Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Diabetes Investig ; 15(6): 693-703, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38634411

RESUMO

AIMS/INTRODUCTION: To investigate the long-term efficacy of various encapsulated xenogeneic islet transplantation, and to explore the impact of different donor porcine genetic traits on islet transplantation outcomes. MATERIALS AND METHODS: Donor porcine islets were obtained from wild-type, α1,3-galactosyltransferase knockout (GTKO) and GTKO with overexpression of membrane cofactor protein genotype. Naked, alginate, alginate-chitosan (AC), alginate-perfluorodecalin (A-PFD) and AC-perfluorodecalin (AC-PFD) encapsulated porcine islets were transplanted into diabetic mice. RESULTS: In vitro assessments showed no differences in the viability and function of islets across encapsulation types and donor porcine islet genotypes. Xenogeneic encapsulated islet transplantation with AC-PFD capsules showed the most favorable long-term outcomes, maintaining normal blood glucose levels for 180 days. A-PFD capsules showed comparable results to AC-PFD capsules, followed by AC capsules and alginate capsules. Conversely, blood glucose levels in naked islet transplantation increased to >300 mg/dL within a week after transplantation. Naked islet transplantation outcomes showed no improvement based on donor islet genotype. However, alginate or AC capsules showed delayed increases in blood glucose levels for GTKO and GTKO with overexpression of membrane cofactor protein porcine islets compared with wild-type porcine islets. CONCLUSION: The AC-PFD capsule, designed to ameliorate both hypoxia and inflammation, showed the highest long-term efficacy in xenogeneic islet transplantation. Genetic modifications of porcine islets with GTKO or GTKO with overexpression of membrane cofactor protein did not influence naked islet transplantation outcomes, but did delay graft failure when encapsulated.


Assuntos
Diabetes Mellitus Experimental , Transplante das Ilhotas Pancreáticas , Transplante Heterólogo , Transplante das Ilhotas Pancreáticas/métodos , Animais , Suínos , Camundongos , Transplante Heterólogo/métodos , Diabetes Mellitus Experimental/terapia , Alginatos , Galactosiltransferases/genética , Sobrevivência de Enxerto , Ilhotas Pancreáticas , Glicemia/análise , Masculino , Genótipo , Doadores de Tecidos
2.
Diabetes Metab J ; 46(5): 677-688, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35124687

RESUMO

BACKGROUND: Neonatal porcine pancreatic cell clusters (NPCCs) have been proposed as an alternative source of ß cells for islet transplantation because of their low cost and growth potential after transplantation. However, the delayed glucose lowering effect due to the immaturity of NPCCs and immunologic rejection remain as a barrier to NPCC's clinical application. Here, we demonstrate accelerated differentiation and immune-tolerant NPCCs by in vitro chemical treatment and microencapsulation. METHODS: NPCCs isolated from 3-day-old piglets were cultured in F-10 media and then microencapsulated with alginate on day 5. Differentiation of NPCCs is facilitated by media supplemented with activin receptor-like kinase 5 inhibitor II, triiodothyronine and exendin-4 for 2 weeks. Marginal number of microencapsulated NPCCs to cure diabetes with and without differentiation were transplanted into diabetic mice and observed for 8 weeks. RESULTS: The proportion of insulin-positive cells and insulin mRNA levels of NPCCs were significantly increased in vitro in the differentiated group compared with the undifferentiated group. Blood glucose levels decreased eventually after transplantation of microencapsulated NPCCs in diabetic mice and normalized after 7 weeks in the differentiated group. In addition, the differentiated group showed nearly normal glucose tolerance at 8 weeks after transplantation. In contrast, neither blood glucose levels nor glucose tolerance were improved in the undifferentiated group. Retrieved graft in the differentiated group showed greater insulin response to high glucose compared with the undifferentiated group. CONCLUSION: in vitro differentiation of microencapsulated immature NPCCs increased the proportion of insulin-positive cells and improved transplant efficacy in diabetic mice without immune rejection.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Ilhotas Pancreáticas , Alginatos/metabolismo , Alginatos/farmacologia , Animais , Animais Recém-Nascidos , Glicemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/cirurgia , Exenatida/farmacologia , Insulina/metabolismo , Camundongos , RNA Mensageiro/metabolismo , RNA Mensageiro/farmacologia , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Suínos , Transplante Heterólogo , Tri-Iodotironina/metabolismo , Tri-Iodotironina/farmacologia
3.
Endocrinol Metab (Seoul) ; 36(1): 146-156, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33677936

RESUMO

BACKGROUND: The microencapsulation is an ideal solution to overcome immune rejection without immunosuppressive treatment. Poor biocompatibility and small molecular antigens secreted from encapsulated islets induce fibrosis infiltration. Therefore, the aims of this study were to improve the biocompatibility of microcapsules by dexamethasone coating and to verify its effect after xenogeneic transplantation in a streptozotocin-induced diabetes mice. METHODS: Dexamethasone 21-phosphate (Dexa) was dissolved in 1% chitosan and was cross-linked with the alginate microcapsule surface. Insulin secretion and viability assays were performed 14 days after microencapsulation. Dexa-containing chitosan-coated alginate (Dexa-chitosan) or alginate microencapsulated porcine islets were transplanted into diabetic mice. The fibrosis infiltration score was calculated from the harvested microcapsules. The harvested microcapsules were stained with trichrome and for insulin and macrophages. RESULTS: No significant differences in glucose-stimulated insulin secretion and islet viability were noted among naked, alginate, and Dexa-chitosan microencapsulated islets. After transplantation of microencapsulated porcine islets, nonfasting blood glucose were normalized in both the Dexa-chitosan and alginate groups until 231 days. The average glucose after transplantation were lower in the Dexa-chitosan group than the alginate group. Pericapsular fibrosis and inflammatory cell infiltration of microcapsules were significantly reduced in Dexa-chitosan compared with alginate microcapsules. Dithizone and insulin were positive in Dexa-chitosan capsules. Although fibrosis and macrophage infiltration was noted on the surface, some alginate microcapsules were stained with insulin. CONCLUSION: Dexa coating on microcapsules significantly suppressed the fibrotic reaction on the capsule surface after transplantation of xenogenic islets containing microcapsules without any harmful effects on the function and survival of the islets.


Assuntos
Quitosana , Diabetes Mellitus Experimental , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Alginatos/metabolismo , Alginatos/farmacologia , Animais , Cápsulas/metabolismo , Cápsulas/farmacologia , Quitosana/metabolismo , Quitosana/farmacologia , Dexametasona/metabolismo , Dexametasona/farmacologia , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/cirurgia , Fibrose , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Camundongos , Suínos
4.
J Diabetes Investig ; 11(2): 268-280, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31872946

RESUMO

There are numerous lines of clinical evidence that inhibition of the renin-angiotensin system (RAS) can prevent and delay the development of diabetes. Also, the role of RAS in the pathogenesis of diabetes, including insulin resistance and ß-cell dysfunction, has been extensively investigated. Nevertheless, this role had not yet been fully shown. A variety of possible protective mechanisms for RAS blockers in the regulation of glucose homeostasis have been suggested. However, the direct effect on pancreatic islet fibrosis has only recently been spotlighted. Various degrees of islet fibrosis are often observed in the islets of patients with type 2 diabetes mellitus, which can be associated with a decrease in ß-cell mass and function in these patients. Pancreatic stellate cells are thought to be deeply involved in this islet fibrosis. In this process, the activation of RAS in islets is shown to transform quiescent pancreatic stellate cells into the activated form, stimulates their proliferation and consequently leads to islet fibrotic destruction. In this article, we introduce existing clinical and experimental evidence for diabetes prevention through inhibition of RAS, and review the responsible local RAS signaling pathways in pancreatic stellate cells. Finally, we propose possible targets for the prevention of islet fibrosis.


Assuntos
Diabetes Mellitus Tipo 2/fisiopatologia , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/fisiopatologia , Células Estreladas do Pâncreas/fisiologia , Sistema Renina-Angiotensina , Animais , Diabetes Mellitus Tipo 2/terapia , Humanos
5.
J Tissue Eng Regen Med ; 12(4): e2110-e2122, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29330944

RESUMO

Hypoxic injury of islets is a major obstacle for encapsulated islet transplantation into the peritoneal cavity. To improve oxygen delivery to encapsulated islets, we integrated 20% of the oxygen carrier material, perfluorodecalin (PFD), in alginate capsules mixed with islets (PFD-alginate). Integration of PFD clearly improved islet viability and decreased reactive oxygen species production compared to islets encapsulated with alginate only (alginate) and naked islets exposed to hypoxia in vitro. In PFD-alginate capsules, HIF-1α expression was minimal, and insulin expression was well maintained. Furthermore, the best islet function represented by glucose-stimulated insulin secretion was observed for the PFD-alginate capsules in hypoxic condition. For the in vivo study, the same number of naked islets and encapsulated islets (alginate and PFD-alginate) was transplanted into streptozotocin-induced diabetic mice. Nonfasting blood glucose levels and the area under the curve for glucose based on intraperitoneal glucose tolerance tests in the PFD-alginate group were lower than in the alginate group. The harvested islets stained positive for insulin in all groups, but the ratio of dead cell area was 4 times higher in the alginate group than in the PFD-alginate group. In conclusion, integration of PFD in alginate microcapsules improved islet function and survival by minimizing the hypoxic damage of islets after intraperitoneal transplantation.


Assuntos
Diabetes Mellitus Experimental/terapia , Fluorocarbonos/química , Secreção de Insulina , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/metabolismo , Alginatos/química , Animais , Cápsulas , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Ilhotas Pancreáticas/patologia , Masculino , Ratos , Ratos Sprague-Dawley
6.
J Tissue Eng Regen Med ; 11(4): 1274-1284, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-26043934

RESUMO

Islet microencapsulation is an attractive strategy for the minimization or avoidance of life-long immunosuppression after transplantation. However, the clinical implementation of this technique is currently limited by incomplete biocompatibility. Thus, the aim of the present study was to demonstrate the improved biocompatibility of rapamycin-containing polyethylene glycol (Rapa-PEG)-coating on alginate microcapsules containing xenogeneic islets. The Rapa-PEG-coating on the alginate layer was observed using scanning electron microscopy (SEM) and the molecular cut-off weight of the microcapsules was approximately 70 kDa. The viabilities of the alginate-encapsulated and Rapa-PEG-coated alginate-encapsulated islets were lower than the viability of the naked islets just after encapsulation, but these the differences diminished over time in culture dishes. Rapa-PEG-coating on the alginate capsules effectively decreased the proliferation of macrophage cells compared to the non-coating and alginate coating of xenogeneic pancreas tissues. Glucose-stimulated insulin secretion did not significantly differ among the groups prior to transplantation. The random blood glucose levels of diabetic mice significantly improved following the transplantation of alginate-encapsulated and Rapa-PEG-coated alginate-encapsulated islets, but there were no significant differences between these two groups. However, there was a significant decrease in the number of microcapsules with fibrotic cell infiltration in the Rapa-PEG-coated alginate microcapsule group compared to the alginate microcapsule group. In conclusion, Rapa-PEG-coating might be an effective technique with which to improve the biocompatibility of microcapsules containing xenogeneic islets. Copyright © 2015 John Wiley & Sons, Ltd.


Assuntos
Alginatos/química , Materiais Revestidos Biocompatíveis/química , Ilhotas Pancreáticas/patologia , Polietilenoglicóis/química , Sirolimo/farmacologia , Transplante Heterólogo , Animais , Materiais Biocompatíveis/farmacologia , Glicemia/metabolismo , Cápsulas , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/patologia , Fibrose , Glucose/farmacologia , Ácido Glucurônico/química , Ácidos Hexurônicos/química , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Células RAW 264.7 , Sus scrofa
7.
Sci Rep ; 6: 23960, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-27044861

RESUMO

The preadipocyte factor 1 (Pref-1) is involved in the proliferation and differentiation of various precursor cells. However, the intracellular signaling pathways that control these processes and the role of Pref-1 in the pancreas remain poorly understood. Here, we showed that Pref-1 induces insulin synthesis and secretion via two independent pathways. The overexpression of Pref-1 activated MAPK signaling, which induced nucleocytoplasmic translocation of FOXO1 and PDX1 and led to the differentiation of human pancreatic ductal cells into ß-like cells and an increase in insulin synthesis. Concurrently, Pref-1 activated Akt signaling and facilitated insulin secretion. A proteomics analysis identified the Rab43 GTPase-activating protein as a downstream target of Akt. A serial activation of both proteins induced various granular protein syntheses which led to enhanced glucose-stimulated insulin secretion. In a pancreatectomised diabetic animal model, exogenous Pref-1 improved glucose homeostasis by accelerating pancreatic ductal and ß-cell regeneration after injury. These data establish a novel role for Pref-1, opening the possibility of applying this molecule to the treatment of diabetes.


Assuntos
Diferenciação Celular , Células Secretoras de Insulina/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Ductos Pancreáticos/citologia , Animais , Proteínas de Ligação ao Cálcio , Linhagem Celular Tumoral , Inibidores Enzimáticos/química , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína Forkhead Box O1/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Glucose/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Insulina/metabolismo , Sistema de Sinalização das MAP Quinases , Neoplasias Pancreáticas/metabolismo , Fosforilação , Proteômica , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Transativadores/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
8.
Transplantation ; 100(2): 334-43, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26479281

RESUMO

BACKGROUND: Clinical application of encapsulated islet transplantation is hindered by low biocompatibility of capsules leading to pericapsular fibrosis and decreased islet viability. To improve biocompatibility, we designed a novel chitosan-coated alginate capsules and compared them to uncoated alginate capsules. METHODS: Alginate capsules were formed by crosslinking with BaCl2, then they were suspended in chitosan solution for 10 minutes at pH 4.5. Xenogeneic islet transplantation, using encapsulated porcine islets in 1,3-galactosyltransferase knockout mice, and allogeneic islet transplantation, using encapsulated canine islets in beagles, were performed without immunosuppressants. RESULTS: The chitosan-alginate capsules showed similar pore size, islet viability, and insulin secretory function compared to alginate capsules, in vitro. Xenogeneic transplantation of chitosan-alginate capsules demonstrated a trend toward superior graft survival (P = 0.07) with significantly less pericapsular fibrosis (cell adhesion score: 3.77 ± 0.41 vs 8.08 ± 0.05; P < 0.001) compared to that of alginate capsules up to 1 year after transplantation. Allogeneic transplantation of chitosan-alginate capsules normalized the blood glucose level up to 1 year with little evidence of pericapsular fibrotic overgrowth on graft explantation. CONCLUSIONS: The efficacy and biocompatibility of chitosan-alginate capsules were demonstrated in xenogeneic and allogeneic islet transplantations using small and large animal models of diabetes. This capsule might be a potential candidate applicable in the treatment of type 1 diabetes mellitus patients, and further studies in nonhuman primates are required.


Assuntos
Alginatos/química , Quitosana/química , Materiais Revestidos Biocompatíveis , Diabetes Mellitus Experimental/cirurgia , Diabetes Mellitus Tipo 1/cirurgia , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/cirurgia , Animais , Compostos de Bário/química , Glicemia/metabolismo , Adesão Celular , Cloretos/química , Reagentes de Ligações Cruzadas/química , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/patologia , Cães , Feminino , Fibrose , Galactosiltransferases/deficiência , Galactosiltransferases/genética , Ácido Glucurônico/química , Sobrevivência de Enxerto , Xenoenxertos , Ácidos Hexurônicos/química , Insulina/sangue , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Transplante das Ilhotas Pancreáticas/efeitos adversos , Masculino , Camundongos , Camundongos Knockout , Porosidade , Células RAW 264.7 , Suínos , Porco Miniatura , Fatores de Tempo , Transplante Homólogo
9.
Xenotransplantation ; 22(4): 249-59, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26174875

RESUMO

BACKGROUND: Hepatocyte transplantation is a promising therapy for acute liver failure. Cell therapy using xenogeneic sources has emerged as an alternative treatment for patients with organ failure due to the shortage of transplantable human organs. The purpose of this study was to improve the survival of mice with acute liver failure by transplanting encapsulated neonatal pig re-aggregated liver cells (NPRLC). METHODS: Liver injury was induced in C57/BL6 male mice by the injection of 600 mg/kg of acetaminophen. Xenogeneic liver cells were isolated from a neonatal pig and processed via re-aggregation and encapsulation to improve the efficiency of the xenogeneic liver cell transplantation. The neonatal pig liver showed abnormal lobule structure. Isolated cells were re-aggregated and intraperitoneally transplanted into acute liver failure mice models. RESULTS: Re-aggregated cells showed significantly enhanced viability and significantly greater synthesis of albumin and urea than cells cultured in monolayers. Further, we observed improved serum levels of ALT/AST, and the survival rate of mice with acute liver failure was improved by the intraperitoneal transplantation of encapsulated hepatocytes (48,000 equivalent (Eq) per mouse). CONCLUSIONS: This study shows that using encapsulated NPRLCs improves the efficacy of xenogeneic liver cell transplantation for the treatment of mice with acute liver failure. Therefore, this may be a good strategy for bridge therapy for the treatment of acute liver failure in humans.


Assuntos
Hepatócitos/transplante , Falência Hepática Aguda/terapia , Transplante Heterólogo/métodos , Acetaminofen/toxicidade , Albuminas/biossíntese , Animais , Animais Recém-Nascidos , Agregação Celular , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , Sobrevivência de Enxerto , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Falência Hepática Aguda/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Esferoides Celulares/transplante , Sus scrofa , Suínos , Porco Miniatura , Ureia/metabolismo
10.
J Korean Med Sci ; 30(7): 991-4, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26130966

RESUMO

Pancreatic islet transplantation is a physiologically advantageous and minimally invasive procedure for the treatment of type 1 diabetes mellitus. Here, we describe the first reported case of successful allogeneic islet transplantation alone, using single-donor, marginal-dose islets in a Korean patient. A 59-yr-old patient with type 1 diabetes mellitus, who suffered from recurrent severe hypoglycemia, received 4,163 islet equivalents/kg from a single brain-death donor. Isolated islets were infused intraportally without any complications. The immunosuppressive regimen was based on the Edmonton protocol, but the maintenance dosage was reduced because of mucositis and leukopenia. Although insulin independence was not achieved, the patient showed stabilized blood glucose concentration, reduced insulin dosage and reversal of hypoglycemic unawareness, even with marginal dose of islets and reduced immunosuppressant. Islet transplantation may successfully improve endogenous insulin production and glycemic stability in subjects with type 1 diabetes mellitus.


Assuntos
Diabetes Mellitus Tipo 1/cirurgia , Hipoglicemia/cirurgia , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/cirurgia , Glicemia/análise , Feminino , Humanos , Terapia de Imunossupressão/métodos , Imunossupressores/uso terapêutico , Ilhotas Pancreáticas/fisiologia , Pessoa de Meia-Idade , República da Coreia , Doadores de Tecidos
11.
Transplantation ; 97(3): 273-9, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24448589

RESUMO

BACKGROUND: Peroxisome proliferator-activated receptor gamma-coactivator-1α (PGC-1α) has recently been implicated as a crucial factor in the glucocorticoid-suppressed expansion and transdifferentiation of porcine neonatal pancreatic cell clusters (NPCCs). However, the molecular mechanism has not been clarified. METHODS: We investigated whether the suppression of PGC-1α expression protects against ß-cell dysfunction induced by dexamethasone (Dx) treatment in vitro and in vivo and determined the mechanism of action of PGC-1α in porcine NPCCs. RESULTS: The reduction in Pdx-1 gene expression caused by either Dx treatment or PGC-1α overexpression was normalized by siPGC-1α. Nuclear FOXO1 and cytoplasmic Pdx-1 were detected after Dx treatment. However, FOXO1 was observed in the cytoplasm, and Pdx-1 was observed in the nucleus after siPGC-1α. Suppression of PGC-1α by siPGC-1α improved the Dx-induced repression of insulin secretion and insulin content. In vivo studies showed that the glucose level in the Ad-siPGC-1α-infected group was significantly lower than that in the Dx-treated group. Insulin expression in the graft tissue disappeared in the Dx-injected group. However, the siPGC-1α- and Dx-treated group showed increased insulin expression and an increase in graft mass, ß-cell mass, and ß-cell % in the graft. Conversely, the Dx-induced ductal cystic area was markedly reduced in the siPGC-1α- and Dx-treated group. CONCLUSIONS: Our results suggest that the transdifferentiation of porcine NPCCs into ß cells is influenced by the duration of the Dx treatment, which might result from the suppression of key pancreatic transcription factors. PGC-1α is an attractive target for modulating the deleterious effects of glucocorticoids on pancreatic stem cells.


Assuntos
Glucocorticoides/metabolismo , Proteínas de Homeodomínio/metabolismo , Células Secretoras de Insulina/citologia , Transplante de Pâncreas , Pâncreas/citologia , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Núcleo Celular/metabolismo , Transdiferenciação Celular , Células Cultivadas , Dexametasona/química , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pâncreas/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Células-Tronco/citologia , Suínos
12.
PLoS One ; 8(11): e79076, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24260156

RESUMO

Surrogate ß-cells derived from stem cells are needed to cure type 1 diabetes, and neonatal liver cells may be an attractive alternative to stem cells for the generation of ß-cells. In this study, we attempted to generate insulin-producing cells from neonatal porcine liver-derived cells using adenoviruses carrying three genes: pancreatic and duodenal homeobox factor1 (PDX1)/VP16, BETA2/NeuroD and v-maf musculo aponeurotic fibrosarcoma oncogene homolog A (MafA), which are all known to play critical roles in pancreatic development. Isolated neonatal porcine liver-derived cells were sequentially transduced with triple adenoviruses and grown in induction medium containing a high concentration of glucose, epidermal growth factors, nicotinamide and a low concentration of serum following the induction of aggregation for further maturation. We noted that the cells displayed a number of molecular characteristics of pancreatic ß-cells, including expressing several transcription factors necessary for ß-cell development and function. In addition, these cells synthesized and physiologically secreted insulin. Transplanting these differentiated cells into streptozotocin-induced immunodeficient diabetic mice led to the reversal of hyperglycemia, and more than 18% of the cells in the grafts expressed insulin at 6 weeks after transplantation. These data suggested that neonatal porcine liver-derived cells can be differentiated into functional insulin-producing cells under the culture conditions presented in this report and indicated that neonatal porcine liver-derived cells (NPLCs) might be useful as a potential source of cells for ß-cell replacement therapy in efforts to cure type I diabetes.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/transplante , Insulina/metabolismo , Fatores de Transcrição Maf Maior/biossíntese , Transativadores/biossíntese , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/terapia , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Glucose/genética , Glucose/metabolismo , Xenoenxertos , Hiperglicemia/genética , Hiperglicemia/metabolismo , Hiperglicemia/terapia , Insulina/genética , Secreção de Insulina , Fígado , Fatores de Transcrição Maf Maior/genética , Camundongos , Suínos , Transativadores/genética
13.
BMB Rep ; 44(11): 713-8, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22118536

RESUMO

In this study, the effects of sitagliptin analogue (SITA) or pioglitazone (PIO) treatment on glucose homeostasis and Β-cell dynamics in animal models of type 2 diabetes--Akita and db/db mice were evaluated. After 4-6 weeks of treatment, both SITA and PIO were shown to lower non-fasting glucose levels and reduced glycemic excursion in the intraperitoneal glucose tolerance test. In addition, both drugs preserved normal islet structure and the proportion of Β-cells in the islets. Compared to the controls, SITA treatment induced a higher Β-cell proliferation rate in Akita mice and a lower rate of apoptosis in db/db mice, whereas PIO treatment induced a lower rate of apoptosis in db/db mice and reduced proliferation rates in Akita mice. In conclusion, both SITA and PIO appear to exert some beneficial effects on the islet structure in addition to glycemic control via different mechanisms that involve Β-cell dynamics in Akita and db/db mice. [BMB reports 2011; 44(11): 713-718].


Assuntos
Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Células Secretoras de Insulina/patologia , Obesidade/complicações , Obesidade/tratamento farmacológico , Pirazinas/uso terapêutico , Tiazolidinedionas/uso terapêutico , Triazóis/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Glicemia/metabolismo , Peso Corporal/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Diabetes Mellitus Experimental/sangue , Jejum/sangue , Teste de Tolerância a Glucose , Homeostase/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Antígeno Ki-67/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/sangue , Tamanho do Órgão/efeitos dos fármacos , Pioglitazona , Pirazinas/administração & dosagem , Pirazinas/farmacologia , Fosfato de Sitagliptina , Tiazolidinedionas/administração & dosagem , Tiazolidinedionas/farmacologia , Triazóis/administração & dosagem , Triazóis/farmacologia
14.
Diabetes Metab J ; 35(2): 119-29, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21738894

RESUMO

BACKGROUND: A limitation in the number of insulin-producing pancreatic beta-cells is a special feature of diabetes. The identification of alternative sources for the induction of insulin-producing surrogate beta-cells is a matter of profound importance. PDX-1/VP16, BETA2/NeuroD, and MafA overexpression have been shown to influence the differentiation and proliferation of pancreatic stem cells. However, few studies have been conducted using adult animal pancreatic stem cells. METHODS: Adult pig pancreatic cells were prepared from the non-endocrine fraction of adult pig pancreata. Porcine neonatal pancreas cell clusters (NPCCs) were prepared from neonatal pigs aged 1-2 days. The dispersed pancreatic cells were infected with PDX-1/VP16, BETA2/NeuroD, and MafA adenoviruses. After infection, these cells were transplanted under the kidney capsules of normoglycemic nude mice. RESULTS: The adenovirus-mediated overexpression of PDX-1, BETA2/NeuroD and MafA induced insulin gene expression in NPCCs, but not in adult pig pancreatic cells. Immunocytochemistry revealed that the number of insulin-positive cells in NPCCs and adult pig pancreatic cells was approximately 2.6- and 1.1-fold greater than those in the green fluorescent protein control group, respectively. At four weeks after transplantation, the relative volume of insulin-positive cells in the grafts increased in the NPCCs, but not in the adult porcine pancreatic cells. CONCLUSION: These data indicate that PDX-1, BETA2/NeuroD, and MafA facilitate the beta-cell differentiation of NPCCs, but not adult pig pancreatic cells. Therefore PDX-1, BETA2/NeuroD, and MafA-induced NPCCs can be considered good sources for the induction of pancreatic beta-cells, and may also have some utility in the treatment of diabetes.

15.
Lab Chip ; 11(6): 1049-53, 2011 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-21283907

RESUMO

Nanochannel membranes have been fabricated for many biological and engineering applications. However, due to low-throughput process, high cost, unsuitable pore geometries, and low chemical/mechanical stability, we could not have obtained optimized nanochannel membranes for biomedical treatments as well as a novel building block for artificial cell membranes. Here, we report a PEO-functionalized straight nanochannel array based on a self-organized porous alumina for a novel biofilter with antifouling, superior immunoprotection and high permeability of nutrients, which have excellent in vivo mechanical stability. Thus, our strategy may provide great advantages in novel membrane biotechnologies such as biofiltration, artificial cells, and drug delivery.


Assuntos
Óxido de Alumínio/química , Nanofibras/química , Polietilenoglicóis/química , Difusão , Humanos , Imunoglobulina G/química , Nanotecnologia
16.
Transplantation ; 90(7): 717-24, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20622751

RESUMO

BACKGROUND: The role of rapamycin in pancreas stem cells remains to be clearly elucidated. Herein, we evaluated the effects of rapamycin on porcine neonatal pancreas cell clusters (NPCCs), which primarily comprised pancreatic precursors, and attempted to find an intracellular mechanism about the harmful effects of rapamycin. METHODS: Porcine NPCCs were treated with rapamycin in a monolayer, and the apoptosis and proliferation were determined via caspase-3 assay and H-thymidine uptake analysis. The expression of transcription factors was assessed via reverse-transcriptase polymerase chain reaction and Western blotting. For the in vivo study, the porcine NPCCs were transplanted into the kidney subcapsules of normal nude mice and treated with rapamycin. RESULTS: Rapamycin treatment significantly reduced the number of ß cells, glucose-stimulated insulin secretion, and the insulin contents in the monolayer-cultured porcine NPCCs. Furthermore, rapamycin treatment increased the apoptosis and inhibited the proliferation of ß cells in the culture dishes. The expressions of the insulin, pancreatic and duodenal homeobox-1, and NeuroD/Beta2 genes were down-regulated via rapamycin treatment. The expression of insulin-like growth factor-II was significantly down-regulated, but the expression of Foxo1 was simultaneously inversely increased, and the translocation of Foxo1 from the cytoplasm to the nucleus was induced by rapamycin treatment. Moreover, rapamycin treatment induced a marked reduction in the relative volume and absolute mass of ß cells in the porcine NPCCs grafts at 8 weeks after transplantation in the normal nude mice. CONCLUSIONS: Here, we demonstrate that rapamycin treatment suppresses the expansion and differentiation of porcine NPCCs, and the alteration of Foxo1 and insulin-like growth factor-II gene expression might be the crucial factors.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Pâncreas/citologia , Sirolimo/farmacologia , Animais , Animais Recém-Nascidos , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Primers do DNA , Replicação do DNA/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Insulina/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pâncreas/efeitos dos fármacos , Pâncreas/fisiologia , Pancreatectomia , Reação em Cadeia da Polimerase/efeitos dos fármacos , Suínos
17.
Biochem Biophys Res Commun ; 390(3): 763-8, 2009 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-19836348

RESUMO

The pathogenesis of post-transplant diabetes mellitus (PTDM) is thought to be partly related to the direct toxic effect of cyclosporine (CsA) on pancreatic beta-cells and the resultant decrease in insulin synthesis and secretion. Although rosiglitazone (Rosi) is an insulin sensitizer, recent data has shown that Rosi also directly protects against beta-cell dysfunction and death. This study was undertaken to clarify the effects of Rosi on CsA-induced beta-cell dysfunction and death. The deterioration in glucose tolerance caused by CsA administration was significantly improved by cotreatment with Rosi. The relative volume and absolute mass of beta-cells were significantly reduced by CsA, whereas combined treatment with Rosi had protective effects. Induction of beta-cell death and increased expression of endoplasmic reticulum (ER) stress markers (CHOP and spliced XBP-1) by CsA were rescued by Rosi. Thus, Rosi signaling directly modulates the ER stress response, promoting beta-cell adaptation and survival. Rosi might be an appropriate drug for preventing and treating CsA-induced PTDM.


Assuntos
Ciclosporina/antagonistas & inibidores , Citoproteção , Diabetes Mellitus/prevenção & controle , Hipoglicemiantes/farmacologia , Imunossupressores/antagonistas & inibidores , Células Secretoras de Insulina/efeitos dos fármacos , Animais , Ciclosporina/efeitos adversos , Ciclosporina/toxicidade , Diabetes Mellitus/induzido quimicamente , Teste de Tolerância a Glucose , Imunossupressores/efeitos adversos , Imunossupressores/toxicidade , Insulina/sangue , Células Secretoras de Insulina/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Rosiglitazona , Tiazolidinedionas , Transplante/efeitos adversos
18.
Biochem Biophys Res Commun ; 365(4): 614-20, 2008 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-18035054

RESUMO

It is well known that the activation of AMP-activated protein kinase (AMPK) represses insulin gene expression and glucose-stimulated insulin secretion. However, how this effect is achieved and the effects of AMPK activation on glucolipotoxicity-induced beta-cell dysfunction have not been elucidated. We investigate whether BETA2 gene expression are involved in the AMPK-mediated regulation of insulin gene expression in normal and dysfunctional beta-cells. BETA2 gene expression and protein levels were significantly decreased by AICAR treatment and those were associated with the suppression of BETA2 promoter activity and DNA binding activity. These results demonstrate that the expressions of BETA2 and insulin gene are positively regulated by glucose and negatively by AMPK. Therefore, AMPK may function as a key molecule, which conveys extracellular metabolic signals into the cells and finely tunes expression of beta-cell specific transcription factors in response to glucose level.


Assuntos
Aminoimidazol Carboxamida/análogos & derivados , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas Quinases/efeitos dos fármacos , Proteínas Quinases/metabolismo , Ribonucleotídeos/administração & dosagem , Transdução de Sinais/fisiologia , Transcrição Gênica/fisiologia , Quinases Proteína-Quinases Ativadas por AMP , Aminoimidazol Carboxamida/administração & dosagem , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Ativação Enzimática/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Ratos , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA