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

RESUMO

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.


Assuntos
Amiloide/metabolismo , Células Secretoras de Insulina/metabolismo , Interleucina-1beta/metabolismo , Transdução de Sinais , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Diabetes Mellitus Tipo 2/terapia , Feminino , Humanos , Células Secretoras de Insulina/patologia , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Transplante das Ilhotas Pancreáticas , Masculino , Camundongos , Camundongos Transgênicos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt
2.
Mol Metab ; 6(8): 833-844, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28752047

RESUMO

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.


Assuntos
Amiloide/metabolismo , Apoptose , Células Secretoras de Insulina/metabolismo , Proteína Antagonista do Receptor de Interleucina 1/metabolismo , Interleucina-1beta/metabolismo , Adolescente , Adulto , Animais , Caspase 8/metabolismo , Linhagem Celular , Células Cultivadas , Proteína Ligante Fas/metabolismo , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Pessoa de Meia-Idade
3.
Diabetes Obes Metab ; 19(5): 682-694, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28058779

RESUMO

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.


Assuntos
Amiloide/agonistas , Apoptose , Interleucina-1beta/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/metabolismo , Receptor fas/agonistas , Adulto , Amiloide/antagonistas & inibidores , Amiloide/química , Amiloide/metabolismo , Animais , Cadáver , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Diabetes Mellitus Tipo 2/cirurgia , Hemizigoto , Humanos , Insulina/metabolismo , Secreção de Insulina , Proteína Antagonista do Receptor de Interleucina 1/genética , Proteína Antagonista do Receptor de Interleucina 1/metabolismo , Interleucina-1beta/antagonistas & inibidores , Interleucina-1beta/genética , Polipeptídeo Amiloide das Ilhotas Pancreáticas/antagonistas & inibidores , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/patologia , Transplante das Ilhotas Pancreáticas/efeitos adversos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Precursores de Proteínas/antagonistas & inibidores , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Interferência de RNA , Proteínas Recombinantes/metabolismo , Técnicas de Cultura de Tecidos , Receptor fas/metabolismo
4.
Diabetologia ; 57(4): 765-75, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24442508

RESUMO

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.


Assuntos
Amiloide/toxicidade , Caspase 8/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/enzimologia , Ilhotas Pancreáticas/citologia , Adulto , Animais , Caspase 3/metabolismo , Caspase 8/genética , Feminino , Humanos , Técnicas In Vitro , Masculino , Camundongos , Pessoa de Meia-Idade
5.
Pancreas ; 43(1): 128-34, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24326367

RESUMO

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.


Assuntos
Adenocarcinoma/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , Neoplasias Pancreáticas/metabolismo , Inibidor 1 da Ativação de Células T com Domínio V-Set/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patologia , Western Blotting , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Linhagem Celular , Linhagem Celular Tumoral , Diabetes Mellitus Tipo 1/genética , Expressão Gênica , Humanos , Imuno-Histoquímica , Insulina/genética , Insulina/metabolismo , Insulinoma/genética , Insulinoma/patologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Inibidor 1 da Ativação de Células T com Domínio V-Set/genética
6.
Biochem J ; 451(2): 227-34, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23390957

RESUMO

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.


Assuntos
Proteína de Ligação a CREB/metabolismo , Incretinas/farmacologia , Células Secretoras de Insulina/metabolismo , Canal de Potássio Kv1.5/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Sirtuína 1/metabolismo , Acetilação/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Proteína de Ligação a CREB/genética , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Polipeptídeo Inibidor Gástrico/metabolismo , Polipeptídeo Inibidor Gástrico/farmacologia , Técnicas de Silenciamento de Genes , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Humanos , Incretinas/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Canal de Potássio Kv1.5/genética , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional , Superfamília Shaker de Canais de Potássio
7.
Islets ; 4(4): 284-95, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22878670

RESUMO

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.


Assuntos
Antígeno CTLA-4/imunologia , Sobrevivência de Enxerto/imunologia , Transplante das Ilhotas Pancreáticas , Transdução de Sinais , Inibidor 1 da Ativação de Células T com Domínio V-Set/metabolismo , Análise de Variância , Animais , Linfócitos B/efeitos dos fármacos , Antígenos CD28/metabolismo , Antígeno CTLA-4/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/cirurgia , Feminino , Fatores de Transcrição Forkhead/metabolismo , Sobrevivência de Enxerto/efeitos dos fármacos , Imunoglobulinas/farmacologia , Insulina/metabolismo , Secreção de Insulina , Interferon gama/metabolismo , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Estatísticas não Paramétricas , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Células Th1/metabolismo , Fatores de Tempo , Transdução Genética , Transplante Homólogo , Inibidor 1 da Ativação de Células T com Domínio V-Set/genética
8.
Am J Pathol ; 181(4): 1296-305, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22902430

RESUMO

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.


Assuntos
Amiloide/metabolismo , Ilhotas Pancreáticas/metabolismo , Técnicas de Cultura de Tecidos/métodos , Alicerces Teciduais/química , Sobrevivência de Tecidos , Apoptose , Caspase 3/metabolismo , Contagem de Células , Ativação Enzimática , Regulação da Expressão Gênica , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Glucagon/patologia , Transportador de Glucose Tipo 2/genética , Transportador de Glucose Tipo 2/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/patologia , Interleucina-1beta/metabolismo , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transativadores/genética , Transativadores/metabolismo , Receptor fas/metabolismo
9.
PLoS One ; 7(1): e28232, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22238573

RESUMO

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.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteína Oncogênica v-akt/antagonistas & inibidores , Linfócitos T/efeitos dos fármacos , Inibidor 1 da Ativação de Células T com Domínio V-Set/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Animais , Anticorpos/metabolismo , Anticorpos/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Proteína Oncogênica v-akt/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/metabolismo , Linfócitos T/fisiologia , Inibidor 1 da Ativação de Células T com Domínio V-Set/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Cell Transplant ; 21(1): 99-111, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21929869

RESUMO

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.


Assuntos
Diabetes Mellitus Experimental/cirurgia , Sobrevivência de Enxerto , Transplante das Ilhotas Pancreáticas/imunologia , Tolerância ao Transplante , Inibidor 1 da Ativação de Células T com Domínio V-Set/genética , Animais , Fatores de Transcrição Forkhead/biossíntese , Interleucina-10/biossíntese , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/imunologia , Teste de Cultura Mista de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Nefrectomia , Linfócitos T Reguladores/imunologia , Transdução Genética , Transplante Homólogo , Inibidor 1 da Ativação de Células T com Domínio V-Set/imunologia
11.
Diabetes ; 60(1): 239-47, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20971966

RESUMO

OBJECTIVE: Differentiation of human embryonic stem (hES) cells to fully developed cell types holds great therapeutic promise. Despite significant progress, the conversion of hES cells to stable, fully differentiated endocrine cells that exhibit physiologically regulated hormone secretion has not yet been achieved. Here we describe an efficient differentiation protocol for the in vitro conversion of hES cells to functional glucagon-producing α- cells. RESEARCH DESIGN AND METHODS: Using a combination of small molecule screening and empirical testing, we developed a six-stage differentiation protocol for creating functional α-cells. An extensive in vitro and in vivo characterization of the differentiated cells was performed. RESULTS: A high rate of synaptophysin expression (>75%) and robust expression of glucagon and the α-cell transcription factor ARX was achieved. After a transient polyhormonal state in which cells coexpress glucagon and insulin, maturation in vitro or in vivo resulted in depletion of insulin and other ß-cell markers with concomitant enrichment of α-cell markers. After transplantation, these cells secreted fully processed, biologically active glucagon in response to physiologic stimuli including prolonged fasting and amino acid challenge. Moreover, glucagon release from transplanted cells was sufficient to reduce demand for pancreatic glucagon, resulting in a significant decrease in pancreatic α-cell mass. CONCLUSIONS: These results indicate that fully differentiated pancreatic endocrine cells can be created via stepwise differentiation of hES cells. These cells may serve as a useful screening tool for the identification of compounds that modulate glucagon secretion as well as those that promote the transdifferentiation of α-cells to ß-cells.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/transplante , Células Secretoras de Glucagon/citologia , Glucagon/metabolismo , Animais , Glicemia/metabolismo , Linhagem Celular , DNA/análise , DNA/genética , Células-Tronco Embrionárias/metabolismo , Glucagon/análise , Células Secretoras de Glucagon/metabolismo , Humanos , Imuno-Histoquímica , Insulina/análise , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Proglucagon/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Gastroenterology ; 138(5): 1966-75, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20138041

RESUMO

BACKGROUND & AIMS: Glucose-dependent insulinotropic polypeptide (GIP) and the proglucagon product glucagon-like peptide-1 (GLP-1) are gastrointestinal hormones that are released in response to nutrient intake and promote insulin secretion. Interestingly, a subset of enteroendocrine cells express both GIP and GLP-1. We sought to determine whether GIP also might be co-expressed with proglucagon in pancreatic alpha-cells. METHODS: We assessed GIP expression via reverse-transcription polymerase chain reaction, in situ hybridization, and immunohistochemistry. We developed a novel bioassay to measure GIP release from isolated islets, compared the biological activities of full-length and truncated GIP, and assessed the impact of immunoneutralization of islet GIP on glucose-stimulated insulin secretion in isolated islets. RESULTS: GIP messenger RNA was present in mouse islets; GIP protein localized to islet alpha-cells of mouse, human, and snake pancreas, based on immunohistochemical analyses. However, using a C-terminal GIP antibody, immunoreactivity was detected in islets from prohormone convertase (PC) 2 knockout but not wild-type mice. Bioactive GIP was secreted from mouse and human islets after arginine stimulation. In the perfused mouse pancreas, GIP(1-42) and amidated GIP(1-30) had equipotent insulinotropic actions. Finally, immunoneutralization of GIP secreted by isolated islets decreased glucose-stimulated insulin secretion. CONCLUSIONS: GIP is expressed in and secreted from pancreatic islets; in alpha-cells, PC2 processes proGIP to yield a truncated but bioactive form of GIP that differs from the PC1/3-derived form from K-cells. Islet-derived GIP promotes islet glucose competence and also could support islet development and/or survival.


Assuntos
Polipeptídeo Inibidor Gástrico/metabolismo , Células Secretoras de Glucagon/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Animais , Boidae , Linhagem Celular , Duodeno/metabolismo , Feminino , Polipeptídeo Inibidor Gástrico/genética , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Humanos , Secreção de Insulina , Ilhotas Pancreáticas/embriologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fragmentos de Peptídeos/metabolismo , Proglucagon/metabolismo , Pró-Proteína Convertase 2/deficiência , Pró-Proteína Convertase 2/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores dos Hormônios Gastrointestinais/genética , Receptores dos Hormônios Gastrointestinais/metabolismo , Fatores de Tempo , Técnicas de Cultura de Tecidos , Transfecção
13.
J Biol Chem ; 284(44): 30372-82, 2009 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-19748889

RESUMO

Glucose-dependent insulinotropic polypeptide (GIP) potentiates glucose-stimulated insulin secretion, insulin biosynthesis, and beta-cell proliferation and survival. In previous studies GIP was shown to promote beta-cell survival by modulating the activity of multiple signaling modules and regulating gene transcription of pro- and anti-apoptotic bcl-2 family proteins. We have now evaluated the mechanisms by which GIP regulates the dynamic interactions between cytoplasmic bcl-2 family members and the mitochondria in INS-1 cells during apoptosis induced by treatment with staurosporine (STS), an activator of the mitochondria-mediated apoptotic pathway. STS induced translocation of bad and bimEL, activation of mitochondrial bax, release of mitochondrial cytochrome c, cleavage of caspase-3, and apoptosis. Each response was significantly diminished by GIP. Using selective enzyme inhibitors, overexpression of dominant-negative Akt, and Akt siRNA, it was demonstrated that GIP promoted beta-cell survival via Akt-dependent suppression of p38 MAPK and JNK and that combined inhibition was sufficient to explain the entire pro-survival responses to GIP during STS treatment. This signaling pathway also explained the pro-survival effects of GIP on INS-1 cells exposed to two other promoters of stress: thapsigargin (endoplasmic reticulum stress) and etoposide (genotoxic stress). Importantly, we discovered that GIP suppressed p38 MAPK and JNK via Akt-mediated changes in the phosphorylation state of the apoptosis signal-regulating kinase 1 in INS-1 cells and human islets, resulting in inhibition of its activity. Inhibition of apoptosis by GIP is therefore mediated via a key pathway involving Akt-dependent inhibition of apoptosis signal-regulating kinase 1, which subsequently prevents the pro-apoptotic actions of p38 MAPK and JNK.


Assuntos
Polipeptídeo Inibidor Gástrico/fisiologia , Células Secretoras de Insulina/citologia , MAP Quinase Quinase Quinase 5/antagonistas & inibidores , Proteína Quinase 8 Ativada por Mitógeno/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Apoptose , Sobrevivência Celular , Células Cultivadas , Humanos , Ilhotas Pancreáticas/citologia , Transdução de Sinais/fisiologia
14.
Life Sci ; 85(15-16): 549-56, 2009 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-19747492

RESUMO

Type 1 diabetes (T1D) is the result of the autoimmune response against pancreatic insulin-producing ss-cells. Its ultimate consequence is beta-cell insufficiency-mediated dysregulation of blood glucose control. In terms of T1D treatment, immunotherapy addresses the cause of T1D, mainly through re-setting the balance between autoimmunity and regulatory mechanisms. Regulatory T cells play an important role in this immune intervention. An alternative T1D treatment is beta-cell replacement, which can reverse the consequence of the disease by replacing destroyed beta-cells in the diabetic pancreas. The applicable insulin-producing cells can be directly obtained from islet transplantation or generated from other cell sources such as autologous adult stem cells, embryonic stem cells, and induced pluripotent stem cells. In this review, we summarize the recent research progress and analyze the possible advantages and disadvantages of these two therapeutic options especially focusing on the potential synergistic effect on T1D treatment. Exploring the optimal combination of immunotherapy and beta-cell replacement will pave the way to the most effective cure for this devastating disease.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Imunoterapia/métodos , Células Secretoras de Insulina/transplante , Terapia Combinada , Diabetes Mellitus Tipo 1/imunologia , Humanos
15.
Transplantation ; 87(4): 482-90, 2009 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-19307783

RESUMO

BACKGROUND: Allogeneic pancreatic islet transplantation has the potential to cure type 1 diabetes. One of the barriers to islet transplantation is the alloreactive T-cell response between donors and recipients. Costimulatory molecules, which play a major role in the regulation of the immune response to antigens during graft rejection, may be used to inhibit allograft destruction. B7-H4 is one such member in the costimulatory family, which has established negative regulatory function of T-cell responses. METHODS: To determine whether local expression of B7-H4 protein can protect beta cells from damage in islet allotransplantation, we have constructed a recombinant adenovirus expressing a B7-H4 complementary deoxyribonucleic acid (Ad-B7-H4). To study the in vivo effects of B7-H4 expression on islet graft survival, adenovirus-transduced islets from donor Balb/c mice were transplanted into streptozotocin-diabetic C57BL/6 mice (n=12). RESULTS: Expression of B7-H4 in islets by Ad-B7-H4 transduction at an optimized condition did not inhibit glucose-stimulated insulin secretion of the treated islets. The recipient mice transplanted with Ad-B7-H4-transduced islets established euglycemia for a longer time (mean 56.5 days), compared with control mice transplanted with Ad-LacZ-transduced islets (mean 14.5 days, [n=12, P<0.001]). Splenocytes isolated from the recipients of Ad-B7-H4-transduced islets showed hyporesponsiveness to alloantigenic stimulation, compared with control recipients. CD45 and insulin staining of the graft transplanted with Ad-B7-H4-transduced islets indicated the preservation of beta cells and decrease of infiltrating immune cells. CONCLUSIONS: Local expression of B7-H4 prolongs islet allograft survival in vivo, suggesting translational potential for beta-cell replacement with reduced immune injury.


Assuntos
Antígeno B7-1/genética , Sobrevivência de Enxerto/fisiologia , Transplante das Ilhotas Pancreáticas/métodos , Transplante das Ilhotas Pancreáticas/fisiologia , Transplante Homólogo/fisiologia , Adenoviridae/genética , Animais , Glicemia/metabolismo , Clonagem Molecular , Feminino , Imuno-Histoquímica , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/fisiologia , Células Secretoras de Insulina/transplante , Ilhotas Pancreáticas/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , RNA/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Inibidor 1 da Ativação de Células T com Domínio V-Set
16.
Biochim Biophys Acta ; 1790(6): 566-74, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19264107

RESUMO

BACKGROUND: Amyloid fibrils created by misfolding and aggregation of proteins are a major pathological feature in a variety of degenerative diseases. Therapeutic approaches including amyloid vaccines and anti-aggregation compounds in models of amyloidosis point to an important role for amyloid in disease pathogenesis. Amyloid deposits derived from the beta-cell peptide islet amyloid polypeptide (IAPP or amylin) are a characteristic of type 2 diabetes and may contribute to loss of beta-cells in this disease. METHODS: We developed a cellular model of rapid amyloid deposition using cultured human islets and observed a correlation between fibril accumulation and beta-cell death. A series of overlapping peptides derived from IAPP was generated. RESULTS: A potent inhibitor (ANFLVH) of human IAPP aggregation was identified. This inhibitory peptide prevented IAPP fibril formation in vitro and in human islet cultures leading to a striking increase in islet cell viability. CONCLUSIONS: These findings indicate an important contribution of IAPP aggregation to beta-cell death in situ and point to therapeutic applications for inhibitors of IAPP aggregation in enhancing beta-cell survival. GENERAL SIGNIFICANCE: Anti-amyloid compounds could potentially reduce the loss of beta-cell mass in type 2 diabetes and maintain healthy human islet cultures for beta-cell replacement therapies.


Assuntos
Amiloide/antagonistas & inibidores , Sobrevivência Celular/fisiologia , Ilhotas Pancreáticas , Sequência de Aminoácidos , Amiloide/metabolismo , Animais , Apoptose/fisiologia , Células Cultivadas , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Ilhotas Pancreáticas/fisiologia , Ilhotas Pancreáticas/ultraestrutura , Dados de Sequência Molecular
17.
World J Surg ; 31(8): 1569-76, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17562107

RESUMO

Canadian surgical contributions to the field of islet transplantation have a rich heritage and a promising future. In this article, some seminal Canadian contributions to this field are reviewed, including contributions at the basic research laboratory and translational applications to bedside therapy of type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/cirurgia , Transplante das Ilhotas Pancreáticas , Animais , Canadá , Humanos , Transplante das Ilhotas Pancreáticas/métodos , Ratos
18.
Transplantation ; 83(1): 24-8, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17220786

RESUMO

BACKGROUND: Islet transplantation can reduce or eliminate the need for insulin in patients with type 1 diabetes. Exenatide is a long acting analogue of Glucagon-like peptide-1 (GLP-1) that augments glucose induced insulin secretion, and may increase beta cell mass. We evaluated the effect of exenatide on insulin secretion after islet transplantation. METHODS: Eleven C-peptide positive islet cell recipients with elevated glucose levels were treated with exenatide for three months. Response was assessed by insulin requirements, meal tolerance tests, and hyperglycemic glucose clamps. RESULTS: Ten patients responded to exenatide. Two patients who had not restarted insulin achieved good glycemic control and one patient who had received 5500 IE/kg in first islet infusion was able to stop insulin. Seven other patients decreased their insulin dose by 39% on exenatide. Hyperglycemic clamp studies showed a rise in second phase insulin release (before exenatide: 246+/-88 pM; during exenatide: 644+/-294 pM, P<0.01). Meal tolerance studies before and one month after stopping exenatide did not show a difference in glucose or C-peptide values. Nausea and vomiting were the major side effects. CONCLUSIONS: Exenatide stimulates insulin secretion in islet transplant recipients. It reduces insulin dose in some patients and may delay the need to resume insulin in others. We did not find any evidence of a trophic effect on islets.


Assuntos
Diabetes Mellitus Tipo 1/cirurgia , Hipoglicemiantes/uso terapêutico , Transplante das Ilhotas Pancreáticas/fisiologia , Peptídeos/uso terapêutico , Peçonhas/uso terapêutico , Adulto , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Diabetes Mellitus Tipo 1/tratamento farmacológico , Relação Dose-Resposta a Droga , Exenatida , Feminino , Técnica Clamp de Glucose , Humanos , Injeções Subcutâneas , Insulina/uso terapêutico , Masculino , Pessoa de Meia-Idade , Peptídeos/administração & dosagem , Peçonhas/administração & dosagem
19.
Cell Transplant ; 15(5): 399-410, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16970282

RESUMO

B7-H4, a recently described member of the B7 family of cosignal molecules, is thought to be involved in the regulation of cellular and humoral immune responses through receptors on activated T and B cells. Human islet cells express positive B7-H4 mRNA in RT-PCR assays, but not B7-H4 protein on cell surface in flow cytometric analyses. To investigate the regulatory effects of activation of the B7-H4 pathway on the function of activated T cells of patients with type 1 diabetes (T1D), we have used our in vitro human experimental system, including human beta-cell antigen-specific T-cell clones and human beta-cell lines CM and HP62, as well as primary islet cells. B7-H4.Ig protein was purified from the culture supernatant of 293T cells transfected by a B7-H4.Ig plasmid (pMIgV, containing a human B7-H4 cDNA and a mouse IgG2a Fc cDNA). Our preliminary studies showed that immobilized fusion protein human B7-H4.Ig (coated with 5 microg/ml for 2 h at 37 degrees C), but not control Ig, clearly inhibited the proliferation of activated CD4+ and CD8+ T cells of patients induced by anti-CD3 antibody in CFSE assays. B7-H4.Ig also arrested cell cycle progression of T cells in G0/G1 phase and induced T-cell apoptosis as measured by BrdU-7-AAD flow cytometric analysis. To determine the cytoprotective effects of B7-H4, we developed transfectants of human beta-cell lines CM and HP62 and islet cells transfected with the B7-H4.Ig plasmid, using empty vector transfectants as controls. The results demonstrate that cell-associated B7-H4.Ig expressed on human beta-cells clearly inhibits the cytotoxicity of the T-cell clones to targeted human beta-cells in 51Cr release cytotoxicity assays. Activation of the B7-H4 pathway may represent a novel immunotherapeutic approach to inhibit T-cell responses for the prevention of beta-cell destruction in T1D.


Assuntos
Antígeno B7-1/metabolismo , Células Secretoras de Insulina/imunologia , Linfócitos T/imunologia , Adolescente , Adulto , Anticorpos Monoclonais/farmacologia , Apoptose/efeitos dos fármacos , Antígeno B7-1/genética , Antígeno B7-1/imunologia , Complexo CD3/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Citotoxicidade Imunológica/efeitos dos fármacos , Feminino , Humanos , Imunoglobulinas/metabolismo , Imunoglobulinas/fisiologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Interferon gama/metabolismo , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/citologia , Linfócitos T/metabolismo , Inibidor 1 da Ativação de Células T com Domínio V-Set
20.
Arch Surg ; 140(8): 735-44, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16103282

RESUMO

HYPOTHESIS: A local multiorgan donor pancreas procurement program can provide a source for optimized isolation of purified viable islets for transplantation into patients with type 1 diabetes mellitus receiving best medical therapy. DESIGN: Prospective before-after cohort study. SETTING: Tertiary referral center. PATIENTS: Glycemic control was assessed in 10 patients with diabetes-induced renal dysfunction who were enrolled in a best medical therapy program and then crossed over to islet transplantation. INTERVENTIONS: Thirty human pancreata were retrieved from local multiorgan donors and consecutively processed with intraductal collagenase perfusion, continuous digestion, and density gradient purification (group 1, n = 9) or similarly processed but impure tissue fractions cultured in vitro and then repurified to retrieve additional islets (group 2, n = 21). Islets were implanted by percutaneous portal embolization, providing more than 10 000 islet equivalents (IE) per kilogram of body weight (infusions from 1-3 donors per patient) under cover of antithymocyte globulin, sirolimus, or mycophenolate mofetil and tacrolimus. MAIN OUTCOME MEASURES: Islet yields, purity, and cell viability (caspase 3, terminal deoxynucleotidyl transferase-mediated biotin-deoxyuridine 5-triphosphate nick-end labeling stain, and insulin secretion in vitro) were compared. In patients, monitored metabolic parameters were C-peptide secretion, insulin requirements, glycemic excursion, and hemoglobin A(1c) (HbA(1c)). RESULTS: For group 1 vs group 2, no differences were observed in pancreas age (43 vs 44 years), cold storage (5 vs 4 hours), or weight (73 vs 82 g). Group 2 yielded 453 690 IE vs 214 109 IE in group 1 (P = .002). Grafts contained 50% or more endocrine cells in both groups. No difference occurred in cell viability or insulin secretion. Islets from 90% of group 2 pancreata met release criteria for transplantation. C-peptide secretion was detected in all recipients and persisted with a median follow-up to 12 months (range, 6-21 months) after full islet transplantation. Daily insulin dependence was reversed in all patients for at least 3 months. Five patients resumed small insulin doses. Compared with the best care program, all patients had improved metabolic stability. The mean +/- SE HbA(1c) level at entry into the study was 7.8% +/- 0.5%, and this decreased to 6.9% +/- 0.2% after best care (P = .38) and further to 6.2% +/- 0.2% at 6 months after transplantation (P = .002 vs entry; P = .15 vs best care; analysis of variance). CONCLUSIONS: Local pancreas donor retrieval with islet isolation and culture conditioning enabled an offer of islets for transplantation for 90% of consecutively processed pancreata. Isolated islets secreted insulin during prolonged follow-up after implantation into patients, yielding metabolic control comparable with that achieved by best medical therapy.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/cirurgia , Insulina/uso terapêutico , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/citologia , Imunologia de Transplantes/fisiologia , Adulto , Análise de Variância , Glicemia/análise , Estudos de Coortes , Diabetes Mellitus Tipo 1/diagnóstico , Feminino , Seguimentos , Rejeição de Enxerto , Sobrevivência de Enxerto , Humanos , Transplante das Ilhotas Pancreáticas/efeitos adversos , Masculino , Pessoa de Meia-Idade , Monitorização Fisiológica/métodos , Probabilidade , Estudos Prospectivos , Medição de Risco , Índice de Gravidade de Doença , Coleta de Tecidos e Órgãos , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA