Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Diabetes Res ; 2016: 1620821, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27413754

RESUMO

Using humanized mice with functional human islets, we investigated whether activating GPR119 by PSN632408, a small molecular agonist, can stimulate human ß-cell regeneration in vivo. Human islets were transplanted under the left kidney capsule of immunodeficient mice with streptozotocin- (STZ-) induced diabetes. The recipient mice were treated with PSN632408 or vehicle and BrdU daily. Human islet graft function in the mice was evaluated by nonfasting glucose levels, oral glucose tolerance, and removal of the grafts. Immunostaining for insulin, glucagon, and BrdU or Ki67 was performed in islet grafts to evaluate α- and ß-cell replication. Insulin and CK19 immunostaining was performed to evaluate ß-cell neogenesis. Four weeks after human islet transplantation, 71% of PSN632408-treated mice achieved normoglycaemia compared with 24% of vehicle-treated mice. Also, oral glucose tolerance was significantly improved in the PSN632408-treated mice. PSN632408 treatment significantly increased both human α- and ß-cell areas in islet grafts and stimulated α- and ß-cell replication. In addition, ß-cell neogenesis was induced from pancreatic duct cells in the islet grafts. Our results demonstrated that activation of GPR119 increases ß-cell mass by stimulating human ß-cell replication and neogenesis. Therefore, GPR119 activators may qualify as therapeutic agents to increase human ß-cell mass in patients with diabetes.


Assuntos
Proliferação de Células/fisiologia , Diabetes Mellitus Experimental/metabolismo , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Regeneração/fisiologia , Ácidos Heterocíclicos/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Glucagon/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , Oxidiazóis/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Regeneração/efeitos dos fármacos
2.
Diabetes ; 64(11): 3873-84, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26185279

RESUMO

An increasing number of therapies have proven effective at reversing hyperglycemia in the nonobese diabetic (NOD) mouse model of type 1 diabetes (T1D), yet situations of successful translation to human T1D are limited. This may be partly due to evaluating the effect of treating immediately at diagnosis in mice, which may not be reflective of the advanced disease state in humans at disease onset. In this study, we treated NOD mice with new-onset as well as established disease using various combinations of four drugs: antithymocyte globulin (ATG), granulocyte-colony stimulating factor (G-CSF), a dipeptidyl peptidase IV inhibitor (DPP-4i), and a proton pump inhibitor (PPI). Therapy with all four drugs induced remission in 83% of new-onset mice and, remarkably, in 50% of NOD mice with established disease. Also noteworthy, disease remission occurred irrespective of initial blood glucose values and mechanistically was characterized by enhanced immunoregulation involving alterations in CD4+ T cells, CD8+ T cells, and natural killer cells. This combination therapy also allowed for effective treatment at reduced drug doses (compared with effective monotherapy), thereby minimizing potential adverse effects while retaining efficacy. This combination of approved drugs demonstrates a novel ability to reverse T1D, thereby warranting translational consideration.


Assuntos
Soro Antilinfocitário/uso terapêutico , Diabetes Mellitus Tipo 1/tratamento farmacológico , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Hiperglicemia/tratamento farmacológico , Inibidores da Bomba de Prótons/uso terapêutico , Animais , Soro Antilinfocitário/farmacologia , Peptídeo C/sangue , Diabetes Mellitus Tipo 1/metabolismo , Inibidores da Dipeptidil Peptidase IV/farmacologia , Quimioterapia Combinada , Feminino , Fator Estimulador de Colônias de Granulócitos/farmacologia , Hiperglicemia/metabolismo , Insulina/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Inibidores da Bomba de Prótons/farmacologia , Resultado do Tratamento
3.
Lancet Diabetes Endocrinol ; 2(9): 710-8, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24997559

RESUMO

BACKGROUND: Type 1 diabetes results from autoimmune destruction of pancreatic ß cells. Findings from preclinical studies suggest that dipeptidyl peptidase-4 inhibitors and proton-pump inhibitors might enhance ß-cell survival and regeneration. We postulated that sitagliptin and lansoprazole would preserve ß-cell function in patients with recent-onset type 1 diabetes. METHODS: We did a double-blind, placebo-controlled, phase 2 trial (REPAIR-T1D). Participants aged 11-36 years, diagnosed with type 1 diabetes within the past 6 months were recruited from Sanford Health Systems (Sioux Falls, SD, USA; Fargo, ND, USA), Children's Hospitals and Clinics of Minnesota (St Paul, MN, USA), and Rady Children's Hospital (San Diego, CA, USA). Participants were randomly assigned (2:1) to receive oral sitagliptin (100 mg for participants ≥18 years, 50 mg for those <18 years) and lansoprazole (60 mg for participants ≥18 years, 30 mg for those <18 years) or matched placebo for 12 months. Randomisation was done by a blocked randomisation process (blocks of three and six), with separate streams for younger (<18 years) and older (≥18 years) participants, and males and females. All participants and personnel remained masked until after the completion of the final 12 month visit, at which time data were unmasked to the analysis team. The primary endpoint was C-peptide response to a mixed meal challenge at 12 months measured as 2 h area under curve. Analysis was by intention to treat. This trial is registered with ClinicalTrials.gov, number NCT01155284. FINDINGS: Between Sept 21, 2010, and May 29, 2012, 46 participants were randomly assigned to the treatment group and 22 to the placebo group; of whom 40 participants in the treatment group and 18 in the placebo group completed the 12-month treatment. At 12 months, the mean change in C-peptide area under curve was -229 pmol/L (95% CI -316 to -142) for the treatment group and -253 pmol/L (-383 to -123) for the placebo group; this difference was not significant (p=0·77). No adverse or serious adverse events were probably or definitely related to the study treatment. INTERPRETATION: Although the expected change in the primary endpoint was not achieved, not all participants had increases in glucagon-like peptide-1 and gastrin concentrations that were expected with treatment. Although participants did not have adverse events related to study drugs, the study is not powered to address safety definitively. Further trials including these drugs might be warranted, but should be designed to ensure appropriate selection of participants and increases in these intermediary hormones. FUNDING: Sanford Research and JDRF.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Inibidores da Dipeptidil Peptidase IV/administração & dosagem , Hipoglicemiantes/administração & dosagem , Células Secretoras de Insulina/efeitos dos fármacos , Lansoprazol/administração & dosagem , Pirazinas/administração & dosagem , Triazóis/administração & dosagem , Adolescente , Adulto , Criança , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/epidemiologia , Método Duplo-Cego , Quimioterapia Combinada , Feminino , Humanos , Células Secretoras de Insulina/metabolismo , Masculino , Fosfato de Sitagliptina , Resultado do Tratamento , Estados Unidos/epidemiologia
4.
PLoS One ; 8(1): e53345, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23382843

RESUMO

BACKGROUND: Activating G-protein coupled receptor 119 (GPR119) by its agonists can stimulate glucagon like peptide-1 (GLP-1) release. GLP-1 is rapidly degraded and inactivated by dipeptidylpeptidase-IV (DPP-IV). We studied the efficiency of combining PSN632408, a GPR119 agonist, with sitagliptin, a DPP-IV inhibitor, on ß-cell regeneration in diabetic mice. MATERIALS & METHODS: Diabetes in C57BL/6 mice was induced by streptozotocin. PSN632408 and sitagliptin alone or in combination were administered to diabetic mice for 7 weeks along with BrdU daily. Nonfasting blood glucose levels were monitored. After treatment, oral glucose tolerance test (OGTT), plasma active GLP-1 levels, ß-cell mass along with α- and ß-cell replication, and ß-cell neogenesis were evaluated. RESULTS: Normoglycemia was not achieved in vehicle-treated mice. By contrast, 32% (6 of 19) of PSN632408-treated diabetic mice, 36% (5 of 14) sitagliptin-treated diabetic mice, and 59% (13 of 22) diabetic mice treated with PSN632408 and sitagliptin combination achieved normoglycemia after 7 weeks treatment. Combination therapy significantly increased plasma active GLP-1 levels, improved glucose clearance, stimulated both α- and ß-cell replication, and augmented ß-cell mass. Furthermore, treatment with combination therapy induced ß-cell neogenesis from pancreatic duct-derived cells. CONCLUSION: Our results demonstrate that combining a GPR119 agonist with a DPP-IV inhibitor may offer a novel therapeutic strategy for stimulating ß-cell regeneration and reversing diabetes.


Assuntos
Dipeptidil Peptidase 4/metabolismo , Células Secretoras de Insulina/citologia , Camundongos Endogâmicos NOD/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Regeneração/efeitos dos fármacos , Ácidos Heterocíclicos/administração & dosagem , Animais , Glicemia/efeitos dos fármacos , Diabetes Mellitus Experimental , Inibidores da Dipeptidil Peptidase IV/administração & dosagem , Peptídeo 1 Semelhante ao Glucagon/sangue , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD/genética , Oxidiazóis/administração & dosagem , Pirazinas/administração & dosagem , Receptores Acoplados a Proteínas G/agonistas , Fosfato de Sitagliptina , Triazóis/administração & dosagem
5.
Diabetes ; 61(9): 2340-8, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22721971

RESUMO

Rapamycin/interleukin-2 (IL-2) combination treatment of NOD mice effectively treats autoimmune diabetes. We performed a phase 1 clinical trial to test the safety and immunologic effects of rapamycin/IL-2 combination therapy in type 1 diabetic (T1D) patients. Nine T1D subjects were treated with 2-4 mg/day rapamycin orally for 3 months and 4.5 × 10(6) IU IL-2 s.c. three times per week for 1 month. ß-Cell function was monitored by measuring C-peptide. Immunologic changes were monitored using flow cytometry and serum analyses. Regulatory T cells (Tregs) increased within the first month of therapy, yet clinical and metabolic data demonstrated a transient worsening in all subjects. The increase in Tregs was transient, paralleling IL-2 treatment, whereas the response of Tregs to IL-2, as measured by STAT5 phosphorylation, increased and persisted after treatment. No differences were observed in effector T-cell subset frequencies, but an increase in natural killer cells and eosinophils occurred with IL-2 therapy. Rapamycin/IL-2 therapy, as given in this phase 1 study, resulted in transient ß-cell dysfunction despite an increase in Tregs. Such results highlight the difficulties in translating therapies to the clinic and emphasize the importance of broadly interrogating the immune system to evaluate the effects of therapy.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Células Secretoras de Insulina/fisiologia , Interleucina-2/administração & dosagem , Sirolimo/administração & dosagem , Linfócitos T Reguladores/imunologia , Animais , Diabetes Mellitus Tipo 1/imunologia , Quimioterapia Combinada , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Interleucina-2/uso terapêutico , Camundongos , Camundongos Endogâmicos NOD
6.
Cell Transplant ; 20(9): 1343-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21396168

RESUMO

Combination therapy with a dipeptidyl peptidase-4 inhibitor (DPP-4i) and a proton pump inhibitor (PPI) raises endogenous levels of GLP-1 and gastrin, respectively, and restores pancreatic ß-cell mass and normoglycemia in nonobese diabetic (NOD) mice with autoimmune diabetes. The aim of this study was to determine whether a DPP-4i and PPI combination could increase ß-cell mass in the adult human pancreas. Pancreatic cells from adult human pancreas donors were implanted in NOD-severe combined immunodeficient (NOD-scid) mice and the mice were treated with a DPP-4i and a PPI for 16 weeks. Human grafts were examined for insulin content and insulin-stained cells. Graft ß-cell function was assessed by intravenous glucose tolerance tests (IVGTT) and by glucose control in human cell-engrafted mice treated with streptozotocin (STZ) to delete mouse pancreatic ß-cells. Plasma GLP-1 and gastrin levels were raised to two- to threefold in DPP-4i- and PPI-treated mice. Insulin content and insulin-stained cells in human pancreatic cell grafts were increased 9- to 13-fold in DPP-4i and PPI-treated mice and insulin-stained cells were colocalized with pancreatic exocrine duct cells. Plasma human C-peptide responses to IVGTT were significantly higher and STZ-induced hyperglycemia was more completely prevented in DPP-4i- and PPI-treated mice with grafts than in vehicle-treated mice with grafts. In conclusion, DPP-4i and PPI combination therapy raises endogenous levels of GLP-1 and gastrin and greatly expands the functional ß-cell mass in adult human pancreatic cells implanted in immunodeficient mice, largely from pancreatic duct cells. This suggests that a DPP-4i and PPI combination treatment may provide a pharmacologic therapy to correct the ß-cell deficit in type 1 diabetes.


Assuntos
Inibidores da Dipeptidil Peptidase IV/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Organogênese/efeitos dos fármacos , Ductos Pancreáticos/citologia , Ductos Pancreáticos/transplante , Inibidores da Bomba de Prótons/farmacologia , Adulto , Animais , Transplante de Células , Quimioterapia Combinada , Feminino , Humanos , Células Secretoras de Insulina/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Ductos Pancreáticos/efeitos dos fármacos
7.
Diabetes ; 57(12): 3281-8, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18835930

RESUMO

OBJECTIVE: Glucagon-like peptide-1 (GLP-1) and gastrin promote pancreatic beta-cell function, survival, and growth. Here, we investigated whether GLP-1 and gastrin can restore the beta-cell mass and reverse hyperglycemia in NOD mice with autoimmune diabetes. RESEARCH DESIGN AND METHODS: Acutely diabetic NOD mice were treated with GLP-1 and gastrin, separately or together, twice daily for 3 weeks. Blood glucose was measured weekly and for a further 5 weeks after treatments, after which pancreatic insulin content and beta-cell mass, proliferation, neogenesis, and apoptosis were measured. Insulin autoantibodies were measured, and adoptive transfer of diabetes and syngeneic islet transplant studies were done to evaluate the effects of GLP-1 and gastrin treatment on autoimmunity. RESULTS: Combination therapy with GLP-1 and gastrin, but not with GLP-1 or gastrin alone, restored normoglycemia in diabetic NOD mice. The GLP-1 and gastrin combination increased pancreatic insulin content, beta-cell mass, and insulin-positive cells in pancreatic ducts, and beta-cell apoptosis was decreased. Insulin autoantibodies were reduced in GLP-1- and gastrin-treated NOD mice, and splenocytes from these mice delayed adoptive transfer of diabetes in NOD-scid mice. Syngeneic islet grafts in GLP-1 -and gastrin-treated NOD mice were infiltrated by leukocytes with a shift in cytokine expression from interferon-gamma to transforming growth factor-beta1, and beta-cells were protected from apoptosis. CONCLUSIONS: Combination therapy with GLP-1 and gastrin restores normoglycemia in diabetic NOD mice by increasing the pancreatic beta-cell mass and downregulating the autoimmune response.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Tipo 1/sangue , Gastrinas/uso terapêutico , Peptídeo 1 Semelhante ao Glucagon/uso terapêutico , Animais , Apoptose , Autoanticorpos/sangue , Glicemia/efeitos dos fármacos , Divisão Celular , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Quimioterapia Combinada , Gastrinas/administração & dosagem , Peptídeo 1 Semelhante ao Glucagon/administração & dosagem , Injeções Intraperitoneais , Anticorpos Anti-Insulina/sangue , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/patologia , Transplante das Ilhotas Pancreáticas , Camundongos , Camundongos Endogâmicos NOD , Valores de Referência
8.
Cell Transplant ; 17(6): 631-40, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18819251

RESUMO

Pancreatic islet transplantation as a treatment for type 1 diabetes is limited by human donor tissue availability. We investigated whether the beta-cell mass in human isolated islets could be expanded by treatments with glucagon-like peptide-1 (GLP-1) and gastrin, peptides reported to stimulate beta-cell growth in mice and rats with deficits in beta-cell mass. Human islets with low endocrine cell purity (7% beta-cells, 4% alpha-cells) and abundant exocrine cells (29% duct cells and 25% acinar cells) were implanted under the renal capsule of nonobese diabetic-severe combined immune deficiency (NOD-scid) mice made diabetic with streptozotocin. The mice were treated with GLP-1 and gastrin, separately and together, daily for 5 weeks. Blood glucose was significantly reduced only in mice implanted with human pancreatic cells and treated with GLP-1 plus gastrin. Correction of hyperglycemia was accompanied by increased insulin content in the human pancreatic cell grafts as well as by increased plasma levels of human C-peptide in the mice. Immunocytochemical examination revealed a fourfold increase in insulin-positive cells in the human pancreatic cell grafts in GLP-1 plus gastrin-treated mice, and most of this increase was accounted for by the appearance of cytokeratin 19-positive pancreatic duct cells expressing insulin. We conclude that combination therapy with GLP-1 and gastrin expands the beta-cell mass in human islets implanted in immunodeficient diabetic mice, largely from pancreatic duct cells associated with the islets, and this is sufficient to ameliorate hyperglycemia in the mice.


Assuntos
Linfócitos B/fisiologia , Diabetes Mellitus Experimental/terapia , Gastrinas/farmacologia , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Ductos Pancreáticos , Animais , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Glicemia/metabolismo , Peptídeo C/metabolismo , Quimioterapia Combinada , Feminino , Humanos , Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Ductos Pancreáticos/citologia , Ductos Pancreáticos/efeitos dos fármacos , Ratos , Transplante Heterólogo
9.
Cell Biochem Biophys ; 48(2-3): 127-37, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17709882

RESUMO

Islet Neogenesis-Associated Protein (INGAP) is a member of the Reg family of proteins implicated in various settings of endogenous pancreatic regeneration. The expression of INGAP and other RegIII proteins has also been linked temporally and spatially with the induction of islet neogenesis in animal models of disease and regeneration. Furthermore, administration of a peptide fragment of INGAP (INGAP peptide) has been demonstrated to reverse chemically induced diabetes as well as improve glycemic control and survival in an animal model of type 1 diabetes. Cultured human pancreatic tissue has also been shown to be responsive to INGAP peptide, producing islet-like structures with function, architecture and gene expression matching that of freshly isolated islets. Likewise, studies in normoglycemic animals show evidence of islet neogenesis. Finally, recent clinical studies suggest an effect of INGAP peptide to improve insulin production in type 1 diabetes and glycemic control in type 2 diabetes.


Assuntos
Antígenos de Neoplasias/fisiologia , Biomarcadores Tumorais/fisiologia , Ilhotas Pancreáticas/fisiologia , Lectinas Tipo C/fisiologia , Regeneração/fisiologia , Animais , Antígenos de Neoplasias/farmacologia , Biomarcadores Tumorais/farmacologia , Ensaios Clínicos Fase II como Assunto , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/metabolismo , Avaliação Pré-Clínica de Medicamentos , Humanos , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Pâncreas/fisiologia , Proteínas Associadas a Pancreatite , Regeneração/efeitos dos fármacos
10.
Cell Biochem Biophys ; 48(2-3): 159-63, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17709885

RESUMO

Type 1 diabetes (T1D) results from autoimmune destruction of the insulin-producing beta-cells in the pancreatic islets of Langerhans by autoreactive T helper 1 (Th1) cells characterized by their cytokine secretory products, interleukin-2 (IL-2) and interferon gamma (IFNgamma). Th1-type cytokines (IL-2 and IFNgamma) correlate with T1D, whereas Th2 (IL-4 and IL-10), Th3 (transforming growth factor beta [TGFbeta]), and T regulatory cell-type cytokines (IL-10 and TGFbeta) correlate with protection from T1D. Paradoxically, however, administrations of Th1-type cytokines (IL-2 and IFNgamma) and immunotherapies that induce Th1-type cytokine responses actually prevent T1D, at least in animal models. Therefore, immunotherapies that inhibit IL-2 production/action will block Th1 cell/cytokine-driven effector mechanisms of pancreatic islet beta-cell destruction; however, anti-IL-2 therapy will not allow immune tolerance to be established. In contrast, immunotherapies that increase IL-2 production/action may correct an immunodeficiency in IL-2 production that appears to underlie the autoimmunity of T1D, thereby restoring immune tolerance to islet beta-cells and prevention of T1D.


Assuntos
Citocinas/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Animais , Apoptose/efeitos dos fármacos , Citocinas/farmacologia , Citocinas/uso terapêutico , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/etiologia , Humanos , Tolerância Imunológica/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , Interleucina-2/metabolismo , Interleucina-2/farmacologia , Interleucina-2/uso terapêutico , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos NOD , Modelos Imunológicos , Sirolimo/farmacologia , Sirolimo/uso terapêutico , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismo
11.
Diabetes ; 54(9): 2596-601, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16123347

RESUMO

Combination therapy with epidermal growth factor (EGF) and gastrin induces beta-cell regeneration in rodents with chemically induced diabetes. We investigated whether EGF plus gastrin could correct hyperglycemia in NOD mice with autoimmune diabetes. Combined treatment with EGF (1 mug/kg) and gastrin (3 mug/kg) for 2 weeks restored normoglycemia after diabetes onset in NOD mice, whereas EGF or gastrin alone did not. Fasting blood glucose remained normal (3.5-6.5 mmol/l) or mildly elevated (<11 mmol/l) in five of six mice (83%) for 10 weeks after EGF plus gastrin treatment was stopped, whereas all mice treated with vehicle or EGF or gastrin alone became severely hyperglycemic (12-35 mmol/l). Pancreatic beta-cell mass was increased threefold and insulin content was increased eightfold in mice treated with EGF plus gastrin compared with pretreatment values. The correction of hyperglycemia correlated significantly with increases in pancreatic beta-cell mass and insulin content. In addition, splenic cells from mice treated with EGF plus gastrin delayed diabetes induction by adoptive transfer of diabetogenic cells into immunodeficient NOD-scid mice, suggesting the induction of immunoregulatory cells in NOD mice treated with EGF plus gastrin. We conclude that a short course of combined EGF and gastrin therapy increases pancreatic beta-cell mass and reverses hyperglycemia in acutely diabetic NOD mice; the impact of this combined therapy may result from the effects of EGF and gastrin on beta-cells, immune cells, or both.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Fator de Crescimento Epidérmico/administração & dosagem , Gastrinas/administração & dosagem , Hiperglicemia/fisiopatologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/fisiologia , Animais , Diabetes Mellitus Tipo 1/fisiopatologia , Quimioterapia Combinada , Feminino , Camundongos , Camundongos Endogâmicos NOD
12.
J Immunol ; 175(2): 1248-56, 2005 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16002729

RESUMO

Nuclear and mitochondrial genomes combine in ALR/Lt mice to produce systemically elevated defenses against free radical damage, rendering these mice resistant to immune-mediated pancreatic islet destruction. We analyzed the mechanism whereby isolated islets from ALR mice resisted proinflammatory stress mediated by combined cytokines (IL-1beta, TNF-alpha, and IFN-gamma) in vitro. Such damage entails both superoxide and NO radical generation, as well as peroxynitrite, resulting from their combination. In contrast to islets from other mouse strains, ALR islets expressed constitutively higher glutathione reductase, glutathione peroxidase, and higher ratios of reduced to oxidized glutathione. Following incubation with combined cytokines, islets from control strains produced significantly higher levels of hydrogen peroxide and NO than islets from ALR mice. Nitrotyrosine was generated in NOD and C3H/HeJ islets but not by ALR islets. Western blot analysis showed that combined cytokines up-regulated the NF-kappaB inducible NO synthase in NOD-Rag and C3H/HeJ islets but not in ALR islets. This inability of cytokine-treated ALR islets to up-regulate inducible NO synthase and produce NO correlated both with reduced kinetics of IkappaB degradation and with markedly suppressed NF-kappaB p65 nuclear translocation. Hence, ALR/Lt islets resist cytokine-induced diabetogenic stress through enhanced dissipation and/or suppressed formation of reactive oxygen and nitrogen species, impaired IkappaB degradation, and blunted NF-kappaB activation. Nitrotyrosylation of beta cell proteins may generate neoantigens; therefore, resistance of ALR islets to nitrotyrosine formation may, in part, explain why ALR mice are resistant to type 1 diabetes when reconstituted with a NOD immune system.


Assuntos
Citocinas/toxicidade , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/patologia , Transporte Ativo do Núcleo Celular , Animais , Biomarcadores/metabolismo , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/fisiopatologia , Feminino , Radicais Livres/metabolismo , Quinase I-kappa B , Proteínas I-kappa B/metabolismo , Imunidade Inata/genética , Mediadores da Inflamação/toxicidade , Ilhotas Pancreáticas/enzimologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos NOD , Camundongos Endogâmicos , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase Tipo II , Estresse Oxidativo , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/metabolismo , Espécies Reativas de Nitrogênio/biossíntese , Fator de Transcrição RelA
13.
J Clin Endocrinol Metab ; 90(6): 3401-9, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15769977

RESUMO

Pancreatic islet transplantation is a viable treatment for type 1 diabetes, but is limited by human donor tissue availability. The combination of epidermal growth factor (EGF) and gastrin induces islet beta-cell neogenesis from pancreatic exocrine duct cells in rodents. In this study we investigated whether EGF and gastrin could expand the beta-cell mass in adult human isolated islets that contain duct as well as endocrine cells. Human islet cells were cultured for 4 wk in serum-free medium (control) or in medium with EGF (0.3 mug/ml), gastrin (1.0 mug/ml), or the combination of EGF and gastrin. beta-Cell numbers were increased in cultures with EGF plus gastrin (+118%) and with EGF (+81%), but not in cultures with gastrin (-3%) or control medium (-62%). After withdrawal of EGF and gastrin and an additional 4 wk in control medium, beta-cell numbers continued to increase only in cultures previously incubated with both EGF and gastrin (+232%). EGF plus gastrin also significantly increased cytokeratin 19-positive duct cells (+678%) in the cultures. Gastrin, alone or in combination with EGF, but not EGF alone, increased the expression of pancreatic and duodenal homeobox factor-1 as well as insulin and C peptide in the cytokeratin 19-positive duct cells. Also, EGF plus gastrin significantly increased beta-cells and insulin content in human islets implanted in immunodeficient nonobese diabetic-severe combined immune deficiency mice as well as insulin secretory responses of the human islet grafts to glucose challenge. In conclusion, combination therapy with EGF and gastrin increases beta-cell mass in adult human pancreatic islets in vitro and in vivo, and this appears to result from the induction of beta-cell neogenesis from pancreatic exocrine duct cells.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Gastrinas/farmacologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/fisiologia , Ductos Pancreáticos/citologia , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Humanos , Ilhotas Pancreáticas/efeitos dos fármacos , Ductos Pancreáticos/efeitos dos fármacos , Fatores de Tempo , Transativadores/genética
15.
Mol Med ; 9(3-4): 96-104, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12865945

RESUMO

Inosine, a naturally occurring purine, was long considered to be an inactive metabolite of adenosine. However, recently inosine has been shown to be an immunomodulator and anti-inflammatory agent. The aim of this study was to determine whether inosine influences anti-inflammatory effects and affects the development of type 1 diabetes in murine models. Type 1 diabetes was induced either chemically by streptozotocin or genetically using the nonobese diabetic mouse (NOD) model. Mice were treated with inosine (100 or 200 mg kg(-1)d(-1)d) and diabetes incidence was monitored. The effect of inosine on pancreas immune cell infiltration, oxidative stress, and cytokine profile also was determined. For the transplantation model islets were placed under the renal capsule of NOD mice and inosine (200 mg kg(-1)d d(-1)d) treatment started the day of islet transplantation. Graft rejection was diagnosed by return of hyperglycemia accompanied by glucosuria and ketonuria. Inosine reduced the incidence of diabetes in both streptozotocin-induced diabetes and spontaneous diabetes in NOD mice. Inosine decreased pancreatic leukocyte infiltration and oxidative stress in addition to switching the cytokine profile from a Th1 to a Th2 profile. Inosine prolonged pancreatic islet graft survival, increased the number of surviving beta cells, and reduced the number of infiltrating leukocytes. Inosine protects against both the development of diabetes and against the rejection of transplanted islets. The purine exerts anti-inflammatory effects in the pancreas, which is its likely mode of action. The use of inosine should be considered as a potential preventative therapy in humans susceptible to developing Type 1 diabetes and as a possible antirejection therapy for islet transplant recipients.


Assuntos
Diabetes Mellitus Experimental/prevenção & controle , Diabetes Mellitus Tipo 1/prevenção & controle , Rejeição de Enxerto/prevenção & controle , Inosina/farmacologia , Transplante das Ilhotas Pancreáticas , Animais , Glicemia/metabolismo , Sobrevivência Celular , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/genética , Feminino , Humanos , Hiperglicemia/tratamento farmacológico , Hiperglicemia/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Leucócitos , Peroxidação de Lipídeos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Naftalenos , Pâncreas/imunologia , Pâncreas/metabolismo , Estreptozocina
16.
Diabetes ; 52(7): 1683-8, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12829633

RESUMO

Poly (ADP-ribose) polymerase (PARP) is a nuclear enzyme that consumes NAD in response to DNA strand breaks. The PARP inhibitor nicotinamide prevents NAD consumption and protects islet beta-cells from chemically induced necrosis but not cytokine-induced apoptosis. Therefore, it is unclear how nicotinamide protects NOD mice from autoimmune diabetes in which apoptosis is the mode of beta-cell death. To investigate the mechanism of diabetes prevention by PARP inhibition, we studied the effects of a novel, potent PARP inhibitor, PJ34, a phenanthridinone derivative, on diabetes development in NOD mice and on diabetes recurrence in diabetic NOD mice transplanted with syngeneic islets. PJ34 administration from age 5 or 15 weeks significantly decreased insulitis, beta-cell destruction and diabetes incidence, and protection from diabetes continued for 12 weeks after PJ34 therapy was stopped. Similarly, syngeneic islet graft survival was prolonged and outlasted therapy in PJ34-treated mice. Immunohistochemical studies revealed significantly fewer leukocytes in islet grafts of PJ34-treated mice, together with increased apoptosis of these cells and decreased expression of the T helper 1-type cytokine interferon (IFN)-gamma. These results suggest that PARP inhibition protects against autoimmune beta-cell destruction in NOD mice by inducing apoptosis of islet-infiltrating leukocytes and decreasing IFN-gamma expression in the islets.


Assuntos
Apoptose/efeitos dos fármacos , Diabetes Mellitus Tipo 1/prevenção & controle , Transplante das Ilhotas Pancreáticas/fisiologia , Ilhotas Pancreáticas/patologia , Leucócitos/patologia , Niacinamida/farmacologia , Fenantrenos/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases , Envelhecimento , Animais , Diabetes Mellitus Tipo 1/imunologia , Inibidores Enzimáticos/farmacologia , Interferon gama/genética , Interleucinas/genética , Leucócitos/citologia , Leucócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos NOD , RNA Mensageiro/genética , Recidiva , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transplante Isogênico
18.
Cell Transplant ; 11(6): 519-28, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12428741

RESUMO

Nonobese diabetic (NOD) mice develop diabetes and destroy syngeneic islet grafts through an autoimmune response. Because transforming growth factor (TGF)-beta1 downregulates immune responses, we tested whether overexpression of TGF-beta1 by gene transfection of NOD mouse islets could protect beta-cells in islet grafts from autoimmune destruction. NOD mouse islet cells were transfected with an adenoviral DNA expression vector encoding porcine latent TGF-beta1 (Ad TGF-beta1) or the adenoviral vector alone (control Ad vector). The frequency of total islet cells expressing TGF-beta1 protein was increased from 12 +/- 1% in control Ad vector-transfected cells to 89 +/- 4% in Ad TGF-beta1-transfected islet cells, and the frequency of beta-cells that expressed TGF-beta1 was increased from 12 +/- 1% to 60 +/- 7%. Also, secretion of TGF-beta1 was significantly increased in islets that overexpressed TGF-beta1. Ad TGF-beta1-transfected NOD mouse islets that overexpressed TGF-beta1 prevented diabetes recurrence after transplantation into diabetic NOD mice for a median of 22 days compared with only 7 days for control Ad vector-transfected islets (p = 0.001). Immunohistochemical examination of the islet grafts revealed significantly more TGF-beta1+ cells and insulin+ cells and significantly fewer CD45+ leukocytes in Ad TGF-beta1-transfected islet grafts. Also, islet beta-cell apoptosis was significantly decreased whereas apoptosis of graft-infiltrating leukocytes was significantly increased in Ad TGF-beta1-transfected islet grafts. These observations demonstrate that overexpression of TGF-beta1, by gene transfection of NOD mouse islets, protects islet beta-cells from apoptosis and autoimmune destruction and delays diabetes recurrence after islet transplantation.


Assuntos
Terapia Genética , Transplante das Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/imunologia , Fator de Crescimento Transformador beta/genética , Animais , Células Cultivadas , Feminino , Regulação da Expressão Gênica , Vetores Genéticos , Rejeição de Enxerto/prevenção & controle , Imuno-Histoquímica , Ilhotas Pancreáticas/citologia , Transplante das Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Endogâmicos NOD , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Suínos , Transfecção/métodos , Fator de Crescimento Transformador beta/metabolismo , Transplante Isogênico
19.
Diabetes ; 51(3): 638-45, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11872661

RESUMO

Sirolimus is an immunosuppressant that inhibits interleukin (IL)-2 signaling of T-cell proliferation but not IL-2-induced T-cell apoptosis. Therefore, we hypothesized that administration of IL-2, together with sirolimus, might shift T-cell proliferation to apoptosis and prevent autoimmune destruction of islet beta-cells. We found that sirolimus and IL-2 therapy of female NOD mice, beginning at age 10 weeks, was synergistic in preventing diabetes development, and disease prevention continued for 13 weeks after stopping sirolimus and IL-2 therapy. Similarly, sirolimus and IL-2 were synergistic in protecting syngeneic islet grafts from recurrent autoimmune destruction after transplantation in diabetic NOD mice, and diabetes did not recur after stopping sirolimus and IL-2 combination therapy. Immunocytochemical examination of islet grafts revealed significantly decreased numbers of leukocytes together with increased apoptosis of these cells in mice treated with sirolimus and IL-2, whereas beta-cells were more numerous, and significantly fewer were apoptotic. In addition, Th1-type cells (gamma-interferon-positive and IL-2(+)) were decreased the most, and Th2-type cells (IL-4(+) and IL-10(+)) and Th3-type cells (transforming growth factor-beta1(+)) were increased the most in islet grafts of sirolimus and IL-2-treated mice. We conclude that 1) combination therapy with sirolimus and IL-2 is synergistic in protecting islet beta-cells from autoimmune destruction; 2) diabetes prevention continues after withdrawal of therapy; and 3) the mechanism of protection involves a shift from Th1- to Th2- and Th3-type cytokine-producing cells, possibly due to deletion of autoreactive Th1 cells.


Assuntos
Doenças Autoimunes/prevenção & controle , Diabetes Mellitus Tipo 1/prevenção & controle , Imunossupressores/uso terapêutico , Interleucina-2/uso terapêutico , Sirolimo/uso terapêutico , Animais , Apoptose , Diabetes Mellitus Tipo 1/imunologia , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Imuno-Histoquímica , Imunossupressores/administração & dosagem , Interferon gama/análise , Interleucina-2/administração & dosagem , Interleucina-2/análise , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Transplante das Ilhotas Pancreáticas , Camundongos , Camundongos Endogâmicos NOD , Sirolimo/administração & dosagem , Fator de Necrose Tumoral alfa/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...