Skip to main content
Molecular Genetics and Metabolism Reports logoLink to Molecular Genetics and Metabolism Reports
. 2017 May 27;12:51–56. doi: 10.1016/j.ymgmr.2017.05.008

Adipose stem cells from obese patients show specific differences in the metabolic regulators vitamin D and Gas5

Laura M Pérez a,b, Beatriz de Lucas a,b, Victoria V Lunyak c, Beatriz G Gálvez a,b,
PMCID: PMC5447652  PMID: 28580301

Abstract

Adipose tissue is a significant source of mesenchymal stem cells for regenerative therapies; however, caution should be taken as their environmental niche can affect their functional properties. We have previously demonstrated the negative impact of obesity on the function of adipose-derived stem cells (ASCs). Here we have evaluated other possible properties and targets that are altered by obesity such as the recently described long non-coding molecule Gas5, which is involved in glucocorticoid resistance. Using ASCs isolated from obese (oASCs) and control subjects (cASCs), we have analyzed additional metabolic and inflammatory conditions that could be related with their impaired therapeutic potential and consequently their possible usefulness in the clinic.

Keywords: Obesity, Adipose stem cells, Inflammation, Glucocorticoids, Gas5

Highlights

  • Altered genetic and metabolic targets by obesity in adipose stem cells population

  • Gas5 involved in glucocorticoid resistance such as altered target

  • Additional metabolic and inflammation conditions on obese adipose stem cells

1. Introduction

Adipose tissue (AT) is a multifunctional and highly dynamic organ that, beyond its traditional role as a lipid storage site, is also a major endocrine organ [1] and an abundant source of stem cells [2]. AT secretes numerous factors, termed adipocytokines, which can originate from any cellular compartment (adipocytes, preadipocytes, immune cells, etc.,), and are major regulators of AT metabolism [3], [4]. AT also serves as a novel source of adult stem cells, termed adipose stem cells (ASCs), which have great potential for therapeutic applications [5]. However, we and others have shown that the environmental niche in which ASCs reside has a profound impact on their functional properties [6], [7], [8].

Changes to AT mass are associated with endocrine and metabolic dysfunction and are linked to obesity [9] and a chronic inflammatory milieu [10], resulting in an altered cytokine profile that is often accompanied by insulin resistance and dyslipidemia [11]. Obese individuals present increased circulating levels of several inflammation markers [12], [13] including Il-6, IL-8 and IL-1b, whose origin in AT is primarily the nonadipocyte component [14]. This overproduction of proinflammatory cytokines and the absence of anti-inflammatory cytokines contribute to the pathophysiology of obesity. AT-derived inflammatory adipocytokines are also secreted into systemic circulation where they exert profound effects on the hypothalmic-pituritary-adrenal axis by upregulating the expression of corticotropin-releasing hormone (CRH) and increasing the production of glucocorticoids (GCs) [15].

GCs have well-established effects on the metabolic regulation of AT homeostasis [16], [17], and are required for glucose and fatty acid metabolism [11], [18]. They also play an important role in inflammation associated with obesity since the glucocorticoid biosynthetic pathway enzyme, 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), is upregulated selectively in AT in obesity. While circulating GC levels are normal in obesity [19], 11β-HSD1 converts inactive cortisone back to active cortisol in AT [20], and increasing local cortisol levels are implicated in the pathogenesis of the metabolic syndrome [21]. The action of GCs on target cells is thought to be mediated by the glucocorticoid receptor (GR), whose binding by GC agonists triggers its translocation into the nucleus [22] where it dimerizes and binds directly to glucocorticoid-responsive elements (GREs) to stimulate the expression and regulation of different genes. GR can be regulated by other factors, such as the recently discovered Gas5 component.

Long noncoding RNAs (lncRNAs) are a novel class of functional RNAs that control gene regulation [23]. Many of them are associated with differentiation and homeostasis of metabolic tissues, such as the growth arrest-specific 5 (Gas5) lncRNA, which is induced under cellular stress or starvation conditions [24]. This lncRNA is a transcript of Gas5, a non-protein coding gene [25], which acts as a repressor of the GR through binding of a decoy GRE to ligand-activated GR [26], thereby suppressing the transcriptional activity of GC-responsive genes. Along this line, vitamin D deficiency is known to be related to obesity [27], and vitamin D can influence adipocytokine production and inflammation in AT [28].

Several independent lines of evidence demonstrate the dysregulated function of ASCs isolated from AT of obese patients [8], [29]. We have also demonstrated impaired differentiation [30], migration [31] and metabolic properties [6], [32] of ASCs isolated from obese individuals. Here, we examined additional metabolic factors in this stem cell population that may have utility for their functional characterization.

2. Materials and methods

2.1. Reagents

Dulbeco's modified Eagle's medium (DMEM) and fetal bovine serum (FBS) was purchased from Sigma (St. Louis MO). Penicillin, streptomycin, l-glutamine and Hepes was from Lonza (Basel, Switzerland). Kits for molecular studies were purchased from Applied Biosystems, Life Technologies (Paisley, UK). Unless otherwise stated, all other reagents were purchased from Sigma-Aldrich.

2.2. Isolation, culture and adipogenesis of ASCs

Human adipose-derived stem cells (ASCs) from non-obese and obese (body mass index < 22 kg/mg2 and > 30 kg/m2, respectively) patients were isolated and characterized as described [30], [33]. Subcutaneous adipose tissue was obtained from patients after bariatric surgery (obese patients) or normal surgery (non-obese patients) at the Hospital Universitario de la Princesa, Madrid (females aged 35–45 years; 5 obese and 5 non-obese). Sample collection conformed to the principles of the Declaration of Helsinki and the NIH Belmont Report. The ethics committee of the Centro Nacional de Investigaciones Cardiovasculares approved the use of human samples. Cells were isolated, sorted and expanded as described [34], and cultured in DMEM supplemented with 10% FBS at 37 °C in a humidified 5% CO2/95% air atmosphere. Adipogenic differentiation was performed using 2 × 104 cells plated into 24-well plates in DMEM/FBS. After 24 h, the medium was exchanged for adipocyte growth medium (STEMPRO Adipogenesis Differentiation Kit) supplemented with 1 μM dexamethasone and cells were cultured for up to 7 d. Triglyceride accumulation was visualized by staining paraformaldehyde (PFA)-fixed cells with Oil Red O, and images were taken with an IX71 inverted microscope.

2.3. Flow cytometry analysis

ASCs were stained with the following antibodies against surface markers: CD45-V450, CD34-PE, CD44-Alexa488, Sca1-PeCy7 and ckit-APC, all at 1:100 (BD Biosciences, San Jose, CA, USA). After incubation for 25 min at 4 °C, stained cells were analyzed by flow cytometry on an LSRFortessa flow cytometer (BD Biosciences, San Jose, CA). Data were analyzed with BD FACSDiva Software.

2.4. Quantitative real-time polymerase chain reaction

Total RNA was extracted from ASCs with TRI Reagent and reverse transcribed using the High Capacity cDNA Reverse Transcription Kit. Quantitative real-time polymerase chain reaction (qRT-PCR) was performed using the primers shown in Table 1. Each cDNA sample was amplified in triplicate using a SYBR Green PCR Master Mix. PCR mixes were loaded in an AB 7900 Fast Real-Time PCR System and quantified using SDS 2.0 software.

Table 1.

Primer sequences for qRT-PCR.

Primer forward sequence (5′-3′) Primer reverse sequence (5′-3′)
β-Actin CACGATGGAGGGGCCGGACTCAT TAAAGACCTCTATGCCAACACAG
Gas5 AGCTGGAAGTTGAAATGG CAAGCCGACTCTCCATACC
IL-1 GGGCCTCAAGGAAAAGAATC TTCTGCTTGAGAGGTGCTGA
IL-6 TACCCCCAGGAGAAGATTCC TTTTCTGCCAGTGCCTCTTT
IL-8 GTGCAGTTTTGCCAAGGAGT CTCTGCACCCAGTTTTCCTT
IL-10 TGCCTTCAGCAGAGTGAAGA GGTCTTGGTTCTCAGCTTGG
MCP-1 CCCCAGTCACCTGCTGTTAT TGGAATCCTGAACCCACTTC
Icam-1 GGCTGGAGCTGTTTGAGAAC ACTGTGGGGTTCAACCTCTG

2.5. Vitamin D analysis

Cells and culture medium were separated by centrifugation at 5725 × g for 5 min at 4 °C. Cell pellets containing equal numbers of each cell line were subjected to three freeze-thaw cycles as described [6] to obtain cellular extracts. Culture medium and cellular extracts were prepared for liquid chromatography-mass spectrometry (LC-MS) to quantify total vitamin D derivatives as described [32]. Samples for each technique were prepared with replicates as quality control. Values are expressed as the “percentage of change” between groups.

2.6. Data analysis

Statistical analysis was performed using the GraphPad Prism software package (GraphPad, San Diego, CA). Comparison between groups was performed by one-way or two-way analysis of variance as required by the assay. Values were expressed as mean ± SEM or mean ± SD, and data was considered significantly different at p < 0.05. Analysis of LC-MS data was carried out by univariate (UVA) and multivariate (MVA) analysis using MATLAB R2015 software (Mathworks, Inc., Natick, USA) and SIMCA P + 12.0.1 software (Umetrics, Umea, Sweden), respectively. For UVA, the non-parametric Mann-Whitney U test with Benjamini-Hochberg post hoc correction (level q < 0.05) was applied, and for MVA, log-transformed and Pareto-scaled data, or just Pareto-scaled data, were used to create multivariate models.

3. Results

3.1. Adipose stem cell isolation and characterization

ASCs derived from subcutaneous depots from non-obese (cASC) and obese (oASC) donors were isolated using an explant method [30], [33]. Cells were characterized by flow cytometry and were positive for the surface markers CD34, CD44, Sca1 and ckit and negative for CD45 (Fig. 1A). Comparable ASC populations were obtained from non-obese and obese subjects as characterized by specific stem cell markers [34].

Fig. 1.

Fig. 1

A. Analysis of cell surface markers by flow cytometry in adipose stem cells from control (cASCs) and obese (oASCs) subjects. Positively stained cells are expressed as a percentage in the plot. Histograms show isotype-matched controls and fluorescence intensity of each cell surface marker and are representative of three independent experiments with similar results. B. Representative image of Oil Red O staining in cASCs and oASCs differentiated for 7 days into adipocytes. Scale bar, 30 μm. C. Gas5 gene expression by cASCs and oASCs under basal conditions (− diff) and at day 7 differentiation (+ diff). ***p < 0.001 (Anova). D. Vitamin D quantification. Graph represents the percentage of change within the cells and in the medium. + p < 0.05. E. Gene expression profile of different cytokines in cASCs and oASCs. *p < 0.05.

3.2. Gas5 expression is enhanced during adipogenesis

We examined Gas5 expression in ASCs grown under normal conditions and after adipocyte differentiation. No differences in the levels of Gas5 mRNA were found between cASC and oASC under basal conditions (Fig. 1B). We induced adipocyte differentiation by culturing cells in STEMPRO medium with dexamethasome. After 7 days, differentiation was confirmed by the presence of accumulated triglycerides evaluated with Oil red O (Fig. 1C), and Gas5 expression was measured. Whereas adipogenesis increased Gas5 expression both in cASC and oASC 4–5-fold, the increase was significantly lower in oASC (Fig. 1B).

3.3. Inflammatory and metabolic conditions

Obesity is associated with insufficiency of vitamin D [35], which acts an antioxidant and has pleiotropic effects including an anti-inflammatory action. Having shown that oASCs have an altered metabolic profile [6], [32], we assessed cellular and secreted vitamin D levels in the two ASC groups. The level of cellular vitamin D was significantly lower (60%) in oASC than in cASC, and this was paralleled by a significant decrease in secreted levels (30%) in oASC (Fig. 1D). This result is line with the findings that individuals with obesity present a reduction in vitamin D.

An inverse relationship between vitamin D levels and inflammation markers has been described in obese patients [36]. To determine whether obesity and vitamin D deficiency resulted in increased cytokine production in ASCs, we measured the expression pattern of a panel of cytokines and Icam-1. Expression of IL-6, IL-8, IL-10 and MCP-1 was higher in oASC than in cASC, whereas no differences in expression were detected for IL-1 and Icam-1. These results suggest an increased inflammatory response in oASC due to obesity and vitamin D reduction.

4. Discussion

While ASCs hold great promise for regenerative medicine applications [37], obesity leads to a reduction in the ASC pool [38], [39] and we have previously shown that AT from obese donors has a negative effect on the ASC population, impacting their stemness and metabolic properties [6], [30], [31].

We show here that whereas oASC and cASC have similar Gas5 expression levels under basal conditions, expression is lower in oASC than cASC during adipogenic differentiation. Gas5 levels are increased during adipogenic differentiation due to the presence of dexamethasone in the medium, but this synthetic glucocorticoid presents a reduced action on oASCs.

Some studies have suggested an association between obesity and vitamin D deficiency, as obese individuals tend to have low vitamin D levels [35]. We found a decrease in vitamin D levels in oASC, both in the medium and in cells, confirming this association. Additionally, obesity results in a blood flow imbalance leading to inflammation, and macrophage infiltration [40], and it has been demonstrated that vitamin D metabolites influence adipcytokine production and the inflammatory response in AT [28]. Accordingly, we detected a cytokine profile imbalance between oASC and cASCs. Thus, obesity affects vitamin D levels and promotes an altered cytokine profile that is inflammatory. It remains to be tested whether vitamin D decreases the release of the main pro-inflammatory molecules in AT.

Our findings allow us to propose the following model (Fig. 2). Obesity provokes a marked inflammatory process within AT, principally in macrophages [41]. This inflammation is associated with changes in the microenvironment (red arrow Fig. 2) characterized by abnormal adipocytokine production and proinflammatory signaling pathway activation [9]. In parallel, GC levels increase due to enhanced levels of 11βHSD1 [11], which results in an altered metabolism due to GC-regulated pathways such as glucose [42] and lipid [43] metabolism. Furthermore, it has been reported that GCs act synergistically with insulin, and can improve or impair their action [11]. GCs are regulated by the noncoding RNA Gas5, which is an associated repressor of the GR [44]. During dexamethasome-induced lipogenesis, Gas5 levels increase to repress GC action [45], but Gas5 levels in oASC are lower than in cASCs (Fig. 1C), hindering lipid metabolism. We have previously shown that ASCs isolated from obese subjects have significantly enhanced apoptosis and also a reduced proliferative capacity [6]. In this regard, GCs regulate apoptosis and exert significant anti-proliferative effects [46], and GC dysregulation supports our previous findings in relation to apoptosis and proliferation.

Fig. 2.

Fig. 2

Adipose tissue inflammation in obese conditions induces alterations in adipose stem cells. Cytokines are increased in obese conditions, triggering an enhanced glucocorticoid production as an anti-inflammatory response. Dexamethasone and adipogenic components promote adipogenic differentiation and induce an altered response in different metabolic pathways. Dex, Dexamethasone; Gas5, Growth arrest-specific 5; GC, Glucocorticoids; GR, Glucocorticoid receptor; GRE Glucocorticoid response element; Glc, Glucose; GLUT, Glucose transporter; Ins, Insulin; IRS, Insulin receptor substrate; VitD, Vitamin D.

Several in vitro studies have demonstrated that vitamin D inhibits chronic inflammation in AT [47]; however, vitamin D levels are decreased in obese subjects [35]. The present findings are in accordance with this and we hypothesize that low amounts of vitamin D may increase the metabolic risk and contribute to the inflammatory process. Furthermore, vitamin D enhances GC responsiveness by increasing its anti-inflammatory activity [48], and we found a misbalance in those components in oASCs. Concomitant with this, we detected increased cytokine expression, and it has been described that chronic activation of pro-inflammatory pathways within AT may impair metabolic control [49]. Thus, the increased inflammatory state and GC release activate anti-inflammatory pathways and GCs down-regulate the expression of several inflammatory genes, including those encoding cytokines [50].

ASCs have been used in several studies and preclinical data indicate that ASCs present high safety and efficacy, supporting their use in cell-based regenerative medicine. The data in the present study confirm and extend our previous findings showing that ASCs present different biological properties due to the obese microenvironmental niche. Obesity-derived signals, inflammatory and others, play a key role in ASC impairment and our present findings can help us to understand the mechanisms responsible for these metabolic alterations.

Funding

This study was supported by a grant from the Spanish Ministry of Economy and Competitiveness (SAF 2015-67911) to BGG. BdL is supported by FPU fellowship from the Spanish Ministry.

Acknowledgements

We would like to thank Dr. Kenneth McCreath for critically reviewing the manuscript.

L.M.P. and B.dL. researched data. All authors contributed to discussion and reviewed the manuscript. L.M.P. and B.G.G. wrote the manuscript and discussed data.

References

  • 1.Coelho M., Oliveira T., Fernandes R. Biochemistry of adipose tissue: an endocrine organ. Arch. Med. Sci. 2013;9:191–200. doi: 10.5114/aoms.2013.33181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gimble J.M., Katz A.J., Bunnell B.A. Adipose-derived stem cells for regenerative medicine. Circ. Res. 2007;100:1249–1260. doi: 10.1161/01.RES.0000265074.83288.09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Coppack S.W. Pro-inflammatory cytokines and adipose tissue. Proc. Nutr. Soc. 2001;60:349–356. doi: 10.1079/pns2001110. [DOI] [PubMed] [Google Scholar]
  • 4.Greenberg A.S., Obin M.S. Obesity and the role of adipose tissue in inflammation and metabolism. Am. J. Clin. Nutr. 2006;83:461S–465S. doi: 10.1093/ajcn/83.2.461S. [DOI] [PubMed] [Google Scholar]
  • 5.Dai R., Wang Z., Samanipour R., Koo K.I., Kim K. Adipose-derived stem cells for tissue engineering and regenerative medicine applications. Stem Cells Int. 2016;2016:6737345. doi: 10.1155/2016/6737345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Perez L.M., Bernal A., de Lucas B., San Martin N., Mastrangelo A., Garcia A., Barbas C., Galvez B.G. Altered metabolic and stemness capacity of adipose tissue-derived stem cells from obese mouse and human. PLoS One. 2015;10 doi: 10.1371/journal.pone.0123397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Pachon-Pena G., Serena C., Ejarque M., Petriz J., Duran X., Oliva-Olivera W., Simo R., Tinahones F.J., Fernandez-Veledo S., Vendrell J. Obesity determines the immunophenotypic profile and functional characteristics of human mesenchymal stem cells from adipose tissue. Stem Cells Transl. Med. 2016;5:464–475. doi: 10.5966/sctm.2015-0161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Onate B., Vilahur G., Camino-Lopez S., Diez-Caballero A., Ballesta-Lopez C., Ybarra J., Moscatiello F., Herrero J., Badimon L. Stem cells isolated from adipose tissue of obese patients show changes in their transcriptomic profile that indicate loss in stemcellness and increased commitment to an adipocyte-like phenotype. BMC Genet. 2013;14:625. doi: 10.1186/1471-2164-14-625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Jung U.J., Choi M.S. Obesity and its metabolic complications: the role of adipokines and the relationship between obesity, inflammation, insulin resistance, dyslipidemia and nonalcoholic fatty liver disease. Int. J. Mol. Sci. 2014;15:6184–6223. doi: 10.3390/ijms15046184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Bastard J.P., Maachi M., Lagathu C., Kim M.J., Caron M., Vidal H., Capeau J., Feve B. Recent advances in the relationship between obesity, inflammation, and insulin resistance. Eur. Cytokine Netw. 2006;17:4–12. [PubMed] [Google Scholar]
  • 11.Lee M.J., Pramyothin P., Karastergiou K., Fried S.K. Deconstructing the roles of glucocorticoids in adipose tissue biology and the development of central obesity. Biochim. Biophys. Acta. 2014;1842:473–481. doi: 10.1016/j.bbadis.2013.05.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Ouchi N., Parker J.L., Lugus J.J., Walsh K. Adipokines in inflammation and metabolic disease. Nat. Rev. Immunol. 2011;11:85–97. doi: 10.1038/nri2921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Fain J.N. Release of interleukins and other inflammatory cytokines by human adipose tissue is enhanced in obesity and primarily due to the nonfat cells. Vitam. Horm. 2006;74:443–477. doi: 10.1016/S0083-6729(06)74018-3. [DOI] [PubMed] [Google Scholar]
  • 14.Trayhurn P., Wood I.S. Adipokines: inflammation and the pleiotropic role of white adipose tissue. Br. J. Nutr. 2004;92:347–355. doi: 10.1079/bjn20041213. [DOI] [PubMed] [Google Scholar]
  • 15.Hermoso M.A., Cidlowski J.A. Putting the brake on inflammatory responses: the role of glucocorticoids. IUBMB Life. 2003;55:497–504. doi: 10.1080/15216540310001642072. [DOI] [PubMed] [Google Scholar]
  • 16.Asensio C., Muzzin P., Rohner-Jeanrenaud F. Role of glucocorticoids in the physiopathology of excessive fat deposition and insulin resistance. Int. J. Obes. Relat. Metab. Disord. 2004;28(Suppl. 4):S45–S52. doi: 10.1038/sj.ijo.0802856. [DOI] [PubMed] [Google Scholar]
  • 17.Gruver-Yates A.L., Cidlowski J.A. Tissue-specific actions of glucocorticoids on apoptosis: a double-edged sword. Cell. 2013;2:202–223. doi: 10.3390/cells2020202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Macfarlane D.P., Forbes S., Walker B.R. Glucocorticoids and fatty acid metabolism in humans: fuelling fat redistribution in the metabolic syndrome. J. Endocrinol. 2008;197:189–204. doi: 10.1677/JOE-08-0054. [DOI] [PubMed] [Google Scholar]
  • 19.Hautanen A., Raikkonen K., Adlercreutz H. Associations between pituitary-adrenocortical function and abdominal obesity, hyperinsulinaemia and dyslipidaemia in normotensive males. J. Intern. Med. 1997;241:451–461. doi: 10.1111/j.1365-2796.1997.tb00002.x. [DOI] [PubMed] [Google Scholar]
  • 20.Tomlinson J.W., Sinha B., Bujalska I., Hewison M., Stewart P.M. Expression of 11beta-hydroxysteroid dehydrogenase type 1 in adipose tissue is not increased in human obesity. J. Clin. Endocrinol. Metab. 2002;87:5630–5635. doi: 10.1210/jc.2002-020687. [DOI] [PubMed] [Google Scholar]
  • 21.Esteves C.L., Kelly V., Begay V., Man T.Y., Morton N.M., Leutz A., Seckl J.R., Chapman K.E. Regulation of adipocyte 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) by CCAAT/enhancer-binding protein (C/EBP) beta isoforms, LIP and LAP. PLoS One. 2012;7 doi: 10.1371/journal.pone.0037953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Oakley R.H., Cidlowski J.A. Cellular processing of the glucocorticoid receptor gene and protein: new mechanisms for generating tissue-specific actions of glucocorticoids. J. Biol. Chem. 2011;286:3177–3184. doi: 10.1074/jbc.R110.179325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kornfeld J.W., Bruning J.C. Regulation of metabolism by long, non-coding RNAs. Front. Genet. 2014;5:57. doi: 10.3389/fgene.2014.00057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Huarte M., Rinn J.L. Large non-coding RNAs: missing links in cancer? Hum. Mol. Genet. 2010;19:R152–R161. doi: 10.1093/hmg/ddq353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Coccia E.M., Cicala C., Charlesworth A., Ciccarelli C., Rossi G.B., Philipson L., Sorrentino V. Regulation and expression of a growth arrest-specific gene (gas5) during growth, differentiation, and development. Mol. Cell. Biol. 1992;12:3514–3521. doi: 10.1128/mcb.12.8.3514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Mayama T., Marr A.K., Kino T. Differential expression of glucocorticoid receptor noncoding RNA repressor Gas5 in autoimmune and inflammatory diseases. Horm. Metab. Res. 2016 doi: 10.1055/s-0042-106898. [DOI] [PubMed] [Google Scholar]
  • 27.Foss Y.J. Vitamin D deficiency is the cause of common obesity. Med. Hypotheses. 2009;72:314–321. doi: 10.1016/j.mehy.2008.10.005. [DOI] [PubMed] [Google Scholar]
  • 28.Ding C., Gao D., Wilding J., Trayhurn P., Bing C. Vitamin D signalling in adipose tissue. Br. J. Nutr. 2012;108:1915–1923. doi: 10.1017/S0007114512003285. [DOI] [PubMed] [Google Scholar]
  • 29.Roldan M., Macias-Gonzalez M., Garcia R., Tinahones F.J., Martin M. Obesity short-circuits stemness gene network in human adipose multipotent stem cells. FASEB J. 2011;25:4111–4126. doi: 10.1096/fj.10-171439. [DOI] [PubMed] [Google Scholar]
  • 30.Perez L.M., Bernal A., San Martin N., Lorenzo M., Fernandez-Veledo S., Galvez B.G. Metabolic rescue of obese adipose-derived stem cells by Lin28/Let7 pathway. Diabetes. 2013;62:2368–2379. doi: 10.2337/db12-1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Perez L.M., Bernal A., San Martin N., Galvez B.G. Obese-derived ASCs show impaired migration and angiogenesis properties. Arch. Physiol. Biochem. 2013;119:195–201. doi: 10.3109/13813455.2013.784339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Mastrangelo A., Panadero M.I., Perez L.M., Galvez B.G., Garcia A., Barbas C., Ruperez F.J. New insight on obesity and adipose-derived stem cells by comprehensive metabolomics. Biochem. J. 2016 doi: 10.1042/BCJ20160241. [DOI] [PubMed] [Google Scholar]
  • 33.Galvez B.G., San Martin N., Rodriguez C. TNF-alpha is required for the attraction of mesenchymal precursors to white adipose tissue in Ob/ob mice. PLoS One. 2009;4:e4444. doi: 10.1371/journal.pone.0004444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Rodeheffer M.S., Birsoy K., Friedman J.M. Identification of white adipocyte progenitor cells in vivo. Cell. 2008;135:240–249. doi: 10.1016/j.cell.2008.09.036. [DOI] [PubMed] [Google Scholar]
  • 35.Fish E., Beverstein G., Olson D., Reinhardt S., Garren M., Gould J. Vitamin D status of morbidly obese bariatric surgery patients. J. Surg. Res. 2010;164:198–202. doi: 10.1016/j.jss.2010.06.029. [DOI] [PubMed] [Google Scholar]
  • 36.Pojednic R.M., Ceglia L., Lichtenstein A.H., Dawson-Hughes B., Fielding R.A. Vitamin D receptor protein is associated with interleukin-6 in human skeletal muscle. Endocrine. 2015;49:512–520. doi: 10.1007/s12020-014-0505-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Lindroos B., Suuronen R., Miettinen S. The potential of adipose stem cells in regenerative medicine. Stem Cell Rev. 2011;7:269–291. doi: 10.1007/s12015-010-9193-7. [DOI] [PubMed] [Google Scholar]
  • 38.Tchoukalova Y., Koutsari C., Jensen M. Committed subcutaneous preadipocytes are reduced in human obesity. Diabetologia. 2007;50:151–157. doi: 10.1007/s00125-006-0496-9. [DOI] [PubMed] [Google Scholar]
  • 39.De Girolamo L., Stanco D., Salvatori L., Coroniti G., Arrigoni E., Silecchia G., Russo M.A., Niada S., Petrangeli E., Brini A.T. Stemness and osteogenic and adipogenic potential are differently impaired in subcutaneous and visceral adipose derived stem cells (ASCs) isolated from obese donors. Int. J. Immunopathol. Pharmacol. 2013;26:11–21. doi: 10.1177/03946320130260S103. [DOI] [PubMed] [Google Scholar]
  • 40.Trayhurn P. Hypoxia and adipose tissue function and dysfunction in obesity. Physiol. Rev. 2013;93:1–21. doi: 10.1152/physrev.00017.2012. [DOI] [PubMed] [Google Scholar]
  • 41.Weisberg S.P., McCann D., Desai M., Rosenbaum M., Leibel R.L., Ferrante A.W., Jr. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Investig. 2003;112:1796–1808. doi: 10.1172/JCI19246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ferris H.A., Kahn C.R. New mechanisms of glucocorticoid-induced insulin resistance: make no bones about it. J. Clin. Investig. 2012;122:3854–3857. doi: 10.1172/JCI66180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Peckett A.J., Wright D.C., Riddell M.C. The effects of glucocorticoids on adipose tissue lipid metabolism. Metab. Clin. Exp. 2011;60:1500–1510. doi: 10.1016/j.metabol.2011.06.012. [DOI] [PubMed] [Google Scholar]
  • 44.Lucafo M., De Iudicibus S., Di Silvestre A., Pelin M., Candussio L., Martelossi S., Tommasini A., Piscianz E., Ventura A., Decorti G. Long noncoding RNA GAS5: a novel marker involved in glucocorticoid response. Curr. Mol. Med. 2015;15:94–99. doi: 10.2174/1566524015666150114122354. [DOI] [PubMed] [Google Scholar]
  • 45.Kino T., Hurt D.E., Ichijo T., Nader N., Chrousos G.P. Noncoding RNA gas5 is a growth arrest- and starvation-associated repressor of the glucocorticoid receptor. Sci. Signal. 2010;3:ra8. doi: 10.1126/scisignal.2000568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Greenstein S., Ghias K., Krett N.L., Rosen S.T. Mechanisms of glucocorticoid-mediated apoptosis in hematological malignancies. Clin. Cancer Res. 2002;8:1681–1694. [PubMed] [Google Scholar]
  • 47.Mutt S.J., Hypponen E., Saarnio J., Jarvelin M.R., Herzig K.H. Vitamin D and adipose tissue-more than storage. Front. Physiol. 2014;5:228. doi: 10.3389/fphys.2014.00228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Zhang Y., Leung D.Y., Goleva E. Vitamin D enhances glucocorticoid action in human monocytes: involvement of granulocyte-macrophage colony-stimulating factor and mediator complex subunit 14. J. Biol. Chem. 2013;288:14544–14553. doi: 10.1074/jbc.M112.427054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Geer E.B., Islam J., Buettner C. Mechanisms of glucocorticoid-induced insulin resistance: focus on adipose tissue function and lipid metabolism. Endocrinol. Metab. Clin. N. Am. 2014;43:75–102. doi: 10.1016/j.ecl.2013.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Caldenhoven E., Liden J., Wissink S., Van de Stolpe A., Raaijmakers J., Koenderman L., Okret S., Gustafsson J.A., Van der Saag P.T. Negative cross-talk between RelA and the glucocorticoid receptor: a possible mechanism for the antiinflammatory action of glucocorticoids. Mol. Endocrinol. 1995;9:401–412. doi: 10.1210/mend.9.4.7659084. [DOI] [PubMed] [Google Scholar]

Articles from Molecular Genetics and Metabolism Reports are provided here courtesy of Elsevier

RESOURCES