Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cell ; 184(13): 3502-3518.e33, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34048700

RESUMEN

Thermogenic adipocytes possess a therapeutically appealing, energy-expending capacity, which is canonically cold-induced by ligand-dependent activation of ß-adrenergic G protein-coupled receptors (GPCRs). Here, we uncover an alternate paradigm of GPCR-mediated adipose thermogenesis through the constitutively active receptor, GPR3. We show that the N terminus of GPR3 confers intrinsic signaling activity, resulting in continuous Gs-coupling and cAMP production without an exogenous ligand. Thus, transcriptional induction of Gpr3 represents the regulatory parallel to ligand-binding of conventional GPCRs. Consequently, increasing Gpr3 expression in thermogenic adipocytes is alone sufficient to drive energy expenditure and counteract metabolic disease in mice. Gpr3 transcription is cold-stimulated by a lipolytic signal, and dietary fat potentiates GPR3-dependent thermogenesis to amplify the response to caloric excess. Moreover, we find GPR3 to be an essential, adrenergic-independent regulator of human brown adipocytes. Taken together, our findings reveal a noncanonical mechanism of GPCR control and thermogenic activation through the lipolysis-induced expression of constitutively active GPR3.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Receptor de Androstano Constitutivo/metabolismo , Lipólisis , Receptores Acoplados a Proteínas G/metabolismo , Termogénesis , Adipocitos/metabolismo , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Frío , Grasas de la Dieta/farmacología , Humanos , Ratones Endogámicos C57BL , Fenotipo , Receptores Acoplados a Proteínas G/genética , Transducción de Señal , Sistema Nervioso Simpático/metabolismo , Transcripción Genética
2.
Nature ; 609(7926): 361-368, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35790189

RESUMEN

Brown adipose tissue (BAT) dissipates energy1,2 and promotes cardiometabolic health3. Loss of BAT during obesity and ageing is a principal hurdle for BAT-centred obesity therapies, but not much is known about BAT apoptosis. Here, untargeted metabolomics demonstrated that apoptotic brown adipocytes release a specific pattern of metabolites with purine metabolites being highly enriched. This apoptotic secretome enhances expression of the thermogenic programme in healthy adipocytes. This effect is mediated by the purine inosine that stimulates energy expenditure in brown adipocytes by the cyclic adenosine monophosphate-protein kinase A signalling pathway. Treatment of mice with inosine increased BAT-dependent energy expenditure and induced 'browning' of white adipose tissue. Mechanistically, the equilibrative nucleoside transporter 1 (ENT1, SLC29A1) regulates inosine levels in BAT: ENT1-deficiency increases extracellular inosine levels and consequently enhances thermogenic adipocyte differentiation. In mice, pharmacological inhibition of ENT1 as well as global and adipose-specific ablation enhanced BAT activity and counteracted diet-induced obesity, respectively. In human brown adipocytes, knockdown or blockade of ENT1 increased extracellular inosine, which enhanced thermogenic capacity. Conversely, high ENT1 levels correlated with lower expression of the thermogenic marker UCP1 in human adipose tissues. Finally, the Ile216Thr loss of function mutation in human ENT1 was associated with significantly lower body mass index and 59% lower odds of obesity for individuals carrying the Thr variant. Our data identify inosine as a metabolite released during apoptosis with a 'replace me' signalling function that regulates thermogenic fat and counteracts obesity.


Asunto(s)
Adipocitos Marrones , Tejido Adiposo Pardo , Metabolismo Energético , Inosina , Adipocitos Marrones/efectos de los fármacos , Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Animales , Metabolismo Energético/efectos de los fármacos , Tranportador Equilibrativo 1 de Nucleósido/antagonistas & inhibidores , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Humanos , Inosina/metabolismo , Inosina/farmacología , Ratones , Obesidad/genética , Obesidad/metabolismo , Termogénesis/genética , Proteína Desacopladora 1/metabolismo
3.
Liver Int ; 40(9): 2228-2241, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32627946

RESUMEN

BACKGROUND & AIMS: Phosphodiesterase-5 inhibitors (PDE-5-I) are used for treatment of erectile dysfunction (ED), which is common in patients with cirrhosis. They may improve portal hypertension (PH), but contradictory data on efficacy and side-effects have been reported. Non-selective beta blockers (NSBB) reduce portal pressure, but might aggravate ED. Thus, we evaluated the combination of PDE-5-I with NSBB and its impact on PH and ED in experimental cirrhosis. METHODS: ED was assessed in cirrhotic patients (n = 86) using standardized questionnaire. Experimental cirrhosis was induced by bile-duct-ligation or carbon-tetrachloride intoxication in rats. Corpus cavernosum pressure - a surrogate of ED -, as well as systemic and portal haemodynamics, were measured in vivo and in situ after acute administration of udenafil alone or in combination with propranolol. mRNA and protein levels of PDE-5 signalling were analysed using PCR and western Blot. RESULTS: ED in humans was related to severity of liver disease and to NSBB treatment. PDE-5 was mainly expressed in hepatic stellate cells and upregulated in human and experimental cirrhosis. Propranolol reduced corpus cavernosum pressure in cirrhotic rats and it was restored by udenafil. Even though udenafil treatment improved PH, it led to a reduction of mean arterial pressure. The combination of udenafil and propranolol reduced portal pressure and hepatic resistance without systemic side-effects. CONCLUSIONS: ED is common with advanced cirrhosis and concomitant NSBB treatment. The combination of PDE-5-I and NSBB improves ED and PH in experimental cirrhosis.


Asunto(s)
Disfunción Eréctil , Hipertensión Portal , Cirrosis Hepática Experimental , Animales , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5 , Disfunción Eréctil/tratamiento farmacológico , Disfunción Eréctil/etiología , Humanos , Hipertensión Portal/tratamiento farmacológico , Masculino , Inhibidores de Fosfodiesterasa 5 , Presión Portal , Ratas , Ratas Sprague-Dawley
4.
Nature ; 516(7531): 395-9, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25317558

RESUMEN

Brown adipose tissue (BAT) is specialized in energy expenditure, making it a potential target for anti-obesity therapies. Following exposure to cold, BAT is activated by the sympathetic nervous system with concomitant release of catecholamines and activation of ß-adrenergic receptors. Because BAT therapies based on cold exposure or ß-adrenergic agonists are clinically not feasible, alternative strategies must be explored. Purinergic co-transmission might be involved in sympathetic control of BAT and previous studies reported inhibitory effects of the purinergic transmitter adenosine in BAT from hamster or rat. However, the role of adenosine in human BAT is unknown. Here we show that adenosine activates human and murine brown adipocytes at low nanomolar concentrations. Adenosine is released in BAT during stimulation of sympathetic nerves as well as from brown adipocytes. The adenosine A2A receptor is the most abundant adenosine receptor in human and murine BAT. Pharmacological blockade or genetic loss of A2A receptors in mice causes a decrease in BAT-dependent thermogenesis, whereas treatment with A2A agonists significantly increases energy expenditure. Moreover, pharmacological stimulation of A2A receptors or injection of lentiviral vectors expressing the A2A receptor into white fat induces brown-like cells-so-called beige adipocytes. Importantly, mice fed a high-fat diet and treated with an A2A agonist are leaner with improved glucose tolerance. Taken together, our results demonstrate that adenosine-A2A signalling plays an unexpected physiological role in sympathetic BAT activation and protects mice from diet-induced obesity. Those findings reveal new possibilities for developing novel obesity therapies.


Asunto(s)
Adenosina/metabolismo , Adipocitos/metabolismo , Tejido Adiposo Pardo/metabolismo , Receptor de Adenosina A2A/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacología , Agonistas del Receptor de Adenosina A2/farmacología , Tejido Adiposo Pardo/efectos de los fármacos , Animales , Células Cultivadas , Cricetinae , Dieta , Humanos , Masculino , Mesocricetus , Ratones , Ratones Endogámicos C57BL , Fenetilaminas/farmacología
5.
Glia ; 67(12): 2329-2342, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31328322

RESUMEN

Adenosine A2A receptors (A2A R) are modulators of various physiological processes essential for brain homeostasis and fine synaptic tuning. In certain neurodegenerative conditions, notably Alzheimer's disease (AD), A2A Rs are pathologically upregulated in neurons but also in astrocytes. In that context, the use of A2A Rs inhibitors, normalizing impaired receptor function, is seen as a potential therapeutic strategy. However, the impact of A2A R alterations, particularly in astrocytes, is not fully understood. Here, we investigated the effect of A2A R overexpression on transcriptional deregulation in primary astrocytic cultures. By performing whole transcriptome analysis, we found that A2A R overexpression promotes robust transcriptional changes, mostly affecting immune response, angiogenesis, and cell activation-related genes. Importantly, we observed that treatment with SCH58261, a selective A2A R antagonist, restored the expression levels of several inflammatory and astrocytic activation-related genes, such as Interleukin-1beta and vimentin. This supports the notion that A2A R blockade could restore some astrocytic dysfunctions associated with abnormal A2A R expression, further arguing for a potential beneficial impact of receptor antagonists in A2A R-induced transcriptional deregulation, inflammation, and astrogliosis. Overall, our findings provide novel insights into the putative impact of A2A R overexpression on transcriptional deregulation in astrocytes, thereby opening novel avenues for the use of A2A R antagonists as potential therapeutic strategy in neurodegenerative diseases.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/farmacología , Astrocitos/fisiología , Receptor de Adenosina A2A/genética , Receptor de Adenosina A2A/metabolismo , Transcripción Genética/fisiología , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Células Cultivadas , Redes Reguladoras de Genes/efectos de los fármacos , Redes Reguladoras de Genes/fisiología , Ratones , Transcripción Genética/efectos de los fármacos
6.
FASEB J ; 27(4): 1621-30, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23303211

RESUMEN

With more than half a billion individuals affected worldwide, obesity has reached pandemic proportions. Development of "brown-like" or "brite" adipocytes within white adipose tissue (WAT) has potential antiobesity and insulin-sensitizing effects. We investigated the role of cyclic GMP (cGMP) signaling, focusing on cGMP-dependent protein kinase I (PKGI) in WAT. PKGI is expressed in murine WAT, primary adipocytes, and 3T3-L1. Treatment of adipocytes with cGMP resulted in increased adipogenesis, with a 54% increase in expression of peroxisome proliferator-activated receptor-γ. Lentiviral overexpression of PKGI further increased adipogenesis, whereas loss of PKGI significantly reduced adipogenic differentiation. In addition to adipogenic effects, PKGI had an antihypertrophic and anti-inflammatory effect via RhoA phosphorylation and reduction of proinflammatory adipokine expression. Moreover, PKGI induced a 4.3-fold increase in abundance of UCP-1 and the development of a brown-like thermogenic program in primary adipocytes. Notably, treatment of C57BL/6 mice with phosphodiesterase inhibitor sildenafil (12 mg/kg/d) for 7 d caused 4.6-fold increase in uncoupling protein-1 expression and promoted establishment of a brown fat cell-like phenotype ("browning") of WAT in vivo. Taken together, PKGI is a key regulator of cell size, adipokine secretion and browning of white fat depots and thus could be a valuable target in developing novel treatments for obesity.


Asunto(s)
Adipogénesis , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , GMP Cíclico/metabolismo , Células 3T3-L1/citología , Adipocitos/metabolismo , Tejido Adiposo Pardo/citología , Tejido Adiposo Blanco/citología , Animales , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Canales Iónicos/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Obesidad/metabolismo , Transducción de Señal , Proteína Desacopladora 1
7.
Nat Metab ; 6(6): 1053-1075, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38684889

RESUMEN

Promoting brown adipose tissue (BAT) activity innovatively targets obesity and metabolic disease. While thermogenic activation of BAT is well understood, the rheostatic regulation of BAT to avoid excessive energy dissipation remains ill-defined. Here, we demonstrate that adenylyl cyclase 3 (AC3) is key for BAT function. We identified a cold-inducible promoter that generates a 5' truncated AC3 mRNA isoform (Adcy3-at), whose expression is driven by a cold-induced, truncated isoform of PPARGC1A (PPARGC1A-AT). Male mice lacking Adcy3-at display increased energy expenditure and are resistant to obesity and ensuing metabolic imbalances. Mouse and human AC3-AT are retained in the endoplasmic reticulum, unable to translocate to the plasma membrane and lack enzymatic activity. AC3-AT interacts with AC3 and sequesters it in the endoplasmic reticulum, reducing the pool of adenylyl cyclases available for G-protein-mediated cAMP synthesis. Thus, AC3-AT acts as a cold-induced rheostat in BAT, limiting adverse consequences of cAMP activity during chronic BAT activation.


Asunto(s)
Adenilil Ciclasas , Tejido Adiposo Pardo , Frío , Adenilil Ciclasas/metabolismo , Adenilil Ciclasas/genética , Tejido Adiposo Pardo/metabolismo , Animales , Ratones , Masculino , Humanos , Termogénesis/genética , Metabolismo Energético , AMP Cíclico/metabolismo , Ratones Noqueados
8.
Commun Biol ; 5(1): 280, 2022 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-35351968

RESUMEN

Pharmacological activation of brown adipose tissue (BAT) is an attractive approach for increasing energy expenditure to counteract obesity. Given the side-effects of known activators of BAT, we studied inhibitors of BAT as a novel, alternative concept to regulate energy expenditure. We focused on G-protein-coupled receptors that are one of the major targets of clinically used drugs. Here, we identify GPR183, also known as EBI2, as the most highly expressed inhibitory G-protein-coupled receptor in BAT among the receptors examined. Activation of EBI2 using its endogenous ligand 7α,25-dihydroxycholesterol significantly decreases BAT-mediated energy expenditure in mice. In contrast, mice deficient for EBI2 show increased energy dissipation in response to cold. Interestingly, only thermogenic adipose tissue depots - BAT and subcutaneous white adipose tissue -respond to 7α,25-dihydroxycholesterol treatment/EBI2 activation but not gonadal white fat, which has the lowest thermogenic capacity. EBI2 activation in brown adipocytes significantly reduces norepinephrine-induced cAMP production, whereas pharmacological inhibition or genetic ablation of EBI2 results in an increased response. Importantly, EBI2 significantly inhibits norepinephrine-induced activation of human brown adipocytes. Our data identify the 7α,25-dihydroxycholesterol/EBI2 signaling pathway as a so far unknown BAT inhibitor. Understanding the inhibitory regulation of BAT might lead to novel pharmacological approaches to increase the activity of thermogenic adipose tissue and whole body energy expenditure in humans.


Asunto(s)
Adipocitos Marrones , Tejido Adiposo Pardo , Metabolismo Energético , Receptores Acoplados a Proteínas G , Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Humanos , Ratones , Norepinefrina/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Termogénesis
9.
Mol Biol Cell ; 33(12): br22, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35947507

RESUMEN

The different adipose tissues (ATs) can be distinguished according to their function. For example, white AT stores energy in form of lipids, whereas brown AT dissipates energy in the form of heat. These functional differences are represented in the respective adipocyte morphology; whereas white adipocytes contain large, unilocular lipid droplets, brown adipocytes contain smaller, multilocular lipid droplets. However, an automated, image analysis pipeline to comprehensively analyze adipocytes in vitro in cell culture as well as ex vivo in tissue sections is missing. We here present AdipoQ, an open-source software implemented as ImageJ plugins that allows us to analyze adipocytes in tissue sections and in vitro after histological and/or immunofluorescent labeling. AdipoQ is compatible with different imaging modalities and staining methods, allows batch processing of large datasets and simple post-hoc analysis, provides a broad band of parameters, and allows combining multiple fluorescent readouts. Therefore AdipoQ is of immediate use not only for basic research but also for clinical diagnosis.


Asunto(s)
Adipocitos , Tejido Adiposo Pardo , Gotas Lipídicas , Lípidos , Programas Informáticos
10.
Microbiome ; 10(1): 96, 2022 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-35739571

RESUMEN

BACKGROUND: Bariatric surgery remains the most effective therapy for adiposity reduction and remission of type 2 diabetes. Although different bariatric procedures associate with pronounced shifts in the gut microbiota, their functional role in the regulation of energetic and metabolic benefits achieved with the surgery are not clear. METHODS: To evaluate the causal as well as the inherent therapeutic character of the surgery-altered gut microbiome in improved energy and metabolic control in diet-induced obesity, an antibiotic cocktail was used to eliminate the gut microbiota in diet-induced obese rats after gastric bypass surgery, and gastric bypass-shaped gut microbiota was transplanted into obese littermates. Thorough metabolic profiling was combined with omics technologies on samples collected from cecum and plasma to identify adaptions in gut microbiota-host signaling, which control improved energy balance and metabolic profile after surgery. RESULTS: In this study, we first demonstrate that depletion of the gut microbiota largely reversed the beneficial effects of gastric bypass surgery on negative energy balance and improved glucolipid metabolism. Further, we show that the gastric bypass-shaped gut microbiota reduces adiposity in diet-induced obese recipients by re-activating energy expenditure from metabolic active brown adipose tissue. These beneficial effects were linked to improved glucose homeostasis, lipid control, and improved fatty liver disease. Mechanistically, these effects were triggered by modulation of taurine metabolism by the gastric bypass gut microbiota, fostering an increased abundance of intestinal and circulating taurine-conjugated bile acid species. In turn, these bile acids activated gut-restricted FXR and systemic TGR5 signaling to stimulate adaptive thermogenesis. CONCLUSION: Our results establish the role of the gut microbiome in the weight loss and metabolic success of gastric bypass surgery. We here identify a signaling cascade that entails altered bile acid receptor signaling resulting from a collective, hitherto undescribed change in the metabolic activity of a cluster of bacteria, thereby readjusting energy imbalance and metabolic disease in the obese host. These findings strengthen the rationale for microbiota-targeted strategies to improve and refine current therapies of obesity and metabolic syndrome. Video Abstract Bariatric Surgery (i.e. RYGB) or the repeated fecal microbiota transfer (FMT) from RYGB donors into DIO (diet-induced obesity) animals induces shifts in the intestinal microbiome, an effect that can be impaired by oral application of antibiotics (ABx). Our current study shows that RYGB-dependent alterations in the intestinal microbiome result in an increase in the luminal and systemic pool of Taurine-conjugated Bile acids (TCBAs) by various cellular mechanisms acting in the intestine and the liver. TCBAs induce signaling via two different receptors, farnesoid X receptor (FXR, specifically in the intestines) and the G-protein-coupled bile acid receptor TGR5 (systemically), finally resulting in metabolic improvement and advanced weight management. BSH, bile salt hydrolase; BAT brown adipose tissue.


Asunto(s)
Diabetes Mellitus Tipo 2 , Derivación Gástrica , Microbiota , Tejido Adiposo/metabolismo , Animales , Ácidos y Sales Biliares , Glucemia , Dieta , Obesidad/metabolismo , Obesidad/cirugía , Ratas , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Taurina , Termogénesis
11.
Mol Cancer ; 9: 31, 2010 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-20137080

RESUMEN

BACKGROUND: The human microbial pathogen Helicobacter pylori resides in the stomach of about fifty percent of the world's population and represents a risk factor for chronic gastritis, peptic ulcers and, in rare cases, gastric cancer. Alterations of the Wnt/beta-catenin signaling pathway have been described in almost every human cancer disease, due to the regulation of target genes being involved in cell cycle control, differentiation, cell migration or stem cell control. Our study aimed to elucidate the role of proximal Wnt signaling components low density lipoprotein receptor-related protein 6 (LRP6) and Dishevelled (Dvl) in the activation of beta-catenin early after infection of gastric epithelial cells with H. pylori. RESULTS: Infection of gastric epithelial NCI-N87 cells with H. pylori induces rapid phosphorylation of the Wnt/beta-catenin pathway co-receptor LRP6 independent of the cytotoxin-associated gene A (CagA) or vacuolating cytotoxin A (VacA). However, bacteria lacking a functional type 4 secretion system (T4SS) failed to induce LRP6 phosphorylation. Further, we identified proteins of the Dvl family, namely Dvl2 and Dvl3, which are involved in LRP6 phosphorylation. H. pylori-induced nuclear accumulation of beta-catenin and its transcriptional activation, and expression of Wnt target genes are strongly reduced in stable knockdown cell lines deficient for LRP6, Dvl2 or Dvl3. CONCLUSION: We analysed the H. pylori-induced activation of Wnt-signaling factors and demonstrate for the first time that the canonical Wnt-signaling proteins LRP6 and Dvl2 and Dvl3 are involved in the regulation of beta-catenin.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Helicobacter pylori/fisiología , Fosfoproteínas/metabolismo , Receptores de LDL/metabolismo , beta Catenina/metabolismo , Proteínas Bacterianas/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Proteínas Dishevelled , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Infecciones por Helicobacter/metabolismo , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad , Modelos Biológicos , Fosforilación , Factores de Transcripción TCF/metabolismo
12.
Exp Cell Res ; 315(20): 3500-8, 2009 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-19665015

RESUMEN

Helicobacter pylori, a microaerophilic gram-negative bacterium, colonizes the human stomach. About 50% of the world's population is infected, and this infection is considered as the major risk factor for the development of gastric adenocarcinomas in 1% of infected subjects. Carcinogenesis is characterized by the process of epithelial-to-mesenchymal transition (EMT), in the course of which fully differentiated epithelial cells turn into depolarized and migratory cells. Concomitant disruption of adherence junctions (AJ) is facilitated by growth factors like hepatocyte growth factor 1 (HGF-1), but has been also shown to depend on ectodomain shedding of E-cadherin. The aim of this study was to investigate the impact of infection with H. pylori of NCI-N87 gastric epithelial cells on the shedding of E-cadherin and HGF-receptor c-Met. Our results show that infection with H. pylori provokes shedding of the surface proteins c-Met and E-cadherin. Evidence is provided that ADAM10 contributes to the shedding of c-Met and E-cadherin.


Asunto(s)
Cadherinas/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/fisiología , Fragmentos de Péptidos/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM10 , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Línea Celular , Progresión de la Enfermedad , Expresión Génica/genética , Glicina/análogos & derivados , Glicina/farmacología , Infecciones por Helicobacter/microbiología , Humanos , Ácidos Hidroxámicos/farmacología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/antagonistas & inhibidores , Metaloendopeptidasas/metabolismo , Inhibidores de Proteasas/farmacología , ARN Interferente Pequeño/genética , Estómago/citología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología , Transfección
13.
Mol Metab ; 40: 101029, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32480042

RESUMEN

OBJECTIVES: Typically, obesity results from an inappropriate balance between energy uptake from nutrient consumption and burning of calories, which leads to a pathological increase in fat mass. Obesity is a major cause of insulin resistance and diabetes. Inhibitory G proteins (Gαi) form a subfamily that is involved in the regulation of adipose tissue function. Among the three Gαi members, i.e. Gαi1, Gαi2, Gαi3, the Gαi2, protein is predominantly expressed in adipose tissue. However, the functions of the Gαi2 isoform in adipose tissue and its impact on the development of obesity are poorly understood. METHODS: By using AdipoqCreERT2 mice, we generated adipocyte-specific Gnai2-deficient mice to study Gαi2 function, specifically in white and brown adipocytes. These mice were fed either a control diet (CD) or a high fat diet (HFD). Mice were examined for obesity development, insulin resistance and glucose intolerance. We examined adipocyte morphology and the development of inflammation in the white adipose tissue. Finally, intracellular cAMP levels as an indicator of Gαi signaling and glycerol release as an indicator of lipolysis rates were measured to verify the impact of Gαi2 on the signaling pathway in brown and white adipocytes. RESULTS: An adipocyte-specific deficiency of Gαi2 significantly reduced diet-induced obesity, leading to decreased fat masses, smaller adipocytes and decreased inflammation in the white adipose tissue relative to littermate controls. Concurrently, oxygen consumption of brown adipocytes and in vivo measured energy expenditure were significantly enhanced. In addition, glucose tolerance and insulin sensitivity of HFD-fed adipocyte-specific Gnai2-deficient mice were improved compared to the respective controls. In the absence of Gαi2, adrenergic stimulation of intracellular adipocyte cAMP levels was increased, which correlated with increased lipolysis and energy expenditure. CONCLUSION: We conclude that adipocyte Gαi2 is a major regulator of adipocyte lipid content in diet-induced obesity by inhibiting adipocyte lipolysis in a cAMP-dependent manner resulting in increased energy expenditure.


Asunto(s)
Tejido Adiposo/metabolismo , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Obesidad/metabolismo , Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Tejido Adiposo/fisiología , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Dieta Alta en Grasa , Metabolismo Energético , Subunidad alfa de la Proteína de Unión al GTP Gi2/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Glucosa/metabolismo , Intolerancia a la Glucosa/metabolismo , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Lipólisis , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/genética , Consumo de Oxígeno
14.
Cell Metab ; 32(1): 56-70.e7, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32589947

RESUMEN

The combination of aging populations with the obesity pandemic results in an alarming rise in non-communicable diseases. Here, we show that the enigmatic adenosine A2B receptor (A2B) is abundantly expressed in skeletal muscle (SKM) as well as brown adipose tissue (BAT) and might be targeted to counteract age-related muscle atrophy (sarcopenia) as well as obesity. Mice with SKM-specific deletion of A2B exhibited sarcopenia, diminished muscle strength, and reduced energy expenditure (EE), whereas pharmacological A2B activation counteracted these processes. Adipose tissue-specific ablation of A2B exacerbated age-related processes and reduced BAT EE, whereas A2B stimulation ameliorated obesity. In humans, A2B expression correlated with EE in SKM, BAT activity, and abundance of thermogenic adipocytes in white fat. Moreover, A2B agonist treatment increased EE from human adipocytes, myocytes, and muscle explants. Mechanistically, A2B forms heterodimers required for adenosine signaling. Overall, adenosine/A2B signaling links muscle and BAT and has both anti-aging and anti-obesity potential.


Asunto(s)
Envejecimiento/metabolismo , Obesidad/metabolismo , Receptor de Adenosina A2B/metabolismo , Adolescente , Adulto , Animales , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Receptor de Adenosina A2B/deficiencia , Transducción de Señal , Adulto Joven
16.
Ann Anat ; 218: 227-235, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29730468

RESUMEN

Obesity and aging are associated with periodontitis, which represents a chronic inflammatory disease of the tooth-supporting tissues, i.e. the periodontium. However, if both risk factors also have a negative impact on crestal alveolar bone in a clinically healthy periodontium, has yet to be elucidated and was analyzed in this in-vivo study. Eight C57BL/6 mice were fed a normal diet during the entire study. Half of these mice were sacrificed at week 19 (group 1: younger lean mice), whereas the other half of the animals were sacrificed at week 25 (group 2: older lean mice). In addition, four mice were fed a high-fat diet until their sacrifice at week 19 (group 3: younger obese mice). Mandibles and maxillae were scanned by micro-computed tomography and, subsequently, the distance between the cementoenamel junction and alveolar bone crest (CEJ-ABC) at all molars was determined. Levels of interleukin-6, cyclooxygenase-2, visfatin and adiponectin in gingival samples were quantified by real-time PCR. For statistical analyses, the Mann-Whitney-U test was applied (p<0.05). As compared to lean mice, obese animals presented a significantly increased CEJ-ABC distance, i.e. reduced alveolar bone crest height, at week 19. The alveolar bone loss was mainly found at the first molars of the mandibles. In animals fed a normal diet, the alveolar bone crest height in the mandibles and maxillae was significantly lower in the older mice as compared to the younger animals. Furthermore, gingival cyclooxygenase-2 and visfatin expressions were higher in the obese versus lean mice and in the older versus younger mice. This in-vivo investigation shows that obesity and older age can result in reduced alveolar bone crest height and suggests that they represent risk factors even in a clinically healthy periodontium.


Asunto(s)
Envejecimiento/patología , Proceso Alveolar/crecimiento & desarrollo , Proceso Alveolar/patología , Obesidad/patología , Pérdida de Hueso Alveolar , Animales , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Citocinas/biosíntesis , Citocinas/genética , Dieta Alta en Grasa , Expresión Génica , Encía/química , Encía/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , Mandíbula/crecimiento & desarrollo , Mandíbula/patología , Maxilar/crecimiento & desarrollo , Maxilar/patología , Ratones , Ratones Endogámicos C57BL , Diente Molar/anatomía & histología , Nicotinamida Fosforribosiltransferasa/biosíntesis , Nicotinamida Fosforribosiltransferasa/genética , Microtomografía por Rayos X
17.
Diabetes ; 67(7): 1226-1236, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29650773

RESUMEN

Activating brown adipose tissue (BAT) could provide a potential approach for the treatment of obesity and metabolic disease in humans. Obesity is associated with upregulation of the endocannabinoid system, and blocking the cannabinoid type 1 receptor (CB1R) has been shown to cause weight loss and to decrease cardiometabolic risk factors. These effects may be mediated partly via increased BAT metabolism, since there is evidence that CB1R antagonism activates BAT in rodents. To investigate the significance of CB1R in BAT function, we quantified the density of CB1R in human and rodent BAT using the positron emission tomography radioligand [18F]FMPEP-d2 and measured BAT activation in parallel with the glucose analog [18F]fluorodeoxyglucose. Activation by cold exposure markedly increased CB1R density and glucose uptake in the BAT of lean men. Similarly, ß3-receptor agonism increased CB1R density in the BAT of rats. In contrast, overweight men with reduced BAT activity exhibited decreased CB1R in BAT, reflecting impaired endocannabinoid regulation. Image-guided biopsies confirmed CB1R mRNA expression in human BAT. Furthermore, CB1R blockade increased glucose uptake and lipolysis of brown adipocytes. Our results highlight that CB1Rs are significant for human BAT activity, and the CB1Rs provide a novel therapeutic target for BAT activation in humans.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Respuesta al Choque por Frío/genética , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB1/metabolismo , Tejido Adiposo Pardo/diagnóstico por imagen , Tejido Adiposo Pardo/patología , Adulto , Animales , Células Cultivadas , Fluorodesoxiglucosa F18 , Humanos , Masculino , Persona de Mediana Edad , Sobrepeso/diagnóstico por imagen , Sobrepeso/genética , Sobrepeso/metabolismo , Tomografía de Emisión de Positrones , Pirrolidinonas , Ratas , Ratas Sprague-Dawley , Termogénesis/genética , Regulación hacia Arriba/genética , Adulto Joven
18.
Sci Rep ; 8(1): 9372, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29921982

RESUMEN

In cirrhotic patients, portal hypertension (PHT) deteriorates survival, yet treatment options are limited. A major contributor to increased intrahepatic vasoconstriction in PHT is dysfunctional nitric-oxide signaling. Soluble guanylate cyclase (sGC) is the receptor of nitric-oxide and can be stimulated by riociguat. Riociguat is approved for pulmonary hypertension but has not been studied in liver cirrhosis. In this study we assessed the effects of riociguat on PHT and liver fibrosis in cholestatic (bile duct ligation, BDL) and toxic (carbon-tetrachloride, CCl4) rat models. In cirrhotic livers sGC expression was upregulated. In BDL rats, riociguat reduced liver fibrosis and decreased portal pressure without affecting systemic hemodynamics. In an early BDL disease stage, riociguat decreased bile duct proliferation, improved sinusoidal vascular dysfunction and inhibited angiogenesis. In advanced BDL riociguat exhibited anti-inflammatory effects. In CCl4 rats the beneficial effects of riociguat treatment were less pronounced and confined to an early disease stage. Similarly, in patients with cholestatic cirrhosis and PHT nitrates (that induce sGC activity) decreased portal pressure more effectively than in patients with non-cholestatic etiology. We also found an improvement of transaminases in patients with pulmonary hypertension receiving riociguat. Our findings support the clinical development of sGC stimulators in patients with cirrhotic PHT.


Asunto(s)
Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/enzimología , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Guanilil Ciclasa Soluble/metabolismo , Animales , Hemodinámica/efectos de los fármacos , Humanos , Hipertensión Portal/tratamiento farmacológico , Hipertensión Portal/enzimología , Masculino , Ratas , Ratas Sprague-Dawley
19.
Mol Metab ; 6(3): 276-287, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28271034

RESUMEN

OBJECTIVE: Obesity is defined as an abnormal increase in white adipose tissue (WAT) and is a major risk factor for type 2 diabetes and cardiovascular disease. Brown adipose tissue (BAT) dissipates energy and correlates with leanness. Natriuretic peptides have been shown to be beneficial for brown adipocyte differentiation and browning of WAT. METHODS: Here, we investigated the effects of an optimized designer natriuretic peptide (CD-NP) on murine adipose tissues in vitro and in vivo. RESULTS: In murine brown and white adipocytes, CD-NP activated cGMP production, promoted adipogenesis, and increased thermogenic markers. Consequently, mice treated for 10 days with CD-NP exhibited increased "browning" of WAT. To study CD-NP effects on diet-induced obesity (DIO), we delivered CD-NP for 12 weeks. Although CD-NP reduced inflammation in WAT, CD-NP treated DIO mice exhibited a significant increase in body mass, worsened glucose tolerance, and hepatic steatosis. Long-term CD-NP treatment resulted in an increased expression of the NP scavenging receptor (NPR-C) and decreased lipolytic activity. CONCLUSIONS: NP effects differed depending on the duration of treatment raising questions about the rational of natriuretic peptide treatment in obese patients.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Venenos Elapídicos/farmacología , Péptido Natriurético Tipo-C/farmacología , Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Adipogénesis/efectos de los fármacos , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Animales , Diabetes Mellitus Tipo 2/complicaciones , Dieta , Venenos Elapídicos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Péptido Natriurético Tipo-C/metabolismo , Obesidad/etiología , Termogénesis/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA