Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Physiol ; 84: 17-40, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-34705480

RESUMEN

ß-Arrestin-1 and -2 (also known as arrestin-2 and -3, respectively) are ubiquitously expressed cytoplasmic proteins that dampen signaling through G protein-coupled receptors. However, ß-arrestins can also act as signaling molecules in their own right. To investigate the potential metabolic roles of the two ß-arrestins in modulating glucose and energy homeostasis, recent studies analyzed mutant mice that lacked or overexpressed ß-arrestin-1 and/or -2 in distinct, metabolically important cell types. Metabolic analysis of these mutant mice clearly demonstrated that both ß-arrestins play key roles in regulating the function of most of these cell types, resulting in striking changes in whole-body glucose and/or energy homeostasis. These studies also revealed that ß-arrestin-1 and -2, though structurally closely related, clearly differ in their metabolic roles under physiological and pathophysiological conditions. These new findings should guide the development of novel drugs for the treatment of various metabolic disorders, including type 2 diabetes and obesity.


Asunto(s)
Diabetes Mellitus Tipo 2 , Glucosa , Animales , Glucosa/metabolismo , Homeostasis , Humanos , Ratones , beta-Arrestina 1/metabolismo , beta-Arrestinas/metabolismo
2.
Proc Natl Acad Sci U S A ; 112(49): E6818-24, 2015 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-26598688

RESUMEN

G protein-coupled receptors (GPCRs) regulate virtually all physiological functions including the release of insulin from pancreatic ß-cells. ß-Cell M3 muscarinic receptors (M3Rs) are known to play an essential role in facilitating insulin release and maintaining proper whole-body glucose homeostasis. As is the case with other GPCRs, M3R activity is regulated by phosphorylation by various kinases, including GPCR kinases and casein kinase 2 (CK2). At present, it remains unknown which of these various kinases are physiologically relevant for the regulation of ß-cell activity. In the present study, we demonstrate that inhibition of CK2 in pancreatic ß-cells, knockdown of CK2α expression, or genetic deletion of CK2α in ß-cells of mutant mice selectively augmented M3R-stimulated insulin release in vitro and in vivo. In vitro studies showed that this effect was associated with an M3R-mediated increase in intracellular calcium levels. Treatment of mouse pancreatic islets with CX4945, a highly selective CK2 inhibitor, greatly reduced agonist-induced phosphorylation of ß-cell M3Rs, indicative of CK2-mediated M3R phosphorylation. We also showed that inhibition of CK2 greatly enhanced M3R-stimulated insulin secretion in human islets. Finally, CX4945 treatment protected mice against diet-induced hyperglycemia and glucose intolerance in an M3R-dependent fashion. Our data demonstrate, for the first time to our knowledge, the physiological relevance of CK2 phosphorylation of a GPCR and suggest the novel concept that kinases acting on ß-cell GPCRs may represent novel therapeutic targets.


Asunto(s)
Quinasa de la Caseína II/fisiología , Insulina/metabolismo , Receptor Muscarínico M3/fisiología , Animales , Células COS , Chlorocebus aethiops , Femenino , Células HEK293 , Humanos , Secreción de Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Naftiridinas/farmacología , Fenazinas
3.
Exp Physiol ; 100(1): 57-68, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25398717

RESUMEN

NEW FINDINGS: What is the central question of this study? Different nerve contributes periods of life are known for their differential sensitivity to interventions, and increased parasympathetic activity affects the development and maintenance of obesity. Thus, we evaluated the involvement of the vagus nerve by performing a vagotomy in young or adult rats that were offered an obesogenic high-fat diet. What is the main finding and its importance? Although the accumulation of adipose tissue decreased in both younger and older groups, the younger rats showed a greater response to the effects of vagotomy in general. In addition to the important role of the parasympathetic activity, we suggest that the vagus nerve contributes to the condition of obesity. Obesity has become a global problem, and this condition develops primarily because of an imbalance between energy intake and expenditure. The high complexity involved in the regulation of energy metabolism results from several factors besides endocrine factors. It has been suggested that obesity could be caused by an imbalance in the autonomous nervous system, which could lead to a condition of high parasympathetic activity in counterpart to low sympathetic tonus. High-fat (HF) diets have been used to induce obesity in experimental animals, and their use in animals leads to insulin resistance, hyperinsulinaemia and high parasympathetic activity, among other disorders. The aim of this work was to evaluate the effects of a vagotomy performed at the initiation of a HF diet at two different stages of life, weaning and adulthood. The vagotomy reduced parasympathetic activity (-32 and -51% in normal fat-fed rats and -43 and -55% in HF diet-fed rats; P < 0.05) and fat depots (-17 and -33%, only in HF diet-fed rats; P < 0.05). High-fat diet-fed rats exhibited fasting hyperinsulinaemia (fivefold higher in young rats and threefold higher in older rats; P < 0.05); however, vagotomy corrected it in younger rats only, and a similar effect was also observed during the glucose tolerance test. The insulin resistance exhibited by the HF diet-fed groups was not altered in the vagotomized rats. We suggest that the vagus nerve, in addition to the important role of parasympathetic activity, contributes to the condition of obesity, and that non-vagal pathways may be involved along with the imbalanced autonomic nervous system.


Asunto(s)
Dieta Alta en Grasa , Síndrome Metabólico/etiología , Obesidad/etiología , Nervio Vago/fisiopatología , Adiposidad , Factores de Edad , Animales , Biomarcadores/sangre , Glucemia/metabolismo , Modelos Animales de Enfermedad , Insulina/sangre , Resistencia a la Insulina , Masculino , Síndrome Metabólico/sangre , Síndrome Metabólico/fisiopatología , Síndrome Metabólico/prevención & control , Obesidad/sangre , Obesidad/fisiopatología , Obesidad/prevención & control , Ratas Wistar , Factores de Tiempo , Vagotomía , Nervio Vago/cirugía , Destete , Aumento de Peso
4.
Cell Physiol Biochem ; 33(4): 1075-86, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24732778

RESUMEN

BACKGROUND/AIMS: Impaired pancreatic beta cell function and insulin secretion/action are a link between obesity and type 2 diabetes, which are worldwide public health burdens. We aimed to characterize the muscarinic acetylcholine receptor (mAChR) M1-M4 subtypes in isolated pancreatic islets from pre-diabetic obese rats that had been treated neonatally with monosodium L-glutamate (MSG). METHODS: At 90 days of age, both the MSG and the control groups underwent biometric and biochemical evaluation. Anti-muscarinic drugs were used to study mAChR function either in vivo or in vitro. RESULTS: The results demonstrated that atropine treatment reduced insulin secretion in the MSG-treated and control groups, whereas treatment with an M2mAChR-selective antagonist increased secretion. Moreover, the insulinostatic effect of an M3mAChR-selective antagonist was significantly higher in the MSG-treated group. M1mAChR and M3mAChR expression was increased in the MSG-obese group by 55% and 73%, respectively. In contrast, M2mAChR expression decreased by 25% in the MSG group, whereas M4mAChR expression was unchanged. CONCLUSIONS: Functional changes in and altered content of the mAChR (M1-M4) subtypes are pivotal to the demand for high pancreatic beta cell insulin secretion in MSG-obese rats, which is directly associated with vagal hyperactivity and peripheral insulin resistance.


Asunto(s)
Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Obesidad/metabolismo , Receptores Muscarínicos/metabolismo , Glutamato de Sodio/farmacología , Animales , Glucemia/análisis , Prueba de Tolerancia a la Glucosa , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Masculino , Agonistas Muscarínicos/farmacología , Antagonistas Muscarínicos/farmacología , Obesidad/patología , Ratas , Ratas Wistar , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M2/metabolismo , Receptor Muscarínico M3/metabolismo , Receptor Muscarínico M4/metabolismo , Receptores Muscarínicos/química
5.
Br J Nutr ; 111(2): 227-35, 2014 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-23841989

RESUMEN

Impaired pancreatic ß-cell function, as observed in the cases of early nutrition disturbance, is a major hallmark of metabolic diseases arising in adulthood. In the present study, we aimed to investigate the function/composition of the muscarinic acetylcholine receptor (mAChR) subtypes, M2 and M3, in the pancreatic islets of adult offspring of rats that were protein malnourished during lactation. Neonates were nursed by mothers that were fed either a low-protein (4 %, LP) or a normal-protein (23 %, NP) diet. Adult rats were pre-treated with anti-muscarinic drugs and subjected to the glucose tolerance test; the function and protein expression levels of M2mAChR and M3mAChR were determined. The LP rats were lean and hypoinsulinaemic. The selective M2mAChR antagonist methoctramine increased insulinaemia by 31 % in the NP rats and 155 % in the LP rats, and insulin secretion was increased by 32 % in the islets of the NP rats and 88 % in those of the LP rats. The selective M3mAChR antagonist 4-diphenylacetoxy-N-methylpiperidine methiodide decreased insulinaemia by 63 % in the NP rats and 40 % in the LP rats and reduced insulin release by 41 % in the islets of the NP rats and 28 % in those of the LP rats. The protein expression levels of M2mAChR and M3mAChR were 57 % higher and 53 % lower, respectively, in the islets of the LP rats than in those of the NP rats. The expression and functional compositions of M2mAChR and M3mAChR were altered in the islets of the LP rats, as a result of metabolic programming caused by the protein-restricted diet, which might be another possible effect involved in the weak insulin secretion ability of the islets of the programmed adult rats.


Asunto(s)
Alimentación Animal/análisis , Proteínas en la Dieta/administración & dosificación , Células Secretoras de Insulina/fisiología , Lactancia/fisiología , Receptores Muscarínicos/clasificación , Receptores Muscarínicos/metabolismo , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Glucemia , Dieta/veterinaria , Femenino , Glucosa/metabolismo , Intolerancia a la Glucosa , Prueba de Tolerancia a la Glucosa , Homeostasis , Masculino , Fenómenos Fisiologicos Nutricionales Maternos , Antagonistas Muscarínicos/farmacología , Embarazo , Ratas , Ratas Wistar
6.
Eur J Nutr ; 53(3): 711-22, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24468940

RESUMEN

Nutrition and lifestyle, particularly over-nutrition and lack of exercise, promote the progression and pathogenesis of obesity and metabolic diseases. Nutrition is likely the most important environmental factor that modulates the expression of genes involved in metabolic pathways and a variety of phenotypes associated with obesity and diabetes. During pregnancy, diet is a major factor that influences the organ developmental plasticity of the foetus. Experimental evidence shows that nutritional factors, including energy, fatty acids, protein, micronutrients, and folate, affect various aspects of metabolic programming. Different epigenetic mechanisms that are elicited by bioactive factors in early critical developmental ages affect the susceptibility to several diseases in adulthood. The beneficial effects promoted by exercise training are well recognised, and physical exercise may be considered one of the more prominent non-pharmacological tools that can be used to attenuate metabolic programming and to consequently ameliorate the illness provoked by metabolic diseases and reduce the prevalence of obesity, type 2 diabetes, and cardiovascular diseases. Literature on the different outcomes of unbalanced diets and the beneficial effects of some bioactive molecules during gestation and lactation on the metabolic health of offspring, as well as the potential mechanisms underlying these effects, was reviewed. The importance of the combined effects of functional nutrition and exercise as reprogramming tools of metabolic programming is discussed in depth. Finally, this review provides recommendations to healthcare providers that may aid in the control of early programming in an attempt to optimise the health of the mother and child.


Asunto(s)
Medicina Basada en la Evidencia , Hiperfagia/fisiopatología , Conducta Materna , Fenómenos Fisiologicos Nutricionales Maternos , Intercambio Materno-Fetal , Síndrome Metabólico/etiología , Conducta Sedentaria , Animales , Desarrollo Infantil , Susceptibilidad a Enfermedades , Epigénesis Genética , Ejercicio Físico , Femenino , Desarrollo Fetal , Humanos , Hiperfagia/dietoterapia , Hiperfagia/metabolismo , Lactante , Recién Nacido , Lactancia/metabolismo , Masculino , Síndrome Metabólico/metabolismo , Síndrome Metabólico/prevención & control , Actividad Motora , Embarazo
7.
Nat Commun ; 15(1): 5129, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38879678

RESUMEN

Glucagon, a hormone released from pancreatic α-cells, is critical for maintaining euglycemia and plays a key role in the pathophysiology of diabetes. To stimulate the development of new classes of therapeutic agents targeting glucagon release, key α-cell signaling pathways that regulate glucagon secretion need to be identified. Here, we focused on the potential importance of α-cell Gs signaling on modulating α-cell function. Studies with α-cell-specific mouse models showed that activation of α-cell Gs signaling causes a marked increase in glucagon secretion. We also found that intra-islet adenosine plays an unexpected autocrine/paracrine role in promoting glucagon release via activation of α-cell Gs-coupled A2A adenosine receptors. Studies with α-cell-specific Gαs knockout mice showed that α-cell Gs also plays an essential role in stimulating the activity of the Gcg gene, thus ensuring proper islet glucagon content. Our data suggest that α-cell enriched Gs-coupled receptors represent potential targets for modulating α-cell function for therapeutic purposes.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs , Células Secretoras de Glucagón , Glucagón , Ratones Noqueados , Transducción de Señal , Glucagón/metabolismo , Animales , Células Secretoras de Glucagón/metabolismo , Ratones , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Adenosina/metabolismo , Receptor de Adenosina A2A/metabolismo , Receptor de Adenosina A2A/genética , Masculino , Ratones Endogámicos C57BL , Islotes Pancreáticos/metabolismo
8.
Endocrinology ; 163(7)2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35641126

RESUMEN

Pancreatic islet beta cells require a fine-tuned endoplasmic reticulum (ER) stress response for normal function; abnormal ER stress contributes to diabetes pathogenesis. Here, we identified a small molecule, SW016789, with time-dependent effects on beta cell ER stress and function. Acute treatment with SW016789 potentiated nutrient-induced calcium influx and insulin secretion, while chronic exposure to SW016789 transiently induced ER stress and shut down secretory function in a reversible manner. Distinct from the effects of thapsigargin, SW016789 did not affect beta cell viability or apoptosis, potentially due to a rapid induction of adaptive genes, weak signaling through the eIF2α kinase PERK, and lack of oxidative stress gene Txnip induction. We determined that SW016789 acted upstream of voltage-dependent calcium channels (VDCCs) and potentiated nutrient- but not KCl-stimulated calcium influx. Measurements of metabolomics, oxygen consumption rate, and G protein-coupled receptor signaling did not explain the potentiating effects of SW016789. In chemical cotreatment experiments, we discovered synergy between SW016789 and activators of protein kinase C and VDCCs, suggesting involvement of these pathways in the mechanism of action. Finally, chronically elevated calcium influx was required for the inhibitory impact of SW016789, as blockade of VDCCs protected human islets and MIN6 beta cells from hypersecretion-induced dysfunction. We conclude that beta cells undergoing this type of pharmacological hypersecretion have the capacity to suppress their function to mitigate ER stress and avoid apoptosis. These results have the potential to uncover beta cell ER stress mitigation factors and add support to beta cell rest strategies to preserve function.


Asunto(s)
Células Secretoras de Insulina , Insulina , Apoptosis , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo
9.
Mol Metab ; 55: 101415, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34883278

RESUMEN

OBJECTIVE: The goal of this study was to determine the glucometabolic effects of acute activation of Gs signaling in skeletal muscle (SKM) in vivo and its contribution to whole-body glucose homeostasis. METHODS: To address this question, we studied mice that express a Gs-coupled designer G protein-coupled receptor (Gs-DREADD or GsD) selectively in skeletal muscle. We also identified two Gs-coupled GPCRs that are endogenously expressed by SKM at relatively high levels (ß2-adrenergic receptor and CRF2 receptor) and studied the acute metabolic effects of activating these receptors in vivo by highly selective agonists (clenbuterol and urocortin 2 (UCN2), respectively). RESULTS: Acute stimulation of GsD signaling in SKM impaired glucose tolerance in lean and obese mice by decreasing glucose uptake selectively into SKM. The acute metabolic effects following agonist activation of ß2-adrenergic and, potentially, CRF2 receptors appear primarily mediated by altered insulin release. Clenbuterol injection improved glucose tolerance by increasing insulin secretion in lean mice. In SKM, clenbuterol stimulated glycogen breakdown. UCN2 injection resulted in decreased glucose tolerance associated with lower plasma insulin levels. The acute metabolic effects of UCN2 were not mediated by SKM Gs signaling. CONCLUSIONS: Selective activation of Gs signaling in SKM causes an acute increase in blood glucose levels. However, acute in vivo stimulation of endogenous Gs-coupled receptors enriched in SKM has only a limited impact on whole-body glucose homeostasis, most likely due to the fact that these receptors are also expressed by pancreatic islets where they modulate insulin release.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Músculo Esquelético/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Clenbuterol/farmacología , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/fisiología , Glucosa/metabolismo , Intolerancia a la Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Secreción de Insulina/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/fisiología , Obesidad/metabolismo , Receptores Adrenérgicos beta 2/metabolismo
10.
Nat Commun ; 13(1): 22, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013148

RESUMEN

Activation of the sympathetic nervous system causes pronounced metabolic changes that are mediated by multiple adrenergic receptor subtypes. Systemic treatment with ß2-adrenergic receptor agonists results in multiple beneficial metabolic effects, including improved glucose homeostasis. To elucidate the underlying cellular and molecular mechanisms, we chronically treated wild-type mice and several newly developed mutant mouse strains with clenbuterol, a selective ß2-adrenergic receptor agonist. Clenbuterol administration caused pronounced improvements in glucose homeostasis and prevented the metabolic deficits in mouse models of ß-cell dysfunction and insulin resistance. Studies with skeletal muscle-specific mutant mice demonstrated that these metabolic improvements required activation of skeletal muscle ß2-adrenergic receptors and the stimulatory G protein, Gs. Unbiased transcriptomic and metabolomic analyses showed that chronic ß2-adrenergic receptor stimulation caused metabolic reprogramming of skeletal muscle characterized by enhanced glucose utilization. These findings strongly suggest that agents targeting skeletal muscle metabolism by modulating ß2-adrenergic receptor-dependent signaling pathways may prove beneficial as antidiabetic drugs.


Asunto(s)
Reprogramación Celular/efectos de los fármacos , Clenbuterol/farmacología , Hipoglucemiantes/farmacología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Animales , Fenómenos Bioquímicos , Clenbuterol/metabolismo , Femenino , Glucosa/metabolismo , Homeostasis , Resistencia a la Insulina , Masculino , Enfermedades Metabólicas , Metabolómica , Ratones , Ratones Noqueados , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal
11.
Trends Endocrinol Metab ; 32(2): 118-129, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33358450

RESUMEN

ß-Arrestin-1 and -2 are intracellular proteins that are able to inhibit signaling via G protein-coupled receptors (GPCRs). However, both proteins can also modulate cellular functions in a G protein-independent fashion. During the past few years, studies with mutant mice selectivity lacking ß-arrestin-1 and/or -2 in metabolically important cell types have led to novel insights into the mechanisms through which ß-arrestins regulate key metabolic processes in vivo, including whole-body glucose and energy homeostasis. The novel information gained from these studies should inform the development of novel drugs, including ß-arrestin- or G protein-biased GPCR ligands, that could prove useful for the therapy of several important pathophysiological conditions, including type 2 diabetes and obesity.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestinas/metabolismo , Animales , Humanos , Ratones , Unión Proteica , Transducción de Señal/fisiología
12.
FEBS J ; 288(8): 2622-2644, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33682344

RESUMEN

The incidence of obesity and type 2 diabetes (T2D) has been increasing steadily worldwide. It is estimated that by 2045 more than 800 million people will be suffering from diabetes. Despite the advancements in modern medicine, more effective therapies for treating obesity and T2D are needed. G protein-coupled receptors (GPCRs) have emerged as important drug targets for various chronic diseases, including obesity, T2D, and liver diseases. During the past two decades, many laboratories worldwide focused on understanding the role of GPCR signaling in regulating glucose metabolism and energy homeostasis. The information gained from these studies can guide the development of novel therapeutic agents. In this review, we summarize recent studies providing insights into the role of GPCR signaling in peripheral, metabolically important tissues such as pancreas, liver, skeletal muscle, and adipose tissue, focusing primarily on the use of mutant animal models and human data.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Resistencia a la Insulina/genética , Obesidad/genética , Receptores Acoplados a Proteínas G/genética , Tejido Adiposo/metabolismo , Diabetes Mellitus Tipo 2/patología , Homeostasis/genética , Humanos , Hígado/metabolismo , Obesidad/patología , Páncreas/metabolismo , Transducción de Señal/genética
13.
Biochem Pharmacol ; 192: 114739, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34418353

RESUMEN

Hepatic insulin resistance (IR) and enhanced hepatic glucose production (HGP) are key features of type 2 diabetes (T2D), contributing to fasting hyperglycemia. Adenosine receptors (ARs) are G protein-coupled and expressed in hepatocytes. Here, we explored the role of hepatic Gi/o-coupled A1AR on insulin resistance and glucose fluxes associated with obesity. We generated a mouse model with hepatocyte-specific deletion of A1AR (A1LΔ/Δ), which was compared with whole body knockout of A1AR or A1AR/A3AR (both Gi-coupled). Selective deletion of hepatic A1AR resulted in a modest improvement in insulin sensitivity. In addition, HFD A1LΔ/Δ mice showed decreased fasting glucose levels. Hyperinsulinemic-euglycemic clamp studies demonstrated enhanced insulin sensitivity with no change in HGP in HFD A1LΔ/Δ mice. Similar to A1LΔ/Δ, fasting blood glucose levels were significantly reduced in whole body A1Δ/Δ and A1Δ/ΔA3Δ/Δ compared to wild-type mice. Taken together, our data support the concept that blocking hepatic A1AR may decrease fasting blood glucose levels without directly affecting hepatocyte glucose metabolism and insulin sensitivity.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Glucosa/metabolismo , Hepatocitos/metabolismo , Resistencia a la Insulina/fisiología , Receptor de Adenosina A1/deficiencia , Animales , Diabetes Mellitus Experimental/genética , Dieta Alta en Grasa/efectos adversos , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Adenosina A1/genética
14.
Nat Commun ; 12(1): 3385, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099679

RESUMEN

Obesity is the key driver of peripheral insulin resistance, one of the key features of type 2 diabetes (T2D). In insulin-resistant individuals, the expansion of beta-cell mass is able to delay or even prevent the onset of overt T2D. Here, we report that beta-arrestin-1 (barr1), an intracellular protein known to regulate signaling through G protein-coupled receptors, is essential for beta-cell replication and function in insulin-resistant mice maintained on an obesogenic diet. Specifically, insulin-resistant beta-cell-specific barr1 knockout mice display marked reductions in beta-cell mass and the rate of beta-cell proliferation, associated with pronounced impairments in glucose homeostasis. Mechanistic studies suggest that the observed metabolic deficits are due to reduced Pdx1 expression levels caused by beta-cell barr1 deficiency. These findings indicate that strategies aimed at enhancing barr1 activity and/or expression in beta-cells may prove useful to restore proper glucose homeostasis in T2D.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/patología , Obesidad/metabolismo , beta-Arrestina 1/metabolismo , Animales , Glucemia/metabolismo , Proliferación Celular , Diabetes Mellitus Tipo 2/patología , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Proteínas de Homeodominio/metabolismo , Humanos , Resistencia a la Insulina , Masculino , Ratones , Ratones Noqueados , Obesidad/etiología , Obesidad/patología , Transactivadores/metabolismo , beta-Arrestina 1/genética
15.
JCI Insight ; 6(24)2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34752420

RESUMEN

Glucagon, a hormone released from pancreatic α cells, plays a key role in maintaining euglycemia. New insights into the signaling pathways that control glucagon secretion may stimulate the development of novel therapeutic agents. In this study, we investigated the potential regulation of α cell function by G proteins of the Gq family. The use of a chemogenetic strategy allowed us to selectively activate Gq signaling in mouse α cells in vitro and in vivo. Acute stimulation of α cell Gq signaling led to elevated plasma glucagon levels, accompanied by increased insulin release and improved glucose tolerance. Moreover, chronic activation of this pathway greatly improved glucose tolerance in obese mice. We also identified an endogenous Gq-coupled receptor (vasopressin 1b receptor; V1bR) that was enriched in mouse and human α cells. Agonist-induced activation of the V1bR strongly stimulated glucagon release in a Gq-dependent fashion. In vivo studies indicated that V1bR-mediated glucagon release played a key role in the counterregulatory hyperglucagonemia under hypoglycemic and glucopenic conditions. These data indicate that α cell Gq signaling represents an important regulator of glucagon secretion, resulting in multiple beneficial metabolic effects. Thus, drugs that target α cell-enriched Gq-coupled receptors may prove useful to restore euglycemia in various pathophysiological conditions.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Hipoglucemiantes/metabolismo , Transducción de Señal/inmunología , Animales , Humanos , Masculino , Ratones
16.
Nat Commun ; 11(1): 2995, 2020 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-32532984

RESUMEN

Adipocyte dysfunction links obesity to insulin resistance and type 2 diabetes. Adipocyte function is regulated by receptor-mediated activation of heterotrimeric G proteins. Little is known about the potential in vivo metabolic roles of Gi-type G proteins expressed by adipocytes, primarily due to the lack of suitable animal models. To address this question, we generated mice lacking functional Gi proteins selectively in adipocytes. Here we report that these mutant mice displayed significantly impaired glucose tolerance and reduced insulin sensitivity when maintained on an obesogenic diet. In contrast, using a chemogenetic strategy, we demonstrated that activation of Gi signaling selectively in adipocytes greatly improved glucose homeostasis and insulin signaling. We also elucidated the cellular mechanisms underlying the observed metabolic phenotypes. Our data support the concept that adipocyte Gi signaling is essential for maintaining euglycemia. Drug-mediated activation of adipocyte Gi signaling may prove beneficial for restoring proper glucose homeostasis in type 2 diabetes.


Asunto(s)
Adipocitos/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Resistencia a la Insulina/genética , Transducción de Señal/genética , Adipocitos/citología , Animales , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Perfilación de la Expresión Génica/métodos , Intolerancia a la Glucosa/genética , Homeostasis/genética , Insulina/sangre , Insulina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Obesidad/sangre , Obesidad/genética , Obesidad/metabolismo
17.
Nat Commun ; 11(1): 1950, 2020 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-32327658

RESUMEN

BDNF signaling in hypothalamic circuitries regulates mammalian food intake. However, whether BDNF exerts metabolic effects on peripheral organs is currently unknown. Here, we show that the BDNF receptor TrkB.T1 is expressed by pancreatic ß-cells where it regulates insulin release. Mice lacking TrkB.T1 show impaired glucose tolerance and insulin secretion. ß-cell BDNF-TrkB.T1 signaling triggers calcium release from intracellular stores, increasing glucose-induced insulin secretion. Additionally, BDNF is secreted by skeletal muscle and muscle-specific BDNF knockout phenocopies the ß-cell TrkB.T1 deletion metabolic impairments. The finding that BDNF is also secreted by differentiated human muscle cells and induces insulin secretion in human islets via TrkB.T1 identifies a new regulatory function of BDNF on metabolism that is independent of CNS activity. Our data suggest that muscle-derived BDNF may be a key factor mediating increased glucose metabolism in response to exercise, with implications for the treatment of diabetes and related metabolic diseases.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Calcio/metabolismo , Células Cultivadas , Glucosa/metabolismo , Intolerancia a la Glucosa , Humanos , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Noqueados , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptor trkB/química , Receptor trkB/genética , Receptor trkB/metabolismo , Transducción de Señal
18.
Sci Adv ; 6(23): eaba1733, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32548266

RESUMEN

A better understanding of the signaling pathways regulating adipocyte function is required for the development of new classes of antidiabetic/obesity drugs. We here report that mice lacking ß-arrestin-1 (barr1), a cytoplasmic and nuclear signaling protein, selectively in adipocytes showed greatly impaired glucose tolerance and insulin sensitivity when consuming an obesogenic diet. In contrast, transgenic mice overexpressing barr1 in adipocytes were protected against the metabolic deficits caused by a high-calorie diet. Barr1 deficiency led to a myogenic reprogramming of brown adipose tissue (BAT), causing elevated plasma myostatin (Mstn) levels, which in turn led to impaired insulin signaling in multiple peripheral tissues. Additional in vivo studies indicated that barr1-mediated suppression of Mstn expression by BAT is required for maintaining euglycemia. These findings convincingly identify barr1 as a critical regulator of BAT function. Strategies aimed at enhancing barr1 activity in BAT may prove beneficial for the treatment of type 2 diabetes.

19.
Sci Adv ; 6(23): eaaz1341, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32537493

RESUMEN

ß-Arrestin-1 and ß-arrestin-2 have emerged as important signaling molecules that modulate glucose fluxes in several peripheral tissues. The potential roles of neuronally expressed ß-arrestins in regulating glucose homeostasis remain unknown. We here report that mice lacking ß-arrestin-1 (barr1) selectively in AgRP neurons displayed impaired glucose tolerance and insulin sensitivity when consuming an obesogenic diet, while mice overexpressing barr1 selectively in AgRP neurons were protected against obesity-associated metabolic impairments. Additional physiological, biochemical, and electrophysiological data indicated that the presence of barr1 is essential for insulin-mediated hyperpolarization of AgRP neurons. As a result, barr1 expressed by AgRP neurons regulates efferent neuronal pathways that suppress hepatic glucose production and promote lipolysis in adipose tissue. Mice lacking ß-arrestin-2 (barr2) selectively in AgRP neurons showed no substantial metabolic phenotypes. Our data suggest that agents able to enhance the activity of barr1 in AgRP neurons may prove beneficial as antidiabetic drugs.

20.
J Clin Invest ; 129(9): 3732-3737, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31184597

RESUMEN

Beta-arrestin-1 and -2 (Barr1 and Barr2, respectively) are intracellular signaling molecules that regulate many important metabolic functions. We previously demonstrated that mice lacking Barr2 selectively in pancreatic beta-cells showed pronounced metabolic impairments. Here we investigated whether Barr1 plays a similar role in regulating beta-cell function and whole body glucose homeostasis. Initially, we inactivated the Barr1 gene in beta-cells of adult mice (beta-barr1-KO mice). Beta-barr1-KO mice did not display any obvious phenotypes in a series of in vivo and in vitro metabolic tests. However, glibenclamide and tolbutamide, two widely used antidiabetic drugs of the sulfonylurea (SU) family, showed greatly reduced efficacy in stimulating insulin secretion in the KO mice in vivo and in perifused KO islets in vitro. Additional in vivo and in vitro studies demonstrated that Barr1 enhanced SU-stimulated insulin secretion by promoting SU-mediated activation of Epac2. Pull-down and co-immunoprecipitation experiments showed that Barr1 can directly interact with Epac2 and that SUs such as glibenclamide promote Barr1/Epac2 complex formation, triggering enhanced Rap1 signaling and insulin secretion. These findings suggest that strategies aimed at promoting Barr1 signaling in beta-cells may prove useful for the development of efficacious antidiabetic drugs.


Asunto(s)
Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Compuestos de Sulfonilurea/química , beta-Arrestina 1/metabolismo , Animales , Genotipo , Gliburida/farmacología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Hipoglucemiantes/farmacología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Fenotipo , Transducción de Señal , Tolbutamida/farmacología , Arrestina beta 2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA