Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
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.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Mol Med Rep ; 9(3): 1044-8, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24401996

RESUMO

Vascular endothelial growth factor (VEGF) is one of the key modulators of angiogenesis. The highly polymorphic promoter and 5' untranslated region of VEGF have been associated with susceptibility to and aggressiveness of several types of cancer. To examine the functional role of VEGF polymorphisms at -634 and -1498 positions, VEGF mRNA and protein in breast cancer tissues were analyzed by quantitative polymerase chain reaction and immunohistochemistry. A dual-luciferase assay was performed to determine promoter activity. The VEGF-634CC genotype demonstrated the highest VEGF mRNA expression. High VEGF mRNA expression was correlated with a tumor size of >2 cm, the presence of lymphovascular invasion and the presence of axillary nodal metastasis. The promoter containing the -1,498T/-634C haplotype exhibited the highest basal promoter activity. These findings suggest that the interaction between -1,498T and -634C polymorphisms increases VEGF expression and is involved in breast cancer aggressiveness.


Assuntos
Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica/genética , Polimorfismo Genético , Fator A de Crescimento do Endotélio Vascular/genética , Adolescente , Adulto , Alelos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Feminino , Genes Reporter , Genótipo , Haplótipos , Humanos , Metástase Linfática , Células MCF-7 , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto Jovem
10.
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
11.
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
12.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA