Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Pflugers Arch ; 471(2): 337-345, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30310992

RESUMO

The clonal INS-1E beta-cell line has proven to be instrumental for numerous studies investigating the mechanisms of glucose-stimulated insulin secretion. The composition of its culture medium has not changed over the years, although some compounds have been recently highlighted for their effects on tissue differentiation. The present study investigated the effects of long-term treatment of INS-1E cells with 1 µM resveratrol on glucose-stimulated insulin secretion, testing an extended glucose dose response. The data demonstrate that chronic exposure to low-dose resveratrol expands the range of the glucose dose response of INS-1E cells beyond 15 mM glucose. We also assessed whether such beneficial effects could be retained after resveratrol withdrawal from the culture medium. This was not the case as INS-1E cells deprived of resveratrol returned to the phenotype of naïve cells, i.e., exhibiting a plateau phase at 15 mM glucose. Of note, although resveratrol has antioxidant properties, it cannot substitute for ß-mercaptoethanol normally present in the medium of INS-1E cells as a reducing agent. In conclusion, the addition of resveratrol as a standard component of the culture medium of INS-1E cells improves glucose-stimulated insulin secretion.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Glucose/metabolismo , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Resveratrol/farmacologia , Animais , Antioxidantes/metabolismo , Linhagem Celular Tumoral , Células Secretoras de Insulina/metabolismo , Mercaptoetanol/farmacologia , Fenótipo , Ratos
2.
Diabetologia ; 58(10): 2307-16, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26099855

RESUMO

AIMS/HYPOTHESIS: Proinflammatory cytokines contribute to beta cell damage in type 1 diabetes in part through activation of endoplasmic reticulum (ER) stress. In rat beta cells, cytokine-induced ER stress involves NO production and consequent inhibition of the ER Ca(2+) transporting ATPase sarco/endoplasmic reticulum Ca(2+) pump 2 (SERCA2B). However, the mechanisms by which cytokines induce ER stress and apoptosis in mouse and human pancreatic beta cells remain unclear. The purpose of this study is to elucidate the role of ER stress on cytokine-induced beta cell apoptosis in these three species and thus solve ongoing controversies in the field. METHODS: Rat and mouse insulin-producing cells, human pancreatic islets and human EndoC-ßH1 cells were exposed to the cytokines IL-1ß, TNF-α and IFN-γ, with or without NO inhibition. A global comparison of cytokine-modulated gene expression in human, mouse and rat beta cells was also performed. The chemical chaperone tauroursodeoxycholic acid (TUDCA) and suppression of C/EBP homologous protein (CHOP) were used to assess the role of ER stress in cytokine-induced apoptosis of human beta cells. RESULTS: NO plays a key role in cytokine-induced ER stress in rat islets, but not in mouse or human islets. Bioinformatics analysis indicated greater similarity between human and mouse than between human and rat global gene expression after cytokine exposure. The chemical chaperone TUDCA and suppression of CHOP or c-Jun N-terminal kinase (JNK) protected human beta cells against cytokine-induced apoptosis. CONCLUSIONS/INTERPRETATION: These observations clarify previous results that were discrepant owing to the use of islets from different species, and confirm that cytokine-induced ER stress contributes to human beta cell death, at least in part via JNK activation.


Assuntos
Estresse do Retículo Endoplasmático/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Citocinas/farmacologia , Inibidores Enzimáticos/farmacologia , Humanos , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Ratos , Ratos Wistar , Ácido Tauroquenodesoxicólico/farmacologia , Fator de Transcrição CHOP/farmacologia , ômega-N-Metilarginina/farmacologia
3.
Diabetologia ; 57(2): 383-91, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24190582

RESUMO

AIMS/HYPOTHESIS: Genome-wide association studies have revealed an association of the transcription factor ETS variant gene 5 (ETV5) with human obesity. However, its role in glucose homeostasis and energy balance is unknown. METHODS: Etv5 knockout (KO) mice were monitored weekly for body weight (BW) and food intake. Body composition was measured at 8 and 16 weeks of age. Glucose metabolism was studied, and glucose-stimulated insulin secretion was measured in vivo and in vitro. RESULTS: Etv5 KO mice are smaller and leaner, and have a reduced BW and lower fat mass than their wild-type controls on a chow diet. When exposed to a high-fat diet, KO mice are resistant to diet-induced BW gain. Despite a greater insulin sensitivity, KO mice have profoundly impaired glucose tolerance associated with impaired insulin secretion. Morphometric analysis revealed smaller islets and a reduced beta cell size in the pancreatic islets of Etv5 KO mice. Knockdown of ETV5 in an insulin-secreting cell line or beta cells from human donors revealed intact mitochondrial and Ca(2+) channel activity, but reduced insulin exocytosis. CONCLUSION/INTERPRETATION: This work reveals a critical role for ETV5 in specifically regulating insulin secretion both in vitro and in vivo.


Assuntos
Peptídeo C/metabolismo , Proteínas de Ligação a DNA/metabolismo , Exocitose/fisiologia , Glucose/metabolismo , Homeostase/fisiologia , Insulina/metabolismo , Fatores de Transcrição/metabolismo , Animais , Composição Corporal , Peso Corporal , Dieta Hiperlipídica , Ingestão de Alimentos , Estudo de Associação Genômica Ampla , Teste de Tolerância a Glucose , Resistência à Insulina , Secreção de Insulina , Camundongos , Camundongos Knockout
4.
Front Immunol ; 13: 852830, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35529868

RESUMO

Deficiency of adenosine deaminase type 2 (DADA2) is an autosomal recessive disease caused by bi-allelic loss-of-function mutations in ADA2. Treatment with anti-TNF is effective for the autoinflammatory and vasculitic components of the disease but does not correct marrow failure or immunodeficiency; and anti-drug antibodies cause loss of efficacy over time. Allogeneic haematopoietic stem cell transplantation may be curative, but graft versus host disease remains a significant concern. Autologous gene therapy would therefore be an attractive longer-term therapeutic option. We investigated whether lentiviral vector (LV)-mediated ADA2 gene correction could rescue the immunophenotype of DADA2 in primary immune cells derived from patients and in cell line models. Lentiviral transduction led to: i) restoration of ADA2 protein expression and enzymatic activity; (ii) amelioration of M1 macrophage cytokine production, IFN-γ and phosphorylated STAT1 expression in patient-derived macrophages; and (iii) amelioration of macrophage-mediated endothelial activation that drives the vasculitis of DADA2. We also successfully transduced human CD34+ haematopoietic stem progenitor cells (HSPC) derived from a DADA2 patient with pure red cell aplasia and observed restoration of ADA2 expression and enzymatic activity in CD34+HSPC, alongside recovery of stem-cell proliferative and colony forming unit capacity. These preclinical data now expand the evidence for the efficacy of gene transfer strategies in DADA2, and strongly support clinical translation of a lentivirus-mediated gene therapy approach to treat DADA2.


Assuntos
Agamaglobulinemia , Terapia Genética , Imunodeficiência Combinada Severa , Vasculite , Adenosina Desaminase/genética , Agamaglobulinemia/terapia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Imunodeficiência Combinada Severa/terapia , Inibidores do Fator de Necrose Tumoral , Vasculite/terapia
5.
Sci Rep ; 11(1): 13705, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34210996

RESUMO

The D614G mutation in the Spike protein of the SARS-CoV-2 has effectively replaced the early pandemic-causing variant. Using pseudotyped lentivectors, we confirmed that the aspartate replacement by glycine in position 614 is markedly more infectious. Molecular modelling suggests that the G614 mutation facilitates transition towards an open state of the Spike protein. To explain the epidemiological success of D614G, we analysed the evolution of 27,086 high-quality SARS-CoV-2 genome sequences from GISAID. We observed striking coevolution of D614G with the P323L mutation in the viral polymerase. Importantly, the exclusive presence of G614 or L323 did not become epidemiologically relevant. In contrast, the combination of the two mutations gave rise to a viral G/L variant that has all but replaced the initial D/P variant. Our results suggest that the P323L mutation, located in the interface domain of the RNA-dependent RNA polymerase, is a necessary alteration that led to the epidemiological success of the present variant of SARS-CoV-2. However, we did not observe a significant correlation between reported COVID-19 mortality in different countries and the prevalence of the Wuhan versus G/L variant. Nevertheless, when comparing the speed of emergence and the ultimate predominance in individual countries, it is clear that the G/L variant displays major epidemiological supremacy over the original variant.


Assuntos
COVID-19/virologia , RNA-Polimerase RNA-Dependente de Coronavírus/genética , Mutação Puntual , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/genética , COVID-19/epidemiologia , RNA-Polimerase RNA-Dependente de Coronavírus/química , Humanos , Modelos Moleculares , Conformação Proteica , SARS-CoV-2/química , Glicoproteína da Espícula de Coronavírus/química
6.
J Biol Chem ; 284(37): 25004-14, 2009 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-19584051

RESUMO

The SLC25 carrier family mediates solute transport across the inner mitochondrial membrane, a process that is still poorly characterized regarding both the mechanisms and proteins implicated. This study investigated mitochondrial glutamate carrier GC1 in insulin-secreting beta-cells. GC1 was cloned from insulin-secreting cells, and sequence analysis revealed hydropathy profile of a six-transmembrane protein, characteristic of mitochondrial solute carriers. GC1 was found to be expressed at the mRNA and protein levels in INS-1E beta-cells and pancreatic rat islets. Immunohistochemistry showed that GC1 was present in mitochondria, and ultrastructural analysis by electron microscopy revealed inner mitochondrial membrane localization of the transporter. Silencing of GC1 in INS-1E beta-cells, mediated by adenoviral delivery of short hairpin RNA, reduced mitochondrial glutamate transport by 48% (p < 0.001). Insulin secretion at basal 2.5 mM glucose and stimulated either by intermediate 7.5 mM glucose or non-nutrient 30 mM KCl was not modified by GC1 silencing. Conversely, insulin secretion stimulated with optimal 15 mM glucose was reduced by 23% (p < 0.005) in GC1 knocked down cells compared with controls. Adjunct of cell-permeant glutamate (5 mM dimethyl glutamate) fully restored the secretory response at 15 mM glucose (p < 0.005). Kinetics of insulin secretion were investigated in perifused isolated rat islets. GC1 silencing in islets inhibited the secretory response induced by 16.7 mM glucose, both during first (-25%, p < 0.05) and second (-33%, p < 0.05) phases. This study demonstrates that insulin-secreting cells depend on GC1 for maximal glucose response, thereby assigning a physiological function to this newly identified mitochondrial glutamate carrier.


Assuntos
Ácido Glutâmico/metabolismo , Insulina/metabolismo , Proteínas de Membrana Transportadoras/fisiologia , Mitocôndrias/metabolismo , Proteínas Mitocondriais/fisiologia , Animais , Linhagem Celular , Inativação Gênica , Glucose/metabolismo , Humanos , Imuno-Histoquímica , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Cinética , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Proteínas de Transporte da Membrana Mitocondrial , Proteínas Mitocondriais/metabolismo , Modelos Biológicos , RNA Interferente Pequeno/metabolismo , Ratos
7.
Biochem J ; 424(3): 459-66, 2009 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-19764902

RESUMO

Transfer of reducing equivalents between cytosolic compartments and the mitochondrial matrix is mediated by NADH shuttles. Among these, the malate-aspartate shuttle has been proposed to play a major role in beta-cells for the control of glucose-stimulated insulin secretion. AGC1 or Aralar1 (aspartate-glutamate carrier 1) is a key component of the malate-aspartate shuttle. Overexpression of AGC1 increases the capacity of the malate-aspartate shuttle, resulting in enhanced metabolism-secretion coupling, both in INS-1E cells and rat islets. In the present study, knockdown of AGC1 was achieved in the same beta-cell models, using adenovirus-mediated delivery of shRNA (small-hairpin RNA). Compared with control INS-1E cells, down-regulation of AGC1 blunted NADH formation (-57%; P<0.05), increased lactate production (+16%; P<0.001) and inhibited glucose oxidation (-22%; P<0.01). This correlated with a reduced secretory response at 15 mM glucose (-25%; P<0.05), while insulin release was unchanged at intermediate 7.5 mM and basal 2.5 mM glucose. In isolated rat islets, efficient AGC1 knockdown did not alter insulin exocytosis evoked by 16.7 mM glucose. However, 4 mM amino-oxyacetate, commonly used to block transaminases of the malate-aspartate shuttle, inhibited glucose-stimulated insulin secretion to similar extents in INS-1E cells (-66%; P<0.01) and rat islets (-56%; P<0.01). These results show that down-regulation of the key component of the malate-aspartate shuttle AGC1 reduced glucose-induced oxidative metabolism and insulin secretion in INS-1E cells, whereas similar AGC1 knockdown in rat islets did not affect their secretory response.


Assuntos
Ilhotas Pancreáticas/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Proteínas Mitocondriais/metabolismo , Interferência de RNA , Trifosfato de Adenosina/metabolismo , Ácido Amino-Oxiacético/farmacologia , Animais , Cálcio/metabolismo , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Glucose/metabolismo , Glucose/farmacologia , Ácido Glutâmico/metabolismo , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Lactatos/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Proteínas de Membrana Transportadoras/genética , Mitocôndrias/metabolismo , Mitocôndrias/fisiologia , Proteínas de Transporte da Membrana Mitocondrial , Proteínas Mitocondriais/genética , Ratos
8.
Adv Exp Med Biol ; 654: 193-216, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20217499

RESUMO

Pancreatic beta-cells are poised to sense glucose and other nutrient secretagogues to regulate insulin exocytosis, thereby maintaining glucose homeostasis. This process requires translation of metabolic substrates into intracellular messengers recognized by the exocytotic machinery. Central to this metabolism-secretion coupling, mitochondria integrate and generate metabolic signals, thereby connecting glucose recognition to insulin exocytosis. In response to a glucose rise, nucleotides and metabolites are generated by mitochondria and participate, together with cytosolic calcium, to the stimulation of insulin release. This review describes the mitochondrion-dependent pathways of regulated insulin secretion. Mitochondrial defects, such as mutations and reactive oxygen species production, are discussed in the context of beta-cell failure that may participate to the etiology of diabetes.


Assuntos
Diabetes Mellitus/metabolismo , Células Secretoras de Insulina/citologia , Mitocôndrias/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Ácidos Graxos/metabolismo , Glucose/metabolismo , Glutamato Desidrogenase/metabolismo , Ácido Glutâmico/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina , Mitocôndrias/metabolismo , Modelos Biológicos , NAD/metabolismo , Espécies Reativas de Oxigênio
9.
Biochim Biophys Acta ; 1777(7-8): 965-72, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18486589

RESUMO

Glutamate is implicated in numerous metabolic and signalling functions that vary according to specific tissues. Glutamate metabolism is tightly controlled by activities of mitochondrial enzymes and transmembrane carriers, in particular glutamate dehydrogenase and mitochondrial glutamate carriers that have been identified in recent years. It is remarkable that, although glutamate-specific enzymes and transporters share similar properties in most tissues, their regulation varies greatly according to particular organs in order to achieve tissue specific functions. This is illustrated in this review when comparing glutamate handling in liver, brain, and pancreatic beta-cells. We describe the main cellular glutamate pathways and their specific functions in different tissues, ultimately contributing to the control of metabolic homeostasis at the organism level.


Assuntos
Glutamato Desidrogenase/genética , Glutamato Desidrogenase/metabolismo , Ácido Glutâmico/metabolismo , Mitocôndrias/metabolismo , Animais , Ácido Aspártico/metabolismo , Proteínas de Transporte/metabolismo , Diabetes Mellitus/metabolismo , Homeostase , Humanos , Especificidade de Órgãos
10.
JAMA Netw Open ; 2(10): e1914274, 2019 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-31664448

RESUMO

Importance: Neuroinflammatory disorders are a range of severe neurological disorders causing brain and spinal inflammation and are now increasingly recognized in the pediatric population. They are often characterized by marked genotypic and phenotypic heterogeneity, complicating diagnostic work in clinical practice and molecular diagnosis. Objective: To develop and evaluate a next-generation sequencing panel targeting genes causing neuroinflammation or mimicking neuroinflammation. Design, Setting, and Participants: Cohort study in which a total of 257 genes associated with monogenic neuroinflammation and/or cerebral vasculopathy, including monogenic noninflammatory diseases mimicking these entities, were selected. A customized enrichment capture array, the neuroinflammation gene panel (NIP), was created. Targeted high-coverage sequencing was applied to DNA samples taken from eligible patients referred to Great Ormond Street Hospital in London, United Kingdom, between January 1, 2017, and January 30, 2019, because of onset of disease early in life, family history, and/or complex neuroinflammatory phenotypes. Main Outcomes and Measures: The main outcome was the percentage of individuals with definitive molecular diagnoses, variant classification, and clinical phenotyping of patients with pathogenic variants identified using the NIP panel. The NIP panel was initially validated in 16 patients with known genetic diagnoses. Results: The NIP was both sensitive (95%) and specific (100%) for detection of known mutations, including gene deletions, copy number variants, small insertions and deletions, and somatic mosaicism with allele fraction as low as 3%. Prospective testing of 60 patients (30 [50%] male; median [range] age, 9.8 [0.8-20] years) presenting with heterogeneous neuroinflammatory phenotypes revealed at least 1 class 5 (clearly pathogenic) variant in 9 of 60 patients (15%); 18 of 60 patients (30%) had at least 1 class 4 (likely pathogenic) variant. Overall, a definitive molecular diagnosis was established in 12 of 60 patients (20%). Conclusions and Relevance: The NIP was associated with molecular diagnosis in this cohort and complemented routine laboratory and radiological workup of patients with neuroinflammation. Unexpected genotype-phenotype associations in patients with pathogenic variants deviating from the classic phenotype were identified. Obtaining an accurate molecular diagnosis in a timely fashion informed patient management, including successful targeted treatment in some instances and early institution of hematopoietic stem cell transplantation in others.


Assuntos
Encefalopatias/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Inflamação/genética , Adolescente , Criança , Pré-Escolar , Genótipo , Humanos , Lactente , Londres , Técnicas de Diagnóstico Molecular , Mutação , Fenótipo , Sensibilidade e Especificidade , Adulto Jovem
11.
FEBS Lett ; 578(3): 224-8, 2004 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-15589824

RESUMO

Pyruvate promotes hyperpolarization of the inner mitochondrial membrane. However, in isolated mitochondria, pyruvate could participate in a futile cycle leading to mitochondrial depolarization. Here, we investigated this paradox in intact human cells by measuring parameters reflecting mitochondrial activation in response to 1 mM pyruvate and 5 mM glucose. NAD(P)H levels were elevated similarly by both substrates. Conversely, pyruvate induced a first transient phase of mitochondrial depolarization before the establishment of the expected sustained hyperpolarization. This correlated with kinetics of cytosolic ATP levels exhibiting a first phase decrease followed by an increase. Therefore, pyruvate transiently depolarizes mitochondria and reduces ATP in intact cells.


Assuntos
Mitocôndrias/metabolismo , Ácido Pirúvico/metabolismo , Trifosfato de Adenosina/metabolismo , Carbonil Cianeto p-Trifluormetoxifenil Hidrazona/farmacologia , Linhagem Celular Tumoral , Citosol/metabolismo , Glucose/metabolismo , Glucose/farmacologia , Humanos , Ionóforos/farmacologia , Cinética , Luciferases/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Proteínas Mitocondriais/metabolismo , Modelos Biológicos , NADP/metabolismo , Osteossarcoma/metabolismo , Ácido Pirúvico/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rotenona/farmacologia , Especificidade por Substrato , Desacopladores/farmacologia
12.
Diabetes ; 61(7): 1708-18, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22498697

RESUMO

Focal adhesion kinase (FAK) acts as an adaptor at the focal contacts serving as a junction between the extracellular matrix and actin cytoskeleton. Actin dynamics is known as a determinant step in insulin secretion. Additionally, FAK has been shown to regulate insulin signaling. To investigate the essential physiological role of FAK in pancreatic ß-cells in vivo, we generated a transgenic mouse model using rat insulin promoter (RIP)-driven Cre-loxP recombination system to specifically delete FAK in pancreatic ß-cells. These RIPcre(+)fak(fl/fl) mice exhibited glucose intolerance without changes in insulin sensitivity. Reduced ß-cell viability and proliferation resulting in decreased ß-cell mass was observed in these mice, which was associated with attenuated insulin/Akt (also known as protein kinase B) and extracellular signal-related kinase 1/2 signaling and increased caspase 3 activation. FAK-deficient ß-cells exhibited impaired insulin secretion with normal glucose sensing and preserved Ca(2+) influx in response to glucose, but a reduced number of docked insulin granules and insulin exocytosis were found, which was associated with a decrease in focal proteins, paxillin and talin, and an impairment in actin depolymerization. This study is the first to show in vivo that FAK is critical for pancreatic ß-cell viability and function through regulation in insulin signaling, actin dynamics, and granule trafficking.


Assuntos
Citoesqueleto de Actina/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Resistência à Insulina , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animais , Cálcio/metabolismo , Caspase 3/biossíntese , Sobrevivência Celular , Exocitose , Feminino , Quinase 1 de Adesão Focal/genética , Intolerância à Glucose/genética , Insulina/genética , Secreção de Insulina , Células Secretoras de Insulina/citologia , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Paxilina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Talina/metabolismo , Vesículas Transportadoras/metabolismo
13.
Diabetes ; 60(3): 838-47, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21266332

RESUMO

OBJECTIVE: The reversible attachment of small ubiquitin-like modifier (SUMO) proteins controls target localization and function. We examined an acute role for the SUMOylation pathway in downstream events mediating insulin secretion. RESEARCH DESIGN AND METHODS: We studied islets and ß-cells from mice and human donors, as well as INS-1 832/13 cells. Insulin secretion, intracellular Ca(2+), and ß-cell exocytosis were monitored after manipulation of the SUMOylation machinery. Granule localization was imaged by total internal reflection fluorescence and electron microscopy; immunoprecipitation and Western blotting were used to examine the soluble NSF attachment receptor (SNARE) complex formation and SUMO1 interaction with synaptotagmin VII. RESULTS: SUMO1 impairs glucose-stimulated insulin secretion by blunting the ß-cell exocytotic response to Ca(2+). The effect of SUMO1 to impair insulin secretion and ß-cell exocytosis is rapid and does not require altered gene expression or insulin content, is downstream of granule docking at the plasma membrane, and is dependent on SUMO-conjugation because the deSUMOylating enzyme, sentrin/SUMO-specific protease (SENP)-1, rescues exocytosis. SUMO1 coimmunoprecipitates with the Ca(2+) sensor synaptotagmin VII, and this is transiently lost upon glucose stimulation. SENP1 overexpression also disrupts the association of SUMO1 with synaptotagmin VII and mimics the effect of glucose to enhance exocytosis. Conversely, SENP1 knockdown impairs exocytosis at stimulatory glucose levels and blunts glucose-dependent insulin secretion from mouse and human islets. CONCLUSIONS: SUMOylation acutely regulates insulin secretion by the direct and reversible inhibition of ß-cell exocytosis in response to intracellular Ca(2+) elevation. The SUMO protease, SENP1, is required for glucose-dependent insulin secretion.


Assuntos
Exocitose/fisiologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteína SUMO-1/metabolismo , Vesículas Secretórias/metabolismo , Sumoilação/fisiologia , Análise de Variância , Animais , Western Blotting , Cálcio/metabolismo , Células Cultivadas , Cisteína Endopeptidases , Endopeptidases/metabolismo , Exocitose/efeitos dos fármacos , Glucose/metabolismo , Glucose/farmacologia , Células HEK293 , Humanos , Imunoprecipitação , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Microscopia Eletrônica , Vesículas Secretórias/efeitos dos fármacos , Sinaptotagminas/metabolismo
14.
Nutr Metab (Lond) ; 8(1): 2, 2011 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-21244699

RESUMO

Current notions about mechanisms by which catch-up growth predisposes to later type 2 diabetes center upon those that link hyperinsulinemia with an accelerated rate of fat deposition (catch-up fat). Using a rat model of semistarvation-refeeding in which catch-up fat is driven solely by elevated metabolic efficiency associated with hyperinsulinemia, we previously reported that insulin-stimulated glucose utilization is diminished in skeletal muscle but increased in white adipose tissue. Here, we investigated the possibility that hyperinsulinemia during catch-up fat can be contributed by changes in the secretory response of pancreatic beta-cells to glucose. Using the rat model of semistarvation-refeeding showing catch-up fat and hyperinsulinemia, we compared isocalorically refed and control groups for potential differences in pancreatic morphology and in glucose-stimulated insulin secretion during in situ pancreas perfusions as well as ex vivo isolated islet perifusions. Between refed and control animals, no differences were found in islet morphology, insulin content, and the secretory responses of perifused isolated islets upon glucose stimulation. By contrast, the rates of insulin secretion from in situ perfused pancreas showed that raising glucose from 2.8 to 16.7 mmol/l produced a much more pronounced increase in insulin release in refed than in control groups (p < 0.01). These results indicate a role for islet secretory hyperresponsiveness to glucose in the thrifty mechanisms that drive catch-up fat through glucose redistribution between skeletal muscle and adipose tissue. Such beta-cell hyperresponsiveness to glucose may be a key event in the link between catch-up growth, hyperinsulinemia and risks for later type 2 diabetes.

15.
J Endocrinol ; 205(1): 79-86, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20061513

RESUMO

Sclerocarya birrea is a plant widely used as traditional medication for the treatment of diabetes in sub-Saharan regions. However, the mechanism of action is unknown and only hypoglycaemic effects of S. birrea extract (SBE) in diabetic rats have been reported to date. Here, we tested aqueous extracts of S. birrea on insulin-secreting INS-1E cells and isolated rat islets. Following 24 h of treatment at 5 microg/ml, the extract markedly potentiated glucose-stimulated insulin secretion. Neither basal insulin release nor non-nutrient stimulation was affected. The potentiation of the secretory response at stimulatory glucose appeared after 12 h of treatment. No acute effects were observed and, at the effective concentration, SBE was safe regarding cell integrity and differentiation. The mechanism of action of the SBE was related to glucose metabolism as both ATP generation and glucose oxidation were enhanced following the 24-h treatment. In streptozotocin-induced diabetic rats, SBE administration corrected glycaemia and restored plasma insulin levels after 2 weeks of treatment. These data show direct action of S. birrea on insulin-secreting cells and favour further delineation for use of the plant in the management of diabetes.


Assuntos
Anacardiaceae , Diabetes Mellitus Experimental/tratamento farmacológico , Glucose/farmacologia , Hiperglicemia/tratamento farmacológico , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Extratos Vegetais/uso terapêutico , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular Tumoral , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/metabolismo , Modelos Animais de Doenças , Glucose/metabolismo , Hiperglicemia/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Insulinoma/metabolismo , Insulinoma/patologia , Masculino , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Casca de Planta , Extratos Vegetais/farmacologia , Caules de Planta , Ratos , Ratos Wistar , Estreptozocina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA