Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Am J Transplant ; 22(8): 1992-2005, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35506189

RESUMO

Pancreatic islet transplantation has therapeutic potential in type 1 diabetes and is also an established therapy in chronic pancreatitis. However, the long-term transplant outcomes are modest. Identifying indicators of graft function will aid the preservation of transplanted islets and glycemic control. We analyzed beta cell prohormone peptide levels in a retrospective cohort of total pancreatectomy autologous islet transplant patients (n = 28). Proinsulin-to-C-peptide (PI/C) and proIAPP-to-total IAPP (proIAPP/IAPP) ratios measured at 3 months post-transplant were significantly higher in patients who remained insulin dependent at 1 year follow-up. In an immuno-deficient mouse model of human islet transplantation, recipient mice that later became hyperglycemic displayed significantly higher PI/C ratios than mice that remained normoglycemic. Histological analysis of islet grafts showed reduced proportional insulin- and proinsulin-positive area, but elevated glucagon-positive area in grafts that experienced greater secretory demand. Increased prohormone convertase 1/3 was detected in glucagon-positive cells, and glucagon-like peptide 1 (GLP-1) area was elevated in grafts from mice that displayed hyperglycemia or elevated plasma PI/C ratios, demonstrating intra-islet incretin production in metabolically challenged human islet grafts. These data indicate that in failing grafts, alpha cell prohormone processing is likely altered, and incomplete beta cell prohormone processing may be an early indicator of insulin dependency.


Assuntos
Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Animais , Peptídeo C , Glucagon , Humanos , Insulina , Camundongos , Proinsulina , Estudos Retrospectivos
2.
Diabetologia ; 60(3): 453-463, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27999871

RESUMO

AIMS/HYPOTHESIS: A contributor to beta cell failure in type 2 diabetes and islet transplants is amyloid formation by aggregation of the beta cell peptide, islet amyloid polypeptide (IAPP). Similar to the proinsulin processing pathway that generates insulin, IAPP is derived from a prohormone precursor, proIAPP, which requires cleavage by prohormone convertase (PC) 1/3 and PC2 in rodent pancreatic beta cells. We hypothesised that loss of PC2 would promote beta cell death and dysfunction in a rodent model of human beta cell proIAPP overexpression. METHODS: We generated an islet transplant model wherein immune-deficient mouse models of diabetes received islets expressing amyloidogenic human proIAPP and lacking PC2, leading to restoration of normoglycaemia accompanied by increased secretion of human proIAPP. Blood glucose levels were analysed for up to 16 weeks in transplant recipients and grafts were assessed for islet amyloid and beta cell number and death. RESULTS: Hyperglycaemia (blood glucose >16.9 mmol/l) returned in 94% of recipients of islets expressing human proIAPP and lacking PC2, whereas recipients of islets that express human proIAPP and normal PC2 levels remained normoglycaemic for at least 16 weeks. Islet graft failure was accompanied by a ∼20% reduction in insulin-positive cells, yet the degree of amyloid deposition and beta cell apoptosis was similar to those of controls expressing human proIAPP with functional PC2 levels. CONCLUSIONS/INTERPRETATION: PC2 deficiency in transplanted mouse islets expressing human proIAPP promotes beta cell loss and graft failure. Our data suggest that impaired NH2-terminal processing and increased secretion of human proIAPP promote beta cell failure.


Assuntos
Amiloide/metabolismo , Células Secretoras de Insulina/metabolismo , Pró-Proteína Convertase 2/metabolismo , Amiloide/genética , Animais , Glicemia/metabolismo , Western Blotting , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Transplante das Ilhotas Pancreáticas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Proinsulina/metabolismo , Pró-Proteína Convertase 1/genética , Pró-Proteína Convertase 1/metabolismo , Pró-Proteína Convertase 2/genética
3.
J Immunol ; 194(7): 3054-64, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25740943

RESUMO

Autoimmune destruction of insulin-producing ß cells in type 1 diabetes and islet transplantation involves a variety of immune pathways but is primarily mediated by self-reactive T cells. Chemokines can modulate local immune responses in inflammation and tumors by recruiting immune cells. We have reported that expression of the chemokine CCL22 in pancreatic ß cells in the NOD mouse prevents autoimmune attack by recruiting T regulatory cells (Tregs), protecting mice from diabetes. In this study we show that invariant NKT cells are also recruited to CCL22-expressing islet transplants and are required for CCL22-mediated protection from autoimmunity. Moreover, CCL22 induces an influx of plasmacytoid dendritic cells, which correlates with higher levels of IDO in CCL22-expressing islet grafts. In addition to its chemotactic properties, we found that CCL22 activates Tregs and promotes their ability to induce expression of IDO by dendritic cells. Islet CCL22 expression thus produces a tolerogenic milieu through the interplay of Tregs, invariant NKT cells, and plasmacytoid dendritic cells, which results in suppression of effector T cell responses and protection of ß cells. The immunomodulatory properties of CCL22 could be harnessed for prevention of graft rejection and type 1 diabetes as well as other autoimmune disorders.


Assuntos
Quimiocina CCL22/metabolismo , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/metabolismo , Animais , Quimiocina CCL22/genética , Quimiotaxia/genética , Quimiotaxia/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Diabetes Mellitus Tipo 1/genética , Modelos Animais de Doenças , Feminino , Expressão Gênica , Imunomodulação/genética , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Ilhotas Pancreáticas/metabolismo , Transplante das Ilhotas Pancreáticas , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Transdução Genética , Transplantes/imunologia , Transplantes/metabolismo
4.
Diabetologia ; 59(6): 1242-6, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26970755

RESUMO

AIMS/HYPOTHESIS: Islet amyloid, a pathological feature of type 2 diabetes, forms from the aggregation of islet amyloid polypeptide (IAPP), a beta cell peptide that is produced and co-secreted with insulin. Cholesterol regulates amyloid-ß processing, deposition and clearance, promoting amyloidogenesis in the brain. ATP-binding cassette transporter 1 (ABCA1) is a cholesterol efflux transporter that when absent increases and when overexpressed reduces brain amyloid-ß deposition in mouse models of Alzheimer's disease. We examined whether alterations in ABCA1 expression and islet cholesterol content could also modulate islet amyloidogenesis. METHODS: Thioflavin S staining for amyloid was performed in islets isolated from mice with beta cell expression of human IAPP (hIAPP (Tg/o)) and cultured for 8 days following cholesterol loading, microRNA-33 overexpression (to reduce ABCA1 expression) or palmitate treatment in the presence or absence of ABCA1 overexpression or mevastatin treatment (to reduce cholesterol synthesis). hIAPP (Tg/o) mice were crossed with beta cell-specific Abca1-knockout mice (hIAPP (Tg/o) Abca1 (ßKO)) and glucose tolerance and amyloid formation were assessed. RESULTS: Cholesterol loading and microRNA-33-induced reduction in islet ABCA1 expression increased Thioflavin S-positive amyloid in hIAPP (Tg/o) islets. Palmitate treatment also increased amyloid formation and this was reduced by both ABCA1 overexpression and mevastatin treatment. hIAPP (Tg/o) Abca1 (ßKO) mice had increased islet cholesterol, accompanied by fasting hyperglycaemia, glucose intolerance, impaired in vivo insulin secretion and an increased islet proinsulin:insulin ratio. Amyloid area was increased in cultured hIAPP (Tg/o) Abca1 (ßKO) islets compared with hIAPP (Tg/o) controls. CONCLUSIONS/INTERPRETATION: These data suggest that elevations in islet cholesterol may lead to increases in IAPP aggregation and islet amyloid formation, further worsening beta cell function and glucose homeostasis.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/deficiência , Amiloidose/metabolismo , Amiloidose/patologia , Colesterol/metabolismo , Transportador 1 de Cassete de Ligação de ATP/genética , Amiloide/metabolismo , Animais , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Lovastatina/análogos & derivados , Lovastatina/farmacologia , Masculino , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Palmitatos/farmacologia , Ratos
5.
Genome Res ; 20(8): 1037-51, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20551221

RESUMO

The liver and pancreas share a common origin and coexpress several transcription factors. To gain insight into the transcriptional networks regulating the function of these tissues, we globally identify binding sites for FOXA2 in adult mouse islets and liver, PDX1 in islets, and HNF4A in liver. Because most eukaryotic transcription factors bind thousands of loci, many of which are thought to be inactive, methods that can discriminate functionally active binding events are essential for the interpretation of genome-wide transcription factor binding data. To develop such a method, we also generated genome-wide H3K4me1 and H3K4me3 localization data in these tissues. By analyzing our binding and histone methylation data in combination with comprehensive gene expression data, we show that H3K4me1 enrichment profiles discriminate transcription factor occupied loci into three classes: those that are functionally active, those that are poised for activation, and those that reflect pioneer-like transcription factor activity. Furthermore, we demonstrate that the regulated presence of H3K4me1-marked nucleosomes at transcription factor occupied promoters and enhancers controls their activity, implicating both tissue-specific transcription factor binding and nucleosome remodeling complex recruitment in determining tissue-specific gene expression. Finally, we apply these approaches to generate novel insights into how FOXA2, PDX1, and HNF4A cooperate to drive islet- and liver-specific gene expression.


Assuntos
Loci Gênicos , Fator 3-beta Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/genética , Histonas/genética , Proteínas de Homeodomínio/genética , Ilhotas Pancreáticas/metabolismo , Fígado/metabolismo , Nucleossomos/genética , Transativadores/genética , Animais , Sequência de Bases , Sítios de Ligação , Perfilação da Expressão Gênica , Fator 3-beta Nuclear de Hepatócito/metabolismo , Fator 4 Nuclear de Hepatócito/metabolismo , Histonas/metabolismo , Proteínas de Homeodomínio/metabolismo , Camundongos , Dados de Sequência Molecular , Nucleossomos/metabolismo , Sequências Reguladoras de Ácido Nucleico , Transativadores/metabolismo
6.
J Immunol ; 187(5): 2755-65, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21813778

RESUMO

Islets from patients with type 2 diabetes exhibit ß cell dysfunction, amyloid deposition, macrophage infiltration, and increased expression of proinflammatory cytokines and chemokines. We sought to determine whether human islet amyloid polypeptide (hIAPP), the main component of islet amyloid, might contribute to islet inflammation by recruiting and activating macrophages. Early aggregates of hIAPP, but not nonamyloidogenic rodent islet amyloid polypeptide, caused release of CCL2 and CXCL1 by islets and induced secretion of TNF-α, IL-1α, IL-1ß, CCL2, CCL3, CXCL1, CXCL2, and CXCL10 by C57BL/6 bone marrow-derived macrophages. hIAPP-induced TNF-α secretion was markedly diminished in MyD88-, but not TLR2- or TLR4-deficient macrophages, and in cells treated with the IL-1R antagonist (IL-1Ra) anakinra. To determine the significance of IL-1 signaling in hIAPP-induced pancreatic islet dysfunction, islets from wild-type or hIAPP-expressing transgenic mice were transplanted into diabetic NOD/SCID recipients implanted with mini-osmotic pumps containing IL-1Ra (50 mg/kg/d) or saline. IL-1Ra significantly improved the impairment in glucose tolerance observed in recipients of transgenic grafts 8 wk following transplantation. Islet grafts expressing hIAPP contained amyloid deposits in close association with F4/80-expressing macrophages. Transgenic grafts contained 50% more macrophages than wild-type grafts, an effect that was inhibited by IL-1Ra. Our results suggest that hIAPP-induced islet chemokine secretion promotes macrophage recruitment and that IL-1R/MyD88, but not TLR2 or TLR4 signaling is required for maximal macrophage responsiveness to prefibrillar hIAPP. These data raise the possibility that islet amyloid-induced inflammation contributes to ß cell dysfunction in type 2 diabetes and islet transplantation.


Assuntos
Citocinas/metabolismo , Interleucina-1/antagonistas & inibidores , Polipeptídeo Amiloide das Ilhotas Pancreáticas/imunologia , Transplante das Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Transdução de Sinais/imunologia , Animais , Diabetes Mellitus Tipo 2/imunologia , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Imuno-Histoquímica , Inflamação/imunologia , Inflamação/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Gut Microbes ; 14(1): 2111951, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35984746

RESUMO

Hyperglycemia and type 2 diabetes (T2D) are caused by failure of pancreatic beta cells. The role of the gut microbiota in T2D has been studied, but causal links remain enigmatic. Obese individuals with or without T2D were included from two independent Dutch cohorts. Human data were translated in vitro and in vivo by using pancreatic islets from C57BL6/J mice and by injecting flagellin into obese mice. Flagellin is part of the bacterial locomotor appendage flagellum, present in gut bacteria including Enterobacteriaceae, which we show to be more abundant in the gut of individuals with T2D. Subsequently, flagellin induces a pro-inflammatory response in pancreatic islets mediated by the Toll-like receptor (TLR)-5 expressed on resident islet macrophages. This inflammatory response is associated with beta-cell dysfunction, characterized by reduced insulin gene expression, impaired proinsulin processing and stress-induced insulin hypersecretion in vitro and in vivo in mice. We postulate that increased systemically disseminated flagellin in T2D is a contributing factor to beta-cell failure in time and represents a novel therapeutic target.


Assuntos
Diabetes Mellitus Tipo 2 , Flagelina , Microbioma Gastrointestinal , Células Secretoras de Insulina , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Diabetes Mellitus Tipo 2/microbiologia , Flagelina/genética , Flagelina/metabolismo , Humanos , Inflamação/metabolismo , Insulina , Células Secretoras de Insulina/metabolismo , Camundongos
8.
Sci Rep ; 9(1): 10829, 2019 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-31346189

RESUMO

The study of primary glucagon-secreting α-cells is hampered by their low abundance and scattered distribution in rodent pancreatic islets. We have designed a double-stranded adeno-associated virus containing a rat proglucagon promoter (700 bp) driving enhanced green fluorescent protein (AAV GCG-EGFP), to specifically identify α-cells. The administration of AAV GCG-EGFP by intraperitoneal or intraductal injection led to EGFP expression selectively in the α-cell population. AAV GCG-EGFP delivery to mice followed by islet isolation, dispersion and separation by FACS for EGFP resulted in an 86% pure population of α-cells. Furthermore, the administration of AAV GCG-EGFP at various doses to adult wild type mice did not significantly alter body weight, blood glucose, plasma insulin or glucagon levels, glucose tolerance or arginine tolerance. In vitro experiments in transgene positive α-cells demonstrated that EGFP expression did not alter the intracellular Ca2+ pattern in response to glucose or adrenaline. This approach may be useful for studying purified primary α-cells and for the in vivo delivery of other genes selectively to α-cells to further probe their function or to manipulate them for therapeutic purposes.


Assuntos
Dependovirus , Células Secretoras de Glucagon/metabolismo , Glucagon/metabolismo , Proteínas de Fluorescência Verde , Animais , Glicemia , Peso Corporal/fisiologia , Insulina/sangue , Ilhotas Pancreáticas/metabolismo , Camundongos , Regiões Promotoras Genéticas , Ratos
9.
Mol Cell Endocrinol ; 477: 48-56, 2018 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-29883690

RESUMO

Overexpression of the X-linked inhibitor of apoptosis (XIAP) prevents islet allograft rejection. We constructed an adeno-associated virus expressing XIAP driven by the rat insulin promoter (dsAAV8-RIP-XIAP) for long-term beta-cell gene expression in vivo. Pancreatic delivery of dsAAV8-RIP-XIAP prevented autoimmune diabetes in 70% of non-obese diabetic (NOD) mice, associated with decreased insulitis. Islets from Balb/c mice transduced with dsAAV8-RIP-XIAP were protected following transplantation into streptozotocin (STZ)-diabetic Bl/6 recipients, associated with decreased graft infiltration. Interestingly, dsAAV8-RIP-XIAP transduction induced expression of lactate dehydrogenase (LDHA) and monocarboxylate transporter 1 (MCT1), two genes normally suppressed in beta cells and involved in production and release of lactate, a metabolite known to suppress local immune responses. Transduction of Balb/c islets with AAV8-RIP-LDHA-MCT1 tended to prolong allograft survival following transplant into STZ-diabetic Bl/6 recipients. These findings suggest that XIAP has therapeutic potential in autoimmune diabetes and raise the possibility that local lactate production may play a role in XIAP-mediated immunomodulation.


Assuntos
Aloenxertos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/prevenção & controle , Rejeição de Enxerto/prevenção & controle , Imunomodulação , Células Secretoras de Insulina/metabolismo , Transplante das Ilhotas Pancreáticas , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Aloenxertos/efeitos dos fármacos , Aloenxertos/metabolismo , Animais , Diabetes Mellitus Tipo 1/patologia , Glucose/farmacologia , Rejeição de Enxerto/imunologia , Humanos , Tolerância Imunológica/efeitos dos fármacos , Imunomodulação/efeitos dos fármacos , Injeções , Insulina/genética , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , L-Lactato Desidrogenase/metabolismo , Ácido Láctico/biossíntese , Camundongos , Camundongos Endogâmicos NOD , Transportadores de Ácidos Monocarboxílicos/metabolismo , Ratos , Simportadores/metabolismo
10.
Endocrinology ; 146(4): 1808-17, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15618358

RESUMO

Islet amyloid polypeptide (IAPP; amylin) is a peptide hormone that is cosecreted with insulin from beta-cells. Impaired processing of proIAPP, the IAPP precursor, has been implicated in islet amyloid formation in type 2 diabetes. We previously showed that proIAPP is processed to IAPP by the prohormone convertases PC1/3 and PC2 at its carboxyl (COOH) and amino (NH(2)) termini, respectively. In this study, we investigated the role of carboxypeptidase E (CPE) in the processing of proIAPP using mice lacking active CPE (Cpe(fat)/Cpe(fat)) and NIT-2 cells, a beta-cell line derived from their islets. Western blot analysis demonstrated that an approximately 6-kDa NH(2)-terminally unprocessed form of proIAPP was elevated approximately 86% in islets from Cpe(fat)/Cpe(fat) mice, compared with wild type. This increase was independent of the development of hyperglycemia (8 wk male) or obesity (18 wk female). Impaired proIAPP processing was associated with a decrease in PC2 (but not PC1/3) and both the 21- and 27-kDa forms of the PC2 chaperone protein 7B2, suggesting that PC2-mediated processing of proIAPP at its NH(2) terminus was impaired in the absence of CPE. Formation of COOH-terminally amidated (pro)IAPP was reduced approximately 75% in NIT-2, compared with NIT-1 beta-cells, supporting a direct role for CPE in maturation of IAPP by removal of its COOH-terminal dibasic residues, the step essential for IAPP amidation. We conclude that lack of CPE in islet beta-cells results in a marked decrease in processing of proIAPP at its NH(2) (but not COOH) terminus that is associated with attenuated levels of PC2 and (pro)7B2 and a great reduction in formation of mature amidated IAPP.


Assuntos
Amiloide/metabolismo , Carboxipeptidase H/fisiologia , Ilhotas Pancreáticas/metabolismo , Animais , Linhagem Celular , Feminino , Masculino , Camundongos , Pró-Proteína Convertase 1/fisiologia , Pró-Proteína Convertase 2/fisiologia
11.
Cell Transplant ; 24(10): 2143-54, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26423995

RESUMO

Manipulation of regulatory T cell (Treg) migration by islet expression of the chemokine CCL22 prevents diabetes in NOD mice and delays recurrent autoimmunity in syngeneic islet transplants. We sought to determine whether attracting Tregs with CCL22 also prevents islet allograft rejection. Isolated Bl/6 mouse islets were transduced overnight with adenovirus expressing CCL22 (Ad-CCL22) downstream of the CMV promoter. Islets were transplanted under the renal capsule of Balb/c recipients made diabetic by streptozotocin. To assess immunologic tolerance, graft-bearing kidneys from recipients of CCL22-expressing islet grafts were removed, and mice received a second transplant of naive islets from the same donor strain or third-party islets into the contralateral kidney. Adenoviral expression of CCL22 conferred prolonged protection of islet allografts in MHC-mismatched, diabetic recipients, maintaining normoglycemia in 75% of recipients for at least 80 days. Increased frequency of Treg cells was observed in islet grafts transduced with Ad-CCL22 compared with untreated grafts. Normoglycemic recipients of CCL22-expressing islet grafts showed complete absence of antidonor antibodies and no lymphocyte proliferation after exposure to donor splenocytes. After removal of the primary graft at day 80, mice that received a second transplant with untreated islets from the same donor strain did not reject the grafts, suggesting the development of tolerance. Expression of CCL22 recruits Treg cells to transplanted islets, prevents activation of alloreactive T-cells and islet allograft failure and induces alloantigen-specific tolerance. Manipulation of Treg cells by CCL22 in transplanted islets may be a novel therapeutic strategy for diabetes.


Assuntos
Aloenxertos/imunologia , Quimiocina CCL22/imunologia , Sobrevivência de Enxerto/imunologia , Tolerância Imunológica/imunologia , Transplante das Ilhotas Pancreáticas/imunologia , Linfócitos T Reguladores/imunologia , Animais , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/terapia , Isoantígenos/imunologia , Camundongos , Tolerância ao Transplante/imunologia , Transplante Homólogo/métodos
12.
Islets ; 3(4): 175-87, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21633187

RESUMO

Diabetes is a multifactorial metabolic disorder defined by the loss of functional pancreatic insulin-producing ß-cells. The functional maturation and dedifferentiation of adult ß-cells is central to diabetes pathogenesis and to ß-cell replacement therapy for the treatment of diabetes. Despite its importance, the dynamics and mechanisms of adult ß-cell maturation remain poorly understood. Using a novel Pdx1/Ins1 dual fluorescent reporter lentiviral vector, we previously found that individual adult human and mouse ß-cells exist in at least two differentiation states distinguishable by the activation of the rat Ins1 promoter and performed the first real-time imaging of the maturation of individual cultured ß-cells. Our previous study focused on transformed (MIN6) ß-cells as a model to investigatethe kinetics of ß-cell maturation. In the present study, we investigated the kinetics of the maturation process in primary human and mouse ß-cells and performed gene expression profiling. Gene expression profiling of FACS purified immature Pdx1 (+) /Ins1 (low) cells and mature Pdx1 (high) /Ins1 (high ) cells from cultures of human islets, mouse islets and MIN6 cells revealed that Pdx1 (+) /Ins1 (low) cells are enriched for multiple genes associated with ß-cell development/progenitor cells, proliferation, apoptosis, as well as genes coding for other islet cell hormones such as glucagon. We also demonstrated that the heterogeneity in ß-cell maturation states previously observed in vitro, can also be found in vivo. Collectively, these experiments contribute to the understanding of maturation, dedifferentiation and plasticity of adult pancreatic ß-cells. The results have significant implications for islet regeneration and for in vitro generation of functional ß-cells to treat diabetes.


Assuntos
Diferenciação Celular , Células Secretoras de Insulina/fisiologia , Adulto , Animais , Linhagem Celular Transformada , Células Cultivadas , Perfilação da Expressão Gênica , Genes Reporter , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/ultraestrutura , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Célula Única , Transativadores/genética , Transativadores/metabolismo
13.
J Clin Invest ; 121(8): 3024-8, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21737880

RESUMO

Type 1 diabetes is characterized by destruction of insulin-producing ß cells in the pancreatic islets by effector T cells. Tregs, defined by the markers CD4 and FoxP3, regulate immune responses by suppressing effector T cells and are recruited to sites of action by the chemokine CCL22. Here, we demonstrate that production of CCL22 in islets after intrapancreatic duct injection of double-stranded adeno-associated virus encoding CCL22 recruits endogenous Tregs to the islets and confers long-term protection from autoimmune diabetes in NOD mice. In addition, adenoviral expression of CCL22 in syngeneic islet transplants in diabetic NOD recipients prevented ß cell destruction by autoreactive T cells and thereby delayed recurrence of diabetes. CCL22 expression increased the frequency of Tregs, produced higher levels of TGF-ß in the CD4+ T cell population near islets, and decreased the frequency of circulating autoreactive CD8+ T cells and CD8+ IFN-γ­producing T cells. The protective effect of CCL22 was abrogated by depletion of Tregs with a CD25-specific antibody. Our results indicate that islet expression of CCL22 recruits Tregs and attenuates autoimmune destruction of ß cells. CCL22-mediated recruitment of Tregs to islets may be a novel therapeutic strategy for type 1 diabetes.


Assuntos
Quimiocina CCL22/fisiologia , Diabetes Mellitus Tipo 1/prevenção & controle , Ilhotas Pancreáticas/citologia , Linfócitos T Reguladores/metabolismo , Animais , Doenças Autoimunes/metabolismo , Autoimunidade , Linfócitos T CD4-Positivos/metabolismo , Quimiocina CCL22/genética , Diabetes Mellitus Tipo 1/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Ratos
14.
Islets ; 2(1): 18-23, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21099289

RESUMO

Clinical pancreatic islet transplantation has great promise as a treatment for type 1 diabetes but despite recent advances, it is still limited by the need for lifelong immunosuppression, restricted availability of donor islets, and uncertainty regarding long-term graft survival. Using a syngeneic, suboptimal islet transplantation model, we asked whether adenoviral overexpression of an anti-apoptotic protein, the X-linked inhibitor of apoptosis protein (XIAP) would protect transplanted islet cells from death and reduce the number of islets required for successful transplantation. Transplantation of 100 XIAP-expressing islets into the kidney capsule of syngeneic Balb/c mice restored euglycemia in 86% of recipients, where transplantation of 100 islets transduced with a control adenovirus expressing LacZ restored euglycemia in only 27% of recipients. Analysis of islet grafts by insulin/TUNEL double immunostaining revealed fewer apoptotic beta-cells in recipients of XIAP- compared with LacZ-expressing grafts (0.8±0.5 vs. 2.4±0.8 double-positive cells/graft), suggesting that XIAP enhances graft success by inhibiting ß-cell apoptosis in the immediate post-transplant period. In summary, XIAP overexpression inhibits beta cell apoptosis in syngeneic islet transplants, thereby reducing the number of islets and decreasing the number of days required to restore euglycemia. These data raise the possibility that ex vivo XIAP gene transfer in islets prior to transplantation has the potential to increase the number of donor islets available for transplantation and may enhance graft function and long-term transplant success.


Assuntos
Apoptose/fisiologia , Glicemia/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/fisiologia , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/fisiologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/fisiologia , Animais , Apoptose/genética , Contagem de Células , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Experimental/terapia , Feminino , Técnicas de Transferência de Genes , Homeostase/genética , Homeostase/fisiologia , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Transplante das Ilhotas Pancreáticas/métodos , Transplante das Ilhotas Pancreáticas/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Estreptozocina , Transplante Isogênico , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo
15.
Islets ; 2(2): 104-11, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21099302

RESUMO

Type 2 diabetes is a metabolic and inflammatory disease characterized by deteriorating islet function and increased levels of inflammatory cytokines. The inflammatory milieu induced in type 2 diabetes exacerbates islet dysfunction and insulin resistance, and therapies that target inflammation can improve glycemic control in patients with type 2 diabetes. Inflammation in type 2 diabetes may be the result of the stimulation of Toll-like receptors (TLRs), one of the many mediators of inflammation. TLRs can be activated by both exogenous and endogenous ligands, and are responsible for activating NFκB and interferon- inducible inflammatory gene expression. We examined the role of the TIR-domain containing adaptor-inducing interferon-ß (TRIF or TICAM-1), a major signaling molecule for TLR3 and TLR4, in b-cell function and glucose homeostasis by examining mice lacking TRIF (Trif⁻(/)⁻), TLR3 (Tlr3⁻(/)⁻) or TLR4 (Tlr4⁻(/)⁻). Male, 10-week old Trif⁻(/)⁻ mice exhibit a moderate but significant increase in fasting blood glucose compared to C57BL/6 controls (12.0 ± 0.9 vs. 9.7 ± 0.4 mM; p < 0.05) as well as impaired glucose tolerance revealed by IPGTT (AUC: 2850 ± 236 vs. 2050 ± 108; p < 0.005) whereas Tlr3⁻(/)⁻ and Tlr4⁻(/)⁻ mice have normal glucose tolerance. Interestingly, Trif⁻(/)⁻ mice have normal insulin sensitivity yet have increased plasma insulin levels (180 ± 22 vs. 89 ± 24 pM; p < 0.05). Islets isolated from Trif⁻(/)⁻ mice have impaired glucose-stimulated insulin secretion, with a diminished first-phase insulin response to glucose. Immunohistological analysis revealed that age-matched Trif⁻(/)⁻ and control mice have normal islet morphology, although Trif⁻(/)⁻ mice have increased b-cell mass (3.5 ± 0.9 vs. 1.7 ± 0.2 mg; p < 0.05). In summary, mice lacking TRIF have hyperglycemia associated with b-cell dysfunction that may be partly compensated for by increased b-cell mass. These studies suggest a role for TLR signaling in glucose homeostasis, and raise the possibility that TRIF signaling is required for normal b-cell function.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Homeostase/genética , Homeostase/fisiologia , Resistência à Insulina/genética , Resistência à Insulina/fisiologia , Células Secretoras de Insulina/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão/genética , Pâncreas/anatomia & histologia , Pâncreas/citologia , Pâncreas/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Receptores Toll-Like/fisiologia
16.
Transplantation ; 88(9): 1075-80, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19898202

RESUMO

BACKGROUND: Islet transplantation is a promising therapy for type 1 diabetes; however, most islet grafts fail within 5 years. Innate immunity has been suggested to play a role in islet allograft rejection, potentially mediated by toll-like receptors (TLRs), a class of innate immune receptors. Lack of TLR4, in particular, has been reported to improve allograft survival. Therefore, we hypothesized that TLRs may be involved in islet allograft rejection, and that deletion of TLR4 may improve islet graft survival. METHODS: Islets were isolated from C57BL/10ScNJ (Tlr4(-/-)) and C57BL/10 (wild-type [WT]) animals and transplanted into Balb/cJ recipients with streptozotocin-induced diabetes. Blood glucose levels were used to determine graft viability and immunostaining to assess graft morphology and immune cell infiltration. The roles of the TLR4 adaptor molecules MyD88 and TLR adaptor molecule 1 (Ticam-1) were assessed using islets isolated from mice lacking MyD88 (MyD88(-/-)), Ticam-1 (Ticam-1(-/-)), or the combined double knockout (MyD88(-/-)/Ticam-1(-/-)). RESULTS: Contrary to our hypothesis, Tlr4(-/-) and WT islet allografts had similar failure rates; grafts failed at 23.2+/-1.2 and 24.5+/-1.5 days posttransplant, respectively (P=NS). Syngeneic grafts of Tlr4(-/-) and WT islets maintained normoglycemia for up to 10 weeks posttransplant, indicating that failure of Tlr4(-/-) islet allografts could not be attributed to an intrinsic defect in Tlr4(-/-) islets. Similarly, islet allotransplants from MyD88(-/-), Ticam-1(-/-), and MyD88(-/-)/Ticam-1(-/-) donors did not have improved allograft survival compared with WT controls. CONCLUSIONS: These findings indicate that islet allograft rejection in mice is independent of TLR4 and the TLR adaptor molecules MyD88 and Ticam-1, speaking against an essential role for TLR signaling in islet allograft rejection.


Assuntos
Diabetes Mellitus Experimental/cirurgia , Transplante das Ilhotas Pancreáticas/patologia , Transdução de Sinais/fisiologia , Receptores Toll-Like/fisiologia , Animais , Glicemia/metabolismo , Linfócitos T CD8-Positivos/imunologia , Primers do DNA , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Tipo 1/cirurgia , Tolerância Imunológica , Imunidade Inata , Transplante das Ilhotas Pancreáticas/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , RNA/genética , RNA/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia , Transplante Homólogo/patologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa