Vous êtes sur la page 1sur 10

Short Article

The Adipose Tissue Microenvironment Regulates


Depot-Specific Adipogenesis in Obesity
Graphical Abstract Authors
Elise Jeffery, Allison Wing,
Brandon Holtrup, ..., Laura Colman,
Ryan Berry, Matthew S. Rodeheffer

Correspondence
matthew.rodeheffer@yale.edu

In Brief
Jeffery et al. look at differences in fat
depot expansion in male and female mice
in response to a high-fat diet.
Subcutaneous white adipose tissue
(SWAT) obesogenic adipogenesis occurs
only in females. Furthermore depot-
specific activation and differentiation of
white adipocyte precursors are
determined by the microenvironment
rather than cell-intrinsic differences.

Highlights
d High-fat diet feeding induces sex-specific patterns of
adipogenesis

d The amount of obesogenic adipogenesis positively correlates


with fat pad weight

d Estrogen potentiates obesogenic adipogenesis in the


inguinal subcutaneous depot

d Depot-specific adipogenesis is regulated by the adipose


tissue microenvironment

Jeffery et al., 2016, Cell Metabolism 24, 142150


July 12, 2016 2016 Elsevier Inc.
http://dx.doi.org/10.1016/j.cmet.2016.05.012
Cell Metabolism

Short Article

The Adipose Tissue Microenvironment Regulates


Depot-Specific Adipogenesis in Obesity
Elise Jeffery,1 Allison Wing,2 Brandon Holtrup,2 Zachary Sebo,2 Jennifer L. Kaplan,3 Rocio Saavedra-Pena,2
Christopher D. Church,3 Laura Colman,3 Ryan Berry,2 and Matthew S. Rodeheffer2,3,4,*
1Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA
2Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
3Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
4Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT 06520, USA

*Correspondence: matthew.rodeheffer@yale.edu
http://dx.doi.org/10.1016/j.cmet.2016.05.012

SUMMARY cific WAT growth are directly relevant to metabolic health and
disease, yet the regulation of this process in vivo is not well
The sexually dimorphic distribution of adipose tissue understood.
influences the development of obesity-associated Subcutaneous and visceral adipose depots display an array of
pathologies. The accumulation of visceral white adi- differences including gene expression (Cohen et al., 2014; Grove
pose tissue (VWAT) that occurs in males is detri- et al., 2010; Karastergiou et al., 2013), developmental lineage
mental to metabolic health, while accumulation of (Chau et al., 2014; Krueger et al., 2014; Sanchez-Gurmaches
and Guertin, 2014), metabolic characteristics (Cohen et al.,
subcutaneous adipose tissue (SWAT) seen in females
2014; Karastergiou and Fried, 2013; Tran et al., 2008), and adipo-
may be protective. Here, we show that adipocyte
kine secretion profiles (Shi et al., 2009). Interestingly, males pref-
hyperplasia contributes directly to the differential fat erentially accumulate visceral adipose tissue, while premeno-
distribution between the sexes. In male mice, high- pausal females accumulate more subcutaneous adipose tissue
fat diet (HFD) induces adipogenesis specifically in (Palmer and Clegg, 2015). In humans, these patterns of adipose
VWAT, while in females HFD induces adipogenesis distribution result in characteristically female pear-shaped
in both VWAT and SWAT in a sex hormone-depen- obesity and male apple-shaped obesity (Gesta et al., 2007;
dent manner. We also show that the activation of Karastergiou et al., 2012). In postmenopausal women, adipose
adipocyte precursors (APs), which drives adipocyte distribution shifts toward a male-like pattern, implicating sex
hyperplasia in obesity, is regulated by the adipose hormones in maintaining a balance between subcutaneous
depot microenvironment and not by cell-intrinsic and visceral depot mass (Palmer and Clegg, 2015). Importantly,
however, the cellular and molecular mechanisms underlying dif-
mechanisms. These findings indicate that APs are
ferential adipose distribution in males and females are unclear.
plastic cells, which respond to both local and
Adipose tissue growth can occur through two distinct mecha-
systemic signals that influence their differentiation nisms: the increase in size of existing adipocytes (hypertrophy)
potential independent of depot origin. Therefore, or the increase in number of adipocytes (hyperplasia). Since
depot-specific AP niches coordinate adipose tissue mature adipocytes are post-mitotic, new adipocytes are derived
growth and distribution. from adipocyte precursor (AP) cells within the stroma of all white
adipose depots (Berry et al., 2014). It was recently shown that the
expansion of visceral adipose tissue in males occurs through
INTRODUCTION adipocyte hyperplasia in both mice (Jeffery et al., 2015; Wang
et al., 2013) and humans (Arner et al., 2013), while male subcu-
The increasing rates of obesity and obesity-associated pathol- taneous adipose tissue in mice does not exhibit significant
ogies around the world highlight the need for an understanding hyperplasia in response to obesogenic stimuli (Jeffery et al.,
of the mechanisms that underlie white adipose tissue (WAT) 2015; Wang et al., 2013). Furthermore, studies of the dynamics
growth in vivo. The distribution of WAT into subcutaneous of human adipose tissue indicate that weight loss is mediated
(SWAT) and visceral (VWAT) depots in obesity has links to primarily by a reduction in adipocyte size with no significant
metabolic disease. Increased VWAT mass is strongly associated reduction in adipocyte number, even after bariatric surgery
with cardiometabolic risk and mortality across ethnicities (Cou- (Spalding et al., 2008). These data suggest that an increase in
tinho et al., 2011; Lee et al., 2014; Nazare et al., 2012; Phillips adipocyte number in obesity may have lasting effects on energy
and Prins, 2008; Pischon et al., 2008; Wormser et al., 2011). In homeostasis and weight maintenance. Taken together, these
contrast, elevated SWAT mass has been associated with im- findings indicate that depot-specific mechanisms are important
provements in plasma lipid profile, insulin sensitivity, blood pres- in the control of WAT growth in obesity; however, the mecha-
sure, and atherosclerosis (Appleton et al., 2013; Heitmann and nisms driving these processes are not known.
Lissner, 2011; Manolopoulos et al., 2010; Snijder et al., 2003). We recently found that in male mice, obesogenic signals such
These data suggest that the mechanisms underlying depot-spe- as high-fat feeding or hyperphagia lead to the proliferation and

142 Cell Metabolism 24, 142150, July 12, 2016 2016 Elsevier Inc.
Figure 1. Adipocyte Hyperplasia Contributes to Depot- and Sex-Specific WAT Growth in Obesity
(A) Body fat as a percentage of total body weight after 8 weeks of SD or HFD feeding in male and female C57Bl/6J Adiponectin-creER; mTmG mice as measured
by magnetic resonance imaging (n = 915).
(B) Weight of individual fat pads from male or female C57Bl/6J Adiponectin-creER; mTmG mice after 8 weeks of SD or HFD feeding (n = 915).
(C) Experimental design for Adiponectin-creER; mTmG adipogenesis experiments.

(legend continued on next page)

Cell Metabolism 24, 142150, July 12, 2016 143


differentiation of APs in VWAT, but not SWAT (Jeffery et al., remain mGFP+ (mG) (Figures 1C, S1A, and S1B) (Jeffery et al.,
2015). The preferential growth of VWAT in male mice can thus 2014, 2015). Notably, our data indicate that the low dose of
be partially attributed to increased hyperplasia; however, the 50 mg/kg tamoxifen does not significantly affect body weight
mechanism(s) underlying depot-specific AP activation remain (Figure S1C). Consistent with previous findings, we observe
unclear. One potential explanation for this phenomenon is that VWAT-specific adipogenesis in males after 8 weeks of HFD
APs in different depots have distinct intrinsic abilities to respond feeding (Figure 1D). Interestingly, in females we find that HFD
to obesogenic signals. Indeed, previous reports have suggested significantly increases adipogenesis in both VWAT and SWAT
that AP-intrinsic mechanisms are responsible for depot-specific depots (Figure 1E), mirroring the similar expansion of these
differences in AP gene expression, in vitro differentiation poten- depots in response to HFD (Figure 1B). We also compared the
tial, and contribution to metabolic disease (Baglioni et al., 2012; average size of the newly formed mTomato+ adipocytes to the
Fried et al., 2015; Loh et al., 2015; Macotela et al., 2012). How- existing mGFP+ adipocytes within each depot after 8 weeks of
ever, the mechanisms that control depot-specific AP activation diet and found no significant differences (Figures 1F and 1G),
in vivo have not been investigated. Here, we show that obeso- which suggests the newly formed cells contribute significantly
genic adipogenesis in SWAT is differentially regulated in males to fat mass.
and females, explaining the differences in WAT distribution be- We next directly assessed the relationship between adipocyte
tween the sexes. Furthermore, through AP transplant studies, formation and WAT depot mass. Indeed, comparison of depot
we find that depot-specific proliferation and differentiation of size to adipocyte hyperplasia in individual animals shows that
APs is determined by the depot microenvironment and not by in male VWAT, female VWAT, and female SWAT depots, adipo-
cell-intrinsic differences in APs. cyte formation is significantly correlated with fat pad size after
8 weeks of diet (Figures 2A2D). Furthermore, whole-body fat
RESULTS mass is significantly correlated with adipogenesis within these
depots (Figure S2). Conversely, there is no correlation between
Adipocyte Hyperplasia Contributes to Depot- and Sex- adipocyte formation and either fat pad mass or body fat mass
Specific WAT Growth in Obesity in the non-adipogenic male SWAT depot (Figures 2B and S2B).
Sex-specific patterns of adipose distribution have been well These data indicate that adipocyte hyperplasia significantly con-
documented in humans (Gesta et al., 2007; Karastergiou et al., tributes to depot- and sex-specific WAT growth in obesity.
2012; Palmer and Clegg, 2015). To determine if C57Bl6/J mice The production of new adipocytes in vivo occurs through the
display similar phenotypes in the high-fat diet (HFD) model of proliferation and differentiation of APs within the adipose tissue
obesity, we measured whole body fat mass (Figure 1A) and fat stroma (Jeffery et al., 2015; Rodeheffer et al., 2008). In males,
pad weight of the perigonadal VWAT and the inguinal SWAT HFD feeding induces the transient proliferation of APs specif-
depots (Figure 1B) after 8 weeks of HFD or standard diet (SD) ically in VWAT during the first week of HFD feeding, followed
feeding. Although the precise anatomic location of these depots by adipogenesis within this depot (Jeffery et al., 2015). To deter-
in mice differ from humans, these mouse depots impact meta- mine whether the same mechanism of transient AP activation
bolism similarly to human visceral adipose tissues (Barzilai also occurs in female mice upon HFD feeding, we assessed
et al., 1999; Foster et al., 2010, 2011; Gabriely and Barzilai, the incorporation of bromodeoxyuridine (BrdU) into APs via
2003) and gluteofemoral adipose tissue (Foster et al., 2013; flow cytometry (Figure 2E). We find that in both the female
Tran et al., 2008), respectively. We find that upon HFD feeding, SWAT and VWAT depots, HFD feeding induces rapid and tran-
males accumulate more fat mass than females (Figure 1A), sient proliferation of APs (Figures 2F, S2E, and S2F), similar to
consistent with previous reports (Grove et al., 2010; Medrikova male VWAT (Jeffery et al., 2015). Furthermore, analysis of mature
et al., 2012). Interestingly, in both SD- and HFD-fed conditions, adipocyte nuclei after a 1-week BrdU pulse and 7-week chase
male VWAT is significantly larger than male SWAT, whereas indicates that HFD induces significant adipocyte formation
these two depots are maintained at similar sizes in females (Fig- from proliferative precursors in both the SWAT and VWAT de-
ure 1B). These data indicate that, similar to humans, mice are pots of female mice (Figure 2G), whereas males on HFD display
sexually dimorphic with regard to fat mass distribution in obesity. increased BrdU-positive adipocyte formation in VWAT (Jeffery
We recently showed that HFD feeding induces adipocyte et al., 2015), but not SWAT (Figure S2G).
hyperplasia in male VWAT, but not male SWAT, after 8 weeks Interestingly, after ovariectomy females display a more male-
of HFD feeding (Jeffery et al., 2015), contributing to the preferen- like pattern of AP activation in response to HFD, with significantly
tial growth of VWAT in males. To determine whether hyperplastic reduced AP proliferation in SWAT (Figure 2H), which suggests
mechanisms influence sex-specific WAT growth, we used the that sex hormones play a role in the activation of SWAT APs.
Adiponectin-creER; mTmG mouse model to track the formation To test whether estrogen influences depot-specific AP activa-
of new mature adipocytes in vivo in response to SD or HFD tion, we administered estrogen to male mice via mini osmotic
feeding. In this model, new adipocytes that form after tamoxifen pumps and assessed their AP proliferation response to HFD
injection are mTomato+ (mT), while pre-existing adipocytes will feeding. We find that estrogen treatment of males results in

(D and E) Representative confocal images (left) and quantification (right) of adipocyte labeling in the indicated depots of Adiponectin-creER; mTmG male (D) or
female (E) mice (males: n = 46; females: n = 1215).
(F and G) Average adipocyte diameter of tdTomato+ (new) and eGFP+ (old) adipocytes from male (F) and female (G) mice (n = 46).
Note: all data from (A)(E) are from tamoxifen-treated animals. Significance was determined by comparing the indicated groups using an unpaired two-tailed
Students t test. Scale bar is 100 mM. Error bars represent mean SEM. See also Figure S1.

144 Cell Metabolism 24, 142150, July 12, 2016


Figure 2. Fat Pad Weight and Adipocyte Precursor Activation Mirror Depot-Specific Patterns of Adipogenesis
(AD) Correlation between adipogenesis and fat pad weight in the male VWAT (A) and SWAT (B) depots and the female VWAT (C) and SWAT (D) depots. Each
point represents one mouse (n = 2126).
(E) Representative flow cytometry plots to measure BrdU incorporation into adipocyte precursors.
(F) BrdU incorporation into APs from the indicated depot of female mice following 1 week of SD or HFD and BrdU treatment (n = 5 per group).

(legend continued on next page)


Cell Metabolism 24, 142150, July 12, 2016 145
significantly increased AP proliferation in SWAT upon HFD depot microenvironment. To test this possibility, we performed
feeding (Figure 2I), suggesting that estrogen influences the transplant experiments with APs derived from the SWAT of
cellular response of the SWAT depot to diet. Taken together, male mice expressing luciferase under the control of the mature
these data show that adipocyte hyperplasia, mediated by the adipocyte-specific leptin promoter (Figure S3A) (Rodeheffer
proliferation and differentiation of APs, is an important compo- et al., 2008), as well as the mT/mG cassette (leptin-luciferase;
nent of depot- and sex-specific WAT growth in obesity. mT/mG mice). These mice allow both non-invasive assessment
of adipocyte formation via luciferase activity and direct observa-
The Adipose Microenvironment Determines Adipocyte tion of transplanted cells via tdTomato fluorescence. Impor-
Precursor Activation tantly, male mice with this genetic background (FVB/NJ) still
We next investigated the mechanisms regulating depot-specific display a VWAT-specific AP proliferation pattern in response to
AP activation. Previous in vitro studies suggest that APs from HFD feeding (Figure S4B). Leptin-luciferase; mT/mG SWAT
SWAT and VWAT exhibit cell-intrinsic differences that control APs were transplanted into the VWAT or SWAT of congenic
their response to environmental cues and differentiation poten- wild-type male recipient mice, and after recovery, mice were
tial (Baglioni et al., 2012; Loh et al., 2015; Macotela et al., placed on HFD for 8 weeks. Luciferase signal was monitored af-
2012). To determine whether cell-intrinsic phenomena explain ter transplant of APs (baseline) and after 8 weeks of HFD feeding
depot-specific AP activation in vivo, we performed a series of (Figure 4A). Excitingly, we find that upon HFD feeding, SWAT-
AP transplant experiments (Figure 3A). In these experiments, derived APs display significantly increased adipogenesis when
APs were isolated from the VWAT or SWAT of tdTomato-ex- transplanted into the VWAT microenvironment (Figures 4B and
pressing mice by fluorescence-activated cell sorting (FACS) 4C), but not when transplanted back into SWAT, despite clear
and then transplanted into either the VWAT or SWAT of congenic engraftment of injected cells (Figures S4CS4E). Furthermore,
wild-type recipient mice. After recovery, the recipient mice were confocal analysis of adipose tissue from the luminescent region
placed on either HFD or SD for 1 week, and the proliferation of of the injected VWAT fat pad shows the presence of lipid-filled,
tdTomato+ donor APs and tdTomato endogenous APs was tdTomato+ adipocytes (Figure 4D), further confirming that HFD
determined by BrdU incorporation. Flow cytometry analysis of induces the differentiation of SWAT-derived APs in the VWAT
APs demonstrates engraftment of tdTomato+ donor APs in microenvironment.
recipient depots and permits the clear distinction between donor
and recipient AP populations (Figures 3B, 3C, and S3A, and DISCUSSION
S3B). In recipient mice fed SD, neither donor nor endogenous
APs are activated in either depot, indicating that transplantation Taken together, these data show that AP proliferation in
alone does not induce AP proliferation (Figures 3D and 3E). In response to HFD is regulated by cell-extrinsic factors in the
recipient mice fed HFD, APs derived from VWAT depots display depot microenvironment. In the inguinal SWAT depot, systemic
high levels of proliferation when injected back into the VWAT levels of estrogen influence this response. The finding that APs
depot, similar to endogenous cells and APs in the uninjected from different WAT depots are functionally interchangeable
contralateral depot, supporting the ability of the transplanted in vivo suggests that developmental lineage does not irreversibly
cells to be activated by obesogenic stimuli (Figure 3D). Surpris- determine depot-specific AP behavior. Rather, APs are function-
ingly, SWAT-derived donor AP cells are also induced to prolifer- ally plastic cells that are influenced by their physiologic context.
ate after HFD feeding when cells are placed in the VWAT micro- We employed the Adiponectin-creER model to study WAT dy-
environment (Figure 3D), but neither VWAT- nor SWAT-derived namics, which utilizes tamoxifen treatment for cre activation and
APs proliferate significantly when injected back into SWAT cell tracing. While higher doses of tamoxifen have detrimental ef-
(Figure 3E). Furthermore, when male SWAT APs are injected fects on WAT (Ye et al., 2015), reports are conflicting about how
into female SWAT, their proliferation is induced by HFD (Fig- the lower tamoxifen dose used here affects WAT (Hesselbarth
ure S3C), despite lower levels of engraftment (35%, 11/31) and et al., 2015; Liu et al., 2015). To control for the potential effects
increased background proliferation, which may be caused by of tamoxifen treatment in the Adiponectin-creER experiments,
sex-mismatched transplants (Volk et al., 2016; Weiss et al., both SD and HFD groups were treated with tamoxifen. Further-
2009). These data indicate that despite their separate develop- more, if the lower doses of tamoxifen used here had long-term
mental lineages (Chau et al., 2014; Krueger et al., 2014), the effects on creER activation, as has been shown for higher doses
depot-specific response of APs to HFD feeding is determined (Ye et al., 2015), we would not detect any adipocyte formation in
by the WAT depot microenvironment, not by cell-intrinsic prop- our lineage-tracing experiments. We also show similar patterns
erties of APs within distinct depots. of depot-specific adipocyte formation using the tamoxifen-inde-
The depot-specific regulation of both AP proliferation (Figures pendent BrdU tracing assay, demonstrating that the trends in
2 and 3) (Jeffery et al., 2015) and adipogenesis (Figure 1) sug- adipogenesis that we report are not an artifact of tamoxifen
gests that AP differentiation is also controlled by the adipose treatment.

(G) BrdU incorporation into adipocyte nuclei as measured via immunofluorescence in paraffin sections following 1 week of BrdU treatment (pulse) and 7 more
weeks of the indicated diet (chase) (n = 810).
(H and I) BrdU incorporation into APs from the indicated depot of ovariectomized female (H) or estrogen-treated male (I) mice following 1 week of SD or HFD and
BrdU treatment (n = 510).
Significance in (A)(D) was determined using Spearmans non-parametric two-tailed correlation analysis. Significance in (F)(I) was determined by comparing the
indicated groups using an unpaired two-tailed Students t test. Error bars represent mean SEM. See also Figure S2.

146 Cell Metabolism 24, 142150, July 12, 2016


Figure 3. Adipocyte Precursor Activation at the Onset of Obesity Is Determined by the Adipose Depot Microenvironment
(A) Experimental design for AP transplant experiment.
(B and C) Representative flow cytometry histogram (B) and dot plot (C) demonstrating engraftment of donor-derived tdTomato+ APs in the recipient depot and
BrdU incorporation into both tdTomato and tdTomato+ populations.
(D and E) BrdU incorporation into APs in donor, endogenous, or contralateral populations after 1 week of the indicated diet and BrdU treatment (n = 38).
Astrisks indicate significance over SD controls. Significance was determined using one-way ANOVA. Error bars represent mean SEM. See also Figure S3.

Similar to our previous studies of male mice (Jeffery et al., fraction, the abundant CD24 preadipocytes are post-mitotic,
2015), the BrdU- and cre-based techniques detect similar while the comparatively rare CD24+ adipocyte progenitors
depot-specific trends in adipogenesis but result in different proliferate in response to obesogenic stimuli, but rapidly lose
quantitative measurements of adipocyte formation, with the CD24 expression upon activation to generate and replenish the
BrdU method consistently displaying a much lower percentage CD24 preadipocytes (Berry and Rodeheffer, 2013; Jeffery
of new cells formed. For example, in the VWAT of HFD-fed fe- et al., 2015). This large pool of post-mitotic preadipocytes can
males, the Adiponectin-creER method identifies 35.8% 1.8% thus contribute to adipocyte formation without re-entering the
of adipocytes as newly formed, while the Brdu method only la- cell cycle and incorporating BrdU. As a result, the cre-based
bels 4% 0.81% of mature adipocytes. This discrepancy is method assesses the adipogenic capacity of both CD24+ and
explained by WAT biology, as the cre-based lineage tracing CD24 adipocyte precursor populations combined, while the
method assesses the contribution of both post-mitotic preadipo- BrdU assay only quantifies the contribution of the CD24+ adipo-
cytes and proliferative adipocyte progenitors, while the BrdU cyte progenitors within the limited time window of the experi-
technique only assesses the contribution of proliferative cells. ment. Therefore, the elevated assessment of adipogenesis in
We have previously shown that within the WAT stromal vascular the cre model compared to the BrdU assay is accounted for by

Cell Metabolism 24, 142150, July 12, 2016 147


Figure 4. High-Fat Diet-Induced Adipogenesis of Adipocyte Precursors Is Determined by the Adipose Depot Microenvironment
(A) Experimental design for AP transplant experiment.
(B) Representative images of mice with luminescent signal overlay following luciferase injection at the indicated time following transplant of APs from the SWAT of
leptin-luciferase; mTmG mice.
(C) Quantification of luminescent signal at the indicated experimental time points following transplant of APs from the SWAT of leptin-luciferase; mTmG mice
(n = 12).
(D) Representative image of VWAT depot with luminescent overlay showing the region with luciferase activity in the left pad where the transplant was performed.
(E) Representative confocal images of the luminescent region in (D) showing tdTomato+ mature adipocytes with LipidTOX staining (yellow crosses) adjacent to
endogenous tdTomato- adipocytes (white asterisks). Exposure time for each luminescent image was 1 min. Scale bar is 100mM.
Significance was determined using an unpaired two-tailed Students t test. Error bars represent mean SEM. See also Figure S4.

the differentiation of the large pool of committed, post-mitotic The clear links between depot- and sex-specific adipogene-
preadipocytes that were formed from the proliferative CD24+ sis, AP activation, and fat pad weight demonstrate the important
adipocyte progenitors prior to the initiation of the experiment. role that AP regulation and adipogenesis play in obesogenic

148 Cell Metabolism 24, 142150, July 12, 2016


WAT growth. Future studies aimed to define these regulatory ACKNOWLEDGMENTS
signals and compare their function in adipogenic and non-adipo-
We thank Ewa Menet in the Yale Flow Cytometry facility for her technical
genic adipose tissue microenvironments may lead to novel ap-
assistance. This work was supported by Womens Health Research at Yale,
proaches to control adipose tissue distribution in a manner NIDDK grant DK090489, ADA Award 7-12-JF-46, and Yale DERC DK045735
that favors metabolically beneficial SWAT growth over detri- to M.S.R. and Lo Fellowships for Excellence in Stem Cell Research to R.B.
mental VWAT growth. and E.J.

EXPERIMENTAL PROCEDURES Received: October 21, 2015


Revised: March 15, 2016
Animals Accepted: May 25, 2016
All animal studies followed guidelines issued by Yale Universitys Institutional Published: June 16, 2016
Animal Care and Use Committee (IACUC). See Supplemental Experimental
Procedures for details on mouse strains. VWAT refers to the perigonadal REFERENCES
WAT and SWAT refers to inguinal WAT in mice. For BrdU experiments, mice
were given 0.40.8 mg/ml BrdU in drinking water, refreshed every 2 days. Appleton, S.L., Seaborn, C.J., Visvanathan, R., Hill, C.L., Gill, T.K., Taylor,
High-fat diet is from Research Diets (D12492). Standard diet is from Harlan A.W., and Adams, R.J.; North West Adelaide Health Study Team (2013).
Laboratories (2018S). Analysis of body composition was performed by Echo Diabetes and cardiovascular disease outcomes in the metabolically healthy
MRI (Echo Medical System). For estrogen treatment, 8-week-old male mice obese phenotype: a cohort study. Diabetes Care 36, 23882394.
were implanted with mini osmotic pumps (Alzet 1004) delivering cyclodex-
Arner, P., Andersson, D.P., Thorne, A., Wiren, M., Hoffstedt, J., Naslund, E.,
tran-coated estrogen (Sigma E4389) at a dose of 2 mg/kg/day in PBS. Mice
Thorell, A., and Ryden, M. (2013). Variations in the size of the major omentum
were allowed to recover for 2 weeks after pump implantation prior to experi-
are primarily determined by fat cell number. J. Clin. Endocrinol. Metab. 98,
ment initiation.
E897E901.
Baglioni, S., Cantini, G., Poli, G., Francalanci, M., Squecco, R., Di Franco, A.,
Transplant Assay
Borgogni, E., Frontera, S., Nesi, G., Liotta, F., et al. (2012). Functional differ-
0.51 million APs were isolated from the indicated depot of male mice and
ences in visceral and subcutaneous fat pads originate from differences in
injected into the indicated pad of 3- to 6-week-old congenic wild-type mice.
the adipose stem cell. PLoS ONE 7, e36569.
After recovery, mice were placed on HFD or SD and treated with BrdU for
1 week and then sacrificed or maintained on diet for 8 weeks prior to analysis. Barzilai, N., She, L., Liu, B.Q., Vuguin, P., Cohen, P., Wang, J., and Rossetti, L.
Left and right adipose tissue depots were excised and analyzed separately (1999). Surgical removal of visceral fat reverses hepatic insulin resistance.
via flow cytometry, microscopy, or luminescence (see Supplemental Experi- Diabetes 48, 9498.
mental Procedures). Berry, R., and Rodeheffer, M.S. (2013). Characterization of the adipocyte
cellular lineage in vivo. Nat. Cell Biol. 15, 302308.
Flow Cytometry Berry, R., Jeffery, E., and Rodeheffer, M.S. (2014). Weighing in on adipocyte
Flow cytometry preparations were performed as described in Jeffery et al. precursors. Cell Metab. 19, 820.
(2015) for BrdU analysis and Berry and Rodeheffer (2013) for cell sorting.
Chau, Y.Y., Bandiera, R., Serrels, A., Martnez-Estrada, O.M., Qing, W., Lee,
M., Slight, J., Thornburn, A., Berry, R., McHaffie, S., et al. (2014). Visceral
Confocal Microscopy and subcutaneous fat have different origins and evidence supports a
The mT/mG quantification experiments were performed as described in Jeff- mesothelial source. Nat. Cell Biol. 16, 367375.
ery et al. (2015), starting the 50 mg/kg tamoxifen treatments at 8 weeks of age.
See the Supplemental Experimental Procedures for further details. For anal- Cohen, P., Levy, J.D., Zhang, Y., Frontini, A., Kolodin, D.P., Svensson, K.J., Lo,
ysis of leptin-luciferase; tdTomato+ adipocytes following transplantation, tis- J.C., Zeng, X., Ye, L., Khandekar, M.J., et al. (2014). Ablation of PRDM16 and
sue from the luminescent region of the VWAT was dissected and stained beige adipose causes metabolic dysfunction and a subcutaneous to visceral
with HCS LipidTOX Green Neutral Lipid Stain (Invitrogen, H34475, used at fat switch. Cell 156, 304316.
1-100) for at least 30 min before being washed in PBS and mounted onto slides Coutinho, T., Goel, K., Correa de Sa, D., Kragelund, C., Kanaya, A.M., Zeller,
in Fluoromount-G (Southern Biotech; 0100-01). M., Park, J.S., Kober, L., Torp-Pedersen, C., Cottin, Y., et al. (2011). Central
obesity and survival in subjects with coronary artery disease: a systematic
Statistical Analysis review of the literature and collaborative analysis with individual subject
Statistical tests used are indicated in the figure legends. All statistical tests data. J. Am. Coll. Cardiol. 57, 18771886.
were performed using GraphPad Prism v.6.02 for Windows (GraphPad Soft- Foster, M.T., Shi, H., Seeley, R.J., and Woods, S.C. (2010). Transplantation
ware). Data are expressed as mean SEM, and p < 0.05 was considered sta- or removal of intra-abdominal adipose tissue prevents age-induced glucose
tistically significant. Outliers were identified with the ROUT method test using insensitivity. Physiol. Behav. 101, 282288.
GraphPad Prism software, setting Q to 10%. Sample sizes for main figures are Foster, M.T., Shi, H., Seeley, R.J., and Woods, S.C. (2011). Removal of intra-
listed in Table S1 and indicate individual animals (biological replicates). abdominal visceral adipose tissue improves glucose tolerance in rats: role of
hepatic triglyceride storage. Physiol. Behav. 104, 845854.
SUPPLEMENTAL INFORMATION Foster, M.T., Softic, S., Caldwell, J., Kohli, R., de Kloet, A.D., and Seeley, R.J.
(2013). Subcutaneous Adipose Tissue Transplantation in Diet-Induced Obese
Supplemental Information includes Supplemental Experimental Procedures, Mice Attenuates Metabolic Dysregulation While Removal Exacerbates It.
four figures, and one table and can be found with this article online at http:// Physiol. Rep. 1, 1.
dx.doi.org/10.1016/j.cmet.2016.05.012.
Fried, S.K., Lee, M.J., and Karastergiou, K. (2015). Shaping fat distribution:
New insights into the molecular determinants of depot- and sex-dependent
AUTHOR CONTRIBUTIONS
adipose biology. Obesity (Silver Spring) 23, 13451352.

E.J. and M.S.R. designed experiments. R.B. generated the FVB/NJ; Leptin- Gabriely, I., and Barzilai, N. (2003). Surgical removal of visceral adipose tissue:
luciferase: mT/mG mice. E.J., A.W., L.C., and C.D.C. performed experiments. effects on insulin action. Curr. Diab. Rep. 3, 201206.
E.J., A.W., B.H., J.L.K., R.S.-P., and Z.S. analyzed data. E.J., A.W., and M.S.R. Gesta, S., Tseng, Y.H., and Kahn, C.R. (2007). Developmental origin of fat:
interpreted data and wrote the manuscript. tracking obesity to its source. Cell 131, 242256.

Cell Metabolism 24, 142150, July 12, 2016 149


Grove, K.L., Fried, S.K., Greenberg, A.S., Xiao, X.Q., and Clegg, D.J. (2010). relations between abdominal subcutaneous and visceral adiposity, liver fat,
A microarray analysis of sexual dimorphism of adipose tissues in high-fat- and cardiometabolic risk profile: the International Study of Prediction of
diet-induced obese mice. Int. J. Obes. 34, 9891000. Intra-Abdominal Adiposity and Its Relationship With Cardiometabolic Risk/
Heitmann, B.L., and Lissner, L. (2011). Hip Hip Hurrah! Hip size inversely Intra-Abdominal Adiposity. Am. J. Clin. Nutr. 96, 714726.
related to heart disease and total mortality. Obes. Rev. 12, 478481. Palmer, B.F., and Clegg, D.J. (2015). The sexual dimorphism of obesity. Mol.
Hesselbarth, N., Pettinelli, C., Gericke, M., Berger, C., Kunath, A., Stumvoll, Cell. Endocrinol. 402, 113119.
M., Bluher, M., and Kloting, N. (2015). Tamoxifen affects glucose and lipid Phillips, L.K., and Prins, J.B. (2008). The link between abdominal obesity and
metabolism parameters, causes browning of subcutaneous adipose tissue the metabolic syndrome. Curr. Hypertens. Rep. 10, 156164.
and transient body composition changes in C57BL/6NTac mice. Biochem.
Pischon, T., Boeing, H., Hoffmann, K., Bergmann, M., Schulze, M.B., Overvad,
Biophys. Res. Commun. 464, 724729.
K., van der Schouw, Y.T., Spencer, E., Moons, K.G., Tjnneland, A., et al.
Jeffery, E., Berry, R., Church, C.D., Yu, S., Shook, B.A., Horsley, V., Rosen, (2008). General and abdominal adiposity and risk of death in Europe.
E.D., and Rodeheffer, M.S. (2014). Characterization of Cre recombinase N. Engl. J. Med. 359, 21052120.
models for the study of adipose tissue. Adipocyte 3, 206211.
Rodeheffer, M.S., Birsoy, K., and Friedman, J.M. (2008). Identification of white
Jeffery, E., Church, C.D., Holtrup, B., Colman, L., and Rodeheffer, M.S. (2015). adipocyte progenitor cells in vivo. Cell 135, 240249.
Rapid depot-specific activation of adipocyte precursor cells at the onset of
obesity. Nat. Cell Biol. 17, 376385. Sanchez-Gurmaches, J., and Guertin, D.A. (2014). Adipocytes arise from
multiple lineages that are heterogeneously and dynamically distributed. Nat.
Karastergiou, K., and Fried, S.K. (2013). Multiple adipose depots increase car-
Commun. 5, 4099.
diovascular risk via local and systemic effects. Curr. Atheroscler. Rep. 15, 361.
Shi, H., Seeley, R.J., and Clegg, D.J. (2009). Sexual differences in the control of
Karastergiou, K., Smith, S.R., Greenberg, A.S., and Fried, S.K. (2012). Sex
energy homeostasis. Front. Neuroendocrinol. 30, 396404.
differences in human adipose tissues - the biology of pear shape. Biol. Sex
Differ. 3, 13. Snijder, M.B., Dekker, J.M., Visser, M., Yudkin, J.S., Stehouwer, C.D., Bouter,
L.M., Heine, R.J., Nijpels, G., and Seidell, J.C. (2003). Larger thigh and hip
Karastergiou, K., Fried, S.K., Xie, H., Lee, M.J., Divoux, A., Rosencrantz, M.A.,
circumferences are associated with better glucose tolerance: the Hoorn study.
Chang, R.J., and Smith, S.R. (2013). Distinct developmental signatures
Obes. Res. 11, 104111.
of human abdominal and gluteal subcutaneous adipose tissue depots.
J. Clin. Endocrinol. Metab. 98, 362371. Spalding, K.L., Arner, E., Westermark, P.O., Bernard, S., Buchholz, B.A.,
Krueger, K.C., Costa, M.J., Du, H., and Feldman, B.J. (2014). Characterization Bergmann, O., Blomqvist, L., Hoffstedt, J., Naslund, E., Britton, T., et al.
of Cre recombinase activity for in vivo targeting of adipocyte precursor cells. (2008). Dynamics of fat cell turnover in humans. Nature 453, 783787.
Stem Cell Reports 3, 11471158. Tran, T.T., Yamamoto, Y., Gesta, S., and Kahn, C.R. (2008). Beneficial effects
Lee, J.J., Beretvas, S.N., and Freeland-Graves, J.H. (2014). Abdominal of subcutaneous fat transplantation on metabolism. Cell Metab. 7, 410420.
adiposity distribution in diabetic/prediabetic and nondiabetic populations: a Volk, V., Schneider, A., Spineli, L.M., Grosshennig, A., and Stripecke, R.
meta-analysis. J. Obes. 2014, 697264. (2016). The gender gap: discrepant human T-cell reconstitution after cord
Liu, L., Zou, P., Zheng, L., Linarelli, L.E., Amarell, S., Passaro, A., Liu, D., and blood stem cell transplantation in humanized female and male mice. Bone
Cheng, Z. (2015). Tamoxifen reduces fat mass by boosting reactive oxygen Marrow Transplant. 51, 596597.
species. Cell Death Dis. 6, e1586. Wang, Q.A., Tao, C., Gupta, R.K., and Scherer, P.E. (2013). Tracking adipo-
Loh, N.Y., Neville, M.J., Marinou, K., Hardcastle, S.A., Fielding, B.A., genesis during white adipose tissue development, expansion and regenera-
Duncan, E.L., McCarthy, M.I., Tobias, J.H., Gregson, C.L., Karpe, F., and tion. Nat. Med. 19, 13381344.
Christodoulides, C. (2015). LRP5 regulates human body fat distribution by Weiss, E.S., Allen, J.G., Patel, N.D., Russell, S.D., Baumgartner, W.A., Shah,
modulating adipose progenitor biology in a dose- and depot-specific A.S., and Conte, J.V. (2009). The impact of donor-recipient sex matching on
fashion. Cell Metab. 21, 262272. survival after orthotopic heart transplantation: analysis of 18 000 transplants
Macotela, Y., Emanuelli, B., Mori, M.A., Gesta, S., Schulz, T.J., Tseng, Y.H., in the modern era. Circ Heart Fail 2, 401408.
and Kahn, C.R. (2012). Intrinsic differences in adipocyte precursor cells from Wormser, D., Kaptoge, S., Di Angelantonio, E., Wood, A.M., Pennells, L.,
different white fat depots. Diabetes 61, 16911699. Thompson, A., Sarwar, N., Kizer, J.R., Lawlor, D.A., Nordestgaard, B.G.,
Manolopoulos, K.N., Karpe, F., and Frayn, K.N. (2010). Gluteofemoral body fat et al.; Emerging Risk Factors Collaboration (2011). Separate and combined as-
as a determinant of metabolic health. Int. J. Obes. 34, 949959. sociations of body-mass index and abdominal adiposity with cardiovascular
Medrikova, D., Jilkova, Z.M., Bardova, K., Janovska, P., Rossmeisl, M., and disease: collaborative analysis of 58 prospective studies. Lancet 377, 1085
Kopecky, J. (2012). Sex differences during the course of diet-induced obesity 1095.
in mice: adipose tissue expandability and glycemic control. Int. J. Obes. 36, Ye, R., Wang, Q.A., Tao, C., Vishvanath, L., Shao, M., McDonald, J.G., Gupta,
262272. R.K., and Scherer, P.E. (2015). Impact of tamoxifen on adipocyte lineage
Nazare, J.A., Smith, J.D., Borel, A.L., Haffner, S.M., Balkau, B., Ross, R., tracing: Inducer of adipogenesis and prolonged nuclear translocation of Cre
Massien, C., Almeras, N., and Despres, J.P. (2012). Ethnic influences on the recombinase. Mol. Metab. 4, 771778.

150 Cell Metabolism 24, 142150, July 12, 2016

Vous aimerez peut-être aussi