Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Endocrine ; 83(2): 378-391, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37752366

RESUMO

PURPOSE: To evaluate the dopaminergic signaling in human adipose tissue in the context of obesity and type 2 diabetes (T2D) and potential direct implications in adipose tissue metabolism. METHODS: mRNA and protein expression of dopamine receptors D1 and D2 (DRD1 and DRD2) were determined in subcutaneous adipose tissue from subjects without or with T2D and with different body weight, and correlated with markers of obesity, hyperglycemia, and insulin resistance. Glucose uptake and lipolysis were measured in adipocytes ex vivo following short-term exposure to dopamine, DRD1 receptor agonist (SKF81297), or DRD2 receptor agonist (bromocriptine). RESULTS: DRD1 and DRD2 gene expression in subcutaneous adipose tissue correlated positively with clinical markers of insulin resistance (e.g. HOMA-IR, insulin, and triglycerides) and central obesity in subjects without T2D. Protein expression of DRD2 in subcutaneous adipose tissue, but not DRD1, is higher in subjects with impaired fasting glucose and T2D and correlated positively with hyperglycemia, HbA1c, and glucose AUC, independent of obesity status. DRD1 and DRD2 proteins were mainly expressed in adipocytes, compared to stromal vascular cells. Dopamine and dopaminergic agonists did not affect adipocyte glucose uptake ex vivo, but DRD1 and DRD2 agonist treatment inhibited isoproterenol-stimulated lipolysis. CONCLUSION: The results suggest that protein expression of DRD2 in subcutaneous adipose tissue is up-regulated with hyperglycemia and T2D. Whether DRD2 protein levels contribute to T2D development or occur as a secondary compensatory mechanism needs further investigation. Additionally, dopamine receptor agonists inhibit adipocyte beta-adrenergic stimulation of lipolysis, which might contribute to the beneficial effects in lipid metabolism as observed in patients taking bromocriptine.


Assuntos
Diabetes Mellitus Tipo 2 , Hiperglicemia , Resistência à Insulina , Estado Pré-Diabético , Humanos , Diabetes Mellitus Tipo 2/metabolismo , Estado Pré-Diabético/metabolismo , Bromocriptina , Dopamina/metabolismo , Tecido Adiposo/metabolismo , Gordura Subcutânea/metabolismo , Glucose/metabolismo , Hiperglicemia/metabolismo , Obesidade/metabolismo , Agonistas de Dopamina , Receptores de Dopamina D2/genética
2.
Ups J Med Sci ; 124(3): 158-167, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31407948

RESUMO

Background: Tissue factor (TF) combined with its ligand FVII initiates blood coagulation and intracellular signaling. Obese and type 2 diabetic subjects have increased TF expression in their adipose tissue and an increased risk for thrombotic complications. Here we address the role of TF/FVII on adipocyte functions. Materials and methods: Subcutaneous fat was obtained by means of needle aspiration from healthy volunteers, and adipocytes were isolated after collagenase digestion. 3T3-L1 fibroblasts kept in culture were differentiated into adipocytes by addition of IBMX, dexamethasone, rosiglitazone, and insulin to the media. Proteins and mRNA were analyzed by western blot and RT-PCR. Coagulation activity was determined by a colorimetric FX-assay. Lipolysis was measured as free glycerol using a colorimetric method. Glucose uptake was evaluated by scintillation counting of D-[U-14C] glucose. Results: In isolated human primary adipocytes we found expression of TF and FVII. TF expression was confirmed in 3T3-L1 adipocytes, and both cell types were found to be procoagulant in a TF/FVIIa-dependent manner. FXa was generated without FVIIa added to the coagulation assay, and active site-inhibited FVIIa blocked FXa formation, supporting our finding of FVII production by human primary adipocytes. There was no evidence for a role of TF in either lipolysis or glucose uptake in our experimental settings. Conclusion: Human primary adipocytes express active TF and FVII, and the TF/FVIIa complex formed on the adipocyte surface can activate substrate FX. Whether the TF/FVIIa complex conveys signaling pathways leading to biological functions and has any biological activity in adipocytes beyond coagulation remains to be elucidated.


Assuntos
Adipócitos/metabolismo , Regulação da Expressão Gênica , Serina Endopeptidases/genética , Tromboplastina/genética , Adipócitos/efeitos dos fármacos , Coagulação Sanguínea/fisiologia , Western Blotting , Células Cultivadas , Meios de Cultivo Condicionados , Dexametasona/farmacologia , Regulação para Baixo , Voluntários Saudáveis , Humanos , Insulina/farmacologia , Lipólise/genética , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Rosiglitazona/farmacologia , Transdução de Sinais , Gordura Subcutânea/citologia , Transfecção
3.
Diabetologia ; 58(11): 2563-72, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26271343

RESUMO

AIMS/HYPOTHESIS: Patients diagnosed with type 1 or type 2 diabetes have elevated levels of coagulation factor VIIa (FVIIa) and its receptor tissue factor (TF) in their bloodstream. This may affect the fate of the beta cells. We aimed to study the effects of TF/FVIIa signalling on cytokine-induced beta cell death and islet function in vitro. METHODS: Human pancreatic islets and MIN-6 beta cells were used to study TF mRNA and protein expression using real-time PCR, immunoblotting and flow cytometry. The effects of TF/FVIIa on cytokine-induced beta cell death were studied in MIN-6 cells and human pancreatic islets using cell-death ELISA and propidium iodide and cleaved caspase-3 staining. Effects of TF/FVIIa on the phosphorylation of p38, extracellular signal-regulated kinase and c-Jun N-terminal kinase (JNK) were investigated by immunoblotting. Glucose-stimulated insulin secretion (GSIS) from human islets was measured with an insulin ELISA. RESULTS: A combination of the cytokines IL-1ß, TNF-α and IFN-γ induced TF expression in human pancreatic islets and in beta cells. TF/FVIIa did not affect basal beta cell death but, independently of downstream coagulation activity, augmented beta cell death in response to cytokines. The effect of TF/FVIIa on cytokine-induced beta cell death was found to be dependent on the stress kinase JNK, since FVIIa addition potentiated cytokine-induced JNK activation and JNK inhibition abolished the effect of TF/FVIIa on cytokine-induced beta cell death. Moreover, TF/FVIIa signalling resulted in inhibition of GSIS from human pancreatic islets. CONCLUSIONS/INTERPRETATION: These results indicate that TF/FVIIa signalling has a negative effect on beta cell function and promotes beta cell death in response to cytokines.


Assuntos
Morte Celular/fisiologia , Citocinas/farmacologia , Fator VIIa/metabolismo , Glucose/farmacologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Tromboplastina/metabolismo , Caspase 3/metabolismo , Morte Celular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator VIIa/genética , Humanos , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , Ilhotas Pancreáticas/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Tromboplastina/genética , Fator de Necrose Tumoral alfa/farmacologia
4.
J Biol Chem ; 289(47): 32379-91, 2014 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-25281742

RESUMO

Tissue factor (TF) binds the serine protease factor VIIa (FVIIa) to form a proteolytically active complex that can trigger coagulation or activate cell signaling. Here we addressed the involvement of tyrosine kinase receptors (RTKs) in TF/FVIIa signaling by antibody array analysis and subsequently found that EphB2 and EphA2 of the Eph RTK family were cleaved in their ectodomains by TF/FVIIa. We used N-terminal Edman sequencing and LC-MS/MS analysis to characterize the cleaved Eph isoforms and identified a key arginine residue at the cleavage site, in agreement with the tryptic serine protease activity of FVIIa. Protease-activated receptor 2 (PAR2) signaling and downstream coagulation activity was non-essential in this context, in further support of a direct cleavage by TF/FVIIa. EphB2 was cleaved by FVIIa concentrations in the subnanomolar range in a number of TF expressing cell types, indicating that the active cellular pool of TF was involved. FVIIa caused potentiation of cell repulsion by the EphB2 ligand ephrin-B1, demonstrating a novel proteolytical event to control Eph-mediated cell segregation. These results define Eph RTKs as novel proteolytical targets of TF/FVIIa and provide new insights into how TF/FVIIa regulates cellular functions independently of PAR2.


Assuntos
Fator VIIa/metabolismo , Receptor EphA2/metabolismo , Receptor EphB2/metabolismo , Tromboplastina/metabolismo , Sequência de Aminoácidos , Sítios de Ligação/genética , Western Blotting , Linhagem Celular Tumoral , Movimento Celular , Células Cultivadas , Dissulfetos/química , Dissulfetos/metabolismo , Fator VII , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Oxirredução , Ligação Proteica , Estrutura Terciária de Proteína , Proteólise , Receptor EphA2/química , Receptor EphA2/genética , Receptor EphB2/química , Receptor EphB2/genética , Análise de Sequência de Proteína , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Especificidade por Substrato , Espectrometria de Massas em Tandem
5.
Thromb Haemost ; 111(4): 748-60, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24336871

RESUMO

The insulin-like growth factor 1 receptor (IGF-1R) is known to promote survival and has also been implicated in the pathogenesis of several disease states, including cardiovascular disorders and cancer. Recently, we showed that binding of coagulation factor VIIa (FVIIa) to its receptor tissue factor (TF) protects cancer cells from TNF-related apoptosis inducing ligand (TRAIL)-induced apoptosis. Here we present evidence that this biological function of TF/FVIIa is dependent on the IGF-1R. IGF-1R inhibitors AG1024 and PPP as well as siRNA-mediated downregulation of IGF-1R, abolished the TF/FVIIa-mediated protection against TRAIL-induced apoptosis. Moreover, FVIIa rapidly induced a time- and concentration-dependent tyrosine phosphorylation of the IGF-1R in MDA-MB-231 breast cancer cells and in primary human monocytes, an event that was accompanied by IGF-1R chromatin binding and gene transcription. We hereby present novel evidence of a cross-talk between the coagulation and IGF-1R signalling systems, and propose that the IGF-1R is a key player in mediating TF/FVIIa-induced cell survival.


Assuntos
Células Epiteliais/fisiologia , Fator VIIa/metabolismo , Monócitos/fisiologia , Receptor IGF Tipo 1/metabolismo , Tromboplastina/metabolismo , Apoptose/efeitos dos fármacos , Coagulação Sanguínea/efeitos dos fármacos , Coagulação Sanguínea/genética , Linhagem Celular Tumoral , Sobrevivência Celular , Citoproteção/efeitos dos fármacos , Citoproteção/genética , Células Epiteliais/efeitos dos fármacos , Humanos , Monócitos/efeitos dos fármacos , Fenciclidina/análogos & derivados , Fenciclidina/farmacologia , Ligação Proteica , RNA Interferente Pequeno/genética , Receptor Cross-Talk , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor IGF Tipo 1/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ativação Transcricional/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Tirfostinas/farmacologia
6.
PLoS One ; 6(9): e24831, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21935477

RESUMO

PURPOSE: Imatinib Mesylate (Gleevec) is a drug that potently counteracts diabetes both in humans and in animal models for human diabetes. We have previously reported that this compound in human pancreatic islets stimulates NF-κB signaling and islet cell survival. The aim of this study was to investigate control of NF-κB post-translational modifications exerted by Imatinib and whether any such effects are associated with altered islet gene expression and chemokine production in vitro. PROCEDURES: Human islets were either left untreated or treated with Imatinib for different timepoints. IκB-α and NF-κB p65 phosphorylation and methylation were assessed by immunoblot analysis. Islet gene expression was assessed using a commercial Pathway Finder microarray kit and RT-PCR. Islet chemokine production was determined by flow cytometric bead array analysis. FINDINGS: Human islet IκB-α and Ser276-p65 phosphorylation were increased by a 20 minute Imatinib exposure. Methylation of p65 at position Lys221 was increased after 60 min of Imatinib exposure and persisted for 3 hours. Microarray analysis of islets exposed to Imatinib for 4 hours revealed increased expression of the inflammatory genes IL-4R, TCF5, DR5, I-TRAF, I-CAM, HSP27 and IL-8. The islet release of IL-8 was augmented in islets cultured over night in the presence of Imatinib. Following 30 hours of Imatinib exposure, the cytokine-induced IκB-α and STAT1 phosphorylation was abolished and diminished, respectively. The cytokine-induced release of the chemokines MIG and IP10 was lower in islets exposed to Imatinib for 30 hours. CONCLUSION: Imatinib by itself promotes a modest activation of NF-κB. However, a prolonged exposure of human islets to Imatinib is associated with a dampened response to cytokines. It is possible that Imatinib induces NF-κB preconditioning of islet cells leading to lowered cytokine sensitivity and a mitigated islet inflammation.


Assuntos
Piperazinas/farmacologia , Pirimidinas/farmacologia , Fator de Transcrição RelA/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Benzamidas , Quimiocinas/metabolismo , Citometria de Fluxo , Proteínas de Choque Térmico HSP27/genética , Humanos , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Mesilato de Imatinib , Técnicas In Vitro , Interleucina-8/genética , Metilação/efeitos dos fármacos , Inibidor de NF-kappaB alfa , Fosforilação/efeitos dos fármacos , Reação em Cadeia da Polimerase em Tempo Real , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Fator de Transcrição RelA/genética
7.
Diabetes ; 60(7): 1946-54, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21617177

RESUMO

OBJECTIVE: Loss of thrombospondin (TSP)-1 in pancreatic islets has been shown to cause islet hyperplasia. This study tested the hypothesis that endothelial-derived TSP-1 is important for ß-cell function. RESEARCH DESIGN AND METHODS: Islet function was evaluated both in vivo and in vitro. Messenger RNA and protein expression were measured by real-time PCR and Western blot, respectively. The role of endothelial-derived TSP-1 for ß-cell function was determined using a transplantation design in which recipient blood vessels either were allowed to grow or not into the transplanted islets. RESULTS: TSP-1-deficient mice were glucose intolerant, despite having an increased ß-cell mass. Moreover, their islets had decreased glucose-stimulated insulin release, (pro)insulin biosynthesis, and glucose oxidation rate, as well as increased expression of uncoupling protein-2 and lactate dehydrogenase-A when compared with control islets. Almost all TSP-1 in normal islets were found to be derived from the endothelium. Transplantation of free and encapsulated neonatal wild-type and TSP-1-deficient islets was performed in order to selectively reconstitute with TSP-1-positive or -negative blood vessels in the islets and supported that the ß-cell defects occurring in TSP-1-deficient islets reflected postnatal loss of the glycoprotein in the islet endothelial cells. Treatment of neonatal TSP-1-deficient mice with the transforming growth factor (TGF)ß-1-activating sequence of TSP-1 showed that reconstitution of TGFß-1 activation prevented the development of decreased glucose tolerance in these mice. Thus, endothelial-derived TSP-1 activates islet TGFß-1 of importance for ß-cells. CONCLUSIONS: Our study indicates a novel role for endothelial cells as functional paracrine support for pancreatic ß-cells.


Assuntos
Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/fisiologia , Trombospondina 1/fisiologia , Animais , Células Endoteliais/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Intolerância à Glucose/fisiopatologia , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Transplante das Ilhotas Pancreáticas/fisiologia , Camundongos , Trombospondina 1/biossíntese , Trombospondina 1/deficiência , Fator de Crescimento Transformador beta/metabolismo
8.
Clin Sci (Lond) ; 118(4): 241-7, 2009 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-19886867

RESUMO

Altered tyrosine kinase signalling has been implicated in several diseases, paving the way for the development of small-molecule TKIs (tyrosine kinase inhibitors). TKIs such as imatinib, sunitinib and dasatinib are clinically used for treating chronic myeloid leukaemia, gastrointestinal stromal tumours and other malignancies. In addition to their use as anti-cancer agents, increasing evidence points towards an anti-diabetic effect of these TKIs. Imatinib and other TKIs counteract diabetes not only in non-obese diabetic mice, but also in streptozotocin diabetic mice, db/db mice, high-fat-treated rats and humans with T2D (Type 2 diabetes). Although the mechanisms of protection need to be investigated further, the effects of imatinib and other TKIs in human T2D and the rapidly growing findings from animal models of T1D (Type 1 diabetes) and T2D are encouraging and give hope to improved treatment of human diabetes. In the present article, we review the anti-diabetic effects of TKIs which appear to involve both protection against beta-cell death and improved insulin sensitivity. Considering the relatively mild side effects of TKIs, we hypothesize that TKIs could be used to treat new-onset T1D, prevent T1D in individuals at high risk of developing the disease, treat the late stages of T2D and improve the outcome of islet transplantation.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Tirosina Quinases/antagonistas & inibidores , Animais , Benzamidas , Ensaios Clínicos como Assunto/métodos , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Tipo 1/prevenção & controle , Humanos , Mesilato de Imatinib , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Piperazinas/uso terapêutico , Pirimidinas/uso terapêutico
9.
J Endocrinol ; 203(2): 271-9, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19696098

RESUMO

Src homology 2 domain-containing protein B (SHB) is an adapter protein involved in the regulation of beta-cell and endothelial cell function. We have recently obtained the Shb knockout mouse, and consequently, the aim of this study was to assess the effect of Shb deletion upon beta-cell function and blood glucose homeostasis. Shb-/- mice display an elevated basal blood glucose concentration, and this increase is maintained during insulin challenge in insulin sensitivity tests. To assess glucose-induced insulin secretion, pancreata were perfused, and it was observed that Shb-/- first phase insulin secretion was blunted during glucose stimulation. Gene expression of Shb-/- islets shortly after isolation was altered, with increased pancreatic and duodenal homeobox gene-1 (Pdx1) gene expression and reduced expression of Vegf-A. Islet culture normalized Pdx1 gene expression. The microvascular density of the Shb-/- islets was reduced, and islet capillary endothelial cell morphology was changed suggesting an altered microvascular function as a contributing cause to the impaired secretory activity. Capacitance measurements of depolarization-induced exocytosis indicate a direct effect on the exocytotic machinery, in particular a dramatic reduction in readily releasable granules, as responsible for the insulin-secretory defect operating in Shb-/- islets. Shb-/- mice exhibited no alteration of islet volume or beta-cell area. In conclusion, loss of Shb impairs insulin secretion, alters islet microvascular morphology, and increases the basal blood glucose concentration. The impaired insulin secretory response is a plausible underlying cause of the metabolic impairment observed in this mutant mouse.


Assuntos
Glucose/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Animais , Glicemia/análise , Células Endoteliais/citologia , Proteínas de Homeodomínio/genética , Homeostase , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Patch-Clamp , Transativadores/genética , Fator A de Crescimento do Endotélio Vascular/genética
10.
Am J Physiol Endocrinol Metab ; 297(5): E1067-77, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19706790

RESUMO

The transcription factor nuclear factor (NF)-κB is known to modulate rates of apoptosis and may therefore play a role in the increased ß-cell death that occurs in type 1 and type 2 diabetes. The aim of the present investigation was to study the expression of NF-κB subunits in human islet cells and whether overexpression of the NF-κB subunit c-Rel affects islet cell survival. We detected expression of p65, Rel-B, p50, p105, p52, and the ribosomal protein S3 (rpS3) in human islet cells. Among these, only p65 and rpS3 were translocated from the cytosolic to the nuclear fraction in response to cytokines. Interestingly, rpS3 participated in p65 binding to the κB-element in gel shift analysis experiments. We observed cytoplasmic c-Rel expression in vivo in 6J mice, and signs of nuclear translocation in ß-cells of infiltrated nonobese diabetic islets. Human islet cells were also dispersed by trypsin treatment and transduced with a c-Rel adenoviral vector. This resulted in increased expression of c-Rel and inhibitory factor κB, increased κB-binding activity, and augmented protein levels of Bcl-X(L,) c-IAP2, and heat shock protein 72. c-Rel expression in human islet cells protected against cytokine-induced caspase 3 activation and cell death. c-Rel protected also against streptozotocin- and H(2)O(2)-induced cell death, in both intact rat islets and human islet cells. We conclude that rpS3 participates in NF-κB signaling and that a genetic increase in the activity of the NF-κB subunit c-Rel results in protection against cell death in human islets.


Assuntos
Ilhotas Pancreáticas/efeitos dos fármacos , NF-kappa B/biossíntese , NF-kappa B/fisiologia , Subunidades Proteicas/biossíntese , Subunidades Proteicas/fisiologia , Adenoviridae/genética , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Western Blotting , Morte Celular/fisiologia , Sobrevivência Celular/fisiologia , Corantes , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Expressão Gênica/fisiologia , Vetores Genéticos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Microscopia Confocal , Proteínas Nucleares/biossíntese , Proteínas Nucleares/isolamento & purificação , Perfusão , Ratos , Ratos Sprague-Dawley , Sais de Tetrazólio , Tiazóis , Transdução Genética
11.
Biochem Biophys Res Commun ; 387(3): 553-7, 2009 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-19615333

RESUMO

Type 1 diabetes may depend on cytokine-induced beta-cell death and therefore the current investigation was performed in order to elucidate this response in Shb-deficient islets. A combination of interleukin-1beta and interferon-gamma caused a diminished beta-cell death response in Shb null islets. Furthermore, the induction of an unfolded protein response (UPR) by adding cyclopiazonic acid did not increase cell death in Shb-deficient islets, despite simultaneous expression of UPR markers. The heat-shock protein Hsp70 was more efficiently induced in Shb knockout islets, providing an explanation for the decreased susceptibility of Shb-deficient islets to cytokines. It is concluded that islets deficient in the Shb protein are less susceptible to cytotoxic conditions, and that this partly depends on their increased ability to induce Hsp70 under such circumstances. Interference with Shb signaling may provide means to improve beta-cell viability under conditions of beta-cell stress.


Assuntos
Apoptose , Diabetes Mellitus Tipo 1/imunologia , Proteínas de Choque Térmico HSP70/biossíntese , Ilhotas Pancreáticas/imunologia , Proteínas Proto-Oncogênicas/metabolismo , Animais , Interferon gama/imunologia , Interferon gama/farmacologia , Interleucina-1beta/imunologia , Interleucina-1beta/farmacologia , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas/genética
12.
Diabetes ; 57(7): 1896-904, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18420486

RESUMO

OBJECTIVE: The transcription factor nuclear factor-kappaB (NF-kappaB) and the mitogen-activated protein kinases (MAPKs) c-Jun NH(2)-terminal kinase (JNK) 1/2 are known to play decisive roles in cytokine-induced damage of rodent beta-cells. The upstream events by which these factors are activated in response to cytokines are, however, uncharacterized. The aim of the present investigation was to elucidate a putative role of the MAPK kinase kinase-1 (MEKK-1) in cytokine-induced signaling. RESEARCH DESIGN AND METHODS: To establish a functional role of MEKK-1, the effects of transient MEKK-1 overexpression in betaTC-6 cells, achieved by lipofection and cell sorting, and MEKK-1 downregulation in betaTC-6 cells and human islet cells, achieved by diced-small interfering RNA treatment, were studied. RESULTS: We observed that overexpression of wild-type MEKK-1, but not of a kinase dead MEKK-1 mutant, resulted in potentiation of cytokine-induced JNK activation, inhibitor of kappaB (IkappaB) degradation, and cell death. Downregulation of MEKK-1 in human islet cells provoked opposite effects, i.e., attenuation of cytokine-induced JNK and MKK4 activation, IkappaB stability, and a less pronounced NF-kappaB translocation. betaTC-6 cells with a downregulated MEKK-1 expression displayed also a weaker cytokine-induced iNOS expression and lower cell death rates. Also primary mouse islet cells with downregulated MEKK-1 expression were protected against cytokine-induced cell death. CONCLUSIONS: MEKK-1 mediates cytokine-induced JNK- and NF-kappaB activation, and this event is necessary for iNOS expression and cell death.


Assuntos
Citocinas/farmacologia , Ilhotas Pancreáticas/enzimologia , MAP Quinase Quinase Quinase 1/metabolismo , NF-kappa B/fisiologia , Animais , Morte Celular , DNA Complementar/genética , Indução Enzimática , Genes Reporter , Humanos , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Óxido Nítrico Sintase Tipo II/biossíntese , Fosforilação , RNA Interferente Pequeno/genética
13.
Endocrinology ; 149(6): 3046-53, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18308848

RESUMO

The aim of the present investigation was to characterize the role of the MAPK kinase kinase-1 (MEKK-1) in stress-induced cell death of insulin producing cells. We observed that transient overexpression of the wild type MEKK-1 protein in the insulin-producing cell lines RIN-5AH and betaTC-6 increased c-Jun N-terminal kinase (JNK) phosphorylation and augmented cell death induced by diethylenetriamine/nitroso-1-propylhydrazino)-1-propanamine (DETA/NO), streptozotocin (STZ), and hydrogen peroxide (H2O2). Furthermore, DETA/NO or STZ induced a rapid threonine phosphorylation of MEKK-1. Silencing of MEKK-1 gene expression in betaTC-6 and human dispersed islet cells, using in vitro-generated diced small interfering RNA, resulted in protection from DETA/NO, STZ, H2O2, and tunicamycin induced cell death. Moreover, in DETA/NO-treated cells diced small interfering RNA-mediated down-regulation of MEKK-1 resulted in decreased activation of JNK but not p38 and ERK. Inhibition of JNK by treatment with SP600125 partially protected against DETA/NO- or STZ-induced cell death. In summary, our results support an essential role for MEKK-1 in JNK activation and stress-induced beta-cell death. Increased understanding of the signaling pathways that augment or diminish beta-cell MEKK-1 activity may aid in the generation of novel therapeutic strategies in the treatment of type 1 diabetes.


Assuntos
Ilhotas Pancreáticas/enzimologia , MAP Quinase Quinase Quinase 1/metabolismo , Animais , Morte Celular , Sobrevivência Celular , Diabetes Mellitus Tipo 1/terapia , Citometria de Fluxo , Genes Reporter , Humanos , Insulinoma/enzimologia , Insulinoma/patologia , Ilhotas Pancreáticas/citologia , MAP Quinase Quinase Quinase 1/genética , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Ratos , Transdução de Sinais , Transfecção , Células Tumorais Cultivadas
14.
Exp Cell Res ; 313(2): 284-91, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17112510

RESUMO

The adaptor protein Shb has previously been shown to regulate apoptosis in response to cytokines and inhibitors of angiogenesis although the mechanisms governing these effects have remained obscure. We currently demonstrate interactions between Shb and c-Abl and that Shb regulates c-Abl kinase activity. The data suggest that c-Abl binds to tyrosine phosphorylated Shb via a concerted effort involving both the c-Abl SH3 and SH2 domains. The biological significance of the Shb/c-Abl interaction was presently tested in overexpression experiments and was found to promote hydrogen peroxide-induced cell death. We also show by Shb knockdown experiments that Shb regulates c-Abl activity and modulates cell death in response to the genotoxic agent cisplatin and the endoplasmic reticulum stress-inducer tunicamycin. The findings are in agreement with the notion of Shb playing a pivotal role in modulating c-Abl pro-apoptotic signaling in response to various stress stimuli.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose , Proteínas Proto-Oncogênicas c-abl/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Morte Celular , Células Cultivadas , Cisplatino/toxicidade , Humanos , Fosforilação , Proteínas Proto-Oncogênicas/genética , Tunicamicina/toxicidade , Tirosina/metabolismo
15.
FASEB J ; 21(2): 618-28, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17135364

RESUMO

It was recently reported that tyrosine kinase inhibitor imatinib mesylate (Gleevec) improves Type 2 diabetes, possibly by decreasing insulin resistance. However, as both Type 2 and Type 1 diabetes are characterized by beta-cell dysfunction and death, we investigated whether imatinib counteracts diabetes by maintaining beta-cell function. We observed that imatinib counteracted diabetes in two animal models, the streptozotocin-injected mouse and the nonobese diabetes mouse, and that this was paralleled by a partial preservation of the beta-cell mass. In addition, imatinib decreased the death of human beta-cells in vitro when exposed to NO, cytokines, and streptozotocin. The imatinib effect was mimicked by siRNA-mediated knockdown of c-Abl mRNA. Imatinib enhanced beta-cell survival by promoting a state similar to ischemic preconditioning, as evidenced by NF-kappaB activation, increased NO and reactive oxygen species production, and depolarization of the inner mitochondrial membrane. Imatinib did not suppress islet cell death in the presence of an NF-kappaB inhibitor, suggesting that NF-kappaB activation is a necessary step in the antiapoptotic action of imatinib. We conclude that imatinib mediates beta-cell survival and that this could contribute to the beneficial effects observed in diabetes.


Assuntos
Apoptose/efeitos dos fármacos , Diabetes Mellitus Tipo 1/prevenção & controle , Ilhotas Pancreáticas/efeitos dos fármacos , NF-kappa B/metabolismo , Piperazinas/farmacologia , Pirimidinas/farmacologia , Animais , Benzamidas , Linhagem Celular Tumoral , Citocinas/metabolismo , Citocinas/farmacologia , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Mesilato de Imatinib , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Endogâmicos , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Compostos Nitrosos/farmacologia , Piperazinas/administração & dosagem , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-abl/genética , Proteínas Proto-Oncogênicas c-kit/genética , Pirimidinas/administração & dosagem , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Estreptozocina/farmacologia
16.
Ann N Y Acad Sci ; 1040: 114-22, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15891014

RESUMO

RNA interference (RNAi) is emerging as a powerful and convenient tool for studying gene function and genetic variation. RNAi is mediated by 21- to 23-nucleotide-long, small interfering RNAs (siRNA) produced from larger double-stranded RNAs in vivo by the RNase III family enzyme Dicer. To overcome the problems associated with the use of predesigned synthetic siRNA molecules, a novel method utilizing the in vitro activity of recombinant Dicer has been developed recently. In nonislet cells, it has been demonstrated that a pool of siRNA, generated by Dicer from in vitro transcribed dsRNA (d-siRNA), mediates convenient, efficient, and reproducible gene silencing in various cell types. The aim of this study was to evaluate the ability of d-siRNA to silence endogenous gene expression in pancreatic islet cells. We observed that liposomal transfection mediates efficient transport of siRNA in up to 90% of dispersed islet cells and that d-siRNA mediates almost complete and nontoxic silencing of an endogenous mRNA, the messenger coding for the nonreceptor tyrosine kinase c-Abl. The approach described here using d-siRNA provides an important tool for elucidating gene function in further studies of pancreatic islets and diabetes pathophysiology.


Assuntos
Inativação Gênica/fisiologia , Ilhotas Pancreáticas/metabolismo , RNA Interferente Pequeno/biossíntese , Ribonuclease III/fisiologia , Animais , Linhagem Celular , Humanos , Masculino , Camundongos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/fisiologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Ribonuclease III/biossíntese , Ribonuclease III/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA