Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BMJ Open ; 14(5): e079574, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38719313

RESUMEN

INTRODUCTION: SARS-CoV-2 mainly infects respiratory endothelial cells, which is facilitated through its spike protein binding to heparan sulphate. Calcium dobesilate (CaD) is a well-established, widely available vasoactive and angioprotective drug interacting with heparan sulphate, with the potential to interfere with the uptake of SARS-CoV-2 by epithelial cells. The CADOVID trial aims to evaluate the efficacy and safety of CaD in reducing the SARS-CoV-2 viral load in non-hospitalised adult patients diagnosed with COVID-19, confirmed by a positive SARS-CoV-2 PCR, including its efficacy to reduce the impact of persistent COVID-19 symptoms. METHODS AND ANALYSIS: This is a randomised, placebo-controlled, double-blind, monocentric phase II trial. Enrolment began in July 2022. A total of 74 adult patients will be randomly allocated to the CaD arm or the placebo group with a 1:1 ratio, respectively. Participants in the intervention arm will receive two capsules of CaD 500 mg two times per day and the placebo arm will receive two matching capsules of mannitol 312.5 mg two times per day, with a treatment period of 7 days for both arms, followed by a 77-day observational period without treatment administration. Participants will be asked to complete secured online questionnaires using their personal smartphone or other electronic device. These include a COVID-19 questionnaire (assessing symptoms, temperature measurement, reporting of concomitant medication and adverse events), a COVID-19 persistent symptoms' questionnaire and the Short Form 12-Item (SF-12) survey. SARS-CoV-2 PCR testing will be performed on nasopharyngeal swabs collected on days 1, 4, 8 and 21. The primary endpoint is the reduction from baseline of SARS-CoV-2 viral load determined by RT-PCR at day 4. ETHICS AND DISSEMINATION: This trial has received approval by the Geneva Regional Research Ethics Committee (2022-00613) and Swissmedic (701339). Dissemination of results will be through presentations at scientific conferences and publication in scientific journals. TRIAL REGISTRATION NUMBER: NCT05305508; Clinicaltrials.gov; Swiss National Clinical Portal Registry (SNCTP 000004938).


Asunto(s)
COVID-19 , Dobesilato de Calcio , SARS-CoV-2 , Carga Viral , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Dobesilato de Calcio/uso terapéutico , Ensayos Clínicos Fase II como Asunto , COVID-19/virología , Tratamiento Farmacológico de COVID-19 , Método Doble Ciego , Pacientes Ambulatorios , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento , Carga Viral/efectos de los fármacos
2.
Mol Cell Endocrinol ; 541: 111503, 2022 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-34763008

RESUMEN

Pancreatic beta cell dysfunction is a hallmark of type 2 diabetes. Growth differentiation factor 15 (GDF15), which is an energy homeostasis regulator, has been shown to improve several metabolic parameters in the context of diabetes. However, its effects on pancreatic beta-cell remain to be identified. We, therefore, performed experiments using cell models and histological sectioning of wild-type and knock-out GDF15 mice to determine the effect of GDF15 on insulin secretion and cell viability. A bioinformatics analysis was performed to identify GDF15-correlated genes. GDF15 prevents glucotoxicity-mediated altered glucose-stimulated insulin secretion (GSIS) and connexin-36 downregulation. Inhibition of endogenous GDF15 reduced GSIS in cultured mouse beta-cells under standard conditions while it had no impact on GSIS in cells exposed to glucolipotoxicity, which is a diabetogenic condition. Furthermore, this inhibition exacerbated glucolipotoxicity-reduced cell survival. This suggests that endogenous GDF15 in beta-cell is required for cell survival but not GSIS in the context of glucolipotoxicity.


Asunto(s)
Conexinas/genética , Glucosa/efectos adversos , Factor 15 de Diferenciación de Crecimiento/fisiología , Células Secretoras de Insulina/fisiología , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Conexinas/metabolismo , Citoprotección/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Glucosa/metabolismo , Factor 15 de Diferenciación de Crecimiento/genética , Insulina/metabolismo , Secreción de Insulina/efectos de los fármacos , Secreción de Insulina/genética , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína delta-6 de Union Comunicante
3.
Physiol Rep ; 9(6): e14786, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33769715

RESUMEN

Beta cell failure is one of the most important features of type 2 diabetes mellitus (T2DM). High-density lipoprotein (HDL) has been proposed to improve ß-cell function. However, the mechanisms involved in this process are still poorly understood. The aim of this study was to investigate the contribution of sphingosine-1-phosphate (S1P) in the impact of HDL treatment on insulin secretion by pancreatic ß-cells and to determine its mechanisms. Primary cultures of ß-cells isolated from rat were treated with or without HDL in the presence or absence of S1P pathway inhibitors and insulin secretion response was analyzed. The S1P content of HDL (HDL-S1P) isolated from T2DM patients was analyzed and correlated to the HDL-induced insulin secretion. The expression of genes involved in the biosynthesis of the insulin was also evaluated. HDL as well as S1P treatment enhanced glucose-stimulated insulin secretion (GSIS). In HDL isolated from T2DM patients, while HDL-S1P was strongly correlated to its pro-secretory capacity (r = 0.633, p = 0.005), HDL-cholesterol and apolipoprotein AI levels were not. HDL-induced GSIS was blocked by the S1P1/3 antagonist but not by the S1P2 antagonist, and was also accompanied by increased intracellular S1P in ß-cells. We also observed that HDL improved GSIS without significant changes in expression levels of insulin biosynthesis genes. Our present study highlights the importance HDL-S1P in GSIS in T2DM patients and demonstrates that HDL induces insulin secretion by a process involving both intra- and extra-cellular sources of S1P independently of an effect on insulin biosynthesis genes.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Lipoproteínas HDL/administración & dosificación , Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Anciano , Animales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Cultivo Primario de Células , Ratas , Esfingosina/metabolismo
4.
Endocrinology ; 161(2)2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31905402

RESUMEN

Characterization of enteroendocrine L cells in diabetes is critical for better understanding of the role of glucagon-like peptide-1 (GLP-1) in physiology and diabetes. We studied L-cell transcriptome changes including microRNA (miRNA) dysregulation in obesity and diabetes. We evaluated the regulation of miRNAs through microarray analyses on sorted enteroendocrine L cells from control and obese glucose-intolerant (I-HFD) and hyperglycemic (H-HFD) mice after 16 weeks of respectively low-fat diet (LFD) or high-fat diet (HFD) feeding. The identified altered miRNAs were studied in vitro using the mouse GLUTag cell line to investigate their regulation and potential biological functions. We identified that let-7e-5p, miR-126a-3p, and miR-125a-5p were differentially regulated in L cells of obese HFD mice compared with control LFD mice. While downregulation of let-7e-5p expression was observed in both I-HFD and H-HFD mice, levels of miR-126a-3p increased and of miR-125a-5p decreased significantly only in I-HFD mice compared with controls. Using miRNA inhibitors and mimics we observed that modulation of let-7e-5p expression affected specifically GLP-1 cellular content and basal release, whereas Gcg gene expression and acute GLP-1 secretion and cell proliferation were not affected. In addition, palmitate treatment resulted in a decrease of let-7e-5p expression along with an increase in GLP-1 content and release, suggesting that palmitate acts on GLP-1 through let-7e-5p. By contrast, modulation of miR-125a-5p and miR-126a-3p in the same conditions did not affect content or secretion of GLP-1. We conclude that decrease of let-7e-5p expression in response to palmitate may constitute a compensatory mechanism contributing to maintaining constant glycemia in obese mice.


Asunto(s)
Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , MicroARNs/metabolismo , Obesidad/metabolismo , Animales , Línea Celular , Dieta Alta en Grasa , Masculino , Ratones Transgénicos , Palmitatos
5.
PLoS One ; 14(3): e0213299, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30849121

RESUMEN

Characterization of endocrine-cell functions and associated molecular signatures in diabetes is crucial to better understand why and by which mechanisms alpha and beta cells cause and perpetuate metabolic abnormalities. The now recognized role of glucagon in diabetes control is a major incentive to have a better understanding of dysfunctional alpha cells. To characterize molecular alterations of alpha cells in diabetes, we analyzed alpha-cell transcriptome from control and diabetic mice using diet-induced obesity model. To this aim, we quantified the expression levels of total mRNAs from sorted alpha and beta cells of low-fat and high-fat diet-treated mice through RNAseq experiments, using a transgenic mouse strain allowing collections of pancreatic alpha- and beta-cells after 16 weeks of diet. We now report that pancreatic alpha cells from obese hyperglycemic mice displayed minor variations of their transcriptome compared to controls. Depending on analyses, we identified 11 to 39 differentially expressed genes including non-alpha cell markers mainly due to minor cell contamination during purification process. From these analyses, we identified three new target genes altered in diabetic alpha cells and potently involved in cellular stress and exocytosis (Upk3a, Adcy1 and Dpp6). By contrast, analysis of the beta-cell transcriptome from control and diabetic mice revealed major alterations of specific genes coding for proteins involved in proliferation and secretion. We conclude that alpha cell transcriptome is less reactive to HFD diet compared to beta cells and display adaptations to cellular stress and exocytosis.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Regulación de la Expresión Génica , Células Secretoras de Glucagón/metabolismo , Islotes Pancreáticos/metabolismo , Obesidad/metabolismo , Transcriptoma , Animales , Células Cultivadas , Células Secretoras de Glucagón/citología , Islotes Pancreáticos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Ratones Transgénicos , Obesidad/etiología , Obesidad/patología
6.
Mol Metab ; 22: 84-95, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30711402

RESUMEN

OBJECTIVE: Diabetes is a complex disease implicating several organs and cell types. Within the islets, dysregulation occurs in both alpha- and beta-cells, leading to defects of insulin secretion and increased glucagon secretion. Dysregulation of alpha-cells is associated with transcriptome changes. We hypothesized that microRNAs (miRNAs) which are negative regulators of mRNA stability and translation could be involved in alpha-cell alterations or adaptations during type 2 diabetes. METHODS: miRNA microarray analyses were performed on pure alpha- and beta-cells from high-fat diet fed obese hyperglycemic mice and low-fat diet fed controls. Then, the most regulated miRNA was overexpressed or inhibited in primary culture of mouse and human alpha-cells to determine its molecular and functional impact. RESULTS: 16 miRNAs were significantly regulated in alpha-cells of obese hyperglycemic mice and 28 in beta-cells. miR-132-3p had the strongest regulation level in alpha-cells, where it was downregulated, while we observed an opposite upregulation in beta-cells. In vitro experiments showed that miR-132-3p, which is inversely regulated by somatostatin and cAMP, is a positive modulator of alpha-cell proliferation and implicated in their resistance to apoptosis. These effects are associated with the regulation of a series of genes, including proliferation and stress markers Mki67 and Bbc3 in mouse and human alpha-cells, potentially involved in miR-132-3p functions. CONCLUSIONS: Downregulation of miR-132-3p in alpha-cells of obese diabetic mice may constitute a compensatory mechanism contributing to keep glucagon-producing cell number constant in diabetes.


Asunto(s)
Regulación hacia Abajo , Células Secretoras de Glucagón/metabolismo , MicroARNs/metabolismo , Obesidad/metabolismo , Animales , Células Cultivadas , Humanos , Ratones , Ratones Obesos
7.
JCI Insight ; 3(7)2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29618657

RESUMEN

Clinical and experimental data indicate a beneficial effect of estrogens on energy and glucose homeostasis associated with improved insulin sensitivity and positive effects on insulin secretion. The aim of the study was to investigate the impact of estrogens on proglucagon-producing cells, pancreatic α cells, and enteroendocrine L cells. The consequences of sexual hormone deprivation were evaluated in ovariectomized mice (ovx). Ovx mice exhibited impaired glucose tolerance during oral glucose tolerance tests (OGTT), which was associated with decreased GLP-1 intestinal and pancreatic secretion and content, an effect that was reversed by estradiol (E2) treatment. Indeed, E2 increased oral glucose-induced GLP-1 secretion in vivo and GLP-1 secretion from primary culture of mouse and human α cells through the activation of all 3 estrogen receptors (ERs), whereas E2-induced GLP-1 secretion from mouse and human intestinal explants occurred only by ERß activation. Underlying the implication of ERß, its selective agonist WAY20070 was able to restore glucose tolerance in ovx mice at least partly through plasma GLP-1 increase. We conclude that E2 directly controls both α- and L cells to increase GLP-1 secretion, in addition to its effects on insulin and glucagon secretion, highlighting the potential beneficial role of the estrogenic pathway and, more particularly, of ERß agonists to prevent type 2 diabetes.


Asunto(s)
Células Enteroendocrinas/metabolismo , Estradiol/metabolismo , Receptor beta de Estrógeno/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Células Secretoras de Glucagón/metabolismo , Animales , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/prevención & control , Células Enteroendocrinas/efectos de los fármacos , Receptor beta de Estrógeno/agonistas , Femenino , Péptido 1 Similar al Glucagón/análisis , Células Secretoras de Glucagón/efectos de los fármacos , Glucosa/administración & dosificación , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovariectomía , Oxazoles/farmacología , Fenoles/farmacología , Cultivo Primario de Células
8.
Artículo en Inglés | MEDLINE | ID: mdl-28439257

RESUMEN

Circadian clocks have been developed in evolution as an anticipatory mechanism allowing for adaptation to the constantly changing light environment due to rotation of the Earth. This mechanism is functional in all light-sensitive organisms. There is a considerable body of evidence on the tight connection between the circadian clock and most aspects of physiology and metabolism. Clocks, operative in the pancreatic islets, have caught particular attention in the last years due to recent reports on their critical roles in regulation of insulin secretion and etiology of type 2 diabetes. While ß-cell clocks have been extensively studied during the last years, α-cell clocks and their role in islet function and orchestration of glucose metabolism stayed unexplored, largely due to the difficulty to isolate α-cells, which represents a considerable technical challenge. Here, we provide a detailed description of an experimental approach for the isolation of separate mouse α- and ß-cell population, culture of isolated primary α- and ß-cells, and their subsequent long-term high-resolution circadian bioluminescence recording. For this purpose, a triple reporter ProGlucagon-Venus/RIP-Cherry/Per2:Luciferase mouse line was established, carrying specific fluorescent reporters for α- and ß-cells, and luciferase reporter for monitoring the molecular clockwork. Flow cytometry fluorescence-activated cell sorting allowed separating pure α- and ß-cell populations from isolated islets. Experimental conditions, developed by us for the culture of functional primary mouse α- and ß-cells for at least 10 days, will be highlighted. Importantly, temporal analysis of freshly isolated α- and ß-cells around-the-clock revealed preserved rhythmicity of core clock genes expression. Finally, we describe the setting to assess circadian rhythm in cultured α- and ß-cells synchronized in vitro. The here-described methodology allows to analyze the functional properties of primary α- and ß-cells under physiological or pathophysiological conditions and to assess the islet cellular clock properties.

9.
Endocrinology ; 157(10): 3832-3843, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27547850

RESUMEN

Glucose homeostasis depends on the coordinated secretion of glucagon, insulin, and Glucagon-like peptide (GLP)-1 by pancreas and intestine. Obesity, which is associated with an increased risk of developing insulin resistance and type 2 diabetes, affects the function of these organs. Here, we investigate the functional and molecular adaptations of proglucagon-producing cells in obese mice to better define their involvement in type 2 diabetes development. We used GLU-Venus transgenic male mice specifically expressing Venus fluorochrome in proglucagon-producing cells. Mice were subjected to 16 weeks of low-fat diet or high-fat diet (HFD) and then subdivided by measuring glycated hemoglobin (HbA1c) in 3 groups: low-fat diet mice and I-HFD (glucose-intolerant) mice with similar HbA1c and H-HFD (hyperglycemic) mice, which exhibited higher HbA1c. At 16 weeks, both HFD groups exhibited similar weight gain, hyperinsulinemia, and insulin resistance. However, I-HFD mice exhibited better glucose tolerance compared with H-HFD mice. I-HFD mice displayed functional and molecular adaptations of enteroendocrine L-cells resulting in increased intestinal GLP-1 biosynthesis and release as well as maintained pancreatic α- and ß-cell functions. By contrast, H-HFD mice exhibited dysfunctional L, α- and ß-cells with increased ß- and L-cell numbers. Administration of the GLP-1R antagonist Exendin9-39 in I-HFD mice led to hyperglycemia and alterations of glucagon secretion without changes in insulin secretion. Our results highlight the cross-talk between islet and intestine endocrine cells and indicate that a compensatory adaptation of L-cell function in obesity plays an important role in preserving glucose homeostasis through the control of pancreatic α-cell functions.


Asunto(s)
Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Células Secretoras de Glucagón/metabolismo , Hiperglucemia/metabolismo , Obesidad/metabolismo , Animales , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa , Glucagón/metabolismo , Hiperglucemia/etiología , Células Secretoras de Insulina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/complicaciones , Fragmentos de Péptidos , Fenotipo
10.
Endocrinology ; 157(2): 536-47, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26696123

RESUMEN

Glucagon and α-cell dysfunction are critical in the development of hyperglycemia during diabetes both in humans and rodents. We hypothesized that α-cell dysfunction leading to dysregulated glucagon secretion in diabetes is due to both a lack of insulin and intrinsic defects. To characterize α-cell dysfunction in diabetes, we used glucagon-Venus transgenic male mice and induced insulinopenic hyperglycemia by streptozotocin administration leading to alterations of glucagon secretion. We investigated the in vivo impact of insulinopenic hyperglycemia on glucagon-producing cells using FACS-sorted α-cells from control and diabetic mice. We demonstrate that increased glucagonemia in diabetic mice is mainly due to increases of glucagon release and biosynthesis per cell compared with controls without changes in α-cell mass. We identified genes coding for proteins involved in glucagon biosynthesis and secretion, α-cell differentiation, and potential stress markers such as the glucagon, Arx, MafB, cMaf, Brain4, Foxa1, Foxa3, HNF4α, TCF7L2, Glut1, Sglt2, Cav2.1, Cav2.2, Nav1.7, Kir6.2/Sur1, Pten, IR, NeuroD1, GPR40, and Sumo1 genes, which were abnormally regulated in diabetic mice. Importantly, insulin treatment partially corrected α-cell function and expression of genes coding for proglucagon, or involved in glucagon secretion, glucose transport and insulin signaling but not those coding for cMAF, FOXA1, and α-cell differentiation markers as well as GPR40, NEUROD1, CAV2.1, and SUMO1. Our results indicate that insulinopenic diabetes induce marked α-cell dysfunction and molecular alteration, which are only partially corrected by in vivo insulin treatment.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Células Secretoras de Glucagón/efectos de los fármacos , Células Secretoras de Glucagón/metabolismo , Glucagón/metabolismo , Insulina/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Diabetes Mellitus Experimental/patología , Glucagón/biosíntesis , Glucagón/genética , Insulina/uso terapéutico , Masculino , Ratones , Ratones Transgénicos , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/genética
11.
Endocrinology ; 155(10): 3781-92, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25057789

RESUMEN

The Forkhead box A transcription factors are major regulators of glucose homeostasis. They show both distinct and redundant roles during pancreas development and in adult mouse ß-cells. In vivo ablation studies have revealed critical implications of Foxa1 on glucagon biosynthesis and requirement of Foxa2 in α-cell terminal differentiation. In order to examine the respective role of these factors in mature α-cells, we used small interfering RNA (siRNA) directed against Foxa1 and Foxa2 in rat primary pancreatic α-cells and rodent α-cell lines leading to marked decreases in Foxa1 and Foxa2 mRNA levels and proteins. Both Foxa1 and Foxa2 control glucagon gene expression specifically through the G2 element. Although we found that Foxa2 controls the expression of the glucagon, MafB, Pou3f4, Pcsk2, Nkx2.2, Kir6.2, and Sur1 genes, Foxa1 only regulates glucagon gene expression. Interestingly, the Isl1 and Gipr genes were not controlled by either Foxa1 or Foxa2 alone but by their combination. Foxa1 and Foxa2 directly activate and bind the promoter region the Nkx2.2, Kir6.2 and Sur1, Gipr, Isl1, and Pou3f4 genes. We also demonstrated that glucagon secretion is affected by the combined effects of Foxa1 and Foxa2 but not by either one alone. Our results indicate that Foxa1 and Foxa2 control glucagon biosynthesis and secretion as well as α-cell differentiation with both common and unique target genes.


Asunto(s)
Diferenciación Celular/genética , Células Secretoras de Glucagón/fisiología , Glucagón/biosíntesis , Glucagón/metabolismo , Factor Nuclear 3-alfa del Hepatocito/fisiología , Factor Nuclear 3-beta del Hepatocito/fisiología , Animales , Sitios de Unión/genética , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Cricetinae , Factor Nuclear 3-alfa del Hepatocito/antagonistas & inhibidores , Factor Nuclear 3-beta del Hepatocito/antagonistas & inhibidores , Proteína Homeobox Nkx-2.2 , Masculino , Regiones Promotoras Genéticas , ARN Interferente Pequeño/farmacología , Ratas
12.
J Diabetes ; 5(2): 102-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23302052

RESUMEN

Type 2 diabetes (T2D) is one of the most common diseases, affecting 5-10% of the population in most countries; the progression of its prevalence has been constant over the past 50 years in all countries worldwide, creating a major public health problem in terms of disease management and financial burden. Although the pathophysiology of T2D has been attributed for decades to insulin resistance and decreased insulin secretion, particularly in response to glucose, the contributing role of glucagon in hyperglycemia has been highlighted since the early 1970s by demonstrating its glycogenolytic, gluconeogenic and ketogenic properties. More recently, the importance of glucagon in diabetes has been highlighted in a model of streptozotocin-induced diabetic mice becoming euglycemic in the absence of glucagon receptors and without insulin treatment. Understanding the dysregulation of α-cells in diabetes will be critical to better define the pathophysiology of diabetes and develop new antidiabetic treatment.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Animales , Células Secretoras de Glucagón/metabolismo , Glucosa/metabolismo , Humanos , Hipoglucemiantes/farmacología , Islotes Pancreáticos/metabolismo , Ratones
13.
Endocrinology ; 153(9): 4204-15, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22778220

RESUMEN

The Pax6 transcription factor is crucial for pancreatic α-cells. Indeed, Pax6-deficient mouse models are characterized by markedly altered α-cell differentiation. Our objective was to investigate the role of Pax6 in glucagon secretion process. We used a Pax6-deficient model in rat primary enriched-α cells with specific small interfering RNA leading to a 70% knockdown of Pax6 expression. We first showed that Pax6 knockdown decreases glucagon biosynthesis as well as glucagon release. Through physiological assays, we demonstrated that the decrease of Pax6 affects specifically acute glucagon secretion in primary α-cell in response to glucose, palmitate, and glucose-dependent insulinotropic peptide (GIP) but not the response to arginine and epinephrine. We identified in Pax6 knockdown model that genes involved in glucagon secretion such as the glucokinase (GCK), G protein-coupled receptor (GPR40), and GIP receptor (GIPR) as well as the corresponding proteins were significantly decreased whereas the insulin receptor (IR) Kir6.2/Sur1, and glucose transporter 1 genes were not affected. We demonstrated that Pax6 directly binds and activates specific elements on the promoter region of the GPR40, GCK, and GIPR genes. Finally, through site-directed mutagenesis experiments, we showed that disruption of Pax6 binding on the GCK, GPR40, and GIPR gene promoters led to specific decreases of their activities in the αTC1.9 glucagon-producing cell line. Hence our results indicate that Pax6 acts on the regulation of glucagon secretion at least through the transcriptional control of GCK, GPR40, and GIPR. We propose that Pax6 is not only critical for glucagon biosynthesis but also for glucagon secretion particularly in response to nutrients.


Asunto(s)
Proteínas del Ojo/metabolismo , Glucagón/metabolismo , Proteínas de Homeodominio/metabolismo , Factores de Transcripción Paired Box/metabolismo , Proteínas Represoras/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Células Cultivadas , Proteínas del Ojo/genética , Glucoquinasa/genética , Glucoquinasa/metabolismo , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Proteínas de Homeodominio/genética , Inmunoprecipitación , Mutagénesis Sitio-Dirigida , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Ratas , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Receptores de Droga/genética , Receptores de Droga/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de la Hormona Gastrointestinal/genética , Receptores de la Hormona Gastrointestinal/metabolismo , Proteínas Represoras/genética , Receptores de Sulfonilureas
14.
Mol Endocrinol ; 26(4): 696-709, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22403172

RESUMEN

The Pax6 transcription factor is crucial for endocrine cell differentiation and function. Indeed, mutations of Pax6 are associated with a diabetic phenotype and a drastic decrease of insulin-positive cell number. Our aim was to better define the ß-cell Pax6 transcriptional network and thus provide further information concerning the role of Pax6 in ß-cell function. We developed a Pax6-deficient model in rat primary ß-cells with specific small interfering RNA leading to a 75% knockdown of Pax6 expression. Through candidate gene approach, we confirmed that Pax6 controls the mRNA levels of the insulin 1 and 2, Pdx1, MafA, GLUT2, and PC1/3 genes in ß-cells. Importantly, we identified new Pax6 target genes coding for GK, Nkx6.1, cMaf, PC2, GLP-1R and GIPR which are all involved in ß-cell function. Furthermore, we demonstrated that Pax6 directly binds and activates specific elements on the promoter region of these genes. We also demonstrated that Pax6 knockdown led to decreases in insulin cell content, in insulin processing, and a specific defect of glucose-induced insulin secretion as well as a significant reduction of GLP-1 action in primary ß-cells. Our results strongly suggest that Pax6 is crucial for ß-cells through transcriptional control of key genes coding for proteins that are involved in insulin biosynthesis and secretion as well as glucose and incretin actions on ß-cells. We provide further evidence that Pax6 represents a key element of mature ß-cell function.


Asunto(s)
Proteínas del Ojo/fisiología , Glucosa/fisiología , Proteínas de Homeodominio/fisiología , Células Secretoras de Insulina/fisiología , Insulina/biosíntesis , Factores de Transcripción Paired Box/fisiología , Precursores de Proteínas/biosíntesis , Proteínas Represoras/fisiología , Animales , Secuencia de Bases , Sitios de Unión , Diferenciación Celular , Células Cultivadas , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Péptido 1 Similar al Glucagón/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Interferencia de ARN , Ratas , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Transcripción Genética
15.
J Biol Chem ; 285(43): 33381-33393, 2010 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-20592023

RESUMEN

The paired box homeodomain Pax6 is crucial for endocrine cell development and function and plays an essential role in glucose homeostasis. Indeed, mutations of Pax6 are associated with diabetic phenotype. Importantly, homozygous mutant mice for Pax6 are characterized by markedly decreased ß and δ cells and absent α cells. To better understand the critical role that Pax6 exerts in glucagon-producing cells, we developed a model of primary rat α cells. To study the transcriptional network of Pax6 in adult and differentiated α cells, we generated Pax6-deficient primary rat α cells and glucagon-producing cells, using either specific siRNA or cells expressing constitutively a dominant-negative form of Pax6. In primary rat α cells, we confirm that Pax6 controls the transcription of the Proglucagon and processing enzyme PC2 genes and identify three new target genes coding for MafB, cMaf, and NeuroD1/Beta2, which are all critical for Glucagon gene transcription and α cell differentiation. Furthermore, we demonstrate that Pax6 directly binds and activates the promoter region of the three genes through specific binding sites and that constitutive expression of a dominant-negative form of Pax6 in glucagon-producing cells (InR1G9) inhibits the activities of the promoters. Finally our results suggest that the critical role of Pax6 action on α cell differentiation is independent of those of Arx and Foxa2, two transcription factors that are necessary for α cell development. We conclude that Pax6 is critical for α cell function and differentiation through the transcriptional control of key genes involved in glucagon gene transcription, proglucagon processing, and α cell differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas del Ojo/metabolismo , Regulación de la Expresión Génica/fisiología , Células Secretoras de Glucagón/metabolismo , Proteínas de Homeodominio/metabolismo , Modelos Biológicos , Factores de Transcripción Paired Box/metabolismo , Proteínas Represoras/metabolismo , Elementos de Respuesta/fisiología , Animales , Línea Celular , Proteínas del Ojo/genética , Células Secretoras de Glucagón/citología , Proteínas de Homeodominio/genética , Humanos , Ratones , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , Proglucagón/biosíntesis , Proglucagón/genética , Ratas , Proteínas Represoras/genética
16.
Mol Cell Biol ; 29(8): 2322-34, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19223471

RESUMEN

Pax6 is important in the development of the pancreas and was previously shown to regulate pancreatic endocrine differentiation, as well as the insulin, glucagon, and somatostatin genes. Prohormone convertase 2 (PC2) is the main processing enzyme in pancreatic alpha cells, where it processes proglucagon to produce glucagon under the spatial and temporal control of 7B2, which functions as a molecular chaperone. To investigate the role of Pax6 in glucagon biosynthesis, we studied potential target genes in InR1G9 alpha cells transfected with Pax6 small interfering RNA and in InR1G9 clones expressing a dominant-negative form of Pax6. We now report that Pax6 controls the expression of the PC2 and 7B2 genes. By binding and transactivation studies, we found that Pax6 indirectly regulates PC2 gene transcription through cMaf and Beta2/NeuroD1 while it activates the 7B2 gene both directly and indirectly through the same transcription factors, cMaf and Beta2/NeuroD1. We conclude that Pax6 is critical for glucagon biosynthesis and processing by directly and indirectly activating the glucagon gene through cMaf and Beta2/NeuroD1, as well as the PC2 and 7B2 genes.


Asunto(s)
Proteínas del Ojo/fisiología , Regulación de la Expresión Génica/fisiología , Proteínas de Homeodominio/fisiología , Proteína 7B2 Secretora Neuroendocrina/genética , Factores de Transcripción Paired Box/fisiología , Proproteína Convertasa 2/genética , Proteínas Represoras/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Línea Celular , Cricetinae , Glucagón/biosíntesis , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteínas del Tejido Nervioso/genética , Factor de Transcripción PAX6 , Proglucagón/metabolismo , Proteínas Proto-Oncogénicas c-maf/genética , Activación Transcripcional
17.
J Biol Chem ; 282(48): 35024-34, 2007 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-17901057

RESUMEN

Specific expression of the glucagon gene in the rat pancreas requires the presence of the G1 element localized at -100/-49 base pairs on the promoter. Although it is known that multiple transcription factors such as Pax-6, Cdx-2/3, c-Maf, Maf-B, and Brain-4 can activate the glucagon gene promoter through G1, their relative importance in vivo is unknown. We first studied the expression of Maf-B, c-Maf, and Cdx-2/3 in the developing and adult mouse pancreas. Although Maf-B was detectable in a progressively increasing number of alpha-cells throughout development and in adulthood, c-Maf and Cdx-2/3 were expressed at low and very low levels, respectively. However, c-Maf but not Cdx-2/3 was detectable in adult islets by Western blot analyses. We then demonstrated the in vivo interactions of Pax-6, Cdx-2/3, Maf-B, and c-Maf but not Brain-4 with the glucagon gene promoter in glucagon-producing cells. Although Pax-6, Cdx-2/3, Maf-B, and c-Maf were all able to bind G1 by themselves, we showed that Pax-6 could interact with Maf-B, c-Maf, and Cdx-2/3 and activate transcription of the glucagon gene promoter. Overexpression of dominant negative forms of Cdx-2/3 and Mafs in alpha-cell lines indicated that Cdx-2/3 and the Maf proteins interact on an overlapping site within G1 and that this binding site is critical in the activation of the glucagon gene promoter. Finally, we show that specific inhibition of Pax-6 and c-Maf but not Cdx-2/3 or Maf-B led to decreases in endogenous glucagon gene expression and that c-Maf binds the glucagon gene promoter in mouse islets. We conclude that Pax-6 and c-Maf interact with G1 to activate basal expression of the glucagon gene.


Asunto(s)
Proteínas del Ojo/fisiología , Regulación de la Expresión Génica , Glucagón/biosíntesis , Proteínas de Homeodominio/fisiología , Factores de Transcripción Paired Box/fisiología , Proteínas Proto-Oncogénicas c-maf/fisiología , Proteínas Represoras/fisiología , Animales , Secuencia de Bases , Línea Celular , Cricetinae , Proteínas del Ojo/genética , Genes Dominantes , Glucagón/genética , Proteínas de Homeodominio/genética , Islotes Pancreáticos/metabolismo , Ratones , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción PAX6 , Factores del Dominio POU/metabolismo , Factores de Transcripción Paired Box/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-maf/genética , Ratas , Proteínas Represoras/genética
18.
Biochem J ; 403(3): 593-601, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17263687

RESUMEN

The transcription factor Nkx6.1 is required for the establishment of functional insulin-producing beta-cells in the endocrine pancreas. Overexpression of Nkx6.1 has been shown to inhibit glucagon gene expression while favouring insulin gene activation. Down-regulation resulted in the opposite effect, suggesting that absence of Nkx6.1 favours glucagon gene expression. To understand the mechanism by which Nkx6.1 suppresses glucagon gene expression, we studied its effect on the glucagon gene promoter activity in non-islet cells using transient transfections and gel-shift analyses. In glucagonoma cells transfected with an Nkx6.1-encoding vector, the glucagon promoter activity was reduced by 65%. In BHK21 cells, Nkx6.1 inhibited by 93% Pax6-mediated activation of the glucagon promoter, whereas Cdx2/3 and Maf stimulations were unaltered. Although Nkx6.1 could interact with both the G1 and G3 element, only the former displayed specificity for Nkx6.1. Mutagenesis of the three potential AT-rich motifs within the G1 revealed that only the Pax6-binding site preferentially interacted with Nkx6.1. Chromatin immunoprecipitation confirmed interaction of Nkx6.1 with the glucagon promoter and revealed a direct competition for binding between Pax6 and Nkx6.1. A weak physical interaction between Pax6 and Nkx6.1 was detected in vitro and in vivo suggesting that Nkx6.1 predominantly inhibits glucagon gene transcription through G1-binding competition. We suggest that cell-specific expression of the glucagon gene may only proceed when Nkx6.1, in combination with Pdx1 and Pax4, are silenced in early alpha-cell precursors.


Asunto(s)
Proteínas del Ojo/antagonistas & inhibidores , Células Secretoras de Glucagón/fisiología , Glucagón/genética , Proteínas de Homeodominio/fisiología , Factores de Transcripción Paired Box/antagonistas & inhibidores , Proteínas Represoras/antagonistas & inhibidores , Transcripción Genética/efectos de los fármacos , Animales , Línea Celular , Cricetinae , Proteínas de Homeodominio/antagonistas & inhibidores , Factor de Transcripción PAX6 , Regiones Promotoras Genéticas/efectos de los fármacos , ARN Mensajero/antagonistas & inhibidores
19.
J Lipid Res ; 46(4): 697-705, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15627654

RESUMEN

The sterol regulatory element binding protein 1 (SREBP-1) is regarded as a major factor involved in the nutritional regulation of lipogenesis. The aim of the present work was to demonstrate its involvement in the response of key genes of glucose and lipid metabolism in liver, adipose tissue, and skeletal muscle during fasting and refeeding. The regulation of hexokinase-2 (HKII) was investigated as a marker of the glucose metabolic pathway and that of FAS was investigated as a marker of the lipogenic pathway. The in vivo association of SREBP-1 with the promoter regions of these genes was determined in the different tissues using chromatin immunoprecipitation assays. Fasting decreased, and refeeding restored, FAS and HKII mRNA and protein levels in each tissue. The concomitant measurement of SREBP-1a and SREBP-1c mRNA levels, of mature SREBP-1 protein abundance in nuclear extracts, and of SREBP-1 interaction with target promoters led to the conclusion that SREBP-1 plays a major role in the response of FAS and HKII genes to nutritional regulation in rodents. These data elucidate the important role of SREBP-1 not only in the regulation of lipid metabolism but also of glucose metabolism and energy homeostasis.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas de Unión al ADN/metabolismo , Ayuno/fisiología , Regulación de la Expresión Génica , Hexoquinasa/genética , Factores de Transcripción/metabolismo , Transcripción Genética/genética , Receptor fas/genética , Tejido Adiposo/metabolismo , Alimentación Animal , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Hígado/metabolismo , Masculino , Músculo Esquelético/metabolismo , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína 1 de Unión a los Elementos Reguladores de Esteroles , Factores de Transcripción/genética
20.
Diabetes ; 53(2): 321-9, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14747281

RESUMEN

Insulin upregulates hexokinase II (HKII) expression in skeletal muscle, and this effect is altered in type 2 diabetic patients. This study was conducted to identify the transcription factors that mediate the effect of insulin on HKII gene expression in human muscle. We have cloned the promoter region of the HKII gene and investigated its regulation in a primary culture of human skeletal muscle cells. We defined a region (-369/-270) that conferred the transcriptional response to insulin. This region contains a sterol regulatory element (SRE) that interacted with the recombinant active form of SRE binding protein-1c (SREBP-1c) in electrophoretic mobility shift assays, and, using chromatin immunoprecipitation assay, we showed that endogenous SREBP-1 interacted directly with the promoter region of the HKII gene in human muscle cells. Mutation of the SRE sequence completely suppressed the response of the promoter to insulin stimulation. Finally, overexpression of the rodent mature form of SREBP-1c (adipocyte determination and differentiation factor-1 [ADD1]-403) was able to reproduce insulin action, whereas a dominant-negative form (ADD1-403R) prevented the effect of insulin on HKII promoter constructs. These results demonstrate that SREBP-1c is involved in the effect of insulin on HKII gene transcription and indicate that it is one of the mediators of insulin action on gene expression in human skeletal muscle.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/fisiología , Proteínas de Unión al ADN/fisiología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hexoquinasa/genética , Insulina/farmacología , Músculo Esquelético/enzimología , Secuencia de Bases , Sitios de Unión , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Células Cultivadas , Clonación Molecular , Proteínas de Unión al ADN/metabolismo , Vectores Genéticos , Hexoquinasa/metabolismo , Humanos , Músculo Esquelético/citología , Regiones Promotoras Genéticas , Proteínas Recombinantes/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...