Interconversion of white and brown adipocytes occurs between anabolic and catabolic states. The molecular mechanism regulating this phenotypic switch remains largely unknown. This study explores the role of tuberous sclerosis complex 1 (TSC1)–mechanistic target of rapamycin (mTOR) signaling in the conversion of brown to white adipose tissue (WAT). A colony of Fabp4-Tsc1−/− mice, in which the Tsc1 gene was specifically deleted by the fatty acid binding protein 4 (FABP4)-Cre, was established. Western blotting and immunostaining demonstrated the absence of TSC1 and activation of ribosomal protein S6 kinase 1, the downstream target of mTOR complex 1 (mTORC1) signaling, in the brown adipose tissues (BATs) of Fabp4-Tsc1−/− mice. Accumulation of lipid droplets in BAT was significantly increased. Levels of brown adipocyte markers were markedly downregulated, while white adipocyte markers were upregulated. Rapamycin reversed the conversion from BAT to WAT in Fabp4-Tsc1−/− mice. Deletion of the Tsc1 gene in cultured brown preadipocytes significantly increased the conversion to white adipocytes. FoxC2 mRNA, the transcriptional factor for brown adipocyte determination, was significantly decreased, while mRNAs for retinoblastoma protein, p107 and RIP140, the transcriptional factors for white adipocyte determination, increased in the BAT of Fabp4-Tsc1−/− mice. Our study demonstrates that TSC1-mTORC1 signaling contributes to the brown-to-white adipocyte phenotypic switch.

In mammals, there exist two different types of adipose tissues: white adipose tissues (WATs) and brown adipose tissues (BATs), which are made up of unilocular white adipocytes and multilocular (ML) brown adipocytes, respectively. White adipocytes contain all of the enzymatic machinery necessary to build triglycerides from fatty acids synthesized de novo or imported from circulating lipoproteins, while brown adipocytes comprise of abundant mitochondria that oxidize fatty acids and generate heat via uncoupling protein 1 (UCP1) in response to cold or excess feeding (13). WAT and BAT are traditionally considered distinct fat depots located in different anatomic sites. However, recent studies have shown that most of the fat depots contain both WAT and BAT, which are interconvertible (4). Differentiated WAT has been demonstrated to acquire certain BAT-like properties and can be converted into an energy-consumption organ under special conditions. Cold exposure or administration of β3-agonist has been reported to induce the emergence of ML brown adipocytes containing a high amount of mitochondria and UCP1 in WAT depots (57).

The molecular mechanism underlying the interconversion between WAT and BAT is currently under active investigation. Study with the transgenic mouse model suggests that C/EBPβ is critical for the brown adipocyte conversion. Transgenic knockin mouse that replaces the C/EBPα gene by C/EBPβ gene (denoted as C/EBP β/β) shows an increase in brown adipocyte-like cells characterized by cAMP accumulation, enhanced mitochondrial biogenesis, and UCP1 expression in WATs (8). In addition, several transcription-related molecules such as retinoblastoma protein (Rb), p107, transcriptional intermediary factor 2, 4E-binding protein 1, and RIP140 (915) have been reported to influence the brown or white fat phenotype selectivity. All of these transcriptional factors act to suppress the brown fat phenotype. Mice lacking p107, Rb, or RIP140 demonstrate a uniform replacement of WAT with BAT and elevated levels of peroxisome proliferator–activated receptor γ coactivator 1α (PGC1α) and UCP1 (13,14). In contrast, FoxC2, a member of the forkhead transcription factor family, induces the emergence of brown fat cells in WAT. WAT in mice overexpressing FoxC2 acquires certain BAT-like properties such as increased levels of PGC1α, UCP1, and two important cAMP pathway proteins: β3-adrenoceptor and the protein kinase A α regulatory subunit 1 (16). While these studies suggest that a group of transcriptional factors is critical for maintaining the WAT identity, the upstream signals or factors linking the extracellular signals with these transcriptional factors to initiate the conversion and therefore determine the fate of BAT versus WAT remain largely unknown.

The mechanistic target of rapamycin (mTOR), an evolutionarily conserved serine-threonine protein kinase, can sense the extracellular energy status to regulate the cell growth and proliferation in a variety of cells and tissues. Two distinct mTOR complexes, mTORC1 and mTORC2, have been characterized. mTORC1 is sensitive to rapamycin and is responsible for phosphorylation of ribosomal S6 kinase 1 (S6K1) and eukaryotic initiation factor 4E-binding protein 1. In contrast, mTORC2 is not inhibited by rapamycin (17,18). In adipocytes, mTOR is proposed to regulate protein synthesis, leptin synthesis and secretion, and adipogenesis (1921). Inhibition of mTORC1 signaling by rapamycin attenuates the mTOR-induced adipogenesis (2224). All of these data suggest that mTOR may serve as the critical intracellular molecule sensing the extracellular energy surplus to increase the energy storage capacity of adipocytes. Whether mTOR signaling controls the interconversion between brown and white adipocytes remains to be explored.

Tuberous sclerosis complex 1 (TSC1) and TSC2 form a heterodimer complex to negatively regulate mTORC1 activity, but might activate mTORC2 (18,25). In order to examine whether the TSC1-mTOR signaling is involved in the fate determination of BAT versus WAT, we generated a colony of transgenic mice Fabp4-Tsc1−/− mice in which TSC1, the negative upstream regulator of mTORC1 signaling, is specifically deleted by fatty acid binding protein 4 (FABP4)-Cre (26). We report in this study that deletion of Tsc1 gene activates mTORC1 signaling in the BAT of Fabp4-Tsc1−/− mice, which subsequently induces the conversion of brown adipocytes into cells with the morphology and gene expression pattern of white adipocytes.

Animals and Animal Care

Fabp4-Cre mice that express the Cre recombinase gene under the control of the FABP4 gene promoter, and Tsc1lox/lox mice in which exons 17 and 18 of Ts1 gene are flanked by loxP sites by homologous recombination, were purchased from The Jackson Laboratory (Bar Harbor, ME). Fabp4-Tsc1−/− mice were generated by breeding Tsc1lox/lox mice with Fabp4-Cre mice. Control experiments were performed using littermate Tsc1lox/lox animals (26). Mice were housed on a 12:12-h light/dark cycle. Regular chow and water were available ad libitum. The animals used in this study were handled in accordance with the Guide for the Care and Use of Laboratory Animals published by the National Institutes of Health (publication no. 85-23, revised 1996). All of the experimental protocols were approved by the Animal Care and Use Committee of Peking University.

Rapamycin Treatment

Rapamycin (Santa Cruz Biotechnology) was initially dissolved in 100% DMSO, stored at −20°C, and further diluted in normal saline immediately before use. Pregnant mice were randomly divided into two groups: control and treatment. At the near-term pregnancy, mice were intramuscularly injected with rapamycin at a dose of 1 mg/kg body weight/day or vehicle for 3 consecutive days before the birth of pups.

Isolation and Primary Culture of Brown Fat Adipocytes

Interscapular BAT was harvested from neonatal Tsc1lox/lox mice and brown adipocytes isolated by digestion with collagenase and mechanistic dispersion as previously described (27). Isolated cells were plated in tissue-culture dishes in DMEM supplemented with 20% FBS. After 4 h of culture at 37°C, cells were rinsed twice with PBS, after which 70% of the initial cells were attached to the dish, forming a monolayer. To induce adipocyte differentiation, cells were cultured for 2 days in 10% FBS-DMEM supplemented with 20 nmol/L insulin, 1 nmol/L T3, 12.5 mmol/L indomethacin, 0.5 mmol/L isobutylmethylxanthine, and 2 mg/mL dexamethasone. On days 3–10, the induction medium was substituted by maintenance medium consisting of DMEM supplemented with 20 nmol/L insulin and 1 nmol/L T3. Fresh maintenance medium was added every 3 days until day 10.

Adenovirus Infection

The Cre adenoviruses were expanded, titrated in 293 cells, and purified by cesium chloride methods as described previously (28). For adenovirus-mediated gene transfer, primary brown fat preadipocytes were cultured to 30–50% confluence and infected with adenovirus for 48 h. Infection efficiency was generally >60%, as judged by green fluorescent protein (GFP) expression observed under the microscope. Infected primary brown fat preadipocytes were differentiated for 10 days and then harvested for subsequent analysis.

RNA Extraction and Quantitative Real-Time PCR Analysis

Total RNA was isolated using the TRIzol reagent. Mitochondrial DNA (mtDNA) was isolated by Mitochondrial DNA Isolation Kit (BioVision Inc., Milpitas, CA). Reverse transcription and quantitative real-time PCR were performed as previously described (29,30). PCR reactions were performed in duplicate, and each experiment was repeated three to five times. Primers used in this study were shown in Table 1.

Table 1

List and sequences of primers used in RT-PCR experiments

Upstream primer (5′-3′)Downstream primer (5′-3′)Gene accession number
Mouse UCP1 GGACGACCCCTAATCTAATG CATTAGATTAGGGGTCGTCC NM_009463.3 
Mouse UCP2 GGAGAGTCAAGGGCTAGT ACTAGCCCTTGACTCTCC NM_011671.4 
Mouse UCP3 ATCAGGATTCTGGCAGGC GCCTGCCAGAATCCTGAT NM_009464.3 
Mouse PGC1α GATTGAAGTGGTGTAGCGAC GTCGCTACACCACTTCAATC NM_008904.2 
Mouse Agt GGAGTGACACCCAGAACA TAGATGGCGAACAGGAAG NM_007428.3 
Mouse Ednra CGGAGATCAACTTTCTGG TGGAGACGATTTCAATGG NM_010332.2 
Mouse annexin A1 CCCTTCCTTCAATGTATCC GCATAGCCAAAACAACCTC NM_010730.2 
Mouse Psat TACAAGGAGGTGGGTCTG ATTCTTCTGAGCACCAGC NM_001205339.1 
Mouse Rb 1 GGATGGAGAAGGACCTGATA GAGGCTGCTTGTGTCTCTGT NM_009029.2 
Mouse p107 GCAAGAGCATCATTCCTACT ACGAAACTCTTGTGGTAGGT NM_011249.2 
Mouse FoxC2 ATCACTCTGAACGGCATC GGCACTTTCACGAAGCAC NM_013519.2 
Mouse RIP140 CCTCGTCACTGCCTGAAG GCACTCAGAGCCAAGTTC AF053062.1 
mtDNA gene CO1 TGCTAGCCGCAGGCATTAC GGGTGCCCAAAGAATCAGAAC NC_013763.1 
Single-copy nuclear gene Ndufv1 CTTCCCCACTGGCCTCAAG CCAAAACCCAGTGATCCAGC NM_133666 
Mouse β-actin ATCTGGCACCACACCTTC AGCCAGGTCCAGACGCA NM_007393 
Upstream primer (5′-3′)Downstream primer (5′-3′)Gene accession number
Mouse UCP1 GGACGACCCCTAATCTAATG CATTAGATTAGGGGTCGTCC NM_009463.3 
Mouse UCP2 GGAGAGTCAAGGGCTAGT ACTAGCCCTTGACTCTCC NM_011671.4 
Mouse UCP3 ATCAGGATTCTGGCAGGC GCCTGCCAGAATCCTGAT NM_009464.3 
Mouse PGC1α GATTGAAGTGGTGTAGCGAC GTCGCTACACCACTTCAATC NM_008904.2 
Mouse Agt GGAGTGACACCCAGAACA TAGATGGCGAACAGGAAG NM_007428.3 
Mouse Ednra CGGAGATCAACTTTCTGG TGGAGACGATTTCAATGG NM_010332.2 
Mouse annexin A1 CCCTTCCTTCAATGTATCC GCATAGCCAAAACAACCTC NM_010730.2 
Mouse Psat TACAAGGAGGTGGGTCTG ATTCTTCTGAGCACCAGC NM_001205339.1 
Mouse Rb 1 GGATGGAGAAGGACCTGATA GAGGCTGCTTGTGTCTCTGT NM_009029.2 
Mouse p107 GCAAGAGCATCATTCCTACT ACGAAACTCTTGTGGTAGGT NM_011249.2 
Mouse FoxC2 ATCACTCTGAACGGCATC GGCACTTTCACGAAGCAC NM_013519.2 
Mouse RIP140 CCTCGTCACTGCCTGAAG GCACTCAGAGCCAAGTTC AF053062.1 
mtDNA gene CO1 TGCTAGCCGCAGGCATTAC GGGTGCCCAAAGAATCAGAAC NC_013763.1 
Single-copy nuclear gene Ndufv1 CTTCCCCACTGGCCTCAAG CCAAAACCCAGTGATCCAGC NM_133666 
Mouse β-actin ATCTGGCACCACACCTTC AGCCAGGTCCAGACGCA NM_007393 

Western Blot Analysis

Protein extracts of BAT tissues or cultured cells were immunoblotted with UCP1 (Santa Cruz Biotechnology), angiotensinogen (Agt; Epitomics), phosphorylated (p-)mTOR (Ser2448; Cell Signaling Technology, Beverly, MA), mTOR (Cell Signaling Technology), p-S6K1 (Thr389; Cell Signaling Technology), S6K1 (Cell Signaling Technology), p-S6 (Ser235/236; Cell Signaling Technology), S6 (Cell Signaling Technology), TSC1 (Cell Signaling Technology), TSC2 (Cell Signaling Technology), and β-actin (Cell Signaling Technology) as previously described (29,30). After incubation with IRDye-conjugated second antibody for 1 h, specific reaction was visualized using the Odyssey infrared imaging system (LI-COR Biosciences, Lincoln, NE).

Immunostaining

The BATs were quickly removed and prepared for immunostaining. Slides were individually incubated with p-S6 (Ser235/236; Cell Signaling Technology) antibody (1:100) in a humid chamber at 4°C overnight. Secondary antibody staining was performed with a biotin-labeled horse anti-rabbit antibody (1:200) for 1 h at room temperature, followed by incubation with a streptavidin-biotin horseradish peroxidase complex (Vector Laboratories, Burlingame, CA). Immunoreactivity was detected using diaminobenzidine substrate (peroxide substrate kit, SK-4100; Vector Laboratories) for 2–5 min. Slides were then counterstained with Mayer hematoxylin before dehydration and mounting. The expression of p-S6 was evaluated by comparing the staining intensities between different samples incubated with specific primary antibody or control IgG under same conditions.

Conventional Electron Microscopy

The BATs were fixed with 3% glutaraldehyde and 1% osmium in 0.1% cacodylate buffer, rinsed with a series of graded acetone solutions, and then embedded in epon 812. After ultrathin sectioning with an ultramicrotome (MT-7000; RMC Products, Tucson, AZ), specimens were stained with uranyl acetate and lead citrate and then observed under the JEM1230 electron microscope (Horiba Corp., Kyoto, Japan).

Statistical Analysis

Data were expressed as mean ± SEM. Data analysis used GraphPad Prism software. One-way ANOVA, Student-Newman-Keul test (comparisons between multiple groups), or unpaired Student t test (between two groups) was used as appropriate. A P value <0.05 denotes statistical significance.

Activation of mTORC1 Signaling in Fabp4-Tsc1−/− Mice

To explore the role of TSC1-mTORC1 signaling in the determination of brown versus white adipocytes, we generated the Fabp4-Tsc1−/− transgenic mice in which the Tsc1 gene was specifically deleted by FABP4-Cre. As showed in Fig. 1A, levels of TSC1 were negligible in the BAT of Fabp4-Tsc1−/− transgenic mice, whereas modest expression was detected in wild-type (WT) littermates. Levels of TSC2 remain unaltered in Fabp4-Tsc1−/− transgenic mice relative to the WT animals. Similar reduction in TSC1 levels was also observed in the brain, skin and liver tissues, whereas no alteration was detected in heart, lung and kidney (Fig. 1B). These data indicate the successful deletion of the Tsc1 gene in the Fabp4-Tsc1−/− transgenic mice. Since TSC1 is the upstream negative regulator of mTORC1 signaling, we next investigated the mTOR activity in the BAT derived from the Fabp4-Tsc1−/− transgenic mice by examining p-mTOR and ribosomal protein S6K1, the downstream target of mTORC1. Relative to the WT mice, p-mTOR (Ser2448) and p-S6K1 (Thr389) signal intensity increased markedly in the BATs of Fabp4-Tsc1−/− mice (knockout [KO]) (Fig. 1A). Further examination by immunohistochemistry showed that immunoreactivity of p-S6 (Ser235/236), the downstream signal of S6K1, increased significantly in brown adipocytes of Fabp4-Tsc1−/− mice relative to the WT littermates (Fig. 1C). All of these observations demonstrate the activation of mTORC1 signaling in BAT of Fabp4-Tsc1−/− mice.

Figure 1

Activation of mTORC1 signaling in Fabp4-Tsc1−/− mice. A: Representative Western blots from three independent experiments showing levels of TSC1, TSC2, p-mTOR (Ser2448), mTOR, p-S6K1 (Thr389), and S6K1 in BATs from WT or Fabp4-Tsc1−/− mice (KO). B: Levels of TSC1 in the brain, skin, liver, heart, lung, and kidney tissues. C: Representative immunohistochemical staining for p-S6 (Ser235/236) in lower (top panel) and higher magnification (bottom panel). β-Actin and IgG were used as the internal controls for Western blotting and the negative control for immunohistochemistry, respectively. Relative protein signal intensity was quantified, normalized to levels of WT mice and expressed as mean ± SEM. *P < 0.05, **P < 0.01 (n = 3).

Figure 1

Activation of mTORC1 signaling in Fabp4-Tsc1−/− mice. A: Representative Western blots from three independent experiments showing levels of TSC1, TSC2, p-mTOR (Ser2448), mTOR, p-S6K1 (Thr389), and S6K1 in BATs from WT or Fabp4-Tsc1−/− mice (KO). B: Levels of TSC1 in the brain, skin, liver, heart, lung, and kidney tissues. C: Representative immunohistochemical staining for p-S6 (Ser235/236) in lower (top panel) and higher magnification (bottom panel). β-Actin and IgG were used as the internal controls for Western blotting and the negative control for immunohistochemistry, respectively. Relative protein signal intensity was quantified, normalized to levels of WT mice and expressed as mean ± SEM. *P < 0.05, **P < 0.01 (n = 3).

Close modal

Acquisition of White Adipocyte Morphological Properties in BATs

Since Fabp4-Tsc1−/− mice died prematurely within 48 h after birth (26), our capability to analyze the development and function of white adipocytes was limited. We thus focused on the BAT, which was already developed perinatally. As shown in Fig. 2A, cells resembling white adipocytes with more lipid vacuoles increased markedly in the BAT derived from Fabp4-Tsc1−/− mice as compared with the WT littermates. These vacuolar structures were further validated to be lipid droplets by Oil Red O staining (Fig. 2B). The area and diameter of Oil Red O–positive lipid droplets were significantly increased in the BAT derived from Fabp4-Tsc1−/− mice as compared with the WT littermates (Fig. 2C). These results suggest a phenotypic conversion of brown adipocytes into cells with morphological property of white adipocytes in the Fabp4-Tsc1−/− mice.

Figure 2

Whitening of BAT in Fabp4-Tsc1−/− mice. A: Hematoxylin-eosin staining of BAT from WT littermates and Fabp4-Tsc1−/− mice (KO). Shown are histological findings in lower (left panels) and higher magnification (right panels). B: Representative results of Oil Red O staining showing the accumulation of lipid droplets in BAT from WT littermates and Fabp4-Tsc1−/− mice (KO). C: The area and diameter of Oil Red O–positive lipid droplets in BAT from WT littermates and Fabp4-Tsc1−/− mice (KO). Data were expressed as mean ± SEM. D: Electron micrographs depicted the architecture of mitochondria in the BAT from Fabp4-Tsc1−/− mice (KO) and WT littermates. Scale bars, 0.5 μm. E: The diameter of mitochondria was quantified using by the National Institutes of Health ImageJ software program. Results were expressed as mean ± SEM. F: Content of mtDNA was measured by quantitative real-time PCR. Ndufv1 gene was used as the internal control. Results were normalized to the control WT littermates and expressed as mean ± SEM. *P < 0.05, **P < 0.01 in comparison with WT animals (n = 6).

Figure 2

Whitening of BAT in Fabp4-Tsc1−/− mice. A: Hematoxylin-eosin staining of BAT from WT littermates and Fabp4-Tsc1−/− mice (KO). Shown are histological findings in lower (left panels) and higher magnification (right panels). B: Representative results of Oil Red O staining showing the accumulation of lipid droplets in BAT from WT littermates and Fabp4-Tsc1−/− mice (KO). C: The area and diameter of Oil Red O–positive lipid droplets in BAT from WT littermates and Fabp4-Tsc1−/− mice (KO). Data were expressed as mean ± SEM. D: Electron micrographs depicted the architecture of mitochondria in the BAT from Fabp4-Tsc1−/− mice (KO) and WT littermates. Scale bars, 0.5 μm. E: The diameter of mitochondria was quantified using by the National Institutes of Health ImageJ software program. Results were expressed as mean ± SEM. F: Content of mtDNA was measured by quantitative real-time PCR. Ndufv1 gene was used as the internal control. Results were normalized to the control WT littermates and expressed as mean ± SEM. *P < 0.05, **P < 0.01 in comparison with WT animals (n = 6).

Close modal

As the major source for mammalian nonshivering thermogenesis, brown adipocytes contain abundant mitochondria. We thus examined the alteration of mitochondrial structure of BAT using the electron microscopy. In the adipocytes from WT mice, large and electron lucid mitochondria with well-developed cristae are observed, while cells from Fabp4-Tsc1−/− mice revealed mitochondrial swelling and cristae rupture (Fig. 2D). The diameter of mitochondria increased significantly in Fabp4-Tsc1−/− mice in comparison with WT littermates (Fig. 2E). Consistent with this observation, content of relative mtDNA decreased markedly in Fabp4-Tsc1−/− mice relative to WT animals (Fig. 2F). These observations indicate the abnormal mitochondrial structure and reduced mtDNA content in Fabp4-Tsc1−/− mice.

Acquisition of White Adipocyte–Typical Gene Profile in BAT

We next examined the gene expression profile in the BAT from Fabp4-Tsc1−/− mice. With one exception, UCP2, all examined brown adipocyte markers such as UCP1, UCP3, and PGC1α decreased significantly by 62 ± 5, 41 ± 12, and 68 ± 7% in the BAT from Fabp4-Tsc1−/− mice relative to the WT littermates, respectively (Fig. 3A). Meanwhile, markers of white adipocytes (31), such as Agt, phosphoserine aminotransferase (Psat), and endothelian receptor type A (Ednra), increased markedly by 90 ± 24.9, 50 ± 16, and 23 ± 8%, respectively (Fig. 3B). To further validate the changes at the protein levels, the typical brown adipocyte marker UCP1 and white adipocyte marker Agt were examined by Western blotting. As shown in Fig. 3C, expression of UCP1 protein was significantly decreased by 56 ± 15%, while Agt increased by 225 ± 42% in the BAT from Fabp4-Tsc1−/− mice relative to the WT littermates.

Figure 3

Gene expression switch from brown to white adipocytes. The mRNA and protein were extracted from the BAT in Fabp4-Tsc1−/− mice (KO) and WT littermates. Real-time PCR and Western blotting were performed to evaluate the expression of brown adipocyte and white adipocyte markers. A: mRNA levels of brown adipocyte markers: UCP1, UCP2, UCP3, and PGC1α. B: mRNA levels of white adipocyte markers: Agt, Psat, Ednra. C: Representative results of Western blot for UCP1 and Agt. β-Actin was used as the loading control for the Western blot. Relative protein signal intensity was quantified, normalized to levels of WT mice, and expressed as mean ± SEM. mRNA expression levels of distinct genes were normalized to internal control β-actin and expressed as mean ± SEM. *P < 0.05, **P < 0.01 (n = 3 or 6).

Figure 3

Gene expression switch from brown to white adipocytes. The mRNA and protein were extracted from the BAT in Fabp4-Tsc1−/− mice (KO) and WT littermates. Real-time PCR and Western blotting were performed to evaluate the expression of brown adipocyte and white adipocyte markers. A: mRNA levels of brown adipocyte markers: UCP1, UCP2, UCP3, and PGC1α. B: mRNA levels of white adipocyte markers: Agt, Psat, Ednra. C: Representative results of Western blot for UCP1 and Agt. β-Actin was used as the loading control for the Western blot. Relative protein signal intensity was quantified, normalized to levels of WT mice, and expressed as mean ± SEM. mRNA expression levels of distinct genes were normalized to internal control β-actin and expressed as mean ± SEM. *P < 0.05, **P < 0.01 (n = 3 or 6).

Close modal

Acquisition of White Adipocyte–Typical Gene Phenotype in Cultured Brown Adipocytes with Reduced Levels of TSC1

To further explore the effect of TSC1-mTORC1 signaling in the brown-to-white adipocyte phenotypic conversion, we infected the cultured brown fat preadipocytes derived from the Tsc1loxp/loxp mice with Cre adenovirus. As shown in Fig. 4A, reduction of TSC1 activated the mTORC1 signaling in cultured brown adipocytes. Consistent with the observations in Fabp4-Tsc1−/− mice, brown adipocyte markers such as UCP1 and UCP3 decreased by 88 ± 3 and 73 ± 10%, respectively, while UCP2 remained unaltered in comparison with control cells infected with GFP adenovirus (Ad) (Fig. 4B). Contradicting the in vivo observations, PGC1α increased modestly. In contrast, white adipocyte markers Agt, Psat, and Ednra increased by 327 ± 13, 183 ± 19, 287 ± 45%, respectively (Fig. 4C). These results further demonstrate that activation of mTORC1 signaling by removal of Tsc1 gene induces the brown-to-white adipocyte phenotypic switch in vitro.

Figure 4

Induction of white adipocyte conversion by activation of mTORC1 signaling in cultured brown adipocytes. Brown adipocytes were cultured and infected with Ad-Cre or Ad-GFP as described in 2research design and methods. Western blotting was performed to detect levels of TSC1, p-mTOR (Ser2448), p-S6K1 (Thr389) and p-S6 (Ser235/236). Real-time PCR was performed to evaluate the expression of brown and white adipocyte markers. A: Representative Western blots from three independent experiments showing the deletion of TSC1 and subsequent activation of mTORC1 signaling. Signal intensity of relative proteins was quantified, normalized to levels of WT mice, and expressed as mean ± SEM. B: Relative expression levels of brown adipocyte markers: UCP1, UCP2, UCP3, and PGC1α in cultured brown adipocytes infected with Ad-Cre or Ad-GFP. C: mRNA levels of white adipocyte marker: Agt, Psat, and Ednra. Levels of mRNA expression were normalized to internal control β-actin and expressed as mean ± SEM. *P < 0.05, **P < 0.01 (n = 3–6).

Figure 4

Induction of white adipocyte conversion by activation of mTORC1 signaling in cultured brown adipocytes. Brown adipocytes were cultured and infected with Ad-Cre or Ad-GFP as described in 2research design and methods. Western blotting was performed to detect levels of TSC1, p-mTOR (Ser2448), p-S6K1 (Thr389) and p-S6 (Ser235/236). Real-time PCR was performed to evaluate the expression of brown and white adipocyte markers. A: Representative Western blots from three independent experiments showing the deletion of TSC1 and subsequent activation of mTORC1 signaling. Signal intensity of relative proteins was quantified, normalized to levels of WT mice, and expressed as mean ± SEM. B: Relative expression levels of brown adipocyte markers: UCP1, UCP2, UCP3, and PGC1α in cultured brown adipocytes infected with Ad-Cre or Ad-GFP. C: mRNA levels of white adipocyte marker: Agt, Psat, and Ednra. Levels of mRNA expression were normalized to internal control β-actin and expressed as mean ± SEM. *P < 0.05, **P < 0.01 (n = 3–6).

Close modal

Rapamycin Reversed the Brown-to-White Adipocyte Phenotypic Conversion In Vivo and In Vitro

In order to investigate whether the effect of mTORC1 activation on the brown-to-white adipocyte phenotypic conversion is reversible, we used rapamycin, a selective inhibitor of mTORC1 signaling. Pregnant mice were randomly divided into two groups: control and treatment. At the near-term pregnancy, pregnant mice were intramuscularly injected with rapamycin at a dose of 1 mg/kg body weight/day or vehicle for 3 consecutive days before the birth of pups. As shown in Fig. 5A, activation of mTORC1 signaling measured by increase in the phosphorylation of S6K1 (Thr389) and S6 (Ser235/236) was detected in the BAT from Fabp4-Tsc1−/− mice treated with control vehicle, whereas rapamycin treatment completely reversed the increase in the mTORC1 signaling. Furthermore, both the decrease in the brown adipocyte markers UCP1, UCP3, and PGC1α and the increase in the white adipocyte markers Agt, Psat, and Ednra in the BAT of Fabp4-Tsc1−/− mice were reversed by maternal administration of rapamycin (Fig. 5B and C).

Figure 5

Administration of rapamycin rescued the brown-to-white adipocyte phenotypic switch in vivo and in vitro. BAT mRNAs from the Fabp4-Tsc1−/− mice (KO) and WT littermates treated with DMSO (as control) or rapamycin were extracted and analyzed as described in 2research design and methods. Brown adipocytes were cultured and infected with Ad-Cre or Ad-GFP with or without rapamycin treatment. Western blotting was performed to detect levels of p-S6K1 (Thr389) and p-S6 (Ser235/236). Real-time PCR was performed to evaluate the expression of brown and white adipocyte markers. β-Actin was used as the loading control. A: Rapamycin reversed the activation of mTORC1 signaling in Fabp4-Tsc1−/− mice (KO). Maternal administration of rapamycin blocked the downregulation of brown adipocyte transcripts UCP1, UCP3, and PGC1α (B) and the upregulation of white adipocyte–typical genes Agt, Psat, and Ednra (C) in the Fabp4-Tsc1−/− mice (KO). D: Rapamycin reversed the activation of mTORC1 signaling in the primary brown adipocytes isolated from Tsc1lox/lox mice infected with Ad-Cre adenoviruses. Rapamycin blocked the downregulation of brown adipocyte transcripts (E) and the upregulation of white adipocyte–typical genes (F) in the primary brown adipocytes isolated from Tsc1lox/lox mice infected with Ad-Cre adenoviruses. Results were normalized to the internal controls β-actin and expressed as mean ± SEM. *P < 0.05, **P < 0.01 in comparison with control; #P < 0.05, ##P < 0.01, relative to rapamycin treatment alone (n = 6).

Figure 5

Administration of rapamycin rescued the brown-to-white adipocyte phenotypic switch in vivo and in vitro. BAT mRNAs from the Fabp4-Tsc1−/− mice (KO) and WT littermates treated with DMSO (as control) or rapamycin were extracted and analyzed as described in 2research design and methods. Brown adipocytes were cultured and infected with Ad-Cre or Ad-GFP with or without rapamycin treatment. Western blotting was performed to detect levels of p-S6K1 (Thr389) and p-S6 (Ser235/236). Real-time PCR was performed to evaluate the expression of brown and white adipocyte markers. β-Actin was used as the loading control. A: Rapamycin reversed the activation of mTORC1 signaling in Fabp4-Tsc1−/− mice (KO). Maternal administration of rapamycin blocked the downregulation of brown adipocyte transcripts UCP1, UCP3, and PGC1α (B) and the upregulation of white adipocyte–typical genes Agt, Psat, and Ednra (C) in the Fabp4-Tsc1−/− mice (KO). D: Rapamycin reversed the activation of mTORC1 signaling in the primary brown adipocytes isolated from Tsc1lox/lox mice infected with Ad-Cre adenoviruses. Rapamycin blocked the downregulation of brown adipocyte transcripts (E) and the upregulation of white adipocyte–typical genes (F) in the primary brown adipocytes isolated from Tsc1lox/lox mice infected with Ad-Cre adenoviruses. Results were normalized to the internal controls β-actin and expressed as mean ± SEM. *P < 0.05, **P < 0.01 in comparison with control; #P < 0.05, ##P < 0.01, relative to rapamycin treatment alone (n = 6).

Close modal

To confirm the specific effect of Tsc1-mTORC1 signaling on BAT, we then infected the primary brown adipocytes isolated from Tsc1lox/lox mice with Ad-Cre and Ad-GFP. Cells were then treated with rapamycin to block the activation of mTORC1 signaling induced by deletion of Tsc1 (Fig. 5D). As shown in Fig. 5E and F, treatment of cells with rapamycin reversed the downregulation of BAT markers and the upregulation of WAT markers.

These data confirm that rapamycin is able to rescue the brown-to-white adipocyte phenotypic switch induced by deletion of Tsc1 gene in the BAT both in mice and cultured brown adipocytes.

Alteration in Levels of FoxC2/Rb, p107, and RIP140

We next investigated the effect of mTORC1 activation on the transcriptional factors related to the fate determination of BAT and WAT. The mRNA level of FoxC2, which decides the fate of BAT, decreased in the BAT from Fabp4-Tsc1−/− mice, while treatment with rapamycin attenuated the downregulation of FoxC2 (Fig. 6A). In contrast, mRNA levels of Rb, p107, and RIP140, which inhibit the differentiation of BAT, were significantly upregulated relative to WT littermates (Fig. 6A). Again, rapamycin reversed the increase in the mRNA levels of Rb (Fig. 6A). Similar effects were observed in cultured brown adipocytes in which mTORC1 signaling was activated by removal of Tsc1 gene using the Cre recombinase. In comparison with control cells infected with GFP Ad, removal of the Tsc1 gene in brown adipocytes resulted in a significant decrease in FoxC2 and a marked increase in Rb, p107, and RIP140 mRNA levels (Fig. 6B). Removal of Tsc1 by Cre recombinase activated the mTORC1 signaling as measured by increase in the phosphorylation of mTOR, S6K1, and S6 (Fig. 4A). These results further demonstrate that activation of mTORC1 signaling by deletion of Tsc1 gene renders the brown adipocytes more prone to convert into white adipocytes.

Figure 6

Alteration in levels of transcriptional factors: FoxC2, Rb, p107, and RIP140. Activation of mTORC1 signaling was achieved by deletion of TSC1 in cultured brown adipocytes as described in 2research design and methods. The mRNA was extracted from either the BAT harvested from the Fabp4-Tsc1−/− mice (KO) and WT littermates or the cultured brown adipocytes infected with Ad-Cre or Ad-GFP. Real-time PCR was performed to evaluate the expression of the related transcription factors, and results were normalized to internal control β-actin and expressed as mean ± SEM. Effects of mTORC1 activation on mRNA levels of transcriptional factors FoxC2, Rb, p107, and RIP140 in the BAT and in cultured brown adipocytes were shown in A and B, respectively. *P < 0.05, **P < 0.01 compared with WT mice treated with DMSO (n = 6). #P < 0.05, ##P < 0.01 compared with KO mice treated with DMSO (n = 6).

Figure 6

Alteration in levels of transcriptional factors: FoxC2, Rb, p107, and RIP140. Activation of mTORC1 signaling was achieved by deletion of TSC1 in cultured brown adipocytes as described in 2research design and methods. The mRNA was extracted from either the BAT harvested from the Fabp4-Tsc1−/− mice (KO) and WT littermates or the cultured brown adipocytes infected with Ad-Cre or Ad-GFP. Real-time PCR was performed to evaluate the expression of the related transcription factors, and results were normalized to internal control β-actin and expressed as mean ± SEM. Effects of mTORC1 activation on mRNA levels of transcriptional factors FoxC2, Rb, p107, and RIP140 in the BAT and in cultured brown adipocytes were shown in A and B, respectively. *P < 0.05, **P < 0.01 compared with WT mice treated with DMSO (n = 6). #P < 0.05, ##P < 0.01 compared with KO mice treated with DMSO (n = 6).

Close modal

Our present study demonstrates that TSC1-mTORC1 signaling regulates the conversion of brown adipocytes into cells with morphological and gene properties of white adipocytes. This conclusion is supported by the following observations: 1) deletion of TSC1 enhanced mTORC1 activity in BAT of Fabp4-Tsc1−/− mice; 2) cells with morphological properties of white adipocytes increased markedly in the BAT from Fabp4-Tsc1−/− mice; 3) white adipocyte typical transcripts increased significantly, whereas brown adipocyte typical genes reduced markedly in the BAT of Fabp4-Tsc1−/− mice; 4) deletion of Tsc1 gene in cultured brown adipocytes induced the conversion into cells with gene properties of white adipocytes; 5) rapamycin rescued the brown-to-white adipocyte phenotypic switch both in the BAT of FABP4-Tsc1−/− mice and in cultured brown adipocytes; and 6) activation of mTORC1 signaling altered the mRNA levels of FoxC2, Rb, p107, and RIP140 both in mice and cultured brown adipocytes, an effect reversible by treatment with rapamycin.

Evidence suggesting the interconversion between brown and white adipocytes has been emerging. Because of its potential as a promising strategy to counteract obesity by promoting energy expenditure, the conversion of white to brown-like adipocytes has been the focus of studies. Typically, browning involves the appearance of ML adipocyte clusters that are dispersed among unilocular WAT. The browning phenomenon varies among WAT depots and is strongly influenced by synergy between signaling and transcription factors that vary depending upon the environmental conditions. Whether brown adipocytes can directly convert into white adipocytes remain under debate. In an early tracing study, no direct switch from predominantly BAT to predominantly WAT in mice was observed during development (32). In contrast, our present studies provide evidence supporting the phenotypic conversion of brown adipocytes into cells with morphological and gene properties of white fat cells. Several previous reports also support the potential conversion from brown to white adipocytes. Studies in humans have found detectable BAT in up to 96% of healthy young adults but less often in older or obese subjects, suggesting that human adipocytes might convert from a brown into a white adipocyte phenotype during aging. Using immunoelectron microscopic analysis to trace the alteration of UCP in mitochondria, convertible adipose tissue was first identified in mice by Loncar (33). In response to cold stress, cells with morphological properties of brown adipocytes appear in inguinal fat mass. Further examination demonstrates that these brownlike adipocytes regain the properties of white adipocytes upon rewarming the animal to 28°C. Perhaps the most convincing evidence comes from the study by Rosenwald et al. (34). Using the lineage tracing approach in Ucp1-GFP mice, they demonstrate that cold-induced brown like cells are converted into cells with the morphology and gene expression pattern of white adipocytes. Moreover, these white-typical adipocytes can convert into brownlike adipocytes on additional cold stimulation. All of these studies suggest the interconversion between brown and white adipocytes. Our studies further support this concept. However, it is worth of noting that our study cannot exclude the contribution of mTOR signaling in other metabolically critical organs because FABP4 is not exclusively expressed in BAT and WAT (26,35,36). While our in vitro studies suggest that mTOR signaling may determines the interconversion between brown and white adipocytes, a portion of the whitening of brown fat in Fabp4-Tsc1−/− transgenic mice could result from increased mTORC1 function in tissue other than adipose tissues such as brain, liver, and skeletal muscles.

Decrease of BAT in obese subjects indicates that long-term excess energy may be accountable for the loss of brown adipocytes, which are likely converted into the white fat cells based on previous observations described above. The molecular mechanism by which excess energy signals brown adipocytes to initiate the conversion remains unknown. The current study supports the concept that mTORC1 signaling is critical for the conversion of brown adipocytes into cells with white adipocyte properties. Activation of mTORC1 signaling may signal the brown adipocytes the oversupply of energy and therefore initiate the whitening of the BAT. This concept is consistent with previous studies demonstrating that mTORC1 can sense nutrient availability to regulate the differentiation of white adipocytes. The mTORC1 signaling has been reported to specifically regulate the activity of peroxisome proliferator–activated receptor-β, which is essential for the adipogenic gene expression program and thus critical for both initiating and maintaining adipogenesis. In contrast, inhibition of mTORC1 with rapamycin significantly reduces the expression of most adipocyte marker genes and the accumulation of intracellular lipid in white adipocytes.

Appearance of white adipocytes in the brown fat depots may also result from the switch of the progenitor cells to differentiate into white adipocytes. Several recent reports have identified adipocyte progenitor cells in adult adipose tissues that can give rise to adipocytes with either brown or white characteristics (3740). Our data cannot exclude the contribution of precursor cells to the formation of white adipocytes in BAT. The appearance of large amount of cells with morphological and gene properties of white adipocytes supports the concept based on previous studies that differentiation of additional white adipocytes de novo from precursors may occur when the interconversion capacity is insufficient.

In summary, our study demonstrates that activation of mTORC1 signaling stimulates the conversion of brown to white adipocytes. If inhibition of mTORC1 signaling is able to block the conversion of brown to white adipocytes, it might be a promising strategy to target the mTORC1 signaling to increase the number of lipid-burning cells to counteract obesity by promoting energy expenditure.

Funding. This work was supported by grants from the National Natural Science Foundation of China (81330010, 81390354, 81370962, 81170795, and 81030012) and American Diabetes Associationhttp://dx.doi.org/10.13039/100001440 grant 1-13-BS-225.

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. X.X., Y.L., and W.Z. contributed to the design of the experiments, interpretation of data, and the writing of the manuscript; performed the experiments; and approved the final version of the manuscript. H.L., H.T., F.Y., Y.X., and J.Z. performed the experiments, analyzed data, and approved the final version of the manuscript. W.Z. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

1.
Cinti
S
.
Adipocyte differentiation and transdifferentiation: plasticity of the adipose organ
.
J Endocrinol Invest
2002
;
25
:
823
835
[PubMed]
2.
Cannon
B
,
Nedergaard
J
.
Brown adipose tissue: function and physiological significance
.
Physiol Rev
2004
;
84
:
277
359
[PubMed]
3.
Cinti
S
.
The adipose organ
.
Prostaglandins Leukot Essent Fatty Acids
2005
;
73
:
9
15
[PubMed]
4.
Cinti
S
.
Transdifferentiation properties of adipocytes in the adipose organ
.
Am J Physiol Endocrinol Metab
2009
;
297
:
E977
E986
[PubMed]
5.
Enerbäck
S
,
Jacobsson
A
,
Simpson
EM
, et al
.
Mice lacking mitochondrial uncoupling protein are cold-sensitive but not obese
.
Nature
1997
;
387
:
90
94
[PubMed]
6.
Karamitri
A
,
Shore
AM
,
Docherty
K
,
Speakman
JR
,
Lomax
MA
.
Combinatorial transcription factor regulation of the cyclic AMP-response element on the Pgc-1alpha promoter in white 3T3-L1 and brown HIB-1B preadipocytes
.
J Biol Chem
2009
;
284
:
20738
20752
[PubMed]
7.
Puigserver
P
,
Wu
Z
,
Park
CW
,
Graves
R
,
Wright
M
,
Spiegelman
BM
.
A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis
.
Cell
1998
;
92
:
829
839
[PubMed]
8.
Manchado
C
,
Yubero
P
,
Viñas
O
, et al
.
CCAAT/enhancer-binding proteins alpha and beta in brown adipose tissue: evidence for a tissue-specific pattern of expression during development
.
Biochem J
1994
;
302
:
695
700
[PubMed]
9.
Tsukiyama-Kohara
K
,
Poulin
F
,
Kohara
M
, et al
.
Adipose tissue reduction in mice lacking the translational inhibitor 4E-BP1
.
Nat Med
2001
;
7
:
1128
1132
[PubMed]
10.
Picard
F
,
Géhin
M
,
Annicotte
J
, et al
.
SRC-1 and TIF2 control energy balance between white and brown adipose tissues
.
Cell
2002
;
111
:
931
941
[PubMed]
11.
Hansen
JB
,
Jørgensen
C
,
Petersen
RK
, et al
.
Retinoblastoma protein functions as a molecular switch determining white versus brown adipocyte differentiation
.
Proc Natl Acad Sci U S A
2004
;
101
:
4112
4117
[PubMed]
12.
Leonardsson
G
,
Steel
JH
,
Christian
M
, et al
.
Nuclear receptor corepressor RIP140 regulates fat accumulation
.
Proc Natl Acad Sci U S A
2004
;
101
:
8437
8442
[PubMed]
13.
Scimè
A
,
Grenier
G
,
Huh
MS
, et al
.
Rb and p107 regulate preadipocyte differentiation into white versus brown fat through repression of PGC-1alpha
.
Cell Metab
2005
;
2
:
283
295
[PubMed]
14.
Hansen
JB
,
Kristiansen
K
.
Regulatory circuits controlling white versus brown adipocyte differentiation
.
Biochem J
2006
;
398
:
153
168
[PubMed]
15.
Powelka
AM
,
Seth
A
,
Virbasius
JV
, et al
.
Suppression of oxidative metabolism and mitochondrial biogenesis by the transcriptional corepressor RIP140 in mouse adipocytes
.
J Clin Invest
2006
;
116
:
125
136
[PubMed]
16.
Cederberg
A
,
Grønning
LM
,
Ahrén
B
,
Taskén
K
,
Carlsson
P
,
Enerbäck
S
.
FOXC2 is a winged helix gene that counteracts obesity, hypertriglyceridemia, and diet-induced insulin resistance
.
Cell
2001
;
106
:
563
573
[PubMed]
17.
Holz
MK
,
Ballif
BA
,
Gygi
SP
,
Blenis
J
.
mTOR and S6K1 mediate assembly of the translation preinitiation complex through dynamic protein interchange and ordered phosphorylation events
.
Cell
2005
;
123
:
569
580
[PubMed]
18.
Huang
J
,
Dibble
CC
,
Matsuzaki
M
,
Manning
BD
.
The TSC1-TSC2 complex is required for proper activation of mTOR complex 2
.
Mol Cell Biol
2008
;
28
:
4104
4115
[PubMed]
19.
Fox
HL
,
Kimball
SR
,
Jefferson
LS
,
Lynch
CJ
.
Amino acids stimulate phosphorylation of p70S6k and organization of rat adipocytes into multicellular clusters
.
Am J Physiol
1998
;
274
:
C206
C213
[PubMed]
20.
Kim
JE
,
Chen
J
.
regulation of peroxisome proliferator-activated receptor-gamma activity by mammalian target of rapamycin and amino acids in adipogenesis
.
Diabetes
2004
;
53
:
2748
2756
[PubMed]
21.
Roh
C
,
Han
J
,
Tzatsos
A
,
Kandror
KV
.
Nutrient-sensing mTOR-mediated pathway regulates leptin production in isolated rat adipocytes
.
Am J Physiol Endocrinol Metab
2003
;
284
:
E322
E330
[PubMed]
22.
El-Chaâr
D
,
Gagnon
A
,
Sorisky
A
.
Inhibition of insulin signaling and adipogenesis by rapamycin: effect on phosphorylation of p70 S6 kinase vs eIF4E-BP1
.
Int J Obes Relat Metab Disord
2004
;
28
:
191
198
[PubMed]
23.
Bell
A
,
Grunder
L
,
Sorisky
A
.
Rapamycin inhibits human adipocyte differentiation in primary culture
.
Obes Res
2000
;
8
:
249
254
[PubMed]
24.
Yeh
WC
,
Bierer
BE
,
McKnight
SL
.
Rapamycin inhibits clonal expansion and adipogenic differentiation of 3T3-L1 cells
.
Proc Natl Acad Sci U S A
1995
;
92
:
11086
11090
[PubMed]
25.
Inoki
K
,
Corradetti
MN
,
Guan
KL
.
Dysregulation of the TSC-mTOR pathway in human disease
.
Nat Genet
2005
;
37
:
19
24
[PubMed]
26.
Xiang
X
,
Yuan
F
,
Zhao
J
, et al
.
Deficiency in pulmonary surfactant proteins in mice with fatty acid binding protein 4-Cre-mediated knockout of the tuberous sclerosis complex 1 gene
.
Exp Physiol
2013
;
98
:
830
841
[PubMed]
27.
Teruel
T
,
Hernandez
R
,
Lorenzo
M
.
Ceramide mediates insulin resistance by tumor necrosis factor-alpha in brown adipocytes by maintaining Akt in an inactive dephosphorylated state
.
Diabetes
2001
;
50
:
2563
2571
[PubMed]
28.
Wang
N
,
Verna
L
,
Chen
NG
, et al
.
Constitutive activation of peroxisome proliferator-activated receptor-gamma suppresses pro-inflammatory adhesion molecules in human vascular endothelial cells
.
J Biol Chem
2002
;
277
:
34176
34181
[PubMed]
29.
Xu
G
,
Li
Y
,
An
W
, et al
.
Gastric mammalian target of rapamycin signaling regulates ghrelin production and food intake
.
Endocrinology
2009
;
150
:
3637
3644
[PubMed]
30.
Li
Y
,
Jiang
C
,
Xu
G
, et al
.
Homocysteine upregulates resistin production from adipocytes in vivo and in vitro
.
Diabetes
2008
;
57
:
817
827
[PubMed]
31.
Kajimura
S
,
Seale
P
,
Tomaru
T
, et al
.
Regulation of the brown and white fat gene programs through a PRDM16/CtBP transcriptional complex
.
Genes Dev
2008
;
22
:
1397
1409
[PubMed]
32.
Moulin
K
,
Truel
N
,
André
M
, et al
.
Emergence during development of the white-adipocyte cell phenotype is independent of the brown-adipocyte cell phenotype
.
Biochem J
2001
;
356
:
659
664
[PubMed]
33.
Loncar
D
.
Convertible adipose tissue in mice
.
Cell Tissue Res
1991
;
266
:
149
161
[PubMed]
34.
Rosenwald
M
,
Perdikari
A
,
Rülicke
T
,
Wolfrum
C
.
Bi-directional interconversion of brite and white adipocytes
.
Nat Cell Biol
2013
;
15
:
659
667
[PubMed]
35.
Martens
K
,
Bottelbergs
A
,
Baes
M
.
Ectopic recombination in the central and peripheral nervous system by aP2/FABP4-Cre mice: implications for metabolism research
.
FEBS Lett
2010
;
584
:
1054
1058
[PubMed]
36.
Lee
KY
,
Russell
SJ
,
Ussar
S
, et al
.
Lessons on conditional gene targeting in mouse adipose tissue
.
Diabetes
2013
;
62
:
864
874
[PubMed]
37.
Schulz
TJ
,
Huang
TL
,
Tran
TT
, et al
.
Identification of inducible brown adipocyte progenitors residing in skeletal muscle and white fat
.
Proc Natl Acad Sci U S A
2011
;
108
:
143
148
[PubMed]
38.
Lee
YH
,
Petkova
AP
,
Mottillo
EP
,
Granneman
JG
.
In vivo identification of bipotential adipocyte progenitors recruited by β3-adrenoceptor activation and high-fat feeding
.
Cell Metab
2012
;
15
:
480
491
[PubMed]
39.
Rodeheffer
MS
,
Birsoy
K
,
Friedman
JM
.
Identification of white adipocyte progenitor cells in vivo
.
Cell
2008
;
135
:
240
249
[PubMed]
40.
Berry
R
,
Rodeheffer
MS
.
Characterization of the adipocyte cellular lineage in vivo
.
Nat Cell Biol
2013
;
15
:
302
308
[PubMed]