12 research outputs found

    Identification et caractérisation de VSTM2A comme un nouveau facteur modulant l'adipogenèse

    Get PDF
    Aujourd’hui encore, l’obésité touche de plus en plus d’individus, et sa complexité reflète son aspect multifactoriel. Parmi les facteurs impliqués dans le développement de l'obésité, on compte la génétique qui expliquerait notamment la susceptibilité de certains êtres humains à devenir obèse. D'un point de vue moléculaire, la cascade adipogénique permettant à des préadipocytes de devenir des adipocytes matures est un phénomène dynamique et bien caractérisé. En revanche, les facteurs définissant le potentiel adipogénique des préadipocytes ainsi que le renouvellement des adipocytes demeurent encore inconnus. Notre objectif a été d’identifier et de caractériser de nouveaux gènes définissant l’identité des préadipocytes et pouvant influencer le développement du tissu adipeux. Par dilutions sériées d’une culture de 3T3-L1, nous avons isolé et amplifié 23 nouvelles lignées cellulaires que nous avons classées selon leur potentiel adipogénique. Une puce à ADN nous a permis d’identifier les gènes s’exprimant différemment entre les lignées à fort et à faible potentiel adipogénique. Nous avons identifié un nouveau gène inconnu de la littérature nommé V-set and transmembrane domain containing 2A (Vstm2a). Les travaux réalisés dans le cadre de ma thèse ont été de comprendre l’implication de ce gène dans le développement de l’adipocyte in vitro et dans la formation du tissu adipeux in vivo. Nous avons d’abord identifié Vstm2a comme le gène le plus différentiellement exprimé entre les différentes lignées cellulaires, son expression étant très élevée dans les lignées à fort potentiel adipogénique. Une corrélation positive s’est dessinée entre l’expression de Vstm2a et Peroxisome Proliferator-Activated Receptor γ2 (Pparγ2) dans les préadipocytes. Lors du développement de l’adipocyte in vitro, l’expression de Vstm2a précède toujours l’activation de Pparγ2, un facteur clé régulant l’adipogenèse. Cette expression précoce est retrouvée in vivo, lors de la formation et de l’expansion du tissu adipeux. Les cellules exprimant Vstm2a au stade développemental se retrouvent à proximité de vaisseaux sanguins et de cellules musculaires, une caractéristique de précurseurs adipeux. Chez l’adulte, ces cellules s’apparentent à la population de précurseurs adipeux identifiée à partir des marqueurs de surface LIN⁻CD29⁺CD34⁺SCA1⁺PDGFRα⁺. Nous avons découvert que VSTM2A est une protéine glycosylée et abondamment sécrétée par des cellules ayant un fort potentiel adipogénique. La surexpression de VSTM2A intracellulaire suffit à favoriser la différenciation adipocytaire de cellules fibroblastiques, entre autres par l’induction de Pparγ2. À l’inverse, sa sous-expression dans les préadipocytes entraine un défaut adipogénique, réduisant l’expression de Pparγ2. Nous avons montré que VSTM2A agit en amplifiant la réponse aux Bone Morphogenetic Protein (BMP) favorisant ainsi l’expression de Pparγ2. Nous proposons un modèle dans lequel VSTM2A est essentiel pour maintenir et amplifier le potentiel adipogénique des préadipocytes. Une meilleure compréhension des facteurs régulant les phases précoces de l'adipogenèse pourrait permettre le développement de nouveaux outils régulant la formation du tissu adipeux.Obesity affects the life of millions of humans worldwide and is the result of complex interactions between genes and environment. Factors involved in the development of obesity include genetics, which may explain why some humans are more susceptible to become obese. From a molecular point of view, the adipogenic cascade allowing preadipocytes to differentiate into mature adipocytes is a dynamic and well characterized phenomenon. However, the factors defining the adipogenic potential of the preadipocytes as well as the renewal of the adipocytes remain unknown. Our objective was to identify and characterize new genes defining the identity of preadipocytes capable of influencing the development of adipose tissue. By performing serial dilutions of a culture of 3T3-L1 preadipocytes, we have isolated and amplified 23 new cell lines that we have classified according to their adipogenic potential. A microarray allowed us to identify the genes differently expressed between high and low adipogenic lines. We have identified a new gene, unknown from the literature, named V-set and transmembrane domain containing 2A (Vstm2a). The objective of my thesis was to understand the implication of this gene in the development of the adipocytes in vitro and in the formation of adipose tissue in vivo. We first identified that Vstm2a was the most differentially expressed gene between the Low and High adipogenic lines, its expression being elevated in lines with a high adipogenic potential. A positive correlation emerged between the expression of Vstm2a and Pparγ2 in the preadipocytes. During adipocyte development in vitro, the expression of Vstm2a always precedes PPARγ2 activation, a key factor regulating adipogenesis. This expression pattern was also found in vivo during adipose tissue formation and expansion. VSTM2A-expressing cells at the developmental stage are found in the vicinity of the vasculature and muscle cells, a characteristic of adipose precursors. In adults, Vstm2a expression is enriched in the adipose precursor population identified with the surface markers LIN⁻CD29⁺CD34⁺SCA1⁺PDGFRα⁺. We found that VSTM2A is a glycosylated protein that is abundantly secreted by cells with a high adipogenic potential. Overexpression of intracellular VSTM2A is sufficient to promote the adipogenic potential of fibroblastic cells, an effect associated with a rise in basal Pparγ2 expression. Conversely, VSTM2A knockdown impairs adipogenesis by reducing the expression of Pparγ2. We found that VSTM2A promotes Pparγ2 expression by amplifying BMP signaling. Here, we propose a model in which VSTM2A is expressed to maintain and amplify the adipogenic potential of adipose precursors. A better understanding of the factors regulating early steps in adipogenesis could allow the development of new tools to regulate adipose tissue formation

    Amplification of Adipogenic Commitment by VSTM2A

    Get PDF
    Despite progress in our comprehension of the mechanisms regulating adipose tissue development, the nature of the factors that functionally characterize adipose precursors is still elusive. Defining the early steps regulating adipocyte development is needed for the generation of tools to control adipose tissue size and function. Here, we report the discovery of V-set and transmembrane domain containing 2A (VSTM2A) as a protein expressed and secreted by committed preadipocytes. VSTM2A expression is elevated in the early phases of adipogenesis in vitro and adipose tissue development in vivo. We show that VSTM2A-producing cells associate with the vasculature and express the common surface markers of adipocyte progenitors. Overexpression of VSTM2A induces adipogenesis, whereas its depletion impairs this process. VSTM2A controls preadipocyte determination at least in part by modulating BMP signaling and PPARgamma2 activation. We propose a model in which VSTM2A is produced to preserve and amplify the adipogenic capability of adipose precursors

    Loss of hepatic DEPTOR alters the metabolic transition to fasting

    Get PDF
    Objective The mechanistic target of rapamycin (mTOR) is a serine/threonine kinase that functions into distinct protein complexes (mTORC1 and mTORC2) that regulates growth and metabolism. DEP-domain containing mTOR-interacting protein (DEPTOR) is part of these complexes and is known to reduce their activity. Whether DEPTOR loss affects metabolism and organismal growth in vivo has never been tested. Methods We have generated a conditional transgenic mouse allowing the tissue-specific deletion of DEPTOR. This model was crossed with CMV-cre mice or Albumin-cre mice to generate either whole-body or liver-specific DEPTOR knockout (KO) mice. Results Whole-body DEPTOR KO mice are viable, fertile, normal in size, and do not display any gross physical and metabolic abnormalities. To circumvent possible compensatory mechanisms linked to the early and systemic loss of DEPTOR, we have deleted DEPTOR specifically in the liver, a tissue in which DEPTOR protein is expressed and affected in response to mTOR activation. Liver-specific DEPTOR null mice showed a reduction in circulating glucose upon fasting versus control mice. This effect was not associated with change in hepatic gluconeogenesis potential but was linked to a sustained reduction in circulating glucose during insulin tolerance tests. In addition to the reduction in glycemia, liver-specific DEPTOR KO mice had reduced hepatic glycogen content when fasted. We showed that loss of DEPTOR cell-autonomously increased oxidative metabolism in hepatocytes, an effect associated with increased cytochrome c expression but independent of changes in mitochondrial content or in the expression of genes controlling oxidative metabolism. We found that liver-specific DEPTOR KO mice showed sustained mTORC1 activation upon fasting, and that acute treatment with rapamycin was sufficient to normalize glycemia in these mice. Conclusion We propose a model in which hepatic DEPTOR accelerates the inhibition of mTORC1 during the transition to fasting to adjust metabolism to the nutritional status. Keywords: DEPTOR; mTOR; Liver; Glucose; Fastin

    Cathepsin G activity lowers plasma LDL and reduces atherosclerosis

    Get PDF
    AbstractCathepsin G (CatG), a serine protease present in mast cells and neutrophils, can produce angiotensin-II (Ang-II) and degrade elastin. Here we demonstrate increased CatG expression in smooth muscle cells (SMCs), endothelial cells (ECs), macrophages, and T cells from human atherosclerotic lesions. In low-density lipoprotein (LDL) receptor-deficient (Ldlr–/–) mice, the absence of CatG reduces arterial wall elastin degradation and attenuates early atherosclerosis when mice consume a Western diet for 3months. When mice consume this diet for 6months, however, CatG deficiency exacerbates atherosclerosis in aortic arch without affecting lesion inflammatory cell content or extracellular matrix accumulation, but raises plasma total cholesterol and LDL levels without affecting high-density lipoprotein (HDL) or triglyceride levels. Patients with atherosclerosis also have significantly reduced plasma CatG levels that correlate inversely with total cholesterol (r=–0.535, P<0.0001) and LDL cholesterol (r=–0.559, P<0.0001), but not with HDL cholesterol (P=0.901) or triglycerides (P=0.186). Such inverse correlations with total cholesterol (r=–0.504, P<0.0001) and LDL cholesterol (r=–0.502, P<0.0001) remain significant after adjusting for lipid lowering treatments among this patient population. Human CatG degrades purified human LDL, but not HDL. This study suggests that CatG promotes early atherogenesis through its elastinolytic activity, but suppresses late progression of atherosclerosis by degrading LDL without affecting HDL or triglycerides

    The oncometabolite 2-hydroxyglutarate activates the mTOR signalling pathway

    Get PDF
    The identification of cancer-associated mutations in the tricarboxylic acid (TCA) cycle enzymes isocitrate dehydrogenases 1 and 2 (IDH1/2) highlights the prevailing notion that aberrant metabolic function can contribute to carcinogenesis. IDH1/2 normally catalyse the oxidative decarboxylation of isocitrate into α-ketoglutarate (αKG). In gliomas and acute myeloid leukaemias, IDH1/2 mutations confer gain-of-function leading to production of the oncometabolite R-2-hydroxyglutarate (2HG) from αKG. Here we show that generation of 2HG by mutated IDH1/2 leads to the activation of mTOR by inhibiting KDM4A, an αKG-dependent enzyme of the Jumonji family of lysine demethylases. Furthermore, KDM4A associates with the DEP domain-containing mTOR-interacting protein (DEPTOR), a negative regulator of mTORC1/2. Depletion of KDM4A decreases DEPTOR protein stability. Our results provide an additional molecular mechanism for the oncogenic activity of mutant IDH1/2 by revealing an unprecedented link between TCA cycle defects and positive modulation of mTOR function downstream of the canonical PI3K/AKT/TSC1-2 pathway

    Altered Lipid Metabolism Impairs Skeletal Muscle Force in Young Rats Submitted to a Short-Term High-Fat Diet

    Get PDF
    Obesity and ensuing disorders are increasingly prevalent in young populations. Prolonged exposure to high-fat diets (HFD) and excessive lipid accumulation were recently suggested to impair skeletal muscle functions in rodents. We aimed to determine the effects of a short-term HFD on skeletal muscle function in young rats. Young male Wistar rats (100–125 g) were fed HFD or a regular chow diet (RCD) for 14 days. Specific force, resistance to fatigue and recovery were tested in extensor digitorum longus (EDL; glycolytic) and soleus (SOL; oxidative) muscles using an ex vivo muscle contractility system. Muscle fiber typing and insulin signaling were analyzed while intramyocellular lipid droplets (LD) were characterized. Expression of key markers of lipid metabolism was also measured. Weight gain was similar for both groups. Specific force was decreased in SOL, but not in EDL of HFD rats. Muscle resistance to fatigue and force recovery were not altered in response to the diets. Similarly, muscle fiber type distribution and insulin signaling were not influenced by HFD. On the other hand, percent area and average size of intramyocellular LDs were significantly increased in the SOL of HFD rats. These effects were consistent with the increased expression of several mediators of lipid metabolism in the SOL muscle. A short-term HFD impairs specific force and alters lipid metabolism in SOL, but not EDL muscles of young rats. This indicates the importance of clarifying the early mechanisms through which lipid metabolism affects skeletal muscle functions in response to obesogenic diets in young populations

    Loss of Human Beta Cell Identity in a Reconstructed Omental Stromal Cell Environment

    No full text
    In human type 2 diabetes, adipose tissue plays an important role in disturbing glucose homeostasis by secreting factors that affect the function of cells and tissues throughout the body, including insulin-producing pancreatic beta cells. We aimed here at studying the paracrine effect of stromal cells isolated from subcutaneous and omental adipose tissue on human beta cells. We developed an in vitro model wherein the functional human beta cell line EndoC-&beta;H1 was treated with conditioned media from human adipose tissues. By using RNA-sequencing and western blotting, we determined that a conditioned medium derived from omental stromal cells stimulates several pathways, such as STAT, SMAD and RELA, in EndoC-&beta;H1 cells. We also observed that upon treatment, the expression of beta cell markers decreased while dedifferentiation markers increased. Loss-of-function experiments that efficiently blocked specific signaling pathways did not reverse dedifferentiation, suggesting the implication of more than one pathway in this regulatory process. Taken together, we demonstrate that soluble factors derived from stromal cells isolated from human omental adipose tissue signal human beta cells and modulate their identity

    Loss of Human Beta Cell Identity in a Reconstructed Omental Stromal Cell Environment

    No full text
    In human type 2 diabetes, adipose tissue plays an important role in disturbing glucose homeostasis by secreting factors that affect the function of cells and tissues throughout the body, including insulin-producing pancreatic beta cells. We aimed here at studying the paracrine effect of stromal cells isolated from subcutaneous and omental adipose tissue on human beta cells. We developed an in vitro model wherein the functional human beta cell line EndoC-βH1 was treated with conditioned media from human adipose tissues. By using RNA-sequencing and western blotting, we determined that a conditioned medium derived from omental stromal cells stimulates several pathways, such as STAT, SMAD and RELA, in EndoC-βH1 cells. We also observed that upon treatment, the expression of beta cell markers decreased while dedifferentiation markers increased. Loss-of-function experiments that efficiently blocked specific signaling pathways did not reverse dedifferentiation, suggesting the implication of more than one pathway in this regulatory process. Taken together, we demonstrate that soluble factors derived from stromal cells isolated from human omental adipose tissue signal human beta cells and modulate their identity

    Lung cancer susceptibility genetic variants modulate HOXB2 expression in the lung

    No full text
    The HOX genes are transcription factors that are expressed in coordinated spatiotemporal patterns to ensure normal development. Ectopic expression may instead lead to the development and progression of tumors. Genetic polymorphisms in the regions of four HOX gene clusters were tested for association with lung cancer in 420 cases and 3,151 controls. The effect of these variants on lung gene expression (expression quantitative trait loci, eQTL) was tested in a discovery set of 409 non-tumor lung samples and validated in two lung eQTL replication sets (n = 287 and 342). The expression levels of HOXB2 were evaluated at the mRNA and protein levels by quantitative real-time PCR and immunohistochemistry in paired tumor and non-tumor lung tissue samples. The most significant SNP associated with lung cancer in the HOXB cluster was rs10853100 located upstream of the HOXB cluster. HOXB2 was the top eQTL-regulated gene with several polymorphisms associated with its mRNA expression levels in lung tissue. This includes the lung cancer SNP rs10853100 that was significantly associated with HOXB2 expression (P=3.39E-7). In the lung eQTL discovery and replication sets, the lung cancer risk allele (T) for rs10853100 was associated with lower HOXB2 expression levels. In paired normal-tumor samples, HOXB2 mRNA and protein levels were significantly reduced in tumors when compared to non-tumor lung tissues. Genetic variants in the HOXB cluster may confer susceptibility to lung cancer by modulating the expression of HOXB2 in the lung
    corecore