Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Clin Exp Immunol ; 211(3): 224-232, 2023 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-36622793

RESUMO

The HLA region is the major genetic risk determinant of Type 1 diabetes. How non-HLA loci contribute to the genetic risk is incompletely understood, but there are indications that at least some impact progression of asymptomatic autoimmunity. We examined whether SNPs in 7 susceptibility loci (INS, SH2B3, PTPN2, PTPN22, CTLA4, CLEC16A, and IL2RA) could improve prediction of the progression from single to multiple autoantibody positivity, and from there on to diagnosis. SNPs were genotyped in persistently autoantibody positive relatives by allelic discrimination qPCR and disease progression was studied by multivariate Cox regression analysis. In our cohort, only the CTLA4 GA genotype (rs3087243, P = 0.002) and the CLEC16A AA genotype (rs12708716, P = 0.021) were associated with accelerated progression from single to multiple autoantibody positivity, but their effects were restricted to presence of HLA-DQ2/DQ8, and IAA as first autoantibody, respectively. The interaction of CTLA4 and HLA-DQ2/DQ8 overruled the effect of DQ2/DQ8 alone. The HLA-DQ2/DQ8-mediated risk of progression to multiple autoantibodies nearly entirely depended on heterozygosity for CTLA4. The SH2B3 TT genotype (rs3184504) was protective for HLA-DQ8 positive subjects (P = 0.003). At the stage of multiple autoantibodies, only the CTLA4 GA genotype was a minor independent risk factor for progression towards clinical diabetes (P = 0.034). Our study shows that non-HLA polymorphisms impact progression of islet autoimmunity in a subgroup-, stage- and SNP-specific way, suggesting distinct mechanisms. If confirmed, these findings may help refine risk assessment, follow-up, and prevention trials in risk groups.


Assuntos
Diabetes Mellitus Tipo 1 , Humanos , Autoanticorpos , Autoimunidade/genética , Antígeno CTLA-4/genética , Diabetes Mellitus Tipo 1/genética , Predisposição Genética para Doença , Genótipo , Lectinas Tipo C/genética , Proteínas de Transporte de Monossacarídeos/genética , Polimorfismo de Nucleotídeo Único , Proteína Tirosina Fosfatase não Receptora Tipo 22/genética
2.
Cell ; 132(2): 197-207, 2008 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-18243096

RESUMO

Novel strategies in diabetes therapy would obviously benefit from the use of beta (beta) cell stem/progenitor cells. However, whether or not adult beta cell progenitors exist is one of the most controversial issues in today's diabetes research. Guided by the expression of Neurogenin 3 (Ngn3), the earliest islet cell-specific transcription factor in embryonic development, we show that beta cell progenitors can be activated in injured adult mouse pancreas and are located in the ductal lining. Differentiation of the adult progenitors is Ngn3 dependent and gives rise to all islet cell types, including glucose responsive beta cells that subsequently proliferate, both in situ and when cultured in embryonic pancreas explants. Multipotent progenitor cells thus exist in the pancreas of adult mice and can be activated cell autonomously to increase the functional beta cell mass by differentiation and proliferation rather than by self-duplication of pre-existing beta cells only.


Assuntos
Células Secretoras de Insulina/citologia , Pâncreas/citologia , Pâncreas/lesões , Células-Tronco/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/isolamento & purificação , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Núcleo Celular/metabolismo , Proliferação de Células , Expressão Gênica , Genes Reporter , Vetores Genéticos , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Insulina/análise , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Queratinas/metabolismo , Lentivirus/genética , Ligadura , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/isolamento & purificação , Proteínas do Tecido Nervoso/metabolismo , Técnicas de Cultura de Órgãos , Ductos Pancreáticos/cirurgia , Células-Tronco/metabolismo , Fatores de Tempo , beta-Galactosidase/metabolismo
3.
Diabetologia ; 64(11): 2511-2516, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34448034

RESUMO

AIMS/HYPOTHESIS: We examined whether the non-HLA susceptibility locus ERBB3/IKZF4 influences progression of type 1 diabetes stage specifically according to sex. METHODS: SNPs of ERBB3 (rs2292239 T/G) and IKZF4 (rs1701704 G/T) were screened by allelic discrimination quantitative PCR assay in first-degree relatives of type 1 diabetes patients who had developed at least one circulating autoantibody. The effect of ERBB3/IKZF4 genotypes and sex, on the progression of single autoantibody positivity to multiple autoantibody positivity and from multiple autoantibody positivity to diabetes, was studied by Kaplan-Meier analysis and multivariate Cox regression. RESULTS: In the cohort of autoantibody-positive first-degree relatives, the risk allele frequencies for ERBB3 rs2292239 (T) and IKZF4 rs1701704 (G) were increased. There was a significant male excess at the stage of multiple autoantibody positivity (p = 0.021). In Kaplan-Meier survival analysis, progression from single to multiple antibody positivity was delayed in female participants with genotype ERBB3 GG (p = 0.018, vs ERBB3 TG+TT) or IKZF4 TT (p = 0.023, vs IKZF4 GT+GG), but not in male participants. In multivariate Cox regression models, the interaction effects between female sex and ERBB3 GG (p = 0.012; HR = 0.305 [95% CI 0.120, 0.773]) or between female sex and IKZF4 TT (p = 0.011; HR = 0.329 [95% CI 0.140, 0.777]) emerged as potential determinants of delayed progression to multiple autoantibodies. The progression from multiple autoantibody positivity to type 1 diabetes appeared not to be influenced by ERBB3/IKZF4. CONCLUSIONS/INTERPRETATION: In siblings and offspring of type 1 diabetes patients, polymorphism in region ERBB3/IKZF4 may affect disease progression at the level of epitope spreading in female individuals. Our findings suggest that interaction between sex and ERBB3/IKZF4 may contribute to the post-pubertal male excess in type 1 diabetes.


Assuntos
Autoanticorpos/sangue , Autoantígenos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Epitopos/imunologia , Fator de Transcrição Ikaros/genética , Receptor ErbB-3/genética , Caracteres Sexuais , Adolescente , Adulto , Criança , Diabetes Mellitus Tipo 1/genética , Progressão da Doença , Feminino , Predisposição Genética para Doença , Humanos , Insulina/imunologia , Masculino , Polimorfismo de Nucleotídeo Único/genética , Modelos de Riscos Proporcionais , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Tirosina Fosfatases Classe 8 Semelhantes a Receptores/imunologia , Transportador 8 de Zinco/imunologia
4.
Diabetologia ; 59(7): 1474-1479, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27130279

RESUMO

AIMS/HYPOTHESIS: Pw1 or paternally-expressed gene 3 (Peg3) encodes a zinc finger transcription factor that is widely expressed during mouse embryonic development and later restricted to multiple somatic stem cell lineages in the adult. The aim of the present study was to define Pw1 expression in the embryonic and adult pancreas and investigate its role in the beta cell cycle in Pw1 wild-type and mutant mice. METHODS: We analysed PW1 expression by immunohistochemistry in pancreas of nonpregant and pregnant mice and following injury by partial duct ligation. Its role in the beta cell cycle was studied in vivo using a novel conditional knockout mouse and in vitro by lentivirus-mediated gene knockdown. RESULTS: We showed that PW1 is expressed in early pancreatic progenitors at E9.5 but becomes progressively restricted to fully differentiated beta cells as they become established after birth and withdraw from the cell cycle. Notably, PW1 expression declines when beta cells are induced to proliferate and loss of PW1 function activates the beta cell cycle. CONCLUSIONS/INTERPRETATION: These results indicate that PW1 is a co-regulator of the beta cell cycle and can thus be considered a novel therapeutic target in diabetes.


Assuntos
Células Secretoras de Insulina/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Pâncreas/metabolismo , Animais , Ciclo Celular/genética , Ciclo Celular/fisiologia , Proliferação de Células/genética , Proliferação de Células/fisiologia , Imuno-Histoquímica , Células Secretoras de Insulina/citologia , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pâncreas/embriologia
5.
Diabetologia ; 59(9): 1834-7, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27053238

RESUMO

The generation of beta(-like) cells to compensate for their absolute or relative shortage in type 1 and type 2 diabetes is an obvious therapeutic strategy. Patients first received grafts of donor islet cells over 25 years ago, but this procedure has not become routine in clinical practice because of a donor cell shortage and (auto)immune problems. Transplantation of differentiated embryonic and induced pluripotent stem cells may overcome some but not all the current limitations. Reprogramming exocrine cells towards functional beta(-like) cells would offer an alternative abundant and autologous source of beta(-like) cells. This review focuses on work by our research group towards achieving such a source of cells. It summarises a presentation given at the 'Can we make a better beta cell?' symposium at the 2015 annual meeting of the EASD. It is accompanied by two other reviews on topics from this symposium (by Amin Ardestani and Kathrin Maedler, DOI: 10.1007/s00125-016-3892-9 , and by Heiko Lickert and colleagues, DOI: 10.1007/s00125-016-3949-9 ) and a commentary by the Session Chair, Shanta Persaud (DOI: 10.1007/s00125-016-3870-2 ).


Assuntos
Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Pâncreas/citologia , Animais , Diferenciação Celular/fisiologia , Humanos , Macrófagos/metabolismo , Fatores de Transcrição/metabolismo
6.
Eur J Immunol ; 45(5): 1482-93, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25645754

RESUMO

Pancreas injury by partial duct ligation (PDL) activates a healing response, encompassing ß-cell neogenesis and proliferation. Macrophages (MΦs) were recently shown to promote ß-cell proliferation after PDL, but they remain poorly characterized. We assessed myeloid cell diversity and the factors driving myeloid cell dynamics following acute pancreas injury by PDL. In naive and sham-operated pancreas, the myeloid cell compartment consisted mainly of two distinct tissue-resident MΦ types, designated MHC-II(lo) and MHC-II(hi) MΦs, the latter being predominant. MHC-II(lo) and MHC-II(hi) pancreas MΦs differed at the molecular level, with MHC-II(lo) MΦs being more M2-activated. After PDL, there was an early surge of Ly6C(hi) monocyte infiltration in the pancreas, followed by a transient MHC-II(lo) MΦ peak and ultimately a restoration of the MHC-II(hi) MΦ-dominated steady-state equilibrium. These intricate MΦ dynamics in PDL pancreas depended on monocyte recruitment by C-C chemokine receptor 2 and macrophage-colony stimulating factor receptor as well as on macrophage-colony stimulating factor receptor-dependent local MΦ proliferation. Functionally, MHC-II(lo) MΦs were more angiogenic. We further demonstrated that, at least in C-C chemokine receptor 2-KO mice, tissue MΦs, rather than Ly6C(hi) monocyte-derived MΦs, contributed to ß-cell proliferation. Together, our study fully characterizes the MΦ subsets in the pancreas and clarifies the complex dynamics of MΦs after PDL injury.


Assuntos
Macrófagos/imunologia , Macrófagos/patologia , Monócitos/imunologia , Monócitos/patologia , Pâncreas/imunologia , Pâncreas/lesões , Animais , Antígenos Ly/metabolismo , Movimento Celular/imunologia , Proliferação de Células , Microambiente Celular/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Ligadura , Ativação de Macrófagos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células Mieloides/classificação , Células Mieloides/imunologia , Células Mieloides/patologia , Pâncreas/patologia , Ductos Pancreáticos/lesões , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Regeneração/imunologia
7.
Development ; 140(4): 751-64, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23325761

RESUMO

Pancreatic multipotent progenitor cells (MPCs) produce acinar, endocrine and duct cells during organogenesis, but their existence and location in the mature organ remain contentious. We used inducible lineage-tracing from the MPC-instructive gene Ptf1a to define systematically in mice the switch of Ptf1a(+) MPCs to unipotent proacinar competence during the secondary transition, their rapid decline during organogenesis, and absence from the mature organ. Between E11.5 and E15.5, we describe tip epithelium heterogeneity, suggesting that putative Ptf1a(+)Sox9(+)Hnf1ß(+) MPCs are intermingled with Ptf1a(HI)Sox9(LO) proacinar progenitors. In the adult, pancreatic duct ligation (PDL) caused facultative reactivation of multipotency factors (Sox9 and Hnf1ß) in Ptf1a(+) acini, which undergo rapid reprogramming to duct cells and longer-term reprogramming to endocrine cells, including insulin(+) ß-cells that are mature by the criteria of producing Pdx1(HI), Nkx6.1(+) and MafA(+). These Ptf1a lineage-derived endocrine/ß-cells are likely formed via Ck19(+)/Hnf1ß(+)/Sox9(+) ductal and Ngn3(+) endocrine progenitor intermediates. Acinar to endocrine/ß-cell transdifferentiation was enhanced by combining PDL with pharmacological elimination of pre-existing ß-cells. Thus, we show that acinar cells, without exogenously introduced factors, can regain aspects of embryonic multipotentiality under injury, and convert into mature ß-cells.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Multipotentes/fisiologia , Organogênese/fisiologia , Pâncreas/embriologia , Recuperação de Função Fisiológica/fisiologia , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Células Acinares/citologia , Animais , Pesos e Medidas Corporais , Técnicas de Introdução de Genes , Camundongos , Microscopia Confocal , Células-Tronco Multipotentes/metabolismo , Pâncreas/fisiologia , Tamoxifeno , Fatores de Tempo
8.
Diabetologia ; 57(7): 1420-7, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24759958

RESUMO

AIMS/HYPOTHESIS: IL-6 was recently shown to control alpha cell expansion. As beta cells expand following partial pancreatic-duct ligation (PDL) in adult mice, we investigated whether PDL also causes alpha cells to expand and whether IL-6 signalling is involved. As alpha cells can reprogramme to beta cells in a number of beta cell (re)generation models, we examined whether this phenomenon also exists in PDL pancreas. METHODS: Total alpha cell volume, alpha cell size and total glucagon content were evaluated in equivalent portions of PDL- and sham-operated mouse pancreases. Proliferation of glucagon(+) cells was assessed by expression of the proliferation marker Ki67. Inter-conversions between alpha and beta cells were monitored in transgenic mice with conditional cell-type-specific labelling. The role of IL-6 in regulating alpha cell proliferation was evaluated by in situ delivery of an IL-6-inactivating antibody. RESULTS: In response to PDL surgery, alpha cell volume in the ligated tissue was increased threefold, glucagon content fivefold and alpha cell size by 10%. Activation of alpha cell proliferation in PDL pancreas required IL-6 signalling. A minor fraction of alpha cells derived from beta cells, whereas no evidence for alpha to beta cell conversion was obtained. CONCLUSIONS/INTERPRETATION: In PDL-injured adult mouse pancreas, new alpha cells are generated mainly by IL-6-dependent self-duplication and seldom by reprogramming of beta cells.


Assuntos
Proliferação de Células/fisiologia , Células Secretoras de Glucagon/citologia , Interleucina-6/metabolismo , Ductos Pancreáticos/citologia , Animais , Tamanho Celular , Células Secretoras de Glucagon/metabolismo , Ligadura , Camundongos , Ductos Pancreáticos/metabolismo
9.
Diabetologia ; 57(1): 140-7, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24121626

RESUMO

AIMS/HYPOTHESIS: Vascular endothelial growth factor (VEGF) has been recognised by loss-of-function experiments as a pleiotropic factor with importance in embryonic pancreas development and postnatal beta cell function. Chronic, nonconditional overexpression of VEGF-A has a deleterious effect on beta cell development and function. We report, for the first time, a conditional gain-of-function study to evaluate the effect of transient VEGF-A overexpression by adult pancreatic beta cells on islet vasculature and beta cell proliferation and survival, under both normal physiological and injury conditions. METHODS: In a transgenicmouse strain, overexpressing VEGF-A in a doxycycline-inducible and beta cell-specific manner, we evaluated the ability of VEGF-A to affect islet vessel density, beta cell proliferation and protection of the adult beta cell mass from toxin-induced injury. RESULTS: Short-term VEGF-A overexpression resulted in islet hypervascularisation, increased beta cell proliferation and protection from toxin-mediated beta cell death, and thereby prevented the development of hyperglycaemia. Extended overexpression of VEGF-A led to impaired glucose tolerance, elevated fasting glycaemia and a decreased beta cell mass. CONCLUSIONS/INTERPRETATION: Overexpression of VEGF-A in beta cells time-dependently affects glycometabolic control and beta cell protection and proliferation. These data nourish further studies to examine the role of controlled VEGF delivery in (pre)clinical applications aimed at protecting and/or restoring the injured beta cell mass.


Assuntos
Diabetes Mellitus/prevenção & controle , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Proliferação de Células , Sobrevivência Celular/fisiologia , Diabetes Mellitus/metabolismo , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Transgênicos , Ratos , Fator A de Crescimento do Endotélio Vascular/genética
10.
Diabetologia ; 56(12): 2647-50, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24026213

RESUMO

AIMS/HYPOTHESIS: Long-term labelling of mice with halogenated thymidine analogues is an established method for quantifying the contribution of beta cell proliferation to in vivo beta cell mass expansion in (re)generation models. The method is believed to give accurate information on the accrued number of cycling beta cells over a period of time. Multiple thymidine analogue labelling is applied for evaluating the duration of postmitotic quiescence in beta cells. We hypothesise, however, that long-term labelling by thymidine analogues hampers beta cell proliferation. METHODS: Thymidine analogues were administered for 7-14 days via the i.p. route to neonatal mice, or via drinking water to young mice with normal pancreases or adult mice with injured pancreases. The proliferation of insulin-positive cells was assessed by their expression of the proliferation markers Ki67 or phosphorylated histone H3 and by their incorporation of nucleotide analogues. RESULTS: In the mouse models of beta cell proliferation investigated herein, long-term administration of thymidine analogues decreased the percentage of Ki67(+) and phosphorylated histone H3(+) beta cells as compared with administration of normal drinking water. Proliferation was restored by washout of the analogue. Labelling with one analogue decreased the subsequent incorporation of another analogue by beta cells. CONCLUSIONS/INTERPRETATION: Long-term labelling with halogenated thymidine analogues is a biased method that underestimates the proliferation and re-division potential of mouse beta cells.


Assuntos
Proliferação de Células/efeitos dos fármacos , Histonas/metabolismo , Células Secretoras de Insulina/metabolismo , Antígeno Ki-67/metabolismo , Timidina/análogos & derivados , Timidina/farmacologia , Animais , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Índice Mitótico , Modelos Animais , Coloração e Rotulagem/métodos , Timidina/administração & dosagem
11.
Genome Res ; 20(6): 722-32, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20395405

RESUMO

The epigenome changes that underlie cellular differentiation in developing organisms are poorly understood. To gain insights into how pancreatic beta-cells are programmed, we profiled key histone methylations and transcripts in embryonic stem cells, multipotent progenitors of the nascent embryonic pancreas, purified beta-cells, and 10 differentiated tissues. We report that despite their endodermal origin, beta-cells show a transcriptional and active chromatin signature that is most similar to ectoderm-derived neural tissues. In contrast, the beta-cell signature of trimethylated H3K27, a mark of Polycomb-mediated repression, clusters with pancreatic progenitors, acinar cells and liver, consistent with the epigenetic transmission of this mark from endoderm progenitors to their differentiated cellular progeny. We also identified two H3K27 methylation events that arise in the beta-cell lineage after the pancreatic progenitor stage. One is a wave of cell-selective de novo H3K27 trimethylation in non-CpG island genes. Another is the loss of bivalent and H3K27me3-repressed chromatin in a core program of neural developmental regulators that enables a convergence of the gene activity state of beta-cells with that of neural cells. These findings reveal a dynamic regulation of Polycomb repression programs that shape the identity of differentiated beta-cells.


Assuntos
Regulação para Baixo , Regulação da Expressão Gênica no Desenvolvimento , Ilhotas Pancreáticas/metabolismo , Pâncreas/embriologia , Proteínas Repressoras/genética , Animais , Separação Celular , Células Cultivadas , Imunoprecipitação da Cromatina , Epigênese Genética , Citometria de Fluxo , Histonas/metabolismo , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Pâncreas/citologia , Proteínas do Grupo Polycomb
13.
J Cell Biol ; 159(2): 303-12, 2002 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-12403815

RESUMO

Regulatory proteins have been identified in embryonic development of the endocrine pancreas. It is unknown whether these factors can also play a role in the formation of pancreatic endocrine cells from postnatal nonendocrine cells. The present study demonstrates that adult human pancreatic duct cells can be converted into insulin-expressing cells after ectopic, adenovirus-mediated expression of the class B basic helix-loop-helix factor neurogenin 3 (ngn3), which is a critical factor in embryogenesis of the mouse endocrine pancreas. Infection with adenovirus ngn3 (Adngn3) induced gene and/or protein expression of NeuroD/beta2, Pax4, Nkx2.2, Pax6, and Nkx6.1, all known to be essential for beta-cell differentiation in mouse embryos. Expression of ngn3 in adult human duct cells induced Notch ligands Dll1 and Dll4 and neuroendocrine- and beta-cell-specific markers: it increased the percentage of synaptophysin- and insulin-positive cells 15-fold in ngn3-infected versus control cells. Infection with NeuroD/beta2 (a downstream target of ngn3) induced similar effects. These data indicate that the Delta-Notch pathway, which controls embryonic development of the mouse endocrine pancreas, can also operate in adult human duct cells driving them to a neuroendocrine phenotype with the formation of insulin-expressing cells.


Assuntos
Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/fisiologia , Proteínas do Tecido Nervoso/genética , Ductos Pancreáticos/citologia , Ductos Pancreáticos/fisiologia , Adulto , Fatores Etários , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Biomarcadores , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio , Humanos , Insulina/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Ilhotas Pancreáticas/embriologia , Proteínas de Membrana/genética , Proteínas Nucleares , Ductos Pancreáticos/embriologia , Fenótipo , Receptores Notch , Fatores de Transcrição , Transcrição Gênica/fisiologia
14.
Free Radic Biol Med ; 42(1): 64-78, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17157194

RESUMO

AMP-activated protein kinase influences cellular metabolism, glucose-regulated gene expression, and insulin secretion of pancreatic beta cells. Its sustained activation by culture at low glucose concentrations or in the presence of 5-aminoimidazole-4-carboxamide riboside (AICAR) was shown to trigger apoptosis in beta cells. This study shows that both low glucose- and AICAR-induced apoptosis are associated with increased formation of mitochondrial superoxide-derived radicals and decreased mitochondrial activity. Mitochondrial dysfunction was reflected by an increased oxidized state of the mitochondrial flavins (FMN/FAD) but not of NAD(P)H. It was accompanied by suppression of glucose oxidation and glucose-induced insulin secretion, while palmitate oxidation appeared unaffected. When the cellular accumulation of superoxide-derived radicals was quenched by the ROS scavengers vitamin E, N-acetylcysteine, or the SOD-mimetic compound MnTBAP, apoptosis was significantly inhibited. Both low glucose and AICAR also elevated the expression of BH3-domain-only Bcl-2 antagonists, and induced caspase-3 activation, causing caspase-dependent truncation of Bcl-2. Overexpression of recombinant human Bcl-2 prevented caspase-3 activation, endogenous Bcl-2 processing, and apoptosis, but did not attenuate oxygen radical formation, AMPK activation, or JNK phosphorylation. We conclude that apoptosis by prolonged AMPK activation in beta cells results from enhanced production of mitochondria-derived oxygen radicals and onset of the intrinsic mitochondrial apoptosis pathway, followed by caspase activation and Bcl-2 cleavage which may amplify the death signal.


Assuntos
Apoptose/fisiologia , Células Secretoras de Insulina/metabolismo , Mitocôndrias/metabolismo , Complexos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases Ativadas por AMP , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Western Blotting , Caspase 3/metabolismo , Linhagem Celular , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Glucose/farmacologia , Peróxido de Hidrogênio/farmacologia , Hipoglicemiantes/metabolismo , Insulina/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Mitocôndrias/patologia , Palmitatos/farmacologia , Fosforilação , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleotídeos/farmacologia
15.
Antioxid Redox Signal ; 9(3): 309-17, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17184175

RESUMO

Excessive formation of oxygen radicals is a well-established mediator of hyperglycemic damage in diabetes to a wide range of tissues, such as neurons, retinal cells, and vascular endothelium. Increased oxygen radical formation is generally considered a toxic side effect of excessive rates of mitochondrial oxidative metabolism and electron transport in high glucose-exposed cells. Along the same line, metabolic oxidative stress is currently also regarded as crucial mediator of beta cell dysfunction and apoptosis under hyperglycemic conditions. Here the authors argue that a healthy beta cell is well equipped to deal adequately with elevated glucose metabolic rates, and demonstrate that decreased glucose catabolism leads to ROS production and apoptosis. They therefore propose that adverse metabolic conditions in poorly controlled diabetes (hyperglycemia and/or dyslipidemia) or genetic defects could decrease the viability of beta cells by interfering with normal glucose sensing and metabolism, rather than by overactivating it. This view is supported by the fragmentary data currently available on the pathways for hypergycemic and hypoglycemic beta cell death.


Assuntos
Apoptose/fisiologia , Glucose/fisiologia , Ilhotas Pancreáticas/citologia , Animais , Diferenciação Celular , Humanos
16.
Diabetes ; 52(10): 2497-502, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14514632

RESUMO

We previously reported that interleukin-1beta (IL-1beta) alone does not cause apoptosis of beta-cells, whereas when combined with gamma-interferon (IFN-gamma) and tumor necrosis factor-alpha (TNF-alpha), it exerts a distinct apoptotic effect. Studies in beta-cell lines indicated that IL-1beta reduced expression of islet brain (IB)-1/JNK interacting protein (JIP)-1, a JNK scaffold protein with antiapoptotic action. We examined whether variations in IB1/JIP-1 expression in purified primary beta-cells affect their susceptibility to cytokine-induced apoptosis. Exposure to IL-1beta for 24 h decreased cellular IB1/JIP-1 content by 66 +/- 17%; this IL-1beta effect was maintained in the presence of TNF-alpha + IFN-gamma, which did not influence IB1/JIP-1 levels by themselves. Addition of IL-1beta to TNF-alpha + IFN-gamma increased apoptosis from 20 +/- 2% to 59 +/- 5%. A similar increase in TNF-alpha + IFN-gamma-induced apoptosis was produced by adenoviral expression of antisense IB1/JIP-1 and was not further enhanced by addition of IL-1beta, indicating that IL-1beta-mediated suppression of IB1/JIP-1 in beta-cells increases their susceptibility to cytokine-induced apoptosis. However, adenovirally mediated overexpression of IB1/JIP-1 also potentiated TNF-alpha + IFN-gamma-induced apoptosis, suggesting that the antiapoptotic effect of IB1/JIP-1 depends on well-defined cellular levels. We conclude that the IB1/JIP-1 level in beta-cells can control their susceptibility to apoptosis independent of JNK signaling.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Apoptose/fisiologia , Proteínas de Transporte/metabolismo , Citocinas/farmacologia , Ilhotas Pancreáticas/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais/fisiologia , Transativadores/metabolismo , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Esquema de Medicação , Combinação de Medicamentos , Sinergismo Farmacológico , Técnicas de Transferência de Genes , Humanos , Interferon gama/genética , Interferon gama/farmacologia , Interleucina-1/administração & dosagem , Ilhotas Pancreáticas/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno , Masculino , Ratos , Ratos Wistar , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/farmacologia
17.
Eur J Pharmacol ; 518(2-3): 90-5, 2005 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-16039647

RESUMO

The drugs troglitazone and metformin are used to reduce the degree of insulin resistance in type 2 diabetes. Both compounds act through different mechanisms which might include opposing effects on the production of adiponectin, an insulin-sensitizer released by adipocytes. This study compared the effects of troglitazone and metformin on adiponectin production by 3T3-L1 adipocytes during 48 h culture. Troglitazone increased adiponectin mRNA and protein expression as well as release, whereas metformin did not affect transcription but reduced protein expression and release. The effect of metformin was also seen with phenformin, and with low-glucose culture, all conditions with a reduced mitochondrial activity and an activated AMP activated protein kinase (AMPK). Addition of the AMPK activator 5-aminoimidazole-4-carboxamide-riboside (AICAR) also caused a decrease in adiponectin protein expression. These data indicate that metformin and troglitazone exert opposing effects on adiponectin expression and release by differentiated 3T3-L1 adipocytes. The metformin-induced suppression involves an activation of AMP activated protein kinase.


Assuntos
Adipócitos/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Metformina/farmacologia , Complexos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Células 3T3-L1 , Proteínas Quinases Ativadas por AMP , Adipócitos/citologia , Adipócitos/metabolismo , Adiponectina , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Northern Blotting , Western Blotting , Cromanos/farmacologia , Meios de Cultivo Condicionados/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Hipoglicemiantes/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Fenformin/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ribonucleotídeos/farmacologia , Tiazolidinedionas/farmacologia , Troglitazona , Fator de Necrose Tumoral alfa/farmacologia
18.
Life Sci ; 77(11): 1273-82, 2005 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-15893773

RESUMO

The adipocyte-derived hormone adiponectin was recently shown to stimulate glucose-utilization and to increase fatty acid oxidation in liver and muscle. The effects were ascribed to adiponectin-receptor mediated activation of the key metabolic regulator AMP-activated protein kinase (AMPK). In pancreatic beta cells, AMPK-activation is known to affect cellular function. We therefore investigated a possible adiponectin-induced activation of AMPK in beta cells. RT-PCR analysis confirmed the expression of adiponectin receptor subtypes 1 and 2 in rat beta cells and showed their expression in insulin-secreting MIN6 cells. Culture with physiological concentrations (2.5 microg/ml) of globular adiponectin was found to increase the phosphorylation of both AMPK and acetylcoA carboxylase (ACC) in these cell types. Like the pharmacological AMPK activator 5-amino-imidazole-4-carboxamide-riboside (AICAR), adiponectin activated AMPK in beta cells and MIN6 cells. In short-term incubations of MIN6 cells with either adiponectin (2.5 microg/ml) or AICAR (1 mM), the flux of glucose-carbon to acyl CoA/cholesterol biosynthetic intermediates was reduced. We conclude that adiponectin induces an activation of AMPK in beta cells, which inhibits their cataplerosis of glucose-carbon to lipids.


Assuntos
Aminoimidazol Carboxamida/análogos & derivados , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Ilhotas Pancreáticas/enzimologia , Complexos Multienzimáticos/biossíntese , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Quinases Ativadas por AMP , Acetil-CoA Carboxilase/metabolismo , Adiponectina , Aminoimidazol Carboxamida/farmacologia , Animais , Western Blotting , Linhagem Celular , Células Cultivadas , Glucose/farmacologia , Lipídeos/biossíntese , Masculino , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleotídeos/farmacologia
19.
J Vis Exp ; (102): e52765, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26273954

RESUMO

Expansion of pancreatic beta cells in vivo or ex vivo, or generation of beta cells by differentiation from an embryonic or adult stem cell, can provide new expandable sources of beta cells to alleviate the donor scarcity in human islet transplantation as therapy for diabetes. Although recent advances have been made towards this aim, mechanisms that regulate beta cell expansion and differentiation from a stem/progenitor cell remain to be characterized. Here, we describe a protocol for an injury model in the adult mouse pancreas that can function as a tool to study mechanisms of tissue remodeling and beta cell proliferation and differentiation. Partial duct ligation (PDL) is an experimentally induced injury of the rodent pancreas involving surgical ligation of the main pancreatic duct resulting in an obstruction of drainage of exocrine products out of the tail region of the pancreas. The inflicted damage induces acinar atrophy, immune cell infiltration and severe tissue remodeling. We have previously reported the activation of Neurogenin (Ngn) 3 expressing endogenous progenitor-like cells and an increase in beta cell proliferation after PDL. Therefore, PDL provides a basis to study signals involved in beta cell dynamics and the properties of an endocrine progenitor in adult pancreas. Since, it still remains largely unclear, which factors and pathways contribute to beta cell neogenesis and proliferation in PDL, a standardized protocol for PDL will allow for comparison across laboratories.


Assuntos
Reprogramação Celular/fisiologia , Células Secretoras de Insulina/citologia , Pâncreas/lesões , Ductos Pancreáticos/cirurgia , Animais , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Humanos , Complicações Intraoperatórias/patologia , Ligadura/métodos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pâncreas/citologia
20.
Stem Cells Transl Med ; 4(6): 555-63, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25848123

RESUMO

UNLABELLED: Macrophages are classically considered detrimental for pancreatic ß-cell survival and function, thereby contributing to ß-cell failure in both type 1 (T1D) and 2 (T2D) diabetes mellitus. In addition, adipose tissue macrophages negatively influence peripheral insulin signaling and promote obesity-induced insulin resistance in T2D. In contrast, recent data unexpectedly uncovered that macrophages are not only able to protect ß cells during pancreatitis but also to orchestrate ß-cell proliferation and regeneration after ß-cell injury. Moreover, by altering their activation state, macrophages are able to improve insulin resistance in murine models of T2D. This review will elaborate on current insights in macrophage heterogeneity and on the evolving role of pancreas macrophages during organogenesis, tissue injury, and repair. Additional identification of macrophage subtypes and of their secreted factors might ultimately translate into novel therapeutic strategies for both T1D and T2D. SIGNIFICANCE: Diabetes mellitus is a pandemic disease, characterized by severe acute and chronic complications. Macrophages have long been considered prime suspects in the pathogenesis of both type 1 and 2 diabetes mellitus. In this concise review, current insights in macrophage heterogeneity and on the, as yet, underappreciated role of alternatively activated macrophages in insulin sensing and ß-cell development/repair are reported. Further identification of macrophage subtypes and of their secreted factors might ultimately translate into novel therapeutic strategies for diabetes mellitus.


Assuntos
Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Macrófagos/metabolismo , Regeneração , Animais , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Diabetes Mellitus Tipo 2/terapia , Humanos , Macrófagos/patologia , Pancreatite/metabolismo , Pancreatite/patologia , Pancreatite/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA