Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Antioxidants (Basel) ; 13(4)2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38671844

RESUMEN

Chronic hyperglycemia induces intrarenal oxidative stress due to the excessive production of reactive oxygen species (ROS), leading to a cascade of events that contribute to the development and progression of diabetic kidney disease (DKD). NOX5, a pro-oxidant NADPH oxidase isoform, has been identified as a significant contributor to renal ROS in humans. Elevated levels of renal ROS contribute to endothelial cell dysfunction and associated inflammation, causing increased endothelial permeability, which can disrupt the renal ecosystem, leading to progressive albuminuria and renal fibrosis in DKD. This study specifically examines the contribution of endothelial cell-specific human NOX5 expression in renal pathology in a transgenic mouse model of DKD. This study additionally compares NOX5 with the previously characterized NADPH oxidase, NOX4, in terms of their relative roles in DKD. Regardless of NOX4 pathway, this study found that endothelial cell-specific expression of NOX5 exacerbates renal injury, albuminuria and fibrosis. This is attributed to the activation of the endothelial mesenchymal transition (EMT) pathway via enhanced ROS formation and the modulation of redox-sensitive factors. These findings underscore the potential therapeutic significance of NOX5 inhibition in human DKD. The study proposes that inhibiting NOX5 could be a promising approach for mitigating the progression of DKD and strengthens the case for the development of NOX5-specific inhibitors as a potential therapeutic intervention.

2.
PLoS One ; 18(7): e0281487, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37418389

RESUMEN

Telomerase reverse transcriptase (TERT) promoter mutations occur frequently in cancer, have been associated with increased TERT expression and cell proliferation, and could potentially influence therapeutic regimens for melanoma. As the role of TERT expression in malignant melanoma and the non-canonical functions of TERT remain understudied, we aimed to extend the current knowledge on the impact of TERT promoter mutations and expression alterations in tumor progression by analyzing several highly annotated melanoma cohorts. Using multivariate models, we found no consistent association for TERT promoter mutations or TERT expression with the survival rate in melanoma cohorts under immune checkpoint inhibition. However, the presence of CD4+ T cells increased with TERT expression and correlated with the expression of exhaustion markers. While the frequency of promoter mutations did not change with Breslow thickness, TERT expression was increased in metastases arising from thinner primaries. As single-cell RNA-sequencing (RNA-seq) showed that TERT expression was associated with genes involved in cell migration and dynamics of the extracellular matrix, this suggests a role of TERT during invasion and metastasis. Co-regulated genes found in several bulk tumors and single-cell RNA-seq cohorts also indicated non-canonical functions of TERT related to mitochondrial DNA stability and nuclear DNA repair. This pattern was also evident in glioblastoma and across other entities. Hence, our study adds to the role of TERT expression in cancer metastasis and potentially also immune resistance.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Telomerasa , Humanos , Linfocitos T CD4-Positivos/patología , Melanoma/genética , Melanoma/patología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Regiones Promotoras Genéticas , Mutación , Reparación del ADN/genética , Telomerasa/genética
4.
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
5.
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
6.
Am J Physiol Endocrinol Metab ; 322(1): E63-E73, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34866401

RESUMEN

In mice, exercise is suggested to activate the mechanistic target of rapamycin complex 2 (mTORC2) in skeletal muscle, and mTORC2 is required for normal muscle glucose uptake during exercise. Whether this translates to human skeletal muscle and what signaling pathways facilitate the exercise-induced mTORC2 activation is unknown. We herein tested the hypothesis that exercise increases mTORC2 activity in human skeletal muscle and investigated if ß2-adrenergic receptor (AR) activation mediates exercise-induced mTORC2 activation. We examined several mTORC2 activity readouts (p-NDRG1 Thr346, p-Akt Ser473, p-mTOR S2481, and p-Akt Thr450) in human skeletal muscle biopsies after uphill walking or cycling exercise. In mouse muscles, we assessed mTORC2 activity readouts following acute activation of muscle ß2-adrenergic or GS signaling and during in vivo and ex vivo muscle contractions. Exercise increased phosphorylation of NDRG1 Thr346 in human soleus, gastrocnemius, and vastus lateralis muscle, without changing p-Akt Ser473, p-Akt Thr450, and p-mTOR Ser2481. In mouse muscle, stimulation of ß2-adrenergic or GS signaling and ex vivo contractions failed to increase p-NDRG1 Thr346, whereas in vivo contractions were sufficient to induce p-NDRG1 Thr346. In conclusion, the mTORC2 activity readout p-NDRG1 Thr346 is a novel exercise-responsive signaling protein in human skeletal muscle. Notably, contraction-induced p-NDRG1 Thr346 appears to require a systemic factor. Unlike exercise, and in contrast to published data obtained in cultured muscles cells, stimulation of ß2-adrenergic signaling is not sufficient to trigger NDRG1 phosphorylation in mature mouse skeletal muscle.NEW & NOTEWORTHY The mTORC2 readout p-NDRG Thr346 is a novel exercise-responsive protein in human skeletal muscle. ß2-AR and GS signaling are not sufficient to induce mTORC2 signaling in adult muscle. In vivo, but not ex vivo, contraction induced p-NDRG Thr346, which indicates requirement of a systemic factor for exercise-induced mTORC2 activation.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Músculo Esquelético/metabolismo , Transducción de Señal/fisiología , Caminata/fisiología , Adulto , Animales , Células Cultivadas , Femenino , Fibroblastos/metabolismo , Voluntarios Sanos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Contracción Muscular/fisiología , Fosforilación/fisiología , Receptores Adrenérgicos beta 2/metabolismo , Adulto Joven
7.
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
8.
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
9.
ACS Pharmacol Transl Sci ; 4(3): 1175-1187, 2021 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-34151207

RESUMEN

Seven-transmembrane receptors signal via G-protein- and ß-arrestin-dependent pathways. We describe a peripheral CB1R antagonist (MRI-1891) highly biased toward inhibiting CB1R-induced ß-arrestin-2 (ßArr2) recruitment over G-protein activation. In obese wild-type and ßArr2-knockout (KO) mice, MRI-1891 treatment reduces food intake and body weight without eliciting anxiety even at a high dose causing partial brain CB1R occupancy. By contrast, the unbiased global CB1R antagonist rimonabant elicits anxiety in both strains, indicating no ßArr2 involvement. Interestingly, obesity-induced muscle insulin resistance is improved by MRI-1891 in wild-type but not in ßArr2-KO mice. In C2C12 myoblasts, CB1R activation suppresses insulin-induced akt-2 phosphorylation, preventable by MRI-1891, ßArr2 knockdown or overexpression of CB1R-interacting protein. MRI-1891, but not rimonabant, interacts with nonpolar residues on the N-terminal loop, including F108, and on transmembrane helix-1, including S123, a combination that facilitates ßArr2 bias. Thus, CB1R promotes muscle insulin resistance via ßArr2 signaling, selectively mitigated by a biased CB1R antagonist at reduced risk of central nervous system (CNS) side effects.

10.
J Neurochem ; 158(3): 603-620, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33540469

RESUMEN

DREADDs (Designer Receptors Exclusively Activated by a Designer Drug) are designer G protein-coupled receptors (GPCRs) that are widely used in the neuroscience field to modulate neuronal activity. In this review, we will focus on DREADD studies carried out with genetically engineered mice aimed at elucidating signaling pathways important for maintaining proper glucose and energy homeostasis. The availability of muscarinic receptor-based DREADDs endowed with selectivity for one of the four major classes of heterotrimeric G proteins (Gs , Gi , Gq , and G12 ) has been instrumental in dissecting the physiological and pathophysiological roles of distinct G protein signaling pathways in metabolically important cell types. The novel insights gained from this work should inform the development of novel classes of drugs useful for the treatment of several metabolic disorders including type 2 diabetes and obesity.


Asunto(s)
Drogas de Diseño/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Drogas de Diseño/farmacología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Transducción de Señal/efectos de los fármacos
11.
Annu Rev Pharmacol Toxicol ; 61: 421-440, 2021 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-32746768

RESUMEN

G protein-coupled receptors (GPCRs) form a superfamily of plasma membrane receptors that couple to four major families of heterotrimeric G proteins, Gs, Gi, Gq, and G12. GPCRs represent excellent targets for drug therapy. Since the individual GPCRs are expressed by many different cell types, the in vivo metabolic roles of a specific GPCR expressed by a distinct cell type are not well understood. The development of designer GPCRs known as DREADDs (designer receptors exclusively activated by a designer drug) that selectively couple to distinct classes of heterotrimeric G proteins has greatly facilitated studies in this area. This review focuses on the use of DREADD technology to explore the physiological and pathophysiological roles of distinct GPCR/G protein cascades in several metabolically important cell types. The novel insights gained from these studies should stimulate the development of GPCR-based treatments for major metabolic diseases such as type 2 diabetes and obesity.


Asunto(s)
Diabetes Mellitus Tipo 2 , Transducción de Señal , Humanos , Hipoglucemiantes , Receptores Acoplados a Proteínas G , Tecnología
12.
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
13.
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.

14.
PLoS Genet ; 15(10): e1008424, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31622341

RESUMEN

Type 2 diabetes (T2D) has become a major health problem worldwide. Skeletal muscle (SKM) is the key tissue for whole-body glucose disposal and utilization. New drugs aimed at improving insulin sensitivity of SKM would greatly expand available therapeutic options. ß-arrestin-1 and -2 (Barr1 and Barr2, respectively) are two intracellular proteins best known for their ability to mediate the desensitization and internalization of G protein-coupled receptors (GPCRs). Recent studies suggest that Barr1 and Barr2 regulate several important metabolic functions including insulin release and hepatic glucose production. Since SKM expresses many GPCRs, including the metabolically important ß2-adrenergic receptor, the goal of this study was to examine the potential roles of Barr1 and Barr2 in regulating SKM and whole-body glucose metabolism. Using SKM-specific knockout (KO) mouse lines, we showed that the loss of SKM Barr2, but not of SKM Barr1, resulted in mild improvements in glucose tolerance in diet-induced obese mice. SKM-specific Barr1- and Barr2-KO mice did not show any significant differences in exercise performance. However, lack of SKM Barr2 led to increased glycogen breakdown following a treadmill exercise challenge. Interestingly, mice that lacked both Barr1 and Barr2 in SKM showed no significant metabolic phenotypes. Thus, somewhat surprisingly, our data indicate that SKM ß-arrestins play only rather subtle roles (SKM Barr2) in regulating whole-body glucose homeostasis and SKM insulin sensitivity.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Músculo Esquelético/metabolismo , Obesidad/metabolismo , beta-Arrestina 1/metabolismo , Arrestina beta 2/metabolismo , Animales , Diabetes Mellitus Tipo 2/etiología , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Glucosa/administración & dosificación , Glucosa/metabolismo , Técnica de Clampeo de la Glucosa , Glucógeno/metabolismo , Humanos , Insulina/metabolismo , Resistencia a la Insulina , Masculino , Ratones , Ratones Noqueados , Obesidad/etiología , Transducción de Señal/genética , beta-Arrestina 1/genética , Arrestina beta 2/genética
15.
Mol Metab ; 27: 83-91, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31272886

RESUMEN

OBJECTIVE: Given the worldwide epidemics of obesity and type 2 diabetes, novel antidiabetic and appetite-suppressing drugs are urgently needed. Adipocytes play a central role in the regulation of whole-body glucose and energy homeostasis. The goal of this study was to examine the metabolic effects of acute and chronic activation of Gs signaling selectively in adipocytes (activated Gs stimulates cAMP production), both in lean and obese mice. METHODS: To address this question, we generated a novel mutant mouse strain (adipo-GsD mice) that expressed a Gs-coupled designer G protein-coupled receptor (Gs DREADD or short GsD) selectively in adipocytes. Importantly, the GsD receptor can only be activated by administration of an exogenous agent (CNO) that is otherwise pharmacologically inert. The adipo-GsD mice were maintained on either regular chow or a high-fat diet and then subjected to a comprehensive series of metabolic tests. RESULTS: Pharmacological (CNO) activation of the GsD receptor in adipocytes of adipo-GsD mice caused profound improvements in glucose homeostasis and protected mice against the metabolic deficits associated with the consumption of a calorie-rich diet. Moreover, chronic activation of Gs signaling in adipocytes led to a striking increase in energy expenditure and reduced food intake, resulting in a decrease in body weight and fat mass when mice consumed a calorie-rich diet. CONCLUSION: Systematic studies with a newly developed mouse model enabled us to assess the metabolic consequences caused by acute or chronic activation of Gs signaling selectively in adipocytes. Most strikingly, chronic activation of this pathway led to reduced body fat mass and restored normal glucose homeostasis in obese mice. These findings are of considerable relevance for the development of novel antidiabetic and anti-obesity drugs.


Asunto(s)
Adipocitos/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo
16.
Diabetes ; 68(6): 1341-1352, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30936140

RESUMEN

Skeletal muscle (SKM) insulin resistance plays a central role in the pathogenesis of type 2 diabetes. Because G-protein-coupled receptors (GPCRs) represent excellent drug targets, we hypothesized that activation of specific functional classes of SKM GPCRs might lead to improved glucose homeostasis in type 2 diabetes. At present, little is known about the in vivo metabolic roles of the various distinct GPCR signaling pathways operative in SKM. In this study, we tested the hypothesis that selective activation of SKM Gq signaling can improve SKM glucose uptake and whole-body glucose homeostasis under physiological and pathophysiological conditions. Studies with transgenic mice expressing a Gq-linked designer GPCR selectively in SKM cells demonstrated that receptor-mediated activation of SKM Gq signaling greatly promoted glucose uptake into SKM and significantly improved glucose homeostasis in obese, glucose-intolerant mice. These beneficial metabolic effects required the activity of SKM AMPK. In contrast, obese mutant mice that lacked both Gαq and Gα11 selectively in SKM showed severe deficits in glucose homeostasis. Moreover, GPCR-mediated activation of Gq signaling also stimulated glucose uptake in primary human SKM cells. Taken together, these findings strongly suggest that agents capable of enhancing SKM Gq signaling may prove useful as novel antidiabetic drugs.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Intolerancia a la Glucosa/metabolismo , Glucosa/metabolismo , Resistencia a la Insulina , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adenilato Quinasa/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Ratones , Ratones Obesos , Ratones Transgénicos , Mioblastos Esqueléticos , Transducción de Señal
17.
Pharmacol Ther ; 189: 71-88, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29684466

RESUMEN

Research on GPR34, which was discovered in 1999 as an orphan G protein-coupled receptor of the rhodopsin-like class, disclosed its physiologic relevance only piece by piece. Being present in all recent vertebrate genomes analyzed so far it seems to improve the fitness of species although it is not essential for life and reproduction as GPR34-deficient mice demonstrate. However, closer inspection of macrophages and microglia, where it is mainly expressed, revealed its relevance in immune cell function. Recent data clearly demonstrate that GPR34 function is required to arrest microglia in the M0 homeostatic non-phagocytic phenotype. Herein, we summarize the current knowledge on its evolution, genomic and structural organization, physiology, pharmacology and relevance in human diseases including neurodegenerative diseases and cancer, which accumulated over the last 20 years.


Asunto(s)
Receptores Lisofosfolípidos/fisiología , Animales , Evolución Molecular , Genómica , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Fenotipo , Receptores Lisofosfolípidos/química , Receptores Lisofosfolípidos/historia
19.
J Biol Chem ; 291(15): 7809-20, 2016 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-26851281

RESUMEN

Designerreceptorsexclusivelyactivated by adesignerdrug (DREADDs) are clozapine-N-oxide-sensitive designer G protein-coupled receptors (GPCRs) that have emerged as powerful novel chemogenetic tools to study the physiological relevance of GPCR signaling pathways in specific cell types or tissues. Like endogenous GPCRs, clozapine-N-oxide-activated DREADDs do not only activate heterotrimeric G proteins but can also trigger ß-arrestin-dependent (G protein-independent) signaling. To dissect the relative physiological relevance of G protein-mediatedversusß-arrestin-mediated signaling in different cell types or physiological processes, the availability of G protein- and ß-arrestin-biased DREADDs would be highly desirable. In this study, we report the development of a mutationally modified version of a non-biased DREADD derived from the M3muscarinic receptor that can activate Gq/11with high efficacy but lacks the ability to interact with ß-arrestins. We also demonstrate that this novel DREADD is activein vivoand that cell type-selective expression of this new designer receptor can provide novel insights into the physiological roles of G protein (Gq/11)-dependentversusß-arrestin-dependent signaling in hepatocytes. Thus, this novel Gq/11-biased DREADD represents a powerful new tool to study the physiological relevance of Gq/11-dependent signaling in distinct tissues and cell types, in the absence of ß-arrestin-mediated cellular effects. Such studies should guide the development of novel classes of functionally biased ligands that show high efficacy in various pathophysiological conditions but display a reduced incidence of side effects.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Hepatocitos/metabolismo , Mapeo de Interacción de Proteínas , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Animales , Arrestinas/metabolismo , Células COS , Calcio/metabolismo , Células Cultivadas , Chlorocebus aethiops , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Mapeo de Interacción de Proteínas/métodos , Mapas de Interacción de Proteínas , beta-Arrestinas
20.
Nat Commun ; 7: 10268, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26743492

RESUMEN

Agouti-related peptide (AgRP) neurons of the hypothalamus play a key role in regulating food intake and body weight, by releasing three different orexigenic molecules: AgRP; GABA; and neuropeptide Y. AgRP neurons express various G protein-coupled receptors (GPCRs) with different coupling properties, including Gs-linked GPCRs. At present, the potential role of Gs-coupled GPCRs in regulating the activity of AgRP neurons remains unknown. Here we show that the activation of Gs-coupled receptors expressed by AgRP neurons leads to a robust and sustained increase in food intake. We also provide detailed mechanistic data linking the stimulation of this class of receptors to the observed feeding phenotype. Moreover, we show that this pathway is clearly distinct from other GPCR signalling cascades that are operative in AgRP neurons. Our data suggest that drugs able to inhibit this signalling pathway may become useful for the treatment of obesity.


Asunto(s)
Proteína Relacionada con Agouti/genética , Ingestión de Alimentos/genética , Hipotálamo/metabolismo , Neuronas/metabolismo , Neuropéptido Y/genética , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Células HEK293 , Humanos , Hipotálamo/citología , Inmunohistoquímica , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Neuropéptido Y/metabolismo , Técnicas de Placa-Clamp , Fosfoproteínas , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transducción de Señal , Ácido gamma-Aminobutírico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...