Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros










Intervalo de año de publicación
1.
Exp Mol Med ; 52(10): 1715-1729, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33028948

RESUMEN

Hyperglycemia-mediated endothelial inflammation participates in the pathogenesis of cardiovascular complications in subjects with diabetes. Previous studies reported that phosphatase and tensin homolog deleted on chromosome ten (PTEN) and SET8 participate in high glucose-mediated endothelial inflammation. In this study, we hypothesize that SET8 regulates PTEN expression, thus contributing to high glucose-mediated vascular endothelial inflammation. Our data indicated that plasma soluble intercellular adhesion molecule-1 (sICAM-1) and endothelial selectin (e-selectin) were increased in patients with diabetes and diabetic rats. PTEN expression was augmented in the peripheral blood mononuclear cells of patients with diabetes and in the aortic tissues of diabetic rats. Our in vitro study indicated that high glucose increased monocyte/endothelial adhesion, endothelial adhesion molecule expression and p65 phosphorylation in human umbilical vein endothelial cells (HUVECs). Moreover, high glucose led to endothelial inflammation via upregulation of PTEN. Furthermore, high glucose inhibited SET8 expression and histone H4 lysine 20 methylation (H4K20me1), a downstream target of SET8. SET8 overexpression reversed the effects of high-glucose treatment. shSET8-mediated endothelial inflammation was counteracted by siPTEN. Furthermore, SET8 was found to interact with FOXO1. siFOXO1 attenuated high glucose-mediated endothelial inflammation. FOXO1 overexpression-mediated endothelial inflammation was counteracted by siPTEN. H4K20me1 and FOXO1 were enriched in the PTEN promoter region. shSET8 increased PTEN promoter activity and augmented the positive effect of FOXO1 overexpression on PTEN promoter activity. Our in vivo study indicated that SET8 was downregulated and FOXO1 was upregulated in the peripheral blood mononuclear cells of patients with diabetes and the aortic tissues of diabetic rats. In conclusion, SET8 interacted with FOXO1 to modulate PTEN expression in vascular endothelial cells, thus contributing to hyperglycemia-mediated endothelial inflammation.


Asunto(s)
Endotelio Vascular/metabolismo , Glucosa/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Fosfohidrolasa PTEN/genética , Adulto , Anciano , Animales , Biomarcadores , Glucemia , Células Cultivadas , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Proteína Forkhead Box O1 , Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Fosfohidrolasa PTEN/metabolismo , Ratas , Vasculitis/etiología , Vasculitis/metabolismo , Vasculitis/patología
2.
Clinics (Sao Paulo) ; 75: e1865, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32935825

RESUMEN

OBJECTIVES: Hypoxia leads to endothelial cell inflammation, apoptosis, and damage, which plays an important role in the complications associated with ischemic cardiovascular disease. As an oxidoreductase, p66Shc plays an important role in the regulation of reactive oxygen species (ROS) production and apoptosis. Ketamine is widely used in clinics. This study was designed to assess the potential protective effect of ketamine against hypoxia-induced injury in human umbilical vein endothelial cells (HUVECs). Moreover, we explored the potential mechanism by which ketamine protected against hypoxia-induced endothelial injury. METHODS: The protective effects of ketamine against hypoxia-induced injury was assessed using cell viability and adhesion assays, quantitative polymerase chain reaction, and western blotting. RESULTS: Our data showed that hypoxia reduced HUVEC viability, increased the adhesion between HUVECs and monocytes, and upregulated the expression of endothelial adhesion molecules at the protein and mRNA levels. Moreover, hypoxia increased ROS accumulation and upregulated p66Shc expression. Furthermore, hypoxia downregulated sirt1 expression in HUVECs. Alternatively, ketamine was shown to reverse the hypoxia-mediated reduction of cell viability and increase in the adhesion between HUVECs and monocytes, ameliorate hypoxia-induced ROS accumulation, and suppress p66Shc expression. Moreover, EX527, a sirt1 inhibitor, reversed the protective effects of ketamine against the hypoxia-mediated reduction of cell viability and increase in adhesion between HUVECs and monocytes. CONCLUSION: Ketamine reduces hypoxia-induced p66Shc expression and attenuates ROS accumulation via upregulating sirt1 in HUVECs, thus attenuating hypoxia-induced endothelial cell inflammation and apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Hipoxia , Ketamina/farmacología , Especies Reactivas de Oxígeno/metabolismo , Supervivencia Celular , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Estrés Oxidativo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Venas Umbilicales
3.
Clinics ; 75: e1865, 2020. graf
Artículo en Inglés | LILACS | ID: biblio-1133469

RESUMEN

OBJECTIVES: Hypoxia leads to endothelial cell inflammation, apoptosis, and damage, which plays an important role in the complications associated with ischemic cardiovascular disease. As an oxidoreductase, p66Shc plays an important role in the regulation of reactive oxygen species (ROS) production and apoptosis. Ketamine is widely used in clinics. This study was designed to assess the potential protective effect of ketamine against hypoxia-induced injury in human umbilical vein endothelial cells (HUVECs). Moreover, we explored the potential mechanism by which ketamine protected against hypoxia-induced endothelial injury. METHODS: The protective effects of ketamine against hypoxia-induced injury was assessed using cell viability and adhesion assays, quantitative polymerase chain reaction, and western blotting. RESULTS: Our data showed that hypoxia reduced HUVEC viability, increased the adhesion between HUVECs and monocytes, and upregulated the expression of endothelial adhesion molecules at the protein and mRNA levels. Moreover, hypoxia increased ROS accumulation and upregulated p66Shc expression. Furthermore, hypoxia downregulated sirt1 expression in HUVECs. Alternatively, ketamine was shown to reverse the hypoxia-mediated reduction of cell viability and increase in the adhesion between HUVECs and monocytes, ameliorate hypoxia-induced ROS accumulation, and suppress p66Shc expression. Moreover, EX527, a sirt1 inhibitor, reversed the protective effects of ketamine against the hypoxia-mediated reduction of cell viability and increase in adhesion between HUVECs and monocytes. CONCLUSION: Ketamine reduces hypoxia-induced p66Shc expression and attenuates ROS accumulation via upregulating sirt1 in HUVECs, thus attenuating hypoxia-induced endothelial cell inflammation and apoptosis.


Asunto(s)
Humanos , Especies Reactivas de Oxígeno/metabolismo , Apoptosis/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Ketamina/farmacología , Hipoxia , Venas Umbilicales , Supervivencia Celular , Estrés Oxidativo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
4.
Immunol Lett ; 202: 16-22, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30055200

RESUMEN

CD8+CD28- regulatory T cells (Tregs) play important roles in chronic viral infections. Programmed death 1 (PD-1) is highly expressed on hepatitis C virus (HCV)-specific CTLs. However, little is known regarding the role of CD8+CD28-PD1+ T cells in hepatitis C. Herein, we found that the frequency of CD8+CD28-PD1+, but not CD8+CD28-PD1- T cells, correlated with markers of chronic hepatitis C virus (HCV) infection and the response to treatment. Our results showed that CD8+CD28-PD1+ T cells were significantly elevated in chronic HCV-infected patients and there was a distinct correlation between the frequency of CD8+CD28-PD1+ T cells and serum levels of HCV RNA. During a 48-week course of treatment with peg-IFN-a2a plus ribavirin, dynamic changes in the frequencies of CD8+CD28-PD1+ T cells were observed, associated with the virologic response. IL-10 secretion may explain the suppressive function of CD8+CD28-PD1+ T cells in chronic HCV-infected patients. Overall, our study demonstrates that PD-1 is an important marker of CD8+CD28- Tregs in chronic HCV infection. Thus, the frequency and regulatory function of CD8+CD28-PD1+ T cells play vital roles in HCV infection and the response to treatment.


Asunto(s)
Antígenos CD28/inmunología , Linfocitos T CD8-positivos/inmunología , Hepatitis C Crónica/inmunología , Interleucina-10/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T Reguladores/inmunología , Adulto , Antivirales/uso terapéutico , Biomarcadores/metabolismo , Linfocitos T CD8-positivos/virología , Femenino , Hepacivirus/efectos de los fármacos , Hepacivirus/inmunología , Hepatitis C Crónica/tratamiento farmacológico , Hepatitis C Crónica/virología , Humanos , Activación de Linfocitos/inmunología , Masculino , ARN Viral/genética , Ribavirina/uso terapéutico , Linfocitos T Reguladores/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...