Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
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
2.
Am J Pathol ; 192(6): 926-942, 2022 06.
Article in English | MEDLINE | ID: mdl-35358473

ABSTRACT

White adipose tissue accumulates at various sites throughout the body, some adipose tissue depots exist near organs whose function they influence in a paracrine manner. Prostate gland is surrounded by a poorly characterized adipose depot called periprostatic adipose tissue (PPAT), which plays emerging roles in prostate-related disorders. Unlike all other adipose depots, PPAT secretes proinflammatory cytokines even in lean individuals and does not increase in volume during obesity. These unique features remain unexplained because of the poor structural and functional characterization of this tissue. This study characterized the structural organization of PPAT in patients compared with abdominopelvic adipose tissue (APAT), an extraperitoneal adipose depot, the accumulation of which is correlated to body mass index. Confocal microscopy followed by three-dimensional reconstructions showed a sparse vascular network in PPAT when compared with that in APAT, suggesting that this tissue is hypoxic. Unbiased comparisons of PPAT and APAT transcriptomes found that most differentially expressed genes were related to the hypoxia response. High levels of the hypoxia-inducible factor 2α confirmed the presence of an adaptive response to hypoxia in PPAT. This chronic hypoxic state was associated with inflammation and fibrosis, which were not further up-regulated by obesity. This fibrosis and inflammation explain the failure of PPAT to expand in obesity and open new mechanistic avenues to explain its role in prostate-related disorders, including cancer.


Subject(s)
Adipose Tissue , Obesity , Adipose Tissue/pathology , Fibrosis , Humans , Hypoxia/pathology , Inflammation/pathology , Male , Obesity/complications
3.
J Invest Dermatol ; 142(9): 2488-2498.e8, 2022 09.
Article in English | MEDLINE | ID: mdl-35150661

ABSTRACT

Obesity is a recognized factor for increased risk and poor prognosis of many cancers, including melanoma. In this study, using genetically engineered mouse models of melanoma (NrasQ61K transgenic expression, associated or not with Cdkn2a heterozygous deletion), we show that obesity increases melanoma initiation and progression by supporting tumor growth and metastasis, thereby reducing survival. This effect is associated with a decrease in p16INK4A expression in tumors. Mechanistically, adipocytes downregulate p16INK4A in melanoma cells through ß-catenin-dependent regulation, which increases cell motility. Furthermore, ß-catenin is directly transferred from adipocytes to melanoma cells in extracellular vesicles, thus increasing its level and activity, which represses CDKN2A transcription. Adipocytes from individuals with obesity have a stronger effect than those from lean individuals, mainly owing to an increase in the number of vesicles secreted, thus increasing the amount of ß-catenin delivered to melanoma cells and, consequently, amplifying their effect. In conclusion, in this study, we reveal that adipocyte extracellular vesicles control p16INK4A expression in melanoma, which promotes tumor progression. This work expands our understanding of the cooperation between adipocytes and tumors, particularly in obesity.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16 , Extracellular Vesicles , Melanoma , Obesity , Adipocytes/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , Extracellular Vesicles/metabolism , Melanoma/genetics , Melanoma/metabolism , Mice , Obesity/genetics , Obesity/metabolism , beta Catenin/metabolism
4.
STAR Protoc ; 2(3): 100629, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34235494

ABSTRACT

Primary human bone marrow adipocytes (BM-Ads) display a specific metabolism that is not recapitulated by in vitro differentiated bone marrow mesenchymal stromal cells. These findings highlight the need for using primary BM-Ads in studies of the metabolic impact of BM-Ads on surrounding cells. Here, we present a protocol for isolating human BM-Ads from bone marrow aspirates and verifying adipocyte suspension purity. These isolated and purified BM-Ads can be used for functional assays or frozen for molecular analyses. For complete details on the use and execution of this protocol, please refer to Attane et al. (2020).


Subject(s)
Adipocytes/cytology , Bone Marrow Cells/cytology , Femur/cytology , Fluorescent Antibody Technique , Humans , Reproducibility of Results
5.
Int J Mol Sci ; 22(4)2021 Feb 17.
Article in English | MEDLINE | ID: mdl-33671469

ABSTRACT

Bone metastasis remains the most frequent and the deadliest complication of prostate cancer (PCa). Mechanisms leading to the homing of tumor cells to bone remain poorly characterized. Role of chemokines in providing navigational cues to migrating cancer cells bearing specific receptors is well established. Bone is an adipocyte-rich organ since 50 to 70% of the adult bone marrow (BM) volume comprise bone marrow adipocytes (BM-Ads), which are likely to produce chemokines within the bone microenvironment. Using in vitro migration assays, we demonstrated that soluble factors released by human primary BM-Ads are able to support the directed migration of PCa cells in a CCR3-dependent manner. In addition, we showed that CCL7, a chemokine previously involved in the CCR3-dependent migration of PCa cells outside of the prostate gland, is released by human BM-Ads. These effects are amplified by obesity and ageing, two clinical conditions known to promote aggressive and metastatic PCa. In human tumors, we found an enrichment of CCR3 in bone metastasis vs. primary tumors at mRNA levels using Oncomine microarray database. In addition, immunohistochemistry experiments demonstrated overexpression of CCR3 in bone versus visceral metastases. These results underline the potential importance of BM-Ads in the bone metastatic process and imply a CCR3/CCL7 axis whose pharmacological interest needs to be evaluated.


Subject(s)
Adipocytes/metabolism , Adipocytes/pathology , Bone Marrow/pathology , Bone and Bones/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, CCR3/metabolism , Aging/pathology , Bone Marrow/drug effects , Bone and Bones/drug effects , Cell Line, Tumor , Chemokine CCL7/metabolism , Chemotaxis/drug effects , Culture Media, Conditioned/pharmacology , Humans , Male , Neoplasm Metastasis , Obesity/complications , Prostatic Neoplasms/complications
6.
J Vis Exp ; (162)2020 08 19.
Article in English | MEDLINE | ID: mdl-32894273

ABSTRACT

Obesity is a major worldwide public health issue that increases the risk to develop cardiovascular diseases, type-2 diabetes, and liver diseases. Obesity is characterized by an increase in adipose tissue (AT) mass due to adipocyte hyperplasia and/or hypertrophia, leading to profound remodeling of its three-dimensional structure. Indeed, the maximal capacity of AT to expand during obesity is pivotal to the development of obesity-associated pathologies. This AT expansion is an important homeostatic mechanism to enable adaptation to an excess of energy intake and to avoid deleterious lipid spillover to other metabolic organs, such as muscle and liver. Therefore, understanding the structural remodeling that leads to the failure of AT expansion is a fundamental question with high clinical applicability. In this article, we describe a simple and fast clearing method that is routinely used in our laboratory to explore the morphology of mouse and human white adipose tissue by fluorescent imaging. This optimized AT clearing method is easily performed in any standard laboratory equipped with a chemical hood, a temperature-controlled orbital shaker and a fluorescent microscope. Moreover, the chemical compounds used are readily available. Importantly, this method allows one to resolve the 3D AT structure by staining various markers to specifically visualize the adipocytes, the neuronal and vascular networks, and the innate and adaptive immune cells distribution.


Subject(s)
Adipose Tissue/pathology , Imaging, Three-Dimensional , Salicylates/pharmacokinetics , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Humans , Mice , Microscopy, Fluorescence , Obesity/metabolism , Obesity/pathology
7.
EMBO J ; 39(3): e102525, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31919869

ABSTRACT

Extracellular vesicles are emerging key actors in adipocyte communication. Notably, small extracellular vesicles shed by adipocytes stimulate fatty acid oxidation and migration in melanoma cells and these effects are enhanced in obesity. However, the vesicular actors and cellular processes involved remain largely unknown. Here, we elucidate the mechanisms linking adipocyte extracellular vesicles to metabolic remodeling and cell migration. We show that adipocyte vesicles stimulate melanoma fatty acid oxidation by providing both enzymes and substrates. In obesity, the heightened effect of extracellular vesicles depends on increased transport of fatty acids, not fatty acid oxidation-related enzymes. These fatty acids, stored within lipid droplets in cancer cells, drive fatty acid oxidation upon being released by lipophagy. This increase in mitochondrial activity redistributes mitochondria to membrane protrusions of migrating cells, which is necessary to increase cell migration in the presence of adipocyte vesicles. Our results provide key insights into the role of extracellular vesicles in the metabolic cooperation that takes place between adipocytes and tumors with particular relevance to obesity.


Subject(s)
Adipocytes/cytology , Extracellular Vesicles/metabolism , Fatty Acids/metabolism , Melanoma/metabolism , Obesity/complications , 3T3 Cells , Adipocytes/metabolism , Animals , Autophagy , Cell Line, Tumor , Cell Movement , Humans , Lipid Metabolism , Male , Mice , Mitochondria/metabolism , Mitochondrial Dynamics , Obesity/metabolism , Oxidation-Reduction
8.
Cell Rep ; 30(4): 949-958.e6, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31995765

ABSTRACT

Under caloric restriction, bone marrow adipocytes (BM-Ads) do not decrease in size compared to white adipocytes, suggesting they harbor unique metabolic properties. We compare human primary BM-Ads with paired subcutaneous adipocytes (SC-Ads) using proteomic and lipidomic approaches. We find that, although SC-Ads and BM-Ads share similar morphological features, they possess distinct lipid metabolism. Although BM-Ad shows enrichment in proteins involved in cholesterol metabolism, correlating with increased free cholesterol content, proteins involved in lipolysis were downregulated. In particular, monoacylglycerol lipase expression is strongly reduced in BM-Ads, leading to monoacylglycerol accumulation. Consequently, basal and induced lipolytic responses are absent in BM-Ads, affirming their differences in metabolic fitness upon caloric restriction. These specific metabolic features are not recapitulated in vitro using common protocols to differentiate bone marrow mesenchymal stem cells. Thus, contrary to classical SC-Ads, BM-Ads display a specific lipid metabolism, as they are devoid of lipolytic activity and exhibit a cholesterol-orientated metabolism.


Subject(s)
Adipocytes/metabolism , Bone Marrow/metabolism , Lipid Metabolism , Proteome/metabolism , Adipocytes/cytology , Adipocytes/enzymology , Adipocytes/ultrastructure , Animals , Bone Marrow/enzymology , Caloric Restriction , Cell Line , Cells, Cultured , Cholesterol/metabolism , Humans , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Lipolysis/physiology , Mice , Microscopy, Electron, Transmission , Monoacylglycerol Lipases/genetics , Monoacylglycerol Lipases/metabolism , Protein Interaction Maps/genetics , Protein Interaction Maps/physiology , Proteome/genetics , Proteomics
9.
Breast Cancer Res ; 21(1): 7, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30654824

ABSTRACT

INTRODUCTION: Clinical studies suggest that obesity, in addition to promoting breast cancer aggressiveness, is associated with a decrease in chemotherapy efficacy, although the mechanisms involved remain elusive. As chemotherapy is one of the main treatments for aggressive or metastatic breast cancer, we investigated whether adipocytes can mediate resistance to doxorubicin (DOX), one of the main drugs used to treat breast cancer, and the mechanisms associated. METHODS: We used a coculture system to grow breast cancer cells with in vitro differentiated adipocytes as well as primary mammary adipocytes isolated from lean and obese patients. Drug cellular accumulation, distribution, and efflux were studied by immunofluorescence, flow cytometry, and analysis of extracellular vesicles. Results were validated by immunohistochemistry in a series of lean and obese patients with cancer. RESULTS: Adipocytes differentiated in vitro promote DOX resistance (with cross-resistance to paclitaxel and 5-fluorouracil) in a large panel of human and murine breast cancer cell lines independently of their subtype. Subcellular distribution of DOX was altered in cocultivated cells with decreased nuclear accumulation of the drug associated with a localized accumulation in cytoplasmic vesicles, which then are expelled into the extracellular medium. The transport-associated major vault protein (MVP), whose expression was upregulated by adipocytes, mediated both processes. Coculture with human mammary adipocytes also induced chemoresistance in breast cancer cells (as well as the related MVP-induced DOX efflux) and their effect was amplified by obesity. Finally, in a series of human breast tumors, we observed a gradient of MVP expression, which was higher at the invasive front, where tumor cells are at close proximity to adipocytes, than in the tumor center, highlighting the clinical relevance of our results. High expression of MVP in these tumor cells is of particular interest since they are more likely to disseminate to give rise to chemoresistant metastases. CONCLUSIONS: Collectively, our study shows that adipocytes induce an MVP-related multidrug-resistant phenotype in breast cancer cells, which could contribute to obesity-related chemoresistance.


Subject(s)
Adipocytes/metabolism , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Obesity/complications , Vault Ribonucleoprotein Particles/metabolism , 3T3 Cells , Adipose Tissue/cytology , Adult , Aged , Animals , Antineoplastic Agents/therapeutic use , Breast/cytology , Breast/pathology , Breast/surgery , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Cell Line, Tumor , Coculture Techniques , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Female , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Humans , Mastectomy , Mice , Middle Aged , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Primary Cell Culture , RNA, Small Interfering/metabolism , Vault Ribonucleoprotein Particles/genetics
10.
Int J Obes (Lond) ; 42(12): 2047-2056, 2018 12.
Article in English | MEDLINE | ID: mdl-29453465

ABSTRACT

BACKGROUND: Adipokines are peptides secreted from white adipose tissue (WAT), which have been linked to WAT dysfunction and metabolic complications of obesity. We set out to identify novel adipokines in subcutaneous WAT (sWAT) linked to insulin resistance (IR). METHODS: Gene expression was determined by microarray and qPCR in obese and non-obese subjects with varying degree of IR. WAT-secreted and circulating protein levels were measured by ELISA. RESULTS: In sWAT of 80 obese women discordant for IR, 44 genes encoding potential adipose-secreted proteins were differentially expressed. Among these, merely two proteins, S100A4 and MXRA5 were released from sWAT in a time-dependent manner (criterion for true adipokines) but only the circulating levels of S100A4 were higher in IR. In two additional cohorts (n = 29 and n = 56), sWAT S100A4 secretion was positively and BMI-independently associated with IR (determined by clamp or HOMA-IR), ATP-III risk score and adipocyte size (hypertrophy). In non-obese (n = 20) and obese subjects before and after bariatric surgery (n = 21), circulating and sWAT-secreted levels were highest in the obese and normalized following weight loss. Serum S100A4 concentrations were higher in subjects with type 2 diabetes. S100A4 sWAT expression associated positively with genes involved in inflammation/extracellular matrix formation and inversely with genes in metabolic pathways. Although S100A4 was expressed in both stromal cells and adipocytes, only the expression in adipocytes associated with BMI. CONCLUSIONS: S100A4 is a novel adipokine associated with IR and sWAT inflammation/adipocyte hypertrophy independently of BMI. Its value as a circulating marker for dysfunctional WAT and IR needs to be validated in larger cohorts.


Subject(s)
Adipokines/metabolism , Adipose Tissue, White/metabolism , Insulin Resistance/physiology , Obesity/metabolism , S100 Calcium-Binding Protein A4/metabolism , Adipokines/blood , Adipose Tissue, White/chemistry , Adult , Biomarkers , Cohort Studies , Female , Humans , Inflammation/metabolism , Male , Middle Aged , Obesity/blood , Obesity/epidemiology , S100 Calcium-Binding Protein A4/blood
11.
World J Diabetes ; 9(1): 25-32, 2018 Jan 15.
Article in English | MEDLINE | ID: mdl-29359026

ABSTRACT

AIM: To study complete dose-dependent effects of obestatin on lipolytic and glucose transport activities in human adipocyte preparations highly responsive to insulin. METHODS: Adipocytes were prepared by liberase digestion from subcutaneous abdominal adipose tissue obtained from overweight subjects undergoing plastic surgery. The index of lipolytic activity was the glycerol released in the incubation medium, while glucose transport was assessed by [3H]-2-deoxyglucose uptake assay. RESULTS: When tested from 0.1 nmol/L to 1 µmol/L, obestatin did not stimulate glycerol release; it did not inhibit the lipolytic effect of isoprenaline and did not alter the insulin antilipolytic effect. Obestatin hardly activated glucose transport at 1 µmol/L only. Moreover, the obestatin stimulation effect was clearly lower than the threefold increase induced by insulin 100 nmol/L. CONCLUSION: Low doses of obestatin cannot directly influence lipolysis and glucose uptake in human fat cells.

12.
Stem Cells ; 35(7): 1799-1814, 2017 07.
Article in English | MEDLINE | ID: mdl-28470788

ABSTRACT

White adipose tissue (WAT) expands in part through adipogenesis, a process involving fat cell generation and fatty acid (FA) storage into triglycerides (TGs). Several findings suggest that inter-individual and regional variations in adipogenesis are linked to metabolic complications. We aimed to identify cellular markers that define human adipocyte progenitors (APs) with pronounced adipogenic/TG storage ability. Using an unbiased single cell screen of passaged human adipose-derived stromal cells (hADSCs), we identified cell clones with similar proliferation rates but discordant capabilities to undergo adipogenic differentiation. Transcriptomic analyses prior to induction of differentiation showed that adipogenic clones displayed a significantly higher expression of CD36, encoding the scavenger receptor CD36. CD36+ hADSCs, in comparison with CD36-cells, displayed almost complete adipogenic differentiation while CD36 RNAi attenuated lipid accumulation. Similar findings were observed in primary CD45-/CD34+/CD31-APs isolated from human WAT where the subpopulation of MSCA1+/CD36+ cells displayed a significantly higher differentiation degree/TG storage capacity than MSCA1+/CD36-cells. Functional analyses in vitro and ex vivo confirmed that CD36 conferred APs an increased capacity to take up FAs thereby facilitating terminal differentiation. Among primary APs from subcutaneous femoral, abdominal and visceral human WAT, the fraction of CD36+ cells was significantly higher in depots associated with higher adipogenesis and reduced metabolic risk (i.e., femoral WAT). We conclude that CD36 marks APs with pronounced adipogenic potential, most probably by facilitating lipid uptake. This may be of value in developing human adipocyte cell clones and possibly in linking regional variations in adipogenesis to metabolic phenotype. Stem Cells 2017;35:1799-1814.


Subject(s)
Adipocytes, White/metabolism , Adipose Tissue, White/metabolism , CD36 Antigens/genetics , Stem Cells/metabolism , Transcriptome , Triglycerides/metabolism , Adipocytes, White/cytology , Adipogenesis/genetics , Adipose Tissue, White/cytology , Adult , Antigens, CD34/genetics , Antigens, CD34/metabolism , Antigens, Surface/genetics , Antigens, Surface/metabolism , Biological Transport , CD36 Antigens/antagonists & inhibitors , CD36 Antigens/metabolism , Cell Differentiation , Cell Proliferation , Female , Gene Expression Profiling , Humans , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Middle Aged , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Single-Cell Analysis , Stem Cells/cytology
13.
Stem Cells Int ; 2016: 1815982, 2016.
Article in English | MEDLINE | ID: mdl-26839555

ABSTRACT

Our knowledge about mesenchymal stem cells has considerably grown in the last years. Since the proof of concept of the existence of such cells in the 70s by Friedenstein et al., a growing mass of reports were conducted for a better definition of these cells and for the reevaluation from the term "mesenchymal stem cells" to the term "mesenchymal stromal cells (MSCs)." Being more than a semantic shift, concepts behind this new terminology reveal the complexity and the heterogeneity of the cells grouped in MSC family especially as these cells are present in nearly all adult tissues. Recently, mesenchymal stromal cell antigen-1 (MSCA-1)/tissue nonspecific alkaline phosphatase (TNAP) was described as a new cell surface marker of MSCs from different tissues. The alkaline phosphatase activity of this protein could be involved in wide range of MSC features described below from cell differentiation to immunomodulatory properties, as well as occurrence of pathologies. The present review aims to decipher and summarize the role of TNAP in progenitor cells from different tissues focusing preferentially on brain, bone marrow, and adipose tissue.

14.
Biochimie ; 124: 112-123, 2016 May.
Article in English | MEDLINE | ID: mdl-26363102

ABSTRACT

The primary cilium is an organelle present in most of the cells of the organism. Ciliopathies, such as the Bardet Biedl and the Alstrom syndromes are associated with obesity. We, and others, have shown that the primary cilium undergoes size modifications during adipocyte differentiation of human adipose stromal cells. We show here that the levels of acetylated α-tubulin, a constituent of the primary cilium, and the expression of HDAC6, the enzyme that deacetylates α-tubulin and is responsible for the loss of the cilium during mitosis, are modulated during adipogenesis. Moreover, during adipocyte differentiation cells that express higher level of HDAC6 are the first to lose their primary cilium. We have investigated the function of HDAC6 on adipocyte differentiation and on the primary cilium. We observe that inhibition of HDAC6 activity leads to a decrease in adipocyte differentiation. This is associated with an inhibition of the initial elongation of the cilium. Interestingly, overexpression of HDAC6 inhibits adipocyte differentiation and blunts the elongation of the primary cilium. In both situations, inhibition of adipocyte differentiation was not associated with an inhibition of the glucocorticoid receptor activity. This indicates that HDAC6 controls adipogenesis through the levels of acetylated α-tubulin. Moreover, we show that although HDAC6 expression increases during adipocyte differentiation it is not sufficient to provoke the loss of the cilium. This suggests the existence of a novel mechanism for the loss of the cilium. Together, these data indicate that HDAC6, and acetylated α-tubulin, are important regulator of adipocyte differentiation.


Subject(s)
Adipocytes/metabolism , Adipogenesis/physiology , Cell Differentiation/physiology , Histone Deacetylases/metabolism , Tubulin/metabolism , Acetylation , Adipocytes/cytology , Cell Line , Cilia/metabolism , Histone Deacetylase 6 , Humans
15.
Stem Cells ; 33(8): 2564-73, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25827082

ABSTRACT

Adipose tissue expansion is well-orchestrated to fulfill the energy demand. It results from adipocyte hypertrophy and hyperplasia due to adipose progenitor cell (APC) expansion and differentiation. Chronic low grade inflammation and hypoxia take place in obese adipose tissue microenvironment. Both of these events were shown to impact the APC pool by promoting increased self-renewal along with a decrease in the APC differentiation potential. However, no common target has been identified so far. Here we show that the immediate early response 3 gene (IER3) is preferentially expressed in APCs and is essential for APC proliferation and self-renewal. Experiments based on RNA interference revealed that impairing IER3 expression altered cell proliferation through ERK1/2 phosphorylation and clonogenicity. IER3 expression was induced by Activin A, which plays a crucial role in adipocyte differentiation as well as by a decrease in oxygen tension through HIF1-induced transcriptional activation. Interestingly, high levels of IER3 were detected in native APCs (CD34+/CD31- cells) isolated from obese patients and conditioned media from obese adipose tissue-macrophages stimulated its expression. Overall, these results indicate that IER3 is a key player in expanding the pool of APC while highlighting the role of distinct effectors found in an obese microenvironment in this process.


Subject(s)
Adipose Tissue/metabolism , Apoptosis Regulatory Proteins/biosynthesis , Gene Expression Regulation/physiology , MAP Kinase Signaling System/physiology , Membrane Proteins/biosynthesis , Stem Cell Niche/physiology , Stem Cells/metabolism , Adipose Tissue/cytology , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Stem Cells/cytology
16.
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
17.
J. physiol. biochem ; 69(3): 575-583, sept. 2013.
Article in English | IBECS | ID: ibc-121676

ABSTRACT

Human abdominal adipose tissue (AAT) can be divided into two compartments according to anatomical location to dermis layer, i.e. superficial and deep compartments (sAAT and dAAT). In morbidly obese patients, dAAT mass has been linked to obesity-associated pathologies. In the present study, we characterized in overweight healthy individuals human sAAT and dAAT cellular composition and adipogenic potential. Twelve paired sAAT and dAAT samples were collected. sAAT compared to dAAT adipocytes are larger. In agreement with increased size, real-time PCR analyses performed on isolated adipocytes showed that sAAT adipocytes exhibited higher leptin transcript levels but also higher expression of genes involved in metabolism including hormone-sensitive lipase compared to dAAT adipocytes. Flow cytometry analyses performed on stroma-vascular fraction (SVF) showed no difference in the numbers of progenitor cells, endothelial cells and macrophages between sAAT and dAAT. Macrophage phenotypes were not distinct between both AAT compartments. However, CD3+ T lymphocyte number was higher in dAAT than in sAAT. Adipogenic potential of dAAT SVF was lower than sAAT SVF whereas the one of isolated progenitor cells was not distinct whatever the AAT compartments. Therefore, in overweight patients, both sAAT and dAAT compartments exhibit differences in terms of adipocytes and T lymphocyte accumulation. dAAT is characterized by higher T lymphocyte accumulation together with smaller less metabolically active adipocytes. The lower adipogenic potential of dAAT SVF is not due to intrinsic progenitor cell properties but more likely to the increased T lymphocyte accumulation (AU)


Subject(s)
Humans , Adipose Tissue/ultrastructure , Subcutaneous Fat/ultrastructure , Overweight , Obesity, Morbid , Obesity, Abdominal , Stem Cells/physiology , T-Lymphocytes/physiology
18.
J Physiol Biochem ; 69(3): 575-83, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23184733

ABSTRACT

Human abdominal adipose tissue (AAT) can be divided into two compartments according to anatomical location to dermis layer, i.e. superficial and deep compartments (sAAT and dAAT). In morbidly obese patients, dAAT mass has been linked to obesity-associated pathologies. In the present study, we characterized in overweight healthy individuals human sAAT and dAAT cellular composition and adipogenic potential. Twelve paired sAAT and dAAT samples were collected. sAAT compared to dAAT adipocytes are larger. In agreement with increased size, real-time PCR analyses performed on isolated adipocytes showed that sAAT adipocytes exhibited higher leptin transcript levels but also higher expression of genes involved in metabolism including hormone-sensitive lipase compared to dAAT adipocytes. Flow cytometry analyses performed on stroma-vascular fraction (SVF) showed no difference in the numbers of progenitor cells, endothelial cells and macrophages between sAAT and dAAT. Macrophage phenotypes were not distinct between both AAT compartments. However, CD3+ T lymphocyte number was higher in dAAT than in sAAT. Adipogenic potential of dAAT SVF was lower than sAAT SVF whereas the one of isolated progenitor cells was not distinct whatever the AAT compartments. Therefore, in overweight patients, both sAAT and dAAT compartments exhibit differences in terms of adipocytes and T lymphocyte accumulation. dAAT is characterized by higher T lymphocyte accumulation together with smaller less metabolically active adipocytes. The lower adipogenic potential of dAAT SVF is not due to intrinsic progenitor cell properties but more likely to the increased T lymphocyte accumulation.


Subject(s)
Adipocytes/pathology , Leptin/metabolism , Overweight/pathology , RNA, Messenger/metabolism , Subcutaneous Fat/pathology , Adipocytes/metabolism , Adult , Cell Count , Endothelial Cells/cytology , Female , Gene Expression , Humans , Leptin/genetics , Macrophages/cytology , Male , Middle Aged , Organ Specificity , Overweight/metabolism , RNA, Messenger/genetics , Stem Cells/cytology , Sterol Esterase/genetics , Sterol Esterase/metabolism , Subcutaneous Fat/metabolism , T-Lymphocytes/cytology
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...