Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
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.
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
3.
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
4.
Sci Rep ; 14(1): 17691, 2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39085384

RESUMO

Glucocorticoids (GCs) are known to stimulate pancreatic beta (ß)-cell apoptosis via several mechanisms, including oxidative stress. Our previous study suggested an increase in dexamethasone-induced pancreatic ß-cell apoptosis via a reduction of glutathione S-transferase P1 (GSTP1), which is an antioxidant enzyme. Imatinib, which is a tyrosine kinase inhibitor, also exerts antioxidant effect. This study aims to test our hypothesis that imatinib would prevent pancreatic ß-cell apoptosis induced by dexamethasone via increased GSTP1 expression and reduced oxidative stress. Our results revealed that dexamethasone significantly increased apoptosis in INS-1 cells when compared to the control, and that imatinib significantly decreased INS-1 cell apoptosis induced by dexamethasone. Moreover, dexamethasone significantly increased superoxide production in INS-1 cells when compared to the control; however, imatinib, when combined with dexamethasone, significantly reduced superoxide production in INS-1 cells. Dexamethasone significantly decreased GSTP1, p-ERK1/2, and BCL2 protein expression, but significantly increased p-JNK, p-p38, and BAX protein expression in INS-1 cells-all compared to control. Importantly, imatinib significantly ameliorated the effect of dexamethasone on the expression of GSTP1, p-ERK1/2, p-JNK, p-p38 MAPK, BAX, and BCL2. Furthermore-6-(7-nitro-2,1,3-benzoxadiazol-4-ylthio) hexanol (NBDHEX), which is a GSTP1 inhibitor, neutralized the protective effect of imatinib against pancreatic ß-cell apoptosis induced by dexamethasone. In conclusion, imatinib decreases pancreatic ß-cell apoptosis induced by dexamethasone via increased GSTP1 expression and reduced oxidative stress.


Assuntos
Apoptose , Dexametasona , Glutationa S-Transferase pi , Mesilato de Imatinib , Células Secretoras de Insulina , Estresse Oxidativo , Mesilato de Imatinib/farmacologia , Dexametasona/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Glutationa S-Transferase pi/metabolismo , Animais , Ratos , Linhagem Celular , Superóxidos/metabolismo
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
Asian Pac J Allergy Immunol ; 20(1): 13-21, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12125913

RESUMO

The estrogen receptor (ER)-alpha protein and ER mRNA were measured in 314 primary breast cancer patients by enzyme immunoassay (EIA) and reverse-transcription polymerase chain reaction (RT-PCR) assay, respectively. The positivity of ER protein was 53% while of ER mRNA was 37.6%. A significant positive association between ER phenotype and ER mRNA was observed (r = 0.40, p < 0.0001) with a positive-negative agreement between them of 71.8%. The percentage of ER-negative, progesterone receptor (PR)-positive breast tumors was 1.9% by EIA and 7% by RT-PCR assay. This may indicate a difference in ER variants in these studied patients. The ER protein and ER mRNA status were inversely related to tumor size and p53 positivity. Also, ER protein was frequently positive in patients with a higher number of lymph node invasions, well to moderate nuclear differentiated tumor cells and negative c-erbB-2 status. The difference of the ER or ER mRNA status regarding ages, menopausal status, tumor stages and histological types was not shown. In the present study, ER mRNA did not demonstrate a closer relationship to prognostic indicators of breast cancer than ER protein. Before including the ER mRNA assessment in routine investigations of breast cancer, its relationship to prognostic factors and survival outcome should be further assessed with a higher number of patients and a longer follow-up time.


Assuntos
Neoplasias da Mama/genética , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Receptores de Estrogênio/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/metabolismo , Receptor alfa de Estrogênio , Feminino , Humanos , Pessoa de Meia-Idade , Prognóstico , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sensibilidade e Especificidade , Estatística como Assunto , Tailândia , Saúde da Mulher , Microglobulina beta-2/metabolismo
14.
J Steroid Biochem Mol Biol ; 139: 25-32, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24120916

RESUMO

Estrogen can improve glucose homeostasis not only in diabetic rodents but also in humans. However, the molecular mechanism by which estrogen prevents pancreatic ß-cell death remains unclear. To investigate this issue, INS-1 cells, a rat insulinoma cell line, were cultured in medium with either 11.1mM or 40mM glucose in the presence or the absence of estrogen. Estrogen significantly reduced apoptotic ß-cell death by decreasing nitrogen-induced oxidative stress and the expression of the ER stress markers GRP 78, ATF6, P-PERK, PERK, uXBP1, sXBP1, and CHOP in INS-1 cells after prolonged culture in medium with 40mM glucose. In contrast, estrogen increased the expression of survival proteins, including sarco/endoplasmic reticulum Ca(2+) ATPase (SERCA-2), Bcl-2, and P-p38, in INS-1 cells after prolonged culture in medium with 40mM glucose. The cytoprotective effect of estrogen was attenuated by addition of the estrogen receptor (ERα and ERß) antagonist ICI 182,780 and the estrogen membrane receptor inhibitor G15. We showed that estrogen decreases not only oxidative stress but also ER stress to protect against 40mM glucose-induced pancreatic ß-cell death.


Assuntos
Apoptose , Estresse do Retículo Endoplasmático , Estradiol/fisiologia , Estrogênios/fisiologia , Glucose/fisiologia , Células Secretoras de Insulina/fisiologia , Animais , Sobrevivência Celular , Citoproteção , Glucose/farmacologia , Estresse Oxidativo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Receptores de Estrogênio/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Resposta a Proteínas não Dobradas , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Endocrinology ; 154(11): 4058-67, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23970784

RESUMO

Male hypogonadism associates with type 2 diabetes, and T can protect pancreatic ß-cells from glucotoxicity. However, the protective mechanism is still unclear. This study thus aims to examine the antiapoptotic mechanism of T in pancreatic ß cells cultured in high-glucose medium. T (0.0005-2 µg/mL) was added to INS-1 cells cultured in basal glucose or high-glucose media. Then cellular apoptosis, oxidative stress, and cell viability were measured. Endoplasmic reticulum (ER) stress markers and sensors and the antiapoptotic protein (B-cell lymphoma 2) were investigated by real-time PCR and Western blot analysis. ER stress markers were also measured in male mouse pancreatic islet cultured in similar conditions. T (0.05 and 0.5 µg/mL) did not have any effect on apoptosis and viability of INS-1 cells cultured in basal glucose medium, but it could reduce apoptosis and increase viability of INS-1 cells cultured in high-glucose medium. The protective effect of T is diminished by androgen receptor inhibitor. T (0.05 µg/mL) could significantly reduce nitrotyrosine levels, mRNA, and protein levels of the ER stress markers and sensor those that were induced when INS-1 cells were cultured in high-glucose medium. It could also significantly increase the survival proteins, sarco/endoplasmic reticulum Ca(2+) ATPase-2, and B-cell lymphoma 2 in INS-1 cells cultured in the same conditions. Similarly, it could reduce ER stress markers and increase sarco/endoplasmic reticulum Ca(2+) ATPase protein levels in male mouse pancreatic islets cultured in high-glucose medium. T can protect against male pancreatic ß-cell apoptosis from glucotoxicity via the reduction of both oxidative stress and ER stress.


Assuntos
Apoptose/efeitos dos fármacos , Glucose/toxicidade , Células Secretoras de Insulina/efeitos dos fármacos , Testosterona/farmacologia , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/fisiologia , Glucose/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos ICR , Receptores Androgênicos , Estresse Fisiológico/efeitos dos fármacos
16.
J Diabetes Complications ; 26(4): 343-7, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22521316

RESUMO

AIMS: PAX4 R192H polymorphism was reported to be associated with maturity onset diabetes of the young (MODY) and early onset-age of type 2 diabetes (T2D). This study aimed to evaluate transcriptional repression activity of PAX4 R192H polymorphism on its target promoters comparing with wild-type PAX4. METHODS: Wild-type PAX4 and PAX4 R192H proteins were expressed in vitro and the cell compartmentalization of each protein was examined after transfection of the plasmid constructs into ßTC3 cells followed by Western-blot analysis. The plasmid containing wild-type PAX4 or PAX4 R192H was co-transfected into ßTC3 and αTC-1.9 cells with insulin or glucagon promoter-reporter construct. Transcriptional repression activities were then determined by dual-luciferase reporter assay. RESULTS: Wild-type PAX4 and PAX4 R192H, which were found to be equally expressed in vitro and transfection systems, were present in the nuclear compartment. Transcriptional repressor activities of PAX4 R192H on human insulin and glucagon promoters were reduced when they were compared with those of wild-type PAX4. CONCLUSIONS: These results suggested that PAX4 R192H polymorphism generated a protein with defect in transcriptional repressor activities on its target genes, which may lead to ß-cell dysfunction associated with MODY and early onset-age of T2D as reported in our previous study.


Assuntos
Diabetes Mellitus Tipo 2/epidemiologia , Diabetes Mellitus Tipo 2/genética , Proteínas de Homeodomínio/genética , Fatores de Transcrição Box Pareados/genética , Polimorfismo Genético/genética , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Idade de Início , Animais , Linhagem Celular , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Glucagon/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Técnicas In Vitro , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Camundongos , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA