Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
Sci Rep ; 11(1): 17796, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34493754

RESUMO

Pancreatic islet cells have plasticity, such as the abilities to dedifferentiate and transdifferentiate. Islet cell conversion to other characteristic cell is largely determined by transcription factors, but significance of expression patterns of these transcription factors in human islet cells remained unclear. Here, we present the NKX6.1-positive ratio of glucagon-positive cells (NKX6.1+/GCG+ ratio) and the ARX-negative ratio of glucagon-positive cells (ARX-/GCG+ ratio) in 34 patients who were not administered antidiabetic agents. Both of NKX6.1+/GCG+ ratio and ARX-/GCG+ ratio negatively associated with relative beta cell area. And these ratios did not have significant correlation with other parameters including age, body mass index, hemoglobin A1c, fasting plasma glucose level or relative alpha-cell area. Our data demonstrate that these expression ratios of transcription factors in glucagon-positive cells closely correlate with the reduction of beta-cell volume in human pancreas.


Assuntos
Transdiferenciação Celular , Regulação da Expressão Gênica , Células Secretoras de Glucagon/metabolismo , Proteínas de Homeodomínio/biossíntese , Células Secretoras de Insulina/metabolismo , Fatores de Transcrição/biossíntese , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/uso terapêutico , Glicemia/análise , Peptídeo C/sangue , Tamanho Celular , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Feminino , Células Secretoras de Glucagon/ultraestrutura , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Intolerância à Glucose/patologia , Hemoglobinas Glicadas/análise , Proteínas de Homeodomínio/genética , Humanos , Células Secretoras de Insulina/ultraestrutura , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Pancreatectomia , Cisto Pancreático/genética , Cisto Pancreático/metabolismo , Cisto Pancreático/patologia , Cisto Pancreático/cirurgia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Pancreaticoduodenectomia , Fatores de Transcrição/genética
2.
Int J Mol Sci ; 21(19)2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-33019671

RESUMO

There are presently no reliable ways to quantify human pancreatic beta cell mass (BCM) in vivo, which prevents an accurate understanding of the progressive beta cell loss in diabetes or following islet transplantation. Furthermore, the lack of beta cell imaging hampers the evaluation of the impact of new drugs aiming to prevent beta cell loss or to restore BCM in diabetes. We presently discuss the potential value of BCM determination as a cornerstone for individualized therapies in diabetes, describe the presently available probes for human BCM evaluation, and discuss our approach for the discovery of novel beta cell biomarkers, based on the determination of specific splice variants present in human beta cells. This has already led to the identification of DPP6 and FXYD2ga as two promising targets for human BCM imaging, and is followed by a discussion of potential safety issues, the role for radiochemistry in the improvement of BCM imaging, and concludes with an overview of the different steps from pre-clinical validation to a first-in-man trial for novel tracers.


Assuntos
Diabetes Mellitus Tipo 1/diagnóstico por imagem , Diabetes Mellitus Tipo 2/diagnóstico por imagem , Células Secretoras de Insulina/ultraestrutura , Transplante das Ilhotas Pancreáticas/diagnóstico por imagem , Compostos Radiofarmacêuticos/química , Anticorpos de Domínio Único/química , 5-Hidroxitriptofano/química , 5-Hidroxitriptofano/farmacocinética , Animais , Biomarcadores/análise , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Dipeptidil Peptidases e Tripeptidil Peptidases/genética , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Exenatida/química , Exenatida/farmacocinética , Radioisótopos de Flúor/química , Radioisótopos de Flúor/farmacocinética , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/transplante , Imageamento por Ressonância Magnética/métodos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Canais de Potássio/genética , Canais de Potássio/metabolismo , Compostos Radiofarmacêuticos/farmacocinética , Anticorpos de Domínio Único/metabolismo , ATPase Trocadora de Sódio-Potássio/genética , ATPase Trocadora de Sódio-Potássio/metabolismo , Tecnécio/química , Tecnécio/metabolismo , Tetrabenazina/análogos & derivados , Tetrabenazina/química , Tetrabenazina/farmacocinética , Tomografia Computadorizada de Emissão de Fóton Único/métodos
3.
Front Immunol ; 11: 1814, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33101266

RESUMO

Beta cell failure and apoptosis following islet inflammation have been associated with autoimmune type 1 diabetes pathogenesis. As conveyors of biological active material, extracellular vesicles (EV) act as mediators in communication with immune effectors fostering the idea that EV from inflamed beta cells may contribute to autoimmunity. Evidence accumulates that beta exosomes promote diabetogenic responses, but relative contributions of larger vesicles as well as variations in the composition of the beta cell's vesiculome due to environmental changes have not been explored yet. Here, we made side-by-side comparisons of the phenotype and function of apoptotic bodies (AB), microvesicles (MV) and small EV (sEV) isolated from an equal amount of MIN6 beta cells exposed to inflammatory, hypoxic or genotoxic stressors. Under normal conditions, large vesicles represent 93% of the volume, but only 2% of the number of the vesicles. Our data reveal a consistently higher release of AB and sEV and to a lesser extent of MV, exclusively under inflammatory conditions commensurate with a 4-fold increase in the total volume of the vesiculome and enhanced export of immune-stimulatory material including the autoantigen insulin, microRNA, and cytokines. Whilst inflammation does not change the concentration of insulin inside the EV, specific Toll-like receptor-binding microRNA sequences preferentially partition into sEV. Exposure to inflammatory stress engenders drastic increases in the expression of monocyte chemoattractant protein 1 in all EV and of interleukin-27 solely in AB suggesting selective sorting toward EV subspecies. Functional in vitro assays in mouse dendritic cells and macrophages reveal further differences in the aptitude of EV to modulate expression of cytokines and maturation markers. These findings highlight the different quantitative and qualitative imprints of environmental changes in subpopulations of beta EV that may contribute to the spread of inflammation and sustained immune cell recruitment at the inception of the (auto-) immune response.


Assuntos
Citocinas/metabolismo , Vesículas Extracelulares/metabolismo , Inflamação/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animais , Apoptose , Hipóxia Celular , Linhagem Celular Tumoral , Dano ao DNA , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/ultraestrutura , Feminino , Inflamação/imunologia , Inflamação/patologia , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/ultraestrutura , Ativação de Macrófagos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos NOD , MicroRNAs/metabolismo , Fenótipo , Células RAW 264.7 , Via Secretória , Transdução de Sinais
4.
Pancreas ; 49(9): 1225-1231, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32898009

RESUMO

OBJECTIVES: We aimed to determine whether responsive insulin-producing cells (IPCs) could be generated from adipose-derived stem cells (ADSCs) isolated from patients with type 1 diabetes mellitus (T1DM). METHODS: We isolated ADSCs from adipose tissue of 4 patients (one patient with T1DM and 3 nondiabetic patients), who underwent surgery and differentiated them into IPCs with using a 2-step xeno-antigen free, 3-dimensional culture method. Characteristics of isolated ADSCs, in vitro cell quality, programmed cell death ligand-1 (PDL-1) expression, and transplantation into streptozotocin induced diabetic nude mice were investigated. RESULTS: Adipose-derived stem cells from T1DM patients and commercially obtained ADSCs showed the same surface markers; CD31CD34CD45CD90CD105CD146. Moreover, the generated IPCs at day 21 demonstrated appropriate autonomous insulin secretion (stimulation index, 3.5; standard deviation, 0.8). Nonfasting blood glucose concentrations of IPC-transplanted mice were normal at 30 days. The normalized rate of IPC-transplanted mice was significantly higher than that of the sham-operated group (P < 0.05). Insulin-producing cells generated from T1DM adipose tissue expressed high levels of PDL-1. CONCLUSIONS: Insulin-producing cells obtained from adipose tissue of T1DM patients are capable of secreting insulin long-term and achieve normoglycemia after transplantation. Expression of PDL-1 suggests the potential for immune circumvention.


Assuntos
Tecido Adiposo/citologia , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/patologia , Células Secretoras de Insulina/citologia , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Animais , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Glucose/farmacologia , Humanos , Insulina/metabolismo , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/ultraestrutura , Camundongos Nus , Microscopia Eletrônica de Transmissão , Células-Tronco/metabolismo
5.
Cells ; 9(1)2020 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-31936632

RESUMO

Nor1, the third member of the Nr4a subfamily of nuclear receptor, is garnering increased interest in view of its role in the regulation of glucose homeostasis. Our previous study highlighted a proapoptotic role of Nor1 in pancreatic beta cells and showed that Nor1 expression was increased in islets isolated from type 2 diabetic individuals, suggesting that Nor1 could mediate the deterioration of islet function in type 2 diabetes. However, the mechanism remains incompletely understood. We herein investigated the subcellular localization of Nor1 in INS832/13 cells and dispersed human beta cells. We also examined the consequences of Nor1 overexpression on mitochondrial function and morphology. Our results show that, surprisingly, Nor1 is mostly cytoplasmic in beta cells and undergoes mitochondrial translocation upon activation by proinflammatory cytokines. Mitochondrial localization of Nor1 reduced glucose oxidation, lowered ATP production rates, and inhibited glucose-stimulated insulin secretion. Western blot and microscopy images revealed that Nor1 could provoke mitochondrial fragmentation via mitophagy. Our study unveils a new mode of action for Nor1, which affects beta-cell viability and function by disrupting mitochondrial networks.


Assuntos
Células Secretoras de Insulina/metabolismo , Mitocôndrias/metabolismo , Membro 3 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Linhagem Celular , Citocinas/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/ultraestrutura , Mitocôndrias/ultraestrutura , Mitofagia , Oxirredução
6.
Theranostics ; 10(1): 398-410, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31903128

RESUMO

Non-invasive imaging of ß-cells represents a desirable preclinical and clinical tool to monitor the change of ß-cell mass and the loss of function during pre-diabetic stages. Although it is widely accepted that manganese (Mn) ions are actively gated by voltage-dependent calcium channels (VDCC) in response to glucose metabolism, little is known on its specificity in vivo for quantification of islet ß-cell function using Mn and magnetic resonance imaging (MRI). On the other hand, glucagon-like-peptide-1 receptor (GLP-1R) represents a validated target for the estimation of ß-cell mass using radiolabeled exendin-4 (Ex4) and positron emission tomography (PET). However, a multiparametric imaging workflow revealing ß-cell mass and function quantitatively is still missing. Methods: We developed a simultaneous PET/MRI protocol to comprehensively quantify in vivo changes in ß-cell mass and function by targeting, respectively, GLP-1R and VDCC coupled with insulin secretion. Differences in the spatial distribution of Mn and radiolabeled Ex4 were monitored overtime in native and transgenic pancreata, characterized by spontaneous pancreatic neuroendocrine tumor development. Follow-up with mass spectrometry imaging (MSI) and autoradiography allowed the ex vivo validation of the specificity of Mn and PET tracer uptake and the detection of endogenous biometals, such as calcium and zinc, throughout the endocrine and exocrine pancreas. Results: Our in vivo data based on a volumetric PET/MRI readout for native pancreata and insulinomas connects uptake of Mn measured at early imaging time points to high non-specific binding by the exocrine tissue, while specific retention was only found 24 h post injection. These results are supported by cross-validation of the spatial distribution of exogenous 55Mn and endogenous 44Ca and 64Zn as well with the specific internalization of the radiolabeled peptide targeting GLP-1R. Conclusion: Simultaneous PET/MR imaging of the pancreas enabled the comprehensive in vivo quantification of ß-cell function and mass using Mn and radiolabeled Ex4. Most important, our data revealed that only late time-point measurements reflect the Mn uptake in the islet ß-cells, while early time points detect non-specific accumulation of Mn in the exocrine pancreas.


Assuntos
Células Secretoras de Insulina , Imageamento por Ressonância Magnética , Pâncreas , Neoplasias Pancreáticas/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Animais , Canais de Cálcio/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/ultraestrutura , Manganês/química , Camundongos , Camundongos Transgênicos , Pâncreas/diagnóstico por imagem , Pâncreas/patologia , Compostos Radiofarmacêuticos/química
7.
Mol Cell Endocrinol ; 503: 110700, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31904405

RESUMO

Genetic and epigenetic factors contribute equally to the pathogenesis of type 1 diabetes mellitus. Sodium butyrate (NaB) has been reported to improve glucose homeostasis by modulation of the p38/ERK MAPK pathway. This work aims to evaluate the effect of NaB on the ultrastructure of pancreatic ß-cells and the PI3/AKT pathway. Juvenile albino male rats were used to establish a type 1 diabetes model using streptozotocin injection and NaB in a pre- and post-treatment schedule. Plasma glucose, insulin levels, and glucose tolerance were evaluated. Light and electron microscopy and immunohistochemistry were performed using Ki-67, caspase-3, and insulin. NaB treatment resulted in a significant improvement in plasma glucose levels, plasma insulin levels/expression, and ameliorated diabetes-induced histological alternations. Additionally, it increased the expression of phosphorylated AKT. These findings provide evidence that NaB may be useful in the treatment of juvenile diabetes.


Assuntos
Ácido Butírico/farmacologia , Diabetes Mellitus Experimental , Inibidores de Histona Desacetilases/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Animais , Ácido Butírico/uso terapêutico , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Glucose/metabolismo , Inibidores de Histona Desacetilases/uso terapêutico , Resistência à Insulina , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/ultraestrutura , Masculino , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos
8.
Diabetes ; 69(3): 342-354, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31836690

RESUMO

Human but not mouse islets transplanted into immunodeficient NSG mice effectively accumulate lipid droplets (LDs). Because chronic lipid exposure is associated with islet ß-cell dysfunction, we investigated LD accumulation in the intact human and mouse pancreas over a range of ages and states of diabetes. Very few LDs were found in normal human juvenile pancreatic acinar and islet cells, with numbers subsequently increasing throughout adulthood. While accumulation appeared evenly distributed in postjuvenile acinar and islet cells in donors without diabetes, LDs were enriched in islet α- and ß-cells from donors with type 2 diabetes (T2D). LDs were also found in the islet ß-like cells produced from human embryonic cell-derived ß-cell clusters. In contrast, LD accumulation was nearly undetectable in the adult rodent pancreas, even in hyperglycemic and hyperlipidemic models or 1.5-year-old mice. Taken together, there appear to be significant differences in pancreas islet cell lipid handling between species, and the human juvenile and adult cell populations. Moreover, our results suggest that LD enrichment could be impactful to T2D islet cell function.


Assuntos
Diabetes Mellitus Tipo 2/patologia , Células Secretoras de Glucagon/patologia , Células Secretoras de Insulina/patologia , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/patologia , Gotículas Lipídicas/patologia , Células Acinares/patologia , Células Acinares/ultraestrutura , Adolescente , Adulto , Fatores Etários , Idoso , Animais , Criança , Pré-Escolar , Diabetes Mellitus Experimental/patologia , Células-Tronco Embrionárias , Feminino , Células Secretoras de Glucagon/ultraestrutura , Humanos , Lactente , Células Secretoras de Insulina/ultraestrutura , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/ultraestrutura , Gotículas Lipídicas/ultraestrutura , Masculino , Camundongos , Microscopia Eletrônica , Microscopia de Fluorescência , Pessoa de Meia-Idade , Ratos , Doadores de Tecidos , Adulto Jovem
9.
Nat Commun ; 10(1): 3700, 2019 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-31420552

RESUMO

Little is known about the role of islet delta cells in regulating blood glucose homeostasis in vivo. Delta cells are important paracrine regulators of beta cell and alpha cell secretory activity, however the structural basis underlying this regulation has yet to be determined. Most delta cells are elongated and have a well-defined cell soma and a filopodia-like structure. Using in vivo optogenetics and high-speed Ca2+ imaging, we show that these filopodia are dynamic structures that contain a secretory machinery, enabling the delta cell to reach a large number of beta cells within the islet. This provides for efficient regulation of beta cell activity and is modulated by endogenous IGF-1/VEGF-A signaling. In pre-diabetes, delta cells undergo morphological changes that may be a compensation to maintain paracrine regulation of the beta cell. Our data provides an integrated picture of how delta cells can modulate beta cell activity under physiological conditions.


Assuntos
Ilhotas Pancreáticas/ultraestrutura , Comunicação Parácrina , Estado Pré-Diabético/patologia , Pseudópodes/ultraestrutura , Células Secretoras de Somatostatina/ultraestrutura , Animais , Glicemia/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/ultraestrutura , Microscopia Intravital , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Imagem Óptica , Optogenética , Estado Pré-Diabético/metabolismo , Pseudópodes/metabolismo , Células Secretoras de Somatostatina/citologia , Células Secretoras de Somatostatina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Cell Rep ; 28(3): 759-772.e10, 2019 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31315053

RESUMO

Mechanisms coordinating pancreatic ß cell metabolism with insulin secretion are essential for glucose homeostasis. One key mechanism of ß cell nutrient sensing uses the mitochondrial GTP (mtGTP) cycle. In this cycle, mtGTP synthesized by succinyl-CoA synthetase (SCS) is hydrolyzed via mitochondrial PEPCK (PEPCK-M) to make phosphoenolpyruvate, a high-energy metabolite that integrates TCA cycling and anaplerosis with glucose-stimulated insulin secretion (GSIS). Several strategies, including xenotopic overexpression of yeast mitochondrial GTP/GDP exchanger (GGC1) and human ATP and GTP-specific SCS isoforms, demonstrated the importance of the mtGTP cycle. These studies confirmed that mtGTP triggers and amplifies normal GSIS and rescues defects in GSIS both in vitro and in vivo. Increased mtGTP synthesis enhanced calcium oscillations during GSIS. mtGTP also augmented mitochondrial mass, increased insulin granule number, and membrane proximity without triggering de-differentiation or metabolic fragility. These data highlight the importance of the mtGTP signal in nutrient sensing, insulin secretion, mitochondrial maintenance, and ß cell health.


Assuntos
Trifosfato de Adenosina/metabolismo , Glucose/metabolismo , Guanosina Trifosfato/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Mitocôndrias/metabolismo , Succinato-CoA Ligases/metabolismo , Animais , Diferenciação Celular/genética , Linhagem Celular , Proliferação de Células/genética , Ciclo do Ácido Cítrico/genética , Homeostase , Humanos , Secreção de Insulina/genética , Secreção de Insulina/fisiologia , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Mitocôndrias/enzimologia , Mitocôndrias/ultraestrutura , Membranas Mitocondriais/metabolismo , Fosforilação Oxidativa , Fosfoenolpiruvato Carboxiquinase (ATP)/metabolismo , Regulação para Cima
11.
Sci Rep ; 9(1): 4965, 2019 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-30899071

RESUMO

Type I diabetes (T1D) is caused by immune-mediated destruction of pancreatic beta cells. This process is triggered, in part, by specific (aa 9-23) epitopes of the insulin Β chain. Previously, fish insulins were used clinically in patients allergic to bovine or porcine insulin. Fish and human insulin differ by two amino acids in the critical immunogenic region (aa 9-23) of the B chain. We hypothesized that ß cells synthesizing fish insulin would be less immunogenic in a mouse model of T1D. Transgenic NOD mice in which Greater Amberjack fish (Seriola dumerili) insulin was substituted for the insulin 2 gene were generated (mouse Ins1-/- mouse Ins2-/- fish Ins2+/+). In these mice, pancreatic islets remained free of autoimmune attack. To determine whether such reduction in immunogenicity is sufficient to protect ß cells from autoimmunity upon transplantation, we transplanted fish Ins2 transgenic (expressing solely Seriola dumerili Ins2), NOD, or B16:A-dKO islets under the kidney capsules of 5 weeks old female NOD wildtype mice. The B:Y16A Β chain substitution has been previously shown to be protective of T1D in NOD mice. NOD mice receiving Seriola dumerili transgenic islet transplants showed a significant (p = 0.004) prolongation of their euglycemic period (by 6 weeks; up to 18 weeks of age) compared to un-manipulated female NOD (diabetes onset at 12 weeks of age) and those receiving B16:A-dKO islet transplants (diabetes onset at 12 weeks of age). These data support the concept that specific amino acid sequence modifications can reduce insulin immunogenicity. Additionally, our study shows that alteration of a single epitope is not sufficient to halt an ongoing autoimmune response. Which, and how many, T cell epitopes are required and suffice to perpetuate autoimmunity is currently unknown. Such studies may be useful to achieve host tolerance to ß cells by inactivating key immunogenic epitopes of stem cell-derived ß cells intended for transplantation.


Assuntos
Células Secretoras de Insulina/imunologia , Insulina/genética , Sequência de Aminoácidos , Animais , Linfócitos T CD4-Positivos/imunologia , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/terapia , Epitopos/imunologia , Humanos , Insulina/química , Células Secretoras de Insulina/ultraestrutura , Transplante das Ilhotas Pancreáticas , Rim/imunologia , Ativação Linfocitária/imunologia , Camundongos Endogâmicos NOD , Camundongos Transgênicos
12.
Nucleic Acids Res ; 47(8): 4039-4053, 2019 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-30722038

RESUMO

FBXW7, a classic tumor suppressor, is a substrate recognition subunit of the Skp1-cullin-F-box (SCF) ubiquitin ligase that targets oncoproteins for ubiquitination and degradation. We recently found that FBXW7 is recruited to DNA damage sites to facilitate nonhomologous end-joining (NHEJ). The detailed underlying molecular mechanism, however, remains elusive. Here we report that the WD40 domain of FBXW7, which is responsible for substrate binding and frequently mutated in human cancers, binds to poly(ADP-ribose) (PAR) immediately following DNA damage and mediates rapid recruitment of FBXW7 to DNA damage sites, whereas ATM-mediated FBXW7 phosphorylation promotes its retention at DNA damage sites. Cancer-associated arginine mutations in the WD40 domain (R465H, R479Q and R505C) abolish both FBXW7 interaction with PAR and recruitment to DNA damage sites, causing inhibition of XRCC4 polyubiquitination and NHEJ. Furthermore, inhibition or silencing of poly(ADP-ribose) polymerase 1 (PARP1) inhibits PAR-mediated recruitment of FBXW7 to the DNA damage sites. Taken together, our study demonstrates that the WD40 domain of FBXW7 is a novel PAR-binding motif that facilitates early recruitment of FBXW7 to DNA damage sites for subsequent NHEJ repair. Abrogation of this ability seen in cancer-derived FBXW7 mutations provides a molecular mechanism for defective DNA repair, eventually leading to genome instability.


Assuntos
Reparo do DNA por Junção de Extremidades , Proteína 7 com Repetições F-Box-WD/genética , Poli(ADP-Ribose) Polimerase-1/genética , Poli Adenosina Difosfato Ribose/metabolismo , Fator de Células-Tronco/genética , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Sítios de Ligação , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos da radiação , Dano ao DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteína 7 com Repetições F-Box-WD/química , Proteína 7 com Repetições F-Box-WD/metabolismo , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Fibroblastos/ultraestrutura , Raios gama , Células HCT116 , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/efeitos da radiação , Células Secretoras de Insulina/ultraestrutura , Modelos Moleculares , Mutação , Poli(ADP-Ribose) Polimerase-1/metabolismo , Poli Adenosina Difosfato Ribose/química , Ligação Proteica , Domínios Proteicos , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Fator de Células-Tronco/metabolismo , Ubiquitinação/efeitos da radiação
13.
Int J Nanomedicine ; 14: 371-382, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30662261

RESUMO

BACKGROUND: Islet cell transplantation is one of the key treatments for type 1 diabetes. Understanding the mechanisms of insulin fusion and exocytosis are of utmost importance for the improvement of the current islet cell transplantation and treatment of diabetes. These phenomena have not been fully evaluated due either to the lack of proper dynamic imaging, or the lack of proper cell preservation during imaging at nanoscales. METHODS: By maintaining the native environment of pancreatic ß-cells between two graphene monolayer sheets, we were able to monitor the subcellular events using in situ graphene liquid cell (GLC)-transmission electron microscopy (TEM) with both high temporal and high spatial resolution. RESULTS: For the first time, the nucleation and growth of insulin particles until the later stages of fusion were imaged at nanometer scales. The release of insulin from plasma membrane involves the degradation of plasma membrane and drastic reductions in the shorter axis of the insulin particles. Sequential exocytosis results indicated the nucleation, growth and attachment of the new insulin particles to the already anchored ones, which is thermodynamically favorable due to the reduction in total surface, further reducing the Gibbs free energy. The retraction of the already anchored insulin toward the cell is also monitored for the first time live at nanoscale resolution. CONCLUSION: Investigation of insulin granule dynamics in ß-cells can be investigated via GLC-TEM. Our findings with this technology open new realms for the development of novel drugs on pathological pancreatic ß-cells, because this approach facilitates observing the effects of the stimuli on the live cells and insulin granules.


Assuntos
Grafite/química , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/ultraestrutura , Microscopia Eletrônica de Transmissão , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Exocitose , Insulina/metabolismo , Fusão de Membrana , Camundongos
14.
Cell Rep ; 24(1): 181-196, 2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-29972779

RESUMO

Conjugated estrogens (CE) delay the onset of type 2 diabetes (T2D) in postmenopausal women, but the mechanism is unclear. In T2D, the endoplasmic reticulum (ER) fails to promote proinsulin folding and, in failing to do so, promotes ER stress and ß cell dysfunction. We show that CE prevent insulin-deficient diabetes in male and in female Akita mice using a model of misfolded proinsulin. CE stabilize the ER-associated protein degradation (ERAD) system and promote misfolded proinsulin proteasomal degradation. This involves activation of nuclear and membrane estrogen receptor-α (ERα), promoting transcriptional repression and proteasomal degradation of the ubiquitin-conjugating enzyme and ERAD degrader, UBC6e. The selective ERα modulator bazedoxifene mimics CE protection of ß cells in females but not in males.


Assuntos
Diabetes Mellitus/metabolismo , Estrogênios/farmacologia , Proinsulina/biossíntese , Dobramento de Proteína , Proteólise , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Diabetes Mellitus/prevenção & controle , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/ultraestrutura , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Degradação Associada com o Retículo Endoplasmático/efeitos dos fármacos , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Indóis/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/ultraestrutura , Masculino , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Dobramento de Proteína/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Elementos de Resposta/genética , Enzimas de Conjugação de Ubiquitina/metabolismo
15.
J Cell Physiol ; 233(12): 9375-9382, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29923197

RESUMO

We aimed to explore the effects of Inflammatory cytokines (IL-1ß, IFN-γ, TNF-α) on pancreatic ß-cells. CCK-8 assay showed that the cell viability decreased after 24 hr treatment of TNF-α, 48 hr of IFN-γ, and 84 hr of IL-1ß. EdU assay illustrated that after 24 hr treatment, there were significantly reduced EdU-labeled red fluorescence cells in TNF-α group while not in IFN-γ and IL-1ß groups. Flow Cytometry results displayed that TNF-α and IFN-γ groups increased apoptosis while IL-1ß group did not. Cell apoptosis results found that there was an increase in the S-phase population of IL-1ß and TNF-α groups, however, there was no significant difference in cell cycle between IFN-γ group and the control. TEM images showed that there were reduction in the number of granules and mitochondria in IL-1ß and IFN-γ groups, in particular paucity of insulin granules and mitochondria in TNF-α group. Radioimmunoassay results presented that TNF-α inhibited glucose-induced insulin secretion, while there were no significant changes in IL-1ß and IFN-γ groups when compared with the control. Metabolomic analysis found amino acid metabolism and Krebs cycle were the most robust altered metabolism pathways after inflammatory cytokines treatments. Overall, the altered amino acid metabolism and Krebs cycle metabolism might be important mechanisms of TNF-α induced mouse pancreatic ß-cells dysfuction.


Assuntos
Células Secretoras de Insulina/citologia , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Mediadores da Inflamação/farmacologia , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/ultraestrutura , Camundongos
16.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(4): 384-389, 2018 Apr 20.
Artigo em Chinês | MEDLINE | ID: mdl-29735436

RESUMO

OBJECTIVE: To observe the protective effects of potassium channel opener nicorandil against cognitive dysfunction in mice with streptozotocin (STZ)-induced diabetes. METHODS: C57BL/6J mouse models of type 1 diabetes mellitus (T1DM) were established by intraperitoneal injection of STZ and received daily treatment with intragastric administration of nicorandil or saline (model group) for 4 consecutive weeks, with normal C57BL/6J mice serving as control. Fasting blood glucose level was recorded every week and Morris water maze was used to evaluate the cognitive behavior of the mice in the 4th week. At the end of the experiment, the mice were sacrificed to observe the ultrastructural changes in the hippocampus and pancreas under transmission electron microscopy; the contents of glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) in the hippocampus and SOD activity and MDA level in the brain tissue were determined. RESULTS: Compared with the control group, the model group showed significantly increased fasting blood glucose (P<0.001), significantly prolonged escape latency (P<0.05) and increased swimming distance (P<0.01) with ultrastructural damage of pancreatic ß cells and in the hippocampus; GIP and GLP-1 contents in the hippocampus (P<0.01) and SOD activity in the brain were significantly decreased (P<0.05) and MDA content was significantly increased in the model group (P<0.05). Compared with the model group, nicorandil treatment did not cause significant changes in fasting blood glucose, but significantly reduced the swimming distance (P<0.05); nicorandil did not improve the ultrastructural changes in pancreatic ß cells but obviously improved the ultrastructures of hippocampal neurons and synapses. Nicorandil also significantly increased the contents of GIP and GLP-1 in the hippocampus (P<0.05), enhanced SOD activity (P<0.05) and decreased MDA level (P<0.01) in the brain tissue. CONCLUSION: Nicorandil improves cognitive dysfunction in mice with STZ-induced diabetes by increasing GIP and GLP-1 contents in the hippocampus and promoting antioxidation to relieve hippocampal injury.


Assuntos
Disfunção Cognitiva/tratamento farmacológico , Diabetes Mellitus Experimental/complicações , Nicorandil/farmacologia , Animais , Glicemia , Diabetes Mellitus Experimental/induzido quimicamente , Polipeptídeo Inibidor Gástrico/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/ultraestrutura , Malondialdeído/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Estreptozocina , Superóxido Dismutase/metabolismo
17.
Cell Death Dis ; 9(5): 481, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29705815

RESUMO

Human islet amyloid polypeptide (hIAPP), or amylin, has the tendency to aggregate into insoluble amyloid fibrils, a typical feature of islets from type 2 diabetes individuals. Thus, we investigated comparatively the impact of hIAPP on key pathways involved in pancreatic beta survival. INS1E-hIAPP cells present a hyperactivation of MTORC1 and an inhibition of autophagy signaling, those cells showing an increase in cell size. Resveratrol, a MTORC1 inhibitor, can reverse TSC2 degradation that occurs in INS1E-hIAPP cells and diminished MTORC1 hyperactivation with concomitant autophagy stimulation. At the same time, a blockade in mitophagy was found in INS1E-hIAPP cells, as compared with control or INS1E-rIAPP cells. Consistently, human amylin overexpression generates a basal induction of nitrotyrosine levels and polyubiquitinated aggregates. Failure of the protein degradation machinery finally results in an accumulation of damaged and fissioned mitochondria, ROS production, and increased susceptibility to endoplasmic reticulum (ER)-stress-induced apoptosis. Overall, hIAPP overexpression in INS1E cells induced MTORC1 activation and mitophagy inhibition, favoring a pro-fission scenario of damaged mitochondria, these cells turn out to be more susceptible to the ER-stress-induced apoptosis and malfunction.


Assuntos
Células Secretoras de Insulina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Mitocôndrias/metabolismo , Dinâmica Mitocondrial , Mitofagia , Animais , Apoptose , Linhagem Celular Tumoral , Estresse do Retículo Endoplasmático , Humanos , Células Secretoras de Insulina/ultraestrutura , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Mitocôndrias/ultraestrutura , Proteólise , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Proteína 2 do Complexo Esclerose Tuberosa/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo , Ubiquitinação , Regulação para Cima
18.
Glycobiology ; 28(6): 382-391, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29548035

RESUMO

Blood group B glycosphingolipids (B-GSLs) are substrates of the lysosomal alpha-galactosidase A (AGAL). Similar to its major substrate-globotriaosylceramide (Gb3Cer)-B-GSLs are not degraded and accumulate in the cells of patients affected by an inherited defect of AGAL activity (Fabry disease-FD).The pancreas is a secretory organ known to have high biosynthesis of blood group GSLs. Herein, we provide a comprehensive overview of the biochemical and structural abnormalities in pancreatic tissue from two male FD patients with blood group B. In both patients, we found major accumulation of a variety of complex B-GSLs carrying predominantly hexa- and hepta-saccharide structures. The subcellular pathology was dominated by deposits containing B-glycoconjugates and autofluorescent ceroid. The contribution of Gb3Cer to the storage was minor. This abnormal storage pattern was specific for the pancreatic acinar epithelial cells. Other pancreatic cell types including those of islets of Langerhans were affected much less or not at all.Altogether, we provide evidence for a key role of B-antigens in the biochemical and morphological pathology of the exocrine pancreas in FD patients with blood group B. We believe that our findings will trigger further studies aimed at assessing the potential pancreatic dysfunction in this disease.


Assuntos
Doença de Fabry/metabolismo , Glicoesfingolipídeos/metabolismo , Pâncreas/metabolismo , Sistema ABO de Grupos Sanguíneos/metabolismo , Células Acinares/metabolismo , Células Acinares/ultraestrutura , Estudos de Casos e Controles , Doença de Fabry/sangue , Doença de Fabry/patologia , Galactose/análise , Galactose/metabolismo , Glicoesfingolipídeos/química , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/ultraestrutura , Masculino , Pessoa de Meia-Idade , Pâncreas/ultraestrutura
19.
Mol Cell Endocrinol ; 473: 186-193, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29409957

RESUMO

It has been reported that the incretin system, including regulated GLP-1 secretion and locally expressed DPP-4, is present in pancreatic islets. In this study we comprehensively evaluated the expression and role of DPP-4 in islet alpha and beta cells from non-diabetic (ND) and type 2 diabetic (T2D) individuals, including the effects of its inhibition on beta cell function and survival. Isolated islets were prepared from 25 ND and 18 T2D organ donors; studies were also performed with the human insulin-producing EndoC-ßH1 cells. Morphological (including confocal microscopy), ultrastructural (electron microscopy, EM), functional (glucose-stimulated insulin secretion), survival (EM and nuclear dyes) and molecular (RNAseq, qPCR and western blot) studies were performed under several different experimental conditions. DPP-4 co-localized with glucagon and was also expressed in human islet insulin-containing cells. Furthermore, DPP-4 was expressed in EndoC-ßH1 cells. The proportions of DPP-4 positive alpha and beta cells and DPP-4 gene expression were significantly lower in T2D islets. A DPP-4 inhibitor protected ND human beta cells and EndoC-ßH1 cells against cytokine-induced toxicity, which was at least in part independent from GLP1 and associated with reduced NFKB1 expression. Finally, DPP-4 inhibition augmented glucose-stimulated insulin secretion, reduced apoptosis and improved ultrastructure in T2D beta cells. These results demonstrate the presence of DPP-4 in human islet alpha and beta cells, with reduced expression in T2D islets, and show that DPP-4 inhibition has beneficial effects on human ND and T2D beta cells. This suggests that DPP-4, besides playing a role in incretin effects, directly affects beta cell function and survival.


Assuntos
Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/patologia , Dipeptidil Peptidase 4/metabolismo , Células Secretoras de Insulina/enzimologia , Idoso , Apoptose/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Citocinas/toxicidade , Citoproteção/efeitos dos fármacos , Inibidores da Dipeptidil Peptidase IV/farmacologia , Regulação para Baixo/efeitos dos fármacos , Feminino , Humanos , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/ultraestrutura , Masculino , Pessoa de Meia-Idade
20.
J Struct Biol ; 201(1): 15-25, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29078993

RESUMO

In cryogenic correlated light and electron microscopy (cryo-CLEM), frozen targets of interest are identified and located on EM grids by fluorescence microscopy and then imaged at higher resolution by cryo-EM. Whilst working with these methods, we discovered that a variety of mammalian cells exhibit strong punctate autofluorescence when imaged under cryogenic conditions (80 K). Autofluorescence originated from multilamellar bodies (MLBs) and secretory granules. Here we describe a method to distinguish fluorescent protein tags from these autofluorescent sources based on the narrower emission spectrum of the former. The method is first tested on mitochondria and then applied to examine the ultrastructural variability of secretory granules within insulin-secreting pancreatic beta-cell-derived INS-1E cells.


Assuntos
Microscopia Crioeletrônica/métodos , Microscopia de Fluorescência/métodos , Mitocôndrias/ultraestrutura , Vesículas Secretórias/ultraestrutura , Animais , Linhagem Celular Tumoral , Fibroblastos/citologia , Fibroblastos/metabolismo , Fluoresceína-5-Isotiocianato/química , Fluorescência , Células HeLa , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/ultraestrutura , Proteínas Luminescentes/química , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Mitocôndrias/metabolismo , Ratos , Vesículas Secretórias/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA