Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Mar 1.
Published in final edited form as: Biochim Biophys Acta. 2013 May 16;1842(3):377–392. doi: 10.1016/j.bbadis.2013.05.006

Sex dimorphism and depot differences in adipose tissue function

Ursula A White 1, Yourka D Tchoukalova 1
PMCID: PMC3926193  NIHMSID: NIHMS481104  PMID: 23684841

Abstract

Obesity, characterized by excessive adiposity, is a risk factor for many metabolic pathologies, such as Type 2 Diabetes mellitus (T2DM). Numerous studies have shown that adipose tissue distribution may be a greater predictor of metabolic health. Upper-body fat (visceral and subcutaneous abdominal) is commonly associated with the unfavorable complications of obesity, while lower-body fat (gluteal-femoral) may be protective. Current research investigations are focused on analyzing the metabolic properties of adipose tissue, in order to better understand the mechanisms that regulate fat distribution in both men and women. This review will highlight the adipose tissue depot- and sex- dependent differences in white adipose tissue function, including adipogenesis, adipose tissue developmental patterning, the storage and release of fatty acids, and secretory function.

Keywords: Adipocyte, Adipose Tissue, Adipogenesis, Fat Distribution, Lipolysis, Fatty Acid Uptake, Adipokine

1. Introduction

Adipocytes are highly specialized cells that form and store fat in adipose tissue and play a major role in energy homeostasis in vertebrate organisms. Obesity results from an energy surplus and is characterized by an increased storage of lipid and expansion of adipose tissue. Obesity modifies the endocrine and metabolic functions of adipose tissue and is a risk factor for many other metabolic diseases, including Type 2 diabetes (T2DM), cardiovascular disease (CVD), atherosclerosis, and hypertension. No longer recognized as just a lipid-storage depot, white adipose tissue (WAT) has additional properties, including insulin sensitivity and secretory function, that contribute to the pathogenesis of obesity and T2DM (reviewed in [1]). Hence, it is well-established that WAT can significantly impact metabolic health, as its development and function greatly influence whole-body metabolism.

Though overall excessive adiposity is associated with serious co-morbidities, the distribution of body fat has been shown to be a stronger predictor of health risk [28]. Numerous studies have described differential correlations of WAT depots and metabolic risk in humans, presumably due to intrinsic differences in function of the adipose tissue [913]. The two types of WAT, visceral (VAT) and subcutaneous (SAT), are defined by location, and the mechanisms and developmental signals that account for each depot’s unique characteristics are steadily emerging. In human subjects, VAT is usually represented by the omental depot (although it also includes the mesenteric fat) and by the perigonadal depot (parametrial and epididymal fat pad) in rodents; while SAT is represented by the abdominal, gluteal and femoral depots in humans and by the inguinal fat pad in rodents. Studies have revealed that central/abdominal (subcutaneous upper body and visceral) fat deposition correlate with an increased susceptibility for metabolic complications [1418], while gluteal-femoral (lower body) adipose tissue is associated with reduced metabolic risk and may be protective against the adverse health effects of obesity in both sexes [16, 1923](reviewed in [24]). Current investigations are focused on elucidating the mechanisms, adipose tissue- secreted factors, and developmental signals that account for regional differences in WAT development and function. It is important to note that the developmental origin of white and brown adipose tissue is distinct (reviewed in [25, 26]), and the physiological role of brown fat in human body weight regulation and obesity-associated metabolic complications is controversial and has not been elucidated (reviewed in [27]). This review will provide an overview of the depot- and sex- dependent differences in important functions of white adipose tissue, such as adipogenesis, expression of developmental patterning transcription factors, the storage and release of fatty acids, and secretory function.

2. Sex Differences in Fat Distribution

Sex differences in adipose tissue distribution are well-supported by many literature findings and are associated with whole body metabolic health. Women generally have higher adiposity relative to men throughout the entire lifespan [28, 29]. However, men often have more adipose tissue distributed in the central or abdominal region (‘android’ or ‘apple’ phenotype), which carries a much greater risk for metabolic disorders [30](reviewed in [31] and [32]). In contrast, women are characterized by less VAT and more SAT [30, 3335], especially in the lower body (‘gynoid’ or ‘pear’ phenotype) [36, 37]. Likewise, though women can also possess the upper-body obese phenotype, the reduced metabolic disease risk in women has been attributed to the propensity to store body fat in the SAT depot, particularly in the gluteal-femoral region. Though the factors and mechanisms that govern this sexual dimorphism are not elucidated, these characteristic android and gynoid fat distribution patterns in men and women appear as early as puberty (reviewed in [38]). Therefore, evidence suggests that this distribution may be predominantly sex hormone-dependent [39] (reviewed in [40]).

Sex steroids are endogenous modulators of adipose tissue development and function, and may also influence, in part, the distribution of SAT versus VAT depots (reviewed in [41]), although little is known about the cellular and molecular mechanisms of this regulation. In menopausal women, who have a decline in circulating estrogen levels [4246], visceral adiposity increases, resulting from a shift towards a central/android body fat distribution. Postmenopausal women were shown to have a much greater increase in VAT volume as compared to premenopausal women [47, 48]. Furthermore, women who received hormone replacement therapy (HRT) had lower waist circumferences and VAT than those who did not receive therapy [4951]. Collectively, these studies suggest that estrogen may modulate WAT distribution by effectively reducing central adiposity in humans. Likewise, the deposition of fat in the gluteal-femoral SAT depots in women versus deposition in the VAT in men may be related to the higher level of estrogen in premenopausal women compared to men (reviewed in [52]).

Androgens may also have depot- and gender- specific effects on adipose tissue distribution. In men, as testosterone declines with age [53] and in polycystic ovary syndrome (PCOS) women, often characterized by a hyper-androgenic state [54, 55] (reviewed in [56]), there is an increase in VAT. Testosterone therapy in aging men decreased visceral fat mass and increased lean muscle mass [57, 58]. In women, testosterone levels correlated positively with significantly increased abdominal fat [59, 60]. In addition, obese postmenopausal women treated with testosterone developed significantly increased visceral fat [61, 62].

Numerous studies have demonstrated that both SAT and VAT in men and women express sex steroid receptors, notably the estrogen (ERα, ERβ and its variants, and G protein-coupled ER) and androgen (AR) receptors [6366]. There is also limited evidence to support regional and sex differences in the expression of sex hormone receptors. ERβ1 expression (mRNA and protein) was much reduced in VAT (intraabdominal) compared to SAT, whereas the expression of ERβ4 and ERβ5 mRNA levels were significantly higher in gluteal SAT compared to abdominal SAT in both men and women, with higher ERβ mRNA levels in women compared with men [67]. Recent findings by Gavin et al. support these results, as their analyses demonstrate that abdominal SAT contained more ERα protein compared to gluteal, and gluteal SAT contained more ERβ protein when compared to abdominal SAT from overweight premenopausal women collected during the follicular phase of the menstrual cycle [68]. Importantly, the waist-to hip ratio negatively correlates with the gluteal ERβ protein abundance and positively with the ERα/ ERβ ratio, indicating that ERs play a role in modulating regional fat distribution. In addition, ERα and ERβ have been reported to mediate distinct, depot-specific actions [66, 67, 69]. Estrogens in vitro up-regulate the expression of both ERα and ERβ mRNA in subcutaneous adipocytes from women, but only the ERα in subcutaneous and visceral adipocytes from men [66, 67, 69]. In addition, though the ERα subtype was present in preadipocytes, estrogens in vitro regulated the ER activity in differentiated but not in confluent preadipocytes, suggesting that the ER becomes functional during the course of adipogenesis [66, 67, 69]. Investigations utilizing knockout models have also yielded novel information regarding the specific roles for estrogen, as well as ERα and ERβ, in the maintenance of WAT lipid and glucose homeostasis [7073](reviewed in [74]). Hence, these studies provide evidence that estrogen may modulate fat accumulation in a depot- specific manner via the differential expression of ERs within WAT.

Sex-specific adipose tissue distribution may also result from the secretion of sex hormones from adipose tissue in a depot-specific manner. Though other tissues account for the majority of sex hormones in circulation, adipose tissue can also contribute a substantial amount of circulating estrogen and testosterone (reviewed in [75, 76]). Furthermore, human WAT depots possess the enzymes and intermediates necessary for sex hormone synthesis in a depot-specific manner (reviewed in [77, 78]). Collectively, these data suggest that both circulating and local adipose tissue production of sex hormones may have important effects on adipose tissue distribution (reviewed in [79]). Further studies are necessary to elucidate the regional effects of sex-steroid hormones in adipose tissue. Determining the expression of steroidogenic metabolizing enzymes and ERs in upper- and lower-body WAT and testing the effects of treatment with sex steroids on the cellular dynamic properties of preadipocytes in both sexes will further advance our understanding of the contribution of sex hormones to WAT metabolism.

3. Adipogenesis

3A. Definition

Although most development occurs during prenatal and early postnatal life (reviewed in [80]), WAT retains the ability to expand during adult life, especially to accommodate energy surplus. Adipose tissue expansion (adipogenesis) occurs in two ways- by increase of existing adipocytes’ size (hypertrophy) or by recruiting new fat cells (hyperplasia). The mechanisms that influence the pattern of expansion via hypertrophy and/or hyperplasia have not been elucidated, as individuals vary by the predominant depot-dependent cellular mechanisms that are involved. Preliminary evidence in rodents suggests that VAT (perigonadal) expands predominantly by adipocyte hypertrophy, while SAT (inguinal) by adipocyte hyperplasia following exposure to a high-fat diet [81]. Findings from Tchoukalova et al. have shown that SAT expansion in response to overfeeding resulted in higher adipocyte hypertrophy in the upper-body (abdominal) SAT depot, relative to femoral SAT depot, which had increased hyperplasia [82]. Indeed, adipocyte morphology is significant, as increased adipocyte size correlates with higher metabolic risk [8385].

Accumulating evidence in human subjects suggests that obesity complications result from the inability of SAT to expand and safely store lipids, which leads to ectopic deposition in other tissues, lipotoxicity, and insulin resistance (reviewed in [8690]). We have shown that the number of small, early-differentiated adipocytes, isolated from the stromal-vascular fraction (SVF) in SAT depots of normal weight men and women, correlates positively with subcutaneous adiposity (particularly in the femoral SAT), and negatively with the visceral fat accumulation [91]. These data indicate that the abundance of adipocytes in the SAT depots is an important predictor for SAT expendability. The aforementioned studies highlight the importance of proliferative adipocyte precursor cells that are capable of undergoing differentiation to supply new, mature adipocytes, in order to support the expansion of SAT, prevent the accumulation of fat in VAT, and maintain healthy adipose dynamics. Therefore, to safely accommodate metabolic demands, an adipocyte precursor pool is thought to remain present and undergo a constant cell turnover in WAT during adult life. Studies by Spalding et al. suggest that approximately 10% of the body’s adipocytes are regenerated each year [92]. In addition, adipocyte number can increase during the development of obesity in adulthood, despite a higher rate of apoptosis (cell death) [93]. Although the term adipogenesis refers to the proliferation and differentiation of adipocyte precursor cells, the apoptosis of adipocytes and their precursors is an important regulator of adipocyte cellularity and is, thus, also mentioned in this section.

3B. Methods for Assessment of Adipogenesis

Very few in vivo data is available regarding the mechanisms that modulate the commitment of adipocyte precursor cells to the preadipocyte, as well as the processes that control the formation of new adipocytes in human WAT. The established approaches to assess adipogenesis in WAT depots involve observing the changes in adipocyte size and/or number or analyzing fat cell size distribution, accompanied by the expression profiles of proteins or genes involved in adipogenesis [94, 95]. Alternative methods include functional assays and immunohistochemistry to determine adipocyte differentiation, proliferation, and susceptibility to apoptosis, using preadipocyte cell lines or primary adipose-derived stem cell cultures (reviewed in [96, 97]). These techniques have been invaluable in providing data regarding various facets of adipogenesis and in identifying proteins or pathways that regulate adipogenesis. However, these in vitro techniques show limited snapshots of select mechanisms of adipogenesis and cannot provide an integrative evaluation of adipogenesis within the natural microenvironment of the adipose tissue.

Additional studies designed to overcome these limitations have been performed by investigators using animal models, in which adipocyte size-distribution analyses were taken at the end of high-fat dietary interventions with gradually increasing durations to show the dynamics of adipocyte cellularity with weight gain, using a cross-sectional design [93] or obtained longitudinally by serial biopsies of inguinal WAT depots [98]. Collectively, these studies suggest an oscillatory pattern of adipose tissue remodeling, involving simultaneous and repetitive cycles of hyperplasia, hypertrophy, and hypoplasia (decreased adipocyte number), presumably reflecting proliferation and differentiation of adipocyte precursor cells, development of mature adipocytes, and apoptosis, respectively. Interestingly, the rate of enlargement of adipocytes (hypertrophy) is proportional to the difference between the lipid load and the storage capacity of adipocytes [98], which may also depend on both the number and metabolic properties of the adipocytes. These findings correlate with other studies, which suggest that the adipose morphology of SAT may be related to adipocyte turnover [99], as subjects with hypertrophy generated 70% less adipocytes per year than those with hyperplasia. Frequent cycling, however, may promote replicative senescence of adipocyte progenitor cells and impairment of their adipogenic function [100]. Though these approaches could yield valuable information, they are not applicable to study human subjects due to the ethical consideration of their invasiveness.

Recently, Spalding and colleagues introduced an innovative method to study adipocyte turnover in humans by measuring the incorporation of 14C derived from above ground nuclear bomb tests in adipocyte DNA [92]. Though the lipid turnover rates calculated by this method are reliable, the 14C-labeling method involves the retrospective analysis over a long period of time (years). However, an additional method, which employs labeling of DNA of adipocytes and the SVF with the stable isotope deuterium (2H) over a short period of time (months), offers an advantage to study in vivo adipogenesis [101]. Our laboratory has refined this method in a rat model by purifying the isolated adipocytes and adipocyte progenitor cells (short-time culturing of SVF) [102]. Importantly, this method provides an advantage over the in vitro techniques for assessment of adipogenesis, as it provides an integrative evaluation of adipogenesis within the natural microenvironment of the adipose tissue, which can also be utilized for intervention studies due to the shorter duration of time. Overall, this method has valuable implications to assess cell turnover in adipose tissue [103]; however, the effectiveness of this method to study human adipogenesis and adipocyte cell turnover in vivo remains to be validated.

3C. Depot- Differences in Adipogenesis

While numerous studies have investigated regional differences in adipose tissue metabolism, few have examined depot-specific differences in adipocyte differentiation [104107]. WAT depots possess significant differences in adipogenesis, as the proliferation and differentiation of both human and rodent primary preadipocytes have been shown to be influenced by the anatomic location of the depot, as well as aging, gender, and metabolic health [82, 89, 108118]. While the differences in adipogenesis and its regulation in SAT and VAT depots have been extensively studied [104, 105, 119], studies comparing adipogenesis between upper-body and lower-body subcutaneous depots are sparse [82, 106]. Early studies using thymidine incorporation into fat cell DNA reported increased proliferation in the visceral (parametrial and retroperitoneal depots) preadipocytes of high fat diet-fed rats [120]. Additional studies in rodents indicated that SAT (inguinal) adipocyte progenitors in rodents are more abundant and have significantly increased proliferation as compared to VAT (perigonadal) adipocyte progenitors in response to high-fat diet [81]. Notably, recent studies by Macotela et al. that highlight the intrinsic differences of VAT(epididymal) versus SAT (inguinal) preadipocyte pools in mice reveal that precursor cells from VAT display less differentiation capacity, and VAT has a decreased percentage of preadipocytes following high fat diet, with subsequent increase in other stromovascular cells (i.e. macrophages). They also demonstrate that preadipocytes from VAT highly express anti-adipogenic factors, as opposed to preadipocytes from SAT, which show higher expression of pro-adipogenic genes [121]. This decrease in the preadipocyte pool may be partially attributed to increased cell death of adipocyte precursor cells in VAT, as increased susceptibility to apoptotic stimuli of VAT (vs. abdominal SAT) preadipocytes have been reported in human subjects [122, 123]. Early human studies from the Kirkland laboratory revealed that preadipocytes from abdominal SAT accumulated more lipids and had higher differentiation capacity and levels of adipocyte markers compared to preadipocytes from VAT in obese subjects [119]. Other studies performed in primary cultures showed that the proliferation capacity of preadipocytes from abdominal SAT precursor cells was higher than in VAT (omental) cells in obese individuals [105]. Overall, the reduced proliferation and differentiation capacity of visceral preadipocytes may account for the increased hypertropy of existent adipocytes and the metabolic abnormalities associated with VAT.

Accumulating evidence suggests that preadipocyte number and cellularity may depend on the metabolic state of the individual. Isakson et al. demonstrated impaired differentiation of preadipocytes from abdominal SAT in obese individuals, when compared to lean [87]. More recent reports showed that abdominal SAT has increased proliferation of adipocyte precursors in increasing obese conditions [124]. Collectively, these findings suggest that the preadipocytes in the abdominal SAT depot of obese individuals have the capacity to proliferate, in response to metabolic demand, but lack the ability to differentiate. Conversely, other studies indicate that the preadipocyte numbers were lower in obese women as compared to lean [88]. However, these observations could be attributed to greater recruitment of preadipocytes to adipogenesis or greater preadipocyte apoptosis.

Recent findings demonstrate that humans with morbid obesity, with corresponding excessive WAT development, had decreased adipocyte precursors in abdominal SAT, compared to individuals with moderate obesity [125]. This decrease was accompanied by smaller mean adipocyte diameter and a marked increase in the expression of adipogenic markers, which suggests increased differentiation of new preadipocytes and subsequent depletion of the adipocyte progenitor pool. Indeed, recent compelling data reported decreased replicative potential, premature cellular senescence, and loss of the differentiation potential of VAT preadipocytes from patients with morbid obesity compared to lean individuals [100]. Likewise, other findings have shown that although thiazolidinediones stimulate adipogenesis, its chronic administration decreases the adipogenic potential of adipocyte progenitor cells in WAT [126]. Hence, depot-specific differences in adipocyte progenitor abundance and proliferation may influence whether a fat depot expands by hypertrophy or hyperplasia, and thus could also have important implications on the development of metabolic disease. Collectively, the aforementioned studies demonstrate that, compared to VAT preadipocytes, the SAT preadipocytes have higher proliferation and differentiation, but are less prone to apoptosis. This suggests the existence of an adipocyte precursor pool with a higher efficiency for adipogenesis that may contribute to a higher contribution of hyperplasia in SAT than VAT in conditions of obesity.

Though the aforementioned data collectively indicate that SAT depots contain a greater number of functional adipocyte progenitors as compared to VAT depots, these findings are inconclusive; as additional analyses of the adipose cellularity of lower-body depots are necessary. Limited investigations indicate that preadipocytes from abdominal SAT of obese women differentiate less readily and are more susceptible to apoptosis as compared to the femoral SAT depot [88]. These results support previous reports in primary cultures showing that abdominal SAT preadipocyte differentiation inversely correlates with increased obesity and central adiposity [127]. Thus, the stromavascular fraction (SVF) of abdominal SAT fat tissue from centrally obese individuals might contain more preadipocytes with impaired differentiation potential compared to femoral SAT. This provides evidence that VAT and abdominal SAT may share similar properties, as previously shown [128].

3D. Sex- Differences in Adipogenesis

Limited studies depict sexual dimorphism in adipogenesis. Cross-sectional comparisons of adipocyte morphology and adipogenesis in abdominal and femoral SAT depots in men and premenopausal women with normal weight show larger femoral vs. abdominal SAT adipocyte size in both sexes, but more abundant small adipocytes (aP2+CD68 population in the SVF) in women than in men, especially in the femoral depot [91, 129]. Interestingly, femoral adipocytes differentiate less than abdominal SAT in vitro but are more resistant to tumor necrosis factor alpha (TNFα)-induced apoptosis, suggesting reduced turnover and utilization of the preadipocyte pool in lower- vs. upper-body fat in women [91]. Thus, it appears that a contribution by microenvironmental factors and the existence of a high abundance of small adipocytes, rather than the inherent dynamic properties of preadipocytes, may explain the higher percent of lower-body SAT in women with normal weight.

With accumulation of adipose tissue in obesity, the number of adipocytes increases in women but not in men [129] (Table 1). While adipocyte size increases in both sexes, the rate of the adipocyte hypertrophy is higher in men than in women, particularly in the lower-body fat. Cross-sectional comparisons of adipocyte morphology and adipogenesis in abdominal SAT and VAT depots in pre- and postmenopausal women reveals adipocyte hypertrophy in both depots but hyperplasia only in the abdominal SAT depot [130]) (Table 1). Although such comparisons have not been done solely in men, the findings of reduced adipogenic capability of VAT preadipocytes compared to abdominal SAT preadipocytes in studies of obese men and women combined and the larger visceral fat accumulation in men [91, 131] suggest greater contribution of adipocyte hypertrophy to the VAT expansion in men than in women.

Table 1.

Dynamics of adipocyte characteristics and adipose tissue remodeling with fat gain by depot and sex

Depots Females Males

Adipocyte
size
Adipocyte
number
Frequency of
fat tissue
remodeling*
Adipocyte
size
Adipocyte
number
Frequency of
fat tissue
remodeling*
Omental ↑↑↑↑* * ↑↑↑
SAT Abdominal ↑/↔
Femoral ↑↑ ↑↑↑ ↑↑
*

Hypothetical interpretations; ↑, increased; ↔, not changed

Because sex hormones are known regulators of WAT function, there is increasing evidence for the effects of sex steroids on preadipocyte kinetics. For example, testosterone supplementation inhibits the commitment of pluripotent mesenchymal stem cells to the adipogenic lineage, while promoting myogenesis [132]. Furthermore, testosterone impairs adipogenesis of 3T3-L1 preadipocytes, via interaction of the androgen-receptor complex with β-catenin in the Wnt signaling pathway [133]. Estrogen stimulates preadipocyte proliferation [134], via non-genomic mechanisms involving MAPK-dependent and c-fos signaling pathways [135]. The mitogenic effect of estrogen is demonstrated in both abdominal SAT and VAT preadipocytes; but women are more responsive than men [136]. Estrogen increases adipogenesis via transcriptional up-regulation of PPARγ and IGFR1 genes in ovariectomised animals only [137]. Progestins stimulate preadipocyte differentiation by increasing the gene expression of adipocyte determination and differentiation 1/sterol regulatory element-binding protein 1 (ADD1/SREBP1) transcription factor, which in turn up-regulates the transcription of the lipogenic enzyme fatty acid synthase (FAS) [138]. More importantly, the transcription of cytochrome P450, one of the components of aromatase, is higher in the femoral than in the abdominal region SAT [139, 140]. In addition, an analysis of the global gene expression in the VAT (perigonadal) fat pads in male vs. female mice found that members of the cytochrome 450 superfamily, including aromatase cytochrome P450 (product of the Cyp 19 gene), and hydroxysteroid dehydrogenases are among the sexually dimorphic genes [140].

Women have fluctuations in their levels of sex hormones, which can be modulated by the menopausal status, the use of contraceptives in premenopausal women, or the receipt of HRT in peri- or post-menopausal women. Therefore, these factors must be considered when formulating a study design to effectively analyze WAT, as sex hormones are effectors of adipose metabolism. Nevertheless, the aforementioned studies included women that took contraceptives [91] and postmenopausal women [130], and additional analyses excluding these women did not change the main results or conclusions of the study.

3E. Summary

Overall, in vitro analyses have provided limited, yet very useful information on the processes of adipocyte turnover. Analyzing the various aspects of adipocyte morphology and adipogenesis (Table1) will contribute to a greater understanding of factors that influence depot- and sex-specific WAT expansion. Recent research investigations have integrated the complex dynamics of adipocyte morphology and adipose tissue remodeling in relation to other aspects of WAT function. An overview of the differences in adipose tissue remodeling cycles among depots in men and women that is based on current published data is suggested in Table 1. The general hypothesis is that the high rate of adipocyte hypertrophy and low contribution of hyperplasia in WAT predispose some individuals to increased susceptibility to apoptosis, increased initiation of a local inflammatory response (infiltration of adipose tissue with immune cells and increased secretion of pro-inflammatory molecules by immune cells and adipocyte precursor cells) [141], and gradual impairment of the adipogenic potential of adipocyte precursor cells [100]. In effect, these events shorten the cycles leading to unfavorable increase in their frequency. Taking into account the depot-differences in adipogenesis and adipocyte morphology, it appears that VAT may be more susceptible to this detrimental adipose remodeling than the SAT depots in both sexes. In addition, the more hypertrophic type of WAT expansion in men vs. women and the less available small adipocytes having high lipid storage capacity may cause accelerated adipose tissue remodeling and subsequent adipose dysfunction in men. In contrast, in SAT depots in women, these cycles may be insignificantly affected, and the local inflammation accompanying the adipocyte hypertrophy may be successfully resolved without any unfavorable functional consequences. Because WAT remodeling and metabolism are very complex, future analyses should involve the assessment of adipogenesis in vivo, in order to integrate factors within the natural microenvironment of the adipose tissue that are not present in vitro.

4. Developmental Programming of Regional Adipose Tissue Function

Transcriptional profiling has revealed limited yet valuable information about potential genetic determinants and intrinsic mechanisms that underlie the depot-specific characteristics of adipose tissue in both men and women. Differences in the expression of developmental genes, many of which are highly conserved transcription factors that act during normal development and remain active in adults, have been reported in the adipose tissue of rodents and humans [142147]. One large group of developmental genes contain a particular DNA sequence (homeobox) coding for a distinguishable but variable protein domain of approximately 60 amino acids (homeodomain) [148]. Genes encoding proteins with similar sequences within the homeodomain are categorized into eleven classes [148]. One interesting family of 39 members that contains the homeodomain sequence from Class 1 is the HOX genes. They are organized in four clusters of 9–11 genes localized on chromosomes 7 (HOXA), 17 (HOXB), 12 (HOXC), and 2 (HOXD). Genes in the four clusters with similar sequences and position on the locus can be aligned with each other into 13 paralogous group, which are organized along the chromosome in an order that parallels their expression along the anterior-posterior body axis in chronological order [149]. Specifically, 3’ HOX genes in groups 1– 3 are expressed earliest during development and primarily control the rostral body segments, followed by the expression of the central HOX genes in groups 4–8, which control the thoracic portion of the body, and finally by the 5’ Hox genes in groups 9–13 that control the lumbo-sacral region. This approach, defined as spatial and temporal co-linearity, enables the HOX genes to convey regional body patterning. Other families of homeodomain proteins, found to be expressed in adipose tissue include Irx (iroquois), hedgehog (Hh), engrailed (En) genes, paired related homeobox (Prrx), Meis homeobox 1 (Meis), pre-B-cell leukemia homeobox I (Pbx), PBX/knotted homeobox (Pknox), short stature homeobox (Shox), and others. T-box (TBX) genes are a family of genes encoding transcription factors that are different from the homeodomain proteins, but also regulate a variety of developmental processes and are expressed in adipose tissue.

4A. Depot-dependent differences in the expression of developmental genes

Studies comparing SAT (inguinal in mice and abdominal SAT in humans) and VAT (epididymal in mice and omental in humans) reveal that adipose tissue from VAT express higher levels of HOXA5 and HOXC8, whereas SAT has higher levels of HOXA10, SHOX2, and EN1, in both mice and humans [104, 142144]. Conversely, TBX15 and Sfrp2 were higher in SAT vs. VAT in mice, but higher in VAT vs. SAT in humans [144]. In addition, microarray analyses identify that gluteal SAT, adipocytes and the SVF express higher levels of HOXA2, HOXA3, HOXA4, HOXA5, HOXA9, HOXB7, HOXB8, HOXC8, and IRX2, but lower levels of HOXA10, HOXC13, and PBX15 compared to those from the abdominal SAT site [146, 147]. Notably, depot-specific variations in gene expression are also observed in preadipocytes [104, 144]. In addition, select developmental genes, TBX15 and HOXA5, demonstrate changes in expression that correlate with levels of obesity (body mass index) and fat distribution (waist-to-hip ratio) [144]. More extensive gene expression analyses reveal that homeobox genes and other pregnancy-associated factors, are distinct between fat cell progenitors of both rodent and human adipose tissue depots [104, 150, 151]. The observed differences in gene expression appear to be intrinsic and persist through in vitro culture and differentiation; hence, the microenvironment does not appear to be an influence. Furthermore, the results from the aforementioned experiments by Tchkonia et al. highlighting the differences in lipid accumulation and differentiation capacity of abdominal SAT versus VAT preadipocytes [119] were associated with distinct patterns of gene expression and conserved over multiple cell generations [145]. Collectively, these data suggest that WAT depots originate from different precursor cells, whose function is presumably controlled by genes involved in development and pattern specification.

Questions arise as to the regulatory mechanisms that drive the homeobox gene expression. Recently, several hormones and their cognate receptors that influence adipogenesis have been shown to regulate select homeobox gene expression. For example, retinoic acid has a well-characterized role in the commitment of mouse embryonic stem cells to the adipocyte lineage [152]. Interestingly, PBX1 has been shown to be induced after following treatment with retinoic acid suggesting its involvement in mediating the retinoic acid action [152]. A siRNA-mediated silencing of PBX1 expression in multipotent adipose derived stem cells (hMADs) demonstrates that PBX1 may play a role in human adipogenesis by maintaining the proliferation of adipocyte precursors and preventing their commitment to the adipocyte lineage [152]. Recently, the expression of PBX1 and PBX3 has been shown to be higher in abdominal SAT compared to gluteal depots [146]. Taken together, these data suggest that the depot-specific differences in preadipocyte pools are established during development.

4B. Sex-dependent differences in the expression of developmental genes

Sex differences in the expression of homeobox genes have also been observed in adipose tissue. Gesta et al. reports that there is higher expression of the HOXC9 gene in SAT than in VAT in males, but not in females [144]. Genome-wide association studies (GWAS) and meta-analyses of GWAS have also identified novel sexually-dimorphic genetic loci associated with upper- or lower-body fat distribution [153, 154], including five genes (RSPO3, TBX15, ITPR2, WARS2 and STAB1) that are differentially expressed between abdominal and gluteal SAT [153]. These studies provide evidence for sexual dimorphism in the associations of certain genetic loci as effectors of adipose tissue distribution. Sex differences in epigenetic regulation by environmental and/or hormonal factors have been reported in both rodents and humans and greatly contribute to adipose tissue distribution [155]. Polymorphisms in the estrogen receptor α gene are also associated with body fat distribution in women [156, 157].

A recent transcriptional study of the homeobox genes in abdominal and gluteal SAT depots in men with abdominal obesity (WHR 0.91) and premenopausal women with more peripheral fat distribution (WHR 0.82) show that the expression of HOXC13 and HOXB8 in the gluteal depot is higher in females than in males, whereas the expression of the HOXA2 gene is higher in males compared to females [146]. The expression of the homeobox gene cofactors PBX1, PBX3, and MEIS 1 is higher in abdominal SAT compared to the gluteal depots only in male subjects [146]. Sex hormones, including estrogen, progesterone, and testosterone, have also been described to regulate homeobox gene expression and mediate diverse functions in both developing and adult tissues (reviewed in [158]). These data further support the notion that the apparent differences in adipose tissue function between obese males and females may be mediated by sex hormones via downstream regulation of homeobox gene transcription.

4C. Summary

Because developmental genes heavily influence cell fate and may regulate the development of WAT in utero, their role in nutritional reprogramming of regional adiposity in both sexes requires further investigation. Given the evidence of their differential expression in adult adipose tissue among depots and between sexes, future studies are warranted to explore the precise role in the regulation of regional WAT expansion and fat distribution. Specific directions are well delineated by Karastergiou and colleagues [159] and include: 1) to validate whether the depot- and sex differences in developmental genes are retained after ex vivo culture; 2) to understand the importance of developmental genes in regulating adipogenesis and the functional properties of mature adipocytes; 3) to determine whether other cellular types from the SVF also exhibit depot-dependent differential gene expression and, if so, to understand the subsequent effect on the microenvironment; 4) to investigate whether fat distribution has contributed to the differences in gene expression profiles found between men and women with peripheral fat distribution, by performing additional comparisons between men and women with the same (upper-body) fat distribution phenotype or between women with upper- vs. lower-body fat distribution; and 5) to determine whether depot differences in HOX gene expression contribute to racial differences in fat distribution.

5. Secretory Function

An important property of adipose tissue is the production of secretory factors (adipokines) that mediate local and whole body metabolism via auto-/paracrine and endocrine mechanisms. Adipose tissue expansion, and subsequent remodeling, is often associated with altered adipokine production. Several WAT-derived factors have been investigated and shown to modulate numerous physiological systems. Notably, many adipokines are also up-regulated in obesity and promote inflammatory responses, insulin resistance, and other metabolic complications (reviewed in [160]). Evidence suggests that variances in adipokine secretion may partly contribute to the differential metabolic risks that are associated with fat distribution, notably the protective properties of lower body WAT (reviewed in [24]).

Leptin, an adipocyte-secreted factor, was shown to be predominantly expressed by isolated subcutaneous adipocytes as opposed to omental adipocytes, particularly in women [161, 162]. In vivo human studies demonstrate that secreted levels of leptin are higher in femoral SAT than abdominal SAT [163]. Another metabolically favorable adipokine, adiponectin, was shown to be positively associated with femoral SAT and gluteal fat mass [164, 165] and negatively associated with VAT [166]. Interestingly, levels of both leptin and adiponectin are higher in women [167171].

Other adipokines have been shown to be highly regulated in obesity/T2DM and may contribute to metabolic dysfunction. Retinol-binding protein 4 (RBP4), a protein released from adipocytes and associated with obesity and insulin resistance, is preferentially produced by VAT and is a marker of intra-abdominal adipose tissue expansion [172]. Plasminogen activator inhibitor 1 (PAI1), an inhibitor of fibrinolysis, is strongly up-regulated in VAT depots, and plasma levels correlate with trunk fat mass in obesity [173], suggesting a plausible link between obesity and thrombotic disorders [174].

5A. Novel Adipokines

In recent years, several novel adipokines have been identified and are at the forefront of scientific research to elucidate their roles in human metabolism and disease. Here, we present the current state of the knowledge of selected adipokines (summarized in Table 3).

Table 3.

Relative alterations in adipokine transcription as a function of regional fat gain in men and women

Depots Females Males

Pro-
inflammatory
Anti-
inflammatory
Compensatory
anti-
inflammatory
Pro-
inflammatory
Anti-
inflammatory
Compensatory
anti-
inflammatory
VAT ↑↑ ND ↑↑↑↑ ND
SAT Abdominal ↑/↔ ↑/↔/↓ + +/ND
Femoral* ++ ↑↑ ↓↓ +/ND
*

Hypothetical interpretations; ND, not detected; ↑, increased; ↓, decreased; ↔, not changed

Dipeptidyl peptidase (DPP)-4

Protein expression of dipeptidyl peptidase (DPP)-4, a novel adipokine previously studied for its role in the incretin system, is substantially elevated in VAT compared with SAT of obese individuals [175]. The main cellular source of DPP4 secretion is differentiated adipocytes, with a lesser contribution from SVF cells and macrophages. DPP4 mediates insulin resistance, as this adipokine decreases insulin action in adipocytes via decreased insulin-stimulated Akt phosphorylation. Serum levels of DPP4 are also elevated in obese subjects, compared to subjects with normal weight, and are highly associated with abdominal SAT adipocyte hypertrophy, insulin resistance, and other components of the metabolic syndrome.

Chemerin

Chemerin, also known as retinoic acid receptor responder 2 (RARRES2) or tazarotene-induced gene 2 (TIG2), is another recently discovered adipocytokine that may influence adipose tissue development, inflammation, and glucose homeostasis [176]. It is synthesized as an inactive precursor, prochemerin, which is converted to its active form through C-terminal cleavage by serine proteases, especially during inflammatory conditions. Chemerin signals through the G-protein coupled seven transmembrane receptors ChemR23 (CMKLR1) [177]) and G protein-coupled receptor 1 (GPR1) [178]). Evidence also suggests that chemerin binds to the C-C chemokine receptor-like 2 (CCLR2), which seems to increase its local concentration and facilitate its presentation to ChemR23 on adjacent cells [179, 180]). CMKLR1−/− mice exhibit decreased adipose tissue TNFα and IL-6 mRNA, but are glucose intolerant compared with wild-type mice due decreased glucose-stimulated insulin secretion and glucose uptake in skeletal muscle and WAT [181]. Both adipocytes [182] and fibroblasts [183] produce chemerin. Adipocyte hypertrophy and the inflammatory microenvironment in adipose tissue enhance chemerin synthesis, as judged by its increased secretion in response to free fatty acids, lipopolysaccharide, and interleukin-1beta [184, 185]. Chemerin acts like a chemokine in that it induces leukocyte migration and increases macrophage adhesion to VCAM-1 and fibronectin [186], and thereby potentiating local inflammation. In contrast, it also exerts potent anti-inflammatory effects on activated macrophages by promoting phagocytosis [187]. It is not clear whether these dual actions are elicited by different chemerin variants; hence, these effects require further analysis and validation.

Obese subjects with metabolic syndrome have elevated levels of circulating and gluteal SAT-secreted chemerin [188]. In addition, circulating chemerin levels are positively correlated with VAT accumulation [189] and low circulating estradiol concentrations [190]. Moreover, measurements of the chemerin expression in paired abdominal SAT and VAT samples in patients with severe obesity reveal higher levels in VAT than in abdominal SAT in men, but the opposite is observed in women. Interestingly, women with PCOS show a pattern similar to men, suggesting a role of androgens in the sexual dimorphism in adipose tissue-derived chemerin secretory function [191]. Similar sex- and adipose tissue depot-specific differences in chemerin mRNA expression levels are also reported by Alfadda A et al. [192], with expression significantly higher in women than men and in abdominal SAT than VAT. Interestingly, they show a significant negative correlation between chemerin mRNA expression in abdominal SAT and circulating chemerin levels, which in this study are associated with obesity markers but not with markers of insulin resistance. Apparently, the auto-/paracrine and endocrine mechanisms of chemerin that regulate adipose tissue function and glucose homeostasis is depot-specific. Further studies of the chemerin transduction pathways in different adipose tissue depots are warranted to unravel these distinct mechanisms.

Lipocalin 2

Lipocalin 2 (also known as neutrophil gelatinase-associated lipocalin and 24p3) belongs to the lipocalin protein superfamily (reviewed in [193]). Lipocalins bind and transport lipophilic substances such as retinoids, arachidonic acid and steroids but the high-affinity endogenous ligands of lipocalin 2 are unknown. Circulating lipocalin 2 levels are positively associated with adiposity, but metabolic endotoxemia and consumption of saturated fat may influence their levels [194]. Involvement of this and possibly other factors could explain the variable lipocalin-2 levels in the circulation of women with PCOS, an insulin resistant state, that are found to be reduced [195], normal [196], or increased [197] compared to healthy women. Likewise, lipocalin 2-deficiency in mice is reported to either improve [198] or impair [199] insulin sensitivity compared with control littermates in the context of diet-induced obesity. The expression of lipocalin 2 in adipose tissue increases with obesity by inflammatory stimuli [200204]. There are sex-and depot-dependent differences in the expression of lipocalin 2 that resemble those of chemerin; i.e. men have higher expression of in VAT than in abdominal SAT, whereas women showed the opposite [191]. Further studies are needed to understand how these differences contribute to the distinct regional adipose tissue function and whole body glucose homeostasis.

Glypican-4 (Gpc4)

The cell surface proteoglycan glypican-4 (Gpc4) was shown to be released primarily by adipocytes, following a proteolytic cleavage of the GPI anchor by lipases, such as the insulin-regulated glycosylphosphatidylinositol-specific phospholipase D (GPLD1). Glypican-4 expression is higher in abdominal SAT versus VAT depots in humans with normal weight [205]. Conversely, with increasing body mass index (BMI) and waste-to-hip ratio (WHR), Gpc4 mRNA levels in abdominal SAT decrease while those in VAT increase, reaching the highest abundance in overweight and obese individuals [206]. In women, serum Gpc4 levels continuously increase from the lean to the obese phenotype. While in men, serum Gpc4 increases from the lean to overweight phenotype, but in obese men, Gpc4 levels decrease to those observed in lean men, possibly due greater reduction of GPLD1 activity as a result of enhanced insulin resistance [206]. Overall, circulating Gpc4 levels positively correlate with increasing BMI and insulin resistance, and additional experiments depict glypican-4 as a novel adipogenesis-promoting and insulin-sensitizing adipose-derived factor [206].

Omentin

Omentin is another new adipocytokine that is secreted mainly by stromavascular cells [207]. Circulating omentin levels are reduced in subjects with obesity and T2DM compared with lean subjects [208210] and lower omentin mRNA levels in gluteal adipose depot have been reported in obese subjects with metabolic syndrome [188]. Omentin gene expression is higher in VAT than in abdominal SAT [191, 211] in both men and women, despite the putative androgen response elements in its promoter region suggesting that other factors may be involved in the regulation [191]. A recent study finds sex as one of the determinants of circulating omentin concentrations, and low testosterone concentrations are related to higher omentin levels [190]. Omentin increases glucose uptake by human adipocytes in vitro through enhanced protein kinase B (AKT) phosphorylation and insulin signal transduction [210]. It also exerts antiinflammatory effects by inhibiting TNFα-induced expression of adhesion molecules in endothelial and vascular smooth muscle cells [212, 213]. These data suggest that omentin may play a positive role to reduce inflammation and increase insulin sensitivity, but its clinical relevance needs to be confirmed in future studies.

Secreted frizzled-related proteins (SFRPs)

A family of five secreted frizzled-related proteins, termed SFRP1–5, has been implicated in the regulation of adipogenesis. They share significant homology with the Frizzled receptors and bind to wingless-type (Wnt) ligands, interfering with the Wnt/β-catenin signaling [214], which exerts adipogenesis-inhibitory action by reducing the expression of pro-adipogenic transcription factors [215]. SFRP1 and SFRP 4 mRNA is up-regulated during adipogenic differentiation, suggesting a proadipogenic role [216, 217]. A comprehensive assessment of all five SFRP proteins in abdominal SAT and VAT depots from lean and obese people in a study by Ehrlund A et al. [218] shows that distinctive members of the SFRP family exhibit different depot-dependent expression patterns as a function of obesity. Specifically, SFRP1 decreases in abdominal SAT but is unchanged in VAT, whereas SFRP2–4 is up-regulated, more noticeably in the VAT depot, and SFRP5 does not change in either depot. SFRP1 in abdominal SAT is negatively associated with BMI and insulin resistance via reducing the secretion of the proinflammatory cytokines and increasing the release of adiponectin. SFRP2–4 mRNA levels in both abdominal depots show opposite relationships, but the mechanisms are unknown. Notably, SFRP1, SFRP2 and SFRP4 are secreted by abdominal SAT explants but not SFRP5, raising speculation about its contribution to the SFRP5 in circulation. Furthermore, the reports on the role of circulating SFRP5 in obesity-related insulin resistance are inconsistent. Some observations in animals and humans show that the levels decline with obesity [219, 220], while others report opposite findings [221, 222]. It is of interest to note that one study finds higher plasma SFRP5 levels in females compared to males, suggesting possible sex-specific regulation. Additional studies are needed to elucidate the role of SFRP2–5 in the adipose tissue function, obesity, and metabolic health.

Vaspin

Vaspin (visceral adipose tissue–derived serine protease inhibitor, serpinA12), is another adipokine associated with insulin-sensitizing effects. In human studies, the vaspin mRNA levels are not detectable in persons with normal weight and in some obese subjects with insulin resistance. However, vaspin expression is induced in VAT and/or SAT of overweight and obese individuals [223]. Administration of vaspin to mice with dietary-induced obesity ameliorates insulin resistance, in part, through normalizing the altered expression of genes relevant to insulin resistance, including leptin, resistin, TNFα, glucose transporter 4, and adiponectin [224]. Some studies suggest that the induction of vaspin gene expression in human adipose tissue may be a compensatory mechanism associated with obesity and insulin resistance. The vaspin mRNA levels vary between VAT and SAT depots, but its regulation appears to be depot specific. Its expression in VAT is strongly predicted by percent body fat whereas insulin sensitivity predicts the SAT vaspin mRNA levels. Interestingly, although no sex-differences are observed for the vaspin expression in adipose tissue, sexual dimorphism has been reported for circulating levels, which were shown to be higher in women [225, 226].

5B. Summary

Overall, there is limited knowledge about the roles of functionally divergent WAT depots in human adipokine production. Nevertheless, many adipokines have been shown to be differentially expressed and secreted, as a function of adipocyte morphology, between WAT depots, specifically abdominal SAT and VAT. Likewise, the relationship between circulating levels of adipokines, their secretion from WAT depots, as well as their endocrine effects, have been heavily evaluated to understand their contribution to the development of obesity and related metabolic disorders. It is important to discern whether these adipokines appear as a compensatory reaction to increased metabolic distress, i.e. adipocyte hypertrophy, in an effort to undergo normal adipose tissue remodeling, or if they have a pathogenic role leading to WAT metabolic dysfunction. Accordingly, select adipokines may exert dual pro- or anti-inflammatory or insulin resistant or insulin sensitive effects, relative to the adipocyte characteristics and WAT remodeling paradigms. A speculative summary of the depot- and sex-dependent differences in novel adipokine production is presented in Table 2. Overall, cytokines that decrease adipocyte insulin resistance and show pro-inflammatory actions, including DPP4, chemerin, and lipocalin 2, appear to be produced more in VAT than SAT depots and more in men than in women. This may result from their secretion from hypertrophic adipocytes, macrophages, and/or adipocyte precursor cells in a more inflammatory state, reflecting a WAT remodeling pattern more frequent in VAT depots and in men (Table 1). While adipokines with insulin sensitizing and/or anti-inflammatory properties, such as Gpc4, omentin, and certain SFRPs, seem to increase mostly in the VAT (more in women than men), in an attempt to ameliorate or resolve the local inflammation, and increase much less or decrease in abdominal SAT (less in women than men). Of note, vaspin, is not detected in many subjects but may also be expressed as part of a compensatory mechanism in response to the development of insulin resistance. Importantly, although the expression of adipokines in lower-body SAT has not been analyzed, we speculate that sex differences in the production will be highly significant, considering the differences in adipocyte cellularity between the two SAT depots between the sexes (Table 1). Further studies of the adipokine expression in upper- and lower-body depots using longitudinal designs will be necessary to test these hypotheses. Likewise, the characterization of sex- and depot-differences in the expression and secretion of the numerous novel adipokines (reviewed in [193, 227]) will provide further insight into the paracrine and endocrine function of adipose tissue depots and the relationship to energy homeostasis and metabolic health.

Table 2.

Notes on depot- and sex differences in the expression of novel adipokines

Molecule Cellular
source(s)
in adipose
tissue
Auto/paracrine
function
Depot-
specific
effects on
mRNA
levels
Sex-specific
effects on
circulating or AT
mRNA levels
Effects of
obesity
DPP4 Adipocytes-
COR(+) size;
SVF; Mφ
↓Insulin signaling
in adipocytes
VAT>SAT
in obese,
↑Serum DPP4
↑DPP4 mRNA
in VAT, SAT
Chemerin SVF
Adipocytes
Pro-/anti-
inflammatory
SAT
mRNA
COR(−)
serum
chemerin
M & PCOS-F:
VAT>SAT F:
SAT>VAT
Serum chemerin
COR(−) serum
estradiol
↑secretion by
adipocyte
hypertrophy &
AT inflammation
Lipocalin
2
Transports
lipophilic
substances
(retinoids,
steroids,
arachidonic acid)
M & PCOS-F: VAT>SAT
F: SAT>VAT
SAT mRNA
COR(−) serum
↑secretion by
AT inflammation
Serum levels &
relation to
insulin
sensitivity-
varying data
Gpc4 Adipocytes
Less by
SVF
↑Insulin signaling
↑Adipocyte
differentiation
SAT>VAT
in lean
Serum Gpc4:
F: ↑ continuously
with ↑BMI
M: ↑ from normal
weight, to overweight
↓ in obese
↑VAT,
COR(+) BMI, WHR
↓SAT, COR(−) BMI, WHR
Serum Gpc4:
COR(+) BMI, IR
Omentin SVF ↑glucose
uptake
by adipocytes
Anti-inflammatory
VAT>SAT AT mRNA: F=M
Serum omentin
COR(−) serum
testorsterone
↓Circulating &
gluteal mRNA
Serum omentin:
COR(−) MetS
SFRPs SAT explant
secretes
SFRP1,−2,−4
↑ adipogenesis
SFRP1: antiinflammatory, ↑adiponectin
SFRP2–4: inverse
to SFRP1
Circulating
SFRP5 levels:
F>M
SFRP1 mRNA:
↓SAT, ↔VAT
SFRP2–4
mRNA: ↑SAT,
↑↑ in VAT
Serum SFRP5:
varying data
Vaspin Adipocytes
Precursors
SVF
↑Glucose uptake
by adipocytes
Anti-inflammatory
↑Adiponectin
VAT:
COR(+) BF
SAT:
COR(+) IR
& WHR
AT mRNA:
F=M, Serum vaspin:
F>M
No mRNA in AT
in lean;
↑compensatory
with ↑BF, IR

Abbreviations: DPP4, dipeptidyl peptidase 4; SV, stroma-vascular; Mφ, macrophages; VAT, visceral adipose tissue; SAT, subcutaneous adipose tissue; COR(+)/(−) , positive or negative correlation; PCOS-F, women with polycystic ovarian syndrome; SVF, stromal-vascular fraction; Gpc4, Glypican 4; BMI, body mass index: body weight (kg) : height (cm)2; WHR, waist to hip (circumferences) ratio; F, female(s); M, male(s); TNFα, tumor necrosis factor alpha; SFRPs, a family of five secreted frizzled-related proteins; Wnt, wingless-type ligands; AT, adipose tissue; ↑, increased; ↓, decreased.

6. Lipid Metabolism

A critical function of adipose tissue is the storage and release of triglycerides (TGs) to provide energy in the form of fatty acids, to be utilized during exercise, fasting, or starvation. Important factors in adipose tissue fatty acid metabolism are the rate of TG storage, primarily via lipoprotein lipase activity, and the rate of lipolysis, which is potently regulated by insulin (fed state) and catecholamines (fasted state) and results in the liberation of free fatty acids (FFAs) as fuel to tissues and organs. Likewise, evidence suggests that upper-body (subcutaneous and visceral) and lower-body fat depots show distinct properties in the rates and amounts of lipolysis and fatty acid uptake, and variations in these factors may be responsible for the depot-specific characteristics of adipose tissue.

6A. Lipolysis and FFA Release from WAT

Numerous findings suggest that there is great heterogeneity in lipolysis between adipose tissue depots of men and women. On average, adipose tissue lipolysis is substantially greater (~40%) in women than in men, even though metabolic health is typically better in women. This is presumably due, in part, to increased fat oxidation and more efficient utilization and disposal of FFA in women [228, 229] (reviewed in [230]). Studies have shown that upper-body adipocytes isolated from lean males and females are highly responsive to lipolytic adrenergic stimulation, as compared to lower body adipocytes [231235]. Importantly, these effects were more pronounced in females. Subsequent in vivo studies in lean individuals support these findings, in that catecholamine-induced lipolysis is greater in upper-body than in lower-body SAT depots [236238]. In addition, both exercise-induced and prolonged fasting-induced lipolytic activity was significantly more marked in abdominal than in gluteal-femoral SAT [239, 240]. Furthermore, in obese women, femoral SAT is less lipolytically active than upper-body fat [14, 241]. A recent study by Gavin et al. demonstrates that premenopausal overweight women perfused with estrogen had an increased lipolytic response in abdominal SAT, while estrogen inhibited lipolysis in gluteal SAT [242]. This study, among others, highlights the role of sex hormones in body fat distribution [243, 244], and suggests that estrogen may be involved in the maintenance of the gynoid body fat distribution in premenopausal women. Additional studies demonstrate that hormone sensitive lipase (HSL), a key enzyme in lipolysis, is differentially expressed between adipose tissue depots (reviewed in [245]), and the rate of action of HSL is higher in the abdominal than the gluteal depot [246]. Importantly, upper-body/visceral adipose tissue lipolysis from both males and upper-body obese females were also shown to be more resistant to the anti-lipolytic effects of insulin when compared to non-obese or lower body obese adults [247250]. Furthermore, FFA concentrations are much greater in upper-body obese individuals following a meal [14, 251], supporting the notion that upper-body adipocytes are more resistant to the anti-lipolytic effects of insulin. In contrast, lower body adipocytes are more responsive to the anti-lipolytic effects of alpha 2 adrenergic agonists and have lower responses to lipolytic agonists (reviewed in [252]). Collectively, these studies suggest that body fat distribution greatly influences lipolysis, as summarized in Table 4. While the regulation of lipolytic processes in various WAT depots require further investigation, studies suggest that the differential levels of lipolysis, as shown in the above analyses, may be mediated, in part, through a higher expression of lipolytic beta-adrenergic receptors in the upper-body depot [253, 254].

Table 4.

Schematic of WAT depot- and sex differences in lipid metabolism

Depots Females Males

WAT depot
mass
Lipolysis/FFA
release
FFA uptake WAT depot
mass
Lipolysis/FFA
release
FFA uptake
VAT ↑↑
SAT Abdominal ↑↑
Gluteal ↓↓ ↑↑
Femoral ↓↓ ↑↑

Sex dimorphism is also evident in these analyses, as abdominal SAT in women greatly exceeds the lipolytic effects seen in men; while femoral SAT had an increased lipolytic response in men but not women [238, 239]. Lipolysis was also much greater in women compared to men following prolonged fasting [255]. Conversely, the stimulation of lipolysis in VAT is much higher in men than women [238, 254].

FFAs that are released from stored TGs can be utilized as required between meals and in the starved state; however, excess FFAs can contribute to the adverse metabolic consequences of obesity by ectopic fat accumulation in hepatic and peripheral tissues. Compared with lean women, systemic FFA release in the fasting state is greater in upper-body obese women, due to their higher fat mass [256]. Likewise, lipolysis-induced FFA release is greatly reduced in lower-body obese women, such that the levels are comparable to non-obese women. Studies have demonstrated that lipolysis of VAT increases, relative to visceral fat mass, which results in the release of excess FFAs into the liver via the portal vein in both obese men and women, but more so in obese women [257]. It is well-established that high levels of FFA can mediate insulin resistance in both muscle [258, 259] and liver [260, 261]. Nevertheless, though VAT-derived FFAs play an important role in abnormalities of hepatic function in obese individuals [262264], VAT may not be a significant contributor of FFAs to peripheral tissues. Hence, upper-body obesity is highly associated with increased FFA release, and abdominal SAT, as opposed to VAT, may be the primary source of excess systemic FFAs in both men and women [14, 241, 248].

6C. Storage and Uptake of FFA in WAT

The storage of intracellular TGs in adipose tissue derived from FFAs depends on the action of lipoprotein lipase (LPL) by adipocytes, as this is the rate-determining step in the uptake of circulating TG-FFA. Several studies have assessed the depot- and sex-specific regulation of LPL. In early studies, LPL expression and activity was shown to be higher in subcutaneous abdominal than in gluteal adipocytes in non-obese men [265]. Conversely, the activity of LPL was higher in gluteal- femoral adipocytes from non-obese women as compared with the abdominal depot [265, 266]. Additional studies demonstrate that LPL activity is lower in VAT versus abdominal SAT-derived adipocytes isolated from women, but higher in the VAT depot in obese men [235, 267]. Likewise, in non-obese or moderately obese men, LPL expression and activity is higher in visceral than in gluteal adipocytes [235]. Additional evidence demonstrates that testosterone was able to suppress LPL activity in the femoral SAT of men, contributing to abdominal fat accumulation [268], while other findings suggest that testosterone does not affect LPL activity in the thigh [269]. Other studies have also demonstrated the role of sex hormones in modulating LPL activity and expression [270, 271]. Taken together, these variances may be due to both sex-specific differences and the complex post-transcriptional regulation of LPL [272]. Nevertheless, it is likely that LPL expression and activity play a critical role in sex-specific adipose tissue depot development and function.

Depot- and sex-specific differences in the direct uptake of circulating FFA have also been described. Overall, short-term meal-derived FFA uptake was shown to be greater in upper-body abdominal SAT versus lower-body SAT in both lean and obese men and women, with women storing more in SAT than men [269, 273, 274]. One point of consideration is that the in vivo rate of FFA uptake in these studies was measured per mass of the entire adipose tissue. Therefore, uptake may be calculated as greater in SAT due to its larger fat mass and actually similar or lower in visceral adipocytes when calculated per cell. Nevertheless, storage of meal-derived fatty acids per gram of adipose tissue lipid following a high fat diet is greatly increased preferentially in the gluteal-femoral SAT of women compared to men [275]. In addition, studies have shown that femoral SAT of women with lower-body obesity more efficiently takes up meal-derived FFAs from circulation [276]. These regional differences were not observed in men; therefore, the aforementioned studies may partially explain sex-dependent body fat distribution, as women typically have more lower-body fat [266]. Conversely, evidence suggests that the uptake of meal-derived FFA in the VAT depot is greater in lean men than women [273, 277]. There are also differences between the upper-body depots, as FFA uptake following a meal was much greater in the VAT than abdominal SAT depot, which was in turn greater than the femoral SAT depot in obese men and women [278]. Furthermore, in men, testosterone induces meal FFA uptake preferentially into abdominal SAT and decreases uptake in VAT [269]. Hence, older men with decreased testosterone develop more VAT.

In the postabsorptive state, lean men are much less efficient at the direct uptake of circulating FFA into the femoral as compared to the abdominal SAT depot; while in obese subjects, direct FFA uptake is increased specifically in the femoral depot of women [279]. Additional studies support these observations, as Koutsari et al. show that women had greater FFA storage than men in both abdominal and femoral SAT in the postabsorptive state [280]. Notably, storage rates were significantly greater in femoral SAT than abdominal SAT in women, whereas the opposite was true in men. The femoral SAT depot of women was also shown to more effectively store FFA during walking conditions, as compared to the abdominal SAT depot and each SAT depot in men [281]. Collectively, these findings suggest that direct adipose tissue FFA storage is significantly greater in women than men.

Overall, the upper-body abdominal fat depot may participate in the short-term daily FFA handling and uptake the flux of dietary fatty acids (reviewed in [282]), while the lower-body gluteal-femoral depot appears to exert its protective properties in long-term fatty acid storage, especially in women (Table 4). Moreover, the storage capacity of gluteal-femoral WAT may influence the level of abdominal/central adiposity, as supported by a recent study by Hernandez et al. depicting the pattern of fat redistribution fat following liposuction [283]. Though some studies suggest that the depot-differences in the above findings may be attributed to LPL activity [277], additional studies are necessary to evaluate the mechanisms underlying FFA storage in upper- and lower-body depots of men and women. Short- and long-term fatty acid metabolism in both upper- and lower-body depots has not been fully elucidated and requires further analysis.

7. Conclusions

Studies support the hypothesis that regional WAT development and expansion greatly impact metabolic health, as fat in different body locations exhibits distinct features and functional characteristics. Additional evidence also highlights sexual dimorphism in adipose tissue function, and there evidence for a plausible role of sex hormones, especially estrogen, in regional WAT function. Yet, the mechanisms that control the regulation of fat distribution in both males and females are poorly understood. Indeed, the influences of adipose tissue depot- and sex- specific effects on adipocyte function, as well as the relationship to metabolic health are not fully elucidated and warrant further analysis. Numerous studies have described depot-and sex-differences in adipose tissue metabolism, such as adipogenesis, the expression of developmental gene signatures, fatty acid handling, and secretory function. Though many of these analyses have focused on the differences between upper-body SAT and VAT, recent investigations have focused on elucidating the differences between upper-and lower-body WAT metabolism, especially the beneficial effects of gluteal-femoral adiposity.

Investigations have provided limited information on the adipocyte proliferation, differentiation, and remodeling processes that are involved in WAT expansion (Table 1). Much remains to be learned about the factors from the microenvironment that influence adipogenesis in adipose tissue depots in both sexes, and how they subsequently contribute to regional fat distribution and metabolic health.

Various studies have yielded the discovery of novel gene loci that are involved in the regulation of body fat distribution in both males and females. Likewise, analyzing the expression patterns of these sex- and depot- specific genes may also provide clues as to the mechanisms underlying regional fat expansion.

Evidence arising from in vitro and in vivo studies suggests that the differential regulation of fatty acid release and uptake in SAT and VAT greatly affect their depot-specific metabolic properties (Table 4). Overall, VAT adipocytes in men exhibit higher rates of fatty acid turnover and lipolysis that in women, leading to a greater release of free fatty acids into the circulation. Such differences provide a link between increased VAT and the metabolic risks associated with obesity. Importantly, the long-term storage of fatty acids in the lower-body (gluteal-femoral) depot, preferentially in women, provides a plausible explanation as to the protective properties of this depot from ectopic fat accumulation and metabolic perturbations. Nevertheless, the exact regulatory mechanisms of fatty acid metabolism remain to be elucidated.

With the accumulating evidence for the beneficial role of the femoral WAT depot, it is important to characterize adipokine expression and secretion in both upper- and lower-body WAT depots; as it is possible that the gluteal-femoral depot could convey protection through a beneficial adipokine profile. These analyses will facilitate categorizing the importance and beneficence of the known adipokines, but, importantly, include the continued discovery of novel adipokines. In addition, accumulating data suggests that adipokines have paracrine effects on preadipocyte cellular kinetics, as well as mature adipocyte metabolic function or secretion, which demonstrates the multiple effects of adipokines on metabolic health (Table 2).

Highlights.

  • Regional fat distribution and sex dimorphism greatly influence metabolic health.

  • Developmental genes may contribute to depot- and sex-specific properties of adipose tissue.

  • Depot and sex influence adipogenesis, secretory function, and fatty acid handling.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Cinti S. The adipose organ at a glance. Dis Model Mech. 2012;5:588–594. doi: 10.1242/dmm.009662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Vague J. La Presse medicale. 1947;55:339. [Not Available] [PubMed] [Google Scholar]
  • 3.Chan JM, Rimm EB, Colditz GA, Stampfer MJ, Willett WC. Obesity, fat distribution, and weight gain as risk factors for clinical diabetes in men. Diabetes care. 1994;17:961–969. doi: 10.2337/diacare.17.9.961. [DOI] [PubMed] [Google Scholar]
  • 4.Carey VJ, Walters EE, Colditz GA, Solomon CG, Willett WC, Rosner BA, Speizer FE, Manson JE. Body fat distribution and risk of non-insulin-dependent diabetes mellitus in women. The Nurses’ Health Study. American journal of epidemiology. 1997;145:614–619. doi: 10.1093/oxfordjournals.aje.a009158. [DOI] [PubMed] [Google Scholar]
  • 5.Okosun IS, Liao Y, Rotimi CN, Prewitt TE, Cooper RS. Abdominal adiposity and clustering of multiple metabolic syndrome in White, Black and Hispanic americans. Annals of epidemiology. 2000;10:263–270. doi: 10.1016/s1047-2797(00)00045-4. [DOI] [PubMed] [Google Scholar]
  • 6.Seidell JC, Perusse L, Despres JP, Bouchard C. Waist and hip circumferences have independent and opposite effects on cardiovascular disease risk factors: the Quebec Family Study. The American journal of clinical nutrition. 2001;74:315–321. doi: 10.1093/ajcn/74.3.315. [DOI] [PubMed] [Google Scholar]
  • 7.Canoy D, Boekholdt SM, Wareham N, Luben R, Welch A, Bingham S, Buchan I, Day N, Khaw KT. Body fat distribution and risk of coronary heart disease in men and women in the European Prospective Investigation Into Cancer and Nutrition in Norfolk cohort: a population-based prospective study. Circulation. 2007;116:2933–2943. doi: 10.1161/CIRCULATIONAHA.106.673756. [DOI] [PubMed] [Google Scholar]
  • 8.Pischon T, Boeing H, Hoffmann K, Bergmann M, Schulze MB, Overvad K, van der Schouw YT, Spencer E, Moons KG, Tjonneland A, Halkjaer J, Jensen MK, Stegger J, Clavel-Chapelon F, Boutron-Ruault MC, Chajes V, Linseisen J, Kaaks R, Trichopoulou A, Trichopoulos D, Bamia C, Sieri S, Palli D, Tumino R, Vineis P, Panico S, Peeters PH, May AM, Bueno-de-Mesquita HB, van Duijnhoven FJ, Hallmans G, Weinehall L, Manjer J, Hedblad B, Lund E, Agudo A, Arriola L, Barricarte A, Navarro C, Martinez C, Quiros JR, Key T, Bingham S, Khaw KT, Boffetta P, Jenab M, Ferrari P, Riboli E. General and abdominal adiposity and risk of death in Europe. The New England journal of medicine. 2008;359:2105–2120. doi: 10.1056/NEJMoa0801891. [DOI] [PubMed] [Google Scholar]
  • 9.Kissebah AH, Vydelingum N, Murray R, Evans DJ, Hartz AJ, Kalkhoff RK, Adams PW. Relation of body fat distribution to metabolic complications of obesity. J Clin Endocrinol Metab. 1982;54:254–260. doi: 10.1210/jcem-54-2-254. [DOI] [PubMed] [Google Scholar]
  • 10.McLaughlin T, Lamendola C, Liu A, Abbasi F. Preferential fat deposition in subcutaneous versus visceral depots is associated with insulin sensitivity. J Clin Endocrinol Metab. 2011;96:E1756–E1760. doi: 10.1210/jc.2011-0615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kovacova Z, Tencerova M, Roussel B, Wedellova Z, Rossmeislova L, Langin D, Polak J, Stich V. The impact of obesity on secretion of adiponectin multimeric isoforms differs in visceral and subcutaneous adipose tissue. Int J Obes (Lond) 2011 doi: 10.1038/ijo.2011.223. [DOI] [PubMed] [Google Scholar]
  • 12.Amati F, Pennant M, Azuma K, Dube JJ, Toledo FG, Rossi AP, Kelley DE, Goodpaster BH. Lower thigh subcutaneous and higher visceral abdominal adipose tissue content both contribute to insulin resistance. Obesity. 2012;20:1115–1117. doi: 10.1038/oby.2011.401. [DOI] [PubMed] [Google Scholar]
  • 13.Michaud A, Drolet R, Noel S, Paris G, Tchernof A. Visceral fat accumulation is an indicator of adipose tissue macrophage infiltration in women. Metabolism. 2011 doi: 10.1016/j.metabol.2011.10.004. [DOI] [PubMed] [Google Scholar]
  • 14.Guo Z, Hensrud DD, Johnson CM, Jensen MD. Regional postprandial fatty acid metabolism in different obesity phenotypes. Diabetes. 1999;48:1586–1592. doi: 10.2337/diabetes.48.8.1586. [DOI] [PubMed] [Google Scholar]
  • 15.Tordjman J, Divoux A, Prifti E, Poitou C, Pelloux V, Hugol D, Basdevant A, Bouillot JL, Chevallier JM, Bedossa P, Guerre-Millo M, Clement K. Structural and inflammatory heterogeneity in subcutaneous adipose tissue: relation with liver histopathology in morbid obesity. Hepatol. 2012 doi: 10.1016/j.jhep.2011.12.015. [DOI] [PubMed] [Google Scholar]
  • 16.Amati F, Pennant M, Azuma K, Dube JJ, Toledo FG, Rossi AP, Kelley DE, Goodpaster BH. Lower Thigh Subcutaneous and Higher Visceral Abdominal Adipose Tissue Content Both Contribute to Insulin Resistance. Obesity (Silver Spring) 2012 doi: 10.1038/oby.2011.401. [DOI] [PubMed] [Google Scholar]
  • 17.Michaud A, Drolet R, Noel S, Paris G, Tchernof A. Visceral fat accumulation is an indicator of adipose tissue macrophage infiltration in women. Metabolism: clinical and experimental. 2012;61:689–698. doi: 10.1016/j.metabol.2011.10.004. [DOI] [PubMed] [Google Scholar]
  • 18.Smith SR, Lovejoy JC, Greenway F, Ryan D, deJonge L, de la Bretonne J, Volafova J, Bray GA. Contributions of total body fat, abdominal subcutaneous adipose tissue compartments, and visceral adipose tissue to the metabolic complications of obesity. Metabolism: clinical and experimental. 2001;50:425–435. doi: 10.1053/meta.2001.21693. [DOI] [PubMed] [Google Scholar]
  • 19.Snijder MB, Dekker JM, Visser M, Yudkin JS, Stehouwer CD, Bouter LM, Heine RJ, Nijpels G, Seidell JC. Larger thigh and hip circumferences are associated with better glucose tolerance: the Hoorn study. Obesity research. 2003;11:104–111. doi: 10.1038/oby.2003.18. [DOI] [PubMed] [Google Scholar]
  • 20.Snijder MB, Dekker JM, Visser M, Bouter LM, Stehouwer CD, Yudkin JS, Heine RJ, Nijpels G, Seidell JC. Trunk fat and leg fat have independent and opposite associations with fasting and postload glucose levels: the Hoorn study. Diabetes Care. 2004;27:372–377. doi: 10.2337/diacare.27.2.372. [DOI] [PubMed] [Google Scholar]
  • 21.Snijder MB, Dekker JM, Visser M, Bouter LM, Stehouwer CD, Kostense PJ, Yudkin JS, Heine RJ, Nijpels G, Seidell JC. Associations of hip and thigh circumferences independent of waist circumference with the incidence of type 2 diabetes: the Hoorn Study. The American journal of clinical nutrition. 2003;77:1192–1197. doi: 10.1093/ajcn/77.5.1192. [DOI] [PubMed] [Google Scholar]
  • 22.Snijder MB, Visser M, Dekker JM, Goodpaster BH, Harris TB, Kritchevsky SB, De Rekeneire N, Kanaya AM, Newman AB, Tylavsky FA, Seidell JC. Seidell, Low subcutaneous thigh fat is a risk factor for unfavourable glucose and lipid levels, independently of high abdominal fat. The Health ABC Study, The Health ABC Study. 2005;48:301–308. doi: 10.1007/s00125-004-1637-7. [DOI] [PubMed] [Google Scholar]
  • 23.Pinnick KE, Neville MJ, Fielding BA, Frayn KN, Karpe F, Hodson L. Gluteofemoral adipose tissue plays a major role in production of the lipokine palmitoleate in humans. Diabetes. 2012;61:1399–1403. doi: 10.2337/db11-1810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Manolopoulos KN, Karpe F, Frayn KN. Gluteofemoral body fat as a determinant of metabolic health. Int J Obes (Lond) 2010;34:949–959. doi: 10.1038/ijo.2009.286. [DOI] [PubMed] [Google Scholar]
  • 25.Lefterova MI, Lazar MA. New developments in adipogenesis. Trends Endocrinol Metab. 2009;20:107–114. doi: 10.1016/j.tem.2008.11.005. [DOI] [PubMed] [Google Scholar]
  • 26.Seale P, Kajimura S, Spiegelman BM. Transcriptional control of brown adipocyte development and physiological function--of mice and men. Genes & development. 2009;23:788–797. doi: 10.1101/gad.1779209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ravussin E, Galgani JE. The implication of brown adipose tissue for humans. Annual review of nutrition. 2011;31:33–47. doi: 10.1146/annurev-nutr-072610-145209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Gallagher D, Visser M, Sepulveda D, Pierson RN, Harris T, Heymsfield SB. How useful is body mass index for comparison of body fatness across age, sex, and ethnic groups? American journal of epidemiology. 1996;143:228–239. doi: 10.1093/oxfordjournals.aje.a008733. [DOI] [PubMed] [Google Scholar]
  • 29.Jackson AS, Stanforth PR, Gagnon J, Rankinen T, Leon AS, Rao DC, Skinner JS, Bouchard C, Wilmore JH. The effect of sex, age and race on estimating percentage body fat from body mass index: The Heritage Family Study. International journal of obesity and related metabolic disorders : journal of the International Association for the Study of Obesity. 2002;26:789–796. doi: 10.1038/sj.ijo.0802006. [DOI] [PubMed] [Google Scholar]
  • 30.Demerath EW, Sun SS, Rogers N, Lee M, Reed D, Choh AC, Couch W, Czerwinski SA, Chumlea WC, Siervogel RM, Towne B. Anatomical patterning of visceral adipose tissue: race sex, and age variation. Obesity (Silver Spring) 2007;15:2984–2993. doi: 10.1038/oby.2007.356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Karastergiou K, Fried SK. Sex differences in human adipose tissues - the biology of pear shape. Biol Sex Differ. 2012;3:13. doi: 10.1186/2042-6410-3-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Geer EB, Shen W. Gender differences in insulin resistance, body composition, and energy balance. Gend Med. 2009;6(Suppl 1):60–75. doi: 10.1016/j.genm.2009.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Schreiner PJ, Terry JG, Evans GW, Hinson WH, Crouse JR, 3rd, Heiss G. Sex-specific associations of magnetic resonance imaging-derived intra-abdominal and subcutaneous fat areas with conventional anthropometric indices. The Atherosclerosis Risk in Communities Study. Am J Epidemiol. 1996;144:335–345. doi: 10.1093/oxfordjournals.aje.a008934. [DOI] [PubMed] [Google Scholar]
  • 34.Despres JP, Couillard C, Gagnon J, Bergeron J, Leon AS, Rao DC, Skinner JS, Wilmore JH, Bouchard C. Race, visceral adipose tissue, plasma lipids, and lipoprotein lipase activity in men and women: the Health, Risk Factors, Exercise Training, and Genetics (HERITAGE) family study. Arteriosclerosis, thrombosis, and vascular biology. 2000;20:1932–1938. doi: 10.1161/01.atv.20.8.1932. [DOI] [PubMed] [Google Scholar]
  • 35.Camhi SM, Bray GA, Bouchard C, Greenway FL, Johnson WD, Newton RL, Ravussin E, Ryan DH, Smith SR, Katzmarzyk PT. The relationship of waist circumference and BMI to visceral, subcutaneous, and total body fat: sex and race differences. Obesity (Silver Spring) 2011;19:402–408. doi: 10.1038/oby.2010.248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Goodpaster BH, Krishnaswami S, Harris TB, Katsiaras A, Kritchevsky SB, Simonsick EM, Nevitt M, Holvoet P, Newman AB. Obesity, regional body fat distribution, and the metabolic syndrome in older men and women. Archives of internal medicine. 2005;165:777–783. doi: 10.1001/archinte.165.7.777. [DOI] [PubMed] [Google Scholar]
  • 37.Yim JE, Heshka S, Albu JB, Heymsfield S, Gallagher D. Femoral-gluteal subcutaneous and intermuscular adipose tissues have independent and opposing relationships with CVD risk. J Appl Physiol. 2008;104:700–707. doi: 10.1152/japplphysiol.01035.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wells JC. Sexual dimorphism of body composition, Best practice & research. Clinical endocrinology & metabolism. 2007;21:415–430. doi: 10.1016/j.beem.2007.04.007. [DOI] [PubMed] [Google Scholar]
  • 39.de Ridder CM, Bruning PF, Zonderland ML, Thijssen JH, Bonfrer JM, Blankenstein MA, Huisveld IA, Erich WB. Body fat mass, body fat distribution, and plasma hormones in early puberty in females. J Clin Endocrinol Metab. 1990;70:888–893. doi: 10.1210/jcem-70-4-888. [DOI] [PubMed] [Google Scholar]
  • 40.Stevens J, Katz EG, Huxley RR. Associations between gender, age and waist circumference. European journal of clinical nutrition. 2010;64:6–15. doi: 10.1038/ejcn.2009.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Shi H, Clegg DJ. Sex differences in the regulation of body weight. Physiol Behav. 2009;97:199–204. doi: 10.1016/j.physbeh.2009.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ley CJ, Lees B, Stevenson JC. Sex- and menopause-associated changes in body-fat distribution. Am J Clin Nutr. 1992;55:950–954. doi: 10.1093/ajcn/55.5.950. [DOI] [PubMed] [Google Scholar]
  • 43.Svendsen OL, Hassager C, Christiansen C. Age- and menopause-associated variations in body composition and fat distribution in healthy women as measured by dual-energy X-ray absorptiometry. Metabolism: clinical and experimental. 1995;44:369–373. doi: 10.1016/0026-0495(95)90168-x. [DOI] [PubMed] [Google Scholar]
  • 44.Toth MJ, Tchernof A, Sites CK, Poehlman ET. Effect of menopausal status on body composition and abdominal fat distribution. International journal of obesity and related metabolic disorders : journal of the International Association for the Study of Obesity. 2000;24:226–231. doi: 10.1038/sj.ijo.0801118. [DOI] [PubMed] [Google Scholar]
  • 45.Piche ME, Lapointe A, Weisnagel SJ, Corneau L, Nadeau A, Bergeron J, Lemieux S. Regional body fat distribution and metabolic profile in postmenopausal women. Metabolism. 2008;57:1101–1107. doi: 10.1016/j.metabol.2008.03.015. [DOI] [PubMed] [Google Scholar]
  • 46.Lovejoy JC, Champagne CM, de Jonge L, Xie H, Smith SR. Increased visceral fat and decreased energy expenditure during the menopausal transition. Int J Obes (Lond) 2008;32:949–958. doi: 10.1038/ijo.2008.25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Kotani K, Tokunaga K, Fujioka S, Kobatake T, Keno Y, Yoshida S, Shimomura I, Tarui S, Matsuzawa Y. Sexual dimorphism of age-related changes in whole-body fat distribution in the obese. Int J Obes Relat Metab Disord. 1994;18:207–202. [PubMed] [Google Scholar]
  • 48.Kanaley JA, Sames C, Swisher L, Swick AG, Ploutz-Snyder LL, Steppan CM, Sagendorf KS, Feiglin D, Jaynes EB, Meyer RA, Weinstock RS. Abdominal fat distribution in pre- and postmenopausal women: The impact of physical activity, age, and menopausal status. Metabolism. 2001;50:976–982. doi: 10.1053/meta.2001.24931. [DOI] [PubMed] [Google Scholar]
  • 49.Perrone G, Liu Y, Capri O, Critelli C, Barillaro F, Galoppi P, Zichella L. Evaluation of the body composition and fat distribution in long-term users of hormone replacement therapy. Gynecol Obstet Invest. 1999;48:52–55. doi: 10.1159/000010134. [DOI] [PubMed] [Google Scholar]
  • 50.Espeland MA, Stefanick ML, Kritz-Silverstein D, Fineberg SE, Waclawiw MA, James MK, Greendale GA. Effect of postmenopausal hormone therapy on body weight and waist and hip girths. Postmenopausal Estrogen-Progestin Interventions Study Investigators. The Journal of clinical endocrinology and metabolism. 1997;82:1549–1556. doi: 10.1210/jcem.82.5.3925. [DOI] [PubMed] [Google Scholar]
  • 51.Munoz J, Derstine A, Gower BA. Fat distribution and insulin sensitivity in postmenopausal women: influence of hormone replacement. Obesity research. 2002;10:424–431. doi: 10.1038/oby.2002.59. [DOI] [PubMed] [Google Scholar]
  • 52.Tchernof A, Poehlman ET, Despres JP. Despres, Body fat distribution, the menopause transition, and hormone replacement therapy. Diabetes & metabolism. 2000;26:12–20. [PubMed] [Google Scholar]
  • 53.Derby CA, Zilber S, Brambilla D, Morales KH, McKinlay JB. Body mass index, waist circumference and waist to hip ratio and change in sex steroid hormones: the Massachusetts Male Ageing Study. Clinical endocrinology. 2006;65:125–131. doi: 10.1111/j.1365-2265.2006.02560.x. [DOI] [PubMed] [Google Scholar]
  • 54.Puder JJ, Varga S, Kraenzlin M, De Geyter C, Keller U, Muller B. Central fat excess in polycystic ovary syndrome: relation to low-grade inflammation and insulin resistance. The Journal of clinical endocrinology and metabolism. 2005;90:6014–6021. doi: 10.1210/jc.2005-1002. [DOI] [PubMed] [Google Scholar]
  • 55.Carmina E, Bucchieri S, Esposito A, Del Puente A, Mansueto P, Orio F, Di Fede G, Rini G. Abdominal fat quantity and distribution in women with polycystic ovary syndrome and extent of its relation to insulin resistance. The Journal of clinical endocrinology and metabolism. 2007;92:2500–2505. doi: 10.1210/jc.2006-2725. [DOI] [PubMed] [Google Scholar]
  • 56.Escobar-Morreale HF, San Millan JL. Abdominal adiposity and the polycystic ovary syndrome. Trends Endocrinol Metab. 2007;18:266–272. doi: 10.1016/j.tem.2007.07.003. [DOI] [PubMed] [Google Scholar]
  • 57.Allan CA, Strauss BJ, Burger HG, Forbes EA, McLachlan RI. Testosterone therapy prevents gain in visceral adipose tissue and loss of skeletal muscle in nonobese aging men. The Journal of clinical endocrinology and metabolism. 2008;93:139–146. doi: 10.1210/jc.2007-1291. [DOI] [PubMed] [Google Scholar]
  • 58.Emmelot-Vonk MH, Verhaar HJ, Nakhai Pour HR, Aleman A, Lock TM, Bosch JL, Grobbee DE, van der Schouw YT. Effect of testosterone supplementation on functional mobility, cognition, and other parameters in older men: a randomized controlled trial. JAMA : the journal of the American Medical Association. 2008;299:39–52. doi: 10.1001/jama.2007.51. [DOI] [PubMed] [Google Scholar]
  • 59.Evans DJ, Hoffmann RG, Kalkhoff RK, Kissebah AH. Kissebah, Relationship of androgenic activity to body fat topography, fat cell morphology, and metabolic aberrations in premenopausal women. J Clin Endocrinol Metab. 1983;57:304–310. doi: 10.1210/jcem-57-2-304. [DOI] [PubMed] [Google Scholar]
  • 60.Haffner SM, Katz MS, Dunn JF. Increased upper body and overall adiposity is associated with decreased sex hormone binding globulin in postmenopausal women. Int J Obes. 1991;15:471–478. [PubMed] [Google Scholar]
  • 61.Lovejoy JC, Bray GA, Bourgeois MO, Macchiavelli R, Rood JC, Greeson C, Partington C. Exogenous androgens influence body composition and regional body fat distribution in obese postmenopausal women--a clinical research center study. J Clin Endocrinol Metab. 1996;81:2198–2203. doi: 10.1210/jcem.81.6.8964851. [DOI] [PubMed] [Google Scholar]
  • 62.Dunaif A. Hyperandrogenic anovulation (PCOS): a unique disorder of insulin action associated with an increased risk of non-insulin-dependent diabetes mellitus. Am J Med. 1995;98:33S–39S. doi: 10.1016/s0002-9343(99)80057-6. [DOI] [PubMed] [Google Scholar]
  • 63.Pedersen SB, Fuglsig S, Sjogren P, Richelsen B. Identification of steroid receptors in human adipose tissue. European journal of clinical investigation. 1996;26:1051–1056. doi: 10.1046/j.1365-2362.1996.380603.x. [DOI] [PubMed] [Google Scholar]
  • 64.Dieudonne MN, Pecquery R, Boumediene A, Leneveu MC, Giudicelli Y. Androgen receptors in human preadipocytes and adipocytes: regional specificities and regulation by sex steroids. Am J Physiol. 1998;274:C1645–C1652. doi: 10.1152/ajpcell.1998.274.6.C1645. [DOI] [PubMed] [Google Scholar]
  • 65.Anwar A, McTernan PG, Anderson LA, Askaa J, Moody CG, Barnett AH, Eggo MC, Kumar S. Site-specific regulation of oestrogen receptor-alpha and -beta by oestradiol in human adipose tissue. obesity & metabolism. 2001;3:338–349. doi: 10.1046/j.1463-1326.2001.00145.x. [DOI] [PubMed] [Google Scholar]
  • 66.Dieudonne MN, Leneveu MC, Giudicelli Y, Pecquery R. Evidence for functional estrogen receptors alpha and beta in human adipose cells: regional specificities and regulation by estrogens. American journal of physiology. Cell physiology. 2004;286:C655–661. doi: 10.1152/ajpcell.00321.2003. [DOI] [PubMed] [Google Scholar]
  • 67.Pedersen SB, Bruun JM, Hube F, Kristensen K, Hauner H, Richelsen B. Demonstration of estrogen receptor subtypes alpha and beta in human adipose tissue: influences of adipose cell differentiation and fat depot localization. Molecular and cellular endocrinology. 2001;182:27–37. doi: 10.1016/s0303-7207(01)00557-3. [DOI] [PubMed] [Google Scholar]
  • 68.Gavin KM, Cooper EE, Hickner RC. Estrogen receptor protein content is different in abdominal than gluteal subcutaneous adipose tissue of overweight-to-obese premenopausal women. Metabolism: clinical and experimental. 2013 doi: 10.1016/j.metabol.2013.02.010. [DOI] [PubMed] [Google Scholar]
  • 69.Hall JM, McDonnell DP. The estrogen receptor beta-isoform (ERbeta) of the human estrogen receptor modulates ERalpha transcriptional activity and is a key regulator of the cellular response to estrogens and antiestrogens. Endocrinology. 1999;140:5566–5578. doi: 10.1210/endo.140.12.7179. [DOI] [PubMed] [Google Scholar]
  • 70.Jones ME, Thorburn AW, Britt KL, Hewitt KN, Wreford NG, Proietto J, Oz OK, Leury BJ, Robertson KM, Yao S, Simpson ER. Aromatase-deficient (ArKO) mice have a phenotype of increased adiposity. Proc Natl Acad Sci U S A. 2000;97:12735–12740. doi: 10.1073/pnas.97.23.12735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Takeda K, Toda K, Saibara T, Nakagawa M, Saika K, Onishi T, Sugiura T, Shizuta Y. Progressive development of insulin resistance phenotype in male mice with complete aromatase (CYP19) deficiency. J Endocrinol. 2003;176:237–246. doi: 10.1677/joe.0.1760237. [DOI] [PubMed] [Google Scholar]
  • 72.Maffei L, Murata Y, Rochira V, Tubert G, Aranda C, Vazquez M, Clyne CD, Davis S, Simpson ER, Carani C. Dysmetabolic syndrome in a man with a novel mutation of the aromatase gene: effects of testosterone, alendronate, and estradiol treatment. The Journal of clinical endocrinology and metabolism. 2004;89:61–70. doi: 10.1210/jc.2003-030313. [DOI] [PubMed] [Google Scholar]
  • 73.Zirilli L, Rochira V, Diazzi C, Caffagni G, Carani C. Human models of aromatase deficiency. The Journal of steroid biochemistry and molecular biology. 2008;109:212–218. doi: 10.1016/j.jsbmb.2008.03.026. [DOI] [PubMed] [Google Scholar]
  • 74.Nilsson S, Gustafsson JA. Estrogen receptors: therapies targeted to receptor subtypes. Clinical pharmacology and therapeutics. 2011;89:44–55. doi: 10.1038/clpt.2010.226. [DOI] [PubMed] [Google Scholar]
  • 75.Simpson ER. Sources of estrogen and their importance. The Journal of steroid biochemistry and molecular biology. 2003;86:225–230. doi: 10.1016/s0960-0760(03)00360-1. [DOI] [PubMed] [Google Scholar]
  • 76.Stocco C. Tissue physiology and pathology of aromatase. Steroids. 2012;77:27–35. doi: 10.1016/j.steroids.2011.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Belanger C, Luu-The V, Dupont P, Tchernof A. Adipose tissue intracrinology: potential importance of local androgen/estrogen metabolism in the regulation of adiposity. Horm Metab Res. 2002;34:737–745. doi: 10.1055/s-2002-38265. [DOI] [PubMed] [Google Scholar]
  • 78.Meseguer A, Puche C, Cabero A. Sex steroid biosynthesis in white adipose tissue. Horm Metab Res. 2002;34:731–736. doi: 10.1055/s-2002-38249. [DOI] [PubMed] [Google Scholar]
  • 79.Mayes JS, Watson GH. Direct effects of sex steroid hormones on adipose tissues and obesity. Obesity reviews : an official journal of the International Association for the Study of Obesity. 2004;5:197–216. doi: 10.1111/j.1467-789X.2004.00152.x. [DOI] [PubMed] [Google Scholar]
  • 80.Poulos SP, Hausman DB, Hausman GJ. The development and endocrine functions of adipose tissue. Mol Cell Endocrinol. 2010;323:20–34. doi: 10.1016/j.mce.2009.12.011. [DOI] [PubMed] [Google Scholar]
  • 81.Joe AW, Yi L, Even Y, Vogl AW, Rossi FM. Depot-specific differences in adipogenic progenitor abundance and proliferative response to high-fat diet. Stem Cells. 2009;27:2563–2570. doi: 10.1002/stem.190. [DOI] [PubMed] [Google Scholar]
  • 82.Tchoukalova YD, Votruba SB, Tchkonia T, Giorgadze N, Kirkland JL, Jensen MD. Regional differences in cellular mechanisms of adipose tissue gain with overfeeding. Proc Natl Acad Sci U S A. 2010;107:18226–18231. doi: 10.1073/pnas.1005259107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Bjorntorp P, Bengtsson C, Blohme G, Jonsson A, Sjostrom L, Tibblin E, Tibblin G, Wilhelmsen L. Adipose tissue fat cell size and number in relation to metabolism in randomly selected middle-aged men and women. Metabolism. 1971;20:927–935. doi: 10.1016/0026-0495(71)90013-8. [DOI] [PubMed] [Google Scholar]
  • 84.Weyer C, Foley JE, Bogardus C, Tataranni PA, Pratley RE. Enlarged subcutaneous abdominal adipocyte size, but not obesity itself, predicts type II diabetes independent of insulin resistance. Diabetologia. 2000;43:1498–1506. doi: 10.1007/s001250051560. [DOI] [PubMed] [Google Scholar]
  • 85.Lundgren M, Svensson M, Lindmark S, Renstrom F, Ruge T, Eriksson JW. Fat cell enlargement is an independent marker of insulin resistance and ‘hyperleptinaemia’. Diabetologia. 2007;50:625–633. doi: 10.1007/s00125-006-0572-1. [DOI] [PubMed] [Google Scholar]
  • 86.Virtue S, Vidal-Puig A, Adipose tissue expandability. lipotoxicity and the Metabolic Syndrome--an allostatic perspective. Biochim Biophys Acta. 2010;1801:338–349. doi: 10.1016/j.bbalip.2009.12.006. [DOI] [PubMed] [Google Scholar]
  • 87.Isakson P, Hammarstedt A, Gustafson B, Smith U. Smith, Impaired preadipocyte differentiation in human abdominal obesity: role of Wnt, tumor necrosis factor-alpha, and inflammation. Diabetes. 2009;58:1550–1557. doi: 10.2337/db08-1770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.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]
  • 89.Hauner H, Ditschuneit HH, Pal SB, Moncayo R, Pfeiffer EF. Fat distribution, endocrine and metabolic profile in obese women with and without hirsutism. Metabolism. 1988;37:281–286. doi: 10.1016/0026-0495(88)90109-6. [DOI] [PubMed] [Google Scholar]
  • 90.Gregoire FM, Johnson PR, Greenwood MR. Comparison of the adipoconversion of preadipocytes derived from lean and obese Zucker rats in serum-free cultures. Int J Obes Relat Metab Disord. 1995;19:664–670. [PubMed] [Google Scholar]
  • 91.Tchoukalova YD, Koutsari C, Votruba SB, Tchkonia T, Giorgadze N, Thomou T, Kirkland JL, Jensen MD. Sex- and Depot-Dependent Differences in Adipogenesis in Normal-Weight Humans. Obesity (Silver Spring) 2010 doi: 10.1038/oby.2010.56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Spalding KL, Arner E, Westermark PO, Bernard S, Buchholz BA, Bergmann O, Blomqvist L, Hoffstedt J, Naslund E, Britton T, Concha H, Hassan M, Ryden M, Frisen J, Arner P. Dynamics of fat cell turnover in humans. Nature. 2008;453:783–787. doi: 10.1038/nature06902. [DOI] [PubMed] [Google Scholar]
  • 93.Strissel KJ, Stancheva Z, Miyoshi H, Perfield JW, 2nd, DeFuria J, Jick Z, Greenberg AS, Obin MS. Adipocyte death, adipose tissue remodeling, and besity complications, Diabetes. 2007;56:2910–2918. doi: 10.2337/db07-0767. [DOI] [PubMed] [Google Scholar]
  • 94.McLaughlin T, Sherman A, Tsao P, Gonzalez O, Yee G, Lamendola C, Reaven GM, Cushman SW. Enhanced proportion of small adipose cells in insulin-resistant vs insulin-sensitive obese individuals implicates impaired adipogenesis. Diabetologia. 2007;50:1707–1715. doi: 10.1007/s00125-007-0708-y. [DOI] [PubMed] [Google Scholar]
  • 95.McLaughlin TM, Liu T, Yee G, Abbasi F, Lamendola C, Reaven G, Tsao P, Cushman S, Sherman A. Pioglitazone Increases the Proportion of Small Cells in Human Abdominal Subcutaneous Adipose Tissue. Obesity (Silver Spring) 2009 doi: 10.1038/oby.2009.380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Lee MJ, Wu Y, Fried SK. A modified protocol to maximize differentiation of human preadipocytes and improve metabolic phenotypes. Obesity (Silver Spring) 2012;20:2334–2340. doi: 10.1038/oby.2012.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Skurk T, Hauner H. Primary culture of human adipocyte precursor cells: expansion and differentiation. Methods Mol Biol. 2012;806:215–226. doi: 10.1007/978-1-61779-367-7_15. [DOI] [PubMed] [Google Scholar]
  • 98.MacKellar J, Cushman SW, Periwal V. Waves of adipose tissue growth in the genetically obese Zucker fatty rat. PLoS One. 2010;5:e8197. doi: 10.1371/journal.pone.0008197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Arner E, Westermark PO, Spalding KL, Britton T, Ryden M, Frisen J, Bernard S, Arner P. Adipocyte turnover: relevance to human adipose tissue morphology. Diabetes. 2010;59:105–109. doi: 10.2337/db09-0942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Roldan M, Macias-Gonzalez M, Garcia R, Tinahones FJ, 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]
  • 101.Strawford A, Antelo F, Christiansen M, Hellerstein MK. Adipose tissue triglyceride turnover, de novo lipogenesis, and cell proliferation in humans measured with 2H2O. Am J Physiol Endocrinol Metab. 2004;286:E577–E588. doi: 10.1152/ajpendo.00093.2003. [DOI] [PubMed] [Google Scholar]
  • 102.Tchoukalova YD, Fitch M, Rogers PM, Covington JD, Henagan TM, Ye J, Hellerstein MK, Ravussin E. In vivo adipogenesis in rats measured by cell kinetics in adipocytes and plastic-adherent stroma-vascular cells in response to high-fat diet and thiazolidinedione. Diabetes. 2012;61:137–144. doi: 10.2337/db10-1768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.White UA, Tchoukalova YD. Implications of 2H-labeling of DNA protocol to measure in vivo cell turnover in adipose tissue. Adipocyte. 2012;1:242–245. doi: 10.4161/adip.20817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Tchkonia T, Lenburg M, Thomou T, Giorgadze N, Frampton G, Pirtskhalava T, Cartwright A, Cartwright M, Flanagan J, Karagiannides I, Gerry N, Forse RA, Tchoukalova Y, Jensen MD, Pothoulakis C, Kirkland JL. Identification of depot-specific human fat cell progenitors through distinct expression profiles and developmental gene patterns. Am J Physiol Endocrinol Metab. 2007;292:E298–E307. doi: 10.1152/ajpendo.00202.2006. [DOI] [PubMed] [Google Scholar]
  • 105.Van Harmelen V, Rohrig K, Hauner H. Comparison of proliferation and differentiation capacity of human adipocyte precursor cells from the omental and subcutaneous adipose tissue depot of obese subjects. Metabolism. 2004;53:632–637. doi: 10.1016/j.metabol.2003.11.012. [DOI] [PubMed] [Google Scholar]
  • 106.Hauner H, Entenmann G. Regional variation of adipose differentiation in cultured stromal-vascular cells from the abdominal and femoral adipose tissue of obese women. Int J Obes. 1991;15:121–126. [PubMed] [Google Scholar]
  • 107.Tchoukalova YD, Koutsari C, Votruba SB, Tchkonia T, Giorgadze N, Thomou T, Kirkland JL, Jensen MD. Sex- and depot-dependent differences in adipogenesis in normal-weight humans. Obesity. 2010;18:1875–1880. doi: 10.1038/oby.2010.56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Maslowska MH, Sniderman AD, MacLean LD, Cianflone K. Regional differences in triacylglycerol synthesis in adipose tissue and in cultured preadipocytes. J Lipid Res. 1993;34:219–228. [PubMed] [Google Scholar]
  • 109.Djian P, Roncari AK, Hollenberg CH. Influence of anatomic site and age on the replication and differentiation of rat adipocyte precursors in culture. J Clin Invest. 1983;72:1200–1208. doi: 10.1172/JCI111075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Djian P, Roncari DA, Hollenberg CH. Adipocyte precursor clones vary in capacity for differentiation. Metabolism. 1985;34:880–883. doi: 10.1016/0026-0495(85)90114-3. [DOI] [PubMed] [Google Scholar]
  • 111.Wang H, Kirkland JL, Hollenberg CH. Varying capacities for replication of rat adipocyte precursor clones and adipose tissue growth. J Clin Invest. 1989;83:1741–1746. doi: 10.1172/JCI114075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Hauner H, Entenmann G, Wabitsch M, Gaillard D, Ailhaud G, Negrel R, Pfeiffer EF. Promoting effect of glucocorticoids on the differentiation of human adipocyte precursor cells cultured in a chemically defined medium. J Clin Invest. 1989;84:1663–1670. doi: 10.1172/JCI114345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Kirkland JL, Hollenberg CH, Gillon WS, Age anatomic site. and the replication and differentiation of adipocyte precursors. Am J Physiol. 1990;258:C206–C210. doi: 10.1152/ajpcell.1990.258.2.C206. [DOI] [PubMed] [Google Scholar]
  • 114.Sztalryd C, Faust IM. Depot-specific features of adipocyte progenitors revealed by primary cultures plated at low density. Int J Obes. 1990;14(Suppl 3):165–175. [PubMed] [Google Scholar]
  • 115.Gregoire F, Todoroff G, Hauser N, Remacle C. The stroma-vascular fraction of rat inguinal and epididymal adipose tissue and the adipoconversion of fat cell precursors in primary culture. Biol Cell. 1990;69:215–222. doi: 10.1016/0248-4900(90)90348-7. [DOI] [PubMed] [Google Scholar]
  • 116.Kirkland JL, Hollenberg CH, Gillon WS. Two preadipocyte subtypes cloned from human omental fat. Obes Res. 1993;1:87–91. doi: 10.1002/j.1550-8528.1993.tb00596.x. [DOI] [PubMed] [Google Scholar]
  • 117.Carraro R, Li ZH, Johnson JE, Jr., Gregerman RI. Adipocytes of old rats produce a decreased amount of differentiation factor for preadipocytes derived from adipose tissue islets. J Gerontol. 1992;47:B198–B201. doi: 10.1093/geronj/47.6.b198. [DOI] [PubMed] [Google Scholar]
  • 118.Kirkland JL, Hollenberg CH, Gillon WS. Effects of fat depot site on differentiation-dependent gene expression in rat preadipocytes. Int J Obes Relat Metab Disord. 1996;20(Suppl 3):S102–S107. [PubMed] [Google Scholar]
  • 119.Tchkonia T, Giorgadze N, Pirtskhalava T, Tchoukalova Y, Karagiannides I, Forse RA, DePonte M, Stevenson M, Guo W, Han J, Waloga G, Lash TL, Jensen MD, Kirkland JL. Fat depot origin affects adipogenesis in primary cultured and cloned human preadipocytes. Am J Physiol Regul Integr Comp Physiol. 2002;282:R1286–R1296. doi: 10.1152/ajpregu.00653.2001. [DOI] [PubMed] [Google Scholar]
  • 120.Ellis JR, McDonald RB, Stern JS. A diet high in fat stimulates adipocyte proliferation in older (22 month) rats. Exp Gerontol. 1990;25:141–148. doi: 10.1016/0531-5565(90)90045-4. [DOI] [PubMed] [Google Scholar]
  • 121.Macotela Y, Emanuelli B, Mori MA, Gesta S, Schulz TJ, Tseng YH, Kahn CR. Intrinsic differences in adipocyte precursor cells from different white fat depots. Diabetes. 2012;61:1691–1699. doi: 10.2337/db11-1753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Niesler CU, Siddle K, Prins JB. Human preadipocytes display a depot-specific susceptibility to apoptosis. Diabetes. 1998;47:1365–1368. doi: 10.2337/diab.47.8.1365. [DOI] [PubMed] [Google Scholar]
  • 123.Papineau D, Gagnon A, Sorisky A. Apoptosis of human abdominal preadipocytes before and after differentiation into adipocytes in culture. Metabolism. 2003;52:987–992. doi: 10.1016/s0026-0495(03)00165-3. [DOI] [PubMed] [Google Scholar]
  • 124.Maumus M, Sengenes C, Decaunes P, Zakaroff-Girard A, Bourlier V, Lafontan M, Galitzky J, Bouloumie A. Evidence of in situ proliferation of adult adipose tissue-derived progenitor cells: influence of fat mass microenvironment and growth. J Clin Endocrinol Metab. 2008;93:4098–4106. doi: 10.1210/jc.2008-0044. [DOI] [PubMed] [Google Scholar]
  • 125.Maumus M, Peyrafitte JA, D’Angelo R, Fournier-Wirth C, Bouloumie A, Casteilla L, Sengenes C, Bourin P. Native human adipose stromal cells: localization, morphology and phenotype. Int J Obes (Lond) 2011;35:1141–1153. doi: 10.1038/ijo.2010.269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Tang W, Zeve D, Seo J, Jo AY, Graff JM. Thiazolidinediones regulate adipose lineage dynamics. Cell Metab. 2011;14:116–122. doi: 10.1016/j.cmet.2011.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Permana PA, Nair S, Lee YH, Luczy-Bachman G, Vozarova De, Courten B, Tataranni PA. Subcutaneous abdominal preadipocyte differentiation in vitro inversely correlates with central obesity. Am J Physiol Endocrinol Metab. 2004;286:E958–E962. doi: 10.1152/ajpendo.00544.2003. [DOI] [PubMed] [Google Scholar]
  • 128.Baglioni S, Francalanci M, Squecco R, Lombardi A, Cantini G, Angeli R, Gelmini S, Guasti D, Benvenuti S, Annunziato F, Bani D, Liotta F, Francini F, Perigli G, Serio M, Luconi M. Characterization of human adult stem-cell populations isolated from visceral and subcutaneous adipose tissue. FASEB J. 2009;23:3494–3505. doi: 10.1096/fj.08-126946. [DOI] [PubMed] [Google Scholar]
  • 129.Tchoukalova YD, Koutsari C, Karpyak MV, Votruba SB, Wendland E, Jensen MD. Subcutaneous adipocyte size and body fat distribution. The American journal of clinical nutrition. 2008;87:56–63. doi: 10.1093/ajcn/87.1.56. [DOI] [PubMed] [Google Scholar]
  • 130.Drolet R, Richard C, Sniderman AD, Mailloux J, Fortier M, Huot C, Rheaume C, Tchernof A. Hypertrophy and hyperplasia of abdominal adipose tissues in women. Int J Obes (Lond) 2008;32:283–291. doi: 10.1038/sj.ijo.0803708. [DOI] [PubMed] [Google Scholar]
  • 131.Tchkonia T, Tchoukalova YD, Giorgadze N, Pirtskhalava T, Karagiannides I, Forse RA, Koo A, Stevenson M, Chinnappan D, Cartwright A, Jensen MD, Kirkland JL. Abundance of two human preadipocyte subtypes with distinct capacities for replication, adipogenesis, and apoptosis varies among fat depots. Am J Physiol Endocrinol Metab. 2005;288:E267–E277. doi: 10.1152/ajpendo.00265.2004. [DOI] [PubMed] [Google Scholar]
  • 132.Singh R, Artaza JN, Taylor WE, Gonzalez-Cadavid NF, Bhasin S. Androgens stimulate myogenic differentiation and inhibit adipogenesis in C3H 10T1/2 pluripotent cells through an androgen receptor-mediated pathway. Endocrinology. 2003;144:5081–5088. doi: 10.1210/en.2003-0741. [DOI] [PubMed] [Google Scholar]
  • 133.Singh R, Artaza JN, Taylor WE, Braga M, Yuan X, Gonzalez-Cadavid NF, Bhasin S. Testosterone inhibits adipogenic differentiation in 3T3-L1 cells: nuclear translocation of androgen receptor complex with beta-catenin and T-cell factor 4 may bypass canonical Wnt signaling to down-regulate adipogenic transcription factors. Endocrinology. 2006;147:141–154. doi: 10.1210/en.2004-1649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Roncari DA, Van RL. Promotion of human adipocyte precursor replication by 17beta-estradiol in culture. J Clin Invest. 1978;62:503–508. doi: 10.1172/JCI109153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Lacasa D, Garcia E, Agli B, Giudicelli Y. Control of rat preadipocyte adipose conversion by ovarian status: regional specificity and possible involvement of the mitogen-activated protein kinase-dependent and c-fos signaling pathways. Endocrinology. 1997;138:2729–2734. doi: 10.1210/endo.138.7.5246. [DOI] [PubMed] [Google Scholar]
  • 136.Anderson LA, McTernan PG, Barnett AH, Kumar S. The effects of androgens and estrogens on preadipocyte proliferation in human adipose tissue: influence of gender and site. J Clin Endocrinol Metab. 2001;86:5045–5051. doi: 10.1210/jcem.86.10.7955. [DOI] [PubMed] [Google Scholar]
  • 137.Dieudonne MN, Pecquery R, Leneveu MC, Giudicelli Y. Opposite effects of androgens and estrogens on adipogenesis in rat preadipocytes: evidence for sex and site-related specificities and possible involvement of insulin-like growth factor 1 receptor and peroxisome proliferator-activated receptor gamma2. Endocrinology. 2000;141:649–656. doi: 10.1210/endo.141.2.7293. [DOI] [PubMed] [Google Scholar]
  • 138.Lacasa D, Le Liepvre X, Ferre P, Dugail I. Progesterone stimulates adipocyte determination and differentiation 1/sterol regulatory element-binding protein 1c gene expression. potential mechanism for the lipogenic effect of progesterone in adipose tissue. J Biol Chem. 2001;276:11512–11516. doi: 10.1074/jbc.M008556200. [DOI] [PubMed] [Google Scholar]
  • 139.Bulun SE, Simpson ER. Competitive reverse transcription-polymerase chain reaction analysis indicates that levels of aromatase cytochrome P450 transcripts in adipose tissue of buttocks, thighs, and abdomen of women increase with advancing age. J Clin Endocrinol Metab. 1994;78:428–432. doi: 10.1210/jcem.78.2.8106632. [DOI] [PubMed] [Google Scholar]
  • 140.Yang X, Schadt EE, Wang S, Wang H, Arnold AP, Ingram-Drake L, Drake TA, Lusis AJ. Tissue-specific expression and regulation of sexually dimorphic genes in mice. Genome Res. 2006;16:995–1004. doi: 10.1101/gr.5217506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Gustafson B, Gogg S, Hedjazifar S, Jenndahl L, Hammarstedt A, Smith U. Inflammation and impaired adipogenesis in hypertrophic obesity in man. Am J Physiol Endocrinol Metab. 2009 doi: 10.1152/ajpendo.00377.2009. [DOI] [PubMed] [Google Scholar]
  • 142.Cantile M, Procino A, D’Armiento M, Cindolo L, Cillo C. HOX gene network is involved in the transcriptional regulation of in vivo human adipogenesis. J Cell Physiol. 2003;194:225–236. doi: 10.1002/jcp.10210. [DOI] [PubMed] [Google Scholar]
  • 143.Vohl MC, Sladek R, Robitaille J, Gurd S, Marceau P, Richard D, Hudson TJ, Tchernof A. A survey of genes differentially expressed in subcutaneous and visceral adipose tissue in men. Obes Res. 2004;12:1217–1222. doi: 10.1038/oby.2004.153. [DOI] [PubMed] [Google Scholar]
  • 144.Gesta S, Bluher M, Yamamoto Y, Norris AW, Berndt J, Kralisch S, Boucher J, Lewis C, Kahn CR. Evidence for a role of developmental genes in the origin of obesity and body fat distribution. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:6676–6681. doi: 10.1073/pnas.0601752103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Tchkonia T, Giorgadze N, Pirtskhalava T, Thomou T, DePonte M, Koo A, Forse RA, Chinnappan D, Martin-Ruiz C, von Zglinicki T, Kirkland JL. Fat depot-specific characteristics are retained in strains derived from single human preadipocytes. Diabetes. 2006;55:2571–2578. doi: 10.2337/db06-0540. [DOI] [PubMed] [Google Scholar]
  • 146.Karastergiou K, Fried SK, Xie H, Lee MJ, Divoux A, Rosencrantz MA, Chang RJ, Smith SR. Distinct Developmental Signatures of Human Abdominal and Gluteal Subcutaneous Adipose Tissue Depots. J Clin Endocrinol Metab. 2012 doi: 10.1210/jc.2012-2953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Heid IM, Jackson AU, Randall JC, Winkler TW, Qi L, Steinthorsdottir V, Thorleifsson G, Zillikens MC, Speliotes EK, Magi R, Workalemahu T, White CC, Bouatia-Naji N, Harris TB, Berndt SI, Ingelsson E, Willer CJ, Weedon MN, Luan J, Vedantam S, Esko T, Kilpelainen TO, Kutalik Z, Li S, Monda KL, Dixon AL, Holmes CC, Kaplan LM, Liang L, Min JL, Moffatt MF, Molony C, Nicholson G, Schadt EE, Zondervan KT, Feitosa MF, Ferreira T, Lango Allen H, Weyant RJ, Wheeler E, Wood AR, Estrada K, Goddard ME, Lettre G, Mangino M, Nyholt DR, Purcell S, Smith AV, Visscher PM, Yang J, McCarroll SA, Nemesh J, Voight BF, Absher D, Amin N, Aspelund T, Coin L, Glazer NL, Hayward C, Heard-Costa NL, Hottenga JJ, Johansson A, Johnson T, Kaakinen M, Kapur K, Ketkar S, Knowles JW, Kraft P, Kraja AT, Lamina C, Leitzmann MF, McKnight B, Morris AP, Ong KK, Perry JR, Peters MJ, Polasek O, Prokopenko I, Rayner NW, Ripatti S, Rivadeneira F, Robertson NR, Sanna S, Sovio U, Surakka I, Teumer A, van Wingerden S, Vitart V, Zhao JH, Cavalcanti-Proenca C, Chines PS, Fisher E, Kulzer JR, Lecoeur C, Narisu N, Sandholt C, Scott LJ, Silander K, Stark K, Tammesoo ML, Teslovich TM, Timpson NJ, Watanabe RM, Welch R, Chasman DI, Cooper MN, Jansson JO, Kettunen J, Lawrence RW, Pellikka N, Perola M, Vandenput L, Alavere H, Almgren P, Atwood LD, Bennett AJ, Biffar R, Bonnycastle LL, Bornstein SR, Buchanan TA, Campbell H, Day IN, Dei M, Dorr M, Elliott P, Erdos MR, Eriksson JG, Freimer NB, Fu M, Gaget S, Geus EJ, Gjesing AP, Grallert H, Grassler J, Groves CJ, Guiducci C, Hartikainen AL, Hassanali N, Havulinna AS, Herzig KH, Hicks AA, Hui J, Igl W, Jousilahti P, Jula A, Kajantie E, Kinnunen L, Kolcic I, Koskinen S, Kovacs P, Kroemer HK, Krzelj V, Kuusisto J, Kvaloy K, Laitinen J, Lantieri O, Lathrop GM, Lokki ML, Luben RN, Ludwig B, McArdle WL, McCarthy A, Morken MA, Nelis M, Neville MJ, Pare G, Parker AN, Peden JF, Pichler I, Pietilainen KH, Platou CG, Pouta A, Ridderstrale M, Samani NJ, Saramies J, Sinisalo J, Smit JH, Strawbridge RJ, Stringham HM, Swift AJ, Teder-Laving M, Thomson B, Usala G, van Meurs JB, van Ommen GJ, Vatin V, Volpato CB, Wallaschofski H, Walters GB, Widen E, Wild SH, Willemsen G, Witte DR, Zgaga L, Zitting P, Beilby JP, James AL, Kahonen M, Lehtimaki T, Nieminen MS, Ohlsson C, Palmer LJ, Raitakari O, Ridker PM, Stumvoll M, Tonjes A, Viikari J, Balkau B, Ben-Shlomo Y, Bergman RN, Boeing H, Smith GD, Ebrahim S, Froguel P, Hansen T, Hengstenberg C, Hveem K, Isomaa B, Jorgensen T, Karpe F, Khaw KT, Laakso M, Lawlor DA, Marre M, Meitinger T, Metspalu A, Midthjell K, Pedersen O, Salomaa V, Schwarz PE, Tuomi T, Tuomilehto J, Valle TT, Wareham NJ, Arnold AM, Beckmann JS, Bergmann S, Boerwinkle E, Boomsma DI, Caulfield MJ, Collins FS, Eiriksdottir G, Gudnason V, Gyllensten U, Hamsten A, Hattersley AT, Hofman A, Hu FB, Illig T, Iribarren C, Jarvelin MR, Kao WH, Kaprio J, Launer LJ, Munroe PB, Oostra B, Penninx BW, Pramstaller PP, Psaty BM, Quertermous T, Rissanen A, Rudan I, Shuldiner AR, Soranzo N, Spector TD, Syvanen AC, Uda M, Uitterlinden A, Volzke H, Vollenweider P, Wilson JF, Witteman JC, Wright AF, Abecasis GR, Boehnke M, Borecki IB, Deloukas P, Frayling TM, Groop LC, Haritunians T, Hunter DJ, Kaplan RC, North KE, O’Connell JR, Peltonen L, Schlessinger D, Strachan DP, Hirschhorn JN, Assimes TL, Wichmann HE, Thorsteinsdottir U, van Duijn CM, Stefansson K, Cupples LA, Loos RJ, Barroso I, McCarthy MI, Fox CS, Mohlke KL, Lindgren CM. Meta-analysis identifies 13 new loci associated with waist-hip ratio and reveals sexual dimorphism in the genetic basis of fat distribution. Nature genetics. 2010;42:949–960. doi: 10.1038/ng.685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Holland PW, Booth HA, Bruford EA. Classification and nomenclature of all human homeobox genes. BMC biology. 2007;5:47. doi: 10.1186/1741-7007-5-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Hombria JC, Lovegrove B. Beyond homeosis--HOX function in morphogenesis and organogenesis. Differentiation; research in biological diversity. 2003;71:461–476. doi: 10.1046/j.1432-0436.2003.7108004.x. [DOI] [PubMed] [Google Scholar]
  • 150.Cartwright MJ, Schlauch K, Lenburg ME, Tchkonia T, Pirtskhalava T, Cartwright A, Thomou T, Kirkland JL. Aging, depot origin, and preadipocyte gene expression. J Gerontol A Biol Sci Med Sci. 2010;65:242–251. doi: 10.1093/gerona/glp213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Yamamoto Y, Gesta S, Lee KY, Tran TT, Saadatirad P, Kahn CR. Adipose depots possess unique developmental gene signatures. Obesity (Silver Spring) 2010;18:872–878. doi: 10.1038/oby.2009.512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Monteiro MC, Sanyal M, Cleary ML, Sengenes C, Bouloumie A, Dani C, Billon N. PBX1: a novel stage-specific regulator of adipocyte development. Stem cells. 2011;29:1837–1848. doi: 10.1002/stem.737. [DOI] [PubMed] [Google Scholar]
  • 153.Heid IM, Jackson AU, Randall JC, Winkler TW, Qi L, Steinthorsdottir V, Thorleifsson G, Zillikens MC, Speliotes EK, Magi R, Workalemahu T, White CC, Bouatia-Naji N, Harris TB, Berndt SI, Ingelsson E, Willer CJ, Weedon MN, Luan J, Vedantam S, Esko T, Kilpelainen TO, Kutalik Z, Li S, Monda KL, Dixon AL, Holmes CC, Kaplan LM, Liang L, Min JL, Moffatt MF, Molony C, Nicholson G, Schadt EE, Zondervan KT, Feitosa MF, Ferreira T, Allen HL, Weyant RJ, Wheeler E, Wood AR, Estrada Magic K, Goddard ME, Lettre G, Mangino M, Nyholt DR, Purcell S, Smith AV, Visscher PM, Yang J, McCarroll SA, Nemesh J, Voight BF, Absher D, Amin N, Aspelund T, Coin L, Glazer NL, Hayward C, Heard-Costa NL, Hottenga JJ, Johansson A, Johnson T, Kaakinen M, Kapur K, Ketkar S, Knowles JW, Kraft P, Kraja AT, Lamina C, Leitzmann MF, McKnight B, Morris AP, Ong KK, Perry JR, Peters MJ, Polasek O, Prokopenko I, Rayner NW, Ripatti S, Rivadeneira F, Robertson NR, Sanna S, Sovio U, Surakka I, Teumer A, van Wingerden S, Vitart V, Zhao JH, Cavalcanti-Proenca C, Chines PS, Fisher E, Kulzer JR, Lecoeur C, Narisu N, Sandholt C, Scott LJ, Silander K, Stark K, Tammesoo ML, Teslovich TM, Timpson NJ, Watanabe RM, Welch R, Chasman DI, Cooper MN, Jansson JO, Kettunen J, Lawrence RW, Pellikka N, Perola M, Vandenput L, Alavere H, Almgren P, Atwood LD, Bennett AJ, Biffar R, Bonnycastle LL, Bornstein SR, Buchanan TA, Campbell H, Day IN, Dei M, Dorr M, Elliott P, Erdos MR, Eriksson JG, Freimer NB, Fu M, Gaget S, Geus EJ, Gjesing AP, Grallert H, Grassler J, Groves CJ, Guiducci C, Hartikainen AL, Hassanali N, Havulinna AS, Herzig KH, Hicks AA, Hui J, Igl W, Jousilahti P, Jula A, Kajantie E, Kinnunen L, Kolcic I, Koskinen S, Kovacs P, Kroemer HK, Krzelj V, Kuusisto J, Kvaloy K, Laitinen J, Lantieri O, Lathrop GM, Lokki ML, Luben RN, Ludwig B, McArdle WL, McCarthy A, Morken MA, Nelis M, Neville MJ, Pare G, Parker AN, Peden JF, Pichler I, Pietilainen KH, Platou CG, Pouta A, Ridderstrale M, Samani NJ, Saramies J, Sinisalo J, Smit JH, Strawbridge RJ, Stringham HM, Swift AJ, Teder-Laving M, Thomson B, Usala G, van Meurs JB, van Ommen GJ, Vatin V, Volpato CB, Wallaschofski H, Walters GB, Widen E, Wild SH, Willemsen G, Witte DR, Zgaga L, Zitting P, Beilby JP, James AL, Kahonen M, Lehtimaki T, Nieminen MS, Ohlsson C, Palmer LJ, Raitakari O, Ridker PM, Stumvoll M, Tonjes A, Viikari J, Balkau B, Ben-Shlomo Y, Bergman RN, Boeing H, Smith GD, Ebrahim S, Froguel P, Hansen T, Hengstenberg C, Hveem K, Isomaa B, Jorgensen T, Karpe F, Khaw KT, Laakso M, Lawlor DA, Marre M, Meitinger T, Metspalu A, Midthjell K, Pedersen O, Salomaa V, Schwarz PE, Tuomi T, Tuomilehto J, Valle TT, Wareham NJ, Arnold AM, Beckmann JS, Bergmann S, Boerwinkle E, Boomsma DI, Caulfield MJ, Collins FS, Eiriksdottir G, Gudnason V, Gyllensten U, Hamsten A, Hattersley AT, Hofman A, Hu FB, Illig T, Iribarren C, Jarvelin MR, Kao WH, Kaprio J, Launer LJ, Munroe PB, Oostra B, Penninx BW, Pramstaller PP, Psaty BM, Quertermous T, Rissanen A, Rudan I, Shuldiner AR, Soranzo N, Spector TD, Syvanen AC, Uda M, Uitterlinden A, Volzke H, Vollenweider P, Wilson JF, Witteman JC, Wright AF, Abecasis GR, Boehnke M, Borecki IB, Deloukas P, Frayling TM, Groop LC, Haritunians T, Hunter DJ, Kaplan RC, North KE, O’Connell JR, Peltonen L, Schlessinger D, Strachan DP, Hirschhorn JN, Assimes TL, Wichmann HE, Thorsteinsdottir U, van Duijn CM, Stefansson K, Cupples LA, Loos RJ, Barroso I, McCarthy MI, Fox CS, Mohlke KL, Lindgren CM. Meta-analysis identifies 13 new loci associated with waist-hip ratio and reveals sexual dimorphism in the genetic basis of fat distribution. Nat Genet. 2010;42:949–960. doi: 10.1038/ng.685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Lindgren CM, Heid IM, Randall JC, Lamina C, Steinthorsdottir V, Qi L, Speliotes EK, Thorleifsson G, Willer CJ, Herrera BM, Jackson AU, Lim N, Scheet P, Soranzo N, Amin N, Aulchenko YS, Chambers JC, Drong A, Luan J, Lyon HN, Rivadeneira F, Sanna S, Timpson NJ, Zillikens MC, Zhao JH, Almgren P, Bandinelli S, Bennett AJ, Bergman RN, Bonnycastle LL, Bumpstead SJ, Chanock SJ, Cherkas L, Chines P, Coin L, Cooper C, Crawford G, Doering A, Dominiczak A, Doney AS, Ebrahim S, Elliott P, Erdos MR, Estrada K, Ferrucci L, Fischer G, Forouhi NG, Gieger C, Grallert H, Groves CJ, Grundy S, Guiducci C, Hadley D, Hamsten A, Havulinna AS, Hofman A, Holle R, Holloway JW, Illig T, Isomaa B, Jacobs LC, Jameson K, Jousilahti P, Karpe F, Kuusisto J, Laitinen J, Lathrop GM, Lawlor DA, Mangino M, McArdle WL, Meitinger T, Morken MA, Morris AP, Munroe P, Narisu N, Nordstrom A, Nordstrom P, Oostra BA, Palmer CN, Payne F, Peden JF, Prokopenko I, Renstrom F, Ruokonen A, Salomaa V, Sandhu MS, Scott LJ, Scuteri A, Silander K, Song K, Yuan X, Stringham HM, Swift AJ, Tuomi T, Uda M, Vollenweider P, Waeber G, Wallace C, Walters GB, Weedon MN, Wellcome Trust Case Control C, Witteman JC, Zhang C, Zhang W, Caulfield MJ, Collins FS, Davey Smith G, Day IN, Franks PW, Hattersley AT, Hu FB, Jarvelin MR, Kong A, Kooner JS, Laakso M, Lakatta E, Mooser V, Morris AD, Peltonen L, Samani NJ, Spector TD, Strachan DP, Tanaka T, Tuomilehto J, Uitterlinden AG, van Duijn CM, Wareham NJ, Hugh W, Procardis C, Waterworth DM, Boehnke M, Deloukas P, Groop L, Hunter DJ, Thorsteinsdottir U, Schlessinger D, Wichmann HE, Frayling TM, Abecasis GR, Hirschhorn JN, Loos RJ, Stefansson K, Mohlke KL, Barroso I, McCarthy MI, Giant C. Genome-wide association scan meta-analysis identifies three Loci influencing adiposity and fat distribution. PLoS Genet. 2009;5:e1000508. doi: 10.1371/journal.pgen.1000508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Nilsson C, Niklasson M, Eriksson E, Bjorntorp P, Holmang A. Imprinting of female offspring with testosterone results in insulin resistance and changes in body fat distribution at adult age in rats. The Journal of clinical investigation. 1998;101:74–78. doi: 10.1172/JCI1353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Okura T, Koda M, Ando F, Niino N, Ohta S, Shimokata H. Association of polymorphisms in the estrogen receptor alpha gene with body fat distribution. International journal of obesity and related metabolic disorders : journal of the International Association for the Study of Obesity. 2003;27:1020–1027. doi: 10.1038/sj.ijo.0802378. [DOI] [PubMed] [Google Scholar]
  • 157.Gallagher CJ, Langefeld CD, Gordon CJ, Campbell JK, Mychaleckyj JC, Bryer-Ash M, Rich SS, Bowden DW, Sale MM. Association of the estrogen receptor-alpha gene with the metabolic syndrome and its component traits in African-American families: the Insulin Resistance Atherosclerosis Family Study. Diabetes. 2007;56:2135–2141. doi: 10.2337/db06-1017. [DOI] [PubMed] [Google Scholar]
  • 158.Daftary GS, Taylor HS. Endocrine regulation of HOX genes. Endocrine reviews. 2006;27:331–355. doi: 10.1210/er.2005-0018. [DOI] [PubMed] [Google Scholar]
  • 159.Karastergiou K, Fried SK, Xie H, Lee MJ, Divoux A, Rosencrantz MA, Chang RJ, Smith SR. Distinct developmental signatures of human abdominal and gluteal subcutaneous adipose tissue depots. The Journal of clinical endocrinology and metabolism. 2013;98:362–371. doi: 10.1210/jc.2012-2953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Bluher M. Are there still healthy obese patients? Curr Opin Endocrinol Diabetes Obes. 2012;19:341–346. doi: 10.1097/MED.0b013e328357f0a3. [DOI] [PubMed] [Google Scholar]
  • 161.Hube F, Lietz U, Igel M, Jensen PB, Tornqvist H, Joost HG, Hauner H. Difference in leptin mRNA levels between omental and subcutaneous abdominal adipose tissue from obese humans. Horm Metab Res. 1996;28:690–693. doi: 10.1055/s-2007-979879. [DOI] [PubMed] [Google Scholar]
  • 162.Montague CT, Prins JB, Sanders L, Zhang J, Sewter CP, Digby J, Byrne CD, O’Rahilly S. Depot-related gene expression in human subcutaneous and omental adipocytes. Diabetes. 1998;47:1384–1391. doi: 10.2337/diabetes.47.9.1384. [DOI] [PubMed] [Google Scholar]
  • 163.Nielsen NB, Hojbjerre L, Sonne MP, Alibegovic AC, Vaag A, Dela F, Stallknecht B. Interstitial concentrations of adipokines in subcutaneous abdominal and femoral adipose tissue. Regul Pept. 2009;155:39–45. doi: 10.1016/j.regpep.2009.04.010. [DOI] [PubMed] [Google Scholar]
  • 164.Snijder MB, Flyvbjerg A, Stehouwer CD, Frystyk J, Henry RM, Seidell JC, Heine RJ, Dekker JM. Relationship of adiposity with arterial stiffness as mediated by adiponectin in older men and women: the Hoorn Study. Eur J Endocrinol. 2009;160:387–395. doi: 10.1530/EJE-08-0817. [DOI] [PubMed] [Google Scholar]
  • 165.Fisher FM, McTernan PG, Valsamakis G, Chetty R, Harte AL, Anwar AJ, Starcynski J, Crocker J, Barnett AH, McTernan CL, Kumar S. Differences in adiponectin protein expression: effect of fat depots and type 2 diabetic status. Horm Metab Res. 2002;34:650–654. doi: 10.1055/s-2002-38246. [DOI] [PubMed] [Google Scholar]
  • 166.Ryo M, Nakamura T, Kihara S, Kumada M, Shibazaki S, Takahashi M, Nagai M, Matsuzawa Y, Funahashi T. Adiponectin as a biomarker of the metabolic syndrome. Circ J. 2004;68:975–981. doi: 10.1253/circj.68.975. [DOI] [PubMed] [Google Scholar]
  • 167.Vettor R, De Pergola G, Pagano C, Englaro P, Laudadio E, Giorgino F, Blum WF, Giorgino R, Federspil G. Gender differences in serum leptin in obese people: relationships with testosterone, body fat distribution and insulin sensitivity. Eur J Clin Invest. 1997;27:1016–1024. doi: 10.1046/j.1365-2362.1997.2270773.x. [DOI] [PubMed] [Google Scholar]
  • 168.Saad MF, Damani S, Gingerich RL, Riad-Gabriel MG, Khan A, Boyadjian R, Jinagouda SD, el-Tawil K, Rude RK, Kamdar V. Sexual dimorphism in plasma leptin concentration. J Clin Endocrinol Metab. 1997;82:579–584. doi: 10.1210/jcem.82.2.3739. [DOI] [PubMed] [Google Scholar]
  • 169.Arita Y, Kihara S, Ouchi N, Takahashi M, Maeda K, Miyagawa J, Hotta K, Shimomura I, Nakamura T, Miyaoka K, Kuriyama H, Nishida M, Yamashita S, Okubo K, Matsubara K, Muraguchi M, Ohmoto Y, Funahashi T, Matsuzawa Y. Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochem Biophys Res Commun. 1999;257:79–83. doi: 10.1006/bbrc.1999.0255. [DOI] [PubMed] [Google Scholar]
  • 170.Nishizawa H, Shimomura I, Kishida K, Maeda N, Kuriyama H, Nagaretani H, Matsuda M, Kondo H, Furuyama N, Kihara S, Nakamura T, Tochino Y, Funahashi T, Matsuzawa Y. Androgens decrease plasma adiponectin, an insulin-sensitizing adipocyte-derived protein. Diabetes. 2002;51:2734–2741. doi: 10.2337/diabetes.51.9.2734. [DOI] [PubMed] [Google Scholar]
  • 171.Cnop M, Havel PJ, Utzschneider KM, Carr DB, Sinha MK, Boyko EJ, Retzlaff BM, Knopp RH, Brunzell JD, Kahn SE. Relationship of adiponectin to body fat distribution, insulin sensitivity and plasma lipoproteins: evidence for independent roles of age and sex. Diabetologia. 2003;46:459–469. doi: 10.1007/s00125-003-1074-z. [DOI] [PubMed] [Google Scholar]
  • 172.Kloting N, Wilke B, Kloting I. Alleles on rat chromosome 4 (D4Got41-Fabp1/Tacr1) regulate subphenotypes of obesity. Obes Res. 2005;13:589–595. doi: 10.1038/oby.2005.63. [DOI] [PubMed] [Google Scholar]
  • 173.Gavi S, Qurashi S, Stuart LM, Lau R, Melendez MM, Mynarcik DC, McNurlan MA, Gelato MC. Influence of age on the association of retinol-binding protein 4 with metabolic syndrome. Obesity (Silver Spring) 2008;16:893–895. doi: 10.1038/oby.2007.138. [DOI] [PubMed] [Google Scholar]
  • 174.Shimomura I, Funahashi T, Takahashi M, Maeda K, Kotani K, Nakamura T, Yamashita S, Miura M, Fukuda Y, Takemura K, Tokunaga K, Matsuzawa Y. Enhanced expression of PAI-1 in visceral fat: possible contributor to vascular disease in obesity. Nat Med. 1996;2:800–803. doi: 10.1038/nm0796-800. [DOI] [PubMed] [Google Scholar]
  • 175.Lamers D, Famulla S, Wronkowitz N, Hartwig S, Lehr S, Ouwens DM, Eckardt K, Kaufman JM, Ryden M, Muller S, Hanisch FG, Ruige J, Arner P, Sell H, Eckel J. Dipeptidyl peptidase 4 is a novel adipokine potentially linking obesity to the metabolic syndrome. Diabetes. 2011;60:1917–1925. doi: 10.2337/db10-1707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Roh SG, Song SH, Choi KC, Katoh K, Wittamer V, Parmentier M, Sasaki S. Chemerin--a new adipokine that modulates adipogenesis via its own receptor. Biochemical and biophysical research communications. 2007;362:1013–1018. doi: 10.1016/j.bbrc.2007.08.104. [DOI] [PubMed] [Google Scholar]
  • 177.Parolini S, Santoro A, Marcenaro E, Luini W, Massardi L, Facchetti F, Communi D, Parmentier M, Majorana A, Sironi M, Tabellini G, Moretta A, Sozzani S. The role of chemerin in the colocalization of NK and dendritic cell subsets into inflamed tissues. Blood. 2007;109:3625–3632. doi: 10.1182/blood-2006-08-038844. [DOI] [PubMed] [Google Scholar]
  • 178.Barnea G, Strapps W, Herrada G, Berman Y, Ong J, Kloss B, Axel R, Lee KJ. The genetic design of signaling cascades to record receptor activation. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:64–69. doi: 10.1073/pnas.0710487105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Zabel BA, Nakae S, Zuniga L, Kim JY, Ohyama T, Alt C, Pan J, Suto H, Soler D, Allen SJ, Handel TM, Song CH, Galli SJ, Butcher EC. Mast cell-expressed orphan receptor CCRL2 binds chemerin and is required for optimal induction of IgE-mediated passive cutaneous anaphylaxis. The Journal of experimental medicine. 2008;205:2207–2220. doi: 10.1084/jem.20080300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Monnier J, Lewen S, O’Hara E, Huang K, Tu H, Butcher EC, Zabel BA. Expression, regulation, and function of atypical chemerin receptor CCRL2 on endothelial cells. J Immunol. 2012;189:956–967. doi: 10.4049/jimmunol.1102871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Ernst MC, Haidl ID, Zuniga LA, Dranse HJ, Rourke JL, Zabel BA, Butcher EC, Sinal CJ. Disruption of the chemokine-like receptor-1 (CMKLR1) gene is associated with reduced adiposity and glucose intolerance. Endocrinology. 2012;153:672–682. doi: 10.1210/en.2011-1490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Goralski KB, McCarthy TC, Hanniman EA, Zabel BA, Butcher EC, Parlee SD, Muruganandan S, Sinal CJ. Chemerin, a novel adipokine that regulates adipogenesis and adipocyte metabolism. The Journal of biological chemistry. 2007;282:28175–28188. doi: 10.1074/jbc.M700793200. [DOI] [PubMed] [Google Scholar]
  • 183.Albanesi C, Scarponi C, Pallotta S, Daniele R, Bosisio D, Madonna S, Fortugno P, Gonzalvo-Feo S, Franssen JD, Parmentier M, De Pita O, Girolomoni G, Sozzani S. Chemerin expression marks early psoriatic skin lesions and correlates with plasmacytoid dendritic cell recruitment. The Journal of experimental medicine. 2009;206:249–258. doi: 10.1084/jem.20080129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Bauer S, Wanninger J, Schmidhofer S, Weigert J, Neumeier M, Dorn C, Hellerbrand C, Zimara N, Schaffler A, Aslanidis C, Buechler C. Sterol regulatory element-binding protein 2 (SREBP2) activation after excess triglyceride storage induces chemerin in hypertrophic adipocytes. Endocrinology. 2011;152:26–35. doi: 10.1210/en.2010-1157. [DOI] [PubMed] [Google Scholar]
  • 185.Kralisch S, Weise S, Sommer G, Lipfert J, Lossner U, Bluher M, Stumvoll M, Fasshauer M. Interleukin-1beta induces the novel adipokine chemerin in adipocytes in vitro. Regulatory peptides. 2009;154:102–106. doi: 10.1016/j.regpep.2009.02.010. [DOI] [PubMed] [Google Scholar]
  • 186.Hart R, Greaves DR. Chemerin contributes to inflammation by promoting macrophage adhesion to VCAM-1 and fibronectin through clustering of VLA-4 and VLA-5. J Immunol. 2010;185:3728–3739. doi: 10.4049/jimmunol.0902154. [DOI] [PubMed] [Google Scholar]
  • 187.Cash JL, Hart R, Russ A, Dixon JP, Colledge WH, Doran J, Hendrick AG, Carlton MB, Greaves DR. Synthetic chemerin-derived peptides suppress inflammation through ChemR23. The Journal of experimental medicine. 2008;205:767–775. doi: 10.1084/jem.20071601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Jialal I, Devaraj S, Kaur H, Adams-Huet B, Bremer AA. Increased Chemerin and Decreased Omentin-1 in Both Adipose Tissue and Plasma in Nascent Metabolic Syndrome. The Journal of clinical endocrinology and metabolism. 2013 doi: 10.1210/jc.2012-3673. [DOI] [PubMed] [Google Scholar]
  • 189.Shin HY, Lee DC, Chu SH, Jeon JY, Lee MK, Im JA, Lee JW. Chemerin levels are positively correlated with abdominal visceral fat accumulation. Clinical endocrinology. 2012;77:47–50. doi: 10.1111/j.1365-2265.2011.04217.x. [DOI] [PubMed] [Google Scholar]
  • 190.Luque-Ramirez M, Martinez-Garcia MA, Montes-Nieto R, Fernandez-Duran E, Insenser M, Alpanes M, Escobar-Morreale HF. Sexual dimorphism in adipose tissue function as evidenced by circulating adipokine concentrations in the fasting state and after an oral glucose challenge. Hum Reprod. 2013 doi: 10.1093/humrep/det097. [DOI] [PubMed] [Google Scholar]
  • 191.Martinez-Garcia MA, Montes-Nieto R, Fernandez-Duran E, Insenser M, Luque-Ramirez M, Escobar-Morreale HF. Evidence for Masculinization of Adipokine Gene Expression in Visceral and Subcutaneous Adipose Tissue of Obese Women With Polycystic Ovary Syndrome (PCOS) The Journal of clinical endocrinology and metabolism. 2013;98:E388–E396. doi: 10.1210/jc.2012-3414. [DOI] [PubMed] [Google Scholar]
  • 192.Alfadda AA, Sallam RM, Chishti MA, Moustafa AS, Fatma S, Alomaim WS, Al-Naami MY, Bassas AF, Chrousos GP, Jo H. Differential patterns of serum concentration and adipose tissue expression of chemerin in obesity: adipose depot specificity and gender dimorphism. Molecules and cells. 2012;33:591–596. doi: 10.1007/s10059-012-0012-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Ouchi N, Parker JL, Lugus JJ, 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]
  • 194.Moreno-Navarrete JM, Manco M, Ibanez J, Garcia-Fuentes E, Ortega F, Gorostiaga E, Vendrell J, Izquierdo M, Martinez C, Nolfe G, Ricart W, Mingrone G, Tinahones F, Fernandez-Real JM. Metabolic endotoxemia and saturated fat contribute to circulating NGAL concentrations in subjects with insulin resistance. Int J Obes (Lond) 2010;34:240–249. doi: 10.1038/ijo.2009.242. [DOI] [PubMed] [Google Scholar]
  • 195.Diamanti-Kandarakis E, Livadas S, Kandarakis SA, Margeli A, Papassotiriou I. Serum concentrations of atherogenic proteins neutrophil gelatinase-associated lipocalin and its complex with matrix metalloproteinase-9 are significantly lower in women with polycystic ovary syndrome: hint of a protective mechanism? European journal of endocrinology / European Federation of Endocrine Societies. 2008;158:525–531. doi: 10.1530/EJE-07-0822. [DOI] [PubMed] [Google Scholar]
  • 196.Koiou E, Tziomalos K, Katsikis I, Kandaraki EA, Kalaitzakis E, Delkos D, Vosnakis C, Panidis D. Weight loss significantly reduces serum lipocalin-2 levels in overweight and obese women with polycystic ovary syndrome. Gynecological endocrinology : the official journal of the International Society of Gynecological Endocrinology. 2012;28:20–24. doi: 10.3109/09513590.2011.588745. [DOI] [PubMed] [Google Scholar]
  • 197.Cakal E, Ozkaya M, Engin-Ustun Y, Ustun Y. Serum lipocalin-2 as an insulin resistance marker in patients with polycystic ovary syndrome. Journal of endocrinological investigation. 2011;34:97–100. doi: 10.1007/BF03347037. [DOI] [PubMed] [Google Scholar]
  • 198.Law IK, Xu A, Lam KS, Berger T, Mak TW, Vanhoutte PM, Liu JT, Sweeney G, Zhou M, Yang B, Wang Y. Lipocalin-2 deficiency attenuates insulin resistance associated with aging and obesity. Diabetes. 2010;59:872–882. doi: 10.2337/db09-1541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Guo H, Jin D, Zhang Y, Wright W, Bazuine M, Brockman DA, Bernlohr DA, Chen X. Lipocalin-2 deficiency impairs thermogenesis and potentiates diet-induced insulin resistance in mice. Diabetes. 2010;59:1376–1385. doi: 10.2337/db09-1735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Yan QW, Yang Q, Mody N, Graham TE, Hsu CH, Xu Z, Houstis NE, Kahn BB, Rosen ED. The adipokine lipocalin 2 is regulated by obesity and promotes insulin resistance. Diabetes. 2007;56:2533–2540. doi: 10.2337/db07-0007. [DOI] [PubMed] [Google Scholar]
  • 201.Zhang J, Wu Y, Zhang Y, Leroith D, Bernlohr DA, Chen X. The role of lipocalin 2 in the regulation of inflammation in adipocytes and macrophages. Mol Endocrinol. 2008;22:1416–1426. doi: 10.1210/me.2007-0420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Cowland JB, Muta T, Borregaard N. IL-1beta-specific up-regulation of neutrophil gelatinase-associated lipocalin is controlled by IkappaB-zeta. J Immunol. 2006;176:5559–5566. doi: 10.4049/jimmunol.176.9.5559. [DOI] [PubMed] [Google Scholar]
  • 203.Wang Y, Lam KS, Kraegen EW, Sweeney G, Zhang J, Tso AW, Chow WS, Wat NM, Xu JY, Hoo RL, Xu A. Lipocalin-2 is an inflammatory marker closely associated with obesity, insulin resistance, and hyperglycemia in humans. Clinical chemistry. 2007;53:34–41. doi: 10.1373/clinchem.2006.075614. [DOI] [PubMed] [Google Scholar]
  • 204.Catalan V, Gomez-Ambrosi J, Rodriguez A, Ramirez B, Silva C, Rotellar F, Gil MJ, Cienfuegos JA, Salvador J, Fruhbeck G. Increased adipose tissue expression of lipocalin-2 in obesity is related to inflammation and matrix metalloproteinase-2 and metalloproteinase-9 activities in humans. J Mol Med (Berl) 2009;87:803–813. doi: 10.1007/s00109-009-0486-8. [DOI] [PubMed] [Google Scholar]
  • 205.Gesta S, Bluher M, Yamamoto Y, Norris AW, Berndt J, Kralisch S, Boucher J, Lewis C, Kahn CR. Evidence for a role of developmental genes in the origin of obesity and body fat distribution. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:6676–6681. doi: 10.1073/pnas.0601752103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Ussar S, Bezy O, Bluher M, Kahn CR. Glypican-4 enhances insulin signaling via interaction with the insulin receptor and serves as a novel adipokine. Diabetes. 2012;61:2289–2298. doi: 10.2337/db11-1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Yang RZ, Lee MJ, Hu H, Pray J, Wu HB, Hansen BC, Shuldiner AR, Fried SK, McLenithan JC, Gong DW. Identification of omentin as a novel depot-specific adipokine in human adipose tissue: possible role in modulating insulin action. American journal of physiology. Endocrinology and metabolism. 2006;290:E1253–E1261. doi: 10.1152/ajpendo.00572.2004. [DOI] [PubMed] [Google Scholar]
  • 208.de Souza Batista CM, Yang RZ, Lee MJ, Glynn NM, Yu DZ, Pray J, Ndubuizu K, Patil S, Schwartz A, Kligman M, Fried SK, Gong DW, Shuldiner AR, Pollin TI, McLenithan JC. Omentin plasma levels and gene expression are decreased in obesity. Diabetes. 2007;56:1655–1661. doi: 10.2337/db06-1506. [DOI] [PubMed] [Google Scholar]
  • 209.Shibata R, Ouchi N, Takahashi R, Terakura Y, Ohashi K, Ikeda N, Higuchi A, Terasaki H, Kihara S, Murohara T. Omentin as a novel biomarker of metabolic risk factors. Diabetology & metabolic syndrome. 2012;4:37. doi: 10.1186/1758-5996-4-37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Tan BK, Adya R, Randeva HS. Omentin: a novel link between inflammation, diabesity, and cardiovascular disease. Trends in cardiovascular medicine. 2010;20:143–148. doi: 10.1016/j.tcm.2010.12.002. [DOI] [PubMed] [Google Scholar]
  • 211.Barth S, Klein P, Horbach T, Dotsch J, Rauh M, Rascher W, Knerr I. Expression of neuropeptide Y, omentin and visfatin in visceral and subcutaneous adipose tissues in humans:relation to endocrine and clinical parameters. Obesity facts. 2010;3:245–251. doi: 10.1159/000319508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Zhong X, Li X, Liu F, Tan H, Shang D. Omentin inhibits TNF-alpha-induced expression of adhesion molecules in endothelial cells via ERK/NF-kappaB pathway. Biochemical and biophysical research communications. 2012;425:401–406. doi: 10.1016/j.bbrc.2012.07.110. [DOI] [PubMed] [Google Scholar]
  • 213.Kazama K, Usui T, Okada M, Hara Y, Yamawaki H. Omentin plays an anti-inflammatory role through inhibition of TNF-alpha-induced superoxide production in vascular smooth muscle cells. European journal of pharmacology. 2012;686:116–123. doi: 10.1016/j.ejphar.2012.04.033. [DOI] [PubMed] [Google Scholar]
  • 214.Kawano Y, Kypta R. Secreted antagonists of the Wnt signalling pathway. J Cell Sci. 2003;116:2627–2634. doi: 10.1242/jcs.00623. [DOI] [PubMed] [Google Scholar]
  • 215.Christodoulides C, Lagathu C, Sethi JK, Vidal-Puig A. Adipogenesis and WNT signalling. Trends Endocrinol Metab. 2009;20:16–24. doi: 10.1016/j.tem.2008.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Lagathu C, Christodoulides C, Tan CY, Virtue S, Laudes M, Campbell M, Ishikawa K, Ortega F, Tinahones FJ, Fernandez-Real JM, Oresic M, Sethi JK, Vidal-Puig A. Secreted frizzled-related protein 1 regulates adipose tissue expansion and is dysregulated in severe obesity. Int J Obes (Lond) 2010;34:1695–1705. doi: 10.1038/ijo.2010.107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Park JR, Jung JW, Lee YS, Kang KS. The roles of Wnt antagonists Dkk1 and sFRP4 during adipogenesis of human adipose tissue-derived mesenchymal stem cells. Cell proliferation. 2008;41:859–874. doi: 10.1111/j.1365-2184.2008.00565.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Ehrlund A, Mejhert N, Lorente-Cebrian S, Astrom G, Dahlman I, Laurencikiene J, Ryden M. Characterization of the Wnt Inhibitors Secreted Frizzled-Related Proteins (SFRPs) in Human Adipose Tissue. The Journal of clinical endocrinology and metabolism. 2013 doi: 10.1210/jc.2012-3416. [DOI] [PubMed] [Google Scholar]
  • 219.Ouchi N, Higuchi A, Ohashi K, Oshima Y, Gokce N, Shibata R, Akasaki Y, Shimono A, Walsh K. Sfrp5 is an anti-inflammatory adipokine that modulates metabolic dysfunction in obesity. Science. 2010;329:454–457. doi: 10.1126/science.1188280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Hu W, Li L, Yang M, Luo X, Ran W, Liu D, Xiong Z, Liu H, Yang G. Circulating Sfrp5 is a signature of obesity-related metabolic disorders and is regulated by glucose and liraglutide in humans. The Journal of clinical endocrinology and metabolism. 2013;98:290–298. doi: 10.1210/jc.2012-2466. [DOI] [PubMed] [Google Scholar]
  • 221.Koza RA, Nikonova L, Hogan J, Rim JS, Mendoza T, Faulk C, Skaf J, Kozak LP. Changes in gene expression foreshadow diet-induced obesity in genetically identical mice. PLoS genetics. 2006;2:e81. doi: 10.1371/journal.pgen.0020081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Okada Y, Sakaue H, Nagare T, Kasuga M. Diet-induced up-regulation of gene expression in adipocytes without changes in DNA methylation. The Kobe journal of medical sciences. 2009;54:E241–E249. [PubMed] [Google Scholar]
  • 223.Kloting N, Berndt J, Kralisch S, Kovacs P, Fasshauer M, Schon MR, Stumvoll M, Bluher M. Vaspin gene expression in human adipose tissue: association with obesity and type 2 diabetes. Biochemical and biophysical research communications. 2006;339:430–436. doi: 10.1016/j.bbrc.2005.11.039. [DOI] [PubMed] [Google Scholar]
  • 224.Hida K, Wada J, Eguchi J, Zhang H, Baba M, Seida A, Hashimoto I, Okada T, Yasuhara A, Nakatsuka A, Shikata K, Hourai S, Futami J, Watanabe E, Matsuki Y, Hiramatsu R, Akagi S, Makino H, Kanwar YS. Visceral adipose tissue-derived serine protease inhibitor: a unique insulin-sensitizing adipocytokine in obesity. Proc Natl Acad Sci U S A. 2005;102:10610–10615. doi: 10.1073/pnas.0504703102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Youn BS, Kloting N, Kratzsch J, Lee N, Park JW, Song ES, Ruschke K, Oberbach A, Fasshauer M, Stumvoll M, Bluher M. Serum vaspin concentrations in human obesity and type 2 diabetes. Diabetes. 2008;57:372–377. doi: 10.2337/db07-1045. [DOI] [PubMed] [Google Scholar]
  • 226.Seeger J, Ziegelmeier M, Bachmann A, Lossner U, Kratzsch J, Bluher M, Stumvoll M, Fasshauer M. Serum levels of the adipokine vaspin in relation to metabolic and renal parameters. The Journal of clinical endocrinology and metabolism. 2008;93:247–251. doi: 10.1210/jc.2007-1853. [DOI] [PubMed] [Google Scholar]
  • 227.Piya MK, McTernan PG, Kumar S. Adipokine inflammation and insulin resistance: the role of glucose, lipids and endotoxin. J Endocrinol. 2013;216:T1–T15. doi: 10.1530/JOE-12-0498. [DOI] [PubMed] [Google Scholar]
  • 228.Nielsen S, Guo Z, Albu JB, Klein S, O’Brien PC, Jensen MD. Energy expenditure, sex, and endogenous fuel availability in humans. The Journal of clinical investigation. 2003;111:981–988. doi: 10.1172/JCI16253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Koutsari C, Basu R, Rizza RA, Nair KS, Khosla S, Jensen MD. Nonoxidative free fatty acid disposal is greater in young women than men. The Journal of clinical endocrinology and metabolism. 2011;96:541–547. doi: 10.1210/jc.2010-1651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Karpe F, Dickmann JR, Frayn KN. Fatty acids, obesity, and insulin resistance: time for a reevaluation. Diabetes. 2011;60:2441–2449. doi: 10.2337/db11-0425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Wahrenberg H, Lonnqvist F, Arner P. Mechanisms underlying regional differences in lipolysis in human adipose tissue. J Clin Invest. 1989;84:458–467. doi: 10.1172/JCI114187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Fried SK, Leibel RL, Edens NK, Kral JG. Lipolysis in intraabdominal adipose tissues of obese women and men. Obes Res. 1993;1:443–448. doi: 10.1002/j.1550-8528.1993.tb00026.x. [DOI] [PubMed] [Google Scholar]
  • 233.Rebuffe-Scrive M, Anderson B, Olbe L, Bjorntorp P. Metabolism of adipose tissue in intraabdominal depots in severely obese men and women. Metabolism. 1990;39:1021–1025. doi: 10.1016/0026-0495(90)90160-e. [DOI] [PubMed] [Google Scholar]
  • 234.Tchernof A, Belanger C, Morisset AS, Richard C, Mailloux J, Laberge P, Dupont P. Regional differences in adipose tissue metabolism in women: minor effect of obesity and body fat distribution. Diabetes. 2006;55:1353–1360. doi: 10.2337/db05-1439. [DOI] [PubMed] [Google Scholar]
  • 235.Boivin A, Brochu G, Marceau S, Marceau P, Hould FS, Tchernof A. Regional differences in adipose tissue metabolism in obese men. Metabolism. 2007;56:533–540. doi: 10.1016/j.metabol.2006.11.015. [DOI] [PubMed] [Google Scholar]
  • 236.Horowitz JF, Klein S. Whole body and abdominal lipolytic sensitivity to epinephrine is suppressed in upper body obese women. Am J Physiol Endocrinol Metab. 2000;278:E1144–E1152. doi: 10.1152/ajpendo.2000.278.6.E1144. [DOI] [PubMed] [Google Scholar]
  • 237.Guo Z, Johnson CM, Jensen MD. Regional lipolytic responses to isoproterenol in women. Am J Physiol. 1997;273:E108–E112. doi: 10.1152/ajpendo.1997.273.1.E108. [DOI] [PubMed] [Google Scholar]
  • 238.Jensen MD, Cryer PE, Johnson CM, Murray MJ. Effects of epinephrine on regional free fatty acid and energy metabolism in men and women. The American journal of physiology. 1996;270:E259–E264. doi: 10.1152/ajpendo.1996.270.2.E259. [DOI] [PubMed] [Google Scholar]
  • 239.Arner P, Kriegholm E, Engfeldt P, Bolinder J. Adrenergic regulation of lipolysis in situ at rest and during exercise. The Journal of clinical investigation. 1990;85:893–898. doi: 10.1172/JCI114516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Gjedsted J, Gormsen LC, Nielsen S, Schmitz O, Djurhuus CB, Keiding S, Orskov H, Tonnesen E, Moller N. Effects of a 3-day fast on regional lipid and glucose metabolism in human skeletal muscle and adipose tissue. Acta Physiol (Oxf) 2007;191:205–216. doi: 10.1111/j.1748-1716.2007.01740.x. [DOI] [PubMed] [Google Scholar]
  • 241.Martin ML, Jensen MD. Effects of body fat distribution on regional lipolysis in obesity. J Clin Invest. 1991;88:609–613. doi: 10.1172/JCI115345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Gavin KM, Cooper EE, Raymer DK, Hickner RC. Estradiol Effects on Subcutaneous Adipose Tissue Lipolysis in Premenopausal Women are Adipose Tissue Depot Specific and Treatment Dependent, American journal of physiology. Endocrinology and metabolism. 2013 doi: 10.1152/ajpendo.00023.2013. [DOI] [PubMed] [Google Scholar]
  • 243.Pedersen SB, Kristensen K, Hermann PA, Katzenellenbogen JA, Richelsen B. Estrogen controls lipolysis by up-regulating alpha2A–adrenergic receptors directly in human adipose tissue through the estrogen receptor alpha. Implications for the female fat distribution. The Journal of clinical endocrinology and metabolism. 2004;89:1869–1878. doi: 10.1210/jc.2003-031327. [DOI] [PubMed] [Google Scholar]
  • 244.Dicker A, Ryden M, Naslund E, Muehlen IE, Wiren M, Lafontan M, Arner P. Effect of testosterone on lipolysis in human pre-adipocytes from different fat depots. Diabetologia. 2004;47:420–428. doi: 10.1007/s00125-003-1324-0. [DOI] [PubMed] [Google Scholar]
  • 245.Arner P. Differences in lipolysis between human subcutaneous and omental adipose tissues. Ann Med. 1995;27:435–438. doi: 10.3109/07853899709002451. [DOI] [PubMed] [Google Scholar]
  • 246.Tan GD, Goossens GH, Humphreys SM, Vidal H, Karpe F. Upper and lower body adipose tissue function: a direct comparison of fat mobilization in humans. Obes Res. 2004;12:114–118. doi: 10.1038/oby.2004.15. [DOI] [PubMed] [Google Scholar]
  • 247.Jensen MD, Haymond MW, Rizza RA, Cryer PE, Miles JM. Influence of body fat distribution on free fatty acid metabolism in obesity. J Clin Invest. 1989;83:1168–1173. doi: 10.1172/JCI113997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Jensen MD. Gender differences in regional fatty acid metabolism before and after meal ingestion. J Clin Invest. 1995;96:2297–2303. doi: 10.1172/JCI118285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Dowling HJ, Fried SK, Pi-Sunyer FX. Insulin resistance in adipocytes of obese women: effects of body fat distribution and race. Metabolism. 1995;44:987–995. doi: 10.1016/0026-0495(95)90094-2. [DOI] [PubMed] [Google Scholar]
  • 250.Meek SE, Nair KS, Jensen MD. Insulin regulation of regional free fatty acid metabolism. Diabetes. 1999;48:10–14. doi: 10.2337/diabetes.48.1.10. [DOI] [PubMed] [Google Scholar]
  • 251.Roust LR, Jensen MD. Postprandial free fatty acid kinetics are abnormal in upper body obesity. Diabetes. 1993;42:1567–1573. doi: 10.2337/diab.42.11.1567. [DOI] [PubMed] [Google Scholar]
  • 252.Leibel RL, Edens NK, Fried SK. Physiologic basis for the control of body fat distribution in humans. Annu Rev Nutr. 1989;9:417–443. doi: 10.1146/annurev.nu.09.070189.002221. [DOI] [PubMed] [Google Scholar]
  • 253.Arner P, Hellstrom L, Wahrenberg H, Bronnegard M. Beta-adrenoceptor expression in human fat cells from different regions. J Clin Invest. 1990;86:1595–1600. doi: 10.1172/JCI114880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Lonnqvist F, Thorne A, Large V, Arner P. Sex differences in visceral fat lipolysis and metabolic complications of obesity. Arterioscler Thromb Vasc Biol. 1997;17:1472–1480. doi: 10.1161/01.atv.17.7.1472. [DOI] [PubMed] [Google Scholar]
  • 255.Soeters MR, Sauerwein HP, Groener JE, Aerts JM, Ackermans MT, Glatz JF, Fliers E, Serlie MJ. Gender-related differences in the metabolic response to fasting. The Journal of clinical endocrinology and metabolism. 2007;92:3646–3652. doi: 10.1210/jc.2007-0552. [DOI] [PubMed] [Google Scholar]
  • 256.Horowitz JF, Coppack SW, Paramore D, Cryer PE, Zhao G, Klein S. Effect of short-term fasting on lipid kinetics in lean and obese women. The American journal of physiology. 1999;276:E278–E284. doi: 10.1152/ajpendo.1999.276.2.E278. [DOI] [PubMed] [Google Scholar]
  • 257.Nielsen S, Guo Z, Johnson CM, Hensrud DD, Jensen MD. Splanchnic lipolysis in human obesity. J Clin Invest. 2004;113:1582–1588. doi: 10.1172/JCI21047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Boden G, Chen X. Effects of fat on glucose uptake and utilization in patients with non-insulin-dependent diabetes. The Journal of clinical investigation. 1995;96:1261–1268. doi: 10.1172/JCI118160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Roden M, Price TB, Perseghin G, Petersen KF, Rothman DL, Cline GW, Shulman GI. Mechanism of free fatty acid-induced insulin resistance in humans. The Journal of clinical investigation. 1996;97:2859–2865. doi: 10.1172/JCI118742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Saloranta C, Franssila-Kallunki A, Ekstrand A, Taskinen MR, Groop L. Modulation of hepatic glucose production by non-esterified fatty acids in type 2 (non-insulin-dependent) diabetes mellitus. Diabetologia. 1991;34:409–415. doi: 10.1007/BF00403179. [DOI] [PubMed] [Google Scholar]
  • 261.Rebrin K, Steil GM, Getty L, Bergman RN. Free fatty acid as a link in the regulation of hepatic glucose output by peripheral insulin. Diabetes. 1995;44:1038–1045. doi: 10.2337/diab.44.9.1038. [DOI] [PubMed] [Google Scholar]
  • 262.Havel RJ, Kane JP, Balasse EO, Segel N, Basso LV. Splanchnic metabolism of free fatty acids and production of triglycerides of very low density lipoproteins in normotriglyceridemic and hypertriglyceridemic humans. The Journal of clinical investigation. 1970;49:2017–2035. doi: 10.1172/JCI106422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Kissebah AH, Adams PW, Wynn V. Plasma free fatty acid and triglyceride transport kinetics in man. Clinical science and molecular medicine. 1974;47:259–278. doi: 10.1042/cs0470259. [DOI] [PubMed] [Google Scholar]
  • 264.Kissebah AH, Alfarsi S, Adams PW, Wynn V. Role of insulin resistance in adipose tissue and liver in the pathogenesis of endogenous hypertriglyceridaemia in man. Diabetologia. 1976;12:563–571. doi: 10.1007/BF01220632. [DOI] [PubMed] [Google Scholar]
  • 265.Arner P, Lithell H, Wahrenberg H, Bronnegard M. Expression of lipoprotein lipase in different human subcutaneous adipose tissue regions. J Lipid Res. 1991;32:423–429. [PubMed] [Google Scholar]
  • 266.Pouliot MC, Despres JP, Moorjani S, Lupien PJ, Tremblay A, Nadeau A, Bouchard C. Regional variation in adipose tissue lipoprotein lipase activity: association with plasma high density lipoprotein levels. Eur J Clin Invest. 1991;21:398–405. doi: 10.1111/j.1365-2362.1991.tb01387.x. [DOI] [PubMed] [Google Scholar]
  • 267.Fried SK, Kral JG. Sex differences in regional distribution of fat cell size and lipoprotein lipase activity in morbidly obese patients. Int J Obes. 1987;11:129–140. [PubMed] [Google Scholar]
  • 268.Ramirez ME, McMurry MP, Wiebke GA, Felten KJ, Ren K, Meikle AW, Iverius PH. Evidence for sex steroid inhibition of lipoprotein lipase in men: comparison of abdominal and femoral adipose tissue. Metabolism. 1997;46:179–185. doi: 10.1016/s0026-0495(97)90299-7. [DOI] [PubMed] [Google Scholar]
  • 269.Marin P, Oden B, Bjorntorp P. Assimilation and mobilization of triglycerides in subcutaneous abdominal and femoral adipose tissue in vivo in men: effects of androgens. J Clin Endocrinol Metab. 1995;80:239–243. doi: 10.1210/jcem.80.1.7829619. [DOI] [PubMed] [Google Scholar]
  • 270.Price TM, O’Brien SN, Welter BH, George R, Anandjiwala J, Kilgore M. Estrogen regulation of adipose tissue lipoprotein lipase--possible mechanism of body fat distribution. American journal of obstetrics and gynecology. 1998;178:101–107. doi: 10.1016/s0002-9378(98)70634-9. [DOI] [PubMed] [Google Scholar]
  • 271.Blouin K, Nadeau M, Perreault M, Veilleux A, Drolet R, Marceau P, Mailloux J, Luu-The V, Tchernof A. Effects of androgens on adipocyte differentiation and adipose tissue explant metabolism in men and women. Clinical endocrinology. 2010;72:176–188. doi: 10.1111/j.1365-2265.2009.03645.x. [DOI] [PubMed] [Google Scholar]
  • 272.Doolittle MH, Ben-Zeev O, Elovson J, Martin D, Kirchgessner TG. The response of lipoprotein lipase to feeding and fasting. Evidence for posttranslational regulation. J Biol Chem. 1990;265:4570–4577. [PubMed] [Google Scholar]
  • 273.Romanski SA, Nelson RM, Jensen MD. Meal fatty acid uptake in adipose tissue: gender effects in nonobese humans. Am J Physiol Endocrinol Metab. 2000;279:E455–E462. doi: 10.1152/ajpendo.2000.279.2.E455. [DOI] [PubMed] [Google Scholar]
  • 274.Koutsari C, Snozek CL, Jensen MD. Plasma NEFA storage in adipose tissue in the postprandial state: sex-related and regional differences. Diabetologia. 2008;51:2041–2048. doi: 10.1007/s00125-008-1126-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Votruba SB, Jensen MD. Sex-specific differences in leg fat uptake are revealed with a high-fat meal. Am J Physiol Endocrinol Metab. 2006;291:E1115–E1123. doi: 10.1152/ajpendo.00196.2006. [DOI] [PubMed] [Google Scholar]
  • 276.Santosa S, Hensrud DD, Votruba SB, Jensen MD. The influence of sex and obesity phenotype on meal fatty acid metabolism before and after weight loss. Am J Clin Nutr. 2008;88:1134–1141. doi: 10.1093/ajcn/88.4.1134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Nguyen TT, Hernandez Mijares A, Johnson CM, Jensen MD. Postprandial leg and splanchnic fatty acid metabolism in nonobese men and women. The American journal of physiology. 1996;271:E965–E972. doi: 10.1152/ajpendo.1996.271.6.E965. [DOI] [PubMed] [Google Scholar]
  • 278.Jensen MD, Sarr MG, Dumesic DA, Southorn PA, Levine JA. Regional uptake of meal fatty acids in humans. Am J Physiol Endocrinol Metab. 2003;285:E1282–E1288. doi: 10.1152/ajpendo.00220.2003. [DOI] [PubMed] [Google Scholar]
  • 279.Shadid S, Koutsari C, Jensen MD. Direct free fatty acid uptake into human adipocytes in vivo: relation to body fat distribution. Diabetes. 2007;56:1369–1375. doi: 10.2337/db06-1680. [DOI] [PubMed] [Google Scholar]
  • 280.Koutsari C, Ali AH, Mundi MS, Jensen MD. Storage of circulating free fatty acid in adipose tissue of postabsorptive humans: quantitative measures and implications for body fat distribution. Diabetes. 2011;60:2032–2040. doi: 10.2337/db11-0154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Koutsari C, Mundi MS, Ali AH, Jensen MD. Storage rates of circulating free fatty acid into adipose tissue during eating or walking in humans. Diabetes. 2012;61:329–338. doi: 10.2337/db11-0748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Frayn KN. Adipose tissue as a buffer for daily lipid flux. Diabetologia. 2002;45:1201–1210. doi: 10.1007/s00125-002-0873-y. [DOI] [PubMed] [Google Scholar]
  • 283.Hernandez TL, Kittelson JM, Law CK, Ketch LL, Stob NR, Lindstrom RC, Scherzinger A, Stamm ER, Eckel RH. Fat redistribution following suction lipectomy: defense of body fat and patterns of restoration. Obesity (Silver Spring) 2011;19:1388–1395. doi: 10.1038/oby.2011.64. [DOI] [PubMed] [Google Scholar]

RESOURCES