Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
PLoS One ; 13(2): e0193184, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29474443

RESUMEN

Amyloid formation in the pancreatic islets due to aggregation of human islet amyloid polypeptide (hIAPP) contributes to reduced ß-cell mass and function in type 2 diabetes (T2D) and islet transplantation. Protein kinase B (PKB) signaling plays a key role in the regulation of ß-cell survival, function and proliferation. In this study, we used human and hIAPP-expressing transgenic mouse islets in culture as two ex vivo models of human islet amyloid formation to: 1. Investigate the effects of amyloid formation on PKB phosphorylation in primary islet ß-cells; 2. Test if inhibition of amyloid formation and/or interleukin-1ß (IL-1ß) signaling in islets can restore the changes in ß-cell phospho-PKB levels mediated by amyloid formation. Human and hIAPP-expressing mouse islets were cultured in elevated glucose with an amyloid inhibitor (Congo red) or embedded within collagen matrix to prevent amyloid formation. To block the IL-1ß signaling, human islets were treated with an IL-1 receptor antagonist (anakinra) or a glucagon-like peptide-1 agonist (exenatide). ß-cell phospho-PKB levels, proliferation, apoptosis, islet IL-1ß levels and amyloid formation were assessed. Amyloid formation in both cultured human and hIAPP-expressing mouse islets reduced ß-cell phospho-PKB levels and increased islet IL-1ß levels, both of which were restored by prevention of amyloid formation either by the amyloid inhibitor or embedding islets in collagen matrix, resulting in improved ß-cell survival. Furthermore, inhibition of IL-1ß signaling by treatment with anakinra or exenatide increased ß-cell phospho-PKB levels, enhanced proliferation and reduced apoptosis in amyloid forming human islets during 7-day culture. These data suggest that amyloid formation leads to reduced PKB phosphorylation in ß-cells which is associated with elevated islet IL-1ß levels. Inhibitors of amyloid or amyloid-induced IL-1ß production may provide a new approach to restore phospho-PKB levels thereby enhance ß-cell survival and proliferation in conditions associated with islet amyloid formation such as T2D and clinical islet transplantation.


Asunto(s)
Amiloide/metabolismo , Células Secretoras de Insulina/metabolismo , Interleucina-1beta/metabolismo , Transducción de Señal , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/terapia , Femenino , Humanos , Células Secretoras de Insulina/patología , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Trasplante de Islotes Pancreáticos , Masculino , Ratones , Ratones Transgénicos , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt
2.
Mol Metab ; 6(8): 833-844, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28752047

RESUMEN

OBJECTIVES: ß-cell dysfunction and apoptosis associated with islet inflammation play a key role in the pathogenesis of type 2 diabetes (T2D). Growing evidence suggests that islet amyloid, formed by aggregation of human islet amyloid polypeptide (hIAPP), contributes to islet inflammation and ß-cell death in T2D. We recently showed the role of interleukin-1ß (IL-1ß)/Fas/caspase-8 apoptotic pathway in amyloid-induced ß-cell death. In this study, we used human islets in culture as an ex vivo model of amyloid formation to: (1) investigate the effects of amyloid on islet levels of the natural IL-1 receptor antagonist (IL-1Ra); (2) examine if modulating the IL-1ß/IL-1Ra balance can prevent amyloid-induced ß-cell Fas upregulation and apoptosis. METHODS: Isolated human islets (n = 10 donors) were cultured in elevated glucose (to form amyloid) with or without a neutralizing human IL-1ß antibody for up to 7 days. Parallel studies were performed with human islets in which amyloid formation was prevented by adeno-siRNA-mediated suppression of hIAPP expression (as control). ß-cell levels of IL-1Ra, Fas, apoptosis as well as islet function, insulin- and amyloid-positive areas, and IL-1Ra release were assessed. RESULTS: Progressive amyloid formation in human islets during culture was associated with alterations in IL-1Ra. Islet IL-1Ra levels were higher at early stages but were markedly reduced at later stages of amyloid formation. Furthermore, IL-1Ra release from human islets was reduced during 7-day culture in a time-dependent manner. These changes in IL-1Ra production and release from human islets during amyloid formation adversely correlated with islet IL-1ß levels, ß-cell Fas expression and apoptosis. Treatment with IL-1ß neutralizing antibody markedly reduced amyloid-induced ß-cell Fas expression and apoptosis, thereby improving islet ß-cell survival and function during culture. CONCLUSIONS: These data suggest that amyloid formation impairs the balance between IL-1ß and IL-1Ra in islets by increasing IL-1ß production and reducing IL-1Ra levels thereby promoting ß-cell dysfunction and death. Restoring the IL-1ß/IL-1Ra ratio may provide an effective strategy to protect islet ß-cells from amyloid toxicity in T2D.


Asunto(s)
Amiloide/metabolismo , Apoptosis , Células Secretoras de Insulina/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Interleucina-1beta/metabolismo , Adolescente , Adulto , Animales , Caspasa 8/metabolismo , Línea Celular , Células Cultivadas , Proteína Ligando Fas/metabolismo , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Persona de Mediana Edad
3.
Diabetes Obes Metab ; 19(5): 682-694, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28058779

RESUMEN

AIMS: Islet amyloid, formed by aggregation of human islet amyloid polypeptide (hIAPP), contributes to ß-cell failure in type 2 diabetes, cultured and transplanted islets. We previously showed that biosynthetic hIAPP aggregates induce ß-cell Fas upregulation and activation of the Fas apoptotic pathway. We used cultured human and hIAPP-expressing mouse islets to investigate: (1) the role of interleukin-1ß (IL-1ß) in amyloid-induced Fas upregulation; and (2) the effects of IL-1ß-induced ß-cell dysfunction on pro-islet amyloid polypeptide (proIAPP) processing and amyloid formation. RESEARCH DESIGN AND METHODS: Human and h IAPP -expressing mouse islets were cultured to form amyloid without or with the IL-1 receptor antagonist (IL-1Ra) anakinra, in the presence or absence of recombinant IL-1ß. Human islets in which amyloid formation was prevented (amyloid inhibitor or Ad-prohIAPP-siRNA) were cultured similarly. ß-cell function, apoptosis, Fas expression, caspase-8 activation, islet IL-1ß, ß-cell area, ß-/α-cell ratio, amyloid formation, and (pro)IAPP forms were assessed. RESULTS: hIAPP aggregates were found to increase IL-1ß levels in cultured human islets that correlated with ß-cell Fas upregulation, caspase-8 activation and apoptosis, all of which were reduced by IL-1Ra treatment or prevention of amyloid formation. Moreover, IL-1Ra improved culture-induced ß-cell dysfunction and restored impaired proIAPP processing, leading to lower amyloid formation. IL-1ß treatment potentiated impaired proIAPP processing and increased amyloid formation in cultured human and h IAPP -expressing mouse islets, which were prevented by IL-1Ra. CONCLUSIONS: IL-1ß plays a dual role by: (1) mediating amyloid-induced Fas upregulation and ß-cell apoptosis; (2) inducing impaired proIAPP processing thereby potentiating amyloid formation. Blocking IL-1ß may provide a new strategy to preserve ß cells in conditions associated with islet amyloid formation.


Asunto(s)
Amiloide/agonistas , Apoptosis , Interleucina-1beta/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/metabolismo , Receptor fas/agonistas , Adulto , Amiloide/antagonistas & inhibidores , Amiloide/química , Amiloide/metabolismo , Animales , Cadáver , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/cirugía , Hemicigoto , Humanos , Insulina/metabolismo , Secreción de Insulina , Proteína Antagonista del Receptor de Interleucina 1/genética , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/genética , Polipéptido Amiloide de los Islotes Pancreáticos/antagonistas & inhibidores , Polipéptido Amiloide de los Islotes Pancreáticos/química , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Islotes Pancreáticos/citología , Islotes Pancreáticos/patología , Trasplante de Islotes Pancreáticos/efectos adversos , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Precursores de Proteínas/antagonistas & inhibidores , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Interferencia de ARN , Proteínas Recombinantes/metabolismo , Técnicas de Cultivo de Tejidos , Receptor fas/metabolismo
4.
Islets ; 8(2): 48-56, 2016 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-26909740

RESUMEN

Worldwide efforts are underway to replace or repair lost or dysfunctional pancreatic ß-cells to cure diabetes. However, it is unclear what the final product of these efforts should be, as ß-cells are thought to be heterogeneous. To enable the analysis of ß-cell heterogeneity in an unbiased and quantitative way, we developed model-free and model-based statistical clustering approaches, and created new software called TraceCluster. Using an example data set, we illustrate the utility of these approaches by clustering dynamic intracellular Ca(2+) responses to high glucose in ∼300 simultaneously imaged single islet cells. Using feature extraction from the Ca(2+) traces on this reference data set, we identified 2 distinct populations of cells with ß-like responses to glucose. To the best of our knowledge, this report represents the first unbiased cluster-based analysis of human ß-cell functional heterogeneity of simultaneous recordings. We hope that the approaches and tools described here will be helpful for those studying heterogeneity in primary islet cells, as well as excitable cells derived from embryonic stem cells or induced pluripotent cells.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Glucosa/metabolismo , Islotes Pancreáticos/citología , Animales , Humanos , Islotes Pancreáticos/metabolismo , Programas Informáticos
5.
J Cell Physiol ; 231(9): 1964-73, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26743772

RESUMEN

Indoleamine 2,3-dioxygenase (IDO) induces immunological tolerance in physiological and pathological conditions. Therefore, we used dermal fibroblasts with stable IDO expression as a cell therapy to: (i) Investigate the factors determining the efficacy of this cell therapy for autoimmune diabetes in non-obese diabetic (NOD) mice; (ii) Scrutinize the potential immunological mechanisms. Newly diabetic NOD mice were randomly injected with either 10 × 10(6) (10M) or 15 × 10(6) (15M) IDO-expressing dermal fibroblasts. Blood glucose levels (BGLs), body weight, plasma kynurenine levels, insulitis severity, islet beta cell function, autoreactive CD8(+) T cells, Th17 cells and regulatory T cells (Tregs) were then investigated in these mice. IL-1ß and cleaved caspase-3 levels were assessed in islets co-cultured with IDO-expressing fibroblasts. BGLs in 83% mice treated with 15M IDO-expressing fibroblasts recovered to normal up to 120 days. However, only 17% mice treated with 10M IDO-expressing cells were reversed to normoglycemia. A 15M IDO-expressing fibroblasts significantly reduced infiltrated immune cells in islets and recovered the functionality of remaining islet beta cells in NOD mice. Additionally, they successfully inhibited autoreactive CD8(+) T cells and Th17 cells as well as increased Tregs in different organs of NOD mice. Islet beta cells co-cultured with IDO-expressing fibroblasts had reduced IL-1ß levels and cell apoptosis. Both cell number and IDO enzymatic activity contributes to the efficiency of IDO cell therapy. Optimized IDO-expressing fibroblasts successfully reverse the progression of diabetes in NOD mice through induction of Tregs as well as inhibition of beta cell specific autoreactive CD8(+) T cells and Th17 cells. J. Cell. Physiol. 231: 1964-1973, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Diabetes Mellitus Experimental/inmunología , Fibroblastos/enzimología , Hiperglucemia/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Células Secretoras de Insulina/inmunología , Animales , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Femenino , Hiperglucemia/inmunología , Células Secretoras de Insulina/enzimología , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Linfocitos T Reguladores/inmunología
6.
Diabetologia ; 57(4): 765-75, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24442508

RESUMEN

AIMS/HYPOTHESIS: Reduced beta cell mass due to increased beta cell apoptosis is a key defect in type 2 diabetes. Islet amyloid, formed by the aggregation of human islet amyloid polypeptide (hIAPP), contributes to beta cell death in type 2 diabetes and in islet grafts in patients with type 1 diabetes. In this study, we used human islets and hIAPP-expressing mouse islets with beta cell Casp8 deletion to (1) investigate the role of caspase-8 in amyloid-induced beta cell apoptosis and (2) test whether caspase-8 inhibition protects beta cells from amyloid toxicity. METHODS: Human islet cells were cultured with hIAPP alone, or with caspase-8, Fas or amyloid inhibitors. Human islets and wild-type or hIAPP-expressing mouse islets with or without caspase-8 expression (generated using a Cre/loxP system) were cultured to form amyloid. Caspase-8 and -3 activation, Fas and FLICE inhibitory protein (FLIP) expression, islet beta cell and amyloid area, IL-1ß levels, and the beta:alpha cell ratio were assessed. RESULTS: hIAPP treatment induced activation of caspase-8 and -3 in islet beta cells (via Fas upregulation), resulting in apoptosis, which was markedly reduced by blocking caspase-8, Fas or amyloid. Amyloid formation in cultured human and hIAPP-expressing mouse islets induced caspase-8 activation, which was associated with Fas upregulation and elevated islet IL-1ß levels. hIAPP-expressing mouse islets with Casp8 deletion had comparable amyloid, IL-1ß and Fas levels with those expressing hIAPP and Casp8, but markedly lower beta cell apoptosis, higher beta:alpha cell ratio, greater beta cell area, and enhanced beta cell function. CONCLUSIONS/INTERPRETATION: Beta cell Fas upregulation by endogenously produced and exogenously applied hIAPP aggregates promotes caspase-8 activation, resulting in beta cell apoptosis. The prevention of amyloid-induced caspase-8 activation enhances beta cell survival and function in islets.


Asunto(s)
Amiloide/toxicidad , Caspasa 8/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/enzimología , Islotes Pancreáticos/citología , Adulto , Animales , Caspasa 3/metabolismo , Caspasa 8/genética , Femenino , Humanos , Técnicas In Vitro , Masculino , Ratones , Persona de Mediana Edad
7.
Pancreas ; 43(1): 128-34, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24326367

RESUMEN

OBJECTIVES: B7-H4 is a negative coregulatory molecule known to be involved in immune response. We study here B7-H4 expression and its possible role in diabetes and cancer development. METHODS: Formalin-fixed, paraffin-processed pancreas samples from patients with type 1 diabetes (T1D), insulinoma, pancreatic ductal adenocarcinoma (PDAC), and normal organ donors were studied by bright-field and multifluorescence immunohistochemistry to examine B7-H4 expression and its colocalization with islet endocrine hormones. Quantitative RT-PCR and Western blot assay were used to examine B7-H4 mRNA and protein expression in the islet and exocrine tissues from normal donors and pancreatic cancer cell lines. RESULTS: B7-H4 protein expression in islet ß cells is decreased in T1D and PDAC, but increased in insulinoma patients when compared to normal controls; the changes in B7-H4 expression are concomitant with insulin expression on the islet ß cells. The insulin/B7-H4 colocalization on the ß cells, expressed in colocalization coefficient Pearson r, is also changed in these islets. CONCLUSIONS: Our observation of altered B7-H4 expression, concomitant with insulin expression, in the pancreatic islets of T1D, PDAC, and insulinoma patients when compared to normal controls suggests that B7-H4 pathway might play an important role in maintenance of ß-cell function, but its exact role remains to be explored.


Asunto(s)
Adenocarcinoma/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , Neoplasias Pancreáticas/metabolismo , Inhibidor 1 de la Activación de Células T con Dominio V-Set/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patología , Western Blotting , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular , Línea Celular Tumoral , Diabetes Mellitus Tipo 1/genética , Expresión Génica , Humanos , Inmunohistoquímica , Insulina/genética , Insulina/metabolismo , Insulinoma/genética , Insulinoma/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Inhibidor 1 de la Activación de Células T con Dominio V-Set/genética
8.
Biochem J ; 451(2): 227-34, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23390957

RESUMEN

The incretins, GIP (glucose-dependent insulinotropic polypeptide) and GLP-1 (glucagon-like peptide-1) are gastrointestinal hormones conferring a number of beneficial effects on ß-cell secretion, survival and proliferation. In a previous study, it was demonstrated that delayed rectifier channel protein Kv2.1 contributes to ß-cell apoptosis and that the prosurvival effects of incretins involve Kv2.1 PTMs (post-translational modifications), including phosphorylation and acetylation. Since Kv1.5 overexpression was also shown to stimulate ß-cell death, the present study was initiated in order to determine whether incretins modulate Kv1.5α-Kvß2 interaction via PTM and the mechanisms involved. GIP and GLP-1 reduced apoptosis in INS-1 ß-cells (clone 832/13) overexpressing Kv1.5, and RNAi (RNA interference)-mediated knockdown of endogenous Kv1.5 attenuated apoptotic ß-cell death. Both GIP and GLP-1 increased phosphorylation and acetylation of Kv1.5 and its Kvß2 protein subunit, leading to their enhanced interaction. Further studies demonstrated that CBP [CREB (cAMP-response-element-binding protein)-binding protein]/SirT1 mediated acetylation/deacetylation and interaction between Kvß2 and Kv1.5 in response to GIP or GLP-1. Incretin regulation of ß-cell function therefore involves the acetylation of multiple Kvα and Kvß subunits.


Asunto(s)
Proteína de Unión a CREB/metabolismo , Incretinas/farmacología , Células Secretoras de Insulina/metabolismo , Canal de Potasio Kv1.5/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Sirtuina 1/metabolismo , Acetilación/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proteína de Unión a CREB/genética , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Polipéptido Inhibidor Gástrico/metabolismo , Polipéptido Inhibidor Gástrico/farmacología , Técnicas de Silenciamiento del Gen , Péptido 1 Similar al Glucagón/metabolismo , Péptido 1 Similar al Glucagón/farmacología , Humanos , Incretinas/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Canal de Potasio Kv1.5/genética , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional , Canales de Potasio de la Superfamilia Shaker
9.
Transplantation ; 95(1): 94-9, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23192157

RESUMEN

BACKGROUND: Allograft rejection is one of the main obstacles for islet transplantation. B7-H4 plays a key role in maintaining T-cell homeostasis by reducing T-cell proliferation and cytokine production. In this study, we investigated whether the endogenous expression of B7-H4 in ß cells from B7-H4 transgenic mice enhances islet allograft survival. METHODS: B7-H4 transgenic C57BL/6 (B6) mice (RIP.B7-H4) were developed by inserting the entire B7-H4 open reading frame under the rat insulin promoter (RIP). B7-H4 protein expression was examined by flow cytometric analysis and immunohistochemical staining. Islet allograft survival was investigated in streptozotocin-induced diabetic recipient BALB/c (H-2d) mice transplanted with 400 islets from RIP.B7-H4 (H-2b) mice under the kidney capsule. The recipient control group received islets from wild-type B6 donors. RESULTS: B7-H4 protein was significantly up-regulated in isolated islets from RIP.B7-H4 compared with wild-type B6 mice (56%±23% vs. 3%±1.2%). B7-H4 was coexpressed with insulin, but not glucagon, suggesting that B7-H4 is expressed in a ß-cell-specific manner. Recipient BALB/c mice transplanted with RIP.B7-H4 islets established euglycemia for 42.3±18.4 days (mean±SD; n=9) compared with controls at 23.1±7.8 days (mean±SD; n=12; P<0.004, log-rank test). CONCLUSIONS: The endogenous expression of B7-H4 in donor ß cells from transgenic mice prolongs islet allograft survival, confirming the negative role of B7-H4 in regulating alloreactive T-cell responses.


Asunto(s)
Supervivencia de Injerto , Trasplante de Islotes Pancreáticos , Inhibidor 1 de la Activación de Células T con Dominio V-Set/fisiología , Animales , Antígenos CD28/fisiología , Antígeno CTLA-4/fisiología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Linfocitos T/inmunología , Trasplante Homólogo
10.
Islets ; 4(4): 284-95, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22878670

RESUMEN

Costimulation blockade is an effective way to prevent allograft rejection. In this study, we tested the efficacy of two negative co-signaling molecules in protecting islet allograft function. We used local expression of B7-H4 by adenoviral transduction of islets (Ad-B7-H4) and systemic administration of CTLA-4.Ig to investigate the outcomes of allograft survival. Five groups of streptozotocin-induced diabetic C57BL/6 mice received 400 islets each from BALB/c donors. The groups consisted of control (G1); CTLA-4.Ig (G2); Ad-LacZ (G3); Ad-B7-H4 (G4); and Ad-B7-H4 and CTLA-4.Ig combined (G5). G1 and G3 developed graft failure on average of two weeks. G2, G4 and G5 survived for 43.8 ± 34.8, 54.7 ± 31.2 and 77.8 ± 21.5 d, respectively. Activated T and B cells in the lymph nodes were significantly controlled by CTLA-4.Ig treatment. Significantly reduced infiltrates were also detected in the allografts of G2 compared with G1. By contrast, B7-H4 significantly inhibited Th1-associated IFN-gamma secretion in the early stage and increased Foxp3 (+) T cells in the long-term surviving allografts. Our study suggests that CTLA-4 and B7-H4 inhibit alloimmune responses through distinct mechanisms, and that combination therapy which activates two negative co-signaling pathways can further enhance islet allograft survival.


Asunto(s)
Antígeno CTLA-4/inmunología , Supervivencia de Injerto/inmunología , Trasplante de Islotes Pancreáticos , Transducción de Señal , Inhibidor 1 de la Activación de Células T con Dominio V-Set/metabolismo , Análisis de Varianza , Animales , Linfocitos B/efectos de los fármacos , Antígenos CD28/metabolismo , Antígeno CTLA-4/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/cirugía , Femenino , Factores de Transcripción Forkhead/metabolismo , Supervivencia de Injerto/efectos de los fármacos , Inmunoglobulinas/farmacología , Insulina/metabolismo , Secreción de Insulina , Interferón gamma/metabolismo , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Estadísticas no Paramétricas , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Células TH1/metabolismo , Factores de Tiempo , Transducción Genética , Trasplante Homólogo , Inhibidor 1 de la Activación de Células T con Dominio V-Set/genética
11.
Am J Pathol ; 181(4): 1296-305, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22902430

RESUMEN

Islet transplantation provides a promising approach for treatment of type 1 diabetes mellitus. Amyloid formation and loss of extracellular matrix are two nonimmune factors contributing to death of isolated human islets. We tested the effects of two types of three-dimensional scaffolds, collagen matrix (CM) and fibroblast-populated collagen matrix (FPCM), on amyloid formation, viability, and function of isolated islets. Islets from cadaveric donors were cultured in FPCM, CM, or two-dimensional plate (2D) for 7 days. After 7 days, compared with the 2D culture condition, CM and FPCM markedly reduced amyloid formation of cultured islets and decreased apoptotic ß-cell rate by ∼75%. IL-1ß and Fas levels were also reduced in scaffold-embedded islets. Furthermore, ß/α cell ratios were increased by ∼18% and ∼36% in CM- and FPCM-embedded islets, respectively. Insulin content and insulin response to elevated glucose were also enhanced by both three-dimensional scaffolds. Moreover, culture in CM and FPCM (but not 2D) preserved insulin, GLUT-2, and PDX-1 mRNA expression. FPCM-embedded islets had significantly higher insulin response and lower amyloid formation than CM-embedded islets. These findings suggest that three-dimensional scaffolds reduce amyloid formation and improve viability and function of human islets in vitro, and that CM and fibroblasts have additive effects in enhancing islet function and reducing amyloid formation. Using this strategy is likely to improve outcome in human islet transplantation.


Asunto(s)
Amiloide/metabolismo , Islotes Pancreáticos/metabolismo , Técnicas de Cultivo de Tejidos/métodos , Andamios del Tejido/química , Supervivencia Tisular , Apoptosis , Caspasa 3/metabolismo , Recuento de Células , Activación Enzimática , Regulación de la Expresión Génica , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Glucagón/patología , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/patología , Interleucina-1beta/metabolismo , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Receptor fas/metabolismo
12.
Diabetes ; 61(8): 2016-29, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22740171

RESUMEN

Diabetes is a chronic debilitating disease that results from insufficient production of insulin from pancreatic ß-cells. Islet cell replacement can effectively treat diabetes but is currently severely limited by the reliance upon cadaveric donor tissue. We have developed a protocol to efficiently differentiate commercially available human embryonic stem cells (hESCs) in vitro into a highly enriched PDX1+ pancreatic progenitor cell population that further develops in vivo to mature pancreatic endocrine cells. Immature pancreatic precursor cells were transplanted into immunodeficient mice with streptozotocin-induced diabetes, and glycemia was initially controlled with exogenous insulin. As graft-derived insulin levels increased over time, diabetic mice were weaned from exogenous insulin and human C-peptide secretion was eventually regulated by meal and glucose challenges. Similar differentiation of pancreatic precursor cells was observed after transplant in immunodeficient rats. Throughout the in vivo maturation period hESC-derived endocrine cells exhibited gene and protein expression profiles that were remarkably similar to the developing human fetal pancreas. Our findings support the feasibility of using differentiated hESCs as an alternative to cadaveric islets for treating patients with diabetes.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/trasplante , Células Secretoras de Insulina/citología , Páncreas/citología , Animales , Línea Celular , Diabetes Mellitus Experimental/terapia , Proteínas de Homeodominio/biosíntesis , Humanos , Insulina/uso terapéutico , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Páncreas/embriología , Proproteína Convertasas/biosíntesis , Ratas , Células Madre/citología , Transactivadores/biosíntesis
13.
Int J Endocrinol ; 2012: 414812, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22611393

RESUMEN

Islet cell transplantation offers a potential cure for type 1 diabetes, but it is challenged by insufficient donor tissue and side effects of current immunosuppressive drugs. Therefore, alternative sources of insulin-producing cells and isletfriendly immunosuppression are required to increase the efficiency and safety of this procedure. Beta cells can be transdifferentiated from precursors or another heterologous (non-beta-cell) source. Recent advances in beta cell regeneration from somatic cells such as fibroblasts could circumvent the usage of immunosuppressive drugs. Therefore, generation of patient-specific beta cells provides the potential of an evolutionary treatment for patients with diabetes.

14.
PLoS One ; 7(1): e28232, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22238573

RESUMEN

B7-H4 is a newly identified B7 homolog that plays an important role in maintaining T-cell homeostasis by inhibiting T-cell proliferation and lymphokine-secretion. In this study, we investigated the signal transduction pathways inhibited by B7-H4 engagement in mouse T cells. We found that treatment of CD3(+) T cells with a B7-H4.Ig fusion protein inhibits anti-CD3 elicited T-cell receptor (TCR)/CD28 signaling events, including phosphorylation of the MAP kinases, ERK, p38, and JNK. B7-H4.Ig treatment also inhibited the phosphorylation of AKT kinase and impaired its kinase activity as assessed by the phosphorylation of its endogenous substrate GSK-3. Expression of IL-2 is also reduced by B7-H4. In contrast, the phosphorylation state of the TCR proximal tyrosine kinases ZAP70 and lymphocyte-specific protein tyrosine kinase (LCK) are not affected by B7-H4 ligation. These results indicate that B7-H4 inhibits T-cell proliferation and IL-2 production through interfering with activation of ERK, JNK, and AKT, but not of ZAP70 or LCK.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteína Oncogénica v-akt/antagonistas & inhibidores , Linfocitos T/efectos de los fármacos , Inhibidor 1 de la Activación de Células T con Dominio V-Set/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Anticuerpos/metabolismo , Anticuerpos/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteína Oncogénica v-akt/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Transducción de Señal/efectos de los fármacos , Linfocitos T/metabolismo , Linfocitos T/fisiología , Inhibidor 1 de la Activación de Células T con Dominio V-Set/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
Cell Transplant ; 21(1): 99-111, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21929869

RESUMEN

Negative cosignaling molecules play an important role in regulating T-cell responses to alloantigen stimulation. We recently reported that adenoviral-mediated transduction of islet allografts with B7-H4 inhibits allograft rejection. In this study, we investigate the mechanism for B7-H4-induced prolongation of mouse islet allograft survival. Streptozotocin-induced diabetic C57BL/6 mice were rendered normoglycemic by renal subcapsular implants of B7-H4-transduced BALB/c islets. Grafts and spleens were removed after days 2, 10, and 60 (n = 8 each) for characterization of kinetics of Foxp3 and interleukin 10 (IL-10) expression. Mixed lymphocyte reaction (MLR) was done at day 60. Ten mice were subjected to nephrectomy at 60 days and then five were implanted with secondary BALB/c islets and five were given third-party CBA/J islets. An increase in Foxp3 and IL-10 mRNA expression was detected in recipients' spleens at day 60 and this was associated with increased quantities of Foxp3(+) cells. Splenocytes at day 60 showed hyporesponsiveness during MLR to alloantigen stimulation. Proliferation was partially restored after CD25(+) T-cell depletion. Secondary BALB/c islets survived for 79 ± 29 days compared with 21 ± 3.6 days for CBA/J islets (p < 0.001). Local expression of B7-H4 induces long-term unresponsiveness to donor-specific alloantigens, and is associated with T regulatory cells, suggesting the development of tolerance.


Asunto(s)
Diabetes Mellitus Experimental/cirugía , Supervivencia de Injerto , Trasplante de Islotes Pancreáticos/inmunología , Tolerancia al Trasplante , Inhibidor 1 de la Activación de Células T con Dominio V-Set/genética , Animales , Factores de Transcripción Forkhead/biosíntesis , Interleucina-10/biosíntesis , Islotes Pancreáticos/citología , Islotes Pancreáticos/inmunología , Prueba de Cultivo Mixto de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Nefrectomía , Linfocitos T Reguladores/inmunología , Transducción Genética , Trasplante Homólogo , Inhibidor 1 de la Activación de Células T con Dominio V-Set/inmunología
16.
Cell Tissue Bank ; 13(4): 569-75, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21818570

RESUMEN

It has been predicted that one of the greatest increase in prevalence of diabetes will happen in the Middle East bear in the next decades. The aim of standard therapeutic strategies for diabetes is better control of complications. In contrast, some new strategies like cell and gene therapy have aimed to cure the disease. In recent years, significant progress has occurred in beta-cell replacement therapies with a progressive improvement of short-term and long term outcomes. In year 2005, considering the impact of the disease in Iran and the promising results of the Edmonton protocol, the funding for establishing a current Good Manufacturing Practice (cGMP) islet processing facility by Endocrinology and Metabolism Research Center was approved by Tehran University of Medical Sciences. Several islet isolations were performed following establishment of cGMP facility and recruitment of all required equipments for process validation and experimental purpose. Finally the first successful clinical islet isolation and transplantation was performed in September 2010. In spite of a high cost of the procedure it is considered beneficial and may prevent long term complications and the costs associated with secondary cares. In this article we will briefly describe our experience in setting up a cGMP islet processing facility which can provide valuable information for regional countries interested to establish similar facilities.


Asunto(s)
Regulación y Control de Instalaciones , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Bancos de Tejidos/normas , Humanos , Irán , Islotes Pancreáticos/citología
17.
J Transplant ; 2011: 418902, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22028949

RESUMEN

Type 1 diabetes (T1D) is a chronic autoimmune disease and characterized by absolute insulin deficiency. ß-cell replacement by islet cell transplantation has been established as a feasible treatment option for T1D. The two main obstacles after islet transplantation are alloreactive T-cell-mediated graft rejection and recurrence of autoimmune diabetes mellitus in recipients. T cells play a central role in determining the outcome of both autoimmune responses and allograft survival. B7-H4, a newly identified B7 homolog, plays a key role in maintaining T-cell homeostasis by reducing T-cell proliferation and cytokine production. The relationship between B7-H4 and allograft survival/autoimmunity has been investigated recently in both islet transplantation and the nonobese diabetic (NOD) mouse models. B7-H4 protects allograft survival and generates donor-specific tolerance. It also prevents the development of autoimmune diabetes. More importantly, B7-H4 plays an indispensable role in alloimmunity in the absence of the classic CD28/CTLA-4 : B7 pathway, suggesting a synergistic/additive effect with other agents such as CTLA-4 on inhibition of unwanted immune responses.

18.
Diabetes ; 60(12): 3246-55, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21984581

RESUMEN

OBJECTIVE: Autoimmune diabetes is a T cell-mediated disease in which insulin-producing ß-cells are destroyed. Autoreactive T cells play a central role in mediating ß-cell destruction. B7-H4 is a negative cosignaling molecule that downregulates T-cell responses. In this study, we aim to determine the role of B7-H4 on regulation of ß-cell-specific autoimmune responses. RESEARCH DESIGN AND METHODS: Prediabetic (aged 3 weeks) female NOD mice (group 1, n = 21) were treated with intraperitoneal injections of B7-H4.Ig at 7.5 mg/kg, with the same amount of mouse IgG (group 2, n = 24), or with no protein injections (group 3, n = 24), every 3 days for 12 weeks. RESULTS: B7-H4.Ig reduced the incidence of autoimmune diabetes, compared with the control groups (diabetic mice 28.6% of group 1, 66.7% of group 2 [P = 0.0081], and 70.8% of group 3 [group 1 vs. 3, P = 0.0035]). Histological analysis revealed that B7-H4 treatment did not block islet infiltration but rather suppressed further infiltrates after 9 weeks of treatment (group 1 vs. 2, P = 0.0003). B7-H4 treatment also reduced T-cell proliferation in response to GAD65 stimulation ex vivo. The reduction of diabetes is not due to inhibition of activated T cells in the periphery but rather to a transient increase of Foxp3(+) CD4(+) T-cell population at one week posttreatment (12.88 ± 1.29 vs. 11.58 ± 1.46%; n = 8; P = 0.03). CONCLUSIONS: Our data demonstrate the protective role of B7-H4 in the development of autoimmune diabetes, suggesting a potential means of preventing type 1 diabetes by targeting the B7-H4 pathway.


Asunto(s)
Diabetes Mellitus Tipo 1/prevención & control , Inhibidor 1 de la Activación de Células T con Dominio V-Set/uso terapéutico , Animales , Autoinmunidad/efectos de los fármacos , Femenino , Factores de Transcripción Forkhead/metabolismo , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ratones , Ratones Endogámicos NOD , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Estado Prediabético/tratamiento farmacológico , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
19.
J Cell Physiol ; 226(7): 1813-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21506112

RESUMEN

Islet transplantation represents a viable treatment for type 1 diabetes. However, due to loss of substantial mass of islets early after transplantation, islets from two or more donors are required to achieve insulin independence. Islet-extracellular matrix disengagement, which occurs during islet isolation process, leads to subsequent islet cell apoptosis and is an important contributing factor to early islet loss. In this study, we developed a fibroblast populated collagen matrix (FPCM) as a novel scaffold to improve islet cell viability and function post-transplantation. FPCM was developed by embedding fibroblasts within type-I collagen and used as scaffold for islet grafts. Viability and insulin secretory function of islets embedded within FPCM was evaluated in vitro and in a syngeneic murine islet transplantation model. Islets embedded within acellular matrix or naked islets were used as control. Islet cell survival and function was markedly improved particularly after embedding within FPCM. The composite scaffold significantly promoted islet isograft survival and reduced the critical islet mass required for diabetes reversal by half (from 200 to 100 islets per recipient). Fibroblast embedded within FPCM produced fibronectin and growth factors and induced islet cell proliferation. No evidence of fibroblast over-growth within composite grafts was noticed. These results confirm that FPCM significantly promotes islet viability and functionality, enhances engraftment of islet grafts and decreases the critical islet mass needed to reverse hyperglycemia. This promising finding offers a new approach to reducing the number of islet donors per recipient and improving islet transplant outcome.


Asunto(s)
Colágeno Tipo I/metabolismo , Diabetes Mellitus Experimental/cirugía , Fibroblastos/trasplante , Supervivencia de Injerto , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/cirugía , Andamios del Tejido , Animales , Apoptosis , Glucemia/metabolismo , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Insulina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Tiempo , Técnicas de Cultivo de Tejidos
20.
Transplantation ; 91(3): 373-8, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21258272

RESUMEN

BACKGROUND: The effect of islet cell transplantation (ICT) on the progression of diabetic microvascular complications is not well understood. METHODS: We have conducted a prospective, crossover, cohort study comparing ICT with intensive medical therapy on the progression of diabetic nephropathy, retinopathy, and neuropathy. RESULTS: The rate of decline in glomerular filtration rate is slower after ICT than on medical therapy. There was significantly more progression of retinopathy in medically treated patients than post-ICT. There was a nonsignificant trend for improved nerve conduction velocity post-ICT. CONCLUSIONS: ICT is associated with less progression of microvascular complications than intensive medical therapy. Multicenter, randomized trials are needed to further study the role of ICT in slowing the progression of diabetic complications.


Asunto(s)
Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/terapia , Angiopatías Diabéticas/etiología , Angiopatías Diabéticas/prevención & control , Progresión de la Enfermedad , Insulina/uso terapéutico , Trasplante de Islotes Pancreáticos , Adulto , Estudios de Cohortes , Estudios Cruzados , Angiopatías Diabéticas/fisiopatología , Nefropatías Diabéticas/epidemiología , Neuropatías Diabéticas/epidemiología , Retinopatía Diabética/epidemiología , Femenino , Humanos , Hipoglucemiantes/uso terapéutico , Incidencia , Masculino , Microvasos/fisiopatología , Persona de Mediana Edad , Estudios Prospectivos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...