Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Biochim Biophys Acta Mol Basis Dis ; 1867(12): 166261, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34455055

RESUMO

Rapamycin insensitive companion of mechanistic target of Rapamycin (Rictor), the key component of mTOR complex 2 (mTORC2), controls both ß-cell proliferation and function. We sought to study whether long chain acyl-CoA synthetase 4 (Acsl4) worked downstream of Rictor/mTORC2 to maintain ß-cell functional mass. We found Acsl4 was positively regulated by Rictor at transcriptional and posttranslational levels in mouse ß-cell. Infecting adenovirus expressing Acsl4 in ß-cell-specific-Rictor-knockout (ßRicKO) islets and Min6 cells knocking down Rictor with lentivirus-expressing siRNA-oligos targeting Rictor(siRic), recovered the ß-cell dysplasia but not dysfunction. Cell bioenergetic experiment performed with Seahorse XF showed that Acsl4 could not rescue the dampened glucose oxidation in Rictor-lacking ß-cell, but further promoted lipid oxidation. Transposase-Accessible Chromatin (ATAC) and H3K27Ac chromatin immunoprecipitation (ChIP) sequencing studies reflected the epigenetic elevated molecular signature for ß-cell dedifferentiation and mitigated oxidative defense/response. These results were confirmed by the observations of elevated acetylation and ubiquitination of FoxO1, increased protein levels of Gpx1 and Hif1an, excessive reactive oxygen species (ROS) production and diminished MafA in Acsl4 overexpressed Rictor-lacking ß-cells. In these cells, antioxidant treatment significantly recovered MafA level and insulin content. Inducing lipid oxidation alone could not mimic the effect of Acsl4 in Rictor lacking ß-cell. Our study suggested that Acsl4 function in ß-cell was context dependent and might facilitate ß-cell dedifferentiation with attenuated Rictor/mTORC2 activity or insulin signaling via posttranslational inhibiting FoxO1 and epigenetically enhancing ROS induced MafA degradation.


Assuntos
Desdiferenciação Celular/genética , Coenzima A Ligases/genética , Proteína Forkhead Box O1/genética , Células Secretoras de Insulina/metabolismo , Proteína Companheira de mTOR Insensível à Rapamicina/genética , Animais , Proliferação de Células/genética , Epigenômica , Regulação da Expressão Gênica/genética , Glutationa Peroxidase/genética , Humanos , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/patologia , Metabolismo dos Lipídeos/genética , Alvo Mecanístico do Complexo 2 de Rapamicina/genética , Camundongos , Oxigenases de Função Mista/genética , Espécies Reativas de Oxigênio/metabolismo , Glutationa Peroxidase GPX1
2.
Front Microbiol ; 12: 797062, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35185821

RESUMO

Studies have shown that the cholesterol-lowering medicine statins alter the gut microbiome, induce chronic metabolic inflammation, and disrupt glycemic homeostasis. In this study, we aimed to investigate whether effects of atorvastatin (Ator) on gut microbiome and metabolic inflammation could be causally correlated. Mice at 8-week age were fed with high-fat diet (HFD) or HFD with Ator (HFD+Ator) for 16 weeks. 16S rRNA sequencing of stool and RNA sequencing of colon tissue were employed to analyze the intestinal alterations that could be induced by Ator. A human colon carcinoma cell line (Caco2) was used for in vitro experiments on barrier function. Compared to HFD, HFD+Ator induced more weight gain, impaired glucose tolerance, and led to gut microbiota dysbiosis, such as suppressing Akkermansia muciniphila in mice. The expressions of tight junction (TJ) proteins were attenuated in the colon, and the serum LPS-binding-protein (LBP) level was elevated in HFD+Ator mice, so as to transcriptionally activate the intestinal nuclear factor-k-gene binding (NF-κB) signaling pathway. Consistently, Ator impaired the barrier function of Caco2, and treatment of supernatant of A. Muciniphila culture could decrease the intestinal permeability and recover the attenuated expression of TJ proteins induced by Ator. In conclusion, long-term use of Ator with HFD may alter gut microbiota, induce intestinal barrier dysfunction, and hence promote chronic inflammation that contributes to disrupted glycemic homeostasis.

4.
Clin Endocrinol (Oxf) ; 92(4): 350-357, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31954081

RESUMO

OBJECTIVE: Polycystic ovary syndrome (PCOS) is associated with an increased prevalence of dysglycaemia, which includes impaired glucose tolerance and type 2 diabetes mellitus (T2DM). Patients with PCOS demonstrate abnormal patterns of steroid hormones. Here, we analyse the correlation between glucose metabolism and serum steroid hormones in PCOS. DESIGN: Observational double-centre study. PATIENTS: 914 patients with PCOS. MEASUREMENTS: We assessed the glucose metabolism status of all patients according to the 1999 WHO criteria. Serum steroid hormones were measured by liquid chromatography-tandem mass spectrometry. RESULTS: The median age of the patients was 26 years (interquartile range: 21-30), and 40.6% (371/914) had abnormal glucose metabolism: 29.3% (268/914) had prediabetes, and 11.3% (103/914) had T2DM. Correlation analysis not adjusting for confounding factors revealed that serum aldosterone, androstenedione, oestrone, pregnenolone and the free androgen index were positively correlated, while progesterone was negatively correlated with the risk of abnormal glucose metabolism. After adjusting for age, body mass index and fasting insulin levels in the logistic regression model, only aldosterone (P = .013), androstenedione (P = .046) and oestrone (P = .014; in quartiles) were correlated with the risk of abnormal glucose metabolism. CONCLUSIONS: This study indicates a high prevalence of prediabetes and T2DM in patients with PCOS. Furthermore, there were positive correlations of serum aldosterone, androstenedione and oestrone with the risk of abnormal glucose metabolism after adjusting for confounding factors.


Assuntos
Diabetes Mellitus Tipo 2 , Síndrome do Ovário Policístico , Androgênios , Índice de Massa Corporal , Feminino , Glucose , Humanos , Recém-Nascido , Esteroides
5.
Diabetes ; 69(1): 48-59, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31649162

RESUMO

Statins are cholesterol-lowering agents that increase the incidence of diabetes and impair glucose tolerance via their detrimental effects on nonhepatic tissues, such as pancreatic islets, but the underlying mechanism has not been determined. In atorvastatin (ator)-treated high-fat diet-fed mice, we found reduced pancreatic ß-cell size and ß-cell mass, fewer mature insulin granules, and reduced insulin secretion and glucose tolerance. Transcriptome profiling of primary pancreatic islets showed that ator inhibited the expression of pancreatic transcription factor, mechanistic target of rapamycin (mTOR) signaling, and small G protein (sGP) genes. Supplementation of the mevalonate pathway intermediate geranylgeranyl pyrophosphate (GGPP), which is produced by 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase, significantly restored the attenuated mTOR activity, v-maf musculoaponeurotic fibrosarcoma oncogene homolog A (MafA) expression, and ß-cell function after ator, lovastatin, rosuvastatin, and fluvastatin treatment; this effect was potentially mediated by sGP prenylation. Rab5a, the sGP in pancreatic islets most affected by ator treatment, was found to positively regulate mTOR signaling and ß-cell function. Rab5a knockdown mimicked the effect of ator treatment on ß-cells. Thus, ator impairs ß-cell function by regulating sGPs, for example, Rab5a, which subsequently attenuates islet mTOR signaling and reduces functional ß-cell mass. GGPP supplementation could constitute a new approach for preventing statin-induced hyperglycemia.


Assuntos
Atorvastatina/farmacologia , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Ácido Mevalônico/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Contagem de Células , Células Cultivadas , Feminino , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/crescimento & desenvolvimento , Masculino , Redes e Vias Metabólicas/genética , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão/genética , Fosfatos de Poli-Isoprenil/farmacologia , Transdução de Sinais/genética
6.
Nat Med ; 23(7): 859-868, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28628112

RESUMO

Emerging evidence has linked the gut microbiome to human obesity. We performed a metagenome-wide association study and serum metabolomics profiling in a cohort of lean and obese, young, Chinese individuals. We identified obesity-associated gut microbial species linked to changes in circulating metabolites. The abundance of Bacteroides thetaiotaomicron, a glutamate-fermenting commensal, was markedly decreased in obese individuals and was inversely correlated with serum glutamate concentration. Consistently, gavage with B. thetaiotaomicron reduced plasma glutamate concentration and alleviated diet-induced body-weight gain and adiposity in mice. Furthermore, weight-loss intervention by bariatric surgery partially reversed obesity-associated microbial and metabolic alterations in obese individuals, including the decreased abundance of B. thetaiotaomicron and the elevated serum glutamate concentration. Our findings identify previously unknown links between intestinal microbiota alterations, circulating amino acids and obesity, suggesting that it may be possible to intervene in obesity by targeting the gut microbiota.


Assuntos
DNA Bacteriano/análise , Disbiose/microbiologia , Microbioma Gastrointestinal/genética , Metaboloma , Obesidade/microbiologia , Adiposidade , Adulto , Animais , Bacteroides/genética , Bacteroides thetaiotaomicron/genética , Cirurgia Bariátrica , Estudos de Casos e Controles , Disbiose/metabolismo , Feminino , Fusobacterium/genética , Gastrectomia , Ácido Glutâmico/sangue , Humanos , Masculino , Metagenoma , Camundongos , Obesidade/metabolismo , Obesidade/cirurgia , Aumento de Peso , Adulto Jovem
7.
Biochim Biophys Acta Gen Subj ; 1861(8): 2039-2047, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28435021

RESUMO

BACKGROUND: Compensation of the pancreatic ß cell functional mass in response to metabolic stress is key to the pathogenesis of Type 2 Diabetes. The mTORC2 pathway governs fuel metabolism and ß cell functional mass. It is unknown whether mTORC2 is required for regulating metabolic stress-induced ß cell compensation. METHODS: We challenged four-week-old ß-cell-specific Rictor (a key component of mTORC2)-knockout mice with a high fat diet (HFD) for 4weeks and measured metabolic and pancreatic morphological parameters. We performed ex vivo experiments to analyse ß cell insulin secretion and electrophysiology characteristics. Adenoviral-mediated overexpression and lentiviral-ShRNA-mediated knocking down proteins were applied in Min6 cells and cultured primary mouse islets. RESULTS: ßRicKO mice showed a significant glucose intolerance and a reduced plasma insulin level and an unchanged level ß cell mass versus the control mice under HFD. A HFD or palmitate treatment enhanced both glucose-induced insulin secretion (GIIS) and the PMA (phorbol 12-myristate 13-acetate)-induced insulin secretion in the control islets but not in the ßRicKO islets. The KO ß cells showed similar glucose-induced Ca2+ influx but lower membrane capacitance increments versus the control cells. The enhanced mTORC2/PKC proteins levels in the control HFD group were ablated by Rictor deletion. Replenishing PKCα by overexpression of PKCα-T638D restored the defective GIIS in ßRicKO islets. CONCLUSIONS: The mTORC2/Rictor pathway modulates ß cell compensatory GIIS under nutrient overload mediated by its phosphorylation of PKCα. GENERAL SIGNIFICANCE: This study suggests that the mTORC2/PKC pathway in ß cells is involved in the pathogenesis of T2D.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Complexos Multiproteicos/fisiologia , Proteína Quinase C-alfa/fisiologia , Transdução de Sinais/fisiologia , Estresse Fisiológico/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Animais , Diabetes Mellitus Tipo 2/etiologia , Dieta Hiperlipídica , Secreção de Insulina , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Acetato de Tetradecanoilforbol/farmacologia
8.
J Endocrinol ; 229(2): 73-83, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26869333

RESUMO

The cellular and molecular mechanisms of glucose-stimulated ß-cell proliferation are poorly understood. Recently, secreted frizzled-related protein 5 (encoded by Sfrp5; a Wnt signaling inhibitor) has been demonstrated to be involved in ß-cell proliferation in obesity. A previous study demonstrated that glucose enhanced Wnt signaling to promote cell proliferation. We hypothesized that inhibition of SFRP5 contributes to glucose-stimulated ß-cell proliferation. In this study, we found that the Sfrp5 level was significantly reduced in high glucose-treated INS-1 cells, primary rat ß-cells, and islets isolated from glucose-infused rats. Overexpression of SFRP5 diminished glucose-stimulated proliferation in both INS-1 cells and primary ß-cells, with a concomitant inhibition of the Wnt signaling pathway and decreased cyclin D2 expression. In addition, we showed that glucose-induced Sfrp5 suppression was modulated by the PI3K/AKT pathway. Therefore, we conclude that glucose inhibits Sfrp5 expression via the PI3K/AKT pathway and hence promotes rat pancreatic ß-cell proliferation.


Assuntos
Adipocinas/metabolismo , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Adipocinas/genética , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Ciclina D2/metabolismo , Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Masculino , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Regulação para Cima , Via de Sinalização Wnt
9.
Exp Ther Med ; 10(1): 67-73, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26170914

RESUMO

Cardiovascular complications are the major causes of morbidity and mortality associated with Type 2 Diabetes. Among the macrovascular complications, diabetic cardiomyopathy (DCM) is generally considered to be inadequately recognized and managed. Bcl-2/adenovirus E1B 19 kDa-interacting protein 3 (BNIP3), is known to play a key role in the initiation of the mitochondrial pathway of apoptosis induced by hypoxia and acidosis in the heart. It is unknown whether BNIP3 is also important for cardiac cell survival or adaption in response to hyperglycemia. Based on the previous finding that BNIP3 was significantly induced in the diabetic rat heart, BNIP3 was transfected in primary rat cardiomyocytes and the H9c2 cell line in the present study. Overexpressed BNIP3 decreased the mitochondrial membrane potential and induced cell apoptosis. When BNIP3 was knocked down, the effect on cell apoptosis was reversed. Transcriptome analysis showed that the genes regulating mitochondrial metabolism, such as carnitine palmitoyltransferase 1b, cytochrome c oxidase subunit VIIIb and creatine kinase (brain), and those regulating cardiac fibrosis, such as matrix metallopeptidase 9, could be the targets of BNIP3 in rat cardiomyocytes. In conclusion, hyperglycemia-induced BNIP3 expression may compromise cardiac cell survival and function. Under the diabetic condition, BNIP3 could be involved in the regulation of mitochondrial function, lipid metabolism and fibrosis. BNIP3 could therefore serve as a potential drug target against diabetic macrovascular complications and, in particular, DCM.

10.
Endocrinology ; 156(1): 48-57, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25343275

RESUMO

The tumor suppressor menin is recognized as a key regulator of ß-cell proliferation. To induce tumorigenesis within the pancreatic ß-cells, floxed alleles of Men1 were selectively ablated using Cre-recombinase driven by the insulin promoter. Despite the ß-cell specificity of the RipCre, glucagon-expressing tumors as well as insulinomas developed in old mutant mice. These glucagon-expressing tumor cells were menin deficient and expressed the mature α-cell-specific transcription factors Brain-specific homeobox POU domain protein 4 (Brn4) and v-maf musculoaponeurotic fibrosarcoma oncogene family, protein B (MafB). Moreover, the inactivation of ß-cell-specific transcription factors was observed in mutant ß-cells. Our work shows that Men1 ablation in the pancreatic ß-cells leads to the inactivation of specific transcription factors, resulting in glucagon-expressing tumor development, which sheds light on the mechanisms of islet tumorigenesis.


Assuntos
Regulação Neoplásica da Expressão Gênica/fisiologia , Glucagon/metabolismo , Glucagonoma/metabolismo , Células Secretoras de Insulina/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogênicas/genética , Animais , Feminino , Deleção de Genes , Genótipo , Células Secretoras de Glucagon/fisiologia , Glucagonoma/genética , Masculino , Camundongos , Camundongos Knockout , Neoplasias Pancreáticas/genética , Fatores de Transcrição
11.
PLoS One ; 8(4): e62399, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23626817

RESUMO

BACKGROUND: To date, the available non-invasive remedies for primary aldosteronism are not satisfactory in clinical practice. The phosphoinositide 3-kinase (PI3Ks)/protein kinase B (PKB or AKT)/mammalian target of rapamycin (mTOR) signaling pathway is essential for tumorigenesis and metastasis in many types of human tumors, including renal cancer, adrenal carcinoma and pheochromocytoma. The possibility that this pathway is also necessary for the pathogenesis of primary aldosteronism has not yet been explored. To answer this question, we investigated the activity of the PI3K/AKT/mTOR signaling pathway in normal adrenal glands (NAGs), primary aldosteronism (PA) patients and NCI-H295R cells. METHODOLOGY/PRINCIPAL FINDINGS: Between January 2005 and December 2011, we retrospectively reviewed the records of 45 patients with PA. We compared clinical characteristics (age, gender and biochemical data) and the expression of phospho-AKT (p-AKT), phospho-mTOR (p-mTOR), phospho-S6 (p-S6) and vascular endothelial growth factor (VEGF) by immunohistochemical staining and western blotting, analyzing 30 aldosterone-producing adenomas (APAs), 15 idiopathic hyperaldosteronism (IHA) tissues and 12 NAGs following nephrectomy for renal tumors (control group). Compared with the control group, most of the PA patients presented with polydipsia, polyuria, resistant hypertension, profound hypokalemia, hyperaldosteronemia and decreased plasma renin activity. Compared with normal zona glomerulosa, the levels of p-AKT, p-mTOR, p-S6 and VEGF were significantly upregulated in APA and IHA. No significant differences were found between APA and IHA in the expression of these proteins. Additionally, positive correlations existed between the plasma aldosterone levels and the expression of p-AKT and p-mTOR. In vitro studies showed that mTOR inhibitor rapamycin could inhibit cell proliferation in NCI-H295R cells in a dose- and time-dependent manner. Furthermore, this inhibitor also decreased aldosterone secretion. CONCLUSIONS: Our data suggest that the PI3K/AKT/mTOR signaling pathway, which was overactivated in APA and IHA compared with normal zona glomerulosa, may mediate aldosterone hypersecretion and participate in the development of PA.


Assuntos
Hiperaldosteronismo/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Glândulas Suprarrenais/metabolismo , Glândulas Suprarrenais/patologia , Adulto , Aldosterona/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Hiperaldosteronismo/etiologia , Hiperaldosteronismo/patologia , Masculino , Pessoa de Meia-Idade , Sirolimo/farmacologia
12.
J Diabetes ; 5(4): 421-8, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23590680

RESUMO

BACKGROUND: Glucagon-like peptide-1 (GLP-1) analogues have emerged as insulin secretagogues and are widely used in type 2 diabetic patients. GLP-1 analogues also demonstrate a promotion of beta cell proliferation and reduction of apoptosis in rodents. In the present study, we investigated the protection of pancreatic beta cells by early use (at the age of 2 weeks) of GLP-1 analogue, liraglutide in Gato-Kakizaki (GK) rats and explored the underlying mechanisms. METHODS: The effects of liraglutide on glucose tolerance were evaluated by intraperitoneal glucose tolerance test (IPGTT) and insulin release tests (IRT). Ki67 and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) immunostaining, Western blots and real-time polymerase chain reaction were applied to evaluate cell proliferation, apoptosis and related gene expressions. RESULTS: Our results demonstrated that early use of liraglutide improved glucose tolerance during liraglutide treatment in GK rats. Liraglutide increased pancreatic insulin contents and markedly reduced beta cell apoptosis. Liraglutide also downregulated pro-apoptotic gene expressions and reduced intra-islet macrophage infiltration. CONCLUSIONS: This experiment reported for the first time that early use of liraglutide could protect beta cell failure in pre-diabetic GK rats through reduction of beta cell apoptosis and ameliorating islet inflammation.


Assuntos
Citoproteção/efeitos dos fármacos , Diabetes Mellitus Experimental/prevenção & controle , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Intolerância à Glucose/tratamento farmacológico , Hipoglicemiantes/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Células Secretoras de Insulina/fisiologia , Liraglutida , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Masculino , Ratos , Fatores de Tempo
13.
Diabetes ; 60(3): 827-37, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21266327

RESUMO

OBJECTIVE: We examined the role of Rictor/mammalian target of rapamycin complex 2 (mTORC2), a key component of the phosphotidylinositol-3-kinase (PI3K)/mTORC2/AKT signaling pathway, in regulating both ß-cell mass and function. RESEARCH DESIGN AND METHODS: Mice with ß-cell-specific deletions of Rictor or Pten were studied to determine the effects of deleting either or both genes on ß-cell mass and glucose homeostasis. RESULTS: Rictor null mice exhibited mild hyperglycemia and glucose intolerance caused by a reduction in ß-cell mass, ß-cell proliferation, pancreatic insulin content, and glucose-stimulated insulin secretion. Islets from these mice exhibited decreased AKT-S473 phosphorylation and increased abundance of FoxO1 and p27 proteins. Conversely, Pten null (ßPtenKO) mice exhibited an increase in ß-cell mass caused by increased cellular proliferation and size. Although ß-cell mass was normal in mice lacking both Rictor and Pten (ßDKO), their ß-cells were larger than those in the ßPtenKO mice. Even though the ß-cell proliferation rate in the ßDKO mice was lower than in the ßPtenKO mice, there was a 12-fold increase the phosphorylation of AKT-T308. CONCLUSIONS: PI3K/AKT signaling through mTORC2/pAKT-S473 plays a key role in maintaining normal ß-cell mass. The phosphorylation of AKT-S473, by negatively regulating that of AKT-T308, is essential for maintaining a balance between ß-cell proliferation and cell size in response to proliferative stimuli.


Assuntos
Proteínas de Transporte/metabolismo , Proliferação de Células , Tamanho Celular , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Transdução de Sinais/fisiologia , Alelos , Análise de Variância , Animais , Apoptose/fisiologia , Western Blotting , Proteínas de Transporte/genética , Imunofluorescência , Genótipo , Glucose/metabolismo , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Hiperglicemia/genética , Hiperglicemia/metabolismo , Camundongos , Camundongos Knockout , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Fosforilação/fisiologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína Companheira de mTOR Insensível à Rapamicina , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Endocr J ; 56(1): 55-63, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18840928

RESUMO

We previously found that cathepsin K (CTSK) played an important role in adipocyte differentiation. However, the underlying molecular mechanism is not clear. Through the time window study, it was observed that CTSK activities were required mainly in the early phases of adipogenic process. At the same time, the expression of type I collagen disappeared. However, type I collagen can still be observed during the whole process when the CTSK inhibitor-E64 was added. The mRNA levels of peroxisome proliferator-activated receptor gamma (PPAR-gamma) and CCAAT/enhancer binding protein alpha (C/EBP-alpha) was also declining. These imply that CTSK may play a role in adipogenesis in early differentiation phases and produce an effect at least partly by degrading type I collagen, which may provides a basis for developing novel therapeutic approaches to treat obesity and the diseases associated with it.


Assuntos
Adipócitos/fisiologia , Catepsinas/fisiologia , Diferenciação Celular , Colágeno Tipo I/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Animais , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Catepsina K , Catepsinas/antagonistas & inibidores , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Colágeno Tipo I/fisiologia , Inibidores de Cisteína Proteinase/farmacologia , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Leucina/análogos & derivados , Leucina/farmacologia , Camundongos , PPAR gama/genética , PPAR gama/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Fatores de Tempo
15.
Life Sci ; 81(2): 160-5, 2007 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-17544010

RESUMO

Changes in AMP-activated protein kinase (AMPK) activity contribute to the regulation of insulin secretion. Troglitazone has been shown to lower serum insulin levels and protect beta cell function. The aim of the present study was to examine the effects of troglitazone on AMPK activity and insulin secretion in beta cells. Isolated rat islets and MIN6 cells were treated for a short (1 h) or a long time (20 h) with troglitazone. One-hour troglitazone treatment activated AMPK and inhibited both glucose-stimulated insulin secretion (GSIS) and the response of insulin secretion to combined stimuli of glucose and palmitate. Long (20 h) treatment with troglitazone caused a sustained phosphorylation of AMPK and acetyl-CoA carboxylase, and increased GSIS after withdrawal of the drug. This study provided evidence that troglitazone activated AMPK in beta cells. In addition to the insulin-sensitizing effects in peripheral tissues, troglitazone also directly inhibits insulin hypersecretion by the elevated glucose and fatty acids, and thus protects beta cells from glucolipotoxicity.


Assuntos
Cromanos/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Hipoglicemiantes/farmacologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Tiazolidinedionas/farmacologia , Animais , Linhagem Celular , Glucose/farmacologia , Immunoblotting , Indicadores e Reagentes , Células Secretoras de Insulina/efeitos dos fármacos , Masculino , Palmitatos/farmacologia , Fosforilação , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Troglitazona
16.
Zhonghua Nei Ke Za Zhi ; 43(10): 764-8, 2004 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-15631831

RESUMO

OBJECTIVE: A case of primary pigmented nodular adrenal disease (PPNAD) was first diagnosed in Ruijin Hospital, Shanghai, China and molecular genetic research was then carried on the proband and his family members. METHODS: History and laboratory tests were routinely taken. Liddle's test, adrenal CT and pituitary magnetic resonance imaging were also carried out. Complete family history was obtained and eight of the family members donated their blood for DNA extraction. Polymerase chain reaction was done on all the exons of PRKAR1A gene and the product of the reaction was sequenced with ABI 3700. The right adrenal of the patient was then resected, part of the tissue was preserved in liquid nitrogen for DNA/RNA extraction and the remaining sent to Department of Pathology. RESULTS: The patient presented an atypical appearance of Cushing's syndrome. His father had a typical history of cardiac myoma. Cortisone level could not be refrained in Liddle's test for the patient. Imaging examination presented a nodular adrenal and a full pituitary. A novel mutation of PRKAR1A-S147N was found in both the patient's and his father's gene. CONCLUSIONS: This is the first patient diagnosed as PPNAD based on his clinical manifestations, laboratory tests and imaging and pathological examinations. According to the history of his father and the results of molecular genetic analysis, the diagnosis of Carney complex can be established on this patient and his father. It is also the first time that this kind of point mutation was found in Chinese people.


Assuntos
Doenças do Córtex Suprarrenal/genética , Síndrome de Cushing/genética , Proteínas Quinases Dependentes de AMP Cíclico/genética , Transtornos da Pigmentação/genética , Doenças do Córtex Suprarrenal/diagnóstico , Adulto , Sequência de Bases , Síndrome de Cushing/diagnóstico , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico , Éxons/genética , Humanos , Masculino , Linhagem , Transtornos da Pigmentação/diagnóstico , Mutação Puntual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA