Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 300
Filtrar
1.
Biomater Sci ; 12(4): 821-836, 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38168805

RESUMO

Islet transplantation holds significant promise as a curative approach for type 1 diabetes (T1D). However, the transition of islet transplantation from the experimental phase to widespread clinical implementation has not occurred yet. One major hurdle in this field is the challenge of insufficient vascularization and subsequent early loss of transplanted islets, especially in non-intraportal transplantation sites. The establishment of a fully functional vascular system following transplantation is crucial for the survival and secretion function of islet grafts. This vascular network not only ensures the delivery of oxygen and nutrients, but also plays a critical role in insulin release and the timely removal of metabolic waste from the grafts. This review summarizes recent advances in effective strategies to improve graft revascularization and enhance islet survival. These advancements include the local release and regulation of angiogenic factors (e.g., vascular endothelial growth factor, VEGF), co-transplantation of vascular fragments, and pre-vascularization of the graft site. These innovative approaches pave the way for the development of effective islet transplantation therapies for individuals with T1D.


Assuntos
Diabetes Mellitus Tipo 1 , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Humanos , Ilhotas Pancreáticas/metabolismo , Diabetes Mellitus Tipo 1/cirurgia , Materiais Biocompatíveis , Fator A de Crescimento do Endotélio Vascular/metabolismo , Transplante das Ilhotas Pancreáticas/fisiologia , Neovascularização Fisiológica
2.
Stem Cell Rev Rep ; 20(3): 839-844, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38153636

RESUMO

BACKGROUND: Insular allograft for unstable type 1 diabetes and autograft in pancreatectomy patients are nowadays considered established procedures with precise indications and predictable outcomes. The clinical outcome of islet transplantation is similar to that of pancreas transplantation, avoiding the complications associated with organ transplantation. OBJECTIVE: We hypothesised that transplantation of islets of Langerhans within an endocrine organ could better promote their engraftment and function. This could help to resolve or ameliorate known pathological conditions such as unstable type 1 diabetes and complicated type 2 diabetes. RATIONALE: Pancreatic islet transplantation is currently performed almost exclusively in the liver. The liver provides a sufficiently favourable environment, although not entirely. The hepatic parenchyma has a lower oxygen tension than the pancreatic parenchyma and the vascular structure of the liver is not typical of an exclusively endocrine organ. Moreover, islet transplantation into the liver is not without complications, including hematoma or portal vein thrombosis. PROPOSED PROJECT: The thyroid gland is the endocrine gland proposed as a 'container'. In fact, it has all the characteristics of 'physio-compatibility' which can address the objectives assumed. It is indeed an ideal site because it is an easily accessible anatomical site that allows islets to be implanted using ultrasound-guided transcutaneous inoculation technique. Moreover, it has physiological and anatomical endocrine affinities with pancreatic islets and, if necessary, it can be removed, using hormone supplementation or replacement therapy. CONCLUSIONS: The thyroid gland may be proposed as an ideal site for islet implantation due to its anatomical and physiocompatibility characteristics.


Assuntos
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Transplante de Pâncreas , Humanos , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/cirurgia , Glândula Tireoide , Pancreatectomia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/cirurgia , Ilhotas Pancreáticas/cirurgia , Transplante das Ilhotas Pancreáticas/métodos , Transplante das Ilhotas Pancreáticas/fisiologia
3.
Tissue Cell ; 79: 101919, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36137362

RESUMO

Type 1 diabetes is an autoimmune disease that emerges with the destruction of beta cells of pancreatic Langerhans islets. Three different therapeutical approaches have been developed so far; pancreas transplantation, islet transplantation, and cell-based therapies. Bioengineering cell sheets for tissue generating is one of the latest approaches that have been used to construct cell-sheets instead of single cells so that it mimics the in vivo environments more. In this study, extra-hepatic functional islet tissue was constructed by transferring the 3-D beta cells and GFP labelled MSCs MSC sheets to the subcutaneous site of rats with STZ-induced diabetes. rBM-MSCs and beta cells were cultured on the 6-well PIPAAm culture dishes. Obtained rBM-MSCs as two-cell sheets and beta cells cultured in droplets with matrigel has transplanted into the dorsal subcutaneous area of diabetic rats. Fasting blood glucose levels and body weights were evaluated for 30 days after transplantation. Immunocytochemistry analysis for the anti-apoptotic, anti-inflammatory, and angiogenetic effects of MSCs on the 30th day of subcutaneous cell transplantation. All recipient rats transplanted with beta-cells with MSCs returned toward normoglycemia by day 5 and remained at this level for 30 days. Immunocytochemical analyses supported that the MSCs and beta cells preserved their viability and function. MSCs secrete cytokines and growth factors TGF-ß and IL-6; MSCs of the important features of the anti-apoptotic and anti-inflammatory properties, thanks to apoptosis of beta cells preserve graft explained by inhibition. In transplantation of MSCs induced angiogenesis and neovascularization, PECAM-1 and GFP positive simultaneously determining endothelial cells was observed indicating. Subcutaneous 3D beta-cell transplantation would be possible with the MSC-sheets as a feeder layer of beta cells. The beta-cell line is glucose-sensitive and has a high insulin release potential, and can be used as an alternative to islets in in vivo transplant studies.


Assuntos
Diabetes Mellitus Experimental , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Ratos , Animais , Células Endoteliais/metabolismo , Células Secretoras de Somatostatina/metabolismo , Transplante das Ilhotas Pancreáticas/fisiologia , Bioengenharia , Insulina/metabolismo
4.
Nat Med ; 28(2): 272-282, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35115708

RESUMO

Human pluripotent stem-cell-derived islets (hPSC-islets) are a promising cell resource for diabetes treatment1,2. However, this therapeutic strategy has not been systematically assessed in large animal models physiologically similar to humans, such as non-human primates3. In this study, we generated islets from human chemically induced pluripotent stem cells (hCiPSC-islets) and show that a one-dose intraportal infusion of hCiPSC-islets into diabetic non-human primates effectively restored endogenous insulin secretion and improved glycemic control. Fasting and average pre-prandial blood glucose levels significantly decreased in all recipients, accompanied by meal or glucose-responsive C-peptide release and overall increase in body weight. Notably, in the four long-term follow-up macaques, average hemoglobin A1c dropped by over 2% compared with peak values, whereas the average exogenous insulin requirement reduced by 49% 15 weeks after transplantation. Collectively, our findings show the feasibility of hPSC-islets for diabetic treatment in a preclinical context, marking a substantial step forward in clinical translation of hPSC-islets.


Assuntos
Diabetes Mellitus Experimental , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Animais , Glicemia , Diabetes Mellitus Experimental/terapia , Humanos , Insulina , Transplante das Ilhotas Pancreáticas/fisiologia , Primatas
5.
Methods Mol Biol ; 2224: 87-98, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33606208

RESUMO

Type 1 diabetes (T1D) is an autoimmune disease, where insulin-producing ß-cells in the pancreas are inappropriately recognized and destroyed by immune cells. Islet transplantation is the most successful cell-based therapy for T1D individuals who experience frequent and severe life-threatening hypoglycemia. However, this therapy is extremely restricted owing to the limited availability of donor pancreas. In recent years, significant progress has been made in generating ß-cells from stem/progenitor cells using different approaches of in vitro differentiation. The insulin production from such in vitro generated ß-cells is still far less than that observed in islet ß-cells. We employed a novel strategy to improve the efficiency of progenitor cell differentiation by performing partial mouse pancreas resection after transplanting in vitro generated insulin-producing cells under the kidney capsule of these mice. Pancreas resection (pancreatectomy) has been shown to induce regenerative pathways, leading to regeneration of almost the entire resected pancreas over 3-5 weeks in mice. We found that in our method, regenerating mouse pancreas promotes better graft differentiation/maturation and insulin production from transplanted cells. In this chapter, we detail the protocols used for transplantation of in vitro differentiated cells in immunocompromised mice, partial pancreatectomy in host (NOD scid) mice, and assessment of graft function. We believe that our protocols provide a solid platform for further studies aimed at understanding growth/differentiation molecules secreted from regenerating pancreas that promote graft maturation.


Assuntos
Diferenciação Celular/fisiologia , Pâncreas/fisiologia , Animais , Diabetes Mellitus Tipo 1/fisiopatologia , Células Secretoras de Insulina/fisiologia , Transplante das Ilhotas Pancreáticas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pancreatectomia/métodos , Células-Tronco/fisiologia
6.
Curr Stem Cell Res Ther ; 14(1): 52-56, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30207243

RESUMO

In the field of stem cell therapy, research on the application of mesenchymal stem cells (MSCs) has flourished because of the various functions. On the other hand, research on the method of cell transplantation has developed from the administration of cell suspensions to cell-sheet engineering and 3D technology. In the trend, a cell transplantation platform named CellSaic, which is a combination of xeno-free recombinant scaffolds in a cell aggregate-like shape, was developed. CellSaic is the cell transplantation platform that can prevent the central necrosis within cell aggregates by arranging the cells and petaloid pieces of recombinant peptide (RCP) in a mosaic. The prevention of central necrosis is the most significant advantage over other 3D culture systems. This review details the unique characteristics of CellSaic including safety examination results and describes its future application for MSC transplantation. Particularly, in the application of MSCs, it has been reported that the MSC CellSaics increased the effect on improving various symptoms compared with MSCs only in the application of the therapy to inflammatory bowel disease (IBD), cerebral infarction, bone cartilage regeneration in joints, and islet transplantation. In accordance with the "One Health" concept, it is anticipated that this technology is expected to contribute to companion animal therapy and human therapy in the future.


Assuntos
Agregação Celular/fisiologia , Transplante de Células-Tronco Mesenquimais/métodos , Fragmentos de Peptídeos/fisiologia , Engenharia Tecidual , Animais , Cartilagem/crescimento & desenvolvimento , Adesão Celular/fisiologia , Infarto Cerebral/terapia , Colite/terapia , Citocinas/metabolismo , Humanos , Doenças Inflamatórias Intestinais/terapia , Transplante das Ilhotas Pancreáticas/fisiologia , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Camundongos , Necrose/fisiopatologia , Neoplasias Pancreáticas/terapia , Transplantes/fisiologia
7.
Stem Cells Transl Med ; 8(3): 209-214, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30417988

RESUMO

We examined data and patterns in clinical islet transplant studies registered on ClinicalTrials.gov (CTgov) for treatment of type 1 diabetes (T1D), with a goal of extracting insights to apply in the design of a pluripotent stem cell-derived islet therapy. Clinical islet transplantation, as a cell therapy (rather than solid organ transplant) is a unique precedent for stem cell-based islet therapies. Registration activity shows that the field is not growing significantly, and newer registrations suggest that the reasons for stagnation include need for a more optimal site of infusion/transplantation, and especially a need for better immune protective strategies to advance a more effective and durable therapy for T1D. Stem Cells Translational Medicine 2019;8:209&214.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Transplante das Ilhotas Pancreáticas/fisiologia , Ilhotas Pancreáticas/citologia , Células-Tronco/citologia , Transplantes/citologia , Diferenciação Celular/fisiologia , Ensaios Clínicos como Assunto , Humanos
8.
Med Sci (Paris) ; 32(4): 401-7, 2016 Apr.
Artigo em Francês | MEDLINE | ID: mdl-27137698

RESUMO

Islet transplantation has set the ground for diabetes cell therapy and is still undergoing various developments that might improve clinical outcomes. Alternative sources for ß-cell replacement strategies are now led by human pluripotent stem cells that demonstrate near-normal ß-cell features after in vitro differentiation and which can reverse diabetes in mice. Yet, their propensity for tumor formation is still unresolved. The adult pancreas is suggested as a reservoir of facultative progenitors that could represent adequate candidates for ß-cell engineering, either in vivo through pharmacological treatment or after expansion in culture. This review focuses on the latest developments in protocols aiming at de novo production of functional ß cells.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/tendências , Diabetes Mellitus/terapia , Adulto , Animais , Diferenciação Celular/fisiologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Humanos , Células Secretoras de Insulina/transplante , Transplante das Ilhotas Pancreáticas/métodos , Transplante das Ilhotas Pancreáticas/fisiologia , Camundongos , Pâncreas/citologia , Pâncreas/fisiologia , Células-Tronco Pluripotentes/fisiologia , Células-Tronco Pluripotentes/transplante , Regeneração
9.
Diabetes ; 65(5): 1208-18, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26936960

RESUMO

Diabetes is associated with loss of functional pancreatic ß-cells, and restoration of ß-cells is a major goal for regenerative therapies. Endogenous regeneration of ß-cells via ß-cell replication has the potential to restore cellular mass; however, pharmacological agents that promote regeneration or expansion of endogenous ß-cells have been elusive. The regenerative capacity of ß-cells declines rapidly with age, due to accumulation of p16(INK4a), resulting in limited capacity for adult endocrine pancreas regeneration. Here, we show that transforming growth factor-ß (TGF-ß) signaling via Smad3 integrates with the trithorax complex to activate and maintain Ink4a expression to prevent ß-cell replication. Importantly, inhibition of TGF-ß signaling can result in repression of the Ink4a/Arf locus, resulting in increased ß-cell replication in adult mice. Furthermore, small molecule inhibitors of the TGF-ß pathway promote ß-cell replication in human islets transplanted into NOD-scid IL-2Rg(null) mice. These data reveal a novel role for TGF-ß signaling in the regulation of the Ink4a/Arf locus and highlight the potential of using small molecule inhibitors of TGF-ß signaling to promote human ß-cell replication.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/antagonistas & inibidores , Células Secretoras de Insulina/efeitos dos fármacos , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Animais , Benzamidas/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/agonistas , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Dioxóis/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Transplante das Ilhotas Pancreáticas/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Receptores de Fatores de Crescimento Transformadores beta/agonistas , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Regeneração/efeitos dos fármacos , Bancos de Tecidos , Fator de Crescimento Transformador beta1/metabolismo , Transplante Heterólogo , Transplante Heterotópico
10.
Diabetes ; 65(2): 451-62, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26581595

RESUMO

Clinical islet transplantation has become an established treatment modality for selected patients with type 1 diabetes. However, a large proportion of transplanted islets is lost through multiple factors, including immunosuppressant-related toxicity, often requiring more than one donor to achieve insulin independence. On the basis of the cytoprotective capabilities of antifreeze proteins (AFPs), we hypothesized that supplementation of islets with synthetic AFP analog antiaging glycopeptide (AAGP) would enhance posttransplant engraftment and function and protect against tacrolimus (Tac) toxicity. In vitro and in vivo islet Tac exposure elicited significant but reversible reduction in insulin secretion in both mouse and human islets. Supplementation with AAGP resulted in improvement of islet survival (Tac(+) vs. Tac+AAGP, 31.5% vs. 67.6%, P < 0.01) coupled with better insulin secretion (area under the curve: Tac(+) vs. Tac+AAGP, 7.3 vs. 129.2 mmol/L/60 min, P < 0.001). The addition of AAGP reduced oxidative stress, enhanced insulin exocytosis, improved apoptosis, and improved engraftment in mice by decreasing expression of interleukin (IL)-1ß, IL-6, keratinocyte chemokine, and tumor necrosis factor-α. Finally, transplant efficacy was superior in the Tac+AAGP group and was similar to islets not exposed to Tac, despite receiving continuous treatment for a limited time. Thus, supplementation with AAGP during culture improves islet potency and attenuates long-term Tac-induced graft dysfunction.


Assuntos
Proteínas Anticongelantes/farmacologia , Imunossupressores/toxicidade , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/efeitos dos fármacos , Tacrolimo/toxicidade , Animais , Apoptose/efeitos dos fármacos , Exocitose , Sobrevivência de Enxerto/efeitos dos fármacos , Humanos , Insulina/metabolismo , Secreção de Insulina , Interleucinas/metabolismo , Ilhotas Pancreáticas/lesões , Ilhotas Pancreáticas/fisiologia , Transplante das Ilhotas Pancreáticas/fisiologia , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/farmacologia
11.
Endocr Pathol ; 26(2): 95-103, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25762503

RESUMO

Diabetes mellitus type 1 is a form of diabetes mellitus that results from the autoimmune destruction of insulin-producing beta cells in the pancreas. The current gold standard therapy for pancreas transplantation has limitations because of the long list of waiting patients and the limited supply of donor pancreas. Mesenchymal stem cells (MSCs), a relatively new potential therapy in various fields, have already made their mark in the young field of regenerative medicine. Recent studies have shown that the implantation of MSCs decreases glucose levels through paracrine influences rather than through direct transdifferentiation into insulin-producing cells. Therefore, these cells may use pro-angiogenic and immunomodulatory effects to control diabetes following the cotransplantation with pancreatic islets. In this review, we present and discuss new approaches of using MSCs in the treatment of diabetes mellitus type 1.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Animais , Diferenciação Celular , Humanos , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/fisiologia , Transplante das Ilhotas Pancreáticas/efeitos adversos , Transplante das Ilhotas Pancreáticas/fisiologia , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia
12.
Transplantation ; 99(6): 1132-43, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25806408

RESUMO

BACKGROUND: Nerve growth factor (NGF), which plays important roles in promoting growth and differentiation of nerve cells, has recently been reported as a regulator in pancreatic ß cells in terms of insulin releasing function. In this study, we examined whether NGF stimulation would promote islet graft survival and function in islet transplantation. METHODS: We found that supplementation of cultured islets with NGF improved the viability of islet cells and induced the production of insulin, vascular endothelial growth factor, and cellular proliferative markers. Because a specific inhibitor of TrkA, K252a, blocked all these effects, we propose that the TrkA receptor is the mediator of NGF stimulation. RESULTS: After transplantation to the kidney subcapsule and liver of syngenic diabetic mice, a higher rate of normoglycemic achievement, increased serum insulin, and improved glucose tolerance were observed in the mice transplanted with NGF-pretreated islet grafts. Histological analysis revealed higher expression of insulin and vascular endothelial growth factor, an increase in proliferative ß cells, and revascularization in NGF-pretreated islet grafts without activation of any inflammatory cells. CONCLUSIONS: The NGF treatment can therefore serve as a new and promising therapeutic tool for improving islet graft viability and function in islet transplantation.


Assuntos
Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Transplante das Ilhotas Pancreáticas/métodos , Fator de Crescimento Neural/farmacologia , Receptor trkA/metabolismo , Animais , Glicemia/metabolismo , Carbazóis/farmacologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citocinas/biossíntese , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Experimental/terapia , Sobrevivência de Enxerto/efeitos dos fármacos , Alcaloides Indólicos/farmacologia , Mediadores da Inflamação/metabolismo , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/fisiologia , Transplante das Ilhotas Pancreáticas/patologia , Transplante das Ilhotas Pancreáticas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Receptor trkA/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
Endocr J ; 60(6): 697-708, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23719783

RESUMO

Experimental transplantation of endocrine tissues has led to significant advances in our understanding of endocrinology and metabolism. Endocrine cell transplantation therapy is expected to be applied to the treatment of metabolic endocriopathies. Restoration of functional pancreatic beta-cell mass or of functional adipose mass are reasonable treatment approaches for patients with diabetes or lipodystrophy, respectively. Human induced pluripotent stem (iPS) cell research is having a great impact on life sciences. Doctors Takahashi and Yamanaka discovered that the forced expression of a set of genes can convert mouse and human somatic cells into a pluripotent state [1, 2]. These iPS cells can differentiate into a variety of cell types. Therefore, iPS cells from patients may be a potential cell source for autologous cell replacement therapy. This review briefly summarizes the current knowledge about transplantation therapy for diabetes mellitus, the development of the endocrine pancreas and adipocytes, and endocrine-metabolic disease-specific iPS cells.


Assuntos
Adipócitos/fisiologia , Diabetes Mellitus/terapia , Células-Tronco Pluripotentes Induzidas/transplante , Células Secretoras de Insulina/transplante , Ilhotas Pancreáticas/fisiologia , Adipócitos/transplante , Animais , Humanos , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/embriologia , Transplante das Ilhotas Pancreáticas/fisiologia , Camundongos
14.
Transplantation ; 95(11): 1396-403, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23558506

RESUMO

BACKGROUND: Several retrospective studies with short-term follow-up have demonstrated a low rate of new-onset diabetes after distal pancreatectomy for benign pancreatic tumors. We sought to determine the long-term diabetes-free survival of patients who underwent islet autotransplantation (IAT) after distal pancreatectomy and to identify any associations between the isolation parameters of autologous islets and diabetes-free survival. METHODS: Among the 37 nondiabetic patients who underwent 50% to 60% partial pancreatectomy, 20 underwent IAT (IAT group; median follow-up period, 61 months). In the IAT group, diabetes-free survival was determined based on annual oral glucose tolerance tests, fasting blood glucose, and hemoglobin A1C. RESULTS: The 7-year diabetes-free survival rate was 51% in the IAT group (median follow-up period, 61 months) and 45% in the 37 study subjects. Diabetes-free survival was significantly prolonged when islet yield per gram of pancreas weight was more than 5154 islet equivalents (IEQ)/g, even in patients with prediabetes and high insulin resistance who had a markedly high rate of diabetes development. The proportion of patients with impaired glucose tolerance at 2 years after distal pancreatectomy was 12 of 16 in the control group, 6 of 7 in patients with islet yields of less than 5154 IEQ/g, and 3 of 11 in patients with islet yields of more than 5154 IEQ/g (P=0.019). CONCLUSIONS: Partial (50%-60%) pancreatectomy for benign pancreatic tumors had a major metabolic consequence, especially in patients with prediabetes and high insulin resistance. In this setting, prolonged diabetes-free survival was observed in patients who underwent IAT when a high islet yield per gram of pancreas was achieved.


Assuntos
Diabetes Mellitus/epidemiologia , Transplante das Ilhotas Pancreáticas/fisiologia , Neoplasias/mortalidade , Neoplasias/cirurgia , Pancreatectomia/métodos , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/cirurgia , Adulto , Idoso , Feminino , Seguimentos , Humanos , Incidência , Resistência à Insulina/fisiologia , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Pâncreas/patologia , Pâncreas/cirurgia , Estado Pré-Diabético/fisiopatologia , Taxa de Sobrevida , Transplante Autólogo , Resultado do Tratamento
15.
Pancreas ; 42(6): 971-6, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23587851

RESUMO

OBJECTIVE: Survival of transplanted islets is limited partly because of the disruption of the islet basement membrane (BM) occurring during isolation. We hypothesized that the embedment of BM extract (BME) could induce a viable cell mass and prolong islet functionality before transplantation. METHODS: A special reconstituted BME that solidifies into a gel at 37°C was used to embed isolated islets in this study. The strategy was used to re-establish the interaction between the islets and peri-islet BM. RESULTS: Islets embedded in BME showed lower caspase-3 levels and higher Akt activity than those in suspension. Moreover, we found for the first time that the expression of α3 integrin and focal adhesion kinase (FAK) and FAK activity was up-regulated in islets after BME embedment. The reverse effect was observed on islet apoptosis when islets rescued from a 24-hour suspension culture were embedded in BME for the next 24 hours. In addition, expression of pancreatic duodenal homeobox factor-1 and phospho-extracellular signal-regulated kinase 1/2 was partially preserved, suggesting the positive effect of BME on islet development. CONCLUSIONS: These results indicate that BME embedment of islets can up-regulate the expression of α3 integrin and its signal transduction, which may improve islet viability.


Assuntos
Membrana Basal/metabolismo , Integrina alfa3/metabolismo , Ilhotas Pancreáticas/metabolismo , Transdução de Sinais/fisiologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Feminino , Sobrevivência de Enxerto/efeitos dos fármacos , Sobrevivência de Enxerto/fisiologia , Proteínas de Homeodomínio/metabolismo , Ilhotas Pancreáticas/citologia , Transplante das Ilhotas Pancreáticas/fisiologia , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Extratos de Tecidos/farmacologia , Transativadores/metabolismo , Regulação para Cima/efeitos dos fármacos
16.
Apoptosis ; 18(6): 681-8, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23536200

RESUMO

Human islet isolation is associated with adverse conditions inducing apoptosis and necrosis. The aim of the present study was to assess whether antiapoptotic preconditioning can improve in vitro and posttransplant function of isolated human islets. A dose-finding study demonstrated that 200 µmol/L of the caspase-3 inhibitor Ac-DEVD-CMK was most efficient to reduce the expression of activated caspase-3 in isolated human islets exposed to severe heat shock. Ac-DEVD-CMK-pretreated or sham-treated islets were transplanted into immunocompetent or immunodeficient diabetic mice and subjected to static glucose incubation to measure insulin and proinsulin secretion. Antiapoptotic pretreatment significantly deteriorated graft function resulting in elevated nonfasting serum glucose when compared to sham-treated islets transplanted into diabetic nude mice (p < 0.01) and into immunocompetent mice (p < 0.05). Ac-DEVD-CMK pretreatment did not significantly change basal and glucose-stimulated insulin release compared to sham-treated human islets but increased the proinsulin release at high glucose concentrations (20 mM) thus reducing the insulin-to-proinsulin ratio in preconditioned islets (p < 0.05). This study demonstrates that the caspase-3 inhibitor Ac-DEVD-CMK interferes with proinsulin conversion in preconditioned islets reducing their potency to cure diabetic mice. The mechanism behind this phenomenon is unclear so far but may be related to the ketone CMK linked to the Ac-DEVD molecule. Further studies are required to identify biocompatible caspase inhibitors suitable for islet preconditioning.


Assuntos
Inibidores de Caspase/farmacologia , Insulina/metabolismo , Transplante das Ilhotas Pancreáticas/fisiologia , Ilhotas Pancreáticas/fisiologia , Proinsulina/metabolismo , Condicionamento Pré-Transplante/métodos , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Caspase 3 , Diabetes Mellitus Experimental/cirurgia , Glucose/farmacologia , Proteínas de Choque Térmico/biossíntese , Temperatura Alta/efeitos adversos , Humanos , Secreção de Insulina , Camundongos , Camundongos Nus , Obtenção de Tecidos e Órgãos/métodos , Transplante Heterólogo/fisiologia
17.
Proc Natl Acad Sci U S A ; 110(6): 2288-93, 2013 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-23345449

RESUMO

Here, we evaluate an alternative approach of preconditioning pancreatic islets before transplantation using a potent agonist of growth-hormone-releasing hormone (GHRH) to promote islet viability and function, and we explore the adrenal gland as an alternative transplantation site for islet engraftment. The endocrine microenvironment of the adrenal represents a promising niche with the unique advantages of exceptional high oxygen tension and local anti-inflammatory and immunosuppressive properties. GHRH agonists have been shown to promote islet graft survival and function, which may help to reduce the islet mass necessary to reverse diabetes. In the present study, the most potent GHRH agonist MR403 was tested on insulinoma cells, isolated rat islets, and adrenal ß-cell cocultures in vitro. GHRH receptor is expressed on both adrenal cells and islets. MR403 caused a significant increase in cell viability and proliferation and revealed an antiapoptotic effect on insulinoma cells. Viability of rat islets was increased after treatment with the agonist and in coculture with adrenal cells. Rat islets were transplanted into diabetic mice to the intraadrenal transplant site and compared with the classical transplants underneath the kidney capsule. Graft function and integration were tested by metabolic follow-up and immunohistochemical staining of intraadrenal grafts. A rapid decrease occurred in blood glucose levels in both models, and all animals reached normoglycemia within the first days after transplantation. Our studies demonstrated that the adrenal may be an attractive site for islet transplantation and that GHRH analogs might allow reduction of the islet mass needed to reverse a diabetic status.


Assuntos
Hormônio Liberador de Gonadotropina/agonistas , Transplante das Ilhotas Pancreáticas/métodos , Condicionamento Pré-Transplante/métodos , Glândulas Suprarrenais/fisiologia , Glândulas Suprarrenais/cirurgia , Animais , Linhagem Celular , Técnicas de Cocultura , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Experimental/cirurgia , Feminino , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/fisiologia , Transplante das Ilhotas Pancreáticas/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores de Neuropeptídeos/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética
18.
Blood ; 120(23): 4455-7, 2012 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-23197582

RESUMO

In this issue of Blood, Christoffersson et al provide data indicating the existence of a new neutrophil subset with angiogenic characteristics.


Assuntos
Transplante das Ilhotas Pancreáticas/fisiologia , Metaloproteinase 9 da Matriz/metabolismo , Neovascularização Fisiológica/fisiologia , Neutrófilos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Feminino , Masculino
19.
Blood ; 120(23): 4653-62, 2012 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-22966168

RESUMO

Recruitment and retention of leukocytes at a site of blood vessel growth are crucial for proper angiogenesis and subsequent tissue perfusion. Although critical for many aspects of regenerative medicine, the mechanisms of leukocyte recruitment to and actions at sites of angiogenesis are not fully understood. In this study, we investigated the signals attracting leukocytes to avascular transplanted pancreatic islets and leukocyte actions at the engraftment site. Expression of the angiogenic stimulus VEGF-A by mouse pancreatic islets was elevated shortly after syngeneic transplantation to muscle. High levels of leukocytes, predominantly CD11b(+)/Gr-1(+)/CXCR4(hi) neutrophils, were observed at the site of engraftment, whereas VEGF-A-deficient islets recruited only half of the amount of leukocytes when transplanted. Acute VEGF-A exposure of muscle increased leukocyte extravasation but not the levels of SDF-1α. VEGF-A-recruited neutrophils expressed 10 times higher amounts of MMP-9 than neutrophils recruited to an inflammatory stimulus. Revascularization of islets transplanted to MMP-9-deficient mice was impaired because blood vessels initially failed to penetrate grafts, and after 2 weeks vascularity was still disturbed. This study demonstrates that VEGF-A recruits a proangiogenic circulating subset of CD11b(+)/Gr-1(+) neutrophils that are CXCR4(hi) and deliver large amounts of the effector protein MMP-9, required for islet revascularization and functional integration after transplantation.


Assuntos
Transplante das Ilhotas Pancreáticas/fisiologia , Metaloproteinase 9 da Matriz/metabolismo , Neovascularização Fisiológica/fisiologia , Neutrófilos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Antígeno CD11b/metabolismo , Quimiocina CXCL12/metabolismo , Feminino , Hipóxia , Imuno-Histoquímica , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/metabolismo , Masculino , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Microscopia de Vídeo , Neovascularização Fisiológica/genética , Infiltração de Neutrófilos , Receptores CXCR4 , Receptores de Quimiocinas/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética
20.
Diabetes Metab Res Rev ; 28(6): 475-84, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22389124

RESUMO

Hypoxia is believed to be a crucial factor involved in cell adaptation to environmental stress. Islet transplantation, especially with immunoisolated islets, interrupts vascular connections, resulting in the substantially decreased delivery of oxygen and nutrients to islet cells. Insulin-producing pancreatic beta cells are known to be highly susceptible to oxygen deficiency. Such susceptibility to hypoxia is believed to be one of the main causes of beta-cell death in the post-transplantation period. Different strategies have been developed for the protection of beta cells against hypoxic injury and for oxygen delivery to transplanted islets. The enhancement of beta-cell defense properties against hypoxia has been achieved using various techniques such as gene transfection, drug supplementation, co-culturing with stem cells and cell selection. Technologies for oxygen delivery to transplanted islets include local neovascularization of subcutaneous sites, electrochemical and photosynthetic oxygen generation, oxygen refuelling of bio-artificial pancreas and whole body oxygenation by using hyperbaric therapy. Progress in the field of oxygen technologies for islet transplantation requires a multidisciplinary approach to explore and optimize the interaction between components of the biological system and different technological processes. This review article focuses mainly on the recently developed strategies for oxygenation and protection from hypoxic injury - to achieve stable and long-term normoglycaemia in diabetic patients with transplanted pancreatic islets.


Assuntos
Hipóxia Celular , Células Secretoras de Insulina/fisiologia , Transplante das Ilhotas Pancreáticas/métodos , Animais , Morte Celular/efeitos dos fármacos , Hipóxia Celular/genética , Separação Celular , Genes bcl-2/genética , Engenharia Genética , Peptídeo 1 Semelhante ao Glucagon/genética , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Precondicionamento Isquêmico/métodos , Transplante das Ilhotas Pancreáticas/fisiologia , Metalotioneína/genética , Neovascularização Fisiológica/efeitos dos fármacos , Oxigênio/administração & dosagem , Pâncreas Artificial , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/genética , Vitamina E/uso terapêutico , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA