Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Dominique Langin is active.

Publication


Featured researches published by Dominique Langin.


The FASEB Journal | 2004

Weight loss regulates inflammation-related genes in white adipose tissue of obese subjects

Karine Clément; Nathalie Viguerie; Christine Poitou; Claire Carette; Véronique Pelloux; Cyrile Anne Curat; Audrey Sicard; Sophie Rome; Arriel Benis; Jean Daniel Zucker; Hubert Vidal; Martine Laville; Gregory S. Barsh; Arnaud Basdevant; Vladimir Stich; Raffaella Cancello; Dominique Langin

Adipose tissue produces inflammation and immunity molecules suspected to be involved in obesity‐related complications. The pattern of expression and the nutritional regulation of these molecules in humans are poorly understood. We analyzed the gene expression profiles of subcutaneous white adipose tissue from 29 obese subjects during very low calorie diet (VLCD) using cDNA microarray and reverse transcription quantitative PCR. The patterns of expression were compared with that of 17 non‐obese subjects. We determined whether the regulated genes were expressed in adipocytes or stromavascular fraction cells. Gene expression profiling identified 100 inflammation‐related transcripts that are regulated in obese individuals when eating a 28 day VLCD but not a 2 day VLCD. Cluster analysis showed that the pattern of gene expression in obese subjects after 28 day VLCD was closer to the profile of lean subjects than to the pattern of obese subjects before VLCD. Weight loss improves the inflammatory profile of obese subjects through a decrease of proinflammatory factors and an increase of anti‐inflammatory molecules. The genes are expressed mostly in the stromavascular fraction of adipose tissue, which is shown to contain numerous macrophages. The beneficial effect of weight loss on obesity‐related complications may be associated with the modification of the inflammatory profile in adipose tissue.— Clément, K., Viguerie, N., Poitou, C., Carette, C., Pelloux, V., Curat, C. A., Sicard, A., Rome, S., Benis, A., Zucker, J.‐D., Vidal, H., Laville, M., Barsh, G. S., Basdevant, A., Stich, V., Cancello R., Langin, D. Weight loss regulates inflammation‐related genes in white adipose tissue of obese subjects. FASEB J. 18, 1657–1669 (2004)


Progress in Lipid Research | 2009

Lipolysis and lipid mobilization in human adipose tissue.

Max Lafontan; Dominique Langin

Triacylglycerol (TAG) stored in adipose tissue (AT) can be rapidly mobilized by the hydrolytic action of the three main lipases of the adipocyte. The non-esterified fatty acids (NEFA) released are used by other tissues during times of energy deprivation. Until recently hormone-sensitive lipase (HSL) was considered to be the key rate-limiting enzyme responsible for regulating TAG mobilization. A novel lipase named adipose triglyceride lipase/desnutrin (ATGL) has been identified as playing an important role in the control of fat cell lipolysis. Additionally perilipin and other proteins of the surface of the lipid droplets protecting or exposing the TAG core of the droplets to lipases are also potent regulators of lipolysis. Considerable progress has been made in understanding the mechanisms of activation of the various lipases. Lipolysis is under tight hormonal regulation. The best understood hormonal effects on AT lipolysis concern the opposing regulation by insulin and catecholamines. Heart-derived natriuretic peptides (i.e., stored in granules in the atrial and ventricle cardiomyocytes and exerting stimulating effects on diuresis and natriuresis) and numerous autocrine/paracrine factors originating from adipocytes and other cells of the stroma-vascular fraction may also participate in the regulation of lipolysis. Endocrine and autocrine/paracrine factors cooperate and lead to a fine regulation of lipolysis in adipocytes. Age, anatomical site, sex, genotype and species differences all play a part in the regulation of lipolysis. The manipulation of lipolysis has therapeutic potential in the metabolic disorders frequently associated with obesity and probably in several inborn errors of metabolism.


Journal of Biological Chemistry | 2003

Acquirement of Brown Fat Cell Features by Human White Adipocytes

Claire Tiraby; Geneviève Tavernier; Corinne Lefort; Dominique Larrouy; Frédéric Bouillaud; Daniel Ricquier; Dominique Langin

Obesity, i.e. an excess of white adipose tissue (WAT), predisposes to the development of type 2 diabetes and cardiovascular disease. Brown adipose tissue is present in rodents but not in adult humans. It expresses uncoupling protein 1 (UCP1) that allows dissipation of energy as heat. Peroxisome proliferator-activated receptor γ (PPARγ) and PPARγ coactivator 1α (PGC-1α) activate mouse UCP1 gene transcription. We show here that human PGC-1α induced the activation of the human UCP1 promoter by PPARγ. Adenovirus-mediated expression of human PGC-1α increased the expression of UCP1, respiratory chain proteins, and fatty acid oxidation enzymes in human subcutaneous white adipocytes. Changes in the expression of other genes were also consistent with brown adipocyte mRNA expression profile. PGC-1α increased the palmitate oxidation rate by fat cells. Human white adipocytes can therefore acquire typical features of brown fat cells. The PPARγ agonist rosiglitazone potentiated the effect of PGC-1α on UCP1 expression and fatty acid oxidation. Hence, PGC-1α is able to direct human WAT PPARγ toward a transcriptional program linked to energy dissipation. However, the response of typical white adipocyte targets to rosiglitazone treatment was not altered by PGC-1α. UCP1 mRNA induction was shown in vivo by injection of the PGC-1α adenovirus in mouse white fat. Alteration of energy balance through an increased utilization of fat in WAT may be a conceivable strategy for the treatment of obesity.


Journal of Clinical Investigation | 1997

Increased uncoupling protein-2 and -3 mRNA expression during fasting in obese and lean humans.

Laurence Millet; Hubert Vidal; Fabrizio Andreelli; Dominique Larrouy; J. P. Riou; D Ricquier; M. Laville; Dominique Langin

Uncoupling protein-2 and -3 (UCP2 and UCP3) are mitochondrial proteins that show high sequence homology with the brown adipocyte-specific UCP1. UCP1 induces heat production by uncoupling respiration from ATP synthesis. UCP2 is widely expressed in human tissues, whereas UCP3 expression seems restricted to skeletal muscle, an important site of thermogenesis in humans. We have investigated the regulation of UCP2 and UCP3 gene expression in skeletal muscle and adipose tissue from lean and obese humans. UCP2 and -3 mRNA levels were not correlated with body mass index (BMI) in skeletal muscle, but a positive correlation (r = 0.55, P < 0.01, n = 22) was found between UCP2 mRNA level in adipose tissue and BMI. The effect of fasting was investigated in eight lean and six obese subjects maintained on a hypocaloric diet (1,045 kJ/d) for 5 d. Calorie restriction induced a similar 2-2.5-fold increase in UCP2 and -3 mRNA levels in lean and obese subjects. To study the effect of insulin on UCP gene expression, six lean and five obese subjects underwent a 3-h euglycemic hyperinsulinemic clamp. Insulin infusion did not modify UCP2 and -3 mRNA levels. In conclusion, the similar induction of gene expression observed during fasting in lean and obese subjects shows that there is no major alteration of UCP2 and -3 gene regulation in adipose tissue and skeletal muscle of obese subjects. The increase in UCP2 and -3 mRNA levels suggests a role for these proteins in the metabolic adaptation to fasting.


Genome Biology | 2008

Adipose tissue transcriptomic signature highlights the pathological relevance of extracellular matrix in human obesity

Corneliu Henegar; Joan Tordjman; Vincent Achard; Danièle Lacasa; Isabelle Cremer; Michèle Guerre-Millo; Christine Poitou; Arnaud Basdevant; Vladimir Stich; Nathalie Viguerie; Dominique Langin; P. Bedossa; Jean-Daniel Zucker; Karine Clément

BackgroundInvestigations performed in mice and humans have acknowledged obesity as a low-grade inflammatory disease. Several molecular mechanisms have been convincingly shown to be involved in activating inflammatory processes and altering cell composition in white adipose tissue (WAT). However, the overall importance of these alterations, and their long-term impact on the metabolic functions of the WAT and on its morphology, remain unclear.ResultsHere, we analyzed the transcriptomic signature of the subcutaneous WAT in obese human subjects, in stable weight conditions and after weight loss following bariatric surgery. An original integrative functional genomics approach was applied to quantify relations between relevant structural and functional themes annotating differentially expressed genes in order to construct a comprehensive map of transcriptional interactions defining the obese WAT. These analyses highlighted a significant up-regulation of genes and biological themes related to extracellular matrix (ECM) constituents, including members of the integrin family, and suggested that these elements could play a major mediating role in a chain of interactions that connect local inflammatory phenomena to the alteration of WAT metabolic functions in obese subjects. Tissue and cellular investigations, driven by the analysis of transcriptional interactions, revealed an increased amount of interstitial fibrosis in obese WAT, associated with an infiltration of different types of inflammatory cells, and suggest that phenotypic alterations of human pre-adipocytes, induced by a pro-inflammatory environment, may lead to an excessive synthesis of ECM components.ConclusionThis study opens new perspectives in understanding the biology of human WAT and its pathologic changes indicative of tissue deterioration associated with the development of obesity.


European Journal of Pharmacology | 1991

Coexistence of three β-adrenoceptor subtypes in white fat cells of various mammalian species

Dominique Langin; Maria P. Portillo; Jean-Sébastien Saulnier-Blache; Max Lafontan

The nature of the beta-adrenoceptors (beta-ARs) of the white fat cells of five mammalian species (rat, hamster, rabbit, dog and humans) was reassessed. The coexistence of at least three beta-ARs on the fat cell (except human) was demonstrated. Comparative binding and lipolysis studies were performed, using recently synthesized compounds selective for the atypical beta-AR of the rat brown fat cell and of the rat colon. beta 1- and beta 2-ARs have previously been identified in all the mammalian white fat cells using [125I]cyanopindolol ([125]CYP) or [3H]dihydroalprenolol. In addition to these receptors, we now demonstrated the existence of a third beta-AR directly involved in adrenergic-mediated lipolysis, and identified it in the white fat cells of the most commonly studied animal species, except humans. This receptor is not detected by the classically used beta-antagonist radioligands, explaining the discrepancies in reports on the nature of the beta-ARs of the adipose tissue. Pharmacological delineation of the third type of beta-AR-induced lipolysis showed this receptor to be rather similar to the previously proposed atypical beta-AR of brown and white rat fat cells. Its pharmacological properties were clarified, using new selective full agonists and partial agonists also acting as non-selective beta 1/beta 2-antagonists. The limits of [125]CYP as a radioligand were reported and the usefulness of BRL 37344, (+/-)-CGP 12177 and phenylethanolaminotralines derivatives (having an atypical beta-activity on intestinal motility) as major tools usable for atypical beta-AR activation was demonstrated. Moreover, confirming our previous results about the nature of the beta-ARs (beta 1- and beta 2-ARs) located in the fat cells of women (Mauriège et al., J. Lipid Res., 1987, 17, 156), no atypical beta-AR-mediated lipolysis was identified in abdominal adipose tissue from healthy women. The possible differences and similarities between this receptor and the recently cloned beta 3-AR are discussed.


Journal of Biological Chemistry | 2009

Contribution of adipose triglyceride lipase and hormone-sensitive lipase to lipolysis in hMADS adipocytes

Véronic Bezaire; Aline Mairal; Carole Ribet; Corinne Lefort; Amandine Girousse; Johan W.E. Jocken; Jurga Laurencikiene; Rodica Anesia; Anne-Marie Rodriguez; Mikael Rydén; Britta M. Stenson; Christian Dani; Gérard Ailhaud; Peter Arner; Dominique Langin

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.


British Journal of Nutrition | 2005

The case for strategic international alliances to harness nutritional genomics for public and personal health

Jim Kaput; Jose M. Ordovas; Lynnette R. Ferguson; Ben van Ommen; Raymond L. Rodriguez; Lindsay H. Allen; Bruce N. Ames; Kevin Dawson; Bruce German; Ronald M. Krauss; Wasyl Malyj; Michael C. Archer; Stephen Barnes; Amelia Bartholomew; Ruth Birk; Peter J. van Bladeren; Kent J. Bradford; Kenneth H. Brown; Rosane Caetano; David Castle; Ruth Chadwick; Stephen L. Clarke; Karine Clément; Craig A. Cooney; Dolores Corella; Ivana Beatrice Manica da Cruz; Hannelore Daniel; Troy Duster; Sven O. E. Ebbesson; Ruan Elliott

Nutrigenomics is the study of how constituents of the diet interact with genes, and their products, to alter phenotype and, conversely, how genes and their products metabolise these constituents into nutrients, antinutrients, and bioactive compounds. Results from molecular and genetic epidemiological studies indicate that dietary unbalance can alter gene-nutrient interactions in ways that increase the risk of developing chronic disease. The interplay of human genetic variation and environmental factors will make identifying causative genes and nutrients a formidable, but not intractable, challenge. We provide specific recommendations for how to best meet this challenge and discuss the need for new methodologies and the use of comprehensive analyses of nutrient-genotype interactions involving large and diverse populations. The objective of the present paper is to stimulate discourse and collaboration among nutrigenomic researchers and stakeholders, a process that will lead to an increase in global health and wellness by reducing health disparities in developed and developing countries.


Proceedings of the Nutrition Society | 1996

Adipocyte hormone-sensitive lipase: a major regulator of lipid metabolism

Dominique Langin; Cecilia Holm; Max Lafontan

Le tissu adipeux joue un r81e important dans le contrde de la balance CnergCtique. La mobilisation des triacylglycCrols par la lipase hormono-sensible (EC 3.1.1.3; LHS) est soumis a un contrBle direct par les hormones et neurotransmetteurs qui modulent les concentrations intracellulaires d’AMP cyclique (AMPc). L’hydrolyse des triacylglycCrols par la LHS constitue l’etape limitante de la lipolyse. La LHS est phosphorylCe sur le site rkgulateur (Ser552 dans la LHS humaine) par la p r o t h e kinase dependante de 1’AMPc (EC 2.7.1.37) lorsque les concentrations intracellulaires d’ AMPc augmentent. Cette phosphorylation conduit B l’activation de l’enzyme. Une deuxikme site de phosphorylation (Ser5.54 dans la LHS humaine) dCnommC site basal est la cible de la protCine kinase activCe par I’AMP. La phosphorylation du site basal ne conduit pas B l’activation de la LHS et empkche la phosphorylation sur le site regulateur. La phosphorylation et I’activation de la protCine kinase activke par 1’AMP constitue donc un mecanisme antilipolytique qui est fonctionnel sur cellules isolCes mais dont l’importance physiologique n’est pas connue. Les ADN complkmentaires de la LHS de plusieurs espkces ont CtC clones et la structure des gbnes de LHS de l’homme et de la souris sont connus. Differents domaines fonctionnels de la protCine ont CtC proposes. Une rCgion d’homologie de sequences en amont de la serine 424 du site catalytique avec cinq enzymes d’organismes procaryotes et une enzyme humaine a ete decrite. La localisation chromosomique du gbne de la LHS est connue chez l’homme (chromosome 19, region q13-1+13.2), le porc et la souris. La mise en evidence de marqueurs polymorphiques dans le g&ne devrait permettre de tester l’hypothkse d’une implication de la LHS dans certaines maladies hereditaires du metabolisme lipidique. L’expression de la LHS varie selon la localisation anatomique du tissu adipeux chez le rat. Cette expression subit Cgalement des variations durant la gestation chez le rat et le cycle annuel chez les mammifbres hibernants. Chez l’homme, les taux d’ARN messagers de la LHS sont diminuCs dans le tissu adipeux de certains patients atteints de cancer. L‘activitC enzymatique totale est diminuee chez les patients atteints d’hyperlipidemie familiale combinCe mais pas chez les patients atteints du syndrome metabolique bien que, dans les deux cas, la lipolyse adipocytaire maximale soit diminuke. Les mkcanismes molCculaires de contr6le de l’expression de la LHS sont pratiquement inconnus.


Trends in Pharmacological Sciences | 2000

β3-Adrenoceptors in the cardiovascular system

Chantal Gauthier; Dominique Langin; Jean-Luc Balligand

beta-Adrenoceptors of the beta1 and beta2 subtypes classically mediate the effects of catecholamines on the contractility of cardiac muscle and the relaxation of vascular smooth muscle. Since the molecular characterization of the beta3-adrenoceptor in 1989, most studies of this adrenoceptor subtype have focused on its control of lipolysis in adipose tissues. However, more recent studies have investigated the involvement of beta3-adrenoceptors in the physiological control of cardiac and vascular contractility. In this article, the pharmacological and molecular evidence that supports the functional role of beta3-adrenoceptors in cardiovasculartissues of various species, including humans, will be discussed. These data might provide new insights into our understanding of the abnormal responsiveness of the cardiovascular system to catecholamines in heart failure and its treatment with beta3-adrenoceptor antagonists.

Collaboration


Dive into the Dominique Langin's collaboration.

Top Co-Authors

Avatar

Vladimir Stich

Charles University in Prague

View shared research outputs
Top Co-Authors

Avatar

Peter Arner

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Wim H. M. Saris

Maastricht University Medical Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arne Astrup

French Institute of Health and Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eva Klimcakova

Charles University in Prague

View shared research outputs
Top Co-Authors

Avatar

Claus Holst

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar

Jan Polak

Charles University in Prague

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge