Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
J Am Heart Assoc ; 12(24): e029745, 2023 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-38084712

RESUMO

BACKGROUND: Cardiac hypertrophy (CH) is a well-established risk factor for many cardiovascular diseases and a primary cause of mortality and morbidity among older adults. Currently, no pharmacological interventions have been specifically tailored to treat CH. OTUD7B (ovarian tumor domain-containing 7B) is a member of the ovarian tumor-related protease (OTU) family that regulates many important cell signaling pathways. However, the role of OTUD7B in the development of CH is unclear. Therefore, we investigated the role of OTUD7B in CH. METHODS AND RESULTS: OTUD7B knockout mice were used to assay the role of OTUD7B in CH after transverse aortic coarctation surgery. We further assayed the specific functions of OTUD7B in isolated neonatal rat cardiomyocytes. We found that OTUD7B expression decreased in hypertrophic mice hearts and phenylephrine-stimulated neonatal rat cardiomyocytes. Furthermore, OTUD7B deficiency exacerbated transverse aortic coarctation surgery-induced myocardial hypertrophy, abnormal cardiac function, and fibrosis. In cardiac myocytes, OTUD7B knockdown promoted phenylephrine stimulation-induced myocardial hypertrophy, whereas OTUD7B overexpression had the opposite effect. An immunoprecipitation-mass spectrometry analysis showed that OTUD7B directly binds to KLF4 (Krüppel-like factor 4). Additional molecular experiments showed that OTUD7B impedes KLF4 degradation by inhibiting lysine residue at 48 site-linked ubiquitination and suppressing myocardial hypertrophy by activating the serine/threonine kinase pathway. CONCLUSIONS: These results demonstrate that the OTUD7B-KLF4 axis is a novel molecular target for CH treatment.


Assuntos
Coartação Aórtica , Fator 4 Semelhante a Kruppel , Camundongos , Ratos , Animais , Cardiomegalia/genética , Cardiomegalia/prevenção & controle , Cardiomegalia/metabolismo , Fenilefrina/farmacologia , Fenilefrina/metabolismo , Camundongos Knockout , Ubiquitinação , Miócitos Cardíacos/metabolismo , Camundongos Endogâmicos C57BL , Endopeptidases/metabolismo , Endopeptidases/farmacologia
2.
Lipids Health Dis ; 21(1): 29, 2022 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-35282837

RESUMO

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide. The fat mass and obesity-associated protein (FTO) has been shown to be involved in obesity; however, its role in NAFLD and the underlying molecular mechanisms remain largely unknown. METHODS: FTO expression was first examined in the livers of patients with NAFLD and animal and cellular models of NAFLD by real-time PCR and Western blotting. Next, its role in lipid accumulation in hepatocytes was assessed both in vitro and in vivo via gene overexpression and knockdown studies. RESULTS: FTO expression was obviously elevated in the livers of mice and humans with hepatic steatosis, probably due to its decreased ubiquitination. FTO overexpression in HepG2 cells induced triglyceride accumulation, whereas FTO knockdown exerted an opposing effect. Consistent with the findings of in vitro studies, adeno-associated viruses 8 (AAV8)-mediated FTO overexpression in the liver promoted hepatic steatosis in C57BL/6J mice. Mechanistically, FTO inhibited the mRNA of peroxisome proliferator-activated receptor α (PPARα) in hepatocytes. Activation of PPARα by its agonist GW7647 reversed lipid accumulation in hepatocytes induced by FTO overexpression. CONCLUSIONS: Overall, FTO expression is increased in NAFLD, and it promotes hepatic steatosis by targeting PPARα.


Assuntos
Hepatopatia Gordurosa não Alcoólica , PPAR alfa , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Animais , Dieta Hiperlipídica , Hepatócitos/metabolismo , Humanos , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Obesidade/complicações , Obesidade/genética , Obesidade/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo
3.
Hepatology ; 75(5): 1218-1234, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34591986

RESUMO

BACKGROUND AND AIMS: NAFLD is considered as the hepatic manifestation of the metabolic syndrome, which includes insulin resistance, obesity and hyperlipidemia. NASH is a progressive stage of NAFLD with severe hepatic steatosis, hepatocyte death, inflammation, and fibrosis. Currently, no pharmacological interventions specifically tailored for NASH are approved. Ovarian tumor domain, ubiquitin aldehyde binding 1 (OTUB1), the founding member of deubiquitinases, regulates many metabolism-associated signaling pathways. However, the role of OTUB1 in NASH is unclarified. METHODS AND RESULTS: We demonstrated that mice with Otub1 deficiency exhibited aggravated high-fat diet-induced and high-fat high-cholesterol (HFHC) diet-induced hyperinsulinemia and liver steatosis. Notably, hepatocyte-specific overexpression of Otub1 markedly alleviated HFHC diet-induced hepatic steatosis, inflammatory responses, and liver fibrosis. Mechanistically, we identified apoptosis signal-regulating kinase 1 (ASK1) as a key candidate target of OTUB1 through RNA-sequencing analysis and immunoblot analysis. Through immunoprecipitation-mass spectrometry analysis, we further found that OTUB1 directly bound to tumor necrosis factor receptor-associated factor 6 (TRAF6) and suppressed its lysine 63-linked polyubiquitination, thus inhibiting the activation of ASK1 and its downstream pathway. CONCLUSIONS: OTUB1 is a key suppressor of NASH that inhibits polyubiquitinations of TRAF6 and attenuated TRAF6-mediated ASK1 activation. Targeting the OTUB1-TRAF6-ASK1 axis may be a promising therapeutic strategy for NASH.


Assuntos
Cisteína Endopeptidases/metabolismo , Hepatopatia Gordurosa não Alcoólica , Animais , Dieta Hiperlipídica , Modelos Animais de Doenças , Fígado , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Transdução de Sinais , Fator 6 Associado a Receptor de TNF
4.
J Adv Res ; 26: 123-135, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33133688

RESUMO

INTRODUCTION: In atherosclerotic lesions, extensive inflammation of the vessel wall contributes to plaque instability. Long noncoding RNAs (lncRNAs) play important roles in diverse biological processes in atherosclerosis. OBJECTIVES: Here, we aim to identify the functional role and regulatory mechanisms of lncRNA hypoxia-inducible factor 1 alpha-antisense RNA 2 (HIF1A-AS2) in atherosclerotic inflammation. METHODS: An atherosclerotic mouse model was induced in ApoE-/- mice by high fat diet (HFD). Endothelial cells (ECs), human aortic smooth muscle cells (SMCs) or human coronary artery endothelial cells (HCAECs) were exposed to ox-LDL to develop the in vitro model. The effects of lncRNA HIF1A-AS2 on inflammation were evaluated by determining levels of inflammatory factors tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß) and interleukin-6 (IL-6) and levels of adhesion molecules vascular cell adhesion molecule 1 (VCAM-1), intercellular adhesion molecule 1 (ICAM-1), and macrophage cationic peptide 1 (MCP-1). RESULTS: It was established that lncRNA HIF1A-AS2 and ATF2 were highly expressed in atherosclerotic ApoE-/- mice. Downregulating lncRNA HIF1A-AS2 in ox-LDL-exposed ECs, SMCs and HCAECs inhibited inflammation by reducing levels of pro-inflammatory factors and adhesion molecules. LncRNA HIF1A-AS2 bound to the transcription factor USF1 to elevate ATF2 expression. USF1 overexpression counteracted the suppressive effect of lncRNA HIF1A-AS2 silencing on ox-LDL-induced inflammation. Knockdown of lncRNA HIF1A-AS2 or ATF2 could also attenuate inflammation in atherosclerotic mice. Collectively, the present study demonstrates that downregulation of lncRNA HIF1A-AS2 represses the binding of USF1 to the ATF2 promoter region and then inhibits ATF2 expression, thereby suppressing atherosclerotic inflammation. CONCLUSION: This study suggests lncRNA HIF1A-AS2 as an promising therapeutic target for atherosclerosis.

5.
J Am Heart Assoc ; 9(22): e017751, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33170082

RESUMO

Background Cardiac hypertrophy (CH) is a physiological response that compensates for blood pressure overload. Under pathological conditions, hypertrophy can progress to heart failure as a consequence of the disorganized growth of cardiomyocytes and cardiac tissue. USP10 (ubiquitin-specific protease 10) is a member of the ubiquitin-specific protease family of cysteine proteases, which are involved in viral infection, oxidative stress, lipid drop formation, and heat shock. However, the role of USP10 in CH remains largely unclear. Here, we investigated the roles of USP10 in CH. Methods and Results Cardiac-specific USP10 knockout (USP10-CKO) mice and USP10-transgenic (USP10-TG) mice were used to examined the role of USP10 in CH following aortic banding. The specific functions of USP10 were further examined in isolated cardiomyocytes. USP10 expression was increased in murine hypertrophic hearts following aortic banding and in isolated cardiomyocytes in response to hypertrophic agonist. Mice deficient in USP10 in the heart exhibited exaggerated cardiac hypertrophy and fibrosis following pressure overload stress, which resulted in worsening of cardiac contractile function. In contrast, cardiac overexpression of USP10 protected against pressure overload-induced maladaptive CH. Mechanistically, we demonstrated that USP10 activation and interaction with Sirt6 in response to angiotensin II led to a marked increase in the ubiquitination of Sirt6 and resulted in Akt signaling downregulation and attenuation of cardiomyocyte hypertrophy. Accordingly, inactivation of USP10 reduced Sirt6 abundance and stability and diminished Sirt6-induced downstream signaling in cardiomyocytes. Conclusions USP10 functions as a Sirt6 deubiquitinase that induces cardiac myocyte hypertrophy and triggers maladaptive CH.


Assuntos
Cardiomegalia/etiologia , Sirtuínas/metabolismo , Ubiquitina Tiolesterase/fisiologia , Angiotensina II , Animais , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Técnicas de Cultura de Células , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Transdução de Sinais/fisiologia
6.
Cell Death Dis ; 11(2): 140, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-32080168

RESUMO

Cardiac hypertrophy (CH) is an independent risk factor for many cardiovascular diseases, and is one of the primary causes of morbidity and mortality in elderly people. Pathological CH involves excessive protein synthesis, increased cardiomyocyte size, and ultimately the development of heart failure. Myotubularin-related protein 14 (MTMR14) is a member of the myotubularin (MTM)-related protein family, which is involved in apoptosis, aging, inflammation, and autophagy. However, its exact function in CH is still unclear. Herein, we investigated the roles of MTMR14 in CH. We show that MTMR14 expression was increased in hypertrophic mouse hearts. Mice deficient in heart MTMR14 exhibited an aggravated aortic-banding (AB)-induced CH phenotype. In contrast, MTMR14 overexpression prevented pressure overload-induced hypertrophy. At the molecular level, prevention of CH in the absence of MTMR14 involved elevations in Akt pathway components, which are key elements that regulate apoptosis and cell proliferation. These results demonstrate that MTMR14 is a new molecular target for the treatment of CH.


Assuntos
Hipertrofia Ventricular Esquerda/enzimologia , Miócitos Cardíacos/enzimologia , Monoéster Fosfórico Hidrolases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Proliferação de Células , Tamanho Celular , Modelos Animais de Doenças , Células HEK293 , Humanos , Hipertrofia Ventricular Esquerda/patologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Hipertrofia Ventricular Esquerda/prevenção & controle , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/patologia , Monoéster Fosfórico Hidrolases/deficiência , Monoéster Fosfórico Hidrolases/genética , Ratos Sprague-Dawley , Transdução de Sinais , Função Ventricular Esquerda , Remodelação Ventricular
7.
Int J Biol Sci ; 15(13): 2815-2829, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31853220

RESUMO

Objective: Recent studies demonstrate circulating serum spexin levels are reduced in obesity or type 2 diabetes mellitus (T2DM) patients and may play a role in glucose metabolism. The mechanism underlying is not known. In this study, we explore whether spexin has a role in insulin resistance and hepatic glucose metabolism. Methods: The correlation between serum spexin levels and the homeostasis model assessment of insulin resistance (HOMA-IR) was studied in newly diagnosed T2DM patients. After intraperitoneal injection of exogenous spexin for 8 weeks, the effect of spexin on exogenous glucose infusion rates (GIR), and hepatic glucose production (HGP) were assessed by extended hyperinsulinemic-euglycemic clamp in high-fat-diet (HFD)-induced rats. Glucose concentration with CRISPR/Cas9-mediated disruption of spexin expression in HepG2 cells culture was observed. Expression of transcription factors (Forkhead box O1, FoxO1 and peroxisome proliferator-activated receptor gamma coactivator 1-alpha, PGC-1α) and key enzymes (G-6-Pase and PEPCK) of gluconeogenesis pathway were observed in vitro and in vivo. Results: The serum spexin level was significantly low in newly diagnosed T2DM patients as compared with healthy patients and significantly negatively correlated with the HOMA-IR values. Exogenous spexin treatment resulted in weight loss and decrease of HOMA-IR value in high-fat-diet (HFD)-induced rats. The exogenous glucose infusion rates (GIR) were higher in the HFD + spexin group than that in the HFD group (358 ± 32 vs. 285 ± 24 µmol/kg/min, P < 0.05). Steady-state hepatic glucose production (HGP) was also suppressed by ~50% in the HFD + spexin group as compared with that in the HFD group. Furthermore, spexin inhibited gluconeogenesis in dose-dependent and time-dependent manner in the insulin-resistant cell model. CRISPR/Cas9-mediated knockdown of spexin in HepG2 cells activated gluconeogenesis. Moreover, spexin was shown regulating gluconeogenesis by inhibiting FoxO1/PGC-1α pathway, and key gluconeogenic enzymes, (PEPCK and G-6-Pase) in both HFD-induced rats and insulin-resistant cells. Conclusions: Spexin plays an important role in insulin resistance in HFD-induced rats and insulin-resistant cells. Regulation of the effects of spexin on insulin resistance may hold therapeutic value for metabolic diseases.


Assuntos
Proteína Forkhead Box O1/metabolismo , Gluconeogênese/fisiologia , Resistência à Insulina/fisiologia , Fígado/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Adulto , Idoso , Animais , Dieta Hiperlipídica/efeitos adversos , Feminino , Cromatografia Gasosa-Espectrometria de Massas , Gluconeogênese/genética , Células Hep G2 , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
8.
Hepatology ; 70(4): 1099-1118, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30820969

RESUMO

Nonalcoholic fatty liver disease (NAFLD) has become the most common cause of chronic liver disease worldwide. Due to the growing economic burden of NAFLD on public health, it has become an emergent target for clinical intervention. DUSP12 is a member of the dual specificity phosphatase (DUSP) family, which plays important roles in brown adipocyte differentiation, microbial infection, and cardiac hypertrophy. However, the role of DUSP12 in NAFLD has yet to be clarified. Here, we reveal that DUSP12 protects against hepatic steatosis and inflammation in L02 cells after palmitic acid/oleic acid treatment. We demonstrate that hepatocyte specific DUSP12-deficient mice exhibit high-fat diet (HFD)-induced and high-fat high-cholesterol diet-induced hyperinsulinemia and liver steatosis and decreased insulin sensitivity. Consistently, DUSP12 overexpression in hepatocyte could reduce HFD-induced hepatic steatosis, insulin resistance, and inflammation. At the molecular level, steatosis in the absence of DUSP12 was characterized by elevated apoptosis signal-regulating kinase 1 (ASK1), which mediates the mitogen-activated protein kinase (MAPK) pathway and hepatic metabolism. DUSP12 physically binds to ASK1, promotes its dephosphorylation, and inhibits its action on ASK1-related proteins, JUN N-terminal kinase, and p38 MAPK in order to inhibit lipogenesis under high-fat conditions. Conclusion: DUSP12 acts as a positive regulator in hepatic steatosis and offers potential therapeutic opportunities for NAFLD.


Assuntos
Apoptose/genética , Fosfatases de Especificidade Dupla/genética , Regulação da Expressão Gênica , MAP Quinase Quinase Quinase 5/genética , Hepatopatia Gordurosa não Alcoólica/genética , Análise de Variância , Animais , Células Cultivadas , Dieta Hiperlipídica , Modelos Animais de Doenças , Regulação para Baixo , Humanos , Resistência à Insulina/genética , Metabolismo dos Lipídeos/genética , Lipogênese/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Distribuição Aleatória , Valores de Referência , Transdução de Sinais/genética
9.
Braz J Med Biol Res ; 51(8): e7299, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29924135

RESUMO

Non-alcoholic fatty liver disease (NAFLD) is a common disease associated with metabolic syndrome and can lead to life-threatening complications like hepatic carcinoma and cirrhosis. Exenatide, a glucagon-like peptide-1 (GLP-1) receptor agonist antidiabetic drug, has the capacity to overcome insulin resistance and attenuate hepatic steatosis but the specific underlying mechanism is unclear. This study was designed to investigate the underlying molecular mechanisms of exenatide therapy on NAFLD. We used in vivo and in vitro techniques to investigate the protective effects of exenatide on fatty liver via fat mass and obesity associated gene (FTO) in a high-fat (HF) diet-induced NAFLD animal model and related cell culture model. Exenatide significantly decreased body weight, serum glucose, insulin, insulin resistance, serum free fatty acid, triglyceride, total cholesterol, low-density lipoprotein, aspartate aminotransferase, and alanine aminotransferase levels in HF-induced obese rabbits. Histological analysis showed that exenatide significantly reversed HF-induced lipid accumulation and inflammatory changes accompanied by decreased FTO mRNA and protein expression, which were abrogated by PI3K inhibitor LY294002. This study indicated that pharmacological interventions with GLP-1 may represent a promising therapeutic strategy for NAFLD.


Assuntos
Dioxigenase FTO Dependente de alfa-Cetoglutarato/efeitos dos fármacos , Fígado Gorduroso/metabolismo , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Peptídeos/farmacologia , Substâncias Protetoras/farmacologia , Peçonhas/farmacologia , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Animais , Glicemia/análise , Peso Corporal/efeitos dos fármacos , Cromonas/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Ingestão de Alimentos/efeitos dos fármacos , Inibidores Enzimáticos/metabolismo , Exenatida , Fígado Gorduroso/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas In Vitro , Insulina/sangue , Masculino , Malondialdeído/análise , Morfolinas/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Obesidade/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Coelhos , Superóxido Dismutase/análise
10.
Lipids Health Dis ; 17(1): 73, 2018 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-29631603

RESUMO

BACKGROUND: The increasing prevalence of non-alcoholic fatty liver disease (NAFLD) has followed the international rise in obesity rates. Multiple mechanisms are involved in NAFLD, including endoplasmic reticulum stress and oxidative stress. Heat shock protein 70 (HSP70), which is abundant in most organisms, is sensitive to stress. However, the role of HSP70 in NAFLD has not been investigated. Here, we investigated the possible role of HSP70 in lipid synthesis. METHODS: C57BL/6 mice were fed a high-fat diet, and HepG2 cells were treated with 0.5 mM palmitic acid (PA). HSP70 expression was detected by qPCR, Western blot and immunohistochemistry. Total cholesterol (TC) and triglyceride (TG) levels were detected by enzyme-linked immunosorbent assay (ELISA). After Hsp70 overexpression and knockdown, TC and TG levels and FAS, SCD, and ACC expression were detected. RESULTS: HSP70 expression was significantly increased in the livers of obese mice. In vitro, HSP70 expression was markedly induced by PA in HepG2 cells. Notably, HSP70 overexpression in HepG2 cells enhanced TC and TG synthesis, in parallel with the upregulation of lipogenic genes, including FAS, SCD and ACC. By contrast, HSP70 knockdown decreased the levels of cellular lipids and the expression of FAS, SCD, and ACC in HepG2 cells. Together, our results suggest that HSP70 may promote lipogenesis in HepG2 cells. CONCLUSIONS: Heat shock protein 70 promotes lipogenesis in HepG2 cells.


Assuntos
Proteínas de Choque Térmico HSP70/metabolismo , Lipogênese/fisiologia , Animais , Dieta Hiperlipídica , Enzimas/genética , Enzimas/metabolismo , Técnicas de Silenciamento de Genes , Proteínas de Choque Térmico HSP70/genética , Células Hep G2 , Humanos , Metabolismo dos Lipídeos/fisiologia , Lipogênese/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/metabolismo , Ácido Palmítico/farmacologia , Triglicerídeos/metabolismo , Regulação para Cima
11.
Biomacromolecules ; 19(2): 438-448, 2018 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-29334216

RESUMO

In this paper, reconstituted high-density lipoprotein (rHDL), a lipoprotein-derived nanovector, was constructed for codelivery of paclitaxel (PTX) and wild-type p53 gene (p53). The particle size and the zeta potential of PTX-DODAB/p53-rHDL nanoparticles were 177.2 nm and -20.06 mV, respectively. Meanwhile, they exhibited great serum stability and satisfactory sustained release characteristics in vitro. PTX-DODAB/pDNA-rHDL nanoparticles simultaneously improved the cellular uptake of PTX and pDNA via scavenger receptor B type I (SR-BI) mediated lysosome-independent internalization and promoted the transfection of pDNA in MCF-7 cells, which were revealed by flow cytometry and confocal laser scanning microscopy analyses. The high p53 protein expression in MCF-7 cells after rHDL-mediated transfection was detected by Western blotting assay. Moreover, PTX-DODAB/p53-rHDL nanoparticles showed superior cytotoxicity and significantly induced apoptosis in SR-BI overexpressed MCF-7 cells. In in vivo studies, PTX-DODAB/p53-rHDL nanoparticles without obvious toxic effects to vessels, blood, or major organs exhibited efficient tumor targeting and encouraging antitumor effects on tumor-bearing nude mice compared with controls. All the results above indicated that PTX-DODAB/p53-rHDL nanoparticles held broad prospects in combination of chemotherapeutics and gene therapeutic agents for cancer-targeted therapy.


Assuntos
Antineoplásicos/administração & dosagem , Apoptose , Terapia Genética/métodos , Lipoproteínas HDL/química , Neoplasias Mamárias Experimentais/terapia , Nanopartículas/química , Animais , Antineoplásicos/uso terapêutico , Feminino , Humanos , Células MCF-7 , Neoplasias Mamárias Experimentais/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/efeitos adversos , Paclitaxel/administração & dosagem , Paclitaxel/uso terapêutico , Compostos de Amônio Quaternário/química , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
12.
Braz. j. med. biol. res ; 51(8): e7299, 2018. graf
Artigo em Inglês | LILACS | ID: biblio-951744

RESUMO

Non-alcoholic fatty liver disease (NAFLD) is a common disease associated with metabolic syndrome and can lead to life-threatening complications like hepatic carcinoma and cirrhosis. Exenatide, a glucagon-like peptide-1 (GLP-1) receptor agonist antidiabetic drug, has the capacity to overcome insulin resistance and attenuate hepatic steatosis but the specific underlying mechanism is unclear. This study was designed to investigate the underlying molecular mechanisms of exenatide therapy on NAFLD. We used in vivo and in vitro techniques to investigate the protective effects of exenatide on fatty liver via fat mass and obesity associated gene (FTO) in a high-fat (HF) diet-induced NAFLD animal model and related cell culture model. Exenatide significantly decreased body weight, serum glucose, insulin, insulin resistance, serum free fatty acid, triglyceride, total cholesterol, low-density lipoprotein, aspartate aminotransferase, and alanine aminotransferase levels in HF-induced obese rabbits. Histological analysis showed that exenatide significantly reversed HF-induced lipid accumulation and inflammatory changes accompanied by decreased FTO mRNA and protein expression, which were abrogated by PI3K inhibitor LY294002. This study indicated that pharmacological interventions with GLP-1 may represent a promising therapeutic strategy for NAFLD.


Assuntos
Animais , Masculino , Coelhos , Peptídeos/farmacologia , Peçonhas/farmacologia , Substâncias Protetoras/farmacologia , Fígado Gorduroso/metabolismo , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Dioxigenase FTO Dependente de alfa-Cetoglutarato/efeitos dos fármacos , Glicemia/análise , Peso Corporal/efeitos dos fármacos , Técnicas In Vitro , Regulação da Expressão Gênica/efeitos dos fármacos , Morfolinas/metabolismo , Cromonas/metabolismo , Modelos Animais de Doenças , Ingestão de Alimentos/efeitos dos fármacos , Inibidores Enzimáticos/metabolismo , Fígado Gorduroso/patologia , Dieta Hiperlipídica , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Exenatida , Insulina/sangue , Malondialdeído/análise , Obesidade/metabolismo
13.
Chin Med J (Engl) ; 127(21): 3771-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25382334

RESUMO

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is a complex disorder and has been closely linked to obesity. The fat mass and obesity-associated (FTO) gene is a newly discovered gene related to obesity, which enhances oxidative stress and lipogenesis in NAFLD. The forkhead transcription factor O1 (FoxO1) is another important gene involved in NAFLD, which causes lipid disorders when insulin resistance appears in the liver. However, the interactions between FTO and FoxO1 during the pathogenesis of NAFLD have not been fully elucidated. This study was designed to identify the relationship between these two factors that are involved in the development of NAFLD. METHODS: This study includes two parts referred to as animal and cell experiments. Twelve female SPF C57BL/6 mice were fed a high-fat diet to serve as an NAFLD animal model. Aspartate aminotransferase (AST), alanine aminotransferase (ALT), total triglyceride (TG), total cholesterol (TC), alkaline phosphatase (ALP), high-density lipoprotein (HDL), and low-density lipoprotein (LDL) were measured. Immunohistochemical analysis was used to detect the expression and histological localization of FTO, FoxO1, and adenosine monophosphate (AMP)-activated protein kinase (AMPK). The L02 cells were exposed to high fat for 24, 48, or 72 hours. Oil red O staining was used to detect intracellular lipid droplets. Reverse transcription-polymerase chain reaction was used for analyzing the levels of FTO and FoxO1 mRNA. RESULTS: At the end of 10 weeks, ALP, ALT, AST, and LDL were significantly increased (P < 0.01), while TC and TG were also significantly higher (P < 0.05). In addition, HDL was significantly decreased (P < 0.05). The FTO and FoxO1 proteins were weakly expressed in the control group, but both FTO and FoxO1 were expressed significantly higher (P < 0.01) in the experimental group, and the expression of the two factors was significantly correlated. AMPK in the high-fat group showed a low level of correlation with FTO, but not with FoxO1. Oil Red O staining results showed that the cells cultured in 50% fetal bovine serum for 24, 48, or 72 hours exhibited steatosis. FTO and FoxO1 mRNA were increased in the high-fat group compared with the normal group (P < 0.01). The expression levels of FTO and FoxO1 mRNA were the highest at 48 hours (P < 0.05). CONCLUSIONS: A high-fat diet leads to higher expression of FTO, phosphorylation of FoxO1, and decreased phosphorylation of AMPK. These results suggest that the interactions between FTO and FoxO1 are closely related to the pathogenesis of NAFLD.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Obesidade/metabolismo , Alanina Transaminase/genética , Alanina Transaminase/metabolismo , Animais , Aspartato Aminotransferases/genética , Aspartato Aminotransferases/metabolismo , Colesterol/metabolismo , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Feminino , Fatores de Transcrição Forkhead/genética , Lipoproteínas HDL/metabolismo , Lipoproteínas LDL/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/patologia , Triglicerídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...