Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 145
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
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.
Immunity ; 50(5): 1232-1248.e14, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31027998

RESUMEN

Regulatory T cells (Treg cells) are important for preventing autoimmunity and maintaining tissue homeostasis, but whether Treg cells can adopt tissue- or immune-context-specific suppressive mechanisms is unclear. Here, we found that the enzyme hydroxyprostaglandin dehydrogenase (HPGD), which catabolizes prostaglandin E2 (PGE2) into the metabolite 15-keto PGE2, was highly expressed in Treg cells, particularly those in visceral adipose tissue (VAT). Nuclear receptor peroxisome proliferator-activated receptor-γ (PPARγ)-induced HPGD expression in VAT Treg cells, and consequential Treg-cell-mediated generation of 15-keto PGE2 suppressed conventional T cell activation and proliferation. Conditional deletion of Hpgd in mouse Treg cells resulted in the accumulation of functionally impaired Treg cells specifically in VAT, causing local inflammation and systemic insulin resistance. Consistent with this mechanism, humans with type 2 diabetes showed decreased HPGD expression in Treg cells. These data indicate that HPGD-mediated suppression is a tissue- and context-dependent suppressive mechanism used by Treg cells to maintain adipose tissue homeostasis.


Asunto(s)
Dinoprostona/análogos & derivados , Dinoprostona/metabolismo , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Grasa Intraabdominal/inmunología , Linfocitos T Reguladores/enzimología , Linfocitos T Reguladores/inmunología , Células 3T3 , Animales , Línea Celular , Diabetes Mellitus Tipo 2/metabolismo , Células HEK293 , Homeostasis/inmunología , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Resistencia a la Insulina/genética , Grasa Intraabdominal/citología , Células Jurkat , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Noqueados , Factor de Transcripción STAT5/metabolismo
3.
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
4.
Mol Cell ; 80(6): 940-954.e6, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33202251

RESUMEN

Mechanisms that control mobilization of cytosolic calcium [Ca2+]i are key for regulation of numerous eukaryotic cell functions. One such paradigmatic mechanism involves activation of phospholipase Cß (PLCß) enzymes by G protein ßγ subunits from activated Gαi-Gßγ heterotrimers. Here, we report identification of a master switch to enable this control for PLCß enzymes in living cells. We find that the Gαi-Gßγ-PLCß-Ca2+ signaling module is entirely dependent on the presence of active Gαq. If Gαq is pharmacologically inhibited or genetically ablated, Gßγ can bind to PLCß but does not elicit Ca2+ signals. Removal of an auto-inhibitory linker that occludes the active site of the enzyme is required and sufficient to empower "stand-alone control" of PLCß by Gßγ. This dependence of Gi-Gßγ-Ca2+ on Gαq places an entire signaling branch of G-protein-coupled receptors (GPCRs) under hierarchical control of Gq and changes our understanding of how Gi-GPCRs trigger [Ca2+]i via PLCß enzymes.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP/genética , Subunidades beta de la Proteína de Unión al GTP/genética , Subunidades gamma de la Proteína de Unión al GTP/genética , Proteínas de Unión al GTP Heterotriméricas/genética , Fosfolipasa C beta/genética , Calcio/metabolismo , Señalización del Calcio/genética , Citosol/metabolismo , Células HEK293 , Humanos , Unión Proteica/genética , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/genética
5.
Nature ; 566(7743): 279-283, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30700909

RESUMEN

Adaptation to the environment and extraction of energy are essential for survival. Some species have found niches and specialized in using a particular source of energy, whereas others-including humans and several other mammals-have developed a high degree of flexibility1. A lot is known about the general metabolic fates of different substrates but we still lack a detailed mechanistic understanding of how cells adapt in their use of basic nutrients2. Here we show that the closely related fasting/starvation-induced forkhead transcription factors FOXK1 and FOXK2 induce aerobic glycolysis by upregulating the enzymatic machinery required for this (for example, hexokinase-2, phosphofructokinase, pyruvate kinase, and lactate dehydrogenase), while at the same time suppressing further oxidation of pyruvate in the mitochondria by increasing the activity of pyruvate dehydrogenase kinases 1 and 4. Together with suppression of the catalytic subunit of pyruvate dehydrogenase phosphatase 1 this leads to increased phosphorylation of the E1α regulatory subunit of the pyruvate dehydrogenase complex, which in turn inhibits further oxidation of pyruvate in the mitochondria-instead, pyruvate is reduced to lactate. Suppression of FOXK1 and FOXK2 induce the opposite phenotype. Both in vitro and in vivo experiments, including studies of primary human cells, show how FOXK1 and/or FOXK2 are likely to act as important regulators that reprogram cellular metabolism to induce aerobic glycolysis.


Asunto(s)
Aerobiosis , Factores de Transcripción Forkhead/metabolismo , Glucólisis , Células 3T3 , Animales , Células Cultivadas , Femenino , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/genética , Humanos , Ácido Láctico/biosíntesis , Ácido Láctico/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/enzimología , Mitocondrias/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Oxidación-Reducción , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Piruvato Deshidrogenasa (Lipoamida)-Fosfatasa/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Complejo Piruvato Deshidrogenasa/química , Complejo Piruvato Deshidrogenasa/metabolismo , Ácido Pirúvico/metabolismo
6.
Int J Mol Sci ; 23(1)2021 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-35008716

RESUMEN

The gap junction protein connexin 43 (Cx43) is associated with increased cell migration and to related changes of the actin cytoskeleton, which is mediated via its C-terminal cytoplasmic tail and is independent of its channel function. Cx43 has been shown to possess an angiogenic potential, however, the role of Cx43 in endothelial cell migration has not yet been investigated. Here, we found that the knock-down of Cx43 by siRNA in human microvascular endothelial cells (HMEC) reduces migration, as assessed by a wound assay in vitro and impaired aortic vessel sprouting ex vivo. Immunoprecipitation of Cx43 revealed an interaction with the tyrosine phosphatase SHP-2, which enhanced its phosphatase activity, as observed in Cx43 expressing HeLa cells compared to cells treated with an empty vector. Interestingly, the expression of a dominant negative substrate trapping mutant SHP-2 (CS) in HMEC, via lentiviral transduction, also impaired endothelial migration to a similar extent as Cx43 siRNA compared to SHP-2 WT. Moreover, the reduction in endothelial migration upon Cx43 siRNA could not be rescued by the introduction of a constitutively active SHP-2 construct (EA). Our data demonstrate that Cx43 and SHP-2 mediate endothelial cell migration, revealing a novel interaction between Cx43 and SHP-2, which is essential for this process.


Asunto(s)
Movimiento Celular , Conexina 43/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Animales , Movimiento Celular/genética , Regulación hacia Abajo/genética , Células HeLa , Humanos , Neovascularización Fisiológica/genética , Unión Proteica , Ratas
7.
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
8.
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
9.
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
10.
Eur J Nucl Med Mol Imaging ; 46(3): 743-750, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30105585

RESUMEN

PURPOSE: Brown adipose tissue (BAT) has emerged as a potential target to combat obesity and diabetes, but novel strategies to activate BAT are needed. Adenosine and A2A receptor (A2AR) agonism activate BAT in rodents, and endogenous adenosine is released locally in BAT as a by-product of noradrenaline, but physiological data from humans is lacking. The purpose of this pilot study was to investigate the effects of exogenous adenosine on human BAT perfusion, and to determine the density of A2ARs in human BAT in vivo for the first time, using PET/CT imaging. METHODS: Healthy, lean men (n = 10) participated in PET/CT imaging with two radioligands. Perfusion of BAT, white adipose tissue (WAT) and muscle was quantified with [15O]H2O at baseline, during cold exposure and during intravenous administration of adenosine. A2AR density of the tissues was quantified with [11C]TMSX at baseline and during cold exposure. RESULTS: Adenosine increased the perfusion of BAT even more than cold exposure (baseline 8.3 ± 4.5, cold 19.6 ± 9.3, adenosine 28.6 ± 7.9 ml/100 g/min, p < 0.01). Distribution volume of [11C]TMSX in BAT was significantly lower during cold exposure compared to baseline. In cold, low [11C]TMSX binding coincided with high concentrations of noradrenaline. CONCLUSIONS: Adenosine administration caused a maximal perfusion effect in human supraclavicular BAT, indicating increased oxidative metabolism. Cold exposure increased noradrenaline concentrations and decreased the density of A2AR available for radioligand binding in BAT, suggesting augmented release of endogenous adenosine. Our results show that adenosine and A2AR are relevant for activation of human BAT, and A2AR provides a future target for enhancing BAT metabolism.


Asunto(s)
Tejido Adiposo Pardo/diagnóstico por imagen , Tejido Adiposo Pardo/metabolismo , Radioisótopos de Oxígeno , Tomografía Computarizada por Tomografía de Emisión de Positrones , Receptor de Adenosina A2A/metabolismo , Agua , Xantinas , Adenosina/farmacología , Tejido Adiposo Pardo/irrigación sanguínea , Tejido Adiposo Pardo/efectos de los fármacos , Adolescente , Adulto , Frío , Metabolismo Energético/efectos de los fármacos , Ácidos Grasos no Esterificados/metabolismo , Femenino , Humanos , Masculino , Norepinefrina/metabolismo , Flujo Sanguíneo Regional/efectos de los fármacos , Adulto Joven
11.
Curr Opin Cardiol ; 34(4): 376-383, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30994530

RESUMEN

PURPOSE OF REVIEW: Cardiovascular and metabolic diseases are closely linked and commonly occur in the same patients. This review focuses on the cyclic guanosine monophosphate (cGMP) system and its crosstalk between metabolism and the cardiovascular system. RECENT FINDINGS: Recent studies suggest that cGMP, which serves as second messenger for nitric oxide and for natriuretic peptides, improves oxidative metabolism and insulin signaling. The clinical evidence is particularly strong for the natriuretic peptide branch of the cGMP system. Clinical trials suggested improvements in insulin sensitivity and reductions in the risk of progressing to type 2 diabetes mellitus. However, further studies are needed. SUMMARY: Enhancing cGMP signaling through nonpharmacological or pharmacological means may improve glucose metabolism in addition to affecting the cardiovascular system. However, excessive cGMP production could have significant unwanted cardiovascular and metabolic effects.


Asunto(s)
Enfermedades Cardiovasculares , GMP Cíclico , Diabetes Mellitus Tipo 2 , Humanos , Óxido Nítrico , Transducción de Señal
12.
Handb Exp Pharmacol ; 251: 337-346, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29633182

RESUMEN

In the last decade, exosomes have gained interest as a new type of intercellular communication between cells and tissues. Exosomes are circulating, cell-derived lipid vesicles smaller than 200 nm that contain proteins and nucleic acids, including microRNAs (miRNAs), and are able to modify cellular targets. Exosomal miRNAs function as signalling molecules that regulate the transcription of their target genes and can cause phenotypic transformation of recipient cells. Recent studies have shown that brown fat secretes exosomes as a form of communication with other metabolic organs such as the liver. Moreover, it has been shown that levels of miRNAs in BAT-derived exosomes change after BAT activation in vitro and in vivo. Thus, BAT-derived exosomes can be used as potential biomarkers of BAT activity. Here, we review the present knowledge about BAT-derived exosomes and their role in metabolism.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Exosomas , MicroARNs , Biomarcadores , Comunicación Celular , Exosomas/metabolismo , Hígado/fisiología , MicroARNs/metabolismo
13.
Handb Exp Pharmacol ; 251: 161-182, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29633180

RESUMEN

Cold-induced activation of brown adipose tissue (BAT) is mediated by norepinephrine and adenosine that are released during sympathetic nerve activation. Both signaling molecules induce an increase in intracellular levels of 3',5'-cyclic adenosine monophosphate (cAMP) in murine and human BAT. In brown adipocytes, cAMP plays a central role, because it activates lipolysis, glucose uptake, and thermogenesis. Another well-studied intracellular second messenger is 3',5'-cyclic guanosine monophosphate (cGMP), which closely resembles cAMP. Several studies have shown that intact cGMP signaling is essential for normal adipogenic differentiation and BAT-mediated thermogenesis in mice. This chapter highlights recent observations, demonstrating the physiological significance of cyclic nucleotide signaling in BAT as well as their potential to induce browning of white adipose tissue (WAT) in mice and humans.


Asunto(s)
Tejido Adiposo Pardo , Tejido Adiposo Blanco/metabolismo , Termogénesis , Tejido Adiposo Blanco/química , Animales , AMP Cíclico/química , GMP Cíclico/química , Humanos , Ratones
14.
Int J Mol Sci ; 20(18)2019 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-31500245

RESUMEN

Vascular remodeling and angiogenesis are required to improve the perfusion of ischemic tissues. The hypoxic environment, induced by ischemia, is a potent stimulus for hypoxia inducible factor 1α (HIF-1α) upregulation and activation, which induce pro-angiogenic gene expression. We previously showed that the tyrosine phosphatase SHP-2 drives hypoxia mediated HIF-1α upregulation via inhibition of the proteasomal pathway, resulting in revascularization of wounds in vivo. However, it is still unknown if SHP-2 mediates HIF-1α upregulation by affecting 26S proteasome activity and how the proteasome is regulated upon hypoxia. Using a reporter construct containing the oxygen-dependent degradation (ODD) domain of HIF-1α and a fluorogenic proteasome substrate in combination with SHP-2 mutant constructs, we show that SHP-2 inhibits the 26S proteasome activity in endothelial cells under hypoxic conditions in vitro via Src kinase/p38 mitogen-activated protein kinase (MAPK) signalling. Moreover, the simultaneous expression of constitutively active SHP-2 (E76A) and inactive SHP-2 (CS) in separate hypoxic wounds in the mice dorsal skin fold chamber by localized magnetic nanoparticle-assisted lentiviral transduction showed specific regulation of proteasome activity in vivo. Thus, we identified a new additional mechanism of SHP-2 mediated HIF-1α upregulation and proteasome activity, being functionally important for revascularization of wounds in vivo. SHP-2 may therefore constitute a potential novel therapeutic target for the induction of angiogenesis in ischemic vascular disease.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/química , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Piel/lesiones , Animales , Hipoxia de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/citología , Células Endoteliales/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas , Nanopartículas de Magnetita , Masculino , Ratones , Mutación , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteolisis , Piel/irrigación sanguínea , Remodelación Vascular
15.
Annu Rev Pharmacol Toxicol ; 55: 207-27, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25149919

RESUMEN

Brown adipose tissue (BAT) was previously regarded as a special type of fat relevant only for defending hibernating animals and newborns against a cold environment. Recently, BAT has received considerable attention following its (re)discovery in humans. Using glucose tracers, multiple laboratories independently found metabolically active BAT in adults. The enormous metabolic powers of BAT in animal models could make it an attractive target for antiobesity therapies in humans. Here, we review the present knowledge on the role of BAT in energy homeostasis and metabolism, focusing on signaling pathways and potential targets for novel therapeutics. We also shine light on ongoing debates, including those about the true color of brown fat in adults, as well as on the requirements for translation of basic research on BAT into clinical medicine.


Asunto(s)
Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Blanco/efectos de los fármacos , Fármacos Antiobesidad/farmacología , Obesidad/tratamiento farmacológico , Adipocitos Marrones/efectos de los fármacos , Adipocitos Marrones/metabolismo , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/fisiopatología , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/fisiopatología , Animales , Descubrimiento de Drogas , Metabolismo Energético/efectos de los fármacos , Humanos , MicroARNs/metabolismo , Terapia Molecular Dirigida , Obesidad/genética , Obesidad/metabolismo , Obesidad/fisiopatología , Fenotipo , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo
16.
Stem Cells ; 35(4): 859-871, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27870307

RESUMEN

Gap junctional intercellular communication (GJIC) has been suggested to be involved in early embryonic development but the actual functional role remained elusive. Connexin (Cx) 43 and Cx45 are co-expressed in embryonic stem (ES) cells, form gap junctions and are considered to exhibit adhesive function and/or to contribute to the establishment of defined communication compartments. Here, we describe the generation of Cx43/Cx45-double deficient mouse ES cells to achieve almost complete breakdown of GJIC. Cre-loxP induced deletion of both, Cx43 and Cx45, results in a block of differentiation in embryoid bodies (EBs) without affecting pluripotency marker expression and proliferation in ES cells. We demonstrate that GJIC-incompetent ES cells fail to form primitive endoderm in EB cultures, representing the inductive key step of further differentiation events. Lentiviral overexpression of either Cx43 or Cx45 in Cx43/45 mutants rescued the observed phenotype, confirming the specificity and indicating a partially redundant function of both connexins. Upon differentiation GJIC-incompetent ES cells exhibit a strikingly altered subcellular localization pattern of the transcription factor NFATc3. Control EBs exhibit significantly more activated NFATc3 in cellular nuclei than mutant EBs suggesting that Cx-mediated communication is needed for synchronized NFAT activation to induce orchestrated primitive endoderm formation. Moreover, pharmacological inhibition of NFATc3 activation by Cyclosporin A, a well-described inhibitor of calcineurin, phenocopies the loss of GJIC in control cells. Stem Cells 2017;35:859-871.


Asunto(s)
Comunicación Celular , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Endodermo/embriología , Endodermo/metabolismo , Uniones Comunicantes/metabolismo , Animales , Apoptosis , Biomarcadores/metabolismo , Calcineurina/metabolismo , Diferenciación Celular , Proliferación Celular , Conexina 43/metabolismo , Conexinas/metabolismo , Endodermo/citología , Gastrulación , Lentivirus/metabolismo , Ratones , Mutagénesis/genética , Factores de Transcripción NFATC/metabolismo , Transducción de Señal
17.
Eur J Nucl Med Mol Imaging ; 45(12): 2244, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30196366

RESUMEN

The original version of this article contained a mistake in the first sentence of the Results section of the Abstract.

18.
Arterioscler Thromb Vasc Biol ; 37(12): 2280-2290, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29025706

RESUMEN

OBJECTIVE: Because of its strategic position between endothelial and smooth muscle cells in microvessels, Cx37 (Connexin 37) plays an important role in myoendothelial gap junctional intercellular communication. We have shown before that NO inhibits gap junctional intercellular communication through gap junctions containing Cx37. However, the underlying mechanism is not yet identified. APPROACH AND RESULTS: Using channel-forming Cx37 mutants exhibiting partial deletions or amino acid exchanges in their C-terminal loops, we now show that the phosphorylation state of a tyrosine residue at position 332 (Y332) in the C-terminus of Cx37 controls the gap junction-dependent spread of calcium signals. Mass spectra revealed that NO protects Cx37 from dephosphorylation at Y332 by inhibition of the protein tyrosine phosphatase SHP-2. Functionally, the inhibition of gap junctional intercellular communication by NO decreased the spread of the calcium signal (induced by mechanical stimulation of individual endothelial cells) from endothelial to smooth muscle cells in intact vessels, while, at the same time, augmenting the calcium signal spreading within the endothelium. Consequently, preincubation of small resistance arteries with exogenous NO enhanced the endothelium-dependent dilator response to acetylcholine in spite of a pharmacological blockade of NO-dependent cGMP formation by the soluable guanylyl cyclase inhibitor ODQ (1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one). CONCLUSIONS: Our results identify a novel mechanism by which NO can increase the efficacy of calcium, rising vasoactive agonists in the microvascular endothelium.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Comunicación Celular/efectos de los fármacos , Conexinas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Extremidad Inferior/irrigación sanguínea , Músculo Liso Vascular/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Animales , Arterias/efectos de los fármacos , Arterias/enzimología , Conexinas/genética , Relación Dosis-Respuesta a Droga , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/enzimología , Células HeLa , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Masculino , Ratones Endogámicos C57BL , Músculo Liso Vascular/enzimología , Óxido Nítrico/farmacología , Donantes de Óxido Nítrico/metabolismo , Fosforilación , Dominios Proteicos , Interferencia de ARN , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Tirosina , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Proteína alfa-4 de Unión Comunicante
19.
Mol Ther ; 25(7): 1616-1627, 2017 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-28434868

RESUMEN

Hypoxia promotes vascularization by stabilization and activation of the hypoxia inducible factor 1α (HIF-1α), which constitutes a target for angiogenic gene therapy. However, gene therapy is hampered by low gene delivery efficiency and non-specific side effects. Here, we developed a gene transfer technique based on magnetic targeting of magnetic nanoparticle-lentivirus (MNP-LV) complexes allowing site-directed gene delivery to individual wounds in the dorsal skin of mice. Using this technique, we were able to control HIF-1α dependent wound healing angiogenesis in vivo via site-specific modulation of the tyrosine phosphatase activity of SHP-2. We thus uncover a novel physiological role of SHP-2 in protecting HIF-1α from proteasomal degradation via a Src kinase dependent mechanism, resulting in HIF-1α DNA-binding and transcriptional activity in vitro and in vivo. Excitingly, using targeting of MNP-LV complexes, we achieved simultaneous expression of constitutively active as well as inactive SHP-2 mutant proteins in separate wounds in vivo and hereby specifically and locally controlled HIF-1α activity as well as the angiogenic wound healing response in vivo. Therefore, magnetically targeted lentiviral induced modulation of SHP-2 activity may be an attractive approach for controlling patho-physiological conditions relying on hypoxic vessel growth at specific sites.


Asunto(s)
Portadores de Fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Nanopartículas de Magnetita/administración & dosificación , Neovascularización Fisiológica , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Cicatrización de Heridas/genética , Animales , Línea Celular , Células Endoteliales/citología , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Hipoxia/genética , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Lentivirus/genética , Lentivirus/metabolismo , Nanopartículas de Magnetita/química , Ratones , Terapia Molecular Dirigida , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteolisis , Piel/lesiones , Piel/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
20.
Pflugers Arch ; 468(11-12): 1803-1807, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27704210

RESUMEN

Fat tissue is well known for its capacity to store energy and its detrimental role in obesity and metaflammation. However, humans possess different types of fat that have different functions in physiology and metabolic diseases. Apart from white adipose tissue (WAT), the body's main energy storage, there is also brown adipose tissue (BAT) that dissipates energy as a defense against cold and maintains energy balance for the whole body. BAT is present not only in newborns but also in adult humans and its mass correlates with leanness. Moreover, "brown-like" adipocytes have been detected in human WAT. These "brown-in-white" (brite) or beige cells can be induced by cold and a broad spectrum of pharmacological substances and, therefore, they are also known as "inducible brown adipocytes." Activation of brown and/or brite adipocytes reduces metabolic diseases, at least in murine models of obesity. Thus, brown/brite adipocytes represent the "brite" side of fat and are potential targets for novel therapeutic approaches for treatment of obesity and obesity-associated diseases.


Asunto(s)
Adipocitos Beige/metabolismo , Adipocitos Marrones/metabolismo , Obesidad/metabolismo , Adipocitos Beige/citología , Adipocitos Marrones/citología , Animales , Metabolismo Energético , Humanos , Obesidad/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA