Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Transl Med ; 14(634): eabl6992, 2022 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-35235340

RESUMO

SERAC1 deficiency is associated with the mitochondrial 3-methylglutaconic aciduria with deafness, (hepatopathy), encephalopathy, and Leigh-like disease [MEGD(H)EL] syndrome, but the role of SERAC1 in mitochondrial physiology remains unknown. Here, we generated Serac1-/- mice that mimic the major diagnostic clinical and biochemical phenotypes of the MEGD(H)EL syndrome. We found that SERAC1 localizes to the outer mitochondrial membrane and is a protein component of the one-carbon cycle. By interacting with the mitochondrial serine transporter protein SFXN1, SERAC1 facilitated and was required for SFXN1-mediated serine transport from the cytosol to the mitochondria. Loss of SERAC1 impaired the one-carbon cycle and disrupted the balance of the nucleotide pool, which led to primary mitochondrial DNA (mtDNA) depletion in mice, HEK293T cells, and patient-derived immortalized lymphocyte cells due to insufficient supply of nucleotides. Moreover, both in vitro and in vivo supplementation of nucleosides/nucleotides restored mtDNA content and mitochondrial function. Collectively, our findings suggest that MEGD(H)EL syndrome shares both clinical and molecular features with the mtDNA depletion syndrome, and nucleotide supplementation may be an effective therapeutic strategy for MEGD(H)EL syndrome.


Assuntos
DNA Mitocondrial , Serina , Animais , Hidrolases de Éster Carboxílico/genética , Hidrolases de Éster Carboxílico/metabolismo , Contratura , DNA Mitocondrial/genética , Células HEK293 , Perda Auditiva Neurossensorial , Histiocitose , Humanos , Camundongos , Mitocôndrias/metabolismo , Mutação , Nucleotídeos/metabolismo , Serina/genética , Serina/metabolismo , Síndrome
2.
Eur J Pharmacol ; 917: 174720, 2022 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-34953801

RESUMO

Autoimmune hepatitis (AIH) is a chronic progressive liver disease that currently does not have a successful therapeutic option. Vitexin, a bioflavonoid isolated from various medicinal plants, possesses a variety of activities; however, whether vitexin protects against AIH remains unclear. Therefore, the current study aims to investigate the hepatoprotective effects and mechanism of action of vitexin in both an experimental autoimmune hepatitis (EAH) mouse model and in D-galactosamine/lipopolysaccharide (D-GalN/LPS)-induced hepatocyte injury. Syngeneic liver antigen S100 was used to establish EAH. Vitexin treatment significantly decreased the infiltration of inflammatory and CD4+ T cells in the liver, reduced ALT and AST levels in the serum and attenuated hepatic injury due to oxidative stress. Moreover, vitexin mitigated the upregulation of Bax and cleaved caspase-3 and the downregulation of Bcl-2 in the livers of AIH mice. These regulations were accompanied by not only increased phosphorylation of AMPK, AKT and GSK-3ß but also activation of Nrf2. Furthermore, vitexin inhibited apoptosis and the overexpression of inflammatory cytokines in D-GalN/LPS-treated AML12 cells. In addition, vitexin enhanced the phosphorylation of AMPK, AKT and GSK-3ß. When AML12 cells were treated with an inhibitor of AMPK/AKT or specific siRNA targeting Nrf2, vitexin did not further induce the activation of Nrf2/HO-1. A molecular docking study confirmed that vitexin could interact with AMPK through hydrogen bonding interactions. In conclusion, vitexin ameliorated hepatic injury in EAH mice through activation of the AMPK/AKT/GSK-3ß pathway and upregulation of the Nrf2 gene.


Assuntos
Fator 2 Relacionado a NF-E2
3.
Hypertens Res ; 44(9): 1175-1185, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34127818

RESUMO

Arterial stiffness has been suggested as an independent cardiovascular risk factor. This systematic review and meta-analysis aimed to quantify the association between brachial-ankle pulse wave velocity (baPWV) and prognosis in atherosclerotic cardiovascular disease (ASCVD) patients. The PubMed, EMBASE, and Cochrane Library databases were searched for cohort studies examining the association of high baPWV with prognosis in ASCVD patients. High baPWV was defined by the cutoffs provided by each study. The outcomes of interest were cardiovascular events, cardiovascular mortality, and all-cause mortality. Hazard ratios (HRs) and 95% confidence intervals (CIs) were combined using the random-effects model with inverse variance weighting. We identified 15 studies that were eligible for inclusion in the meta-analysis. The overall HRs and 95% CIs of cardiovascular events, cardiovascular mortality, and all-cause mortality for high baPWV were 2.55 (1.61-4.03), 2.66 (1.88-3.76), and 1.77 (1.09-2.87), respectively. The association between baPWV and cardiovascular events remained significant, irrespective of determination methods for cutoffs of baPWV, classification of ASCVD, outcome definitions, Newcastle-Ottawa Scale score and average age, independent of age and hypertension. Significantly higher HRs were observed in the subgroups of >3 years follow-up duration (p for interaction: 0.04), cutoff points by ROC curves (p for interaction: 0.04) and an average age of <65 years (p for interaction: 0.01). A 1 standard deviation increase in baPWV was associated with a 1.41-fold (1.24-1.60) increase in the risk of cardiovascular events. High baPWV is independently associated with an increased risk of cardiovascular events, cardiovascular mortality, and all-cause mortality in ASCVD patients.


Assuntos
Doenças Cardiovasculares , Rigidez Vascular , Idoso , Índice Tornozelo-Braço , Doenças Cardiovasculares/diagnóstico , Humanos , Prognóstico , Análise de Onda de Pulso , Fatores de Risco
4.
J Clin Lipidol ; 15(3): 466-476, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34006456

RESUMO

BACKGROUND: Lipoprotein(a) has been suggested as an independent risk factor for cardiovascular events in patients with coronary heart disease (CHD). OBJECTIVE: This study aimed to investigate the association of lipoprotein(a) with long-term poor prognosis following acute coronary syndromes (ACS) in advanced-age patients. METHODS: We enrolled 536 patients aged ≥80 years hospitalized for ACS and plasma lipoprotein(a) concentrations were measured at admission. The primary outcomes were hard CHD events (a composite of fatal or non-fatal myocardial infarction, and CHD death). The secondary outcomes included major adverse cardiovascular events (MACEs), all-cause death and cardiac death. RESULTS: During a median 66-month follow-up, 89 hard CHD events occurred. The optimal cutoff points of lipoprotein(a) levels were obtained from ROC curve analyses. Kaplan-Meier curves showed a significantly higher cumulative incidence of hard CHD events, MACEs, all-cause death and cardiac death in high lipoprotein(a) group than that in low lipoprotein(a) group. Multivariate Cox proportional hazards analyses revealed that elevated lipoprotein(a) levels were independently associated with an increased risk of hard CHD events [hazard ratio (HR): 1.714, 95% confidence interval (95%CI): 1.114-2.638], MACEs (HR 1.354, 95%CI: 1.024-1.790), all-cause death (HR 1.804, 95%CI: 1.286-2.532) and cardiac death (HR 1.891, 95%CI: 1.112-3.217). Furthermore, adding lipoprotein(a) to the prognostic model for hard CHD events improved the C-statistic value (P < 0.05). CONCLUSION: Elevated lipoprotein(a) levels were associated with an increased risk of hard CHD events, MACEs, all-cause death and cardiac death in the advanced-age patients with ACS, which indicated that routine screening for lipoprotein(a) might aid prognosis and risk assessment.


Assuntos
Síndrome Coronariana Aguda/sangue , Síndrome Coronariana Aguda/patologia , Lipoproteína(a)/sangue , Idoso de 80 Anos ou mais , Feminino , Seguimentos , Humanos , Masculino , Prognóstico , Fatores de Risco
5.
PLoS One ; 10(6): e0129665, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26061278

RESUMO

Acidic fibroblast growth factor (FGF1) has been suggested to enhance the functional activities of endothelial progenitor cells (EPCs). The Forkhead homeobox type O transcription factors (FOXOs), a key substrate of the survival kinase Akt, play important roles in regulation of various cellular processes. We previously have shown that FOXO3a is the main subtype of FOXOs expressed in EPCs. Here, we aim to determine whether FGF1 promotes EPC function through Akt/FOXO3a pathway. Human peripheral blood derived EPCs were transduced with adenoviral vectors either expressing a non-phosphorylable, constitutively active triple mutant of FOXO3a (Ad-TM-FOXO3a) or a GFP control (Ad-GFP). FGF1 treatment improved functional activities of Ad-GFP transduced EPCs, including cell viability, proliferation, antiapoptosis, migration and tube formation, whereas these beneficial effects disappeared by Akt inhibitor pretreatment. Moreover, EPC function was declined by Ad-TM-FOXO3a transduction and failed to be attenuated even with FGF1 treatment. FGF1 upregulated phosphorylation levels of Akt and FOXO3a in Ad-GFP transduced EPCs, which were repressed by Akt inhibitor pretreatment. However, FGF1 failed to recover Ad-TM-FOXO3a transduced EPCs from dysfunction. These data indicate that FGF1 promoting EPC function is at least in part mediated through Akt/FOXO3a pathway. Our study may provide novel ideas for enhancing EPC angiogenic ability and optimizing EPC transplantation therapy in the future.


Assuntos
Células Progenitoras Endoteliais/efeitos dos fármacos , Fator 1 de Crescimento de Fibroblastos/farmacologia , Fatores de Transcrição Forkhead/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Apoptose , Movimento Celular , Proliferação de Células , Células Cultivadas , Células Progenitoras Endoteliais/metabolismo , Células Progenitoras Endoteliais/fisiologia , Proteína Forkhead Box O3 , Humanos , Sistemas do Segundo Mensageiro
6.
J Cell Physiol ; 230(9): 2098-107, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25640014

RESUMO

Cell loss due to apoptosis induced by oxidative stress is a major hurdle for endothelial progenitor cells (EPCs)-based therapy. Sirtuin 1 (SIRT1) plays important roles in many pathophysiological processes by deacetylating various substrates, including forkhead transcription factor (FOXO). However, after deacetylation, the fate of FOXO protein remains to be explored. In the present study, we investigated whether SIRT1 exerted a protective effect on hydrogen peroxide (H(2)O(2))-induced EPCs apoptosis and, if so, what the underlying mechanism might be. EPCs were isolated and obtained from human umbilical cord blood by density gradient centrifugation and identified by morphology, tube formation ability, cell surface markers, and the ability to take up acetylated low-density lipoprotein (Dil-Ac-LDL) and bind ulex europaeus agglutinin 1 (FITC-UEA-1). Immunofluorescence showed that SIRT1 is localized in the nucleus of EPCs in the presence or absence of H(2)O(2). SIRT1 protein level in EPCs was increased by the treatment with H(2)O(2) for 24 h. Incubation of EPCs with H(2)O(2) dose dependently induced EPCs apoptosis. SIRT1 overexpression reduced the rate of EPCs apoptosis induced by H(2)O(2), whereas SIRT1 downregulation and EX527, a specific SIRT1 inhibitor, exerted the opposite effect. SIRT1 overexpression decreased the total FOXO3a protein expression, whereas SIRT1 downregulation and EX527 increased the amount of FOXO3a protein. SIRT1 reduced FOXO3a transcriptional activity according to Bim expression. Co-immunoprecipitation assay showed that SIRT1 could bind to FOXO3a, reduce its acetylation level and increase its ubiquitination level. To sum up, our work demonstrated that SIRT1 had a pivotally protective role in the regulation of EPCs apoptosis induced by H(2)O(2) and that SIRT1 protected against apoptosis by inhibiting FOXO3a via FOXO3a ubiquitination and subsequent degradation.


Assuntos
Apoptose/efeitos dos fármacos , Fatores de Transcrição Forkhead/genética , Estresse Oxidativo/genética , Sirtuína 1/genética , Células Progenitoras Endoteliais/efeitos dos fármacos , Sangue Fetal , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/antagonistas & inibidores , Humanos , Peróxido de Hidrogênio/administração & dosagem , Proteólise/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Ubiquitinação/genética
7.
PLoS One ; 9(8): e101703, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25093499

RESUMO

Endothelial dysfunction is involved in the pathogenesis of many cardiovascular diseases such as atherosclerosis. Endothelial progenitor cells (EPCs) have been considered to be of great significance in therapeutic angiogenesis. Furthermore, the Forkhead box O (FOXO) transcription factors are known to be important regulators of cell cycle. Therefore, we investigated the effects of changes in FOXO3a activity on cell proliferation and cell cycle regulatory proteins in EPCs. The constructed recombinant adenovirus vectors Ad-TM (triple mutant)-FOXO3a, Ad-shRNA-FOXO3a and the control Ad-GFP were transfected into EPCs derived from human umbilical cord blood. Assessment of transfection efficiency using an inverted fluorescence microscope and flow cytometry indicated a successful transfection. Additionally, the expression of FOXO3a was markedly increased in the Ad-TM-FOXO3a group but was inhibited in the Ad-shRNA-FOXO3a group as seen by western blotting. Overexpression of FOXO3a suppressed EPC proliferation and modulated expression of the cell cycle regulatory proteins including upregulation of the cell cycle inhibitor p27(kip1) and downregulation of cyclin-dependent kinase 2 (CDK2), cyclin D1 and proliferating cell nuclear antigen (PCNA). In the Ad-shRNA-FOXO3a group, the results were counter-productive. Furthermore, flow cytometry for cell cycle analysis suggested that the active mutant of FOXO3a caused a noticeable increase in G1- and S-phase frequencies, while a decrease was observed after FOXO3a silencing. In conclusion, these data demonstrated that FOXO3a could possibly inhibit EPC proliferation via cell cycle arrest involving upregulation of p27(kip1) and downregulation of CDK2, cyclin D1 and PCNA.


Assuntos
Proteínas de Ciclo Celular/genética , Proliferação de Células/genética , Células Progenitoras Endoteliais/fisiologia , Fatores de Transcrição Forkhead/genética , Pontos de Checagem do Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Ciclina D1/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Proteína Forkhead Box O3 , Regulação da Expressão Gênica/fisiologia , Humanos , Antígeno Nuclear de Célula em Proliferação/metabolismo , Interferência de RNA , Transfecção , Regulação para Cima
8.
Microvasc Res ; 90: 48-54, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23920411

RESUMO

OBJECTIVES: Increased oxidative stress has been suggested to contribute to the functional impairment of endothelial progenitor cells (EPCs). The Forkhead box O transcription factors (FoxOs) are critical regulators involved in various cellular processes including cell apoptosis. Here, we investigated whether FoxOs are required in oxidative stress induced EPC apoptosis. METHODS AND RESULTS: EPCs were cultured from cord blood derived mononuclear cells and treated with hydrogen peroxide (H2O2) for induction of oxidative stress. Incubation with H2O2 dose dependently reduced viability and increased apoptosis in EPCs. Western blotting showed that EPCs predominantly expressed FoxO3a and the expression was markedly increased upon H2O2 treatment. Transduction with adenoviral vectors expressing either a wide-type or a non-phosphorylatable, constitutively active mutant of FoxO3a led to further increased apoptosis of EPCs after H2O2 treatment. Conversely, FoxO3a silencing rescued EPCs from these H2O2 induced deleterious effects. Overexpression of FoxO3a also increased the level of the pro-apoptotic protein Bim, whereas FoxO3a silencing downregulated H2O2 induced Bim expression. Furthermore, Matrigel assay demonstrated that FoxO3a overexpression significantly impaired the tube forming ability of EPCs, whereas its silencing completely protected EPCs from H2O2 induced decrease of capillary formation. CONCLUSIONS: These data suggest that oxidative stress induced impairment of EPC survival is mediated through a FoxO3a dependant mechanism, possibly by transcriptional regulation of Bim. Our data indicate FoxO3a as a potential therapeutic target for improvement of EPC number and function in patients with ischemic heart disease.


Assuntos
Células Endoteliais/efeitos dos fármacos , Fatores de Transcrição Forkhead/metabolismo , Peróxido de Hidrogênio/toxicidade , Oxidantes/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Humanos , Proteínas de Membrana/metabolismo , Mutação , Neovascularização Fisiológica/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/metabolismo , Células-Tronco/patologia , Transdução Genética , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...