Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Cell Biochem ; 124(9): 1309-1323, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37555250

RESUMO

Prolonged administration of dexamethasone, a potent anti-inflammatory drug, can lead to steroid-induced diabetes. Imatinib, a medication commonly prescribed for chronic myeloid leukemia (CML), has been shown to improve diabetes in CML patients. Our recent study demonstrated that dexamethasone induces pancreatic ß-cell apoptosis by upregulating the expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its receptor, death receptor 5 (DR5). We hypothesized that imatinib may protect against dexamethasone-induced pancreatic ß-cell apoptosis by reducing the expression of TRAIL and DR5, thereby favorably modulating downstream effectors in apoptotic pathways. We test this hypothesis by assessing the effects of imatinib on dexamethasone-induced apoptosis in rat insulinoma cell line cells. As anticipated, dexamethasone treatment led to increased TRAIL and DR5 expression, as well as an elevation in superoxide production. Conversely, expression of the TRAIL decoy receptor (DcR1) was decreased. Moreover, key effectors in the extrinsic and intrinsic apoptosis pathways, such as B-cell lymphoma 2 (BCL-2) associated X (BAX), nuclear factor kappa B (NF-κb), P73, caspase 8, and caspase 9, were upregulated, while the antiapoptotic protein BCL-2 was downregulated. Interestingly and importantly, imatinib at a concentration of 10 µM reversed the effect of dexamethasone on TRAIL, DR5, DcR1, superoxide production, BAX, BCL-2, NF-κB, P73, caspase 3, caspase 8, and caspase 9. Similar effects of imatinib on dexamethasone-induced TRAIL and DR5 expression were also observed in isolated mouse islets. Taken together, our findings suggest that imatinib protects against dexamethasone-induced pancreatic ß-cell apoptosis by reducing TRAIL and DR5 expression and modulating downstream effectors in the extrinsic and intrinsic apoptosis pathways.


Assuntos
NF-kappa B , Superóxidos , Animais , Camundongos , Ratos , Apoptose , Proteína X Associada a bcl-2/metabolismo , Caspase 8/metabolismo , Caspase 9/metabolismo , Linhagem Celular Tumoral , Dexametasona/farmacologia , Mesilato de Imatinib/farmacologia , Ligantes , NF-kappa B/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Superóxidos/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
2.
Biochem Biophys Res Commun ; 529(3): 826-833, 2020 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-32684311

RESUMO

Maturity-onset diabetes of the young type 3 (MODY3) is caused by mutations in a gene encoding transcription factor hepatocyte nuclear factor 1-alpha (HNF1A). Although the roles of HNF1A in regulation of hepatic and pancreatic genes to maintain glucose homeostasis were investigated, the functions of HNF1A are not completely elucidated. To better understand the functions of HNF1A, we characterized mutations of HNF1A in Thai MODY3 patients and studied the functions of wild-type HNF1A and variant proteins. We demonstrate for the first time that HNF1A upregulates transactivation of an anti-apoptotic gene BCL2 Like 1 (BCL2L1) and that all the identified HNF1A variants including p.D80V, p.R203C, p.P475L, and p.G554fsX556, reduce this ability. The four HNF1A variants impair HNF1A function in promoting INS-1 cell transition from G1 to S phase of cell cycle, which thereby retard cell growth. This finding indicates the role of HNF1A in beta-cell viability by upregulation of anti-apoptotic gene expression and also reaffirms its role in beta-cell growth through cell cycle control.


Assuntos
Diabetes Mellitus Tipo 2/genética , Fator 1-alfa Nuclear de Hepatócito/genética , Células Secretoras de Insulina/citologia , Ativação Transcricional , Proteína bcl-X/genética , Adulto , Sequência de Aminoácidos , Animais , Linhagem Celular , Proliferação de Células , Feminino , Células HeLa , Fator 1-alfa Nuclear de Hepatócito/química , Humanos , Células Secretoras de Insulina/metabolismo , Masculino , Mutação , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Alinhamento de Sequência
3.
Biol Pharm Bull ; 40(9): 1506-1514, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28867733

RESUMO

Gastric motility disturbance is commonly found in long-standing hyperglycemia. Both delayed and rapid gastric emptying has been reported in diabetes. However, very few studies have followed the changes in gastric emptying during disease progression in diabetes because of technical limitations. 13C-Acetic acid breath test is a validated method which is non-invasive and can be used repeatedly or serially to evaluate gastric emptying changes in animal. We investigated the gastric emptying changes in different stages of diabetes using 13C-acetic acid breath test, as well as its related mechanisms involving interstitial cells of Cajal (ICCs), and stem cell factor (SCF) in streptozotocin-induced diabetic rats. The results showed that gastric emptying was accelerated at the early stage (12 weeks of diabetes) whereas intramuscular ICCs (ICC-IM) networks were not different from normal group. At long-term stage (28 weeks of diabetes), gastric emptying had returned to normal pattern with no delayed. ICC-IM networks were decreased in the diabetic group compared to 12th weeks, and were lower than in the normal group at the same time point. SCF levels were constantly high in the diabetic group than in the normal group. This result indicated that 13C-acetic acid breath test is useful to track the alteration in gastric emptying during disease progression. The change of gastric emptying was not found to be significantly associated with ICC-IM. Elevated SCF may help to preserve ICC-IM, especially in the early phase of diabetes.


Assuntos
Ácido Acético/análise , Testes Respiratórios/métodos , Diabetes Mellitus Experimental/fisiopatologia , Esvaziamento Gástrico , Animais , Glicemia/análise , Peso Corporal , Contagem de Células , Diabetes Mellitus Experimental/patologia , Progressão da Doença , Células Intersticiais de Cajal/patologia , Masculino , Monitorização Fisiológica , Proteínas Proto-Oncogênicas c-kit/biossíntese , Ratos , Ratos Sprague-Dawley , Fator de Células-Tronco/biossíntese
4.
J Hum Genet ; 61(11): 943-949, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27334367

RESUMO

We have previously identified PAX4 mutations causing MODY9 and a recent genome-wide association study reported a susceptibility locus of type 2 diabetes (T2D) near PAX4. In this study, we aim to investigate the association between PAX4 polymorphisms and T2D in Thai patients and examine functions of PAX4 variant proteins. PAX4 rs2233580 (R192H) and rs712701 (P321H) were genotyped in 746 patients with T2D and 562 healthy normal control subjects by PCR and restriction-fragment length polymorphism method. PAX4 variant proteins were investigated for repressor function on human insulin and glucagon promoters and for cell viability and apoptosis upon high glucose exposure. Genotype and allele frequencies of PAX4 rs2233580 were more frequent in patients with T2D than in control subjects (P=0.001 and 0.0006, respectively) with odds ratio of 1.66 (P=0.001; 95% confidence interval, 1.22-2.27). PAX4 rs712701 was not associated with T2D but it was in linkage disequilibrium with rs2233580. The 192H/321H (A/A) haplotype was more frequent in T2D patients than in controls (9.5% vs 6.6%; P=0.009). PAX4 R192H, but not PAX4 P321H, impaired repression activities on insulin and glucagon promoters and decreased transcript levels of genes required to maintain ß-cell function, proliferation and survival. Viability of ß-cell was reduced under glucotoxic stress condition for the cells overexpressing either PAX4 R192H or PAX4 P321H or both. Thus these PAX4 polymorphisms may increase T2D risk by defective transcription regulation of target genes and/or decreased ß-cell survival in high glucose condition.


Assuntos
Substituição de Aminoácidos , Códon , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Estudos de Associação Genética , Proteínas de Homeodomínio/genética , Fatores de Transcrição Box Pareados/genética , Polimorfismo Genético , Alelos , Animais , Glicemia , Estudos de Casos e Controles , Linhagem Celular , Sobrevivência Celular , Diabetes Mellitus Tipo 2/diagnóstico , Éxons , Feminino , Expressão Gênica , Frequência do Gene , Genótipo , Glucagon/genética , Glucagon/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Razão de Chances , Fatores de Transcrição Box Pareados/metabolismo , Regiões Promotoras Genéticas , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Estresse Fisiológico , Ativação Transcricional
5.
J Med Assoc Thai ; 98(6): 528-34, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26219155

RESUMO

BACKGROUND: Adiposity is an inflammatory condition contributing to the morbidity and mortality of several disorders, including type 2 diabetes mellitus (T2D) and cardiovascular disease. OBJECTIVE: To compare cardiometabolic risk factors between obese and non-obese Thai patients with T2DM MATERIAL AND METHOD: The cross-sectional study was done in 20 obese (BM >25 kg/m2) and 20 non-obese (BMI 23 kg/m2) T2DM Researchers measured fasting plasma glucose and lipids, serum levels of insulin, leptin, adiponectin, and soluble tumor necrosis factor-alpha receptors type 1 and 2 (sTNF-R] andsTNF-R2). Insulin sensitivity check index (QUICIKI) and insulin resistance index (HOMA-IR) were calculated. RESULTS: Thai obese adults with T2DMhad greater amounts ofsTNF-R2 and HOMA-IR, higher ratios of leptin/adiponectin, and more incidences of hypertension and hypertriglyceridemia in comparison to non-obese counterparts. Additionally, HOMA-IR values in non-obese T2DMwere greater than those reported among non-diabetic Thai adults. A reverse association between inflammatory markers (both sTNF-Rs) andHDLC was detected. Leptin/adiponectin ratios correlated directly with HOMA-IR, serum insulin, plasma triglycerides and BMI, whereas HOMA-IR did not relate to any studied plasma lipid. CONCLUSION: The present study demonstrated an increased cardiometabolic risk in obese T2DM adults than non-obese T2DM adults among the Thai population. The leptin/adiponectin ratio may be more relevant to predict the risk of cardiovascular events in T2DMpatients than HOMA-IR.


Assuntos
Doenças Cardiovasculares/epidemiologia , Diabetes Mellitus Tipo 2/fisiopatologia , Resistência à Insulina , Obesidade/fisiopatologia , Adiponectina/sangue , Adiposidade , Adulto , Idoso , Povo Asiático , Biomarcadores/metabolismo , Doenças Cardiovasculares/etiologia , Estudos Transversais , Diabetes Mellitus Tipo 2/sangue , Feminino , Humanos , Insulina/sangue , Leptina/sangue , Lipídeos/sangue , Masculino , Pessoa de Meia-Idade , Fatores de Risco , Triglicerídeos/sangue
6.
Biochem Biophys Res Commun ; 450(4): 1485-91, 2014 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-25019989

RESUMO

Dengue virus (DENV) is a positive-strand RNA virus of the Flavivirus family with 4 different serotypes. Clinical manifestations of DENV infection include dengue fever, dengue hemorrhagic fever, and dengue shock syndrome. Following DENV infection, apoptosis of hepatic cells is observed both in vitro and in vivo. However, the molecular mechanisms revealing how viral components affect cellular apoptosis remain unclear. In the present study, the role of death domain-associated protein 6 (Daxx) in DENV-mediated apoptosis was characterized by RNA interference and overexpression studies, and the anti-apoptotic function of Daxx during DENV infection was identified. Furthermore, the viral component, DENV capsid protein (DENV C), interacted with Daxx to disrupt interaction between Daxx and NF-κB. The liberated NF-κB activated the promoter of CD137, which is a member of the TNF family, and is previously shown to induce apoptosis during DENV infection. In summary, DENV C disrupts Daxx and NF-κB interaction to induce CD137-mediated apoptosis during DENV infection.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose/fisiologia , Vírus da Dengue/fisiologia , NF-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/fisiologia , Sequência de Bases , Proteínas Correpressoras , Primers do DNA , Células Hep G2 , Humanos , Chaperonas Moleculares , Reação em Cadeia da Polimerase , Ligação Proteica
7.
J Med Assoc Thai ; 97(1): 77-84, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24701733

RESUMO

BACKGROUND: The prevalence of metabolic syndrome (MS) accompanied with cardiometabolic complications has progressively increased in Thailand. The roles of insulin resistance, leptin, adiponectin, and free testosterone as prognostic indicators of MS among Thai population were evaluated MATERIAL AND METHOD: Men and women aged 34 to 89 years (n = 308) having 0-5 criteria of MS according to NCEP III with Asian-specific cut-points for waist circumference were enrolled in this cross-sectional study. Blood glucose, lipids, insulin, leptin, adiponectin, and free testosterone were measured RESULTS: Each component of MS, especially the enlarged waist, adversely affected insulin sensitivity. MS subjects were at higher risk for developing insulin resistance, decreasing of plasma adiponectin, and increasing of leptin and the leptin/ adiponectin ratio in comparison to non-MS individuals. The hormonal changes that have been shown to be associated with increased cardiometabolic risk were amplifiedas more MS criteria have been met. Odds ratios of increased leptin/adiponectin ratio among MS group were highest in comparison to others. Free testosterone levels declined with age and did not discriminate men with MS. CONCLUSION: The results indicate the benefit of hormonal assessment, particularly the leptin/adiponectin ratio in identifying MS individuals with high cardiometabolic disease risk.


Assuntos
Síndrome Metabólica/sangue , Adiponectina/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , Glicemia/análise , Estudos Transversais , Feminino , Humanos , Insulina/sangue , Leptina/sangue , Lipídeos/sangue , Masculino , Pessoa de Meia-Idade , Testosterona/sangue , Tailândia , População Urbana
8.
J Med Assoc Thai ; 95(11): 1472-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23252215

RESUMO

OBJECTIVE: Alterations of hormones involved in food intake can lead to obesity and related-diseases. The aim of the present study was to measure plasma levels of appetite-related hormones: insulin, leptin, adiponectin, acylated ghrelin, and cortisol in connection with eating behaviors among obese and non-obese women. MATERIAL AND METHOD: The present study was performed in 53 non-obese and 33 obese Thai women (BMI < 23 and > or = 25 kg/m2 respectively), aged 25 to 45 years. Saliva and fasting blood samples were collected for hormone measurements. Subjects 'eating behavior was evaluated using Thai version of the Three-factor eating questionnaire (TFEQ) and their stress status was assessed by the Thai stress test (TST). RESULTS: In comparison to non-obese individuals, obese women showed higher disinhibition eating, plasma glucose, insulin, HOMA insulin resistance index, leptin, and triglyceride levels but lesser plasma adiponectin and HDLC. Lower adiponectin was directly associated with higher disinhibition eating. Plasma leptin related positively to fat mass and insulin resistance but negatively to acylated ghrelin level. The trend towards increased acylated ghrelin after adjusted for age, obesity and eating behaviors was shown in stress women. CONCLUSION: Increased insulin resistance, high leptin, and reduced adiponectin accompanied with disinhibition eating have been detected in obese women.


Assuntos
Obesidade/sangue , Adiponectina/sangue , Adulto , Análise de Variância , Antropometria , Glicemia/análise , Comportamento Alimentar , Feminino , Grelina/sangue , Humanos , Hidrocortisona/sangue , Insulina/sangue , Resistência à Insulina , Leptina/sangue , Pessoa de Meia-Idade , Radioimunoensaio , Análise de Regressão , Estatísticas não Paramétricas , Inquéritos e Questionários , Tailândia , Triglicerídeos/sangue
9.
Sci Rep ; 12(1): 12950, 2022 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-35902739

RESUMO

Steroid-induced diabetes is a well-known metabolic side effect of long-term use of glucocorticoid (GC). Our group recently demonstrated dexamethasone-induced pancreatic ß-cell apoptosis via upregulation of TRAIL and TRAIL death receptor (DR5). Genistein protects against pancreatic ß-cell apoptosis induced by toxic agents. This study aimed to investigate the cytoprotective effect of genistein against dexamethasone-induced pancreatic ß-cell apoptosis in cultured rat insulinoma (INS-1) cell line and in isolated mouse islets. In the absence of genistein, dexamethasone-induced pancreatic ß-cell apoptosis was associated with upregulation of TRAIL, DR5, and superoxide production, but downregulation of TRAIL decoy receptor (DcR1). Dexamethasone also activated the expression of extrinsic and intrinsic apoptotic proteins, including Bax, NF-κB, caspase-8, and caspase-3, but suppressed the expression of the anti-apoptotic Bcl-2 protein. Combination treatment with dexamethasone and genistein protected against pancreatic ß-cell apoptosis, and reduced the effects of dexamethasone on the expressions of TRAIL, DR5, DcR1, superoxide production, Bax, Bcl-2, NF-κB, caspase-8, and caspase-3. Moreover, combination treatment with dexamethasone and genistein reduced the expressions of TRAIL and DR5 in isolated mouse islets. The results of this study demonstrate the cytoprotective effect of genistein against dexamethasone-induced pancreatic ß-cell apoptosis in both cell line and islets via reduced TRAIL and DR5 protein expression.


Assuntos
Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Ligante Indutor de Apoptose Relacionado a TNF , Animais , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Caspase 3/metabolismo , Caspase 8/metabolismo , Linhagem Celular Tumoral , Dexametasona/efeitos adversos , Genisteína/farmacologia , Camundongos , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Superóxidos/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Proteína X Associada a bcl-2/metabolismo
10.
J Mol Endocrinol ; 67(3): 95-106, 2021 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-34223821

RESUMO

Long-term medication with dexamethasone - a synthetic glucocorticoid (GC) drug - results in hyperglycemia, or steroid-induced diabetes. Although recent studies revealed that dexamethasone directly induces pancreatic ß-cell apoptosis, its molecular mechanisms remain unclear. In our initial analysis of mRNA transcripts, we discovered the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) pathway may be involved in dexamethasone-induced pancreatic ß-cell apoptosis. In the present study, a mechanism of dexamethasone-induced pancreatic ß-cell apoptosis through the TRAIL pathway was investigated in cultured cells and isolated mouse islets. INS-1 cells were cultured with and without dexamethasone in the presence or absence of a glucocorticoid receptor (GR) inhibitor, RU486. We found that dexamethasone induced pancreatic ß-cell apoptosis in association with the upregulation of TNSF10 (TRAIL) mRNA and protein expression. Moreover, dexamethasone upregulated the TRAIL death receptor (DR5) protein but suppressed the decoy receptor (DcR1) protein. Similar findings were observed in mouse isolated islets: dexamethasone increased TRAIL and DR5 compared to that of control mice. Furthermore, dexamethasone stimulated pro-apoptotic signaling including superoxide production, caspase-8, -9, and -3 activities, NF-κB, and Bax but repressed the anti-apoptotic protein, Bcl-2. All these effects were inhibited by the GR-inhibitor, RU486. Furthermore, knock-down DR5 decreased dexamethasone-induced caspase 3 activity. Caspase-8 and caspase-9 inhibitors protected pancreatic ß-cells from dexamethasone-induced apoptosis. Taken together, dexamethasone induced pancreatic ß-cell apoptosis by binding to the GR and inducing DR5 and TRAIL pathway.


Assuntos
Apoptose/efeitos dos fármacos , Dexametasona/farmacologia , Regulação da Expressão Gênica , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Animais , Biomarcadores , Caspase 3/metabolismo , Caspase 8/metabolismo , Inibidores de Caspase/farmacologia , Técnicas de Silenciamento de Genes , Masculino , Camundongos , NF-kappa B/metabolismo , Estresse Oxidativo , Proteólise , Receptores de Glucocorticoides/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
11.
Biochem Biophys Res Commun ; 383(1): 68-72, 2009 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-19336222

RESUMO

A novel frameshift mutation attributable to 14-nucleotide insertion in hepatocyte nuclear factor-1alpha (HNF-1alpha) encoding a truncated HNF-1alpha (G554fsX556) with 76-amino acid deletion at its carboxyl terminus was identified in a Thai family with maturity-onset diabetes of the young (MODY). The wild-type and mutant HNF-1alpha proteins were expressed by in vitro transcription and translation (TNT) assay and by transfection in HeLa cells. The wild-type and mutant HNF-1alpha could similarly bind to human glucose-transporter 2 (GLUT2) promoter examined by electrophoretic mobility shift assay (EMSA). However, the transactivation activities of mutant HNF-1alpha on human GLUT2 and rat L-type pyruvate kinase (L-PK) promoters in HeLa cells determined by luciferase reporter assay were reduced to approximately 55-60% of the wild-type protein. These results suggested that the functional defect of novel truncated HNF-1alpha (G554fsX556) on the transactivation of its target-gene promoters would account for the beta-cell dysfunction associated with the pathogenesis of MODY.


Assuntos
Diabetes Mellitus Tipo 2/genética , Transportador de Glucose Tipo 2/genética , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Deleção de Sequência , Ativação Transcricional/genética , Animais , Ensaio de Desvio de Mobilidade Eletroforética , Células HeLa , Fator 1-alfa Nuclear de Hepatócito/genética , Humanos , Regiões Promotoras Genéticas , Piruvato Quinase/genética , Ratos
12.
Sci Rep ; 8(1): 12256, 2018 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-30115961

RESUMO

Hyperglycemia stimulates several pathways to induce pancreatic ß-cell apoptosis. In our previous study by mRNA analysis, we demonstrated that B-cell translocation gene 2 (BTG2) expression was up-regulated in INS-1 cells cultured under high glucose conditions, but this effect was reversed by estrogen. In the present study, we demonstrated that BTG2 mRNA and protein expressions in both INS-1 cells and mouse pancreatic islets increased under high glucose conditions compared to those cultured under basal glucose conditions, while in the presence of estrogen, the BTG2 mRNA and protein expressions decreased. SiRNA-BTG2 significantly reduced cell apoptosis, cleaved-caspase 3, and Bax, compared to the siRNA-control in INS-1 cultured under high glucose conditions. We further demonstrated that BTG2 promoter activity was activated under high glucose conditions whereas estrogen significantly reduced it. The effects of estrogen on BTG2 expression were inhibited by estrogen receptor inhibitors. Also, under high glucose conditions, p53 and Bax mRNA and protein expressions increased, but they decreased in the presence of estrogen. Again, the effect of estrogen on p53 and Bax expression was inhibited by estrogen receptor inhibitors. Taken together, this study demonstrates that estrogen reduces pancreatic ß-cell apoptosis under high glucose conditions via suppression of BTG2, p53, and Bax expressions.


Assuntos
Apoptose/efeitos dos fármacos , Estradiol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Proteínas Imediatamente Precoces/genética , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Proteínas Supressoras de Tumor/genética , Animais , Relação Dose-Resposta a Droga , Células Secretoras de Insulina/metabolismo , Camundongos , Regiões Promotoras Genéticas/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
13.
J Clin Endocrinol Metab ; 92(7): 2821-6, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17426099

RESUMO

CONTEXT: Six maturity onset diabetes of the young (MODY) genes have been discovered to date but account for a small proportion of MODY among Asians, suggesting the existence of other MODY genes in this racial group. OBJECTIVE: The aim of this study was to investigate whether or not genetic variants in PAX4, a crucial transcription factor in beta-cell development, contribute to MODY in Thais. DESIGN AND METHODS: We screened PAX4 coding sequences in 46 MODY probands without mutation in known MODY genes and in 74 nondiabetic controls using PCR-single-stranded conformational polymorphism analysis followed by direct sequencing. Genotyping of variants identified was done by PCR-restriction fragment length polymorphism analysis. RESULTS: Eight sequence differences were identified. Two novel variations (R164W and IVS7-1G>A) were found in two different probands. Neither was found in the 74 nondiabetic controls and additional 270 healthy subjects of Thai origin. R164W segregated with diabetes in the family of the proband and in vitro studies showed that it impairs the repressor activity of PAX4 on the insulin and glucagon promoters. The remaining six variants were previously described and observed in both groups. One of them, R192H, was three times more frequent in MODY probands than in 342 nondiabetic controls (minor allele frequency = 0.196 vs. 0.064; P < 0.00001). The same variant was associated with a younger age at diagnosis among 254 Thai subjects with adult-onset type 2 diabetes (44.6 +/- 15 vs. 49.7 +/- 11 yr; P = 0.048). CONCLUSIONS: We have identified two possible pathogenic mutations of PAX4, R164W, and IVS7-1G>A. For one of these, we have shown evidence of segregation with diabetes and a functional impact on PAX4 activity. Single-nucleotide polymorphism R192H might influence the age at onset of diabetes.


Assuntos
Povo Asiático/genética , Diabetes Mellitus Tipo 2/etnologia , Diabetes Mellitus Tipo 2/genética , Proteínas de Homeodomínio/genética , Fatores de Transcrição Box Pareados/genética , Adulto , Idade de Início , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Mutação Puntual , Polimorfismo Conformacional de Fita Simples , Tailândia
14.
J Med Assoc Thai ; 90(5): 956-61, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17596052

RESUMO

BACKGROUND: It is known that males are more susceptible to develop type 2 diabetes than females. Estrogen has a protective effect on pancreatic islet against toxic agent such as amyloid. The role of estrogen in protection pancreatic islet against high glucose is still unknown. OBJECTIVE: Administration of estrogen in an ovariectomised animal shows a protective effect against type 2 diabetes. The present study aimed to determine the direct effect of estrogen on the islet function after prolonged culture in high glucose. MATERIAL AND METHOD: Estrogen (10-1 M in ethanol) was co-cultured with mouse pancreatic islets in normal glucose medium (11.1 mM) for 3 hours or with normal and high glucose medium (40 mM) for 10 days. RESULTS: Estrogen increased glucose-induced insulin secretion in islet culture in normal glucose medium for both 3-hour and 10-day culture. Prolonged exposure of pancreatic islet to high glucose generated impaired glucose-induced insulin secretion, which was partially abrogated by the presence of 10(-5) M estrogen. CONCLUSION: These results indicated a direct effect of estrogen on improving insulin secretion from mouse pancreatic islets that has been impaired by prolonged exposure to high glucose.


Assuntos
Glicemia , Estrogênios/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Animais , Diabetes Mellitus Tipo 2/prevenção & controle , Humanos , Técnicas In Vitro , Secreção de Insulina , Masculino , Camundongos , Fatores de Tempo
15.
J Med Assoc Thai ; 90(10): 2150-7, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18041436

RESUMO

OBJECTIVE: To evaluate the benefits of glucomannan supplement on glycemic and lipid controls in type 2 diabetic patients. MATERIAL AND METHOD: A single-blind, placebo-controlled, crossover trial with two treatments separated by a 2-week washout period was performed in 10 men and 10 women with type 2 diabetes mellitus. Two separated protocols of experiments were sequentially followed. Initially, purified glucomannan (1 g) or placebo was ingested 30 min before 75-g glucose load to evaluate their effects on glucose absorption and insulin secretion in oral glucose tolerance test (OGTT). Later, the glycemic and lipid changes after 4-week intervention with 3 g/day glucomannan comparing to the placebo were determined. The standard OGTT was performed before and after ending of each intervention. RESULTS: Glucomannan taken before performing the OGTT can lower the rise of blood glucose and insulin from 1 to 2 hour in comparison with the placebo, though a statistically significance of insulin was not achieved. Long-term glucomannan supplement significantly reduced the 120-min glucose area under the curve of OGTT. Glucomannan also decreased the rise of low-density lipoprotein cholesterol (LDL-C). Reductions of HOMA-insulin resistance index and body mass index were detected in glucomannan-treated group though the former was shown only in females. No within- and between-group differences of insulin, fructosamine, and other lipids were observed in glucomannan- nor placebo- treated groups. CONCLUSION: In type 2 diabetes, pre-prandial glucomannan ingestion attenuated a rise of blood glucose without significantly affecting insulin levels. Long-term supplement of glucomannan to the regular diabetic regimen lessened post challenge glucose AUC and impeded the rise of LDL-C. Supplement of glucomannan may be beneficial to the glycemic and lipid controls in type 2 diabetes mellitus.


Assuntos
Glicemia/efeitos dos fármacos , Catárticos/farmacologia , Diabetes Mellitus Tipo 2/fisiopatologia , Índice Glicêmico , Insulina , Lipídeos/sangue , Mananas/farmacologia , Adulto , Idoso , Área Sob a Curva , Índice de Massa Corporal , LDL-Colesterol/efeitos dos fármacos , Feminino , Teste de Tolerância a Glucose , Indicadores Básicos de Saúde , Humanos , Resistência à Insulina , Masculino , Pessoa de Meia-Idade , Tailândia
16.
Sci Rep ; 7(1): 16639, 2017 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-29192236

RESUMO

Chronic exposure of pancreatic ß-cells to high glucose levels results in ß-cell dysfunction and death. These effects can be protected by estrogen. The local pancreatic renin-angiotensin system (RAS) has been shown as a novel pathological pathway of high-glucose-induced cell death. The effect of estrogen on pancreatic RAS is still unknown. This study examines whether estrogen protects against pancreatic ß-cell death caused by glucotoxicity via a decrease in the pancreatic ß-cell RAS pathway. When INS-1 cells were cultured in a high glucose medium, cell death was significantly higher than when the cells were cultured in a basal glucose medium; similarly, there were also higher levels of AGTR1 and p47 ph ° x mRNA, and protein expression. Moreover, the addition of 10-8 M 17ß-estradiol to INS-1 cells cultured in a high glucose medium markedly reduced cell death, AGTR1 and p47 ph ° x mRNA levels, and protein expression. Similar results were demonstrated in the pancreatic islets. The presence of 10-8 M 17ß-estradiol, losartan, or a combination of both, in a high glucose medium had similar levels of reduction of p47 ph ° x mRNA and protein expression, compared with those cultured in high glucose. Taken together, estrogen protected pancreatic ß-cells from high-glucose-induced cell death by reducing the AGTR1 pathway.


Assuntos
Estrogênios/metabolismo , Expressão Gênica , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Receptor Tipo 1 de Angiotensina/genética , Animais , Apoptose , Caspase 3/metabolismo , Morte Celular , Sobrevivência Celular/genética , Células Cultivadas , Estrogênios/farmacologia , Glucose/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Losartan/farmacologia , Camundongos , Estresse Oxidativo , RNA Mensageiro/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Transdução de Sinais
17.
Acta Diabetol ; 53(2): 205-16, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25951767

RESUMO

AIMS: Paired box 4 (PAX4) mutations cause maturity-onset diabetes of the young, type 9 (MODY9). The molecular defect and alteration of PAX4 function associated with the mutation PAX4 IVS7-1G>A in a family with MODY9 and severe diabetic complications were studied. METHODS: We investigated the functional consequences of PAX4 IVS7-1G>A on mRNA splicing using minigene assays. Wild-type and mutant PAX4 were expressed in mouse pancreatic ß- and α-cell lines, and protein levels and translocation of PAX4 into the nucleus were determined. We also examined transcriptional repression of PAX4 target-gene promoters and ß-cell viability under diabetic-like (high-glucose) conditions. RESULTS: PAX4 IVS7-1G>A disrupts an acceptor splice site, causing an adjacent cryptic splice site within exon 8 to be used, resulting in a three-nucleotide deletion and glutamine deletion at position 250 (p.Q250del). Wild-type and PAX4 Q250del proteins were expressed at similar levels and could translocate normally into the nucleus in ßTC3 and αTC1.9 cells. However, the repressor functions of PAX4 Q250del on human insulin and glucagon promoters in INS-1 832/13 and αTC1.9 cells were significantly decreased, compared with that of wild-type PAX4. Moreover, the rate of apoptosis was increased in INS-1 cells over-expressing PAX4 Q250del when cultured in high-glucose conditions. CONCLUSIONS: PAX4 IVS7-1G>A caused aberrant mRNA splicing and PAX4 Q250 deletion. The mutation impaired PAX4 repressor functions on target-gene promoters and increased susceptibility to apoptosis upon high glucose exposure. Thus, PAX4 IVS7-1G>A contributes to the pathogenesis of diabetes in this MODY9 family through ß-cell dysfunction.


Assuntos
Processamento Alternativo/genética , Diabetes Mellitus Tipo 2/genética , Proteínas de Homeodomínio/genética , Fatores de Transcrição Box Pareados/genética , Adulto , Animais , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Deleção de Genes , Glucose/farmacologia , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Camundongos , Mutação/genética , Proteínas Repressoras/efeitos dos fármacos , Translocação Genética
18.
J Mol Endocrinol ; 35(1): 39-48, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16087720

RESUMO

Type 2 diabetes is characterized by islet dysfunction resulting in hyperglycemia, which can then lead to further deterioration in islet function. A possible mechanism for hyperglycemia-induced islet dysfunction is the accumulation of advanced glycation end products (AGE). The DBA/2 mouse develops pancreatic islet dysfunction when exposed to a high glucose environment and/or obesity-induced insulin resistance. To determine the biochemical cause of dysfunction, DBA/2 and C57BL/6 control islets were incubated in 11.1 mM or 40 mM glucose in the absence or presence of the AGE inhibitor aminoguanidine (AG) for 10 days. Basal (2.8 mM glucose) insulin release was increased in both DBA/2 and C57BL/6 islets incubated with 40 mM vs 11.1 mM glucose for 10 days. Chronic exposure to hyperglycemia decreased glucose (20 mM)-stimulated insulin secretion in DBA/2 but not in C57BL/6 islets. AG significantly increased fold-induced insulin release in high glucose cultured DBA/2 mouse islets, but did not affect C57BL/6 islet function. DBA/2 islet glucokinase was significantly reduced following 40 mM glucose culture, compared with 11.1 mM glucose cultured DBA/2 islets and 40 mM glucose cultured C57BL/6 islets. Incubation of islets with AG resulted in a normalization of DBA/2 islet glucokinase levels. In conclusion, chronic high glucose-induced increases in AGE can result in islet dysfunction and this is associated with reduced glucokinase levels in a mouse model with susceptibility to islet failure.


Assuntos
Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/fisiopatologia , Glucoquinase/metabolismo , Glucose/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/fisiopatologia , Animais , Sequência de Bases , Sobrevivência Celular , DNA/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Glucose/administração & dosagem , Transportador de Glucose Tipo 2 , Produtos Finais de Glicação Avançada/metabolismo , Hexoquinase/metabolismo , Técnicas In Vitro , Insulina/genética , Secreção de Insulina , Ilhotas Pancreáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Proteínas de Transporte de Monossacarídeos/metabolismo , Proinsulina/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
19.
J Endocrinol ; 224(3): 215-24, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25512346

RESUMO

Hypogonadism in men is associated with an increased incidence of type 2 diabetes. Supplementation with testosterone has been shown to protect pancreatic ß-cell against apoptosis due to toxic substances including streptozotocin and high glucose. One of the pathological mechanisms of glucose-induced pancreatic ß-cell apoptosis is the induction of the local rennin-angiotensin-aldosterone system (RAAS). The role of testosterone in regulation of the pancreatic RAAS is still unknown. This study aims to investigate the protective action of testosterone against glucotoxicity-induced pancreatic ß-cell apoptosis via alteration of the pancreatic RAAS pathway. Rat insulinoma cell line (INS-1) cells or isolated male mouse islets were cultured in basal and high-glucose media in the presence or absence of testosterone, losartan, and angiotensin II (Ang II), then cell apoptosis, cleaved caspase 3 expression, oxidative stress, and expression of angiotensin II type 1 receptor (AGTR1) and p47(phox) mRNA and protein were measured. Testosterone and losartan showed similar effects in reducing pancreatic ß-cell apoptosis. Testosterone significantly reduced expression of AGTR1 protein in INS-1 cells cultured in high-glucose medium or high-glucose medium with Ang II. Testosterone decreased the expression of AGTR1 and p47(phox) mRNA and protein in comparison with levels in cells cultured in high-glucose medium alone. Furthermore, testosterone attenuated superoxide production when co-cultured with high-glucose medium. In contrast, when cultured in basal glucose, supplementation of testosterone did not have any effect on cell apoptosis, oxidative stress, and expression of AGT1R and p47(phox). In addition, high-glucose medium did not increase cleaved caspase 3 in AGTR1 knockdown experiments. Thus, our results indicated that testosterone prevents pancreatic ß-cell apoptosis due to glucotoxicity through reduction of the expression of ATGR1 and its signaling pathway.


Assuntos
Apoptose/efeitos dos fármacos , Glucose/toxicidade , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/genética , Testosterona/farmacologia , Animais , Apoptose/genética , Células Cultivadas , Citoproteção/efeitos dos fármacos , Citoproteção/genética , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Ratos , Receptor Tipo 1 de Angiotensina/metabolismo
20.
Virus Res ; 197: 92-100, 2015 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-25523420

RESUMO

Dengue virus (DENV) infection associates with renal disorders. Patients with dengue hemorrhagic fever and acute kidney injury have a high mortality rate. Increased levels of cytokines may contribute to the pathogenesis of DENV-induced kidney injury. Currently, molecular mechanisms how DENV induces kidney cell injury has not been thoroughly investigated. Excessive cytokine production may be involved in this process. Using human cytokine RT(2) Profiler PCR array, 14 genes including IP-10, RANTES, IL-8, CXCL-9 and MIP-1ß were up-regulated more than 2 folds in DENV-infected HEK 293 cells compared to that of mock-infected HEK 293 cells. In the present study, RANTES was suppressed by the NF-κB inhibitor, compound A (CpdA), in DENV-infected HEK 293 cells implying the role of NF-κB in RANTES expression. Chromatin immunoprecipitation (ChIP) assay showed that NF-κB binds more efficiently to its binding sites on the RANTES promoter in NS5-transfected HEK 293 cells than in HEK 293 cells expressing the vector lacking NS5 gene. To further examine whether the NS5-activated RANTES promoter is mediated through NF-κB, the two NF-κB binding sites on the RANTES promoter were mutated and this promoter was coupled to the luciferase cDNA. The result showed that when both binding sites of NF-κB in the RANTES promoter were mutated, the ability of NS5 to induce the luciferase activity was significantly decreased. Therefore, DENV NS5 activates RANTES production by increasing NF-κB binding to its binding sites on the RANTES promoter.


Assuntos
Quimiocina CCL5/biossíntese , Vírus da Dengue/imunologia , NF-kappa B/metabolismo , Proteínas não Estruturais Virais/imunologia , Linhagem Celular , Imunoprecipitação da Cromatina , Perfilação da Expressão Gênica , Humanos , Análise em Microsséries
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA