Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Lipid Res ; 59(3): 452-461, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29343538

RESUMEN

The recent characterization of functional brown adipose tissue in adult humans has opened new perspectives for regulation of energy expenditure with respect to obesity and diabetes. Furthermore, dietary recommendations have taken into account the insufficient dietary intake of ω3 PUFAs and the concomitant excessive intake of ω6 PUFA associated with the occurrence of overweight/obesity. We aimed to study whether ω3 PUFAs could play a role in the recruitment and function of energy-dissipating brown/brite adipocytes. We show that ω3 PUFA supplementation has a beneficial effect on the thermogenic function of adipocytes. In vivo, a low dietary ω6:ω3 ratio improved the thermogenic response of brown and white adipose tissues to ß3-adrenergic stimulation. This effect was recapitulated in vitro by PUFA treatment of hMADS adipocytes. We pinpointed the ω6-derived eicosanoid prostaglandin (PG)F2α as the molecular origin because the effects were mimicked with a specific PGF2α receptor agonist. PGF2α level in hMADS adipocytes was reduced in response to ω3 PUFA supplementation. The recruitment of thermogenic adipocytes is influenced by the local quantity of individual oxylipins, which is controlled by the ω6:ω3 ratio of available lipids. In human nutrition, energy homeostasis may thus benefit from the implementation of a more balanced dietary ω6:ω3 ratio.


Asunto(s)
Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Blanco/efectos de los fármacos , Suplementos Dietéticos , Ácidos Grasos Omega-3/administración & dosificación , Ácidos Grasos Omega-3/farmacología , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Células Cultivadas , Humanos , Oxilipinas/metabolismo , Receptores de Prostaglandina/agonistas , Receptores de Prostaglandina/metabolismo
2.
Biochim Biophys Acta ; 1861(4): 285-93, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26775637

RESUMEN

Brite adipocytes recently discovered in humans are of considerable importance in energy expenditure by converting energy excess into heat. This property could be useful in the treatment of obesity, and nutritional aspects are relevant to this important issue. Using hMADS cells as a human cell model which undergoes a white to a brite adipocyte conversion, we had shown previously that arachidonic acid, the major metabolite of the essential nutrient Ω6-linoleic acid, plays a major role in this process. Its metabolites PGE2 and PGF2 alpha inhibit this process via a calcium-dependent pathway, whereas in contrast carbaprostacyclin (cPGI2), a stable analog of prostacyclin, activates white to brite adipocyte conversion. Herein, we show that cPGI2 generates via its cognate cell-surface receptor IP-R, a cyclic AMP-signaling pathway involving PKA activity which in turn induces the expression of UCP1. In addition, cPGI2 activates the pathway of nuclear receptors of the PPAR family, i.e. PPARα and PPARγ, which act separately from IP-R to up-regulate the expression of key genes involved in the function of brite adipocytes. Thus dual pathways are playing in concert for the occurrence of a browning process of human white adipocytes. These results make prostacyclin analogs as a new class of interesting molecules to treat obesity and associated diseases.


Asunto(s)
Adipocitos Marrones/efectos de los fármacos , Adipocitos Blancos/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Fármacos Antiobesidad/farmacología , Epoprostenol/análogos & derivados , PPAR alfa/agonistas , PPAR gamma/agonistas , Receptores de Prostaglandina/agonistas , Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Metabolismo Energético/efectos de los fármacos , Activación Enzimática , Epoprostenol/farmacología , Humanos , Lactante , Canales Iónicos/genética , Canales Iónicos/metabolismo , Masculino , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR gamma/metabolismo , Fenotipo , Interferencia de ARN , Receptores de Epoprostenol , Receptores de Prostaglandina/metabolismo , Transducción de Señal/efectos de los fármacos , Termogénesis/efectos de los fármacos , Factores de Tiempo , Transfección , Proteína Desacopladora 1
3.
Stem Cells ; 32(6): 1578-90, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24375761

RESUMEN

Adipose tissue contains thermogenic adipocytes (i.e., brown and brite/beige) that oxidize nutrients at exceptionally high rates via nonshivering thermogenesis. Its recent discovery in adult humans has opened up new avenues to fight obesity and related disorders such as diabetes. Here, we identified miR-26a and -26b as key regulators of human white and brite adipocyte differentiation. Both microRNAs are upregulated in early adipogenesis, and their inhibition prevented lipid accumulation while their overexpression accelerated it. Intriguingly, miR-26a significantly induced pathways related to energy dissipation, shifted mitochondrial morphology toward that seen in brown adipocytes, and promoted uncoupled respiration by markedly increasing the hallmark protein of brown fat, uncoupling protein 1. By combining in silico target prediction, transcriptomics, and an RNA interference screen, we identified the sheddase ADAM metallopeptidase domain 17 (ADAM17) as a direct target of miR-26 that mediated the observed effects on white and brite adipogenesis. These results point to a novel, critical role for the miR-26 family and its downstream effector ADAM17 in human adipocyte differentiation by promoting characteristics of energy-dissipating thermogenic adipocytes.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipogénesis/genética , MicroARNs/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAM17 , Adipocitos Marrones/citología , Adipocitos Marrones/ultraestructura , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/ultraestructura , Adulto , Diferenciación Celular/genética , Preescolar , Frío , Simulación por Computador , Humanos , Lactante , Canales Iónicos , Masculino , MicroARNs/genética , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas Mitocondriales , Transducción de Señal/genética , Transcriptoma/genética , Proteína Desacopladora 1 , Regulación hacia Arriba/genética
4.
Biochem Biophys Res Commun ; 440(4): 786-91, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24134848

RESUMEN

Chondrogenesis has been widely investigated in vitro using bone marrow-derived mesenchymal stromal cells (BM-MSCs) or primary chondrocytes. However, their use raises some issues partially circumvented by the availability of Adipose tissue-derived MSCs. Herein; we characterized the chondrogenic potential of human Multipotent Adipose-Derived Stem (hMADS) cells, and their potential use as pharmacological tool. hMADS cells are able to synthesize matrix proteins including COMP, Aggrecan and type II Collagen. Furthermore, hMADS cells express BMP receptors in a similar manner to BM-MSC, and BMP6 treatment of differentiated cells prevents expression of the hypertrophic marker type X Collagen. We tested whether IL-1ß and nicotine could impact chondrocyte differentiation. As expected, IL-1ß induced ADAMTS-4 gene expression and modulated negatively chondrogenesis while these effects were reverted in the presence of the IL-1 receptor antagonist. Nicotine, at concentrations similar to those observed in blood of smokers, exhibited a dose dependent increase of Aggrecan expression, suggesting an unexpected protective effect of the drug under these conditions. Therefore, hMADS cells represent a valuable tool for the analysis of in vitro chondrocyte differentiation and to screen for potentially interesting pharmacological drugs.


Asunto(s)
Tejido Adiposo/citología , Condrocitos/citología , Condrogénesis/fisiología , Células Madre Multipotentes/citología , Proteínas ADAM/genética , Proteína ADAMTS4 , Agrecanos/biosíntesis , Proteína Morfogenética Ósea 6/farmacología , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Separación Celular , Condrogénesis/genética , Colágeno Tipo X/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Interleucina-1beta/farmacología , Células Madre Multipotentes/efectos de los fármacos , Células Madre Multipotentes/metabolismo , Nicotina/farmacología , Procolágeno N-Endopeptidasa/genética
5.
J Exp Med ; 201(9): 1397-405, 2005 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-15867092

RESUMEN

Here, we report the isolation of a human multipotent adipose-derived stem (hMADS) cell population from adipose tissue of young donors. hMADS cells display normal karyotype; have active telomerase; proliferate >200 population doublings; and differentiate into adipocytes, osteoblasts, and myoblasts. Flow cytometry analysis indicates that hMADS cells are CD44+, CD49b+, CD105+, CD90+, CD13+, Stro-1(-), CD34-, CD15-, CD117-, Flk-1(-), gly-A(-), CD133-, HLA-DR(-), and HLA-I(low). Transplantation of hMADS cells into the mdx mouse, an animal model of Duchenne muscular dystrophy, results in substantial expression of human dystrophin in the injected tibialis anterior and the adjacent gastrocnemius muscle. Long-term engraftment of hMADS cells takes place in nonimmunocompromised animals. Based on the small amounts of an easily available tissue source, their strong capacity for expansion ex vivo, their multipotent differentiation, and their immune-privileged behavior, our results suggest that hMADS cells will be an important tool for muscle cell-mediated therapy.


Asunto(s)
Tejido Adiposo/citología , Diferenciación Celular , Distrofina/metabolismo , Regulación de la Expresión Génica , Inmunocompetencia/fisiología , Células Madre Multipotentes/trasplante , Animales , Niño , Preescolar , Cartilla de ADN , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Lactante , Cariotipificación , Masculino , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo
6.
J Lipid Res ; 51(8): 2352-61, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20410018

RESUMEN

The prevalence of obesity has steadily increased over the last few decades. During this time, populations of industrialized countries have been exposed to diets rich in fat with a high content of linoleic acid and a low content of alpha-linolenic acid compared with recommended intake. To assess the contribution of dietary fatty acids, male and female mice fed a high-fat diet (35% energy as fat, linoleic acid:alpha-linolenic acid ratio of 28) were mated randomly and maintained after breeding on the same diet for successive generations. Offspring showed, over four generations, a gradual enhancement in fat mass due to combined hyperplasia and hypertrophy with no change in food intake. Transgenerational alterations in adipokine levels were accompanied by hyperinsulinemia. Gene expression analyses of the stromal vascular fraction of adipose tissue, over generations, revealed discrete and steady changes in certain important players, such as CSF3 and Nocturnin. Thus, under conditions of genome stability and with no change in the regimen over four generations, we show that a Western-like fat diet induces a gradual fat mass enhancement, in accordance with the increasing prevalence of obesity observed in humans.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Dieta , Grasas de la Dieta/farmacología , Mundo Occidental , Adipoquinas/sangre , Tejido Adiposo/citología , Animales , Peso Corporal/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Humanos , Hiperplasia/etiología , Hiperplasia/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Obesidad/etiología , Obesidad/metabolismo , Fenotipo , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Factores de Tiempo , Ácido alfa-Linolénico/metabolismo , Ácido alfa-Linolénico/farmacología
7.
J Biol Chem ; 284(27): 18282-91, 2009 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-19433586

RESUMEN

Lipolysis is the catabolic pathway by which triglycerides are hydrolyzed into fatty acids. Adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL) have the capacity to hydrolyze in vitro the first ester bond of triglycerides, but their respective contributions to whole cell lipolysis in human adipocytes is unclear. Here, we have investigated the roles of HSL, ATGL, and its coactivator CGI-58 in basal and forskolin-stimulated lipolysis in a human white adipocyte model, the hMADS cells. The hMADS adipocytes express the various components of fatty acid metabolism and show lipolytic capacity similar to primary cultured adipocytes. We show that lipolysis and fatty acid esterification are tightly coupled except in conditions of stimulated lipolysis. Immunocytochemistry experiments revealed that acute forskolin treatment promotes HSL translocation from the cytosol to small lipid droplets and redistribution of ATGL from the cytosol and large lipid droplets to small lipid droplets, resulting in enriched colocalization of the two lipases. HSL or ATGL overexpression resulted in increased triglyceride-specific hydrolase capacity, but only ATGL overexpression increased whole cell lipolysis. HSL silencing had no effect on basal lipolysis and only partially reduced forskolin-stimulated lipolysis. Conversely, silencing of ATGL or CGI-58 significantly reduced basal lipolysis and essentially abolished forskolin-stimulated lipolysis. Altogether, these results suggest that ATGL/CGI-58 acts independently of HSL and precedes its action in the sequential hydrolysis of triglycerides in human hMADS adipocytes.


Asunto(s)
Adipocitos/enzimología , Metabolismo Energético/fisiología , Lipasa/metabolismo , Lipólisis/fisiología , Esterol Esterasa/metabolismo , 1-Acilglicerol-3-Fosfato O-Aciltransferasa , Adipocitos/citología , Adipocitos/efectos de los fármacos , Células Cultivadas , Colforsina/farmacología , Citosol/enzimología , Esterificación/fisiología , Ácidos Grasos/metabolismo , Proteínas Fluorescentes Verdes/genética , Humanos , Hidrólisis , Lipasa/genética , ARN Interferente Pequeño , Esterol Esterasa/genética
8.
Stem Cells ; 27(11): 2753-60, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19697348

RESUMEN

In contrast to the earlier contention, adult humans have been shown recently to possess active brown adipose tissue with a potential of being of metabolic significance. Up to now, brown fat precursor cells have not been available for human studies. We have shown previously that human multipotent adipose-derived stem (hMADS) cells exhibit a normal karyotype and high self-renewal ability; they are known to differentiate into cells that exhibit the key properties of human white adipocytes, that is, uncoupling protein two expression, insulin-stimulated glucose uptake, lipolysis in response to beta-agonists and atrial natriuretic peptide, and release of adiponectin and leptin. Herein, we show that, upon chronic exposure to a specific PPARgamma but not to a PPARbeta/delta or a PPARalpha agonist, hMADS cell-derived white adipocytes are able to switch to a brown phenotype by expressing both uncoupling protein one (UCP1) and CIDEA mRNA. This switch is accompanied by an increase in oxygen consumption and uncoupling. The expression of UCP1 protein is associated to stimulation of respiration by beta-AR agonists, including beta3-AR agonist. Thus, hMADS cells represent an invaluable cell model to screen for drugs stimulating the formation and/or the uncoupling capacity of human brown adipocytes that could help to dissipate excess caloric intake of individuals.


Asunto(s)
Adipocitos Marrones/citología , Tejido Adiposo Blanco/citología , Diferenciación Celular , Células Madre Multipotentes/citología , Adipocitos Marrones/efectos de los fármacos , Adipocitos Marrones/metabolismo , Andrógenos , Western Blotting , Diferenciación Celular/efectos de los fármacos , Línea Celular , Respiración de la Célula/efectos de los fármacos , Células Cultivadas , Preescolar , Humanos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Masculino , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Células Madre Multipotentes/efectos de los fármacos , Células Madre Multipotentes/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Receptores Adrenérgicos beta 3/genética , Receptores Adrenérgicos beta 3/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rosiglitazona , Tiazolidinedionas/farmacología , Proteína Desacopladora 1
9.
Cells ; 9(11)2020 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-33171828

RESUMEN

Thermogenic brown and brite adipocytes convert chemical energy from nutrients into heat. Therapeutics that regulate brown adipocyte recruitment and activity represent interesting strategies to control fat mass such as in obesity or cachexia. The peroxisome proliferator-activated receptor (PPAR) family plays key roles in the maintenance of adipose tissue and in the regulation of thermogenic activity. Activation of these receptors induce browning of white adipocyte. The purpose of this work was to characterize the role of carnosic acid (CA), a compound used in traditional medicine, in the control of brown/brite adipocyte formation and function. We used human multipotent adipose-derived stem (hMADS) cells differentiated into white or brite adipocytes. The expression of key marker genes was determined using RT-qPCR and western blotting. We show here that CA inhibits the browning of white adipocytes and favors decreased gene expression of thermogenic markers. CA treatment does not affect ß-adrenergic response. Importantly, the effects of CA are fully reversible. We used transactivation assays to show that CA has a PPARα/γ antagonistic action. Our data pinpoint CA as a drug able to control PPAR activity through an antagonistic effect. These observations shed some light on the development of natural PPAR antagonists and their potential effects on thermogenic response.


Asunto(s)
Abietanos/farmacología , Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Receptores Activados del Proliferador del Peroxisoma/antagonistas & inhibidores , Rosmarinus/química , Adipocitos Beige/efectos de los fármacos , Adipocitos Beige/metabolismo , Adipocitos Marrones/efectos de los fármacos , Adipocitos Blancos/efectos de los fármacos , Animales , Biomarcadores/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Lipólisis/efectos de los fármacos , Ratones , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Rosiglitazona/farmacología , Termogénesis/efectos de los fármacos , Termogénesis/genética
10.
Biochem Biophys Res Commun ; 390(2): 247-51, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-19800867

RESUMEN

Obesity has emerged as a global health problem with more than 1.1 billion adults to be classified as overweight or obese, and is associated with type 2 diabetes, cardiovascular disease, and several cancers. Since obesity is characterized by an increased size and/or number of adipocytes, elucidating the molecular events governing adipogenesis is of utmost importance. Recent findings indicate that microRNAs (miRNAs) - small non-protein-coding RNAs that function as post-transcriptional gene regulators - are involved in the regulatory network of adipogenesis. Whereas only a single human miRNA is known so far to be functional in adipogenesis as pro-adipogenic, several mouse miRNAs have been identified very recently as adipogenic regulators, thereby stimulating demand for studying the functional role of miRNAs during adipogenesis in human. Here, we demonstrate that miR-27b abundance decreased during adipogenesis of human multipotent adipose-derived stem (hMADS) cells. Overexpression of miR-27b blunted induction of PPARgamma and C/EBPalpha, two key regulators of adipogenesis, during early onset of adipogenesis and repressed adipogenic marker gene expression and triglyceride accumulation at late stages. PPARgamma has a predicted and highly conserved binding site in its 3'UTR and was indeed confirmed to be a direct target of miR-27b. Thus, these results suggest that the anti-adipogenic effect of miR-27b in hMADS cells is due, at least in part, to suppression of PPARgamma.


Asunto(s)
Adipocitos/fisiología , Adipogénesis/genética , Regulación de la Expresión Génica , MicroARNs/metabolismo , PPAR gamma/metabolismo , Regiones no Traducidas 3'/metabolismo , Adipocitos/metabolismo , Animales , Secuencia de Bases , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Humanos , Ratones , PPAR gamma/genética
11.
Stem Cells ; 26(9): 2399-407, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18583541

RESUMEN

Osteoporosis constitutes a major worldwide public health burden characterized by enhanced skeletal fragility. Bone metabolism is the combination of bone resorption by osteoclasts and bone formation by osteoblasts. Whereas increase in bone resorption is considered as the main contributor of bone loss that may lead to osteoporosis, this loss is accompanied by increased bone marrow adiposity. Osteoblasts and adipocytes share the same precursor cell and an inverse relationship exists between the two lineages. Therefore, identifying signaling pathways that stimulate mesenchymal stem cells osteogenesis at the expense of adipogenesis is of major importance for developing new therapeutic treatments. For this purpose, we identified by transcriptomic analysis the oxytocin receptor pathway as a potential regulator of the osteoblast/adipocyte balance of human multipotent adipose-derived stem (hMADS) cells. Both oxytocin (OT) and carbetocin (a stable OT analogue) negatively modulate adipogenesis while promoting osteogenesis in both hMADS cells and human bone marrow mesenchymal stromal cells. Consistent with these observations, ovariectomized (OVX) mice and rats, which become osteoporotic and exhibit disequilibrium of this balance, have significant decreased OT levels compared to sham-operated controls. Subcutaneous OT injection reverses bone loss in OVX mice and reduces marrow adiposity. Clinically, plasma OT levels are significantly lower in postmenopausal women developing osteoporosis than in their healthy counterparts. Taken together, these results suggest that plasma OT levels represent a novel diagnostic marker for osteoporosis and that OT administration holds promise as a potential therapy for this disease.


Asunto(s)
Células Madre Mesenquimatosas/patología , Osteoporosis/patología , Oxitocina/fisiología , Adipogénesis , Anciano , Anciano de 80 o más Años , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Células Cultivadas , Preescolar , Femenino , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Persona de Mediana Edad , Osteoblastos/metabolismo , Osteoblastos/patología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteoclastos/patología , Osteogénesis , Osteoporosis/metabolismo , Osteoporosis/terapia , Osteoporosis Posmenopáusica/sangre , Ovariectomía , Oxitocina/análogos & derivados , Oxitocina/sangre , Oxitocina/farmacología , Ratas , Receptores de Oxitocina/metabolismo
12.
BMC Genomics ; 9: 340, 2008 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-18637193

RESUMEN

BACKGROUND: A reciprocal relationship between bone and fat development in osteoporosis is clinically well established. Some of the key molecular regulators involved in this tissue replacement process have been identified. The detailed mechanisms governing the differentiation of mesenchymal stem cells (MSC) - the key cells involved - are however only now beginning to emerge. In an attempt to address the regulation of the adipocyte/osteoblast balance at the level of gene transcription in a comprehensive and unbiased manner, we performed a large-scale gene expression profiling study using a unique cellular model, human multipotent adipose tissue-derived stem cells (hMADS). RESULTS: The analysis of 1606 genes that were found to be differentially expressed between adipogenesis and osteoblastogenesis revealed gene repression to be most prevalent prior to commitment in both lineages. Computational analyses suggested that this gene repression is mediated by miRNAs. The transcriptional activation of lineage-specific molecular processes in both cases occurred predominantly after commitment. Analyses of the gene expression data and promoter sequences produced a set of 65 genes that are candidates for genes involved in the process of adipocyte/osteoblast commitment. Four of these genes were studied in more detail: LXRalpha and phospholipid transfer protein (PLTP) for adipogenesis, the nuclear receptor COUP-TF1 and one uncharacterized gene, TMEM135 for osteoblastogenesis. PLTP was secreted during both early and late time points of hMADS adipocyte differentiation. LXRalpha, COUP-TF1, and the transmembrane protein TMEM135 were studied in primary cultures of differentiating bone marrow stromal cells from healthy donors and were found to be transcriptionally activated in the corresponding lineages. CONCLUSION: Our results reveal gene repression as a predominant early mechanism before final cell commitment. We were moreover able to identify 65 genes as candidates for genes controlling the adipocyte/osteoblast balance and to further evaluate four of these. Additional studies will explore the precise role of these candidate genes in regulating the adipogenesis/osteoblastogenesis switch.


Asunto(s)
Adipogénesis/genética , Perfilación de la Expresión Génica , Células Madre Multipotentes/metabolismo , Osteoblastos/metabolismo , Regiones no Traducidas 3'/genética , Línea Celular , Linaje de la Célula , Biología Computacional , Regulación hacia Abajo , Regulación del Desarrollo de la Expresión Génica , Humanos , MicroARNs/metabolismo , Modelos Genéticos , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
13.
BMC Mol Biol ; 9: 26, 2008 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-18302751

RESUMEN

BACKGROUND: It is well established that adipose tissue plays a key role in energy storage and release but is also a secretory organ and a source of stem cells. Among different lineages, stem cells are able to differentiate into adipocytes and osteoblasts. As secreted proteins could regulate the balance between both lineages, we aimed at characterizing the secretome of human multipotent adipose-derived stem cell (hMADS) at an early step of commitment to adipocytes and osteoblasts. RESULTS: A proteomic approach, using mono-dimensional electrophoresis and tandem mass spectrometry, allowed us to identify a total of 73 proteins at day 0 and day 3 of adipocyte and osteoblast differentiation. Analysis of identified proteins showed that 52 % corresponded to classical secreted proteins characterized by a signal peptide, that 37 % previously described in the extracellular compartment were devoid of signal peptide and that 11 % neither exhibited a signal peptide nor had been previously described extracellularly. These proteins were classified into 8 clusters according to their function. Quantitative analysis has been performed for 8 candidates: PAI-1, PEDF, BIGH3, PTX3, SPARC, ENO1, GRP78 and MMP2. Among them, PAI-1 was detected at day 0 and day 3 of osteoblast differentiation but never in adipocyte secretome. Furthermore we showed that PAI-1 mRNA was down-regulated in the bone of ovariectomized mice. CONCLUSION: Given its regulation during the early events of hMADS cell differentiation and its status in ovariectomized mice, PAI-1 could play a role in the adipocyte/osteoblast balance and thus in bone diseases such as osteoporosis.


Asunto(s)
Adipocitos/fisiología , Diferenciación Celular , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/fisiología , Proteoma/análisis , Animales , Células Cultivadas , Chaperón BiP del Retículo Endoplásmico , Femenino , Humanos , Masculino , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos C57BL , Ovariectomía , Plasminógeno/fisiología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Factores de Tiempo
14.
Biochem Biophys Res Commun ; 374(1): 64-8, 2008 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-18611392

RESUMEN

Stathmin-like 2 (STMN2) protein, a neuronal protein of the stathmin family, has been implicated in the microtubule regulatory network as a crucial element of cytoskeletal regulation. Herein, we describe that STMN2 expression increases at both mRNA and protein levels during osteogenesis of human mesenchymal stem cells derived from adipose tissue (hMADS cells) and bone marrow (hBMS cells), whereas it decreases to undetectable levels during adipogenesis. STMN2 protein is localized in both Golgi and cytosolic compartments. Its expression appears modulated in osteoblasts by nerve growth factor, dexamethasone or RhoA kinase inhibitor Y-27632 which are known effectors of osteogenesis. Thus STMN2 appears a novel marker of osteogenesis and osteoblast per se, that could play a role in the regulation of the adipocyte/osteoblast balance.


Asunto(s)
Proteínas de la Membrana/biosíntesis , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Adipocitos/metabolismo , Adipogénesis , Tejido Adiposo/metabolismo , Amidas/farmacología , Biomarcadores/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Citosol/metabolismo , Dexametasona/farmacología , Aparato de Golgi/metabolismo , Humanos , Proteínas de la Membrana/genética , Células Madre Mesenquimatosas/citología , Neuronas/metabolismo , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Estatmina , Quinasas Asociadas a rho/antagonistas & inhibidores
15.
Arterioscler Thromb Vasc Biol ; 27(6): 1340-5, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17363694

RESUMEN

OBJECTIVES: This study examines the role of insulin and angiotensin II in high-density lipoprotein (HDL) metabolism by focusing on the regulation and function of scavenger receptor type-BI (SR-BI) in adipose tissue. METHODS AND RESULTS: Insulin or angiotensin II injection in wild-type mice induced a decrease in circulating HDL and it was associated with the translocation of SR-BI from intracellular sites to the plasma membrane of adipose tissue. Refeeding upregulated adipose HDL selective cholesteryl esters uptake and SR-BI proteins through transcriptional and posttranscriptional mechanisms. This occurred along with a decrease in serum HDL and an increase in adipose cholesterol content. Similar results were obtained with transgenic mice overexpressing locally angiotensinogen in adipose tissue. In adipose 3T3-L1 cell line, HDL induced lipogenesis by increasing liver X receptor binding activity. This mechanism was dependent of insulin and angiotensin II. CONCLUSIONS: Our results raise the possibility that adipose tissue SR-BI translocation might be a link between adipose tissue lipid storage and HDL clearance.


Asunto(s)
Tejido Adiposo/metabolismo , Adiposidad , Angiotensina II/metabolismo , HDL-Colesterol/metabolismo , Insulina/metabolismo , Lipogénesis , Receptores Depuradores de Clase B/metabolismo , Células 3T3-L1 , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Adiposidad/efectos de los fármacos , Adiposidad/genética , Angiotensina II/farmacología , Angiotensinógeno/genética , Angiotensinógeno/metabolismo , Animales , Membrana Celular/metabolismo , Colesterol/análogos & derivados , Colesterol/metabolismo , HDL-Colesterol/sangre , Proteínas de Unión al ADN/metabolismo , Ingestión de Alimentos , Epidídimo/metabolismo , Homeostasis , Insulina/farmacología , Lipogénesis/efectos de los fármacos , Lipogénesis/genética , Receptores X del Hígado , Masculino , Ratones , Ratones Transgénicos , Receptores Nucleares Huérfanos , Transporte de Proteínas , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Depuradores de Clase B/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Factores de Tiempo , Transcripción Genética , Triglicéridos/metabolismo
16.
Br J Nutr ; 100(3): 461-70, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18307824

RESUMEN

A positive energy balance (energy intake>energy expenditure), in which total fat intake plays an important role, is commonly regarded as a major factor contributing to obesity. Adipose tissue development, i.e. both size (hypertrophy) and number (hyperplasia), is stimulated by high dietary fat intake during early postnatal development, a susceptibility that now appears to continue well into adulthood. Recent human and animal studies suggest that by altering rates of adipocyte differentiation and proliferation, differences in the composition of dietary fat may also contribute to adipose tissue development. At least in rodent models, the relative intake of n-6 to n-3 PUFA is clearly emerging as a new factor in this development. In these models, higher linoleate intake raises tissue arachidonic acid, which increases prostacyclin production and, in turn, stimulates signalling pathways implicated in adipogenesis. Signalling pathways stimulated by arachidonic acid probably include phospholipase and/or cyclo-oxygenase activation and may be linked as much to relatively low intake of n-3 PUFA as to excessive dietary linoleate. One factor potentially contributing to oversight about the apparent role of dietary n-6 PUFA (especially excess dietary linoleate) in adipose tissue development is the historical overestimation of linoleate requirements and the enthusiasm for higher intake of 'essential fatty acids'. More research is needed to address whether disequilibration of dietary PUFA intake contributes to the risk of obesity in humans.


Asunto(s)
Adipogénesis/fisiología , Grasas de la Dieta/efectos adversos , Ácidos Grasos Omega-6/efectos adversos , Obesidad/etiología , Composición Corporal , Niño , Grasas de la Dieta/metabolismo , Ácidos Grasos Omega-6/metabolismo , Humanos , Obesidad/metabolismo , Factores de Riesgo , Estados Unidos
17.
Prog Lipid Res ; 45(3): 203-36, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16516300

RESUMEN

The importance of a high fat intake in the increasing prevalence of childhood and adult obesity remains controversial. Moreover, qualitative changes (i.e. the fatty acid composition of fats) have been largely disregarded. Herein is reviewed the role of polyunsaturated fatty acids (PUFAs) of the n-6 series in promoting adipogenesis in vitro and favouring adipose tissue development in rodents during the gestation/suckling period. Epidemiological data from infant studies as well as the assessment of the fatty acid composition of mature breast milk and infant formulas over the last decades in the Western industrialized world are revisited and appear consistent with animal data. Changes over decades in the intake of n-6 and n-3 PUFAs, with a striking increase in the linoleic acid/alpha-linolenic ratio, are observed. In adults, using a consumption model based upon production data, similar changes in the PUFA content of ingested lipids have been found for France, and are associated with an increase of fat consumption over the last 40 years. These profound quantitative and qualitative alterations can be traced in the food chain and shown to be due to changes in human dietary habits as well as in the feeding pattern of breeding stock. If prevention of obesity is a key issue for future generations, agricultural and food industry policies should be thoroughly reevaluated.


Asunto(s)
Tejido Adiposo/crecimiento & desarrollo , Ácidos Grasos Omega-6/administración & dosificación , Obesidad/etiología , Adipogénesis/fisiología , Adulto , Animales , Grasas de la Dieta/administración & dosificación , Ácidos Grasos Omega-3/administración & dosificación , Conducta Alimentaria , Humanos , Lactante , Fenómenos Fisiológicos Nutricionales del Lactante , Recién Nacido , Leche Humana/química , Obesidad/fisiopatología
18.
Chem Biodivers ; 5(8): 1513-1520, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18729088

RESUMEN

Auranofin is a gold-based antiarthritic drug in clinical use for more that 25 years. However, in spite of a long established use, its specific effects on bone metabolism are still greatly controversial. We have analyzed in vitro the actions of auranofin on human multipotent adipose-derived stem (hMADS) cells, used as a model for bone metabolism, since these cells were reported to undergo osteogenesis both in vitro and in vivo. Cytotoxicity of auranofin on hMADS cells, differentiated into osteoblasts, was initially assessed. Thereafter, the consequences of exposure to nontoxic but clinically relevant auranofin concentrations were analyzed by monitoring the seleno-protein glutathione peroxidase 3 or alkaline phosphatase, a characteristic biomarker of osteogenesis. Notably, we found that chronic treatment with auranofin alters only weakly the levels of alkaline phosphatase, thus implying an overall modest effect on osteogenesis. In contrast, auranofin turned out to greatly affect glutathione peroxidase 3 activity. The possible medical implications of these findings are discussed.


Asunto(s)
Tejido Adiposo/citología , Artritis/tratamiento farmacológico , Auranofina/farmacología , Modelos Biológicos , Osteogénesis/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos , Auranofina/química , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Preescolar , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Perfilación de la Expresión Génica , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Humanos , Masculino , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Biochimie ; 136: 3-11, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28034718

RESUMEN

Oxylipins are bioactive metabolites derived from the oxygenation of ω3 and ω6 polyunsaturated fatty acids, triggered essentially by cyclooxygenase and lipoxygenase activities. Oxylipins are involved in the development and function of adipose tissue and their productions are strictly related to diet quality and quantity. Oxylipins signal via cell surface membrane (G Protein-coupled receptors) and nuclear receptors (peroxisome proliferator-activated receptors), two pathways playing a pivotal role in adipocyte biology. In this review, we made an attempt to cover the available knowledge about synthesis and molecular function of oxylipins known to modulate adipogenesis, adipocyte function and phenotype conversion, with a focus on their interaction with peroxisome proliferator-activated nuclear receptor family.


Asunto(s)
Adipogénesis/fisiología , Oxilipinas/metabolismo , Receptores Activados del Proliferador del Peroxisoma/fisiología , Receptores Acoplados a Proteínas G/fisiología , Animales , Humanos
20.
C R Biol ; 329(8): 570-7; discussion 653-5, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16860275

RESUMEN

Adipose tissue contains various types of cells that include preadipocytes and adipocytes. Studies have emphasized that (i) preadipocytes secrete factors involved in their own differentiation and (ii) adipocytes acquire the ability to communicate systemically with other organs (brain, liver, skeletal muscle) and locally with other cells (preadipocytes, endothelial cells and monocytes/macrophages). Adipocytes secrete proteins exhibiting either beneficial (leptin, adiponectin) or deleterious effects (angiotensinogen). Associated to the effect of secretory products from macrophages (cytokines), a disturbance in the balance between these various secreted factors leads to the development of a metabolic syndrome.


Asunto(s)
Tejido Adiposo/fisiología , Tejido Adiposo/fisiopatología , Síndrome Metabólico/fisiopatología , Adipocitos/citología , Adipocitos/fisiología , Adiponectina/fisiología , Tejido Adiposo/citología , Angiotensinógeno/fisiología , Comunicación Celular , Humanos , Leptina/fisiología , Macrófagos/citología , Macrófagos/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA