Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Adolesc Health ; 67(5S): S24-S31, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33246530

RESUMO

PURPOSE: This article explores differences between the current health status of adolescents and the targets set in Healthy China 2030 (HC2030). METHODS: Global and domestic policies and strategies relating to adolescent health were reviewed. Data from the Global Burden of Disease Study (1990-2016) and the Chinese National Survey on Students' Constitution and Health (1985-2014) were used to analyze time trends and geographical distributions of health indicators for adolescents aged 10-19 years in China. RESULTS: The Chinese government has released many health policies over the past 3 decades. In this context, there has been a major decline in all-cause mortality and stunting. However, gaps between the current health status of adolescents and the targets set in HC2030 were numerous. The prevalence of obesity and poor vision increased rapidly, and few adolescents meet the physical activity and fitness targets set in HC2030. Urban/rural differences were marked for some indicators such as obesity, whereas for other indicators (e.g., stunting), there remained notable differences across provinces. CONCLUSIONS: Many long-standing health problems of adolescents have improved, but new problems related to noncommunicable disease risks have emerged and should be a prominent focus for policy action under HC2030.


Assuntos
Saúde do Adolescente , Nível de Saúde , População Rural , Adolescente , Adulto , Criança , China/epidemiologia , Política de Saúde , Estilo de Vida Saudável , Humanos , Obesidade/epidemiologia , Prevalência , População Urbana , Transtornos da Visão/epidemiologia , Adulto Jovem
2.
Acta Physiol (Oxf) ; 229(2): e13438, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31900993

RESUMO

AIMS: Pathogenic variants of the SCN5A gene can cause Brugada syndrome (BrS) and long QT syndrome (LQTS), which predispose individuals to potentially fatal ventricular arrhythmias and sudden cardiac death. SCN5A encodes the NaV 1.5 protein, the pore forming α-subunit of the voltage-dependent cardiac Na+ channel. Using a WW domain, the E3 ubiquitin ligase Nedd4-2 binds to the PY-motif ([L/P]PxY) within the C-terminus of NaV 1.5, which results in decreased protein expression and current through NaV 1.5 ubiquitination. Here, we investigate the role of E3 ubiquitin ligase Nedd4-2-mediated NaV 1.5 degradation in the pathological mechanisms of the BrS-associated variant SCN5A-p.L1239P and LQTS-associated variant SCN5A-p.Y1977N. METHODS AND RESULTS: Using a combination of molecular biology, biochemical and electrophysiological approaches, we examined the expression, function and Nedd4-2 interactions of SCN5A-p.L1239P and SCN5A-p.Y1977N. SCN5A-p.L1239P is characterized as a loss-of-function, whereas SCN5A-p.Y1977N is a gain-of-function variant of the NaV 1.5 channel. Sequence alignment shows that BrS-associated SCN5A-p.L1239P has a new Nedd4-2-binding site (from LLxY to LPxY). This new Nedd4-2-binding site increases the interaction between NaV 1.5 and Nedd4-2, enhancing ubiquitination and degradation of the NaV 1.5 channel. Disruption of the new Nedd4-2-binding site of SCN5A-p.L1239P restores NaV 1.5 expression and function. However, the LQTS-associated SCN5A-p.Y1977N disrupts the usual Nedd4-2-binding site (from PPxY to PPxN). This decreases NaV 1.5-Nedd4-2 interaction, preventing ubiquitination and degradation of NaV 1.5 channels. CONCLUSIONS: Our data suggest that the PY-motif plays an essential role in modifying the expression/function of NaV 1.5 channels through Nedd4-2-mediated ubiquitination. Alterations of NaV 1.5-Nedd4-2 interaction represent a novel pathological mechanism for NaV 1.5 channel diseases caused by SCN5A variants.


Assuntos
Síndrome de Brugada/metabolismo , Síndrome do QT Longo/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/química , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Motivos de Aminoácidos , Síndrome de Brugada/enzimologia , Células HEK293 , Humanos , Síndrome do QT Longo/enzimologia , Ligação Proteica
3.
Arthritis Res Ther ; 21(1): 134, 2019 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-31159863

RESUMO

BACKGROUND: We previously found that high-mobility group box protein 1 (HMGB1) promoted cell proliferation, migration, invasion, and autophagy in rheumatoid arthritis fibroblast-like synoviocytes (RA-FLS), but little is known about its regulatory mechanism. The aim of this study was to investigate the regulatory mechanism of HMGB1 at the posttranscription level. METHODS: Real-time qPCR, CCK-8 cell proliferation assay, transwell cell migration assay, enzyme-linked immunosorbent assay (ELISA), and western blotting were used in this study. The targeting relationship between miRNA and mRNA was presented by the luciferase reporter assay. RESULTS: MiR-449a was downregulated in RA synovial tissue and inhibited RA-FLS proliferation, migration, and IL-6 production. MiR-449a directly targeted HMGB1 and inhibited its expression. Yin Yang 1(YY1) negatively regulated miR-449a expression and formed a mutual inhibition loop in RA-FLS. MiR-449a inhibited TNFα-mediated HMGB1 and YY1 overexpression and IL-6 production. CONCLUSIONS: Our results reveal the regulatory mechanism of HMGB1 in RA and demonstrate that miR-449a is a crucial molecule in RA pathogenesis and a suitable candidate for miRNA replacement therapies in RA.


Assuntos
Artrite Reumatoide/genética , Regulação da Expressão Gênica , Proteína HMGB1/genética , Inflamação/genética , MicroRNAs/genética , Sinoviócitos/patologia , Fator de Transcrição YY1/genética , Idoso , Apoptose , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Western Blotting , Movimento Celular , Proliferação de Células , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Proteína HMGB1/biossíntese , Humanos , Inflamação/metabolismo , Inflamação/patologia , Masculino , MicroRNAs/biossíntese , Pessoa de Meia-Idade , RNA/genética , Sinoviócitos/metabolismo , Fator de Transcrição YY1/biossíntese
4.
Int J Toxicol ; 36(4): 314-324, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28535735

RESUMO

Clenbuterol, a ß2-selective adrenergic receptor agonist, is illicitly used in weight loss and performance enhancement and animal production. Increasing evidence demonstrates that clenbuterol induces various kinds of arrhythmias and QTc interval prolongation. However, little is known about the underlying mechanism. Most drugs are associated with QTc prolongation through interfering with human ether-a-go-go-related gene (hERG) K+ channels. The present study aims to investigate the effects and underlying mechanisms of clenbuterol on the hERG channel. HEK 293 cells were transfected with wild type and Y652A or F656A mutants of the hERG channel and treated with clenbuterol. The hERG current was recorded using whole-cell patch-clamp technique, and protein level was evaluated by Western blot. We found that clenbuterol decreases the mature form of the hERG protein at the cell membrane in a concentration- and time-dependent manner, without affecting the immature form. Correspondingly, clenbuterol chronic treatment reduced hERG current to a greater extent compared to acute treatment. In the presence of Brefeldin A (BFA), which was used to block hERG channel trafficking to cell membrane, clenbuterol reduced hERG on plasma membrane to a greater extent than BFA alone. In addition, the hERG channel's drug binding sites mutant Y652A and F656A abolished clenbuterol-mediated hERG reduction and current blockade. In conclusion, clenbuterol reduces hERG channel expression and current by promoting the channel degradation. The effect of clenbuterol on the hERG channel is related to the drug-binding sites, Tyr-652 and Phe-656, located on the S6 domain. This biophysical mechanism may underlie clenbuterol-induced QTc prolongation or arrhythmia.


Assuntos
Clembuterol/farmacologia , Canais de Potássio Éter-A-Go-Go/metabolismo , Proteólise/efeitos dos fármacos , Agonistas Adrenérgicos beta/farmacologia , Arritmias Cardíacas/induzido quimicamente , Sítios de Ligação/genética , Células Cultivadas , Relação Dose-Resposta a Droga , Canais de Potássio Éter-A-Go-Go/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Mutação , Técnicas de Patch-Clamp
5.
BMC Cardiovasc Disord ; 17(1): 6, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-28056793

RESUMO

BACKGROUND: The association between admission hyperglycemia and adverse outcomes in patients with non-ST-segment elevation myocardial infarction (NSTEMI) undergoing percutaneous coronary intervention (PCI) has not been well studied, and the optimal plasma glucose cut-off values for prognosis for NSTEMI patients with and without diabetes have not been determined. METHODS: According to glucose level and diabetes status, consecutive NSTEMI patients undergoing PCI (n = 890) were divided into four groups: without diabetes mellitus (DM) and admission plasma glucose (APG) <144 or ≥144 mg/dL; or with DM and APG <180 or ≥180 mg/dL. All patients were followed up at 30 days and 3 years after discharge, and the outcomes were assessed. RESULTS: Admission hyperglycemia was found in 44 and 28% of the DM and non-DM patients, respectively. Multivariable analyses showed that the APG level was an independent predictor of 30-day and 3-year MACEs. Receiver operating characteristic curve analysis revealed that the appropriate cut-off values were 178 and 145 mg/dL for patients with and without DM, respectively, or 157 mg/dL for all patients. CONCLUSIONS: Admission hyperglycemia may be used to predict 30-day and 3-year MACEs in patients with NSTEMI undergoing PCI, irrespective of diabetes status. However, the optimal admission glucose cut-off values for predicting prognosis differ for patients with or without DM.


Assuntos
Glicemia/análise , Diabetes Mellitus/sangue , Infarto do Miocárdio sem Supradesnível do Segmento ST/terapia , Admissão do Paciente , Intervenção Coronária Percutânea , Idoso , Biomarcadores/sangue , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/mortalidade , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio sem Supradesnível do Segmento ST/sangue , Infarto do Miocárdio sem Supradesnível do Segmento ST/diagnóstico , Infarto do Miocárdio sem Supradesnível do Segmento ST/mortalidade , Intervenção Coronária Percutânea/efeitos adversos , Intervenção Coronária Percutânea/mortalidade , Valor Preditivo dos Testes , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento
6.
J Membr Biol ; 248(6): 1181-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26362340

RESUMO

Recent studies have shown that the sensitivity of apamin-sensitive K(+) current (I KAS, mediated by apamin-sensitive small conductance calcium-activated potassium channels subunits) to intracellular Ca(2+) is increased in heart failure (HF), leading to I KAS upregulation, action potential duration shortening, early after depolarization, and recurrent spontaneous ventricular fibrillation. We hypothesized that casein kinase 2 (CK2) interacted with small conductance calcium-activated potassium channels (SK) is decreased in HF, and protein phosphatase 2A (PP2A) is increased on the opposite, upregulating the sensitivity of I KAS to intracellular Ca(2+) in HF. Rat model of volume-overload HF was established by an abdominal arteriovenous fistula procedure. The expression of SK channels, PP2A and CK2 was detected by Western blot analysis. Interaction and colocalization of CK2 with SK channel were detected by co-immunoprecipitation analysis and double immunofluorescence staining. In HF rat left ventricle, SK3 was increased by 100 % (P < 0.05), and SK2 was not significantly changed. PP2A protein was increased by 94.7 % in HF rats (P < 0.05), whereas the level of CK2 was almost unchanged. We found that CK2 colocalized with SK2 and SK3 in rat left ventricle. With anti-CK2α antibody, SK2 and SK3 were immunoprecipitated, the level of precipitated SK2 decreased by half, whereas precipitated SK3 was almost unchanged. In conclusion, the increased expression of total PP2A and decreased interaction of CK2 with SK2 may underlie enhanced sensitivity of I KAS to intracellular Ca(2+) in volume-overload HF rat.


Assuntos
Apamina/metabolismo , Caseína Quinase II/metabolismo , Insuficiência Cardíaca/metabolismo , Potássio/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Potenciais de Ação , Animais , Modelos Animais de Doenças , Ecocardiografia , Quinases do Centro Germinativo , Insuficiência Cardíaca/diagnóstico , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Masculino , Miócitos Cardíacos/metabolismo , Ligação Proteica , Proteína Fosfatase 2/metabolismo , Transporte Proteico , Ratos , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Regulação para Cima
7.
Zhongguo Dang Dai Er Ke Za Zhi ; 17(4): 395-9, 2015 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-25919563

RESUMO

OBJECTIVE: To study the protocol of construction of the mutation E292V and M723I of hABCA3 gene associated with neonatal respiratory distress syndrome, as well as their eukaryotic green fluorescent protein expression rectors, and to examine the expression of mutation proteins in human lung carcinoma epithelial cells (A549). METHODS: Site-directed mutagenesis method based on overlap extension PCR was used to introduce mutations in the two sites which were E292V and M723I in the ABCA3. The PCR fragments were subcloned to PEGFP-C2 vectors to construct the eukaryotic green fluorescent protein expression rectors. A549 cells were transiently transfected with the recombinants using Lipofectamine 2000 and the transfection efficiency was confirmed through GFP signal. The expression and location of recombinants were detected by FV1000 laser scanning microscope. RESULTS: Direct sequence analysis confirmed an A to T transition at position 875 in E292V and a G to A transition at position 2169 in M723I. Recombinants were transfected to A549 cells and both wild type and mutant ABCA3 proteins were expressed in the cytoplasm. CONCLUSIONS: The eukaryotic green fluorescent protein expression rectors of wild type and mutant ABCA3 gene were constructed and they were successfully expressed in A549 cells. This experiment provides a basis for subsequent research.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Mutagênese Sítio-Dirigida , Linhagem Celular Tumoral , Proteínas de Fluorescência Verde/genética , Humanos , Transfecção
8.
Mol Cell Biochem ; 397(1-2): 267-76, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25185755

RESUMO

Pulmonary arterial remodeling is characterized by excessive proliferation, migration, and pro-differentiation and fibrotic activation of adventitial fibroblasts in pulmonary arterial hypertension (PAH) process. Several lines of evidence indicate that serotonin (5-HT) plays a central role in the pathogenesis of pulmonary arterial remodeling. In the present study, we investigated whether 5-HT is directly involved in the functional regulation of pulmonary artery adventitial fibroblasts (PAFs). Incubation of cultured rat PAFs with 5-HT caused a dose-dependent stimulation of cell proliferation, migration activity, and a time-dependent increase of α-SMA expression, a marker of fibroblast differentiation into myofibroblasts, and adventitia fibrosis, evaluating connective tissue growth factor (CTGF) and extracellular matrix (ECM) mRNAs and proteins. These effects were attenuated by the 5-HT2A receptor antagonist, ketanserin and mimicked by the 5-HT2A receptor agonist DOI. 5-HT-induced fibroblasts phenotypic alterations and ECM accumulation were dependent on stimulation of transforming growth factor (TGF)-ß1 as demonstrated using a neutralizing antibody. 5-HT also caused Smad3 phosphorylation and ketanserin diminished 5-HT-induced Smad3 activation. These results demonstrated that 5-HT can directly activate PAFs through 5-HT2A receptor and promote fibroblasts phenotypic alterations and adventitia fibrosis depending on the signaling of the TGF-ß1/Smad3 pathway.


Assuntos
Túnica Adventícia/metabolismo , Fibroblastos/metabolismo , Hipertensão Pulmonar/metabolismo , Artéria Pulmonar/metabolismo , Receptor 5-HT2A de Serotonina/metabolismo , Serotonina/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Túnica Adventícia/patologia , Animais , Proliferação de Células/efeitos dos fármacos , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Fibroblastos/patologia , Fibrose/metabolismo , Fibrose/patologia , Hipertensão Pulmonar/patologia , Ketanserina/farmacologia , Masculino , Fosforilação/efeitos dos fármacos , Artéria Pulmonar/patologia , Ratos , Ratos Sprague-Dawley , Antagonistas da Serotonina/farmacologia , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA