Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 331
Filtrar
1.
Mol Metab ; 61: 101495, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35421610

RESUMEN

OBJECTIVE: The mechanisms by which glucose stimulates insulin secretion from ß-cells are well established and involve inhibition of ATP-sensitive K+ (KATP) channels, followed by a rise in [Ca2+]c that triggers exocytosis. However, the mechanisms by which glucose controls glucagon release from α-cells are much less known. In particular, it is debated whether the sugar controls glucagon secretion by changing α-cell [Ca2+]c, and whether KATP channels or paracrine factors are involved. The present study addresses these issues. METHODS: We tested the effect of a decrease or an increase of glucose concentration (Gx, with x = concentration in mM) on α-cell [Ca2+]c and glucagon secretion. α-cell [Ca2+]c was monitored using GluCreGCaMP6f mice expressing the Ca2+-sensitive fluorescent protein, GCaMP6f, specifically in α-cells. [Ca2+]c was compared between dispersed α-cells and α-cells within islets to evaluate the potential contribution of an indirect effect of glucose. The same protocols were used for experiments of glucagon secretion from whole islets and [Ca2+]c measurements to test if changes in glucagon release mirror those in α-cell [Ca2+]c. RESULTS: Blockade of KATP channels by sulfonylureas (tolbutamide 100 µM or gliclazide 25 µM) strongly increased [Ca2+]c in both dispersed α-cells and α-cells within islets. By contrast, glucose had no effect on [Ca2+]c in dispersed α-cells, whereas it affected it in α-cells within islets. The effect of glucose was however different in islets expressing (Sst+/+) or not somatostatin (SST) (Sst-/-). Decreasing glucose concentration from G7 to G1 modestly increased α-cell [Ca2+]c, but to a slightly larger extent in Sst+/+ islets than in Sst-/- islets. This G1-induced [Ca2+]c rise was also observed in the continuous presence of sulfonylureas in both Sst+/+ and Sst-/- islets. Increasing glucose concentration from G7 to G20 did not affect α-cell [Ca2+]c in Sst+/+ islets which remained low, whereas it strongly increased it in Sst-/- islets. The observations that this increase was seen only in α-cells within islets but never in dispersed α-cells and that it was abrogated by the gap junction inhibitor, carbenoxolone, point to an indirect effect of G20 and suggest that, in Sst-/- islets, G20-stimulated ß-cells entrain α-cells whereas, in Sst+/+ islets, the concomitant release of SST keeps α-cell [Ca2+]c at low levels. The [Ca2+]c lowering effect of endogenous SST is also supported by the observation that SST receptor antagonists (SSTR2/3) increased [Ca2+]c in α-cells from Sst+/+ islets. All these [Ca2+]c changes induced parallel changes in glucagon release. To test if glucose also controls glucagon release independently of [Ca2+]c changes, additional experiments were performed in the continuous presence of 30 mM K+ and the KATP channel opener diazoxide (250 µM). In these conditions, α-cell [Ca2+]c within islets was elevated and its steady-state level was unaffected by glucose. However, decreasing the glucose concentration from G7 to G1 stimulated glucagon release whereas increasing it from G1 to G15 inhibited it. These effects were also evident in Sst-/- islets, and opposite to those on insulin secretion. CONCLUSIONS: We propose a model according to which glucose controls α-cell [Ca2+]c and glucagon secretion through multiple mechanisms. Increasing the glucose concentration modestly decreases [Ca2+]c in α-cells independently of their KATP channels and partly via SST. The involvement of SST increases with the glucose concentration, and one major effect of SST is to keep α-cell [Ca2+]c at low levels by counteracting the effect of an entrainment of α-cells by ß-cells when ß-cells become stimulated by glucose. All these [Ca2+]c changes induce parallel changes in glucagon release. Glucose also decreases the efficacy of Ca2+ on exocytosis by an attenuating pathway that is opposite to the well-established amplifying pathway controlling insulin release in ß-cells.


Asunto(s)
Calcio , Exocitosis , Células Secretoras de Glucagón , Glucagón , Glucosa , Somatostatina , Adenosina Trifosfato , Animales , Calcio/metabolismo , Cationes Bivalentes/metabolismo , Exocitosis/fisiología , Glucagón/biosíntesis , Glucagón/metabolismo , Células Secretoras de Glucagón/metabolismo , Glucosa/análisis , Glucosa/metabolismo , Canales KATP/metabolismo , Ratones , Somatostatina/metabolismo
2.
J Biol Chem ; 296: 100297, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33460647

RESUMEN

The nutrient sensor O-GlcNAc transferase (OGT) catalyzes posttranslational addition of O-GlcNAc onto target proteins, influencing signaling pathways in response to cellular nutrient levels. OGT is highly expressed in pancreatic glucagon-secreting cells (α-cells), which secrete glucagon in response to hypoglycemia. The objective of this study was to determine whether OGT is necessary for the regulation of α-cell mass and function in vivo. We utilized genetic manipulation to produce two α-cell specific OGT-knockout models: a constitutive glucagon-Cre (αOGTKO) and an inducible glucagon-Cre (i-αOGTKO), which effectively delete OGT in α-cells. Using approaches including immunoblotting, immunofluorescent imaging, and metabolic phenotyping in vivo, we provide the first insight on the role of O-GlcNAcylation in α-cell mass and function. αOGTKO mice demonstrated normal glucose tolerance and insulin sensitivity but displayed significantly lower glucagon levels during both fed and fasted states. αOGTKO mice exhibited significantly lower α-cell glucagon content and α-cell mass at 6 months of age. In fasting, αOGTKO mice showed impaired pyruvate stimulated gluconeogenesis in vivo and reduced glucagon secretion in vitro. i-αOGTKO mice showed similarly reduced blood glucagon levels, defective in vitro glucagon secretion, and normal α-cell mass. Interestingly, both αOGTKO and i-αOGTKO mice had no deficiency in maintaining blood glucose homeostasis under fed or fasting conditions, despite impairment in α-cell mass and function, and glucagon content. In conclusion, these studies provide a first look at the role of OGT signaling in the α-cell, its effect on α-cell mass, and its importance in regulating glucagon secretion in hypoglycemic conditions.


Asunto(s)
Glucemia/metabolismo , Células Secretoras de Glucagón/enzimología , Glucagón/biosíntesis , N-Acetilglucosaminiltransferasas/genética , Obesidad/genética , Acilación/efectos de los fármacos , Animales , Ayuno/metabolismo , Femenino , Efecto Fundador , Glucagón/deficiencia , Células Secretoras de Glucagón/efectos de los fármacos , Células Secretoras de Glucagón/patología , Gluconeogénesis/efectos de los fármacos , Gluconeogénesis/genética , Prueba de Tolerancia a la Glucosa , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/metabolismo , Resistencia a la Insulina , Integrasas/genética , Integrasas/metabolismo , Masculino , Ratones , Ratones Noqueados , N-Acetilglucosaminiltransferasas/deficiencia , Obesidad/enzimología , Obesidad/patología , Ácido Pirúvico/metabolismo , Ácido Pirúvico/farmacología
3.
Cytokine ; 138: 155379, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33271384

RESUMEN

BACKGROUND: Blood has been the usual biological fluid for measuring analytes, but there is mounting evidence that saliva may be also useful for detecting cytokines in a noninvasive way. Thus, in this study we aimed to determine concentration of cytokines and other analytes in saliva from a population of healthy children. METHODS: We collected un-stimulated whole saliva samples from clinically healthy children, and concentration of 17 cytokines and 12 other analytes were measured in supernatants. All values were adjusted by albumin content and were log-transformed before multivariate statistical analysis. RESULTS: We included 114 children (53.5% females) between 6.0 and 11.9 years old. The highest concentrations (medians, pg/µg albumin) were seen for visfatin (183.70) and adiponectin (162.26) and the lowest for IL-13 and IL-2 (~0.003). Albumin concentration was associated with age (rS = 0.39, p < 0.001). In the multivariate analysis, five analytes (C peptide, ghrelin, GLP-1, glucagon, leptin) inversely correlated with age and positively with height-for-age. Age was also positively associated with PAI-1, while height-for-age was also positively associated with insulin and visfatin. Finally, BMI-for-age had a positive correlation with GM-CSF and insulin. CONCLUSIONS: Herein, we provided concentration values for 29 analytes in saliva from healthy children that may be useful as preliminary reference framework in the clinical research setting.


Asunto(s)
Citocinas/metabolismo , Saliva/metabolismo , Adiponectina/biosíntesis , Factores de Edad , Estatura , Péptido C/biosíntesis , Niño , Citocinas/biosíntesis , Femenino , Ghrelina/biosíntesis , Glucagón/biosíntesis , Péptido 1 Similar al Glucagón/biosíntesis , Humanos , Insulina/metabolismo , Interleucina-13/biosíntesis , Interleucina-2/biosíntesis , Leptina/biosíntesis , Masculino , Análisis Multivariante , Nicotinamida Fosforribosiltransferasa/biosíntesis , Valores de Referencia
4.
Proc Natl Acad Sci U S A ; 117(49): 31219-31230, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33229527

RESUMEN

Type 1 diabetes (T1D) results from the autoimmune destruction of ß cells, so cure of firmly established T1D requires both reversal of autoimmunity and restoration of ß cells. It is known that ß cell regeneration in nonautoimmune diabetic mice can come from differentiation of progenitors and/or transdifferentiation of α cells. However, the source of ß cell regeneration in autoimmune nonobese diabetic (NOD) mice remains unclear. Here, we show that, after reversal of autoimmunity by induction of haploidentical mixed chimerism, administration of gastrin plus epidermal growth factor augments ß cell regeneration and normalizes blood glucose in the firmly established diabetic NOD mice. Using transgenic NOD mice with inducible lineage-tracing markers for insulin-producing ß cells, Sox9+ ductal progenitors, Nestin+ mesenchymal stem cells, and glucagon-producing α cells, we have found that both reactivation of dysfunctional low-level insulin expression (insulinlo) ß cells and neogenesis contribute to the regeneration, with the latter predominantly coming from transdifferentiation of α cells. These results indicate that, after reversal of autoimmunity, reactivation of ß cells and transdifferentiation of α cells can provide sufficient new functional ß cells to reach euglycemia in firmly established T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Células Secretoras de Insulina/metabolismo , Células Precursoras de Linfocitos B/metabolismo , Regeneración/genética , Animales , Autoinmunidad/genética , Glucemia/efectos de los fármacos , Transdiferenciación Celular/genética , Quimerismo , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Factor de Crecimiento Epidérmico/farmacología , Femenino , Gastrinas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Glucagón/biosíntesis , Células Secretoras de Glucagón/metabolismo , Insulina/genética , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/patología , Células Madre Mesenquimatosas/inmunología , Ratones , Ratones Endogámicos NOD/genética , Células Precursoras de Linfocitos B/efectos de los fármacos
5.
EBioMedicine ; 50: 306-316, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31780397

RESUMEN

BACKGROUND: The understanding of the regulation of glucagon secretion by pancreatic islet α-cells remains elusive. We aimed to develop an in vitro model for investigating the function of human α-cells under direct influence of glucose and other potential regulators. METHODS: Highly purified human α-cells from islets of deceased donors were re-aggregated in the presence or absence of ß-cells in culture, evaluated for glucagon secretion under various treatment conditions, and compared to that of intact human islets and non-sorted islet cell aggregates. FINDINGS: The pure human α-cell aggregates maintained proper glucagon secretion capability at low concentrations of glucose, but failed to respond to changes in ambient glucose concentration. Addition of purified ß-cells, but not the secreted factors from ß-cells at low or high concentrations of glucose, partly restored the responsiveness of α-cells to glucose with regulated glucagon secretion. The EphA stimulator ephrinA5-fc failed to mimic the inhibitory effect of ß-cells on glucagon secretion. Glibenclamide inhibited glucagon secretion from islets and the α- and ß-mixed cell-aggregates, but not from the α-cell-only aggregates, at 2.0 mM glucose. INTERPRETATION: This study validated the use of isolated and then re-aggregated human islet cells for investigating α-cell function and paracrine regulation, and demonstrated the importance of cell-to-cell contact between α- and ß-cells on glucagon secretion. Loss of proper ß- and α-cell physical interaction in islets likely contributes to the dysregulated glucagon secretion in diabetic patients. Re-aggregated select combinations of human islet cells provide unique platforms for studying islet cell function and regulation.


Asunto(s)
Comunicación Celular , Células Secretoras de Glucagón/metabolismo , Glucagón/biosíntesis , Células Secretoras de Insulina/metabolismo , Adulto , Anciano , Biomarcadores , Células Cultivadas , Técnicas de Cocultivo , Femenino , Citometría de Flujo , Glucosa/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Modelos Biológicos , Comunicación Paracrina , Adulto Joven
6.
Int J Mol Sci ; 20(17)2019 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-31443356

RESUMEN

The gut-derived incretin hormones glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are secreted after meal ingestion and work in concert to promote postprandial insulin secretion. Furthermore, GLP-1 inhibits glucagon secretion when plasma glucose concentrations are above normal fasting concentrations while GIP acts glucagonotropically at low glucose levels. A dual incretin receptor agonist designed to co-activate GLP-1 and GIP receptors was recently shown to elicit robust improvements of glycemic control (mean haemoglobin A1c reduction of 1.94%) and massive body weight loss (mean weight loss of 11.3 kg) after 26 weeks of treatment with the highest dose (15 mg once weekly) in a clinical trial including overweight/obese patients with type 2 diabetes. Here, we describe the mechanisms by which the two incretins modulate alpha cell secretion of glucagon, review the effects of co-administration of GLP-1 and GIP on glucagon secretion, and discuss the potential role of glucagon in the therapeutic effects observed with novel unimolecular dual GLP-1/GIP receptor agonists. For clinicians and researchers, this manuscript offers an understanding of incretin physiology and pharmacology, and provides mechanistic insight into future antidiabetic and obesity treatments.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón/agonistas , Glucagón/biosíntesis , Receptores de la Hormona Gastrointestinal/agonistas , Animales , Diabetes Mellitus Tipo 2/metabolismo , Polipéptido Inhibidor Gástrico/metabolismo , Polipéptido Inhibidor Gástrico/farmacología , Péptido 1 Similar al Glucagón/metabolismo , Péptido 1 Similar al Glucagón/farmacología , Humanos
7.
Postgrad Med ; 131(4): 241-250, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30724638

RESUMEN

Treatment of older adults with type 2 diabetes (T2D) is complex because they represent a heterogeneous group with a broad range of comorbidities, functional abilities, socioeconomic status, and life expectancy. Older adults with T2D are at high risk of recurring hypoglycemia, a condition associated with marked morbidity and mortality, because their counter-regulatory mechanism to hypoglycemia is attenuated, and recurring hypoglycemic episodes can lead to hypoglycemia unawareness. In addition, polypharmacy, a result of multiple chronic comorbidities (including heart disease, stroke, and chronic kidney disease), can increase the risk of severe hypoglycemia, especially when patients are taking sulfonylureas or insulin. Often the signs of hypoglycemia are nonspecific (sweating, dizziness, confusion, visual disturbances) and are mistaken for neurological symptoms or dementia. Consequences of hypoglycemia include acute and long-term cognitive changes, cardiac arrhythmia and myocardial infarction, serious falls, frailty, and death, often resulting in hospitalization, which come at a high economic cost. The American Diabetes Association has recently added three new recommendations regarding hypoglycemia in the elderly, highlighting individualized pharmacotherapy with glucose-lowering agents with a low risk of hypoglycemia and proven cardiovascular safety, avoidance of overtreatment, and simplifying treatment regimens while maintaining HbA1c targets. Thus, glycemic goals can be relaxed in the older population as part of individualized care, and physicians must make treatment decisions that best serve their patients' circumstances. This article highlights the issues faced by older people with T2D, the risk factors for hypoglycemia in this population, and the challenges faced by health care providers regarding glycemic management in this patient group.


Asunto(s)
Envejecimiento/fisiología , Complicaciones de la Diabetes/epidemiología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemia/inducido químicamente , Hipoglucemiantes/uso terapéutico , Factores de Edad , Anciano , Anciano de 80 o más Años , Glucemia , Automonitorización de la Glucosa Sanguínea/instrumentación , Automonitorización de la Glucosa Sanguínea/normas , Enfermedades Cardiovasculares/epidemiología , Continuidad de la Atención al Paciente , Demencia/epidemiología , Diabetes Mellitus Tipo 2/fisiopatología , Glucagón/biosíntesis , Hospitalización/estadística & datos numéricos , Humanos , Hipoglucemia/fisiopatología , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/efectos adversos , Insulina/biosíntesis , Atención Dirigida al Paciente , Guías de Práctica Clínica como Asunto , Factores de Riesgo , Factores de Tiempo
8.
Stem Cell Res Ther ; 9(1): 277, 2018 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-30359326

RESUMEN

BACKGROUND: Transplantation of pancreatic ß cells generated in vitro from pluripotent stem cells (hPSCs) such as embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs) has been proposed as an alternative therapy for diabetes. Though many differentiation protocols have been developed for this purpose, lentivirus-mediated forced expression of transcription factors (TF)-PDX1 and NKX6.1-has been at the forefront for its relatively fast and straightforward approach. However, considering that such cells will be used for therapeutic purposes in the future, it is desirable to develop a procedure that does not leave any footprint on the genome, as any changes of DNAs could potentially be a source of unintended, concerning effects such as tumorigenicity. In this study, we attempted to establish a novel protocol for rapid and footprint-free hESC differentiation into a pancreatic endocrine lineage by using synthetic mRNAs (synRNAs) encoding PDX1 and NKX6.1. We also tested whether siPOU5F1, which reduces the expression of pluripotency gene POU5F1 (also known as OCT4), can enhance differentiation as reported previously for mesoderm and endoderm lineages. METHODS: synRNA-PDX1 and synRNA-NKX6.1 were synthesized in vitro and were transfected five times to hESCs with a lipofection reagent in a modified differentiation culture condition. siPOU5F1 was included only in the first transfection. Subsequently, cells were seeded onto a low attachment plate and aggregated by an orbital shaker. At day 13, the degree of differentiation was assessed by quantitative RT-PCR (qRT-PCR) and immunohistochemistry for endocrine hormones such as insulin, glucagon, and somatostatin. RESULTS: Both PDX1 and NKX6.1 expression were detected in cells co-transfected with synRNA-PDX1 and synRNA-NKX6.1 at day 3. Expression levels of insulin in the transfected cells at day 13 were 450 times and 14 times higher by qRT-PCR compared to the levels at day 0 and in cells cultured without synRNA transfection, respectively. Immunohistochemically, pancreatic endocrine hormones were not detected in cells cultured without synRNA transfection but were highly expressed in cells transfected with synRNA-PDX1, synRNA-NKX6.1, and siPOU5F1 at as early as day 13. CONCLUSIONS: In this study, we report a novel protocol for rapid and footprint-free differentiation of hESCs to endocrine cells.


Asunto(s)
Proteínas de Homeodominio/genética , Células Madre Embrionarias Humanas/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/biosíntesis , ARN Mensajero/síntesis química , Transactivadores/genética , Diferenciación Celular , Ingeniería Celular/métodos , Células Cultivadas , Regulación de la Expresión Génica , Glucagón/biosíntesis , Proteínas de Homeodominio/metabolismo , Células Madre Embrionarias Humanas/citología , Humanos , Células Secretoras de Insulina/citología , Lípidos/química , Factor 3 de Transcripción de Unión a Octámeros/antagonistas & inhibidores , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Somatostatina/biosíntesis , Transactivadores/metabolismo , Transfección
9.
EBioMedicine ; 28: 210-224, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29396301

RESUMEN

The circadian system controls the behavior and multiple physiological functions. In mammals, the suprachiasmatic nucleus (SCN) acts as the master pacemaker and regulates the circadian clocks of peripheral tissues. The SCN receives information regarding the light-dark cycle and is thus synchronized to the external 24-hour environment. In contrast, peripheral clocks, such as the liver clock, receive information from the SCN and other factors; in particular, food intake which leads to insulin secretion induces strong entrainment of the liver clock. On the other hand, the liver clock of insulin-depleted mice treated with streptozotocin (STZ) has been shown to be entrained by scheduled feeding, suggesting that insulin is not necessary for entrainment of the liver clock by feeding. In this study, we aimed to elucidate additional mechanism on entraining liver clock by feeding a protein-only diet and/or amino-acid administration which does not increase insulin levels. We demonstrated that protein-only diet and cysteine administration elicit entrainment of the liver clock via glucagon secretion and/or insulin-like growth factors (IGF-1) production. Our findings suggest that glucagon and/or IGF-1 production are additional key factors in food-induced entrainment.


Asunto(s)
Relojes Circadianos , Cisteína/farmacología , Dieta , Proteínas en la Dieta/farmacología , Glucagón/biosíntesis , Factor I del Crecimiento Similar a la Insulina/biosíntesis , Hígado/metabolismo , Animales , Cisteína/administración & dosificación , Ratones Endogámicos ICR , Podofilino/farmacología , Transducción de Señal/efectos de los fármacos , Estreptozocina
10.
J Clin Endocrinol Metab ; 103(4): 1408-1417, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29408994

RESUMEN

Context: Complete loss of ß-cell function in patients with type 1 diabetes mellitus (T1DM) may lead to an increased risk of severe hypoglycemia. Objective: We aimed to determine the impact of C-peptide status on glucagon response and endogenous glucose production (EGP) during hypoglycemia in patients with T1DM. Design and Setting: We conducted an open, comparative trial. Patients: Ten C-peptide positive (C-pos) and 11 matched C-peptide negative (C-neg) patients with T1DM were enrolled. Intervention: Plasma glucose was normalized over the night fast, and after a steady-state (baseline) plateau all patients underwent a hyperinsulinemic, stepwise hypoglycemic clamp with glucose plateaus of 5.5, 3.5, and 2.5 mmol/L and a recovery phase of 4.0 mmol/L. Blood glucagon was measured with a specific and highly sensitive glucagon assay. EGP was determined with a stable isotope tracer technique. Main Outcome Measure: Impact of C-peptide status on glucagon response and EGP during hypoglycemia. Results: Glucagon concentrations were significantly lower in C-pos and C-neg patients than previously reported. At baseline, C-pos patients had higher glucagon concentrations than C-neg patients (8.39 ± 4.6 vs 4.19 ± 2.4 pmol/L, P = 0.016, mean ± standard deviation) but comparable EGP rates (2.13 ± 0.2 vs 2.04 ± 0.3 mg/kg/min, P < 0.391). In both groups, insulin suppressed glucagon levels, but hypoglycemia revealed significantly higher glucagon concentrations in C-pos than in C-neg patients. EGP was significantly higher in C-pos patients at hypoglycemia (2.5 mmol/L) compared with C-neg patients. Conclusions: Glucagon concentrations and EGP during hypoglycemia were more pronounced in C-pos than in C-neg patients, which indicates that preserved ß-cell function may contribute to counterregulation during hypoglycemia in patients with T1DM.


Asunto(s)
Glucemia/biosíntesis , Péptido C/fisiología , Diabetes Mellitus Tipo 1/sangre , Glucagón/biosíntesis , Hipoglucemia/sangre , Adulto , Concienciación , Péptido C/sangre , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Esquema de Medicación , Epinefrina/sangre , Femenino , Glucagón/sangre , Técnica de Clampeo de la Glucosa/métodos , Humanos , Hipoglucemia/psicología , Insulina/administración & dosificación , Insulina/uso terapéutico , Masculino , Persona de Mediana Edad , Norepinefrina/sangre , Adulto Joven
12.
J Cell Biol ; 216(12): 4299-4311, 2017 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-29025873

RESUMEN

The recent demonstration that pancreatic α cells can be continuously regenerated and converted into ß-like cells upon ectopic expression of Pax4 opened new avenues of research in the endocrine cell differentiation and diabetes fields. To determine whether such plasticity was also shared by δ cells, we generated and characterized transgenic animals that express Pax4 specifically in somatostatin-expressing cells. We demonstrate that the ectopic expression of Pax4 in δ cells is sufficient to induce their conversion into functional ß-like cells. Importantly, this conversion induces compensatory mechanisms involving the reactivation of endocrine developmental processes that result in dramatic ß-like cell hyperplasia. Importantly, these ß-like cells are functional and can partly reverse the consequences of chemically induced diabetes.


Asunto(s)
Diabetes Mellitus Experimental/genética , Expresión Génica Ectópica , Proteínas de Homeodominio/genética , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Paired Box/genética , Células Secretoras de Somatostatina/metabolismo , Animales , Proliferación Celular , Transdiferenciación Celular/genética , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/terapia , Terapia Genética/métodos , Glucagón/biosíntesis , Glucagón/genética , Proteínas de Homeodominio/metabolismo , Insulina/biosíntesis , Insulina/genética , Células Secretoras de Insulina/citología , Masculino , Ratones , Ratones Transgénicos , Factores de Transcripción Paired Box/metabolismo , Somatostatina/biosíntesis , Somatostatina/genética , Células Secretoras de Somatostatina/citología , Estreptozocina
13.
J Neuroendocrinol ; 29(1)2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27891681

RESUMEN

There is increasing evidence to suggest that type 2 diabetes mellitus (T2D), a pandemic metabolic disease, may be caused by ß-cell dedifferentiation (ßCD). However, there is currently no universal definition of ßCD, and the underlying mechanism is poorly understood. We hypothesise that a high-glucose in vitro environment mimics hyperglycaemia in vivo and that ß cells grown in this milieu over a long period will undergo dedifferentiation. In the present study, we report that the pancreatic ß cell line mouse insulinoma 6 (MIN6) grown under a high-glucose condition did not undergo massive cell death but exhibited a glucose-stimulated insulin-secreting profile similar to that of immature ß cells. The expression of insulin and the glucose-sensing molecule glucose transporter 2 (Glut2) in late passage MIN6 cells was significantly lower than the early passage at both the RNA and protein levels. Mechanistically, these cells also expressed significantly less of the 'pancreatic and duodenal homebox1' (Pdx1) ß-cell transcription factor. Finally, passaged MIN6 cells dedifferentiated to demonstrate some features of ß-cell precursors, as well as neuroendocrine markers, in addition to expressing both glucagon and insulin. Thus, we concluded that high-glucose passaged MIN6 cells passaged MIN6 cells. provide a cellular model of ß-cell dedifferentiation that can help researchers develop a better understanding of this process. These findings provide new insights that may enhance knowledge of the pathophysiology of T2D and facilitate the establishment of a novel strategy by which this disease can be treated.


Asunto(s)
Biomarcadores/metabolismo , Desdiferenciación Celular/fisiología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Glucagón/biosíntesis , Glucosa/farmacología , Transportador de Glucosa de Tipo 2/biosíntesis , Proteínas de Homeodominio/biosíntesis , Insulina/biosíntesis , Insulina/metabolismo , Secreción de Insulina , Ratones , Proteínas del Tejido Nervioso/biosíntesis , Transactivadores/biosíntesis , Factores de Transcripción/biosíntesis
14.
Tissue Cell ; 48(6): 567-576, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27823763

RESUMEN

In the pancreas of many mammals including humans, endocrine islet cells can be integrated with the nervous system components into neuro-insular complexes. The mechanism of the formation of such complexes is not clearly understood. The present study evaluated the interactions between the nervous system components, epithelial cells and endocrine cells in the human pancreas. Foetal pancreas, gestational age 19-23 weeks (13 cases) and 30-34 weeks (7 cases), were studied using double immunohistochemical labeling with neural markers (S100 protein and beta III tubulin), epithelial marker (cytokeratin 19 (CK19)) and antibodies to insulin and glucagon. We first analyse the structure of neuro-insular complexes using confocal microscopy and provide immunohistochemical evidences of the presence of endocrine cells within the ganglia or inside the nerve bundles. We showed that the nervous system components contact with the epithelial cells located in ducts or in clusters outside the ductal epithelium and form complexes with separate epithelial cells. We observed CK19-positive cells inside the ganglia and nerve bundles which were located separately or were integrated with the islets. Therefore, we conclude that neuro-insular complexes may forms as a result of integration between epithelial cells and nervous system components at the initial stages of islets formation.


Asunto(s)
Células Endocrinas/metabolismo , Células Epiteliales/metabolismo , Sistema Nervioso/metabolismo , Páncreas/metabolismo , Feto , Ganglios/crecimiento & desarrollo , Ganglios/metabolismo , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Glucagón/biosíntesis , Humanos , Insulina/biosíntesis , Queratina-19/biosíntesis , Sistema Nervioso/crecimiento & desarrollo , Páncreas/embriología , Páncreas/crecimiento & desarrollo , Proteínas S100/biosíntesis , Tubulina (Proteína)/biosíntesis
15.
Oncotarget ; 7(46): 74415-74426, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27769070

RESUMEN

Amplification or overexpression of MYCN is involved in development and maintenance of multiple malignancies. A subset of these tumors originates from neural precursors, including the most aggressive forms of the childhood tumors, neuroblastoma and medulloblastoma. In order to model the spectrum of MYCN-driven neoplasms in mice, we transgenically overexpressed MYCN under the control of the human GFAP-promoter that, among other targets, drives expression in neural progenitor cells. However, LSL-MYCN;hGFAP-Cre double transgenic mice did neither develop neural crest tumors nor tumors of the central nervous system, but presented with neuroendocrine tumors of the pancreas and, less frequently, the pituitary gland. Pituitary tumors expressed chromogranin A and closely resembled human pituitary adenomas. Pancreatic tumors strongly produced and secreted glucagon, suggesting that they derived from glucagon- and GFAP-positive islet cells. Interestingly, 3 out of 9 human pancreatic neuroendocrine tumors expressed MYCN, supporting the similarity of the mouse tumors to the human system. Serial transplantations of mouse tumor cells into immunocompromised mice confirmed their fully transformed phenotype. MYCN-directed treatment by AuroraA- or Brd4-inhibitors resulted in significantly decreased cell proliferation in vitro and reduced tumor growth in vivo. In summary, we provide a novel mouse model for neuroendocrine tumors of the pancreas and pituitary gland that is dependent on MYCN expression and that may help to evaluate MYCN-directed therapies.


Asunto(s)
Expresión Génica , Proteína Ácida Fibrilar de la Glía/genética , Glucagón/biosíntesis , Proteína Proto-Oncogénica N-Myc/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Proteína Ácida Fibrilar de la Glía/metabolismo , Glucagonoma/genética , Glucagonoma/metabolismo , Glucagonoma/patología , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , Proteína Proto-Oncogénica N-Myc/metabolismo , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/patología , Neoplasias Hipofisarias/patología , Transcriptoma
16.
Anim Sci J ; 87(9): 1090-8, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27150835

RESUMEN

The regulatory mechanisms underlying food intake in chickens have been a focus of research in recent decades to improve production efficiency when raising chickens. Lines of evidence have revealed that a number of brain-gut peptides function as a neurotransmitter or peripheral satiety hormone in the regulation of food intake both in mammals and chickens. Glucagon, a 29 amino acid peptide hormone, has long been known to play important roles in maintaining glucose homeostasis in mammals and birds. However, the glucagon gene encodes various peptides that are produced by tissue-specific proglucagon processing: glucagon is produced in the pancreas, whereas oxyntomodulin (OXM), glucagon-like peptide (GLP)-1 and GLP-2 are produced in the intestine and brain. Better understanding of the roles of these peptides in the regulation of energy homeostasis has led to various physiological roles being proposed in mammals. For example, GLP-1 functions as an anorexigenic neurotransmitter in the brain and as a postprandial satiety hormone in the peripheral circulation. There is evidence that OXM and GLP-2 also induce anorexia in mammals. Therefore, it is possible that the brain-gut peptides OXM, GLP-1 and GLP-2 play physiological roles in the regulation of food intake in chickens. More recently, a novel GLP and its specific receptor were identified in the chicken brain. This review summarizes current knowledge about the role of glucagon-related peptides in the regulation of food intake in chickens.


Asunto(s)
Pollos/genética , Pollos/fisiología , Ingestión de Alimentos/genética , Ingestión de Alimentos/fisiología , Glucagón/genética , Glucagón/fisiología , Neurotransmisores , Animales , Encéfalo/metabolismo , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Glucagón/biosíntesis , Péptido 1 Similar al Glucagón/biosíntesis , Péptido 1 Similar al Glucagón/genética , Péptido 1 Similar al Glucagón/fisiología , Péptido 2 Similar al Glucagón/biosíntesis , Péptido 2 Similar al Glucagón/genética , Péptido 2 Similar al Glucagón/fisiología , Glucosa/metabolismo , Homeostasis/genética , Homeostasis/fisiología , Mucosa Intestinal/metabolismo , Especificidad de Órganos , Oxintomodulina/biosíntesis , Oxintomodulina/genética , Oxintomodulina/fisiología , Páncreas/metabolismo
17.
Diabetes ; 65(6): 1534-55, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26895793

RESUMEN

Mitochondrial metabolism plays an integral role in glucose-stimulated insulin secretion (GSIS) in ß-cells. In addition, the diabetogenic role of glucagon released from α-cells plays a major role in the etiology of both type 1 and type 2 diabetes because unopposed hyperglucagonemia is a pertinent contributor to diabetic hyperglycemia. Titrating expression levels of the mitochondrial protein mitoNEET is a powerful approach to fine-tune mitochondrial capacity of cells. Mechanistically, ß-cell-specific mitoNEET induction causes hyperglycemia and glucose intolerance due to activation of a Parkin-dependent mitophagic pathway, leading to the formation of vacuoles and uniquely structured mitophagosomes. Induction of mitoNEET in α-cells leads to fasting-induced hypoglycemia and hypersecretion of insulin during GSIS. MitoNEET-challenged α-cells exert potent antiapoptotic effects on ß-cells and prevent cellular dysfunction associated with mitoNEET overexpression in ß-cells. These observations identify that reduced mitochondrial function in α-cells exerts potently protective effects on ß-cells, preserving ß-cell viability and mass.


Asunto(s)
Células Secretoras de Glucagón/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas de Unión a Hierro/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Apoptosis , Supervivencia Celular , Glucagón/biosíntesis , Glucosa/metabolismo , Intolerancia a la Glucosa/etiología , Hiperglucemia/etiología , Insulina/metabolismo , Secreción de Insulina , Ratones , Receptor Cross-Talk
18.
Bosn J Basic Med Sci ; 16(1): 1-7, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26773171

RESUMEN

Pancreatic islet α-cell tumours that overexpress proglucagon are typically associated with the glucagonoma syndrome, a rare disease entity characterised by necrolytic migratory erythema, impaired glucose tolerance, thromboembolic complications and psychiatric disturbances. Paraneoplastic phenomena associated with enteric overexpression of proglucagon-derived peptides are less well recognized and include gastrointestinal dysfunction and hyperinsulinaemic hypoglycaemia. The diverse clinical manifestations associated with glucagon-expressing tumours can be explained, in part, by the repertoire of tumorally secreted peptides liberated through differential post-translational processing of tumour-derived proglucagon. Proglucagon-expressing tumours may be divided into two broad biochemical subtypes defined by either secretion of glucagon or GLP-1, GLP-2 and the glucagon-containing peptides, glicentin and oxyntomodulin, due to an islet α-cell or enteroendocrine L-cell pattern of proglucagon processing, respectively. In the current review we provide an updated overview of the clinical presentation of proglucagon-expressing tumours in relation to known physiological actions of proglucagon-derived peptides and suggest that detailed biochemical characterisation of the peptide repertoire secreted from these tumours may provide new opportunities for diagnosis and clinical management.


Asunto(s)
Células Secretoras de Glucagón/metabolismo , Glucagón/biosíntesis , Glucagonoma/metabolismo , Islotes Pancreáticos/citología , Neoplasias Pancreáticas/metabolismo , Animales , Enfermedades Gastrointestinales/metabolismo , Regulación de la Expresión Génica , Glicentina/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Humanos , Hipoglucemia/metabolismo , Oxintomodulina/metabolismo , Páncreas/metabolismo , Fragmentos de Péptidos , Péptidos/química , Fenotipo , Proglucagón/metabolismo , Dominios Proteicos
19.
J Biomol Screen ; 21(4): 325-32, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26676097

RESUMEN

Glucose homeostasis is primarily controlled by two opposing hormones, insulin and glucagon, and diabetes results when insulin fails to inhibit glucagon action. Recent efforts to control glucagon in diabetes have focused on antagonizing the glucagon receptor, which is effective in lowering blood glucose levels but leads to hyperglucogonemia in rodents. An alternative strategy would be to control glucagon production with small molecules. In pursuit of this goal, we developed a homogeneous AlphaScreen assay for measuring glucagon in cell culture media and used this in a high-throughput screen to discover synthetic compounds that inhibited glucagon secretion from an alpha cell-like cell line. Some of these compounds inhibited transcription of the glucagon gene.


Asunto(s)
Células Secretoras de Glucagón/efectos de los fármacos , Glucagón/antagonistas & inhibidores , Hipoglucemiantes/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Biotina/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cricetinae , Expresión Génica , Genes Reporteros , Glucagón/biosíntesis , Glucagón/genética , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Hipoglucemiantes/química , Cinética , Luciferasas/antagonistas & inhibidores , Luciferasas/genética , Luciferasas/metabolismo , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Estreptavidina/química
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...