Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Diabetes ; 72(11): 1641-1651, 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37625134

RESUMO

Extracellular (e)ATP, a potent proinflammatory molecule, is released by dying/damaged cells at the site of inflammation and is degraded by the membrane ectonucleotidases CD39 and CD73. In this study, we sought to unveil the role of eATP degradation in autoimmune diabetes. We then assessed the effect of soluble CD39 (sCD39) administration in prevention and reversal studies in NOD mice as well as in mechanistic studies. Our data showed that eATP levels were increased in hyperglycemic NOD mice compared with prediabetic NOD mice. CD39 and CD73 were found expressed by both α- and ß-cells and by different subsets of T cells. Importantly, prediabetic NOD mice displayed increased frequencies of CD3+CD73+CD39+ cells within their pancreata, pancreatic lymph nodes, and spleens. The administration of sCD39 into prediabetic NOD mice reduced their eATP levels, abrogated the proliferation of CD4+- and CD8+-autoreactive T cells, and increased the frequency of regulatory T cells, while delaying the onset of T1D. Notably, concomitant administration of sCD39 and anti-CD3 showed a strong synergism in restoring normoglycemia in newly hyperglycemic NOD mice compared with monotherapy with anti-CD3 or with sCD39. The eATP/CD39 pathway plays an important role in the onset of T1D, and its targeting might represent a potential therapeutic strategy in T1D.

2.
Diabetes ; 68(10): 1990-2003, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31371518

RESUMO

Tumors induce tolerance toward their antigens by producing the chemokine CCL21, leading to the formation of tertiary lymphoid organs (TLOs). Ins2-CCL21 transgenic, nonobese diabetic (NOD) mice express CCL21 in pancreatic ß-cells and do not develop autoimmune diabetes. We investigated by which mechanisms CCL21 expression prevented diabetes. Ins2-CCL21 mice develop TLOs by 4 weeks of age, consisting of naive CD4+ T cells compartmentalized within networks of CD45-gp38+CD31- fibroblastic reticular cell (FRC)-like cells. Importantly, 12-week-old Ins2-CCL21 TLOs contained FRC-like cells with higher contractility, regulatory, and anti-inflammatory properties and enhanced expression of ß-cell autoantigens compared with nontransgenic NOD TLOs found in inflamed islets. Consistently, transgenic mice harbored fewer autoreactive T cells and a higher proportion of regulatory T cells in the islets. Using adoptive transfer and islet transplantation models, we demonstrate that TLO formation in Ins2-CCL21 transgenic islets is critical for the regulation of autoimmunity, and although the effect is systemic, the induction is mediated locally likely by lymphocyte trafficking through TLOs. Overall, our findings suggest that CCL21 promotes TLOs that differ from inflammatory TLOs found in type 1 diabetic islets in that they resemble lymph nodes, contain FRC-like cells expressing ß-cell autoantigens, and are able to induce systemic and antigen-specific tolerance leading to diabetes prevention.


Assuntos
Quimiocina CCL21/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/metabolismo , Pâncreas/metabolismo , Células Estromais/metabolismo , Animais , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos
4.
Diabetologia ; 62(7): 1237-1250, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31087105

RESUMO

AIMS/HYPOTHESIS: Autoimmune attack against the insulin-producing beta cells in the pancreatic islets results in type 1 diabetes. However, despite considerable research, details of the type 1 diabetes immunopathology in situ are not fully understood mainly because of difficult access to the pancreatic islets in vivo. METHODS: Here, we used direct non-invasive confocal imaging of islets transplanted in the anterior chamber of the eye (ACE) to investigate the anti-islet autoimmunity in NOD mice before, during and after diabetes onset. ACE-transplanted islets allowed longitudinal studies of the autoimmune attack against islets and revealed the infiltration kinetics and in situ motility dynamics of fluorescence-labelled autoreactive T cells during diabetes development. Ex vivo immunostaining was also used to compare immune cell infiltrations into islet grafts in the eye and kidney as well as in pancreatic islets of the same diabetic NOD mice. RESULTS: We found similar immune infiltration in native pancreatic and ACE-transplanted islets, which established the ACE-transplanted islets as reliable reporters of the autoimmune response. Longitudinal studies in ACE-transplanted islets identified in vivo hallmarks of islet inflammation that concurred with early immune infiltration of the islets and preceded their collapse and hyperglycaemia onset. A model incorporating data on ACE-transplanted islet degranulation and swelling allowed early prediction of the autoimmune attack in the pancreas and prompted treatments to intercept type 1 diabetes. CONCLUSIONS/INTERPRETATION: The current findings highlight the value of ACE-transplanted islets in studying early type 1 diabetes pathogenesis in vivo and underscore the need for timely intervention to halt disease progression.


Assuntos
Diabetes Mellitus Tipo 1/diagnóstico por imagem , Animais , Autoimunidade/fisiologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/cirurgia , Sobrevivência de Enxerto/fisiologia , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/cirurgia , Transplante das Ilhotas Pancreáticas , Camundongos , Camundongos Endogâmicos NOD
5.
Cell Metab ; 27(3): 549-558.e4, 2018 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-29514065

RESUMO

Every animal species has a signature blood glucose level or glycemic set point. These set points are different, and the normal glycemic levels (normoglycemia) of one species would be life threatening for other species. Mouse normoglycemia can be considered diabetic for humans. The biological determinants of the glycemic set point remain unclear. Here we show that the pancreatic islet imposes its glycemic set point on the organism, making it the bona fide glucostat in the body. Moreover, and in contrast to rodent islets, glucagon input from the alpha cell to the insulin-secreting beta cell is necessary to fine-tune the distinctive human set point. These findings affect transplantation and regenerative approaches to treat diabetes because restoring normoglycemia may require more than replacing only the beta cells. Furthermore, therapeutic strategies using glucagon receptor antagonists as hypoglycemic agents need to be reassessed, as they may reset the overall glucostat in the organism.


Assuntos
Células Secretoras de Glucagon/metabolismo , Glucagon/metabolismo , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Animais , Diabetes Mellitus Experimental , Humanos , Hipoglicemiantes/farmacologia , Transplante das Ilhotas Pancreáticas , Macaca fascicularis , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Comunicação Parácrina , Receptores de Glucagon/antagonistas & inibidores
6.
Am J Transplant ; 18(3): 590-603, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29068143

RESUMO

Islet encapsulation may allow transplantation without immunosuppression, but thus far islets in large microcapsules transplanted in the peritoneal cavity have failed to reverse diabetes in humans. We showed that islet transplantation in confined well-vascularized sites like the epididymal fat pad (EFP) improved graft outcomes, but only conformal coated (CC) islets can be implanted in these sites in curative doses. Here, we showed that CC using polyethylene glycol (PEG) and alginate (ALG) was not immunoisolating because of its high permselectivity and strong allogeneic T cell responses. We refined the CC composition and explored PEG and islet-like extracellular matrix (Matrigel; MG) islet encapsulation (PEG MG) to improve capsule immunoisolation by decreasing its permselectivity and immunogenicity while allowing physiological islet function. Although the efficiency of diabetes reversal of allogeneic but not syngeneic CC islets was lower than that of naked islets, we showed that CC (PEG MG) islets from fully MHC-mismatched Balb/c mice supported long-term (>100 days) survival after transplantation into diabetic C57BL/6 recipients in the EFP site (750-1000 islet equivalents/mouse) in the absence of immunosuppression. Lack of immune cell penetration and T cell allogeneic priming was observed. These studies support the use of CC (PEG MG) for islet encapsulation and transplantation in clinically relevant sites without chronic immunosuppression.


Assuntos
Separação Celular/métodos , Diabetes Mellitus Experimental/terapia , Sobrevivência de Enxerto , Transplante das Ilhotas Pancreáticas/instrumentação , Ilhotas Pancreáticas/citologia , Neovascularização Fisiológica , Polietilenoglicóis/química , Aloenxertos , Animais , Cápsulas , Ilhotas Pancreáticas/imunologia , Transplante das Ilhotas Pancreáticas/métodos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
7.
Acta Biomater ; 49: 272-283, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27915019

RESUMO

Islet transplantation is a promising therapy for Type 1 diabetes mellitus; however, host inflammatory and immune responses lead to islet dysfunction and destruction, despite potent systemic immunosuppression. Grafting of poly(ethylene glycol) (PEG) to the periphery of cells or tissues can mitigate inflammation and immune recognition via generation of a steric barrier. Herein, we sought to evaluate the complementary impact of islet PEGylation with a short-course immunotherapy on the survival of fully-MHC mismatched islet allografts (DBA/2 islets into diabetic C57BL/6J recipients). Anti-Lymphocyte Function-associated Antigen 1 (LFA-1) antibody was selected as a complementary, transient, systemic immune monotherapy. Islets were PEGylated via an optimized protocol, with resulting islets exhibiting robust cell viability and function. Following transplantation, a significant subset of diabetic animals receiving PEGylated islets (60%) or anti-LFA-1 antibody (50%) exhibited long-term (>100d) normoglycemia. The combinatorial approach proved synergistic, with 78% of the grafts exhibiting euglycemia long-term. Additional studies examining graft cellular infiltrates at early time points characterized the local impact of the transplant protocol on graft survival. Results illustrate the capacity of a simple polymer grafting approach to impart significant immunoprotective effects via modulation of the local transplant environment, while short-term immunotherapy serves to complement this effect. STATEMENT OF SIGNIFICANCE: We believe this study is important and of interest to the biomaterials and transplant community for several reasons: 1) it provides an optimized protocol for the PEGylation of islets, with minimal impact on the coated islets, which can be easily translated for clinical applications; 2) this optimized protocol demonstrates the benefits of islet PEGylation in providing modest immunosuppression in a murine model; 3) this work demonstrates the combinatory impact of PEGylation with short-course immunotherapy (via LFA-1 blockage), illustrating the capacity of PEGylation to complement existing immunotherapy; and 4) it suggests macrophage phenotype shifting as the potential mechanism for this observed benefit.


Assuntos
Sobrevivência de Enxerto/imunologia , Imunoterapia , Transplante das Ilhotas Pancreáticas , Polietilenoglicóis/química , Animais , Antígenos de Histocompatibilidade/metabolismo , Antígeno-1 Associado à Função Linfocitária/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Modelos Animais , Sobrevivência de Tecidos , Transplante Homólogo
8.
Transplantation ; 101(5): 1025-1035, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27525644

RESUMO

BACKGROUND: Understanding the effects of capsule composition and transplantation site on graft outcomes of encapsulated islets will aid in the development of more effective strategies for islet transplantation without immunosuppression. METHODS: Here, we evaluated the effects of transplanting alginate (ALG)-based microcapsules (Micro) in the confined and well-vascularized epididymal fat pad (EFP) site, a model of the human omentum, as opposed to free-floating in the intraperitoneal cavity (IP) in mice. We also examined the effects of reinforcing ALG with polyethylene glycol (PEG). To allow transplantation in the EFP site, we minimized capsule size to 500 ± 17 µm. Unlike ALG, PEG resists osmotic stress, hence we generated hybrid microcapsules by mixing PEG and ALG (MicroMix) or by coating ALG capsules with a 15 ± 2 µm PEG layer (Double). RESULTS: We found improved engraftment of fully allogeneic BALB/c islets in Micro capsules transplanted in the EFP (median reversal time [MRT], 1 day) versus the IP site (MRT, 5 days; P < 0.01) in diabetic C57BL/6 mice and of Micro encapsulated (MRT, 8 days) versus naked (MRT, 36 days; P < 0.01) baboon islets transplanted in the EFP site. Although in vitro viability and functionality of islets within MicroMix and Double capsules were comparable to Micro, addition of PEG to ALG in MicroMix capsules improved engraftment of allogeneic islets in the IP site, but resulted deleterious in the EFP site, probably due to lower biocompatibility. CONCLUSIONS: Our results suggest that capsule composition and transplant site affect graft outcomes through their effects on nutrient availability, capsule stability, and biocompatibility.


Assuntos
Alginatos/administração & dosagem , Transplante das Ilhotas Pancreáticas/métodos , Polietilenoglicóis/administração & dosagem , Animais , Cápsulas , Epididimo , Ácido Glucurônico/administração & dosagem , Ácidos Hexurônicos/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Omento , Avaliação de Resultados em Cuidados de Saúde
9.
Diabetes ; 65(5): 1350-61, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26916086

RESUMO

Transplantation of pancreatic islets is a therapeutic option to preserve or restore ß-cell function. Our study was aimed at developing a clinically applicable protocol for extrahepatic transplantation of pancreatic islets. The potency of islets implanted onto the omentum, using an in situ-generated adherent, resorbable plasma-thrombin biologic scaffold, was evaluated in diabetic rat and nonhuman primate (NHP) models. Intraomental islet engraftment in the biologic scaffold was confirmed by achievement of improved metabolic function and preservation of islet cytoarchitecture, with reconstitution of rich intrainsular vascular networks in both species. Long-term nonfasting normoglycemia and adequate glucose clearance (tolerance tests) were achieved in both intrahepatic and intraomental sites in rats. Intraomental graft recipients displayed lower levels of serum biomarkers of islet distress (e.g., acute serum insulin) and inflammation (e.g., leptin and α2-macroglobulin). Importantly, low-purity (30:70% endocrine:exocrine) syngeneic rat islet preparations displayed function equivalent to that of pure (>95% endocrine) preparations after intraomental biologic scaffold implantation. Moreover, the biologic scaffold sustained allogeneic islet engraftment in immunosuppressed recipients. Collectively, our feasibility/efficacy data, along with the simplicity of the procedure and the safety of the biologic scaffold components, represented sufficient preclinical testing to proceed to a pilot phase I/II clinical trial.


Assuntos
Materiais Biocompatíveis , Diabetes Mellitus Experimental/cirurgia , Hiperglicemia/prevenção & controle , Transplante das Ilhotas Pancreáticas/métodos , Pâncreas Artificial , Alicerces Teciduais , Animais , Materiais Biocompatíveis/efeitos adversos , Materiais Biocompatíveis/química , Biomarcadores/sangue , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/patologia , Estudos de Viabilidade , Feminino , Terapia de Imunossupressão/efeitos adversos , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/ultraestrutura , Transplante das Ilhotas Pancreáticas/efeitos adversos , Transplante das Ilhotas Pancreáticas/imunologia , Transplante das Ilhotas Pancreáticas/patologia , Macaca fascicularis , Masculino , Microscopia Eletrônica de Varredura , Omento , Pâncreas Artificial/efeitos adversos , Plasma/química , Plasma/metabolismo , Ratos Endogâmicos Lew , Ratos Endogâmicos WF , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Propriedades de Superfície , Trombina/efeitos adversos , Trombina/química , Trombina/metabolismo , Engenharia Tecidual , Alicerces Teciduais/efeitos adversos , Alicerces Teciduais/química , Transplante Heterólogo/efeitos adversos , Transplante Heterotópico/efeitos adversos , Transplante Isogênico/efeitos adversos
10.
Biotechnol Bioeng ; 112(9): 1916-26, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25786390

RESUMO

With a view toward reduction of graft loss, we explored pancreatic islet transplantation within fibrin matrices rendered pro-angiogenic by incorporation of minimal doses of vascular endothelial growth factor-A165 and platelet-derived growth factor-BB presented complexed to a fibrin-bound integrin-binding fibronectin domain. Engineered matrices allowed for extended release of pro-angiogenic factors and for their synergistic signaling with extracellular matrix-binding domains in the post-transplant period. Aprotinin addition delayed matrix degradation and prolonged pro-angiogenic factor availability within the graft. Both subcutaneous (SC) and epididymal fat pad (EFP) sites were evaluated. We show that in the SC site, diabetes reversal in mice transplanted with 1,000 IEQ of syngeneic islets was not observed for islets transplanted alone, while engineered matrices resulted in a diabetes median reversal time (MDRT) of 38 days. In the EFP site, the MDRT with 250 IEQ of syngeneic islets within the engineered matrices was 24 days versus 86 days for islets transplanted alone. Improved function of engineered grafts was associated with enhanced and earlier (by day 7) angiogenesis. Our findings show that by engineering the transplant site to promote prompt re-vascularization, engraftment and long-term function of islet grafts can be improved in relevant extrahepatic sites.


Assuntos
Fibrina/química , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Becaplermina , Proliferação de Células/efeitos dos fármacos , Humanos , Hidrogéis/química , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-sis/química , Proteínas Proto-Oncogênicas c-sis/deficiência , Proteínas Proto-Oncogênicas c-sis/farmacologia , Fator A de Crescimento do Endotélio Vascular/química
11.
Proc Natl Acad Sci U S A ; 111(29): 10514-9, 2014 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-24982192

RESUMO

Encapsulation of islets of Langerhans may represent a way to transplant islets in the absence of immunosuppression. Traditional methods for encapsulation lead to diffusional limitations imposed by the size of the capsules (600-1,000 µm in diameter), which results in core hypoxia and delayed insulin secretion in response to glucose. Moreover, the large volume of encapsulated cells does not allow implantation in sites that might be more favorable to islet cell engraftment. To address these issues, we have developed an encapsulation method that allows conformal coating of islets through microfluidics and minimizes capsule size and graft volume. In this method, capsule thickness, rather than capsule diameter, is constant and tightly defined by the microdevice geometry and the rheological properties of the immiscible fluids used for encapsulation within the microfluidic system. We have optimized the method both computationally and experimentally, and found that conformal coating allows for complete encapsulation of islets with a thin (a few tens of micrometers) continuous layer of hydrogel. Both in vitro and in vivo in syngeneic murine models of islet transplantation, the function of conformally coated islets was not compromised by encapsulation and was comparable to that of unencapsulated islets. We have further demonstrated that the structural support conferred by the coating materials protected islets from the loss of function experienced by uncoated islets during ex vivo culture.


Assuntos
Materiais Revestidos Biocompatíveis/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Microfluídica/instrumentação , Alginatos/farmacologia , Animais , Agregação Celular , Simulação por Computador , Desenho de Equipamento , Ácido Glucurônico/farmacologia , Ácidos Hexurônicos/farmacologia , Hidrodinâmica , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Microesferas , Modelos Biológicos , Polietilenoglicóis/farmacologia , Reprodutibilidade dos Testes
12.
Diabetes ; 63(2): 701-14, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24101677

RESUMO

Podocytes are a major component of the glomerular filtration barrier, and their ability to sense insulin is essential to prevent proteinuria. Here we identify the insulin downstream effector GLUT4 as a key modulator of podocyte function in diabetic nephropathy (DN). Mice with a podocyte-specific deletion of GLUT4 (G4 KO) did not develop albuminuria despite having larger and fewer podocytes than wild-type (WT) mice. Glomeruli from G4 KO mice were protected from diabetes-induced hypertrophy, mesangial expansion, and albuminuria and failed to activate the mammalian target of rapamycin (mTOR) pathway. In order to investigate whether the protection observed in G4 KO mice was due to the failure to activate mTOR, we used three independent in vivo experiments. G4 KO mice did not develop lipopolysaccharide-induced albuminuria, which requires mTOR activation. On the contrary, G4 KO mice as well as WT mice treated with the mTOR inhibitor rapamycin developed worse adriamycin-induced nephropathy than WT mice, consistent with the fact that adriamycin toxicity is augmented by mTOR inhibition. In summary, GLUT4 deficiency in podocytes affects podocyte nutrient sensing, results in fewer and larger cells, and protects mice from the development of DN. This is the first evidence that podocyte hypertrophy concomitant with podocytopenia may be associated with protection from proteinuria.


Assuntos
Regulação da Expressão Gênica/fisiologia , Transportador de Glucose Tipo 4/metabolismo , Podócitos/citologia , Podócitos/metabolismo , Albuminúria , Animais , Tamanho Celular , Nefropatias Diabéticas , Doxorrubicina/toxicidade , Feminino , Barreira de Filtração Glomerular/citologia , Barreira de Filtração Glomerular/patologia , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Transportador de Glucose Tipo 4/genética , Lipopolissacarídeos/toxicidade , Camundongos
13.
Immunol Res ; 57(1-3): 185-96, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24242759

RESUMO

The ultimate goal of diabetes therapy is the restoration of physiologic metabolic control. For type 1 diabetes, research efforts are focused on the prevention or early intervention to halt the autoimmune process and preserve ß cell function. Replacement of pancreatic ß cells via islet transplantation reestablishes physiologic ß cell function in patients with diabetes. Emerging research shows that microRNAs (miRNAs), noncoding small RNA molecules produced by a newly discovered class of genes, negatively regulate gene expression. MiRNAs recognize and bind to partially complementary sequences of target messenger RNA (mRNA), regulating mRNA translation and affecting gene expression. Correlation between miRNA signatures and genome-wide RNA expression allows identification of multiple miRNA-mRNA pairs in biological processes. Because miRNAs target functionally related genes, they represent an exciting and indispensable approach for biomarkers and drug discovery. We are studying the role of miRNA in the context of islet immunobiology. Our research aims at understanding the mechanisms underlying pancreatic ß cell loss and developing clinically relevant approaches for preservation and restoration of ß cell function to treat insulin-dependent diabetes. Herein, we discuss some of our recent efforts related to the study of miRNA in islet inflammation and islet engraftment. Our working hypothesis is that modulation of the expression of specific microRNAs in the transplant microenvironment will be of assistance in enhancing islet engraftment and promoting long-term function.


Assuntos
Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , MicroRNAs/genética , Animais , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/terapia , Sobrevivência de Enxerto/genética , Humanos , Inflamação/genética , Inflamação/metabolismo , Ilhotas Pancreáticas/patologia , MicroRNAs/metabolismo , Neovascularização Fisiológica/genética
14.
Immunol Res ; 57(1-3): 210-21, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24233663

RESUMO

The focus of our research is on islet immunobiology. We are exploring novel strategies that could be of assistance in the treatment and prevention of type 1 diabetes, as well as in the restoration of metabolic control via transplantation of insulin producing cells (i.e., islet cells). The multiple facets of diabetes and ß-cell replacement encompass different complementary disciplines, such as immunology, cell biology, pharmacology, and bioengineering, among others. Through their interaction and integration, a transdisciplinary dimension is needed in order to address and overcome all aspects of the complex puzzle toward a successful clinical translation of a biological cure for diabetes.


Assuntos
Ilhotas Pancreáticas/imunologia , Trifosfato de Adenosina/metabolismo , Animais , Microambiente Celular , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/terapia , Humanos , Oxigenoterapia Hiperbárica , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/transplante , Ilhotas Pancreáticas/metabolismo , Transplante das Ilhotas Pancreáticas/métodos , Transdução de Sinais
15.
Circulation ; 127(4): 463-75, 2013 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-23250993

RESUMO

BACKGROUND: Heart transplantation is a lifesaving procedure for patients with end-stage heart failure. Despite much effort and advances in the field, current immunosuppressive regimens are still associated with poor long-term cardiac allograft outcomes, and with the development of complications, including infections and malignancies, as well. The development of a novel, short-term, and effective immunomodulatory protocol will thus be an important achievement. The purine ATP, released during cell damage/activation, is sensed by the ionotropic purinergic receptor P2X7 (P2X7R) on lymphocytes and regulates T-cell activation. Novel clinical-grade P2X7R inhibitors are available, rendering the targeting of P2X7R a potential therapy in cardiac transplantation. METHODS AND RESULTS: We analyzed P2X7R expression in patients and mice and P2X7R targeting in murine recipients in the context of cardiac transplantation. Our data demonstrate that P2X7R is specifically upregulated in graft-infiltrating lymphocytes in cardiac-transplanted humans and mice. Short-term P2X7R targeting with periodate-oxidized ATP promotes long-term cardiac transplant survival in 80% of murine recipients of a fully mismatched allograft. Long-term survival of cardiac transplants was associated with reduced T-cell activation, T-helper cell 1/T-helper cell 17 differentiation, and inhibition of STAT3 phosphorylation in T cells, thus leading to a reduced transplant infiltrate and coronaropathy. In vitro genetic upregulation of the P2X7R pathway was also shown to stimulate T-helper cell 1/T-helper cell 17 cell generation. Finally, P2X7R targeting halted the progression of coronaropathy in a murine model of chronic rejection as well. CONCLUSIONS: P2X7R targeting is a novel clinically relevant strategy to prolong cardiac transplant survival.


Assuntos
Trifosfato de Adenosina/análogos & derivados , Rejeição de Enxerto/tratamento farmacológico , Rejeição de Enxerto/mortalidade , Transplante de Coração/mortalidade , Antagonistas do Receptor Purinérgico P2X/farmacologia , Receptores Purinérgicos P2X7/metabolismo , Trifosfato de Adenosina/farmacologia , Adulto , Animais , Doença Crônica , Modelos Animais de Doenças , Feminino , Rejeição de Enxerto/imunologia , Transplante de Coração/imunologia , Humanos , Imunocompetência/efeitos dos fármacos , Imunocompetência/imunologia , Isoantígenos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/imunologia , Fator de Transcrição STAT3/metabolismo , Sobreviventes/estatística & dados numéricos , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Células Th17/efeitos dos fármacos , Células Th17/imunologia
16.
Cell Transplant ; 22(7): 1123-35, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23031502

RESUMO

Clinical islet transplantation has demonstrated success in treating type 1 diabetes. A current limitation is the intrahepatic portal vein transplant site, which is prone to mechanical stress and inflammation. Transplantation of pancreatic islets into alternative sites is preferable, but challenging, as it may require a three-dimensional vehicle to confer mechanical protection and to confine islets to a well-defined, retrievable space where islet neovascularization can occur. We have fabricated biostable, macroporous scaffolds from poly(dimethylsiloxane) (PDMS) and investigated islet retention and distribution, metabolic function, and glucose-dependent insulin secretion within these scaffolds. Islets from multiple sources, including rodents, nonhuman primates, and humans, were tested in vitro. We observed high islet retention and distribution within PDMS scaffolds, with retention of small islets (< 100 µm) improved through the postloading addition of fibrin gel. Islets loaded within PDMS scaffolds exhibited viability and function comparable to standard culture conditions when incubated under normal oxygen tensions, but displayed improved viability compared to standard two-dimensional culture controls under low oxygen tensions. In vivo efficacy of scaffolds to support islet grafts was evaluated after transplantation in the omental pouch of chemically induced diabetic syngeneic rats, which promptly achieved normoglycemia. Collectively, these results are promising in that they indicate the potential for transplanting islets into a clinically relevant, extrahepatic site that provides spatial distribution of islets as well as intradevice vascularization.


Assuntos
Dimetilpolisiloxanos/química , Transplante das Ilhotas Pancreáticas , Adulto , Animais , Sobrevivência Celular/efeitos dos fármacos , Pré-Escolar , Feminino , Humanos , Hidrogéis/química , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Masculino , Pessoa de Meia-Idade , Omento/patologia , Porosidade , Primatas , Ratos , Ratos Endogâmicos Lew , Estresse Mecânico , Adulto Jovem
17.
J Transplant ; 2012: 723614, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22655170

RESUMO

Nonspecific inflammation in the transplant microenvironment results in ß-cell dysfunction and death influencing negatively graft outcome. MicroRNA (miRNA) expression and gene target regulation in transplanted islets are not yet well characterized. We evaluated the impact of inflammation on miRNA expression in transplanted rat islets. Islets exposed in vitro to proinflammatory cytokines and explanted syngeneic islet grafts were evaluated by miRNA arrays. A subset of 26 islet miRNAs was affected by inflammation both in vivo and in vitro. Induction of miRNAs was dependent on NF-κB, a pathway linked with cytokine-mediated islet cell death. RT-PCR confirmed expression of 8 miRNAs. The association between these miRNAs and mRNA target-predicting algorithms in genome-wide RNA studies of ß-cell inflammation identified 238 potential miRNA gene targets. Several genes were ontologically associated with regulation of insulin signaling and secretion, diabetes, and islet physiology. One of the most activated miRNAs was miR-21. Overexpression of miR-21 in insulin-secreting MIN6 cells downregulated endogenous expression of the tumor suppressor Pdcd4 and of Pclo, a Ca(2+) sensor protein involved in insulin secretion. Bioinformatics identified both as potential targets. The integrated analysis of miRNA and mRNA expression profiles revealed potential targets that may identify molecular targets for therapeutic interventions.

18.
Diabetes ; 61(7): 1769-78, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22566533

RESUMO

We evaluated the effects of hyperbaric oxygen therapy (HOT) on autoimmune diabetes development in nonobese diabetic (NOD) mice. Animals received no treatment or daily 60-min HOT 100% oxygen (HOT-100%) at 2.0 atmospheres absolute and were monitored for diabetes onset, insulitis, infiltrating cells, immune cell function, and ß-cell apoptosis and proliferation. Cyclophosphamide-induced diabetes onset was reduced from 85.3% in controls to 48% after HOT-100% (P < 0.005) and paralleled by lower insulitis. Spontaneous diabetes incidence reduced from 85% in controls to 65% in HOT-100% (P = 0.01). Prediabetic mice receiving HOT-100% showed lower insulitis scores, reduced T-cell proliferation upon stimulation in vitro (P < 0.03), increased CD62L expression in T cells (P < 0.04), reduced costimulation markers (CD40, DC80, and CD86), and reduced major histocompatibility complex class II expression in dendritic cells (DCs) (P < 0.025), compared with controls. After autoimmunity was established, HOT was less effective. HOT-100% yielded reduced apoptosis (transferase-mediated dUTP nick-end labeling-positive insulin-positive cells; P < 0.01) and increased proliferation (bromodeoxyuridine incorporation; P < 0.001) of insulin-positive cells compared with controls. HOT reduces autoimmune diabetes incidence in NOD mice via increased resting T cells and reduced activation of DCs with preservation of ß-cell mass resulting from decreased apoptosis and increased proliferation. The safety profile and noninvasiveness makes HOT an appealing adjuvant therapy for diabetes prevention and intervention trials.


Assuntos
Proliferação de Células , Diabetes Mellitus Tipo 1/prevenção & controle , Oxigenoterapia Hiperbárica , Células Secretoras de Insulina/fisiologia , Animais , Apoptose/imunologia , Antígeno B7-1/imunologia , Antígeno B7-2/biossíntese , Antígeno B7-2/imunologia , Antígenos CD40/biossíntese , Antígenos CD40/imunologia , Ciclofosfamida/efeitos adversos , Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/induzido quimicamente , Diabetes Mellitus Tipo 1/imunologia , Feminino , Genes MHC da Classe II/imunologia , Imunossupressores/efeitos adversos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/imunologia , Selectina L/biossíntese , Selectina L/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos NOD , Pancreatite/imunologia , Pancreatite/prevenção & controle , Linfócitos T/imunologia
19.
Cell Transplant ; 21(7): 1349-60, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22305457

RESUMO

Ischemic preconditioning (IPC) confers tissue resistance to subsequent ischemia in several organs. The protective effects are obtained by applying short periods of warm ischemia followed by reperfusion prior to extended ischemic insults to the organs. In the present study, we evaluated whether IPC can reduce pancreatic tissue injury following cold ischemic preservation. Rat pancreata were exposed to IPC (10 min of warm ischemia followed by 10 min of reperfusion) prior to ~18 h of cold preservation before assessment of organ injury or islet isolation. Pancreas IPC improved islet yields (964 ± 336 vs. 711 ± 204 IEQ/pancreas; p = 0.004) and lowered islet loss after culture (33 ± 10% vs. 51 ± 14%; p = 0.0005). Islet potency in vivo was well preserved with diabetes reversal and improved glucose clearance. Pancreas IPC reduced levels of NADPH-dependent oxidase, a source of reactive oxygen species, in pancreas homogenates versus controls (78.4 ± 45.9 vs. 216.2 ± 53.8 RLU/µg; p = 0.002). Microarray genomic analysis of pancreata revealed upregulation of 81 genes and downregulation of 454 genes (greater than twofold change) when comparing IPC-treated glands to controls, respectively, and showing a decrease in markers of apoptosis and oxidative stress. Collectively, our study demonstrates beneficial effects of IPC of the pancreas prior to cold organ preservation and provides evidence of the key role of IPC-mediated modulation of oxidative stress pathways. The use of IPC of the pancreas may contribute to increasing the quality of donor pancreas for transplantation and to improving organ utilization.


Assuntos
Precondicionamento Isquêmico , Preservação de Órgãos , Pâncreas/fisiologia , Animais , Glicemia/análise , Separação Celular , Diabetes Mellitus Experimental/cirurgia , Regulação da Expressão Gênica , Ilhotas Pancreáticas/citologia , Transplante das Ilhotas Pancreáticas , Masculino , Camundongos , Camundongos Nus , NADPH Oxidases/metabolismo , Estresse Oxidativo , Ratos , Ratos Endogâmicos Lew
20.
Cell Transplant ; 21(6): 1321-39, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22195604

RESUMO

We sought to assess the potential of human cord blood-derived mesenchymal stem cells (CB-MSCs) to derive insulin-producing, glucose-responsive cells. We show here that differentiation protocols based on stepwise culture conditions initially described for human embryonic stem cells (hESCs) lead to differentiation of cord blood-derived precursors towards a pancreatic endocrine phenotype, as assessed by marker expression and in vitro glucose-regulated insulin secretion. Transplantation of these cells in immune-deficient animals shows human C-peptide production in response to a glucose challenge. These data suggest that human cord blood may be a promising source for regenerative medicine approaches for the treatment of diabetes mellitus.


Assuntos
Glucose/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Células-Tronco Mesenquimais/citologia , Cordão Umbilical/citologia , Animais , Biomarcadores/metabolismo , Peptídeo C/metabolismo , Diferenciação Celular , Células Cultivadas , Células Endócrinas/citologia , Células Endócrinas/metabolismo , Humanos , Imunofenotipagem , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/transplante , Fatores de Transcrição Maf/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Fatores de Transcrição/metabolismo , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...