothelium is crucial in atherosclerosis development. Connexins (Cxs) which form hemichannels or gap junctions, modulate monocyte-endothelium interaction. We previously found that rutaecarpine, an active ingredient of the Chinese herbal medicine Evodia, reversed the altered Cx expression induced by oxidized low-density lipoprotein (ox-LDL) in human umbilical vein endothelial cells, and consequently decreases the adhesive properties of endothelial cells to monocytes. This study further investigated the effect of rutaecarpine on Cx expression in monocytes exposed to ox-LDL. In cultured human monocytic cell line THP-1, ox-LDL rapidly reduced the level of atheroprotective Cx37 but enhanced that of atherogenic Cx43, thereby inhibiting adenosine triphosphate release through hemichannels. Pretreatment with rutaecarpine recovered the expression of Cx37 but inhibited the upregulation of Cx43 induced by ox-LDL, thereby improving adenosine triphosphate-dependent hemichannel activity and preventing monocyte adhesion. These effects of rutaecarpine were attenuated by capsazepine, an antagonist of transient receptor potential vanilloid subtype 1. The antiadhesive effects of rutaecarpine were also attenuated by hemichannel blocker 18α-GA. This study provides additional evidence that rutaecarpine can modulate Cx expression through transient receptor potential vanilloid subtype 1 activation in monocytes, which contributes to the antiadhesive properties of rutaecarpine.


Asunto(s)
Conexinas/efectos de los fármacos , Endotelio Vascular/metabolismo , Alcaloides Indólicos/farmacología , Lipoproteínas LDL/metabolismo , Monocitos/metabolismo , Quinazolinas/farmacología , Adenosina Trifosfato/metabolismo , Aterosclerosis/fisiopatología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Factores de Tiempo
10.
Biochem Biophys Res Commun ; 459(3): 553-9, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25749339

RESUMEN

Recent studies have shown that OPN (osteopontin) plays critical roles in cell survival, differentiation, bio-mineralization, cancer and cardiovascular remodeling. However, its roles in the differentiation of brown adipocytes and the underlying mechanisms remain unclear. Therefore, the aim of this study was to investigate the roles of OPN in the brown adipogenesis and the underlying mechanisms. It was shown that the OPN successfully induced the differentiation of 3T3-L1 white preadipocytes into the PRDM16(+) (PRD1-BF1-RIZ1 homologous domain containing 16) and UCP-1(+) (uncoupling protein-1) brown adipocytes in a concentration and time-dependent manner. Also, activation of PI3K (phosphatidylinositol 3-kinase)-AKT pathway was required for the OPN-induced brown adipogenesis. The findings suggest OPN plays an important role in promoting the differentiation of the brown adipocytes and might provide a potential novel therapeutic approach for the treatment of obesity and related disorders.


Asunto(s)
Adipocitos Blancos/citología , Adipocitos Blancos/metabolismo , Adipogénesis/fisiología , Osteopontina/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células 3T3-L1 , Adipogénesis/genética , Animales , Diferenciación Celular , Integrina alfaVbeta3/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Osteopontina/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal
11.
J Cardiovasc Pharmacol ; 66(2): 148-58, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25915512

RESUMEN

We have recently shown that DJ-1 is implicated in the delayed cardioprotective effect of hypoxic preconditioning (HPC) against hypoxia/reoxygenation (H/R) injury as an endogenous protective protein. This study aims to further investigate the underlying mechanism by which DJ-1 mediates the delayed cardioprotection of HPC against H/R-induced oxidative stress. Using a well-characterized cellular model of HPC from rat heart-derived H9c2 cells, we found that HPC promoted nuclear factor erythroid 2-related factor 2 (Nrf2) and its cytoplasmic inhibitor Kelch-like ECH-associated protein-1 (Keap1) dissociation and resulted in increased nuclear translocation, antioxidant response element-binding, and transcriptional activity of Nrf2 24 hours after HPC, with subsequent upregulation of manganese superoxide dismutase (MnSOD) and heme oxygenase-1 (HO-1), which provided delayed protection against H/R-induced oxidative stress in normal H9c2 cells. However, the aforementioned effects of HPC were abolished in DJ-1-knockdown H9c2 cells, which were restored by restoration of DJ-1 expression. Importantly, we showed that inhibition of the Nrf2 pathway in H9c2 cells mimicked the effects of DJ-1 knockdown and abolished HPC-derived induction of antioxidative enzymes (MnSOD and HO-1) and the delayed cardioprotection. In addition, inhibition of Nrf2 also reversed the effects of restored DJ-1 expression on induction of antioxidative enzymes and delayed cardioprotection by HPC in DJ-1-knockdown H9c2 cells. Taken together, this work revealed that activation of Nrf2 pathway and subsequent upregulation of antioxidative enzymes could be a critical mechanism by which DJ-1 mediates the delayed cardioprotection of HPC against H/R-induced oxidative stress in H9c2 cells.


Asunto(s)
Antioxidantes/metabolismo , Proteínas Asociadas a Microtúbulos/fisiología , Miocitos Cardíacos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Regulación hacia Arriba/fisiología , Animales , Hipoxia de la Célula/fisiología , Línea Celular , Técnicas de Silenciamiento del Gen/métodos , Humanos , Precondicionamiento Isquémico Miocárdico/métodos , Proteína Desglicasa DJ-1 , Ratas , Transducción de Señal/fisiología
12.
Mol Cell Biochem ; 385(1-2): 33-41, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24048861

RESUMEN

It has been well demonstrated that hypoxic preconditioning (HPC) can attenuate hypoxia/reoxygenation (H/R)-induced oxidant stress and elicit delayed cardioprotection by upregulating the expression of multiple antioxidative enzymes such as heme oxygenase-1 (HO-1), manganese superoxide dismutase (MnSOD) and so on. However, the underlying mechanisms of HPC-induced upregulation of antioxidative enzymes are not fully understood. Nuclear factor erythroid 2-related factor 2 (Nrf2) is an essential transcription factor that regulates expression of several antioxidant genes via binding to the antioxidant response element (ARE) and plays a crucial role in cellular defence against oxidative stress. Here, we wondered whether activation of the Nrf2-ARE pathway is responsible for the induction of antioxidative enzymes by HPC and contributes to the delayed cardioprotection of HPC. Cellular model of HPC from rat heart-derived H9c2 cells was induced 24 h prior to H/R. The results showed that HPC efficiently attenuated H/R-induced viability loss and lactate dehydrogenase leakage. In addition, HPC increased nuclear translocation and ARE binding of Nrf2 during the late phase, upregulated the expression of antioxidative enzymes (HO-1 and MnSOD), inhibited H/R-induced oxidant stress. However, when Nrf2 was specifically knocked down by siRNA, the induction of antioxidative enzymes by HPC was completely abolished and, as a result, the inhibitory effect of HPC on H/R-induced oxidant stress was reversed, and the delayed cardioprotection induced by HPC was also abolished. These results suggest that HPC upregulates antioxidative enzymes through activating the Nrf2-ARE pathway and confers delayed cardioprotection against H/R-induced oxidative stress.


Asunto(s)
Antioxidantes/metabolismo , Cardiotónicos/metabolismo , Precondicionamiento Isquémico Miocárdico , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Factor 2 Relacionado con NF-E2/metabolismo , Regulación hacia Arriba , Animales , Elementos de Respuesta Antioxidante/genética , Hipoxia de la Célula , Línea Celular , Núcleo Celular/metabolismo , Técnicas de Silenciamiento del Gen , Unión Proteica , Transporte de Proteínas , Ratas , Transducción de Señal , Estrés Fisiológico
13.
Cell Biochem Funct ; 31(8): 643-51, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23281015

RESUMEN

It has been well accepted that increased reactive oxygen species (ROS) and the subsequent oxidative stress is one of the major causes of ischemia/reperfusion (I/R) injury. DJ-1 protein, as a multifunctional intracellular protein, plays an important role in regulating cell survival and antioxidant stress. Here, we wondered whether DJ-1 overexpression attenuates simulated ischemia/reperfusion (sI/R)-induced oxidative stress. A rat cDNA encoding DJ-1 was inserted into a mammalian expression vector. After introduction of this construct into H9c2 myocytes, stable clones were obtained. Western blot analysis of the derived clones showed a 2.6-fold increase in DJ-1 protein expressing. Subsequently, the DJ-1 gene-transfected and control H9c2 cells were subjected to sI/R, and then cell viability, lactate dehydrogenase, malondialdehyde, intracellular ROS and antioxidant enzymes (superoxide dismutase, catalase and glutathione peroxidase) were measured appropriately. The results showed that stable overexpression of DJ-1 efficiently attenuated sI/R-induced viability loss and lactate dehydrogenase leakage. Additionally, stable overexpression of DJ-1 inhibited sI/R-induced the elevation of ROS and MDA contents followed by the increase of antioxidant enzymes (superoxide dismutase, catalase and glutathione peroxidase) activities and expression. Our data indicate that overexpression of DJ-1 attenuates ROS generation, enhances the cellular antioxidant capacity and prevents sI/R-induced oxidative stress, revealing a novel mechanism of cardioprotection. Importantly, DJ-1 overexpression may be an important part of a protective strategy against ischemia/reperfusion injury.


Asunto(s)
Hipoxia/genética , Hipoxia/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Estrés Oxidativo/genética , Animales , Células Cultivadas , Peroxirredoxinas , Proteína Desglicasa DJ-1 , Ratas
14.
Inflammation ; 46(2): 623-638, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36401667

RESUMEN

Vascular endothelium dysfunction caused by endothelium inflammation is a trigger of numerous cardiovascular diseases. Vascular endothelium inflammation often occurs in patients with obesity. Asprosin (ASP) derived from white adipose tissue plays important roles in maintaining glucose homeostasis. However, effect of ASP on the vascular endothelium inflammation induced by hyperlipidemia and its underlying mechanism remains largely unclear. In this study, models of vascular endothelium inflammation were established to investigate the effect of ASP on the endothelium inflammation both in vivo and in vitro. Our data in vivo showed that recombinant ASP or high-fat diet (HFD) significantly increased the circulating levels of IL-6 and TNF-α and enhanced the adhesion of macrophages to endothelia characterized by the expression increase of CD68, ICAM-1, and VCAM-1 in rats. However, neutralization of ASP with an ASP specific antibody (AASP) significantly antagonized the changes induced by HFD. Similarly, our data in vitro also showed that ASP treatment elevated the expressions of IL-6, TNF-α, and ICAM-1 as well as VCAM-1. More important, our data revealed that the pro-inflammation effect of ASP was achieved by activating the IKKß-NF-κBp65 pathway other than the oxidative stress pathway both in vivo and in vitro. In conclusion, our results demonstrate that ASP is a pro-inflammation player in the obesity-associated endothelium dysfunction. The findings would provide a novel target for the prevention and treatment of obesity-related cardiovascular diseases.


Asunto(s)
Enfermedades Cardiovasculares , Hiperlipidemias , Ratas , Animales , Quinasa I-kappa B/metabolismo , Transducción de Señal , Molécula 1 de Adhesión Intercelular/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-6/metabolismo , Hiperlipidemias/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Inflamación/metabolismo , Endotelio Vascular/metabolismo , Obesidad/metabolismo , FN-kappa B/metabolismo
15.
Mol Nutr Food Res ; 67(18): e2200763, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37436078

RESUMEN

SCOPE: Apigenin (AP) has many pharmacological activities, including anti-inflammation, hyperlipidemia-lowering, and so on. Previous studies show that AP can reduce lipid accumulation in adipocytes in vitro. However, it remains unclear whether and how AP can promote fat-browning. Therefore, mouse obesity model and preadipocyte induction model in vitro are used to investigate the effects of AP on glycolipid metabolism, browning and autophagy as well as the possible mechanisms. METHODS AND RESULTS: The obese mice are intragastrically administrated with AP (0.1 mg g-1  d-1 ) for 4 weeks; meanwhile, the differentiating preadipocytes are respectively treated with the indicated concentrations of AP for 48 h. Metabolic phenotype, lipid accumulation, and fat-browning are respectively evaluated by morphological, functional, and specific markers analysis. The results show that AP treatment alleviates the body weight, glycolipid metabolic disorder, and insulin resistance in the obese mice , which is contributed to the pro-browning effects of AP in vivo and in vitro. Moreover, the study finds that the pro-browning effect of AP is accomplished through autophagy inhibition mediated by the activation of PI3K-Akt-mTOR pathway. CONCLUSIONS: The findings highlight that autophagy inhibition promotes the browning of white adipocytes and suggest that AP would prevent and treat obesity and the associated metabolic disorders.


Asunto(s)
Apigenina , Fosfatidilinositol 3-Quinasas , Animales , Ratones , Apigenina/farmacología , Ratones Obesos , Fosfatidilinositol 3-Quinasas/metabolismo , Obesidad/metabolismo , Peso Corporal , Adipocitos Blancos/metabolismo , Dieta Alta en Grasa , Autofagia , Lípidos/farmacología , Tejido Adiposo Blanco , Tejido Adiposo Pardo , Ratones Endogámicos C57BL
16.
J Physiol Biochem ; 79(3): 529-541, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36781604

RESUMEN

Acacetin (ACA), a flavone isolated from Chinese traditional medical herbs, has numerous pharmacological activities. However, little is known about the roles in white fat browning and energy metabolism. In the present study, we investigated whether and how ACA would improve energy metabolism in vivo and in vitro. ACA (20 mg/kg) was intraperitoneally injected to the mice with obesity induced by HFD for 14 consecutive days (in vivo); differentiated 3T3-L1 adipocytes were treated with ACA (20 µmol/L and 40 µmol/L) for 24 h (in vitro). The metabolic profile, lipid accumulation, fat-browning and mitochondrial contents, and so on were respectively detected. The results in vivo showed that ACA significantly reduced the body weight and visceral adipose tissue weight, alleviated the energy metabolism disorder, and enhanced the browning-related protein expressions in adipose tissue of rats. Besides, the data in vitro revealed that ACA significantly reduced the lipid accumulation, induced the expressions of the browning-related proteins and cAMP-dependent protein kinase A (PKA), and increased the mitochondrium contents, especially enhanced the energy metabolism of adipocytes; however, treatment with beta-adrenergic receptor blocker (propranolol, Pro) or adenyl cyclase (AC) inhibitor (SQ22536, SQ) abrogated the ACA-mediated effects. The data demonstrate that ACA alleviates the energy metabolism disorder through the pro-browning effects mediated by the AC-cAMP pathway. The findings would provide the experimental foundation for ACA to prevent and treat obesity and related metabolism disorders.


Asunto(s)
Flavonas , Enfermedades Metabólicas , Ratones , Ratas , Animales , Obesidad/metabolismo , Tejido Adiposo Blanco/metabolismo , Metabolismo Energético , Flavonas/farmacología , Flavonas/uso terapéutico , Flavonas/metabolismo , Enfermedades Metabólicas/metabolismo , Lípidos/uso terapéutico , Células 3T3-L1 , Tejido Adiposo Pardo/metabolismo , Adipocitos Blancos/metabolismo , Dieta Alta en Grasa/efectos adversos
17.
Obesity (Silver Spring) ; 31(3): 732-743, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36693798

RESUMEN

OBJECTIVE: The aim of the study was to investigate the contribution of asprosin (ASP), a fasting-induced hormone involved in metabolic disorders, to vascular endothelial dysfunction in obesity models. METHODS: Primary rat thoracic aortic endothelial cells treated with palmitic acid and mice fed with a high-fat diet (HFD) were used as the obesity models. The role and mechanism of ASP in endothelial dysfunction were investigated by the means of morphologic, functional, and genetic analysis. RESULTS: ASP aggravated the endothelial dysfunction induced by either palmitic acid in vitro or an HFD in vivo, characterized as the impairment of endothelium-dependent vasodilation, reduction of nitric oxide levels, elevation of malondialdehyde levels, and inhibition of phosphoinositide 3-kinase-AKT-endothelial nitric oxide synthase signaling. However, adipose conditional knockout of ASP or ASP neutralization significantly alleviated the endothelial dysfunction induced by an HFD. Mechanistically, ASP enhanced mitochondrial fission, and inhibition of the fission through knockdown of dynamin-related protein 1 (a fission-hallmark factor) rescued the endothelial dysfunction and the disturbance to mitochondrial dynamics induced by ASP. CONCLUSIONS: The findings demonstrate that ASP causes and even exacerbates vascular endothelial dysfunction through promoting mitochondrial fission in obesity, suggesting that ASP can act as an early predictive marker of blood vessel dysfunction and become a novel potential therapeutic target for obesity-related cardiovascular diseases.


Asunto(s)
Dinámicas Mitocondriales , Ácido Palmítico , Animales , Ratones , Ratas , Dieta Alta en Grasa , Células Endoteliales/metabolismo , Endotelio Vascular , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III/metabolismo , Obesidad/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Vasodilatación
18.
Mol Cell Biochem ; 370(1-2): 231-40, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22878563

RESUMEN

Myocardial preconditioning is a powerful phenomenon that can attenuate ischemia/reperfusion-induced oxidant stress and elicit delayed cardioprotection. Its mechanisms involve activation of intracellular signaling pathways and up-regulation of the protective antioxidant proteins. DJ-1 protein, as a multifunctional intracellular protein, plays an important role in attenuating oxidant stress and promoting cell survival. In the present study, we investigated whether DJ-1 is up-regulated during the late phase of hypoxic preconditioning (HP) and the up-regulation of DJ-1 is mediated by extracellular-regulated kinase 1/2 (ERK1/2) signaling pathway. Rat heart-derived H9c2 cells were exposed to HP. Twenty-four hours later cells were subjected to hypoxia/reoxygenation (H/R) and then cell viability, lactate dehydrogenase (LDH), intracellular reactive oxygen species (ROS), ERK1/2 phosphorylation, and DJ-1 protein were measured appropriately. The results showed that HP efficiently attenuated H/R-induced viability loss and LDH leakage. In addition, HP promoted ERK1/2 activation, up-regulated DJ-1 protein expression, inhibited H/R induced the elevation of ROS. However, when ERK1/2 phosphorylation was specifically inhibited by U0126, the increase in DJ-1 expression occurring during HP was almost completely abolished and, as a result, the delayed cardioprotection induced by HP was abolished, and the inhibitory effect of HP on H/R-induced oxidant stress was also reversed. Furthermore, knocking down DJ-1 by siRNA attenuated the delayed cardioprotection induced by HP. Our data indicate that HP can up-regulate DJ-1 protein expression through the ERK1/2-dependent signaling pathway. Importantly, DJ-1 might be involved in the delayed cardioprotective effect of HP against H/R injury.


Asunto(s)
Hipoxia/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocardio/enzimología , Miocardio/patología , Regulación hacia Arriba , Animales , Butadienos/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Técnicas de Silenciamiento del Gen , L-Lactato Deshidrogenasa/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , FN-kappa B/metabolismo , Nitrilos/farmacología , Oxígeno , Proteína Desglicasa DJ-1 , ARN Interferente Pequeño/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos
19.
Acta Pharmacol Sin ; 33(4): 490-5, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22407227

RESUMEN

AIM: To compare the effects of cholecalciferol (800 IU/d) and calcitriol (0.25 µg/d) on calcium metabolism and bone turnover in Chinese postmenopausal women with vitamin D insufficiency. METHODS: One hundred Chinese postmenopausal women aged 63.8±7.0 years and with serum 25-hydroxyvitamin D [25(OH)D] concentration <30 ng/mL were recruited. The subjects were divided into 2 groups based on the age and serum 25(OH)D concentration: 50 subjects (group A) received cholecalciferol (800 IU/d), and 50 subjects (group B) received calcitriol (0.25 µg/d) for 3 months. In addition, all the subjects received Caltrate D (calcium plus 125 IU cholecalciferol) daily in the form of one pill. The markers of calcium metabolism and bone turnover, including the serum levels of calcium, phosphorus, alkaline phosphatase, intact parathyroid hormone, 25(OH)D and ß-CrossLaps of type I collagen containing cross-linked C-telopeptide (ß-CTX), were measured before and after the intervention. RESULTS: After the 3-month intervention, the serum 25(OH)D concentration in group A was significantly increased from 16.01 ± 5.0 to 20.02 ± 4.5 ng/mL, while that in group B had no significant change. The serum calcium levels in both the groups were significantly increased (group A: from 2.36 ± 0.1 to 2.45 ± 0.1 mmol/L; group B: from 2.36 ± 0.1 to 2.44 ± 0.1 mmol/L). The levels of serum intact parathyroid hormone in both the groups were significantly decreased (group A: from 48.56 ± 12.8 to 39.59 ± 12.6 pg/mL; group B: from 53.67 ± 20.0 to 40.32 ± 15.4 pg/mL). The serum levels of ß-CTX in both the groups were also significantly decreased (group A: from 373.93 ± 135.3 to 325.04 ± 149.0 ng/L; group B: from 431.00 ± 137.1 to 371.74 ± 185.0 ng/L). CONCLUSION: We concluded that both cholecalciferol (800 IU/d) and calcitriol (0.25 µg/d) plus Caltrate D modifies the serum calcium and bone turnover markers in Chinese postmenopausal women with vitamin D insufficiency. In addition, cholecalciferol (800 IU/d) significantly increased the serum 25(OH)D concentration.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Calcitriol/farmacología , Calcio/sangre , Colecalciferol/farmacología , Posmenopausia/efectos de los fármacos , Deficiencia de Vitamina D/sangre , Anciano , Huesos/efectos de los fármacos , Calcio/metabolismo , Femenino , Humanos , Persona de Mediana Edad , Vitamina D/análogos & derivados , Vitamina D/sangre , Deficiencia de Vitamina D/complicaciones
20.
Can J Physiol Pharmacol ; 90(7): 873-80, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22693949

RESUMEN

We have shown recently that sasanquasaponin (SQS) can inhibit ischemia/reperfusion-induced elevation of intracellular Cl(-) concentration ([Cl(-)](i)) and elicit cardioprotection by up-regulating anion exchanger 3 (AE(3)) expression. In the present study, we futher analysed the intracellular signal transduction pathways by which SQS up-regulates AE(3) expression and elicits cardioprotection. Cardiomyocytes were incubated for 24 h with or without 10 µmol/L SQS, followed by simulated ischemia/reperfusion (sI/R). NO formation, Ras activity, and extracellular-regulated kinase 1/2 (ERK1/2) phosphorylation were measured appropriately. We showed that SQS pretreatment efficiently attenuated viability loss and lactate dehydrogenase leakage induced by sI/R in cardiomyocytes. Moreover, SQS induced NO production and promoted Ras activation, which futher promoted extracellular-regulated kinase 1/2 (ERK1/2) phosphorylation. These effects were paralleled by an increase in AE(3) expression. However, when the cardiomyocytes were treated with 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-l-oxyl-3-oxide (c-PTIO; an NO scavenger), S-trans-trans-farnesylthiosalicylic acid (FTS) (a Ras inhibitor), U0126 (an ERK1/2 inhibitor), respectively, the increase in AE(3) expression occurring during SQS pretreatment was almost completely abolished and, as a result, SQS-induced cardioprotection was prevented. Our findings indicate that SQS might up-regulate AE(3) expression through NO/Ras/ERK1/2 signal pathway to elicit cardioprotection in cultured cardiomyocytes.


Asunto(s)
Antiportadores/metabolismo , Cardiotónicos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Óxido Nítrico/metabolismo , Saponinas/farmacología , Proteínas ras/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , L-Lactato Deshidrogenasa/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA