Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Arterioscler Thromb Vasc Biol ; 43(5): 684-696, 2023 05.
Article in English | MEDLINE | ID: mdl-36924232

ABSTRACT

BACKGROUND: Adaptation of fat depots to change in fuel availability is critical for metabolic flexibility and cardiometabolic health. The mechanisms responsible for fat depot-specific lipid sensing and shuttling remain elusive. Adipose tissue microvascular endothelial cells (AT-EC) regulates bidirectional fatty acid fluxes depending on fed or fasted state. How AT-EC sense and adapt to metabolic changes according to AT location remains to be established. METHODS: We combined transcriptional analysis of native human AT-EC together with in vitro approaches in primary human AT-EC and in vivo and ex vivo studies of mice under fed and fasted conditions. RESULTS: Transcriptional large-scale analysis of human AT-EC isolated from gluteofemoral and abdominal subcutaneous AT revealed that the endothelium exhibits a fat depot-specific signature associated with lipid handling and Notch signaling enrichment. We uncovered a functional link between metabolic status and endothelial DLL4 (delta-like canonical notch ligand 4), which decreases with fasting. DLL4 regulates fatty acid uptake through nontranscriptional modulation of macropinocytosis-dependent long chain fatty acid uptake. Importantly, the changes in DLL4 expression, in response to energy transition state, is impaired under obesogenic conditions, an early alteration coinciding with a defect in systemic fatty acid fluxes adaptation and a resistance to weight loss. CONCLUSIONS: DLL4 is a major actor in the adaptive mechanisms of AT-EC to regulate lipid fluxes. It likely contributes to fat depot-dependent metabolism in response to energy transition states. AT-EC alteration with obesity may favor metabolic inflexibility and the development of cardiometabolic disorders.


Subject(s)
Cardiovascular Diseases , Endothelial Cells , Mice , Humans , Animals , Endothelial Cells/metabolism , Fatty Acids/metabolism , Obesity/genetics , Obesity/metabolism , Fasting , Endothelium/metabolism , Cardiovascular Diseases/metabolism , Calcium-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism
2.
Aging Cell ; 22(3): e13776, 2023 03.
Article in English | MEDLINE | ID: mdl-36617688

ABSTRACT

Senescence is a key event in the impairment of adipose tissue (AT) function with obesity and aging but the underlying molecular and cellular players remain to be fully defined, particularly with respect to the human AT progenitors. We have found distinct profiles of senescent progenitors based on AT location between stroma from visceral versus subcutaneous AT. In addition to flow cytometry, we characterized the location differences with transcriptomic and proteomic approaches, uncovering the genes and developmental pathways that are underlying replicative senescence. We identified key components to include INBHA as well as SFRP4 and GREM1, antagonists for the WNT and BMP pathways, in the senescence-associated secretory phenotype and NOTCH3 in the senescence-associated intrinsic phenotype. Notch activation in AT progenitors inhibits adipogenesis and promotes myofibrogenesis independently of TGFß. In addition, we demonstrate that NOTCH3 is enriched in the premyofibroblast progenitor subset, which preferentially accumulates in the visceral AT of patients with an early obesity trajectory. Herein, we reveal that NOTCH3 plays a role in the balance of progenitor fate determination preferring myofibrogenesis at the expense of adipogenesis. Progenitor NOTCH3 may constitute a tool to monitor replicative senescence and to limit AT dysfunction in obesity and aging.


Subject(s)
Cellular Senescence , Proteomics , Humans , Cellular Senescence/genetics , Adipose Tissue/metabolism , Aging/metabolism , Obesity/metabolism
3.
Cells ; 11(22)2022 11 09.
Article in English | MEDLINE | ID: mdl-36428969

ABSTRACT

Adipose tissue (AT) expansion either through hypertrophy or hyperplasia is determinant in the link between obesity and metabolic alteration. The present study aims to profile the unhealthy subcutaneous and visceral AT (SAT, VAT) expansion in obesity and in the outcomes of bariatric surgery (BS). The repartition of adipocytes according to diameter and the numbers of progenitor subtypes and immune cells of SAT and VAT from 161 obese patients were determined by cell imaging and flow cytometry, respectively. Associations with insulin resistance (IR) prior to BS as well as with the loss of excessive weight (EWL) and IR at 1 and 3 years post-BS were studied; prior to BS, SAT and VAT, unhealthy expansions are characterized by the accumulation of adipogenic progenitors and CD4+ T lymphocytes and by adipocyte hypertrophy and elevated macrophage numbers, respectively. Such SAT stromal profile and VAT adipocyte hypertrophy are associated with adverse BS outcomes. Finally, myofibrogenic progenitors are a common determinant of weight and IR trajectories post-BS; the study suggests that adipogenesis in SAT and adipocyte hypertrophy in VAT are common determinants of metabolic alterations with obesity and of the weight loss and metabolic response to bariatric surgery. The data open up new avenues to better understand and predict individual outcomes in response to changes in energy balance.


Subject(s)
Bariatric Surgery , Insulin Resistance , Humans , Adipocytes/metabolism , Obesity/metabolism , Insulin Resistance/physiology , Stromal Cells/metabolism , Hypertrophy
4.
Cell Mol Life Sci ; 78(7): 3485-3501, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33313981

ABSTRACT

The incidence of disorders associated with low inflammatory state, such as chronic kidney disease, increases in the elderly. The accumulation of senescent cells during aging and the senescence-associated secretory phenotype, which leads to inflammaging, is known to be deleterious and account for progressive organ dysfunction. To date, the cellular actors implicated in chronic inflammation in the kidney during aging are still not well characterized. Using the DECyt method, based on hierarchical clustering of flow cytometry data, we showed that aging was associated with significant changes in stromal cell diversity in the kidney. In particular, we identified two cell populations up-regulated with aging, the mesenchymal stromal cell subset (kMSC) expressing CD73 and the monocyte-derived Ly6C+ CCR2+ macrophage subset expressing pro-inflammatory cytokines. Aged CD73+ kMSCs depicted senescence associated features with low proliferation rate, increased DNA damage foci and Ccl2 expression. Using co-cultures experiments, we showed that aged CD73+ kMSC promoted monocyte activation and secretion of inflammatory cytokines albeit less efficiently than young CD73+ kMSCs. In the context of ageing, increased frequency of CD73+ kMSC subpopulations could provide additional niche factors to newly recruited monocytes favoring a positive regulatory loop in response to local inflammation. Interfering with such partnership during aging could be a valuable approach to regulate kidney inflammaging and to limit the risk of developing chronic kidney disease in the elderly.


Subject(s)
Cellular Microenvironment/immunology , Cellular Senescence/immunology , Inflammation/immunology , Kidney/immunology , Macrophages/immunology , Monocytes/immunology , Receptors, CCR2/metabolism , Animals , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Cytokines/metabolism , Inflammation/metabolism , Inflammation/pathology , Kidney/metabolism , Kidney/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Monocytes/pathology
5.
J Pharmacol Exp Ther ; 371(2): 555-566, 2019 11.
Article in English | MEDLINE | ID: mdl-31270215

ABSTRACT

Novel mechanisms and health benefits have been recently suggested for the antidepressant drug phenelzine (PHE), known as a nonselective monoamine oxidase inhibitor. They include an antilipogenic action that could have an impact on excessive fat accumulation and obesity-related metabolic alterations. We evaluated the metabolic effects of an oral PHE treatment on mice fed a high-fat diet (HFD). Eleven-week-old male C57BL/6 mice were fed a HFD and either a 0.028% PHE solution (HFD + PHE) or water to drink for 11 weeks. PHE attenuated the increase in body weight and adiposity without affecting food consumption. Energy efficiency was lower in HFD + PHE mice. Lipid content was reduced in subcutaneous fat pads, liver, and skeletal muscle. In white adipose tissue (WAT), PHE reduced sterol regulatory element-binding protein-1c and phosphoenolpyruvate carboxykinase mRNA levels, inhibited amine-induced lipogenesis, and did not increase lipolysis. Moreover, HFD + PHE mice presented diminished levels of hydrogen peroxide release in subcutaneous WAT and reduced expression of leukocyte transmigration markers and proinflammatory cytokines in visceral WAT and liver. PHE reduced the circulating levels of glycerol, triacylglycerols, high-density lipoprotein cholesterol, and insulin. Insulin resistance was reduced, without affecting glucose levels and glucose tolerance. In contrast, PHE increased rectal temperature and slightly increased energy expenditure. The mitigation of HFD-induced metabolic disturbances points toward a promising role for PHE in obesity treatment and encourages further research on its mechanisms of action. SIGNIFICANCE STATEMENT: Phenelzine reduces body fat, markers of oxidative stress, inflammation, and insulin resistance in high-fat diet mice. Semicarbazide-sensitive amine oxidase, monoamine oxidase, phosphoenolpyruvate carboxykinase, and sterol regulatory element-binding protein-1c are involved in the metabolic effects of phenelzine. Phenelzine could be potentially used for the treatment of obesity-related complications.


Subject(s)
Adipose Tissue/drug effects , Adipose Tissue/metabolism , Diet, High-Fat/adverse effects , Insulin Resistance/physiology , Monoamine Oxidase Inhibitors/administration & dosage , Phenelzine/administration & dosage , Administration, Oral , Animals , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Oxidative Stress/physiology , Treatment Outcome
6.
Am J Clin Nutr ; 109(6): 1499-1510, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30869115

ABSTRACT

BACKGROUND: The adipose tissue (AT) is a secretory organ producing a wide variety of factors that participate in the genesis of metabolic disorders linked to excess fat mass. Weight loss improves obesity-related disorders. OBJECTIVES: Transcriptomic studies on human AT, and a combination of analyses of transcriptome and proteome profiling of conditioned media from adipocytes and stromal cells isolated from human AT, have led to the identification of apolipoprotein M (apoM) as a putative adipokine. We aimed to validate apoM as novel adipokine, investigate the relation of AT APOM expression with metabolic syndrome and insulin sensitivity, and study the regulation of its expression in AT and secretion during calorie restriction-induced weight loss. METHODS: We examined APOM mRNA level and secretion in AT from 485 individuals enrolled in 5 independent clinical trials, and in vitro in human multipotent adipose-derived stem cell adipocytes. APOM expression and secretion were measured during dieting. RESULTS: APOM was expressed in human subcutaneous and visceral AT, mainly by adipocytes. ApoM was released into circulation from AT, and plasma apoM concentrations correlate with AT APOM mRNA levels. In AT, APOM expression inversely correlated with adipocyte size, was lower in obese compared to lean individuals, and reduced in subjects with metabolic syndrome and type 2 diabetes. Regardless of fat depot, there was a positive relation between AT APOM expression and systemic insulin sensitivity, independently of fat mass and plasma HDL cholesterol. In human multipotent adipose-derived stem cell adipocytes, APOM expression was enhanced by insulin-sensitizing peroxisome proliferator-activated receptor agonists and inhibited by tumor necrosis factor α, a cytokine that causes insulin resistance. In obese individuals, calorie restriction increased AT APOM expression and secretion. CONCLUSIONS: ApoM is a novel adipokine, the expression of which is a hallmark of healthy AT and is upregulated by calorie restriction. AT apoM deserves further investigation as a potential biomarker of risk for diabetes and cardiovascular diseases.


Subject(s)
Adipokines/genetics , Apolipoproteins M/genetics , Obesity/diet therapy , Obesity/genetics , Adipocytes/metabolism , Adipokines/metabolism , Apolipoproteins M/metabolism , Caloric Restriction , Clinical Trials as Topic , Cohort Studies , Cross-Sectional Studies , Female , Humans , Longitudinal Studies , Male , Middle Aged , Obesity/metabolism
7.
Arterioscler Thromb Vasc Biol ; 38(5): 1134-1146, 2018 05.
Article in English | MEDLINE | ID: mdl-29545239

ABSTRACT

OBJECTIVE: Adipose tissue (AT) dysfunction associated with obesity or aging is a major cause for lipid redistribution and the progression of cardiometabolic disorders. Our goal is to decipher the contribution of human AT microvascular endothelial cells (ECs) in the maintenance of fatty acid (FA) fluxes and the impact of senescence on their function. APPROACH AND RESULTS: We used freshly isolated primary microvascular ECs from human AT. Our data identified the endothelial FA handling machinery including FATPs (FA transport proteins) FATP1, FATP3, FATP4, and CD36 as well as FABP4 (FA binding protein 4). We showed that PPARγ (peroxisome proliferator-activated receptor gamma) regulates the expression of FATP1, CD36, and FABP4 and is a major regulator of FA uptake in human AT EC (hATEC). We provided evidence that endothelial PPARγ activity is modulated by senescence. Indeed, the positive regulation of FA transport by PPARγ agonist was abolished, whereas the emergence of an inflammatory response was favored in senescent hATEC. This was associated with the retention of nuclear FOXO1 (forkhead box protein O1), whereas nuclear PPARγ translocation was impaired. CONCLUSIONS: These data support the notion that PPARγ is a key regulator of primary hATEC function including FA handling and inflammatory response. However, the outcome of PPARγ activation is modulated by senescence, a phenomenon that may impact the ability of hATEC to properly respond to and handle lipid fluxes. Finally, our work highlights the role of hATEC in the regulation of FA fluxes and reveals that dysfunction of these cells with accelerated aging is likely to participate to AT dysfunction and the redistribution of lipids.


Subject(s)
Abdominal Fat/blood supply , Cellular Senescence , Endothelial Cells/metabolism , Fatty Acids/metabolism , Inflammation/metabolism , Microvessels/metabolism , PPAR gamma/metabolism , Active Transport, Cell Nucleus , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Endothelial Cells/ultrastructure , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Female , Forkhead Box Protein O1/metabolism , Humans , Inflammation/genetics , Inflammation/pathology , Microvessels/ultrastructure , PPAR gamma/genetics , Signal Transduction
8.
Bio Protoc ; 8(3): e2711, 2018 Feb 05.
Article in English | MEDLINE | ID: mdl-34179253

ABSTRACT

Most studies of human adipose tissue (AT) metabolism and functionality have been performed in vitro on isolated mature adipocyte or in situ using the microdialysis technique (Lafontan, 2012). However, these approaches have several limitations. The use of mature isolated adipocytes is limiting as adipocytes are not in their physiological environment and the collagenase digestion process could affect both adipocyte survival and functionality. While metabolic studies using microdialysis have brought the advantage of studying the lipolytic response of the adipose tissue in situ, it provides only qualitative measures but does not give any information on the contribution of different adipose tissue cell components. Moreover, the number of microdialysis probes that can be used concomitantly in one subject is limited and can be influenced by local blood flow changes and by the molecular size cut-off of the microdialysis probe. Here we present a protocol to assess adipose tissue functionality ex vivo in AT explants allowing the studies of adipose tissue in its whole context, for several hours. In addition, the isolation of the different cell components to evaluate the cell-specific impact of lipolysis can be performed. We recently used the present protocol and demonstrated that fatty acid release during lipolysis impacts directly on a specific cell subset present in the adipose tissue stroma-vascular compartment. This assay can be adapted to address other research questions such as the effects of hormones or drugs treatment on the phenotype of the various cell types present in adipose tissue ( Gao et al., 2016 ).

9.
Stem Cells ; 33(4): 1277-91, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25523907

ABSTRACT

Obesity-associated inflammation contributes to the development of metabolic diseases. Although brite adipocytes have been shown to ameliorate metabolic parameters in rodents, their origin and differentiation remain to be characterized in humans. Native CD45-/CD34+/CD31- cells have been previously described as human adipocyte progenitors. Using two additional cell surface markers, MSCA1 (tissue nonspecific alkaline phosphatase) and CD271 (nerve growth factor receptor), we are able to partition the CD45-/CD34+/CD31- cell population into three subsets. We establish serum-free culture conditions without cell expansion to promote either white/brite adipogenesis using rosiglitazone, or bone morphogenetic protein 7 (BMP7), or specifically brite adipogenesis using 3-isobuthyl-1-methylxanthine. We demonstrate that adipogenesis leads to an increase of MSCA1 activity, expression of white/brite adipocyte-related genes, and mitochondriogenesis. Using pharmacological inhibition and gene silencing approaches, we show that MSCA1 activity is required for triglyceride accumulation and for the expression of white/brite-related genes in human cells. Moreover, native immunoselected MSCA1+ cells exhibit brite precursor characteristics and the highest adipogenic potential of the three progenitor subsets. Finally, we provided evidence that MSCA1+ white/brite precursors accumulate with obesity in subcutaneous adipose tissue (sAT), and that local BMP7 and inflammation regulate brite adipogenesis by modulating MSCA1 in human sAT. The accumulation of MSCA1+ white/brite precursors in sAT with obesity may reveal a blockade of their differentiation by immune cells, suggesting that local inflammation contributes to metabolic disorders through impairment of white/brite adipogenesis. Stem Cells 2015;33:1277-1291.


Subject(s)
Adipocytes, White/immunology , Adipocytes, White/metabolism , Adipogenesis/physiology , Antigens, Surface/biosynthesis , Immunity, Cellular/physiology , Adult , Aged , Cells, Cultured , Female , Humans , Middle Aged
10.
Biochim Biophys Acta ; 1841(1): 88-96, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24120919

ABSTRACT

Lysophosphatidic acid (LPA) is a pro-fibrotic mediator acting via specific receptors (LPARs) and is synthesized by autotaxin, that increases with obesity. We tested whether LPA could play a role in adipose tissue (AT)-fibrosis associated with obesity. Fibrosis [type I, III, and IV collagens (COL), fibronectin (FN), TGFß, CTGF and αSMA] and inflammation (MCP1 and F4/80) markers were quantified: (i) in vivo in inguinal (IAT) and perigonadic (PGAT) AT from obese-diabetic db/db mice treated with the LPAR antagonist Ki16425 (5mg/kg/day ip for 7 weeks); and (ii) in vitro in human AT explants in primary culture for 72h in the presence of oleoyl-LPA (10µM) and/or Ki16425 (10µM) and/or the HIF-1α inhibitor YC-1 (100µM). Treatment of db/db mice with Ki16425 reduced Col I and IV mRNAs in IAT and PGAT while Col III mRNAs were only reduced in IAT. This was associated with reduction of COL protein staining in both IAT and PGAT. AT explants showed a spontaneous and time-dependent increase in ATX expression and production of LPA in the culture medium, along with increased levels of Col I and III, TGFß and αSMA mRNAs and of COL protein staining. In vitro fibrosis was blocked by Ki16425 and was further amplified by oleoyl-LPA. LPA-dependent in vitro fibrosis was blocked by co-treatment with YC1. Our results show that endogenous and exogenous LPA exert a pro-fibrotic activity in AT in vivo and in vitro. This activity could be mediated by an LPA1R-dependent pathway and could involve HIF-1α.


Subject(s)
Adipose Tissue/metabolism , Isoxazoles/toxicity , Lysophospholipids/metabolism , Propionates/toxicity , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Actins/biosynthesis , Adipose Tissue/pathology , Animals , Collagen/biosynthesis , Enzyme Activators/pharmacology , Female , Fibrosis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Indazoles/pharmacology , Male , Mice , Mice, Obese , Receptors, Lysophosphatidic Acid/metabolism , Tissue Culture Techniques , Transforming Growth Factor beta/biosynthesis
11.
BMC Endocr Disord ; 13: 54, 2013 Nov 25.
Article in English | MEDLINE | ID: mdl-24267103

ABSTRACT

BACKGROUND: Allograft inflammatory factor 1 (AIF-1) is a putative obesity gene. Our aim was to examine the expression of AIF-1 in human white adipose tissue (WAT) in relation to obesity and metabolic phenotypes in women. METHODS: WAT secretion of AIF-1 was determined in subcutaneous adipose tissue pieces in vitro by ELISA from 5 subjects. mRNA expression of AIF-1 was determined by RT-qPCR in the isolated cell fractions of adipose tissue (n = 5-6 per group), in subcutaneous and visceral WAT pieces from non-obese (n = 12) and obese women (n = 23), and in some subcutaneous WAT also before and after weight reduction (n = 10). Finally, adipose AIF-1 mRNA was related to metabolic phenotypes in 96 subjects with a wide range of BMI. RESULTS: AIF-1 was secreted in a time dependent fashion from WAT. The major source of AIF-1 was WAT resident macrophages. Expression of AIF-1 was similar in visceral and subcutaneous WAT and was two-fold increased in obese women (P < 0.01). AIF-1 mRNA expression levels were normalized after weight reduction (P < 0.01). Expression of AIF-1 was inversely correlated with insulin sensitivity as assessed by insulin tolerance test (KITT), and circulating levels of adiponectin (P = 0.02), and positively correlated with insulin resistance as estimated by HOMA (=0.0042). CONCLUSIONS: AIF-1 is a novel adipokine produced mainly by macrophages within human WAT. Its expression is increased in obese women and associates with unfavourable metabolic phenotypes. AIF-1 may play a paracrine role in the regulation of WAT function through cross-talk between macrophages and other cell types within the adipose tissue.

12.
Diabetologia ; 56(11): 2524-37, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23963324

ABSTRACT

AIMS/HYPOTHESIS: Circulating lipopolysaccharide-binding protein (LBP) is an acute-phase reactant known to be increased in obesity. We hypothesised that LBP is produced by adipose tissue (AT) in association with obesity. METHODS: LBP mRNA and LBP protein levels were analysed in AT from three cross-sectional (n = 210, n = 144 and n = 28) and three longitudinal (n = 8, n = 25, n = 20) human cohorts; in AT from genetically manipulated mice; in isolated adipocytes; and in human and murine cell lines. The effects of a high-fat diet and exposure to lipopolysaccharide (LPS) and peroxisome proliferator-activated receptor (PPAR)γ agonist were explored. Functional in vitro and ex vivo experiments were also performed. RESULTS: LBP synthesis and release was demonstrated to increase with adipocyte differentiation in human and mouse AT, isolated adipocytes and human and mouse cell lines (Simpson-Golabi-Behmel syndrome [SGBS], human multipotent adipose-derived stem [hMAD] and 3T3-L1 cells). AT LBP expression was robustly associated with inflammatory markers and increased with metabolic deterioration and insulin resistance in two independent cross-sectional human cohorts. AT LBP also increased longitudinally with weight gain and excessive fat accretion in both humans and mice, and decreased with weight loss (in two other independent cohorts), in humans with acquired lipodystrophy, and after ex vivo exposure to PPARγ agonist. Inflammatory agents such as LPS and TNF-α led to increased AT LBP expression in vivo in mice and in vitro, while this effect was prevented in Cd14-knockout mice. Functionally, LBP knockdown using short hairpin (sh)RNA or anti-LBP antibody led to increases in markers of adipogenesis and decreased adipocyte inflammation in human adipocytes. CONCLUSIONS/INTERPRETATION: Collectively, these findings suggest that LBP might have an essential role in inflammation- and obesity-associated AT dysfunction.


Subject(s)
Acute-Phase Proteins/metabolism , Adipocytes/metabolism , Adipose Tissue/pathology , Carrier Proteins/metabolism , Inflammation/metabolism , Membrane Glycoproteins/metabolism , Obesity/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adult , Animals , Humans , In Vitro Techniques , Insulin Resistance/physiology , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Rosiglitazone , Thiazolidinediones/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
13.
PLoS One ; 7(2): e31274, 2012.
Article in English | MEDLINE | ID: mdl-22355352

ABSTRACT

OBJECTIVE: The present study was undertaken to characterize the remodeling phenotype of human adipose tissue (AT) macrophages (ATM) and to analyze their paracrine effects on AT progenitor cells. RESEARCH DESIGN AND METHODS: The phenotype of ATM, immunoselected from subcutaneous (Sc) AT originating from subjects with wide range of body mass index and from paired biopsies of Sc and omental (Om) AT from obese subjects, was studied by gene expression analysis in the native and activated states. The paracrine effects of ScATM on the phenotype of human ScAT progenitor cells (CD34(+)CD31(-)) were investigated. RESULTS: Two main ATM phenotypes were distinguished based on gene expression profiles. For ScAT-derived ATM, obesity and adipocyte-derived factors favored a pro-fibrotic/remodeling phenotype whereas the OmAT location and hypoxic culture conditions favored a pro-angiogenic phenotype. Treatment of native human ScAT progenitor cells with ScATM-conditioned media induced the appearance of myofibroblast-like cells as shown by expression of both α-SMA and the transcription factor SNAIL, an effect mimicked by TGFß1 and activinA. Immunohistochemical analyses showed the presence of double positive α-SMA and CD34 cells in the stroma of human ScAT. Moreover, the mRNA levels of SNAIL and SLUG in ScAT progenitor cells were higher in obese compared with lean subjects. CONCLUSIONS: Human ATM exhibit distinct pro-angiogenic and matrix remodeling/fibrotic phenotypes according to the adiposity and the location of AT, that may be related to AT microenvironment including hypoxia and adipokines. Moreover, human ScAT progenitor cells have been identified as target cells for ScATM-derived TGFß and as a potential source of fibrosis through their induction of myofibroblast-like cells.


Subject(s)
Adipose Tissue/metabolism , Macrophages/metabolism , Myofibroblasts/cytology , Obesity/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transforming Growth Factor beta/metabolism , Adipose Tissue/cytology , Biomarkers/metabolism , Blotting, Western , Body Composition , Body Mass Index , Cells, Cultured , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Macrophages/cytology , Myofibroblasts/metabolism , Oligonucleotide Array Sequence Analysis , Omentum/cytology , Omentum/metabolism , Phenotype , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Subcutaneous Fat/cytology , Subcutaneous Fat/metabolism , Transforming Growth Factor beta/genetics
14.
Methods Mol Biol ; 702: 151-61, 2011.
Article in English | MEDLINE | ID: mdl-21082401

ABSTRACT

The present method describes an immunoselection/depletion approach to isolate the native human adipose tissue-derived progenitor cells that are free from endothelial cells and immune cells by the use of magnetic nanobeads and microbeads coupled to antibodies. Moreover, methods to isolate and to analyse the distinct cell populations that constitute the microenvironment of the human adipose tissue progenitor cells, i.e. mature adipocytes, endothelial cells, and macrophages, are mentioned.


Subject(s)
Adipose Tissue/cytology , Cell Separation/methods , Cytokines/metabolism , Lymphocytes/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Antigens, CD34/metabolism , Cell Differentiation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Flow Cytometry , Humans , Lipopolysaccharide Receptors/metabolism , Lymphocytes/cytology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/cytology , Stromal Cells/metabolism
15.
Diabetes ; 59(11): 2755-63, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20713685

ABSTRACT

OBJECTIVE: Regional differences among adipose depots in capacities for fatty acid storage, susceptibility to hypoxia, and inflammation likely contribute to complications of obesity. We defined the properties of endothelial cells (EC) isolated from subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) biopsied in parallel from obese subjects. RESEARCH DESIGN AND METHODS: The architecture and properties of the fat tissue capillary network were analyzed using immunohistochemistry and flow cytometry. CD34(+)/CD31(+) EC were isolated by immunoselection/depletion. Expression of chemokines, adhesion molecules, angiogenic factor receptors, as well as lipogenic and senescence-related genes were assayed by real-time PCR. Fat cell size and expression of hypoxia-dependent genes were determined in adipocytes from both fat depots. RESULTS: Hypoxia-related genes were more highly expressed in VAT than SAT adipocytes. VAT adipocytes were smaller than SAT adipocytes. Vascular density and EC abundance were higher in VAT. VAT-EC exhibited a marked angiogenic and inflammatory state with decreased expression of metabolism-related genes, including endothelial lipase, GPIHBP1, and PPAR gamma. VAT-EC had enhanced expression of the cellular senescence markers, IGFBP3 and γ-H2AX, and decreased expression of SIRT1. Exposure to VAT adipocytes caused more EC senescence-associated ß-galactosidase activity than SAT adipocytes, an effect reduced in the presence of vascular endothelial growth factor A (VEGFA) neutralizing antibodies. CONCLUSIONS: VAT-EC exhibit a more marked angiogenic and proinflammatory state than SAT-EC. This phenotype may be related to premature EC senescence. VAT-EC may contribute to hypoxia and inflammation in VAT.


Subject(s)
Adipose Tissue/metabolism , Adipose Tissue/pathology , Cellular Senescence/physiology , Obesity/metabolism , Obesity/pathology , Adipocytes/cytology , Adipocytes/metabolism , Adipocytes/pathology , Adult , Biopsy , Body Mass Index , Chemokine CCL20/genetics , Female , Gene Expression Regulation , Humans , Hypercholesterolemia/genetics , Hypercholesterolemia/metabolism , Hypercholesterolemia/pathology , Hypertension/genetics , Hypertension/metabolism , Hypertension/pathology , Immunohistochemistry/methods , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Male , Middle Aged , Obesity/genetics , Reference Values , Subcutaneous Fat/metabolism , Subcutaneous Fat/pathology
16.
Arterioscler Thromb Vasc Biol ; 29(10): 1608-14, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19644053

ABSTRACT

OBJECTIVE: Adipose tissue (AT) plays a major role in the low-grade inflammatory state associated with obesity. The aim of the present study was to characterize the human AT lymphocytes (ATLs) and to analyze their interactions with adipocytes. METHODS AND RESULTS: Human ATL subsets were characterized by flow cytometry in subcutaneous ATs from 92 individuals with body mass index (BMI) ranging from 19 to 43 kg/m(2) and in paired biopsies of subcutaneous and visceral AT from 45 class II/III obese patients. CD3(+) ATLs were composed of effector and memory CD4(+) helper and CD8(+) cytotoxic T cells. The number of ATLs correlated positively with BMI and was higher in visceral than subcutaneous AT. Mature adipocytes stimulated the migration of ATLs and released the chemokine CCL20, the receptor of which (CCR6) was expressed in ATLs. The expression of adipocyte CCL20 was positively correlated with BMI and increased in visceral compared to subcutaneous adipocytes. ATLs expressed inflammatory markers and released interferon gamma (IFN gamma). Progenitor and adipocyte treatment with ATL-conditioned media reduced the insulin-mediated upregulation of lipogenic enzymes, an effect involving IFN gamma. CONCLUSIONS: Therefore, crosstalk occurs between adipocytes and lymphocytes within human AT involving T cell chemoattraction by adipocytes and modulation of lipogenesis by ATLs.


Subject(s)
Adipocytes/immunology , Chemokine CCL20/physiology , Lipogenesis , Obesity/immunology , T-Lymphocytes/physiology , Adiposity , Adult , Body Mass Index , CD3 Complex/analysis , Chemokine CCL20/analysis , Female , Flow Cytometry , Humans , Immunophenotyping , Interferon-gamma/physiology , Middle Aged , Subcutaneous Fat/immunology
17.
J Clin Endocrinol Metab ; 94(8): 3003-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19470636

ABSTRACT

CONTEXT: Follistatin is a glycoprotein that binds and neutralizes biological activities of TGFbeta superfamily members including activin and myostatin. We previously identified by expression profiling that follistatin levels in white adipose tissue (WAT) were regulated by obesity. OBJECTIVE: The objective of the study was to elucidate the role of follistatin in human WAT and obesity. DESIGN: We measured secreted follistatin protein from WAT biopsies and fat cells in vitro. We also quantified follistatin mRNA expression in sc and visceral WAT and in WAT-fractionated cells and related it to obesity status, body region, and cellular origin. We investigated the effects of follistatin on adipocyte differentiation of progenitor cells in vitro. PARTICIPANTS: Women (n = 66) with a wide variation in body mass index were recruited by advertisement and from a clinic for weight-reduction therapy. RESULTS: WAT secreted follistatin in vitro. Follistatin mRNA levels in sc but not visceral WAT were decreased in obesity and restored to nonobese levels after weight reduction. Follistatin mRNA levels were high in the stroma-vascular fraction of WAT and low in adipocytes. Recombinant follistatin treatment promoted adipogenic differentiation of progenitor cells and neutralized the inhibitory action of myostatin on differentiation in vitro. Moreover, activin and myostatin signaling receptors were detected in WAT and adipocytes. CONCLUSION: Follistatin is a new adipokine important for adipogenesis. Down-regulated WAT expression of follistatin in obesity may counteract adiposity but could, by inhibiting adipogenesis, contribute to hypertrophic obesity (large fat cells) and insulin resistance.


Subject(s)
Adipogenesis , Follistatin/physiology , Activin Receptors, Type II/genetics , Adipose Tissue, White/metabolism , Adult , Cell Differentiation , Cells, Cultured , Fatty Acid-Binding Proteins/genetics , Female , Follistatin/genetics , Humans , Mesenchymal Stem Cells/cytology , Middle Aged , PPAR gamma/genetics , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/analysis , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics
18.
J Clin Endocrinol Metab ; 93(10): 4098-106, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18682517

ABSTRACT

CONTEXT: Adipocyte formation in human adult adipose tissue (hAT) originates from resident progenitor cell differentiation in the stroma vascular fraction of the AT. The processes involved in the self-renewal of this cell population remain to be defined. OBJECTIVE: The objective was to study in situ and in vitro hAT progenitor cell (defined as CD34(+)/CD31(-) cells) proliferation. DESIGN AND PARTICIPANTS: In situ progenitor cell proliferation was assessed by immunohistochemistry and flow cytometry analyses on hAT from lean to obese subjects using the proliferation marker Ki-67. The effects of adipokines, hypoxia, and conditioned media (CM) from adipocytes, capillary endothelial cells, and macrophages isolated by an immunoselection approach were studied on hAT progenitor cell growth. Cell death in hAT was assessed by the terminal deoxynucleotidyl transferase-mediated dUTP-fluorescein end labeling method. RESULTS: Ki-67-positive staining was observed in AT progenitor cells. Fat mass enlargement in obese patients was associated with an increased Ki-67(+) progenitor cell population together with a new fraction of small adipocytes and increased cell death. HIF-1alpha mRNA expression in freshly harvested progenitor cells was positively correlated with body mass index. Adipocyte- and capillary endothelial cell-CM, hypoxia, leptin, IL-6, lysophosphatidic acid, and vascular endothelial growth factor, all increased hAT progenitor cell proliferation in vitro. Macrophage-CM had an antiproliferative effect that was suppressed by an antioxidant. CONCLUSIONS: The fraction of proliferative progenitor cells in adult hAT is modulated by the degree of adiposity. Changes in the progenitor cell microenvironment involving adipokines, hypoxia, and oxidative stress might play a key role in the control of the self-renewal of the local pool of AT progenitor cells.


Subject(s)
Adipose Tissue/physiology , Cell Proliferation , Extracellular Fluid/physiology , Human Development/physiology , Stem Cells/physiology , Adipokines/pharmacology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adult , Antigens, CD34/genetics , Antigens, CD34/metabolism , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cell Size/drug effects , Cells, Cultured , Culture Media, Conditioned/pharmacology , Female , Humans , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Oxygen/pharmacology , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Stem Cells/cytology , Stem Cells/metabolism
19.
FEBS Lett ; 582(15): 2219-24, 2008 Jun 25.
Article in English | MEDLINE | ID: mdl-18501713

ABSTRACT

Spatial and temporal control of ovine prion protein (Prnp) gene expression was achieved in mice using two transgenes: a Prnp minigene with tet-operator sequences inserted 5' to exon 1 and a mouse neurofilament genomic clone carrying the chimeric-repressor TRSID cDNA. In bi-transgenic mice, ovine PrP(C) expression could be reversibly controlled in neuronal cells by doxycycline treatment whereas it remains constant in other cell types. Overall, this model opens opportunities to assess the involvement of cell types in prion diseases and PrP physiological function. It demonstrates the potentiality of the TRSID-silencer to precisely control temporal and spatial gene expression in vivo.


Subject(s)
PrPC Proteins/genetics , Sheep/genetics , Silencer Elements, Transcriptional , Transgenes , Animals , Down-Regulation , Gene Expression , Mice , Mice, Transgenic , Neurons/metabolism
20.
Gene ; 404(1-2): 1-9, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17905541

ABSTRACT

Glycogen debranching enzyme (AGL) is a multifunctional enzyme acting in the glycogen degradation pathway. In humans, the AGL activity deficiency causes a type III glycogen storage disease (Cori-Forbes disease). One particularity of AGL gene expression lies in the multiple alternative splicing in its 5' region. The AGL gene was localized on ECA5q14-q15. The sequence of the equine cDNA was determined to be 7.5 kb in length with an open reading frame of 4602 bp. The gene is 69 kb long and contains 35 exons. The equine AGL gene has an ubiquitous expression and presents five tissue-dependent cDNA variants arising from alternative splicing of the first exons. The equine skeletal muscle and heart contain four out of six variants previously described in humans and the equine liver express three of these four human variants. We identified a new alternative splicing variant expressed in equine skeletal and heart muscles. All these mRNA variants most probably encode only two different protein isoforms of 1533 and 1377 amino-acids. Four SNPs were detected in the mRNA. The equine in silico promoter sequence reveals a structure similar to those of other mammalian species. The disposition of the transcription factor biding sites does not correlate to the transcription start sites of tissue-specific variants.


Subject(s)
Glycogen Debranching Enzyme System/genetics , Horses/genetics , Polymorphism, Genetic , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Chromosomes/genetics , DNA, Complementary/chemistry , DNA, Complementary/genetics , Gene Expression , Genomics , Glycogen Debranching Enzyme System/metabolism , Molecular Sequence Data , Promoter Regions, Genetic , RNA, Messenger/analysis , RNA, Messenger/metabolism , Sequence Alignment , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL