Comprehensive Physiology Wiley Online Library

Transcriptional Regulation of Adipogenesis

Full Article on Wiley Online Library



ABSTRACT

Adipocytes are the defining cell type of adipose tissue. Once considered a passive participant in energy storage, adipose tissue is now recognized as a dynamic organ that contributes to several important physiological processes, such as lipid metabolism, systemic energy homeostasis, and whole‐body insulin sensitivity. Therefore, understanding the mechanisms involved in its development and function is of great importance. Adipocyte differentiation is a highly orchestrated process which can vary between different fat depots as well as between the sexes. While hormones, miRNAs, cytoskeletal proteins, and many other effectors can modulate adipocyte development, the best understood regulators of adipogenesis are the transcription factors that inhibit or promote this process. Ectopic expression and knockdown approaches in cultured cells have been widely used to understand the contribution of transcription factors to adipocyte development, providing a basis for more sophisticated in vivo strategies to examine adipogenesis. To date, over two dozen transcription factors have been shown to play important roles in adipocyte development. These transcription factors belong to several families with many different DNA‐binding domains. While peroxisome proliferator‐activated receptor gamma (PPARγ) is undoubtedly the most important transcriptional modulator of adipocyte development in all types of adipose tissue, members of the CCAAT/enhancer‐binding protein, Krüppel‐like transcription factor, signal transducer and activator of transcription, GATA, early B cell factor, and interferon‐regulatory factor families also regulate adipogenesis. The importance of PPARγ activity is underscored by several covalent modifications that modulate its activity and its ability to modulate adipocyte development. This review will primarily focus on the transcriptional control of adipogenesis in white fat cells and on the mechanisms involved in this fine‐tuned developmental process. © 2017 American Physiological Society. Compr Physiol 7:635‐674, 2017.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1. Different shades of adipocytes: white, brown, and beige. White adipocytes comprise the majority of fat cells in mice and men. The primary function of white adipocytes is lipid storage. Brown adipocytes largely function to produce heat. Beige adipocytes have properties similar to both brown and white adipocytes. There is evidence that each type of fat cell comes from a different type of preadipocyte. However, there is also evidence that beige adipocytes result from the transdifferentiation of white adipocytes. Although the characterization of each cell type is well understood, the developmental origins of these different types of cells from preadipocytes and their adipocyte progenitor cells is an emerging area of research.
Figure 2. Figure 2. In vitro studies of adipocyte development. Preadipocytes are cells capable of differentiating into adipocytes with the appropriate hormonal cocktail, which varies for adipogenesis of brown, white, or beige preadipocytes. Examples of preadipocytes include 3T3‐L1 and 3T3‐F442A cells. Nonprecursor cells, fibroblasts and other cell types, do not undergo adipogenesis in the presence of the appropriate hormonal cocktail. Examples of nonprecursor cells include NIH 3T3, Swiss 3T3, and Balb/c fibroblasts. Nonprecursor cells can undergo adipogenesis with ectopic expression of PPARγ or other transcription factors.
Figure 3. Figure 3. Structural motifs of DBD s in transcription factors that regulate adipocyte development. Transcription factors have at least one DBD that mediates their capacity to bind to DNA and regulate transcription. Leucine zipper is a dimerization domain of the bZIP class that contains periodic repetition of leucine residues, which coordinates its ability to bind to DNA; C/EBPs and AP‐1 proteins are examples of leucine zipper proteins. In the zinc‐finger structure, a zinc ion is coordinated by a combination of cysteine and histidine residues. Zinc fingers arise from multiple different folding arrangements (“fold groups”). The classic Cys2His2 zinc finger motif is shown. KLFs, GATAs, and Zfps, as well as all of the nuclear receptors are examples of zinc‐finger proteins. Helix‐loop‐helix is characterized by two α‐helices connected by a loop. SREBP and EBFs are helix‐loop‐helix transcription factors. Helix‐turn‐helix is characterized by two α‐helices joined by a short strand of amino acids and is found in IRFs that regulate adipogenesis. Some adipocyte transcription factors such as KLFs and SREBPs contain more than one structural motif to facilitate DNA binding.
Figure 4. Figure 4. Transcription factors that promote adipogenesis. Many transcription factors are induced during adipocyte differentiation. Some of these, like members of the AP‐1 family, are induced during clonal expansion. Others, like PPARγ, have been shown to promote adipocyte differentiation in vitro and in vivo. The relative timing of the induction of each transcription factor is indicated.
Figure 5. Figure 5. Transcription factors that inhibit adipogenesis. Many specific transcription factors have been shown to inhibit adipocyte development. The expression of most of these transcription factors decreases with adipocyte development. Ectopic expression and knockdown approaches have been used to demonstrate the inhibitory actions of these transcription factors. The relative timing of the action of each transcription factor is indicated.
Figure 6. Figure 6. PPARγ exists in two isoforms, γ1 and γ2. PPARγ proteins are members of the large nuclear hormone receptor superfamily of transcription factors that also includes steroid receptors and TRs. PPARγ1 has 30 fewer amino acids in the N‐terminal region than PPARγ2. The covalent modifications are indicated for both PPARγ proteins. Unlike the other modifications, SUMOylation at lysine 365 in the ligand‐binding domain of PPARγ1 (lysine 395 of PPARγ2) and phosphorylation at serine 243 (serine 273 of PPARγ2) do not appear to play a role in adipocyte development.
Figure 7. Figure 7. Covalent modifications of PPARγ1 and PPARγ2 proteins that modulate PPARγ activity and/or adipocyte development. Serine phosphorylation at residue 112 was the first covalent modification shown to regulate and inhibit PPARγ activity. Since then, several other modifications have been identified. Some of these modifications are closely spaced, such as SUMOylation and phosphorylation. The multiple inhibitory covalent modifications of PPARγ underscore the importance of regulating the activity of this highly adipogenic transcription factor.
Figure 8. Figure 8. Covalent modifications of PPARγ2 that are associated with decreased transcriptional activity and inhibition of adipogenesis. The majority of PPARγ covalent modifications are associated with decreased transcriptional activity and inhibition of adipogenesis. The proposed O‐GlcNAcylation site at threonine84 is based on studies of threonine54 in PPARγ1. All of the other sites shown in this figure have been confirmed by in vitro experiments. To date, only serine112 has been studied in a rigorous manner in vivo.
Figure 9. Figure 9. Covalent modifications of PPARγ proteins that correlate with increased transcriptional activity. Both PPARγ1 and PPARγ2 are highly labile proteins that are targeted to the UPP for degradation. Following ligand binding, there is evidence that ubiquitylation of PPARγ proteins occurs in the ligand‐binding domain when PPARγ is transcriptionally active, and inhibition of proteasome activity is accompanied by increased PPARγ ubiquitylation and sustained transcriptional activity. There is also evidence that PPARγ transcriptional activity is activated by acetylation of lysine293 of PPARγ2 (lysine263 of PPARγ1).
Figure 10. Figure 10. The role of C/EBP proteins in adipogenesis. Both C/EBPβ and C/EBPδ are induced early during adipogenesis. C/EBPβ is highly responsive to induction by MIX, a component of the hormonal induction cocktail that increases cAMP signaling. C/EBPδ is activated by DEX, a synthetic GC that promotes adipogenesis. These C/EBP proteins increase the expression of both PPARγ and C/EBPα. Both PPARγ and C/EBPα can regulate the expression of one another. In addition, C/EBPβ is responsible for Wnt/β‐catenin inhibition and regulation of MCE, a process in which growth‐arrested preadipocytes re‐enter the cell cycle before they differentiate into adipocytes.
Figure 11. Figure 11. Wnt/β‐Catenin signaling pathway and inhibition of adipocyte development. Wnt agonists such as Wnt6/10a/10b, NELL1, and Grem2 bind to Frizzled receptors and LRP coreceptors and act through the intracellular protein Dvl, blocking the activity of GSK3β (glycogen synthase kinase‐3) and promoting cytosolic accumulation of β‐catenin, which then translocates to the nucleus and binds to the promoter region of the adipogenic genes PPARγ and C/EBPα, repressing their transcription and, consequently, adipogenesis. WISP2, when secreted, can activate the canonical Wnt pathway and attenuate PPARγ activation; it can also act in the cytosol, where it interacts with ZFP423, preventing it from translocating to the nucleus and activating PPARγ expression.
Figure 12. Figure 12. Wnt/β‐Catenin signaling pathway and activation of adipocyte development. Wnt antagonists such as DKK1, sFRP1/4, XBP1, TREM2, and NRX bind to Frizzled receptors and LRP coreceptors, inactivating the cytosolic protein Dvl. GSK3β is then active and phosphorylates β‐catenin which is thereby targeted for ubiquitylation and subsequent degradation by the UPP. In addition, when BMP4 is present, ZFP423 dissociates from WISP2 and can then translocate to the nucleus and interact with the PPARγ promoter to enhance its expression and promote adipocyte differentiation.
Figure 13. Figure 13. TGF‐β and BMPs act via Smads to regulate adipogenesis. BMP2 and 4 bind to their serine/threonine kinase receptors I and recruit type II receptors, which then transphosphorylate the type I receptors. The activated heterotetrameric complexes phosphorylate R‐Smads (1/5/8), which associate with co‐Smad (Smad4). The Smad complex associates with Shn‐2, translocates to the nucleus, and binds with C/EBPα to induce PPARγ expression. Conversely, PPARγ expression can be inhibited by Tob2, which prevents the Smad complex from translocating into the nucleus and can also block C/EBPα from binding to the PPARγ promoter in the nucleus. Additionally, TGF‐β binds to its serine/threonine kinase receptors II that recruit and transphosphorylate type I receptors. Activated heterotetrameric complexes phosphorylate R‐Smads (2/3), which associate with co‐Smad (Smad4). The Smad complexes then translocate to the nucleus and inhibit PPARγ expression.
Figure 14. Figure 14. Homology of nuclear receptors that modulate adipocyte development. Steroid hormone receptors are transcription factors that bind to DNA and regulate the transcriptional activity of many genes. All of these proteins, like PPARγ, are part of the nuclear receptor superfamily and share the same domains types. The A/B domain located in the N‐terminal part of the protein possesses a ligand‐independent transactivation function (AF‐1). DNA binding occurs at the C domain, the most conserved part of steroid receptors, as well as at the D domain located immediately downstream. The E domain is where the ligand binds, and it can also contribute to receptor dimerization. Some receptors such as ER also have an F domain.
Figure 15. Figure 15. The effect of steroid hormones on adipogenesis. (A) Sex Steroids. Estrogen (E) and the primary androgen, testosterone, enter preadipocytes by diffusing through the plasma membrane. Testosterone is converted to DHT, which has a high affinity for the AR. Estrogen binds to its receptor (ER), forming a homodimer, which translocates to the nucleus and binds EREs in a number of genes. Similarly, DHT bound to AR forms a homodimer that binds AREs in a variety of genes. The majority of the evidence suggests that ER and AR inhibit adipogenesis. (B) Adrenal steroids. Cortisol, the primary GC, and aldosterone, the primary mineralocorticoid (MC), diffuse through the plasma membrane. GC/GR homodimers translocate to the nucleus where they modulate the expression of many genes that have glucocorticoid response elements (GREs). Lipin 1 and KLF15 are GC/GR target genes that are induced during adipogenesis. It is well known that both GR and mineralocorticoid receptor (MR) are expressed in preadipocytes. However, recent studies demonstrate that the levels of GR are several hundred fold higher than MR, and loss of MR does not impact adipogenesis, suggesting that actions of MC on adipogenesis are mediated by GR.
Figure 16. Figure 16. Homology of ER and ERRs. Some of the members of nuclear receptor superfamily are considered “orphans” since no ligands have been discovered. Three of these orphan receptors are named ERRs because they have 68% homology with ER in the DBD (domain C). Since these receptors have only 36% homology in the ligand‐binding domain (domain E), they cannot bind estrogen, but they can bind to the same DNA response elements as estrogen receptors. Notably, the majority of studies demonstrate that ER and ERR have opposite effects on adipogenesis.
Figure 17. Figure 17. The activation of TRs promotes adipogenesis. Although TRs have similarities to steroid hormones, there are a couple of key differences in the pathway. First, thyroid hormone does not diffuse through the plasma membrane, but requires a transport protein such as monocarboxylate transporter 8 (MCT8). Another difference is that un‐liganded TRs bind to DNA at thyroid response elements (TRE) and repress transcription. When thyroid hormone, T3, is present, it is transported through the membrane, travels through the cytosol to the nucleus where it binds its receptor (TR) and activates gene expression. Like PPARγ, TRs dimerize with RXRα and its associated ligand, retinoic acid (RA). In preadipocytes, TRs promote differentiation, particularly to beige and brown adipocytes.


Figure 1. Different shades of adipocytes: white, brown, and beige. White adipocytes comprise the majority of fat cells in mice and men. The primary function of white adipocytes is lipid storage. Brown adipocytes largely function to produce heat. Beige adipocytes have properties similar to both brown and white adipocytes. There is evidence that each type of fat cell comes from a different type of preadipocyte. However, there is also evidence that beige adipocytes result from the transdifferentiation of white adipocytes. Although the characterization of each cell type is well understood, the developmental origins of these different types of cells from preadipocytes and their adipocyte progenitor cells is an emerging area of research.


Figure 2. In vitro studies of adipocyte development. Preadipocytes are cells capable of differentiating into adipocytes with the appropriate hormonal cocktail, which varies for adipogenesis of brown, white, or beige preadipocytes. Examples of preadipocytes include 3T3‐L1 and 3T3‐F442A cells. Nonprecursor cells, fibroblasts and other cell types, do not undergo adipogenesis in the presence of the appropriate hormonal cocktail. Examples of nonprecursor cells include NIH 3T3, Swiss 3T3, and Balb/c fibroblasts. Nonprecursor cells can undergo adipogenesis with ectopic expression of PPARγ or other transcription factors.


Figure 3. Structural motifs of DBD s in transcription factors that regulate adipocyte development. Transcription factors have at least one DBD that mediates their capacity to bind to DNA and regulate transcription. Leucine zipper is a dimerization domain of the bZIP class that contains periodic repetition of leucine residues, which coordinates its ability to bind to DNA; C/EBPs and AP‐1 proteins are examples of leucine zipper proteins. In the zinc‐finger structure, a zinc ion is coordinated by a combination of cysteine and histidine residues. Zinc fingers arise from multiple different folding arrangements (“fold groups”). The classic Cys2His2 zinc finger motif is shown. KLFs, GATAs, and Zfps, as well as all of the nuclear receptors are examples of zinc‐finger proteins. Helix‐loop‐helix is characterized by two α‐helices connected by a loop. SREBP and EBFs are helix‐loop‐helix transcription factors. Helix‐turn‐helix is characterized by two α‐helices joined by a short strand of amino acids and is found in IRFs that regulate adipogenesis. Some adipocyte transcription factors such as KLFs and SREBPs contain more than one structural motif to facilitate DNA binding.


Figure 4. Transcription factors that promote adipogenesis. Many transcription factors are induced during adipocyte differentiation. Some of these, like members of the AP‐1 family, are induced during clonal expansion. Others, like PPARγ, have been shown to promote adipocyte differentiation in vitro and in vivo. The relative timing of the induction of each transcription factor is indicated.


Figure 5. Transcription factors that inhibit adipogenesis. Many specific transcription factors have been shown to inhibit adipocyte development. The expression of most of these transcription factors decreases with adipocyte development. Ectopic expression and knockdown approaches have been used to demonstrate the inhibitory actions of these transcription factors. The relative timing of the action of each transcription factor is indicated.


Figure 6. PPARγ exists in two isoforms, γ1 and γ2. PPARγ proteins are members of the large nuclear hormone receptor superfamily of transcription factors that also includes steroid receptors and TRs. PPARγ1 has 30 fewer amino acids in the N‐terminal region than PPARγ2. The covalent modifications are indicated for both PPARγ proteins. Unlike the other modifications, SUMOylation at lysine 365 in the ligand‐binding domain of PPARγ1 (lysine 395 of PPARγ2) and phosphorylation at serine 243 (serine 273 of PPARγ2) do not appear to play a role in adipocyte development.


Figure 7. Covalent modifications of PPARγ1 and PPARγ2 proteins that modulate PPARγ activity and/or adipocyte development. Serine phosphorylation at residue 112 was the first covalent modification shown to regulate and inhibit PPARγ activity. Since then, several other modifications have been identified. Some of these modifications are closely spaced, such as SUMOylation and phosphorylation. The multiple inhibitory covalent modifications of PPARγ underscore the importance of regulating the activity of this highly adipogenic transcription factor.


Figure 8. Covalent modifications of PPARγ2 that are associated with decreased transcriptional activity and inhibition of adipogenesis. The majority of PPARγ covalent modifications are associated with decreased transcriptional activity and inhibition of adipogenesis. The proposed O‐GlcNAcylation site at threonine84 is based on studies of threonine54 in PPARγ1. All of the other sites shown in this figure have been confirmed by in vitro experiments. To date, only serine112 has been studied in a rigorous manner in vivo.


Figure 9. Covalent modifications of PPARγ proteins that correlate with increased transcriptional activity. Both PPARγ1 and PPARγ2 are highly labile proteins that are targeted to the UPP for degradation. Following ligand binding, there is evidence that ubiquitylation of PPARγ proteins occurs in the ligand‐binding domain when PPARγ is transcriptionally active, and inhibition of proteasome activity is accompanied by increased PPARγ ubiquitylation and sustained transcriptional activity. There is also evidence that PPARγ transcriptional activity is activated by acetylation of lysine293 of PPARγ2 (lysine263 of PPARγ1).


Figure 10. The role of C/EBP proteins in adipogenesis. Both C/EBPβ and C/EBPδ are induced early during adipogenesis. C/EBPβ is highly responsive to induction by MIX, a component of the hormonal induction cocktail that increases cAMP signaling. C/EBPδ is activated by DEX, a synthetic GC that promotes adipogenesis. These C/EBP proteins increase the expression of both PPARγ and C/EBPα. Both PPARγ and C/EBPα can regulate the expression of one another. In addition, C/EBPβ is responsible for Wnt/β‐catenin inhibition and regulation of MCE, a process in which growth‐arrested preadipocytes re‐enter the cell cycle before they differentiate into adipocytes.


Figure 11. Wnt/β‐Catenin signaling pathway and inhibition of adipocyte development. Wnt agonists such as Wnt6/10a/10b, NELL1, and Grem2 bind to Frizzled receptors and LRP coreceptors and act through the intracellular protein Dvl, blocking the activity of GSK3β (glycogen synthase kinase‐3) and promoting cytosolic accumulation of β‐catenin, which then translocates to the nucleus and binds to the promoter region of the adipogenic genes PPARγ and C/EBPα, repressing their transcription and, consequently, adipogenesis. WISP2, when secreted, can activate the canonical Wnt pathway and attenuate PPARγ activation; it can also act in the cytosol, where it interacts with ZFP423, preventing it from translocating to the nucleus and activating PPARγ expression.


Figure 12. Wnt/β‐Catenin signaling pathway and activation of adipocyte development. Wnt antagonists such as DKK1, sFRP1/4, XBP1, TREM2, and NRX bind to Frizzled receptors and LRP coreceptors, inactivating the cytosolic protein Dvl. GSK3β is then active and phosphorylates β‐catenin which is thereby targeted for ubiquitylation and subsequent degradation by the UPP. In addition, when BMP4 is present, ZFP423 dissociates from WISP2 and can then translocate to the nucleus and interact with the PPARγ promoter to enhance its expression and promote adipocyte differentiation.


Figure 13. TGF‐β and BMPs act via Smads to regulate adipogenesis. BMP2 and 4 bind to their serine/threonine kinase receptors I and recruit type II receptors, which then transphosphorylate the type I receptors. The activated heterotetrameric complexes phosphorylate R‐Smads (1/5/8), which associate with co‐Smad (Smad4). The Smad complex associates with Shn‐2, translocates to the nucleus, and binds with C/EBPα to induce PPARγ expression. Conversely, PPARγ expression can be inhibited by Tob2, which prevents the Smad complex from translocating into the nucleus and can also block C/EBPα from binding to the PPARγ promoter in the nucleus. Additionally, TGF‐β binds to its serine/threonine kinase receptors II that recruit and transphosphorylate type I receptors. Activated heterotetrameric complexes phosphorylate R‐Smads (2/3), which associate with co‐Smad (Smad4). The Smad complexes then translocate to the nucleus and inhibit PPARγ expression.


Figure 14. Homology of nuclear receptors that modulate adipocyte development. Steroid hormone receptors are transcription factors that bind to DNA and regulate the transcriptional activity of many genes. All of these proteins, like PPARγ, are part of the nuclear receptor superfamily and share the same domains types. The A/B domain located in the N‐terminal part of the protein possesses a ligand‐independent transactivation function (AF‐1). DNA binding occurs at the C domain, the most conserved part of steroid receptors, as well as at the D domain located immediately downstream. The E domain is where the ligand binds, and it can also contribute to receptor dimerization. Some receptors such as ER also have an F domain.


Figure 15. The effect of steroid hormones on adipogenesis. (A) Sex Steroids. Estrogen (E) and the primary androgen, testosterone, enter preadipocytes by diffusing through the plasma membrane. Testosterone is converted to DHT, which has a high affinity for the AR. Estrogen binds to its receptor (ER), forming a homodimer, which translocates to the nucleus and binds EREs in a number of genes. Similarly, DHT bound to AR forms a homodimer that binds AREs in a variety of genes. The majority of the evidence suggests that ER and AR inhibit adipogenesis. (B) Adrenal steroids. Cortisol, the primary GC, and aldosterone, the primary mineralocorticoid (MC), diffuse through the plasma membrane. GC/GR homodimers translocate to the nucleus where they modulate the expression of many genes that have glucocorticoid response elements (GREs). Lipin 1 and KLF15 are GC/GR target genes that are induced during adipogenesis. It is well known that both GR and mineralocorticoid receptor (MR) are expressed in preadipocytes. However, recent studies demonstrate that the levels of GR are several hundred fold higher than MR, and loss of MR does not impact adipogenesis, suggesting that actions of MC on adipogenesis are mediated by GR.


Figure 16. Homology of ER and ERRs. Some of the members of nuclear receptor superfamily are considered “orphans” since no ligands have been discovered. Three of these orphan receptors are named ERRs because they have 68% homology with ER in the DBD (domain C). Since these receptors have only 36% homology in the ligand‐binding domain (domain E), they cannot bind estrogen, but they can bind to the same DNA response elements as estrogen receptors. Notably, the majority of studies demonstrate that ER and ERR have opposite effects on adipogenesis.


Figure 17. The activation of TRs promotes adipogenesis. Although TRs have similarities to steroid hormones, there are a couple of key differences in the pathway. First, thyroid hormone does not diffuse through the plasma membrane, but requires a transport protein such as monocarboxylate transporter 8 (MCT8). Another difference is that un‐liganded TRs bind to DNA at thyroid response elements (TRE) and repress transcription. When thyroid hormone, T3, is present, it is transported through the membrane, travels through the cytosol to the nucleus where it binds its receptor (TR) and activates gene expression. Like PPARγ, TRs dimerize with RXRα and its associated ligand, retinoic acid (RA). In preadipocytes, TRs promote differentiation, particularly to beige and brown adipocytes.
References
 1.Adams M, Reginato MJ, Shao D, Lazar MA, Chatterjee VK. Transcriptional activation by peroxisome proliferator‐activated receptor gamma is inhibited by phosphorylation at a consensus mitogen‐activated protein kinase site. J Biol Chem 272: 5128‐5132, 1997.
 2.Addison WN, Fu MM, Yang HX, Lin Z, Nagano K, Gori F, Baron R. Direct transcriptional repression of Zfp423 by Zfp521 mediates a bone morphogenic protein‐dependent osteoblast versus adipocyte lineage commitment switch. Mol Cell Biol 34: 3076‐3085, 2014.
 3.Akerblad P, Mansson R, Lagergren A, Westerlund S, Basta B, Lind U, Thelin A, Gisler R, Liberg D, Nelander S, Bamberg K, Sigvardsson M. Gene expression analysis suggests that EBF‐1 and PPARgamma2 induce adipogenesis of NIH‐3T3 cells with similar efficiency and kinetics. Physiol Genomics 23: 206‐216, 2005.
 4.Amato AA, Rajagopalan S, Lin JZ, Carvalho BM, Figueira AC, Lu J, Ayers SD, Mottin M, Silveira RL, Souza PC, Mourao RH, Saad MJ, Togashi M, Simeoni LA, Abdalla DS, Skaf MS, Polikparpov I, Lima MC, Galdino SL, Brennan RG, Baxter JD, Pitta IR, Webb P, Phillips KJ, Neves FA. GQ‐16, a novel peroxisome proliferator‐activated receptor gamma (PPARgamma) ligand, promotes insulin sensitization without weight gain. J Biol Chem 287: 28169‐28179, 2012.
 5.Amini Z, Boyd B, Doucet J, Ribnicky DM, Stephens JM. St. John's Wort inhibits adipocyte differentiation and induces insulin resistance in adipocytes. Biochem Biophys Res Commun 388: 146‐149, 2009.
 6.Arner P, Kulyte A. MicroRNA regulatory networks in human adipose tissue and obesity. Nat Rev Endocrinol 11: 276‐288, 2015.
 7.Asada M, Rauch A, Shimizu H, Maruyama H, Miyaki S, Shibamori M, Kawasome H, Ishiyama H, Tuckermann J, Asahara H. DNA binding‐dependent glucocorticoid receptor activity promotes adipogenesis via Kruppel‐like factor 15 gene expression. Lab Invest 91: 203‐215, 2011.
 8.Bahn YJ, Lee KP, Lee SM, Choi JY, Seo YS, Kwon KS. Nucleoredoxin promotes adipogenic differentiation through regulation of Wnt/beta‐catenin signaling. J Lipid Res 56: 294‐303, 2015.
 9.Banerjee SS, Feinberg MW, Watanabe M, Gray S, Haspel RL, Denkinger DJ, Kawahara R, Hauner H, Jain MK. The Kruppel‐like factor KLF2 inhibits peroxisome proliferator‐activated receptor‐gamma expression and adipogenesis. J Biol Chem 278: 2581‐2584, 2003.
 10.Banks AS, McAllister FE, Camporez JP, Zushin PJ, Jurczak MJ, Laznik‐Bogoslavski D, Shulman GI, Gygi SP, Spiegelman BM. An ERK/Cdk5 axis controls the diabetogenic actions of PPARgamma. Nature 517: 391‐395, 2015.
 11.Barak Y, Nelson MC, Ong ES, Jones YZ, Ruiz‐Lozano P, Chien KR, Koder A, Evans RM. PPAR gamma is required for placental, cardiac, and adipose tissue development. Mol Cell 4: 585‐595, 1999.
 12.Barbatelli G, Murano I, Madsen L, Hao Q, Jimenez M, Kristiansen K, Giacobino JP, De MR, Cinti S. The emergence of cold‐induced brown adipocytes in mouse white fat depots is determined predominantly by white to brown adipocyte transdifferentiation. Am J Physiol Endocrinol Metab 298: E1244‐E1253, 2010.
 13.Barroso I, Gurnell M, Crowley VE, Agostini M, Schwabe JW, Soos MA, Maslen GL, Williams TD, Lewis H, Schafer AJ, Chatterjee VK, O'Rahilly S. Dominant negative mutations in human PPARgamma associated with severe insulin resistance, diabetes mellitus and hypertension. Nature 402: 880‐883, 1999.
 14.Baugh JE, Jr., Floyd ZE, Stephens JM. The modulation of STAT5A/GR complexes during fat cell differentiation and in mature adipocytes. Obesity (Silver Spring) 15: 583‐590, 2007.
 15.Bennett CN, Ross SE, Longo KA, Bajnok L, Hemati N, Johnson KW, Harrison SD, MacDougald OA. Regulation of Wnt signaling during adipogenesis. J Biol Chem 277: 30998‐31004, 2002.
 16.Berry DC, Jiang Y, Graff JM. Emerging roles of adipose progenitor cells in tissue development, homeostasis, expansion and thermogenesis. Trends Endocrinol Metab 27: 574‐585, 2016.
 17.Bertheuil N, Chaput B, Menard C, Varin A, Garrido I, Grolleau JL, Sensebe L, Watier E, Tarte K. [Adipose‐derived stromal cells: History, isolation, immunomodulatory properties and clinical perspectives]. Ann Chir Plast Esthet 60: 94‐102, 2015.
 18.Billon N, Iannarelli P, Monteiro MC, Glavieux‐Pardanaud C, Richardson WD, Kessaris N, Dani C, Dupin E. The generation of adipocytes by the neural crest. Development 134: 2283‐2292, 2007.
 19.Birsoy K, Chen Z, Friedman J. Transcriptional regulation of adipogenesis by KLF4. Cell Metab 7: 339‐347, 2008.
 20.Bjorntorp P. Adipose tissue distribution and function. Int J Obes 15(Suppl 2): 67‐81, 1991.
 21.Blouin K, Veilleux A, Luu‐The V, Tchernof A. Androgen metabolism in adipose tissue: Recent advances. Mol Cell Endocrinol 301: 97‐103, 2009.
 22.Blumberg JM, Tzameli I, Astapova I, Lam FS, Flier JS, Hollenberg AN. Complex role of the vitamin D receptor and its ligand in adipogenesis in 3T3‐L1 cells. J Biol Chem 281: 11205‐11213, 2006.
 23.Bowers RR, Kim JW, Otto TC, Lane MD. Stable stem cell commitment to the adipocyte lineage by inhibition of DNA methylation: Role of the BMP‐4 gene. Proc Natl Acad Sci U S A 103: 13022‐13027, 2006.
 24.Bujalska IJ, Gathercole LL, Tomlinson JW, Darimont C, Ermolieff J, Fanjul AN, Rejto PA, Stewart PM. A novel selective 11beta‐hydroxysteroid dehydrogenase type 1 inhibitor prevents human adipogenesis. J Endocrinol 197: 297‐307, 2008.
 25.Burgermeister E, Chuderland D, Hanoch T, Meyer M, Liscovitch M, Seger R. Interaction with MEK causes nuclear export and downregulation of peroxisome proliferator‐activated receptor gamma. Mol Cell Biol 27: 803‐817, 2007.
 26.Burgermeister E, Seger R. MAPK kinases as nucleo‐cytoplasmic shuttles for PPARgamma. Cell Cycle 6: 1539‐1548, 2007.
 27.Butterwith SC, Wilkie RS, Clinton M. Treatment of pluripotential C3H 10T1/2 fibroblasts with bone morphogenetic protein‐4 induces adipocyte commitment. Biochem Soc Trans 24: 163S, 1996.
 28.Cadigan KM, Nusse R. Wnt signaling: A common theme in animal development. Genes Dev 11: 3286‐3305, 1997.
 29.Camp HS, Tafuri SR. Regulation of peroxisome proliferator‐activated receptor gamma activity by mitogen‐activated protein kinase. J Biol Chem 272: 10811‐10816, 1997.
 30.Camp HS, Tafuri SR, Leff T. c‐Jun N‐terminal kinase phosphorylates peroxisome proliferator‐activated receptor‐gamma1 and negatively regulates its transcriptional activity. Endocrinology 140: 392‐397, 1999.
 31.Cannon B, Hedin A, Nedergaard J. Exclusive occurrence of thermogenin antigen in brown adipose tissue. FEBS Lett 150: 129‐132, 1982.
 32.Cao Y, Gomes SA, Rangel EB, Paulino EC, Fonseca TL, Li J, Teixeira MB, Gouveia CH, Bianco AC, Kapiloff MS, Balkan W, Hare JM. S‐nitrosoglutathione reductase‐dependent PPARgamma denitrosylation participates in MSC‐derived adipogenesis and osteogenesis. J Clin Invest 125: 1679‐1691, 2015.
 33.Cao Z, Umek RM, McKnight SL. Regulated expression of three C/EBP isoforms during adipose conversion of 3T3‐L1 cells. Genes Dev 5: 1538‐1552, 1991.
 34.Caprio M, Antelmi A, Chetrite G, Muscat A, Mammi C, Marzolla V, Fabbri A, Zennaro MC, Feve B. Antiadipogenic effects of the mineralocorticoid receptor antagonist drospirenone: Potential implications for the treatment of metabolic syndrome. Endocrinology 152: 113‐125, 2011.
 35.Caprio M, Feve B, Claes A, Viengchareun S, Lombes M, Zennaro MC. Pivotal role of the mineralocorticoid receptor in corticosteroid‐induced adipogenesis. FASEB J 21: 2185‐2194, 2007.
 36.Carey M. The enhanceosome and transcriptional synergy. Cell 92: 5‐8, 1998.
 37.Cawthorn WP, Bree AJ, Yao Y, Du B, Hemati N, Martinez‐Santibanez G, MacDougald OA. Wnt6, Wnt10a and Wnt10b inhibit adipogenesis and stimulate osteoblastogenesis through a beta‐catenin‐dependent mechanism. Bone 50: 477‐489, 2012.
 38.Cernkovich ER, Deng J, Bond MC, Combs TP, Harp JB. Adipose‐specific disruption of signal transducer and activator of transcription 3 increases body weight and adiposity. Endocrinology 149: 1581‐1590, 2008.
 39.Cervantes‐Camacho C, Beltran‐Langarica A, Ochoa‐Uribe AK, Marsch‐Moreno M, Ayala‐Sumuano JT, Velez‐delValle C, Kuri‐Harcuch W. The transient expression of Klf4 and Klf5 during adipogenesis depends on GSK3beta activity. Adipocyte 4: 248‐255, 2015.
 40.Chapman AB, Knight DM, Ringold GM. Glucocorticoid regulation of adipocyte differentiation: Hormonal triggering of the developmental program and induction of a differentiation‐dependent gene. J Cell Biol 101: 1227‐1235, 1985.
 41.Chazenbalk G, Singh P, Irge D, Shah A, Abbott DH, Dumesic DA. Androgens inhibit adipogenesis during human adipose stem cell commitment to preadipocyte formation. Steroids 78: 920‐926, 2013.
 42.Chen C, Peng Y, Peng Y, Peng J, Jiang S. miR‐135a‐5p inhibits 3T3‐L1 adipogenesis through activation of canonical Wnt/beta‐catenin signaling. J Mol Endocrinol 52: 311‐320, 2014.
 43.Chen C, Xiang H, Peng YL, Peng J, Jiang SW. Mature miR‐183, negatively regulated by transcription factor GATA3, promotes 3T3‐L1 adipogenesis through inhibition of the canonical Wnt/beta‐catenin signaling pathway by targeting LRP6. Cell Signal 26: 1155‐1165, 2014.
 44.Chen Z, Torrens JI, Anand A, Spiegelman BM, Friedman JM. Krox20 stimulates adipogenesis via C/EBPbeta‐dependent and ‐independent mechanisms. Cell Metab 1: 93‐106, 2005.
 45.Cho SY, Park PJ, Shin HJ, Kim YK, Shin DW, Shin ES, Lee HH, Lee BG, Baik JH, Lee TR. (‐)‐Catechin suppresses expression of Kruppel‐like factor 7 and increases expression and secretion of adiponectin protein in 3T3‐L1 cells. Am J Physiol Endocrinol Metab 292: E1166‐E1172, 2007.
 46.Cho YM, Kim DH, Kwak SN, Jeong SW, Kwon OJ. X‐box binding protein 1 enhances adipogenic differentiation of 3T3‐L1 cells through the downregulation of Wnt10b expression. FEBS Lett 587: 1644‐1649, 2013.
 47.Choi JH, Banks AS, Estall JL, Kajimura S, Bostrom P, Laznik D, Ruas JL, Chalmers MJ, Kamenecka TM, Bluher M, Griffin PR, Spiegelman BM. Anti‐diabetic drugs inhibit obesity‐linked phosphorylation of PPARgamma by Cdk5. Nature 466: 451‐456, 2010.
 48.Choi JH, Banks AS, Kamenecka TM, Busby SA, Chalmers MJ, Kumar N, Kuruvilla DS, Shin Y, He Y, Bruning JB, Marciano DP, Cameron MD, Laznik D, Jurczak MJ, Schurer SC, Vidovic D, Shulman GI, Spiegelman BM, Griffin PR. Antidiabetic actions of a non‐agonist PPARgamma ligand blocking Cdk5‐mediated phosphorylation. Nature 477: 477‐481, 2011.
 49.Choi JH, Choi SS, Kim ES, Jedrychowski MP, Yang YR, Jang HJ, Suh PG, Banks AS, Gygi SP, Spiegelman BM. Thrap3 docks on phosphoserine 273 of PPARgamma and controls diabetic gene programming. Genes Dev 28: 2361‐2369, 2014.
 50.Choi SS, Kim ES, Koh M, Lee SJ, Lim D, Yang YR, Jang HJ, Seo KA, Min SH, Lee IH, Park SB, Suh PG, Choi JH. A novel non‐agonist peroxisome proliferator‐activated receptor gamma (PPARgamma) ligand UHC1 blocks PPARgamma phosphorylation by cyclin‐dependent kinase 5 (CDK5) and improves insulin sensitivity. J Biol Chem 289: 26618‐26629, 2014.
 51.Christodoulides C, Lagathu C, Sethi JK, Vidal‐Puig A. Adipogenesis and WNT signalling. Trends Endocrinol Metab 20: 16‐24, 2009.
 52.Christodoulides C, Laudes M, Cawthorn WP, Schinner S, Soos M, O'Rahilly S, Sethi JK, Vidal‐Puig A. The Wnt antagonist Dickkopf‐1 and its receptors are coordinately regulated during early human adipogenesis. J Cell Sci 119: 2613‐2620, 2006.
 53.Chung SS, Ahn BY, Kim M, Choi HH, Park HS, Kang S, Park SG, Kim YB, Cho YM, Lee HK, Chung CH, Park KS. Control of adipogenesis by the SUMO‐specific protease SENP2. Mol Cell Biol 30: 2135‐2146, 2010.
 54.Chung SS, Lee JS, Kim M, Ahn BY, Jung HS, Lee HM, Kim JW, Park KS. Regulation of Wnt/beta‐catenin signaling by CCAAT/enhancer binding protein beta during adipogenesis. Obesity (Silver Spring) 20: 482‐487, 2012.
 55.Chusyd DE, Wang D, Huffman DM, Nagy TR. Relationships between rodent white adipose fat pads and human white adipose fat depots. Front Nutr 3: 10, 2016.
 56.Cianferotti L, Demay MB. VDR‐mediated inhibition of DKK1 and SFRP2 suppresses adipogenic differentiation of murine bone marrow stromal cells. J Cell Biochem 101: 80‐88, 2007.
 57.Cinti S. The adipose organ. Prostaglandins Leukot Essent Fatty Acids 73: 9‐15, 2005.
 58.Clouthier DE, Comerford SA, Hammer RE. Hepatic fibrosis, glomerulosclerosis, and a lipodystrophy‐like syndrome in PEPCK‐TGF‐beta1 transgenic mice. J Clin Invest 100: 2697‐2713, 1997.
 59.Correa D, Hesse E, Seriwatanachai D, Kiviranta R, Saito H, Yamana K, Neff L, Atfi A, Coillard L, Sitara D, Maeda Y, Warming S, Jenkins NA, Copeland NG, Horne WC, Lanske B, Baron R. Zfp521 is a target gene and key effector of parathyroid hormone‐related peptide signaling in growth plate chondrocytes. Dev Cell 19: 533‐546, 2010.
 60.Crandall DL, Busler DE, Novak TJ, Weber RV, Kral JG. Identification of estrogen receptor beta RNA in human breast and abdominal subcutaneous adipose tissue. Biochem Biophys Res Commun 248: 523‐526, 1998.
 61.Crispi S, Sanzari E, Monfregola J, De FN, Fimiani G, Ambrosio R, D'Urso M, Ursini MV. Characterization of the human STAT5A and STAT5B promoters: Evidence of a positive and negative mechanism of transcriptional regulation. FEBS Lett 562: 27‐34, 2004.
 62.Cristancho AG, Lazar MA. Forming functional fat: A growing understanding of adipocyte differentiation. Nat Rev Mol Cell Biol 12: 722‐734, 2011.
 63.Cypess AM, Lehman S, Williams G, Tal I, Rodman D, Goldfine AB, Kuo FC, Palmer EL, Tseng YH, Doria A, Kolodny GM, Kahn CR. Identification and importance of brown adipose tissue in adult humans. N Engl J Med 360: 1509‐1517, 2009.
 64.Danforth E, Jr. Failure of adipocyte differentiation causes type II diabetes mellitus? Nat Genet 26: 13, 2000.
 65.Dang DT, Pevsner J, Yang VW. The biology of the mammalian Kruppel‐like family of transcription factors. Int J Biochem Cell Biol 32: 1103‐1121, 2000.
 66.Darnell JE, Jr. STATs and gene regulation. Science 277: 1630‐1635, 1997.
 67.De PG, Xu XF, Yang SM, Giorgino R, Bjorntorp P. Up‐regulation of androgen receptor binding in male rat fat pad adipose precursor cells exposed to testosterone: Study in a whole cell assay system. J Steroid Biochem Mol Biol 37: 553‐558, 1990.
 68.Deng J, Hua K, Caveney EJ, Takahashi N, Harp JB. Protein inhibitor of activated STAT3 inhibits adipogenic gene expression. Biochem Biophys Res Commun 339: 923‐931, 2006.
 69.Deng J, Hua K, Lesser SS, Harp JB. Activation of signal transducer and activator of transcription‐3 during proliferative phases of 3T3‐L1 adipogenesis. Endocrinology 141: 2370‐2376, 2000.
 70.Deslypere JP, Verdonck L, Vermeulen A. Fat tissue: A steroid reservoir and site of steroid metabolism. J Clin Endocrinol Metab 61: 564‐570, 1985.
 71.Dhavan R, Tsai LH. A decade of CDK5. Nat Rev Mol Cell Biol 2: 749‐759, 2001.
 72.Di PN, Panel V, Hayes S, Bagattin A, Meruvu S, Pandolfi A, Hugendubler L, Fejes‐Toth G, Naray‐Fejes‐Toth A, Mueller E. Serum‐ and glucocorticoid‐inducible kinase 1 (SGK1) regulates adipocyte differentiation via forkhead box O1. Mol Endocrinol 24: 370‐380, 2010.
 73.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. Am J Physiol Cell Physiol 286: C655‐C661, 2004.
 74.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 274: C1645‐C1652, 1998.
 75.Dieudonne MN, Pecquery R, Leneveu MC and 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 141: 649‐656, 2000.
 76.Dieudonne MN, Pecquery R, Leneveu MC, Jaubert AM, Giudicelli Y. Androgen receptors in cultured rat adipose precursor cells during proliferation and differentiation: Regional specificities and regulation by testosterone. Endocrine 3: 537‐541, 1995.
 77.Diezko R, Suske G. Ligand binding reduces SUMOylation of the peroxisome proliferator‐activated receptor gamma (PPARgamma) activation function 1 (AF1) domain. PLoS One 8: e66947, 2013.
 78.Distel RJ, Ro HS, Rosen BS, Groves DL, Spiegelman BM. Nucleoprotein complexes that regulate gene expression in adipocyte differentiation: Direct participation of c‐fos. Cell 49: 835‐844, 1987.
 79.Doney AS, Fischer B, Leese G, Morris AD, Palmer CN. Cardiovascular risk in type 2 diabetes is associated with variation at the PPARG locus: A Go‐DARTS study. Arterioscler Thromb Vasc Biol 24: 2403‐2407, 2004.
 80.Du C, Ma X, Meruvu S, Hugendubler L, Mueller E. The adipogenic transcriptional cofactor ZNF638 interacts with splicing regulators and influences alternative splicing. J Lipid Res 55: 1886‐1896, 2014.
 81.Eguchi J, Yan QW, Schones DE, Kamal M, Hsu CH, Zhang MQ, Crawford GE, Rosen ED. Interferon regulatory factors are transcriptional regulators of adipogenesis. Cell Metab 7: 86‐94, 2008.
 82.Eisenstein A, Carroll SH, Johnston‐Cox H, Farb M, Gokce N, Ravid K. An adenosine receptor‐Kruppel‐like factor 4 protein axis inhibits adipogenesis. J Biol Chem 289: 21071‐21081, 2014.
 83.Ellwood K, Huang W, Johnson R, Carey M. Multiple layers of cooperativity regulate enhanceosome‐responsive RNA polymerase II transcription complex assembly. Mol Cell Biol 19: 2613‐2623, 1999.
 84.Escalona‐Nandez I, Guerrero‐Escalera D, Estanes‐Hernandez A, Ortiz‐Ortega V, Tovar AR, Perez‐Monter C. The activation of peroxisome proliferator‐activated receptor gamma is regulated by Kruppel‐like transcription factors 6 & 9 under steatotic conditions. Biochem Biophys Res Commun 458: 751‐756, 2015.
 85.Fajas L, Auboeuf D, Raspe E, Schoonjans K, Lefebvre AM, Saladin R, Najib J, Laville M, Fruchart JC, Deeb S, Vidal‐Puig A, Flier J, Briggs MR, Staels B, Vidal H, Auwerx J. The organization, promoter analysis, and expression of the human PPARgamma gene. J Biol Chem 272: 18779‐18789, 1997.
 86.Floyd ZE, Stephens JM. Interferon‐gamma‐mediated activation and ubiquitin‐proteasome‐dependent degradation of PPARgamma in adipocytes. J Biol Chem 277: 4062‐4068, 2002.
 87.Floyd ZE, Stephens JM. STAT5A promotes adipogenesis in nonprecursor cells and associates with the glucocorticoid receptor during adipocyte differentiation. Diabetes 52: 308‐314, 2003.
 88.Floyd ZE, Stephens JM. Control of peroxisome proliferator‐activated receptor gamma2 stability and activity by SUMOylation. Obes Res 12: 921‐928, 2004.
 89.Forman BM, Tontonoz P, Chen J, Brun RP, Spiegelman BM, Evans RM. 15‐Deoxy‐delta 12, 14‐prostaglandin J2 is a ligand for the adipocyte determination factor PPAR gamma. Cell 83: 803‐812, 1995.
 90.Gaillard D, Wabitsch M, Pipy B, Negrel R. Control of terminal differentiation of adipose precursor cells by glucocorticoids. J Lipid Res 32: 569‐579, 1991.
 91.Gimble JM, Guilak F. Differentiation potential of adipose derived adult stem (ADAS) cells. Curr Top Dev Biol 58: 137‐160, 2003.
 92.Girdwood D, Bumpass D, Vaughan OA, Thain A, Anderson LA, Snowden AW, Garcia‐Wilson E, Perkins ND, Hay RT. P300 transcriptional repression is mediated by SUMO modification. Mol Cell 11: 1043‐1054, 2003.
 93.Granneman JG. Renaissance of brown adipose tissue research: Integrating the old and new. Int J Obes Suppl 5: S7‐S10, 2015.
 94.Green H, Kehinde O. Formation of normally differentiated subcutaneous fat pads by an established preadipose cell line. J Cell Physiol 101: 169‐171, 1979.
 95.Gregoire F, De BN, Hauser N, Heremans H, Van DJ, Remacle C. Interferon‐gamma and interleukin‐1 beta inhibit adipoconversion in cultured rodent preadipocytes. J Cell Physiol 151: 300‐309, 1992.
 96.Gubelmann C, Schwalie PC, Raghav SK, Roder E, Delessa T, Kiehlmann E, Waszak SM, Corsinotti A, Udin G, Holcombe W, Rudofsky G, Trono D, Wolfrum C, Deplancke B. Identification of the transcription factor ZEB1 as a central component of the adipogenic gene regulatory network. Elife 3: e03346, 2014.
 97.Guo L, Huang JX, Liu Y, Li X, Zhou SR, Qian SW, Liu Y, Zhu H, Huang HY, Dang YJ, Tang QQ. Transactivation of Atg4b by C/EBPbeta promotes autophagy to facilitate adipogenesis. Mol Cell Biol 33: 3180‐3190, 2013.
 98.Gupta RK, Arany Z, Seale P, Mepani RJ, Ye L, Conroe HM, Roby YA, Kulaga H, Reed RR, Spiegelman BM. Transcriptional control of preadipocyte determination by Zfp423. Nature 464: 619‐623, 2010.
 99.Gustafson B, Hammarstedt A, Hedjazifar S, Smith U. Restricted adipogenesis in hypertrophic obesity: The role of WISP2, WNT, and BMP4. Diabetes 62: 2997‐3004, 2013.
 100.Hamm JK, Park BH, Farmer SR. A role for C/EBPbeta in regulating peroxisome proliferator‐activated receptor gamma activity during adipogenesis in 3T3‐L1 preadipocytes. J Biol Chem 276: 18464‐18471, 2001.
 101.Hammarstedt A, Hedjazifar S, Jenndahl L, Gogg S, Grunberg J, Gustafson B, Klimcakova E, Stich V, Langin D, Laakso M, Smith U. WISP2 regulates preadipocyte commitment and PPARgamma activation by BMP4. Proc Natl Acad Sci U S A 110: 2563‐2568, 2013.
 102.Harp JB, Franklin D, Vanderpuije AA, Gimble JM. Differential expression of signal transducers and activators of transcription during human adipogenesis. Biochem Biophys Res Commun 281: 907‐912, 2001.
 103.Hasegawa R, Tomaru Y, de HM, Suzuki H, Hayashizaki Y, Shin JW. Identification of ZNF395 as a novel modulator of adipogenesis. Exp Cell Res 319: 68‐76, 2013.
 104.Hata K, Nishimura R, Ikeda F, Yamashita K, Matsubara T, Nokubi T, Yoneda T. Differential roles of Smad1 and p38 kinase in regulation of peroxisome proliferator‐activating receptor gamma during bone morphogenetic protein 2‐induced adipogenesis. Mol Biol Cell 14: 545‐555, 2003.
 105.Hauser S, Adelmant G, Sarraf P, Wright HM, Mueller E, Spiegelman BM. Degradation of the peroxisome proliferator‐activated receptor gamma is linked to ligand‐dependent activation. J Biol Chem 275: 18527‐18533, 2000.
 106.He H, Chen K, Wang F, Zhao L, Wan X, Wang L, Mo Z. miR‐204‐5p promotes the adipogenic differentiation of human adipose‐derived mesenchymal stem cells by modulating DVL3 expression and suppressing Wnt/beta‐catenin signaling. Int J Mol Med 35: 1587‐1595, 2015.
 107.He W, Barak Y, Hevener A, Olson P, Liao D, Le J, Nelson M, Ong E, Olefsky JM, Evans RM. Adipose‐specific peroxisome proliferator‐activated receptor gamma knockout causes insulin resistance in fat and liver but not in muscle. Proc Natl Acad Sci U S A 100: 15712‐15717, 2003.
 108.Heine PA, Taylor JA, Iwamoto GA, Lubahn DB, Cooke PS. Increased adipose tissue in male and female estrogen receptor‐alpha knockout mice. Proc Natl Acad Sci U S A 97: 12729‐12734, 2000.
 109.Heldin CH, Miyazono K, ten DP. TGF‐beta signalling from cell membrane to nucleus through SMAD proteins. Nature 390: 465‐471, 1997.
 110.Hibi M, Oishi S, Matsushita M, Yoneshiro T, Yamaguchi T, Usui C, Yasunaga K, Katsuragi Y, Kubota K, Tanaka S, Saito M. Brown adipose tissue is involved in diet‐induced thermogenesis and whole‐body fat utilization in healthy humans. Int J Obes (Lond) 40: 1655‐1661, 2016.
 111.Himms‐Hagen J, Melnyk A, Zingaretti MC, Ceresi E, Barbatelli G, Cinti S. Multilocular fat cells in WAT of CL‐316243‐treated rats derive directly from white adipocytes. Am J Physiol Cell Physiol 279: C670‐C681, 2000.
 112.Holmstrom S, Van Antwerp ME, Iniguez‐Lluhi JA. Direct and distinguishable inhibitory roles for SUMO isoforms in the control of transcriptional synergy. Proc Natl Acad Sci U S A 100: 15758‐15763, 2003.
 113.Honda T, Ishii A, Inui M. Regulation of adipocyte differentiation of 3T3‐L1 cells by PDZRN3. Am J Physiol Cell Physiol 304: C1091‐C1097, 2013.
 114.Horton JD, Shimomura I, Brown MS, Hammer RE, Goldstein JL, Shimano H. Activation of cholesterol synthesis in preference to fatty acid synthesis in liver and adipose tissue of transgenic mice overproducing sterol regulatory element‐binding protein‐2. J Clin Invest 101: 2331‐2339, 1998.
 115.Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM. Increased adipose tissue expression of tumor necrosis factor‐alpha in human obesity and insulin resistance. J Clin Invest 95: 2409‐2415, 1995.
 116.Hu E, Kim JB, Sarraf P, Spiegelman BM. Inhibition of adipogenesis through MAP kinase‐mediated phosphorylation of PPARgamma. Science 274: 2100‐2103, 1996.
 117.Hu E, Tontonoz P, Spiegelman BM. Transdifferentiation of myoblasts by the adipogenic transcription factors PPAR gamma and C/EBP alpha. Proc Natl Acad Sci U S A 92: 9856‐9860, 1995.
 118.Hua X, Wu J, Goldstein JL, Brown MS, Hobbs HH. Structure of the human gene encoding sterol regulatory element binding protein‐1 (SREBF1) and localization of SREBF1 and SREBF2 to chromosomes 17p11.2 and 22q13. Genomics 25: 667‐673, 1995.
 119.Huang H, Song TJ, Li X, Hu L, He Q, Liu M, Lane MD, Tang QQ. BMP signaling pathway is required for commitment of C3H10T1/2 pluripotent stem cells to the adipocyte lineage. Proc Natl Acad Sci U S A 106: 12670‐12675, 2009.
 120.Ignotz RA, Massague J. Type beta transforming growth factor controls the adipogenic differentiation of 3T3 fibroblasts. Proc Natl Acad Sci U S A 82: 8530‐8534, 1985.
 121.Ihunnah CA, Wada T, Philips BJ, Ravuri SK, Gibbs RB, Kirisci L, Rubin JP, Marra KG, Xie W. Estrogen sulfotransferase/SULT1E1 promotes human adipogenesis. Mol Cell Biol 34: 1682‐1694, 2014.
 122.Ijichi N, Ikeda K, Horie‐Inoue K, Yagi K, Okazaki Y, Inoue S. Estrogen‐related receptor alpha modulates the expression of adipogenesis‐related genes during adipocyte differentiation. Biochem Biophys Res Commun 358: 813‐818, 2007.
 123.Iniguez‐Lluhi JA, Pearce D. A common motif within the negative regulatory regions of multiple factors inhibits their transcriptional synergy. Mol Cell Biol 20: 6040‐6050, 2000.
 124.Itoh T, Fairall L, Amin K, Inaba Y, Szanto A, Balint BL, Nagy L, Yamamoto K, Schwabe JW. Structural basis for the activation of PPARgamma by oxidized fatty acids. Nat Struct Mol Biol 15: 924‐931, 2008.
 125.Jack BH, Crossley M. GATA proteins work together with friend of GATA (FOG) and C‐terminal binding protein (CTBP) co‐regulators to control adipogenesis. J Biol Chem 285: 32405‐32414, 2010.
 126.Jeffery E, Wing A, Holtrup B, Sebo Z, Kaplan JL, Saavedra‐Pena R, Church CD, Colman L, Berry R, Rodeheffer MS. The adipose tissue microenvironment regulates depot‐specific adipogenesis in obesity. Cell Metab 24: 142‐150, 2016.
 127.Jeong S, Yoon M. 17beta‐Estradiol inhibition of PPARgamma‐induced adipogenesis and adipocyte‐specific gene expression. Acta Pharmacol Sin 32: 230‐238, 2011.
 128.Ji S, Doumit ME, Hill RA. Correction: Regulation of adipogenesis and key adipogenic gene expression by 1, 25‐dihydroxyvitamin D in 3T3‐L1 cells. PLoS One 10: e0134199, 2015.
 129.Ji S, Doumit ME, Hill RA. Regulation of adipogenesis and key adipogenic gene expression by 1, 25‐dihydroxyvitamin D in 3T3‐L1 cells. PLoS One 10: e0126142, 2015.
 130.Ji S, Park SY, Roth J, Kim HS, Cho JW. O‐GlcNAc modification of PPARgamma reduces its transcriptional activity. Biochem Biophys Res Commun 417: 1158‐1163, 2012.
 131.Jiang X, Ye X, Guo W, Lu H, Gao Z. Inhibition of HDAC3 promotes ligand‐independent PPARgamma activation by protein acetylation. J Mol Endocrinol 53: 191‐200, 2014.
 132.Jimenez MA, Akerblad P, Sigvardsson M, Rosen ED. Critical role for Ebf1 and Ebf2 in the adipogenic transcriptional cascade. Mol Cell Biol 27: 743‐757, 2007.
 133.Jin W, Takagi T, Kanesashi SN, Kurahashi T, Nomura T, Harada J, Ishii S. Schnurri‐2 controls BMP‐dependent adipogenesis via interaction with Smad proteins. Dev Cell 10: 461‐471, 2006.
 134.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 97: 12735‐12740, 2000.
 135.Ju D, He J, Zhao L, Zheng X, Yang G. Estrogen related receptor alpha‐induced adipogenesis is PGC‐1beta‐dependent. Mol Biol Rep 39: 3343‐3354, 2012.
 136.Jung YS, Lee JM, Kim DK, Lee YS, Kim KS, Kim YH, Kim J, Lee MS, Lee IK, Kim SH, Cho SJ, Jeong WI, Lee CH, Harris RA, Choi HS. The orphan nuclear receptor ERRgamma regulates hepatic CB1 receptor‐mediated fibroblast growth factor 21 gene expression. PLoS One 11: e0159425, 2016.
 137.Kang S, Akerblad P, Kiviranta R, Gupta RK, Kajimura S, Griffin MJ, Min J, Baron R, Rosen ED. Regulation of early adipose commitment by Zfp521. PLoS Biol 10: e1001433, 2012.
 138.Kang S, Bajnok L, Longo KA, Petersen RK, Hansen JB, Kristiansen K, MacDougald OA. Effects of Wnt signaling on brown adipocyte differentiation and metabolism mediated by PGC‐1alpha. Mol Cell Biol 25: 1272‐1282, 2005.
 139.Katoh D, Nishizuka M, Osada S, Imagawa M. Fad104, a positive regulator of adipocyte differentiation, suppresses invasion and metastasis of melanoma cells by inhibition of STAT3 activity. PLoS One 10: e0117197, 2015.
 140.Kawai M, Namba N, Mushiake S, Etani Y, Nishimura R, Makishima M, Ozono K. Growth hormone stimulates adipogenesis of 3T3‐L1 cells through activation of the Stat5A/5B‐PPARgamma pathway. J Mol Endocrinol 38: 19‐34, 2007.
 141.Kawamura Y, Tanaka Y, Kawamori R, Maeda S. Overexpression of Kruppel‐like factor 7 regulates adipocytokine gene expressions in human adipocytes and inhibits glucose‐induced insulin secretion in pancreatic beta‐cell line. Mol Endocrinol 20: 844‐856, 2006.
 142.Keay S, Grossberg SE. Interferon inhibits the conversion of 3T3‐L1 mouse fibroblasts into adipocytes. Proc Natl Acad Sci U S A 77: 4099‐4103, 1980.
 143.Kelly KA, Gimble JM. 1,25‐Dihydroxy vitamin D3 inhibits adipocyte differentiation and gene expression in murine bone marrow stromal cell clones and primary cultures. Endocrinology 139: 2622‐2628, 1998.
 144.Kennell JA, MacDougald OA. Wnt signaling inhibits adipogenesis through beta‐catenin‐dependent and ‐independent mechanisms. J Biol Chem 280: 24004‐24010, 2005.
 145.Khanna‐Gupta A. Sumoylation and the function of CCAAT enhancer binding protein alpha (C/EBP alpha). Blood Cells Mol Dis 41: 77‐81, 2008.
 146.Kilroy G, Carter LE, Newman S, Burk DH, Manuel J, Moller A, Bowtell DD, Mynatt RL, Ghosh S, Floyd ZE. The ubiquitin ligase Siah2 regulates obesity‐induced adipose tissue inflammation. Obesity (Silver Spring) 23: 2223‐2232, 2015.
 147.Kilroy G, Kirk‐Ballard H, Carter LE, Floyd ZE. The ubiquitin ligase Siah2 regulates PPARgamma activity in adipocytes. Endocrinology 153: 1206‐1218, 2012.
 148.Kilroy GE, Zhang X, Floyd ZE. PPAR‐gamma AF‐2 domain functions as a component of a ubiquitin‐dependent degradation signal. Obesity (Silver Spring) 17: 665‐673, 2009.
 149.Kim HJ, Cha JY, Seok JW, Choi Y, Yoon BK, Choi H, Yu JH, Song SJ, Kim A, Lee H, Kim D, Han JY, Kim JW. Dexras1 links glucocorticoids to insulin‐like growth factor‐1 signaling in adipogenesis. Sci Rep 6: 28648, 2016.
 150.Kim J, Cantwell CA, Johnson PF, Pfarr CM, Williams SC. Transcriptional activity of CCAAT/enhancer‐binding proteins is controlled by a conserved inhibitory domain that is a target for sumoylation. J Biol Chem 277: 38037‐38044, 2002.
 151.Kim JB, Spiegelman BM. ADD1/SREBP1 promotes adipocyte differentiation and gene expression linked to fatty acid metabolism. Genes Dev 10: 1096‐1107, 1996.
 152.Kim JH, Park KW, Lee EW, Jang WS, Seo J, Shin S, Hwang KA, Song J. Suppression of PPARgamma through MKRN1‐mediated ubiquitination and degradation prevents adipocyte differentiation. Cell Death Differ 21: 594‐603, 2014.
 153.Kim JY, van de Wall E, Laplante M, Azzara A, Trujillo ME, Hofmann SM, Schraw T, Durand JL, Li H, Li G, Jelicks LA, Mehler MF, Hui DY, Deshaies Y, Shulman GI, Schwartz GJ, Scherer PE. Obesity‐associated improvements in metabolic profile through expansion of adipose tissue. J Clin Invest 117: 2621‐2637, 2007.
 154.Kimura H, Fujimori K. Activation of early phase of adipogenesis through Kruppel‐like factor KLF9‐mediated, enhanced expression of CCAAT/enhancer‐binding protein beta in 3T3‐L1 cells. Gene 534: 169‐176, 2014.
 155.Kindblom JM, Gevers EF, Skrtic SM, Lindberg MK, Gothe S, Tornell J, Vennstrom B, Ohlsson C. Increased adipogenesis in bone marrow but decreased bone mineral density in mice devoid of thyroid hormone receptors. Bone 36: 607‐616, 2005.
 156.Kishimoto K, Nishizuka M, Katoh D, Kato A, Osada S, Imagawa M. FAD104, a regulatory factor of adipogenesis, acts as a novel regulator of calvarial bone formation. J Biol Chem 288: 31772‐31783, 2013.
 157.Klaus S, Choy L, Champigny O, Cassard‐Doulcier AM, Ross S, Spiegelman B, Ricquier D. Characterization of the novel brown adipocyte cell line HIB 1B. Adrenergic pathways involved in regulation of uncoupling protein gene expression. J Cell Sci 107(Pt 1): 313‐319, 1994.
 158.Kliewer SA, Lenhard JM, Willson TM, Patel I, Morris DC, Lehmann JM. A prostaglandin J2 metabolite binds peroxisome proliferator‐activated receptor gamma and promotes adipocyte differentiation. Cell 83: 813‐819, 1995.
 159.Knebel B, Kotzka J, Lehr S, Hartwig S, Avci H, Jacob S, Nitzgen U, Schiller M, Marz W, Hoffmann MM, Seemanova E, Haas J, Muller‐Wieland D. A mutation in the c‐fos gene associated with congenital generalized lipodystrophy. Orphanet J Rare Dis 8: 119, 2013.
 160.Kong J, Li YC. Molecular mechanism of 1,25‐dihydroxyvitamin D3 inhibition of adipogenesis in 3T3‐L1 cells. Am J Physiol Endocrinol Metab 290: E916‐E924, 2006.
 161.Kramer AH, Edkins AL, Hoppe HC, Prinsloo E. Dynamic mitochondrial localisation of STAT3 in the cellular adipogenesis model 3T3‐L1. J Cell Biochem 116: 1232‐1240, 2015.
 162.Kretzschmar K, Watt FM. Lineage tracing. Cell 148: 33‐45, 2012.
 163.Kroker AJ, Bruning JB. Review of the structural and dynamic mechanisms of PPARgamma partial agonism. PPAR Res 2015: 816856, 2015.
 164.Kruglikov IL, Scherer PE. Dermal adipocytes: From irrelevance to metabolic targets? Trends Endocrinol Metab 27: 1‐10, 2016.
 165.Kubo M, Ijichi N, Ikeda K, Horie‐Inoue K, Takeda S, Inoue S. Modulation of adipogenesis‐related gene expression by estrogen‐related receptor gamma during adipocytic differentiation. Biochim Biophys Acta 1789: 71‐77, 2009.
 166.Kumari M, Wang X, Lantier L, Lyubetskaya A, Eguchi J, Kang S, Tenen D, Roh HC, Kong X, Kazak L, Ahmad R, Rosen ED. IRF3 promotes adipose inflammation and insulin resistance and represses browning. J Clin Invest 126: 2839‐2854, 2016.
 167.Kuzmochka C, Abdou HS, Hache RJ, Atlas E. Inactivation of histone deacetylase 1 (HDAC1) but not HDAC2 is required for the glucocorticoid‐dependent CCAAT/enhancer‐binding protein alpha (C/EBPalpha) expression and preadipocyte differentiation. Endocrinology 155: 4762‐4773, 2014.
 168.Lacasa D, Agli B, Moynard D, Giudicelli Y. Evidence for a regional‐specific control of rat preadipocyte proliferation and differentiation by the androgenic status. Endocrine 3: 789‐793, 1995.
 169.Lacasa D, Garcia E, Henriot D, Agli B, Giudicelli Y. Site‐related specificities of the control by androgenic status of adipogenesis and mitogen‐activated protein kinase cascade/c‐fos signaling pathways in rat preadipocytes. Endocrinology 138: 3181‐3186, 1997.
 170.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 ) 34: 1695‐1705, 2010.
 171.Lagathu C, Christodoulides C, Virtue S, Cawthorn WP, Franzin C, Kimber WA, Nora ED, Campbell M, Medina‐Gomez G, Cheyette BN, Vidal‐Puig AJ, Sethi JK. Dact1, a nutritionally regulated preadipocyte gene, controls adipogenesis by coordinating the Wnt/beta‐catenin signaling network. Diabetes 58: 609‐619, 2009.
 172.Lea‐Currie YR, Monroe D, McIntosh MK. Dehydroepiandrosterone and related steroids alter 3T3‐L1 preadipocyte proliferation and differentiation. Comp Biochem Physiol C Pharmacol Toxicol Endocrinol 123: 17‐25, 1999.
 173.Lee K, Um SH, Rhee DK, Pyo S. Interferon‐alpha inhibits adipogenesis via regulation of JAK/STAT1 signaling. Biochim Biophys Acta 1860: 2416‐2427, 2016.
 174.Lee MJ, Fried SK. The glucocorticoid receptor, not the mineralocorticoid receptor, plays the dominant role in adipogenesis and adipokine production in human adipocytes. Int J Obes (Lond) 38: 1228‐1233, 2014.
 175.Leow SC, Poschmann J, Too PG, Yin J, Joseph R, McFarlane C, Dogra S, Shabbir A, Ingham PW, Prabhakar S, Leow MK, Lee YS, Ng KL, Chong YS, Gluckman PD, Stunkel W. The transcription factor SOX6 contributes to the developmental origins of obesity by promoting adipogenesis. Development 143: 950‐961, 2016.
 176.Levacher C, Sztalryd C, Kinebanyan MF, Picon L. Effects of thyroid hormones on adipose tissue development in Sherman and Zucker rats. Am J Physiol 246: C50‐C56, 1984.
 177.Li D, Yea S, Li S, Chen Z, Narla G, Banck M, Laborda J, Tan S, Friedman JM, Friedman SL, Walsh MJ. Kruppel‐like factor‐6 promotes preadipocyte differentiation through histone deacetylase 3‐dependent repression of DLK1. J Biol Chem 280: 26941‐26952, 2005.
 178.Li P, Fan W, Xu J, Lu M, Yamamoto H, Auwerx J, Sears DD, Talukdar S, Oh D, Chen A, Bandyopadhyay G, Scadeng M, Ofrecio JM, Nalbandian S, Olefsky JM. Adipocyte NCoR knockout decreases PPARgamma phosphorylation and enhances PPARgamma activity and insulin sensitivity. Cell 147: 815‐826, 2011.
 179.Li Y, Zhang J, Schopfer FJ, Martynowski D, Garcia‐Barrio MT, Kovach A, Suino‐Powell K, Baker PR, Freeman BA, Chen YE, Xu HE. Molecular recognition of nitrated fatty acids by PPAR gamma. Nat Struct Mol Biol 15: 865‐867, 2008.
 180.Liang WC, Wang Y, Liang PP, Pan XQ, Fu WM, Yeung VS, Lu YF, Wan DC, Tsui SK, Tsang SY, Ma WB, Zhang JF, Waye MM. MiR‐25 suppresses 3T3‐L1 adipogenesis by directly targeting KLF4 and C/EBPalpha. J Cell Biochem 116: 2658‐2666, 2015.
 181.Lilja L, Yang SN, Webb DL, Juntti‐Berggren L, Berggren PO, Bark C. Cyclin‐dependent kinase 5 promotes insulin exocytosis. J Biol Chem 276: 34199‐34205, 2001.
 182.List EO, Berryman DE, Funk K, Gosney ES, Jara A, Kelder B, Wang X, Kutz L, Troike K, Lozier N, Mikula V, Lubbers ER, Zhang H, Vesel C, Junnila RK, Frank SJ, Masternak MM, Bartke A, Kopchick JJ. The role of GH in adipose tissue: Lessons from adipose‐specific GH receptor gene‐disrupted mice. Mol Endocrinol 27: 524‐535, 2013.
 183.Liu B, Wang T, Mei W, Li D, Cai R, Zuo Y, Cheng J. Small ubiquitin‐like modifier (SUMO) protein‐specific protease 1 de‐SUMOylates Sharp‐1 protein and controls adipocyte differentiation. J Biol Chem 289: 22358‐22364, 2014.
 184.Liu Y, Park F, Pietrusz JL, Jia G, Singh RJ, Netzel BC, Liang M. Suppression of 11beta‐hydroxysteroid dehydrogenase type 1 with RNA interference substantially attenuates 3T3‐L1 adipogenesis. Physiol Genomics 32: 343‐351, 2008.
 185.Liu YY, Ayers S, Milanesi A, Teng X, Rabi S, Akiba Y, Brent GA. Thyroid hormone receptor sumoylation is required for preadipocyte differentiation and proliferation. J Biol Chem 290: 7402‐7415, 2015.
 186.Longo KA, Wright WS, Kang S, Gerin I, Chiang SH, Lucas PC, Opp MR, MacDougald OA. Wnt10b inhibits development of white and brown adipose tissues. J Biol Chem 279: 35503‐35509, 2004.
 187.Luther J, Ubieta K, Hannemann N, Jimenez M, Garcia M, Zech C, Schett G, Wagner EF, Bozec A. Fra‐2/AP‐1 controls adipocyte differentiation and survival by regulating PPARgamma and hypoxia. Cell Death Differ 21: 655‐664, 2014.
 188.Mandrup S, Loftus TM, MacDougald OA, Kuhajda FP, Lane MD. Obese gene expression at in vivo levels by fat pads derived from s.c. implanted 3T3‐F442A preadipocytes. Proc Natl Acad Sci U S A 94: 4300‐4305, 1997.
 189.Margoni A, Fotis L, Papavassiliou AG. The transforming growth factor‐beta/bone morphogenetic protein signalling pathway in adipogenesis. Int J Biochem Cell Biol 44: 475‐479, 2012.
 190.Mariash CN. Thyroid hormone and the adipocyte. J Clin Endocrinol Metab 88: 5603‐5604, 2003.
 191.Mathieu PS, Loboa EG. Cytoskeletal and focal adhesion influences on mesenchymal stem cell shape, mechanical properties, and differentiation down osteogenic, adipogenic, and chondrogenic pathways. Tissue Eng Part B Rev 18: 436‐444, 2012.
 192.McGillicuddy FC, Chiquoine EH, Hinkle CC, Kim RJ, Shah R, Roche HM, Smyth EM, Reilly MP. Interferon gamma attenuates insulin signaling, lipid storage, and differentiation in human adipocytes via activation of the JAK/STAT pathway. J Biol Chem 284: 31936‐31944, 2009.
 193.Meirhaeghe A, Fajas L, Gouilleux F, Cottel D, Helbecque N, Auwerx J, Amouyel P. A functional polymorphism in a STAT5B site of the human PPAR gamma 3 gene promoter affects height and lipid metabolism in a French population. Arterioscler Thromb Vasc Biol 23: 289‐294, 2003.
 194.Meraz MA, White JM, Sheehan KC, Bach EA, Rodig SJ, Dighe AS, Kaplan DH, Riley JK, Greenlund AC, Campbell D, Carver‐Moore K, DuBois RN, Clark R, Aguet M, Schreiber RD. Targeted disruption of the Stat1 gene in mice reveals unexpected physiologic specificity in the JAK‐STAT signaling pathway. Cell 84: 431‐442, 1996.
 195.Meruvu S, Hugendubler L, Mueller E. Regulation of adipocyte differentiation by the zinc finger protein ZNF638. J Biol Chem 286: 26516‐26523, 2011.
 196.Mikkonen L, Hirvonen J, Janne OA. SUMO‐1 regulates body weight and adipogenesis via PPARgamma in male and female mice. Endocrinology 154: 698‐708, 2013.
 197.Mishra A, Zhu XG, Ge K, Cheng SY. Adipogenesis is differentially impaired by thyroid hormone receptor mutant isoforms. J Mol Endocrinol 44: 247‐255, 2010.
 198.Moitra J, Mason MM, Olive M, Krylov D, Gavrilova O, Marcus‐Samuels B, Feigenbaum L, Lee E, Aoyama T, Eckhaus M, Reitman ML, Vinson C. Life without white fat: A transgenic mouse. Genes Dev 12: 3168‐3181, 1998.
 199.Monajemi H, Zhang L, Li G, Jeninga EH, Cao H, Maas M, Brouwer CB, Kalkhoven E, Stroes E, Hegele RA, Leff T. Familial partial lipodystrophy phenotype resulting from a single‐base mutation in deoxyribonucleic acid‐binding domain of peroxisome proliferator‐activated receptor‐gamma. J Clin Endocrinol Metab 92: 1606‐1612, 2007.
 200.Mori T, Sakaue H, Iguchi H, Gomi H, Okada Y, Takashima Y, Nakamura K, Nakamura T, Yamauchi T, Kubota N, Kadowaki T, Matsuki Y, Ogawa W, Hiramatsu R, Kasuga M. Role of Kruppel‐like factor 15 (KLF15) in transcriptional regulation of adipogenesis. J Biol Chem 280: 12867‐12875, 2005.
 201.Mueller E, Drori S, Aiyer A, Yie J, Sarraf P, Chen H, Hauser S, Rosen ED, Ge K, Roeder RG, Spiegelman BM. Genetic analysis of adipogenesis through peroxisome proliferator‐activated receptor gamma isoforms. J Biol Chem 277: 41925‐41930, 2002.
 202.Nedergaard J, Bengtsson T, Cannon B. Unexpected evidence for active brown adipose tissue in adult humans. Am J Physiol Endocrinol Metab 293: E444‐E452, 2007.
 203.Nimitphong H, Holick MF, Fried SK, Lee MJ. 25‐hydroxyvitamin D(3) and 1,25‐dihydroxyvitamin D(3) promote the differentiation of human subcutaneous preadipocytes. PLoS One 7: e52171, 2012.
 204.Nordstrom SM, Tran JL, Sos BC, Wagner KU, Weiss EJ. Disruption of JAK2 in adipocytes impairs lipolysis and improves fatty liver in mice with elevated GH. Mol Endocrinol 27: 1333‐1342, 2013.
 205.Ohshima T, Koga H, Shimotohno K. Transcriptional activity of peroxisome proliferator‐activated receptor gamma is modulated by SUMO‐1 modification. J Biol Chem 279: 29551‐29557, 2004.
 206.Oishi Y, Manabe I, Tobe K, Tsushima K, Shindo T, Fujiu K, Nishimura G, Maemura K, Yamauchi T, Kubota N, Suzuki R, Kitamura T, Akira S, Kadowaki T, Nagai R. Kruppel‐like transcription factor KLF5 is a key regulator of adipocyte differentiation. Cell Metab 1: 27‐39, 2005.
 207.Okazaki R, Inoue D, Shibata M, Saika M, Kido S, Ooka H, Tomiyama H, Sakamoto Y, Matsumoto T. Estrogen promotes early osteoblast differentiation and inhibits adipocyte differentiation in mouse bone marrow stromal cell lines that express estrogen receptor (ER) alpha or beta. Endocrinology 143: 2349‐2356, 2002.
 208.Oppenheimer JH, Schwartz HL, Lane JT, Thompson MP. Functional relationship of thyroid hormone‐induced lipogenesis, lipolysis, and thermogenesis in the rat. J Clin Invest 87: 125‐132, 1991.
 209.Park H, Park H, Pak HJ, Yang DY, Kim YH, Choi WJ, Park SJ, Cho JA, Lee KW. miR‐34a inhibits differentiation of human adipose tissue‐derived stem cells by regulating cell cycle and senescence induction. Differentiation 90: 91‐100, 2015.
 210.Park M, Yi JW, Kim EM, Yoon IJ, Lee EH, Lee HY, Ji KY, Lee KH, Jang JH, Oh SS, Yun CH, Kim SH, Lee KM, Song MG, Kim DH, Kang HS. Triggering receptor expressed on myeloid cells 2 (TREM2) promotes adipogenesis and diet‐induced obesity. Diabetes 64: 117‐127, 2015.
 211.Pascual G, Fong AL, Ogawa S, Gamliel A, Li AC, Perissi V, Rose DW, Willson TM, Rosenfeld MG, Glass CK. A SUMOylation‐dependent pathway mediates transrepression of inflammatory response genes by PPAR‐gamma. Nature 437: 759‐763, 2005.
 212.Pedram A, Razandi M, Blumberg B, Levin ER. Membrane and nuclear estrogen receptor alpha collaborate to suppress adipogenesis but not triglyceride content. FASEB J 30: 230‐240, 2016.
 213.Pei H, Yao Y, Yang Y, Liao K, Wu JR. Kruppel‐like factor KLF9 regulates PPARgamma transactivation at the middle stage of adipogenesis. Cell Death Differ 18: 315‐327, 2011.
 214.Perez‐Mancera PA, Bermejo‐Rodriguez C, Gonzalez‐Herrero I, Herranz M, Flores T, Jimenez R, Sanchez‐Garcia I. Adipose tissue mass is modulated by SLUG (SNAI2). Hum Mol Genet 16: 2972‐2986, 2007.
 215.Phan J, Peterfy M, Reue K. Lipin expression preceding peroxisome proliferator‐activated receptor‐gamma is critical for adipogenesis in vivo and in vitro. J Biol Chem 279: 29558‐29564, 2004.
 216.Poulos SP, Dodson MV, Culver MF, Hausman GJ. The increasingly complex regulation of adipocyte differentiation. Exp Biol Med (Maywood ) 241: 449‐456, 2016.
 217.Price NL, Holtrup B, Kwei SL, Wabitsch M, Rodeheffer M, Bianchini L, Suarez Y, Fernandez‐Hernando C. SREBP‐1c/microRNA 33b genomic loci control adipocyte differentiation. Mol Cell Biol 36: 1180‐1193, 2016.
 218.Price TM, O'Brien SN. Determination of estrogen receptor messenger ribonucleic acid (mRNA) and cytochrome P450 aromatase mRNA levels in adipocytes and adipose stromal cells by competitive polymerase chain reaction amplification. J Clin Endocrinol Metab 77: 1041‐1045, 1993.
 219.Qiang L, Wang L, Kon N, Zhao W, Lee S, Zhang Y, Rosenbaum M, Zhao Y, Gu W, Farmer SR, Accili D. Brown remodeling of white adipose tissue by SirT1‐dependent deacetylation of Ppargamma. Cell 150: 620‐632, 2012.
 220.Quach JM, Walker EC, Allan E, Solano M, Yokoyama A, Kato S, Sims NA, Gillespie MT, Martin TJ. Zinc finger protein 467 is a novel regulator of osteoblast and adipocyte commitment. J Biol Chem 286: 4186‐4198, 2011.
 221.Rajakumari S, Wu J, Ishibashi J, Lim HW, Giang AH, Won KJ, Reed RR, Seale P. EBF2 determines and maintains brown adipocyte identity. Cell Metab 17: 562‐574, 2013.
 222.Rangwala SM, Rhoades B, Shapiro JS, Rich AS, Kim JK, Shulman GI, Kaestner KH and Lazar MA. Genetic modulation of PPARgamma phosphorylation regulates insulin sensitivity. Dev Cell 5: 657‐663, 2003.
 223.Rao TP, Kuhl M. An updated overview on Wnt signaling pathways: A prelude for more. Circ Res 106: 1798‐1806, 2010.
 224.Ren D, Collingwood TN, Rebar EJ, Wolffe AP, Camp HS. PPARgamma knockdown by engineered transcription factors: Exogenous PPARgamma2 but not PPARgamma1 reactivates adipogenesis. Genes Dev 16: 27‐32, 2002.
 225.Rey‐Lopez JP, de Rezende LF, Pastor‐Valero M, Tess BH. The prevalence of metabolically healthy obesity: A systematic review and critical evaluation of the definitions used. Obes Rev 15: 781‐790, 2014.
 226.Richard AJ, Amini ZJ, Ribnicky DM and Stephens JM. St. John's Wort inhibits insulin signaling in murine and human adipocytes. Biochim Biophys Acta 1822: 557‐563, 2012.
 227.Richard AJ, Burris TP, Sanchez‐Infantes D, Wang Y, Ribnicky DM, Stephens JM. Artemisia extracts activate PPARgamma, promote adipogenesis, and enhance insulin sensitivity in adipose tissue of obese mice. Nutrition 30: S31‐S36, 2014.
 228.Richard AJ, Fuller S, Fedorcenco V, Beyl R, Burris TP, Mynatt R, Ribnicky DM, Stephens JM. Artemisia scoparia enhances adipocyte development and endocrine function in vitro and enhances insulin action in vivo. PLoS One 9: e98897, 2014.
 229.Richard AJ, Stephens JM. Emerging roles of JAK‐STAT signaling pathways in adipocytes. Trends Endocrinol Metab 22: 325‐332, 2011.
 230.Rivero S, Diaz‐Guerra MJ, Monsalve EM, Laborda J, Garcia‐Ramirez JJ. DLK2 is a transcriptional target of KLF4 in the early stages of adipogenesis. J Mol Biol 417: 36‐50, 2012.
 231.Rosen E, Eguchi J, Xu Z. Transcriptional targets in adipocyte biology. Expert Opin Ther Targets 13: 975‐986, 2009.
 232.Rosen ED, Sarraf P, Troy AE, Bradwin G, Moore K, Milstone DS, Spiegelman BM, Mortensen RM. PPAR gamma is required for the differentiation of adipose tissue in vivo and in vitro. Mol Cell 4: 611‐617, 1999.
 233.Rossmeisl M, Barbatelli G, Flachs P, Brauner P, Zingaretti MC, Marelli M, Janovska P, Horakova M, Syrovy I, Cinti S, Kopecky J. Expression of the uncoupling protein 1 from the aP2 gene promoter stimulates mitochondrial biogenesis in unilocular adipocytes in vivo. Eur J Biochem 269: 19‐28, 2002.
 234.Rubin CS, Hirsch A, Fung C, Rosen OM. Development of hormone receptors and hormonal responsiveness in vitro. Insulin receptors and insulin sensitivity in the preadipocyte and adipocyte forms of 3T3‐L1 cells. J Biol Chem 253: 7570‐7578, 1978.
 235.Ruiz‐Ojeda FJ, Ruperez AI, Gomez‐Llorente C, Gil A, Aguilera CM. Cell models and their application for studying adipogenic differentiation in relation to obesity: A review. Int J Mol Sci 17: 1040, 2016.
 236.Sanchez‐Gurmaches J, Guertin DA. Adipocytes arise from multiple lineages that are heterogeneously and dynamically distributed. Nat Commun 5: 4099, 2014.
 237.Sanchez‐Gurmaches J, Hung CM, Guertin DA. Emerging complexities in adipocyte origins and identity. Trends Cell Biol 26: 313‐326, 2016.
 238.Sargis RM, Johnson DN, Choudhury RA, Brady MJ. Environmental endocrine disruptors promote adipogenesis in the 3T3‐L1 cell line through glucocorticoid receptor activation. Obesity (Silver Spring) 18: 1283‐1288, 2010.
 239.Sarjeant K, Stephens JM. Adipogenesis. Cold Spring Harb Perspect Biol 4: a008417, 2012.
 240.Saykally JN, Dogan S, Cleary MP, Sanders MM. The ZEB1 transcription factor is a novel repressor of adiposity in female mice. PLoS One 4: e8460, 2009.
 241.Schuh R, Aicher W, Gaul U, Cote S, Preiss A, Maier D, Seifert E, Nauber U, Schroder C, Kemler R, et al. A conserved family of nuclear proteins containing structural elements of the finger protein encoded by Kruppel, a Drosophila segmentation gene. Cell 47: 1025‐1032, 1986.
 242.Schupp M, Lazar MA. Endogenous ligands for nuclear receptors: Digging deeper. J Biol Chem 285: 40409‐40415, 2010.
 243.Scroyen I, Bauters D, Vranckx C, Lijnen HR. The anti‐adipogenic potential of COUP‐TFII is mediated by downregulation of the notch target gene Hey1. PLoS One 10: e0145608, 2015.
 244.Seale P. Transcriptional regulatory circuits controlling brown fat development and activation. Diabetes 64: 2369‐2375, 2015.
 245.Semirale AA, Zhang XW, Wiren KM. Body composition changes and inhibition of fat development in vivo implicates androgen in regulation of stem cell lineage allocation. J Cell Biochem 112: 1773‐1786, 2011.
 246.Semple RK, Chatterjee VK, O'Rahilly S. PPAR gamma and human metabolic disease. J Clin Invest 116: 581‐589, 2006.
 247.Sewter C, Blows F, Considine R, Vidal‐Puig A, O'Rahilly S. Differential effects of adiposity on peroxisomal proliferator‐activated receptor gamma1 and gamma2 messenger ribonucleic acid expression in human adipocytes. J Clin Endocrinol Metab 87: 4203‐4207, 2002.
 248.Sha H, He Y, Chen H, Wang C, Zenno A, Shi H, Yang X, Zhang X, Qi L. The IRE1alpha‐XBP1 pathway of the unfolded protein response is required for adipogenesis. Cell Metab 9: 556‐564, 2009.
 249.Shang CA, Waters MJ. Constitutively active signal transducer and activator of transcription 5 can replace the requirement for growth hormone in adipogenesis of 3T3‐F442A preadipocytes. Mol Endocrinol 17: 2494‐2508, 2003.
 250.Shao D, Rangwala SM, Bailey ST, Krakow SL, Reginato MJ, Lazar MA. Interdomain communication regulating ligand binding by PPAR‐gamma. Nature 396: 377‐380, 1998.
 251.Shao M, Ishibashi J, Kusminski CM, Wang QA, Hepler C, Vishvanath L, MacPherson KA, Spurgin SB, Sun K, Holland WL, Seale P, Gupta RK. Zfp423 maintains white adipocyte identity through suppression of the beige cell thermogenic gene program. Cell Metab 23: 1167‐1184, 2016.
 252.Shen J, James AW, Zhang X, Pang S, Zara JN, Asatrian G, Chiang M, Lee M, Khadarian K, Nguyen A, Lee KS, Siu RK, Tetradis S, Ting K, Soo C. Novel Wnt regulator NEL‐like molecule‐1 antagonizes adipogenesis and augments osteogenesis induced by bone morphogenetic protein 2. Am J Pathol 186: 419‐434, 2016.
 253.Shimano H, Shimomura I, Hammer RE, Herz J, Goldstein JL, Brown MS, Horton JD. Elevated levels of SREBP‐2 and cholesterol synthesis in livers of mice homozygous for a targeted disruption of the SREBP‐1 gene. J Clin Invest 100: 2115‐2124, 1997.
 254.Shimomura I, Shimano H, Horton JD, Goldstein JL, Brown MS. Differential expression of exons 1a and 1c in mRNAs for sterol regulatory element binding protein‐1 in human and mouse organs and cultured cells. J Clin Invest 99: 838‐845, 1997.
 255.Shuai K, Schindler C, Prezioso VR, Darnell JE, Jr. Activation of transcription by IFN‐gamma: Tyrosine phosphorylation of a 91‐kD DNA binding protein. Science 258: 1808‐1812, 1992.
 256.Shuai K, Stark GR, Kerr IM, Darnell JE, Jr. A single phosphotyrosine residue of Stat91 required for gene activation by interferon‐gamma. Science 261: 1744‐1746, 1993.
 257.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 147: 141‐154, 2006.
 258.Soukas A, Socci ND, Saatkamp BD, Novelli S, Friedman JM. Distinct transcriptional profiles of adipogenesis in vivo and in vitro. J Biol Chem 276: 34167‐34174, 2001.
 259.Stark GR, Darnell JE, Jr. The JAK‐STAT pathway at twenty. Immunity 36: 503‐514, 2012.
 260.Stephens JM, Butts MD, Pekala PH. Regulation of transcription factor mRNA accumulation during 3T3‐L1 preadipocyte differentiation by tumour necrosis factor‐alpha. J Mol Endocrinol 9: 61‐72, 1992.
 261.Stephens JM, Morrison RF, Pilch PF. The expression and regulation of STATs during 3T3‐L1 adipocyte differentiation. J Biol Chem 271: 10441‐10444, 1996.
 262.Stephens JM, Morrison RF, Wu Z, Farmer SR. PPARgamma ligand‐dependent induction of STAT1, STAT5A, and STAT5B during adipogenesis. Biochem Biophys Res Commun 262: 216‐222, 1999.
 263.Stephens JM, Morrison RF, Wu Z, Farmer SR. PPARgamma ligand‐dependent induction of STAT1, STAT5A, and STAT5B during adipogenesis. Biochem Biophys Res Commun 262: 216‐222, 1999.
 264.Stewart WC, Morrison RF, Young SL, Stephens JM. Regulation of signal transducers and activators of transcription (STATs) by effectors of adipogenesis: Coordinate regulation of STATs 1, 5A, and 5B with peroxisome proliferator‐activated receptor‐gamma and C/AAAT enhancer binding protein‐alpha. Biochim Biophys Acta 1452: 188‐196, 1999.
 265.Stewart WC, Pearcy LA, Floyd ZE, Stephens JM. STAT5A expression in Swiss 3T3 cells promotes adipogenesis in vivo in an athymic mice model system. Obesity (Silver Spring) 19: 1731‐1734, 2011.
 266.Stine RR, Shapira SN, Lim HW, Ishibashi J, Harms M, Won KJ, Seale P. EBF2 promotes the recruitment of beige adipocytes in white adipose tissue. Mol Metab 5: 57‐65, 2016.
 267.Stockl S, Bauer RJ, Bosserhoff AK, Gottl C, Grifka J, Grassel S. Sox9 modulates cell survival and adipogenic differentiation of multipotent adult rat mesenchymal stem cells. J Cell Sci 126: 2890‐2902, 2013.
 268.Su SC, Tsai LH. Cyclin‐dependent kinases in brain development and disease. Annu Rev Cell Dev Biol 27: 465‐491, 2011.
 269.Sue N, Jack BH, Eaton SA, Pearson RC, Funnell AP, Turner J, Czolij R, Denyer G, Bao S, Molero‐Navajas JC, Perkins A, Fujiwara Y, Orkin SH, Bell‐Anderson K, Crossley M. Targeted disruption of the basic Kruppel‐like factor gene (Klf3) reveals a role in adipogenesis. Mol Cell Biol 28: 3967‐3978, 2008.
 270.Takada I, Mihara M, Suzawa M, Ohtake F, Kobayashi S, Igarashi M, Youn MY, Takeyama K, Nakamura T, Mezaki Y, Takezawa S, Yogiashi Y, Kitagawa H, Yamada G, Takada S, Minami Y, Shibuya H, Matsumoto K, Kato S. A histone lysine methyltransferase activated by non‐canonical Wnt signalling suppresses PPAR‐gamma transactivation. Nat Cell Biol 9: 1273‐1285, 2007.
 271.Takahashi A, Morita M, Yokoyama K, Suzuki T, Yamamoto T. Tob2 inhibits peroxisome proliferator‐activated receptor gamma2 expression by sequestering Smads and C/EBPalpha during adipocyte differentiation. Mol Cell Biol 32: 5067‐5077, 2012.
 272.Tang QQ, Gronborg M, Huang H, Kim JW, Otto TC, Pandey A, Lane MD. Sequential phosphorylation of CCAAT enhancer‐binding protein beta by MAPK and glycogen synthase kinase 3beta is required for adipogenesis. Proc Natl Acad Sci U S A 102: 9766‐9771, 2005.
 273.Tang QQ, Otto TC, Lane MD. Mitotic clonal expansion: A synchronous process required for adipogenesis. Proc Natl Acad Sci U S A 100: 44‐49, 2003.
 274.Teglund S, McKay C, Schuetz E, van Deursen JM, Stravopodis D, Wang D, Brown M, Bodner S, Grosveld G, Ihle JN. Stat5a and Stat5b proteins have essential and nonessential, or redundant, roles in cytokine responses. Cell 93: 841‐850, 1998.
 275.Tian L, Wang C, Hagen FK, Gormley M, Addya S, Soccio R, Casimiro MC, Zhou J, Powell MJ, Xu P, Deng H, Sauve AA, Pestell RG. Acetylation‐defective mutant of Ppargamma is associated with decreased lipid synthesis in breast cancer cells. Oncotarget 5: 7303‐7315, 2014.
 276.Todoric J, Strobl B, Jais A, Boucheron N, Bayer M, Amann S, Lindroos J, Teperino R, Prager G, Bilban M, Ellmeier W, Krempler F, Muller M, Wagner O, Patsch W, Pospisilik JA, Esterbauer H. Cross‐talk between interferon‐gamma and hedgehog signaling regulates adipogenesis. Diabetes 60: 1668‐1676, 2011.
 277.Tong Q, Dalgin G, Xu H, Ting CN, Leiden JM, Hotamisligil GS. Function of GATA transcription factors in preadipocyte‐adipocyte transition. Science 290: 134‐138, 2000.
 278.Tong Q, Tsai J, Tan G, Dalgin G, Hotamisligil GS. Interaction between GATA and the C/EBP family of transcription factors is critical in GATA‐mediated suppression of adipocyte differentiation. Mol Cell Biol 25: 706‐715, 2005.
 279.Tontonoz P, Graves RA, Budavari AI, Erdjument‐Bromage H, Lui M, Hu E, Tempst P, Spiegelman BM. Adipocyte‐specific transcription factor ARF6 is a heterodimeric complex of two nuclear hormone receptors, PPAR gamma and RXR alpha. Nucleic Acids Res 22: 5628‐5634, 1994.
 280.Tontonoz P, Hu E, Graves RA, Budavari AI, Spiegelman BM. mPPAR gamma 2: Tissue‐specific regulator of an adipocyte enhancer. Genes Dev 8: 1224‐1234, 1994.
 281.Tontonoz P, Kim JB, Graves RA, Spiegelman BM. ADD1: A novel helix‐loop‐helix transcription factor associated with adipocyte determination and differentiation. Mol Cell Biol 13: 4753‐4759, 1993.
 282.Tontonoz P, Spiegelman BM. Fat and beyond: The diverse biology of PPARgamma. Annu Rev Biochem 77: 289‐312, 2008.
 283.Torti FM, Torti SV, Larrick JW, Ringold GM. Modulation of adipocyte differentiation by tumor necrosis factor and transforming growth factor beta. J Cell Biol 108: 1105‐1113, 1989.
 284.Tse MC, Liu X, Yang S, Ye K, Chan CB. Fyn regulates adipogenesis by promoting PIKE‐A/STAT5a interaction. Mol Cell Biol 33: 1797‐1808, 2013.
 285.Tseng YH, Kokkotou E, Schulz TJ, Huang TL, Winnay JN, Taniguchi CM, Tran TT, Suzuki R, Espinoza DO, Yamamoto Y, Ahrens MJ, Dudley AT, Norris AW, Kulkarni RN, Kahn CR. New role of bone morphogenetic protein 7 in brown adipogenesis and energy expenditure. Nature 454: 1000‐1004, 2008.
 286.Utreras E, Futatsugi A, Rudrabhatla P, Keller J, Iadarola MJ, Pant HC, Kulkarni AB. Tumor necrosis factor‐alpha regulates cyclin‐dependent kinase 5 activity during pain signaling through transcriptional activation of p35. J Biol Chem 284: 2275‐2284, 2009.
 287.Vanacker JM, Pettersson K, Gustafsson JA, Laudet V. Transcriptional targets shared by estrogen receptor‐related receptors (ERRs) and estrogen receptor (ER) alpha, but not by ERbeta. EMBO J 18: 4270‐4279, 1999.
 288.Vegiopoulos A, Herzig S. Glucocorticoids, metabolism and metabolic diseases. Mol Cell Endocrinol 275: 43‐61, 2007.
 289.Vinson CR, Sigler PB, McKnight SL. Scissors‐grip model for DNA recognition by a family of leucine zipper proteins. Science 246: 911‐916, 1989.
 290.Visweswaran M, Schiefer L, Arfuso F, Dilley RJ, Newsholme P, Dharmarajan A. Wnt antagonist secreted frizzled‐related protein 4 upregulates adipogenic differentiation in human adipose tissue‐derived mesenchymal stem cells. PLoS One 10: e0118005, 2015.
 291.von KA, Tzieply N, Jennewein C, Brune B. Casein‐kinase‐II‐dependent phosphorylation of PPARgamma provokes CRM1‐mediated shuttling of PPARgamma from the nucleus to the cytosol. J Cell Sci 123: 192‐201, 2010.
 292.Wade GN, Gray JM, Bartness TJ. Gonadal influences on adiposity. Int J Obes 9(Suppl 1): 83‐92, 1985.
 293.Waite KJ, Floyd ZE, Arbour‐Reily P, Stephens JM. Interferon‐gamma‐induced regulation of peroxisome proliferator‐activated receptor gamma and STATs in adipocytes. J Biol Chem 276: 7062‐7068, 2001.
 294.Waku T, Shiraki T, Oyama T, Maebara K, Nakamori R, Morikawa K. The nuclear receptor PPARgamma individually responds to serotonin‐ and fatty acid‐metabolites. EMBO J 29: 3395‐3407, 2010.
 295.Wang D, Zhou Y, Lei W, Zhang K, Shi J, Hu Y, Shu G, Song J. Signal transducer and activator of transcription 3 (STAT3) regulates adipocyte differentiation via peroxisome‐proliferator‐activated receptor gamma (PPARgamma). Biol Cell 102: 1‐12, 2010.
 296.Wang EA, Israel DI, Kelly S, Luxenberg DP. Bone morphogenetic protein‐2 causes commitment and differentiation in C3H10T1/2 and 3T3 cells. Growth Factors 9: 57‐71, 1993.
 297.Wang L, Di LJ. Wnt/beta‐catenin mediates AICAR effect to increase GATA3 expression and inhibit adipogenesis. J Biol Chem 290: 19458‐19468, 2015.
 298.Wang QA, Tao C, Gupta RK, Scherer PE. Tracking adipogenesis during white adipose tissue development, expansion and regeneration. Nat Med 19: 1338‐1344, 2013.
 299.Wang QA, Tao C, Jiang L, Shao M, Ye R, Zhu Y, Gordillo R, Ali A, Lian Y, Holland WL, Gupta RK, Scherer PE. Distinct regulatory mechanisms governing embryonic versus adult adipocyte maturation. Nat Cell Biol 17: 1099‐1111, 2015.
 300.Wang RN, Green J, Wang Z, Deng Y, Qiao M, Peabody M, Zhang Q, Ye J, Yan Z, Denduluri S, Idowu O, Li M, Shen C, Hu A, Haydon RC, Kang R, Mok J, Lee MJ, Luu HL, Shi LL. Bone Morphogenetic Protein (BMP) signaling in development and human diseases. Genes Dis 1: 87‐105, 2014.
 301.Wang Y, Sul HS. Pref‐1 regulates mesenchymal cell commitment and differentiation through Sox9. Cell Metab 9: 287‐302, 2009.
 302.Wankhade UD, Shen M, Kolhe R, Fulzele S. Advances in adipose‐derived stem cells isolation, characterization, and application in regenerative tissue engineering. Stem Cells Int 2016: 3206807, 2016.
 303.Wegrzyn J, Potla R, Chwae YJ, Sepuri NB, Zhang Q, Koeck T, Derecka M, Szczepanek K, Szelag M, Gornicka A, Moh A, Moghaddas S, Chen Q, Bobbili S, Cichy J, Dulak J, Baker DP, Wolfman A, Stuehr D, Hassan MO, Fu XY, Avadhani N, Drake JI, Fawcett P, Lesnefsky EJ, Larner AC. Function of mitochondrial Stat3 in cellular respiration. Science 323: 793‐797, 2009.
 304.Wei S, Du M, Jiang Z, Hausman GJ, Zhang L, Dodson MV. Long noncoding RNAs in regulating adipogenesis: New RNAs shed lights on obesity. Cell Mol Life Sci 73: 2079‐2087, 2016.
 305.Weiss MJ and Orkin SH. GATA transcription factors: Key regulators of hematopoiesis. Exp Hematol 23: 99‐107, 1995.
 306.Werman A, Hollenberg A, Solanes G, Bjorbaek C, Vidal‐Puig AJ, Flier JS. Ligand‐independent activation domain in the N terminus of peroxisome proliferator‐activated receptor gamma (PPARgamma). Differential activity of PPARgamma1 and ‐2 isoforms and influence of insulin. J Biol Chem 272: 20230‐20235, 1997.
 307.White UA, Stephens JM. The gp130 receptor cytokine family: Regulators of adipocyte development and function. Curr Pharm Des 17: 340‐346, 2011.
 308.Wiper‐Bergeron N, Salem HA, Tomlinson JJ, Wu D, Hache RJ. Glucocorticoid‐stimulated preadipocyte differentiation is mediated through acetylation of C/EBPbeta by GCN5. Proc Natl Acad Sci U S A 104: 2703‐2708, 2007.
 309.Wu J, Bostrom P, Sparks LM, Ye L, Choi JH, Giang AH, Khandekar M, Virtanen KA, Nuutila P, Schaart G, Huang K, Tu H, van Marken Lichtenbelt WD, Hoeks J, Enerback S, Schrauwen P, Spiegelman BM. Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human. Cell 150: 366‐376, 2012.
 310.Wu J, Srinivasan SV, Neumann JC, Lingrel JB. The KLF2 transcription factor does not affect the formation of preadipocytes but inhibits their differentiation into adipocytes. Biochemistry 44: 11098‐11105, 2005.
 311.Wu Q, Tang SG, Yuan ZM. Gremlin 2 inhibits adipocyte differentiation through activation of Wnt/beta‐catenin signaling. Mol Med Rep 12: 5891‐5896, 2015.
 312.Wu Z, Bucher NL, Farmer SR. Induction of peroxisome proliferator‐activated receptor gamma during the conversion of 3T3 fibroblasts into adipocytes is mediated by C/EBPbeta, C/EBPdelta, and glucocorticoids. Mol Cell Biol 16: 4128‐4136, 1996.
 313.Xie X, Zhou X, Chen W, Long L, Li W, Yang X, Li S, Wang L. L312, a novel PPARgamma ligand with potent anti‐diabetic activity by selective regulation. Biochim Biophys Acta 1850: 62‐72, 2015.
 314.Yahagi N, Shimano H, Hasty AH, Matsuzaka T, Ide T, Yoshikawa T, Amemiya‐Kudo M, Tomita S, Okazaki H, Tamura Y, Iizuka Y, Ohashi K, Osuga J, Harada K, Gotoda T, Nagai R, Ishibashi S, Yamada N. Absence of sterol regulatory element‐binding protein‐1 (SREBP‐1) ameliorates fatty livers but not obesity or insulin resistance in Lep(ob)/Lep(ob) mice. J Biol Chem 277: 19353‐19357, 2002.
 315.Yamashita D, Yamaguchi T, Shimizu M, Nakata N, Hirose F, Osumi T. The transactivating function of peroxisome proliferator‐activated receptor gamma is negatively regulated by SUMO conjugation in the amino‐terminal domain. Genes Cells 9: 1017‐1029, 2004.
 316.Yang SH, Sharrocks AD. SUMO promotes HDAC‐mediated transcriptional repression. Mol Cell 13: 611‐617, 2004.
 317.Yarwood SJ, Sale EM, Sale GJ, Houslay MD, Kilgour E, Anderson NG. Growth hormone‐dependent differentiation of 3T3‐F442A preadipocytes requires Janus kinase/signal transducer and activator of transcription but not mitogen‐activated protein kinase or p70 S6 kinase signaling. J Biol Chem 274: 8662‐8668, 1999.
 318.Ye R, Wang QA, Tao C, Vishvanath L, Shao M, McDonald JG, Gupta RK, Scherer PE. Impact of tamoxifen on adipocyte lineage tracing: Inducer of adipogenesis and prolonged nuclear translocation of Cre recombinase. Mol Metab 4: 771‐778, 2015.
 319.Yeh WC, Cao Z, Classon M, McKnight SL. Cascade regulation of terminal adipocyte differentiation by three members of the C/EBP family of leucine zipper proteins. Genes Dev 9: 168‐181, 1995.
 320.Ying H, Araki O, Furuya F, Kato Y, Cheng SY. Impaired adipogenesis caused by a mutated thyroid hormone alpha1 receptor. Mol Cell Biol 27: 2359‐2371, 2007.
 321.Yokoyama C, Wang X, Briggs MR, Admon A, Wu J, Hua X, Goldstein JL, Brown MS. SREBP‐1, a basic‐helix‐loop‐helix‐leucine zipper protein that controls transcription of the low density lipoprotein receptor gene. Cell 75: 187‐197, 1993.
 322.You L, Pan L, Chen L, Chen JY, Zhang X, Lv Z, Fu D. Suppression of zinc finger protein 467 alleviates osteoporosis through promoting differentiation of adipose derived stem cells to osteoblasts. J Transl Med 10: 11, 2012.
 323.Yu H, He K, Wang L, Hu J, Gu J, Zhou C, Lu R, Jin Y. Stk40 represses adipogenesis through translational control of CCAAT/enhancer‐binding proteins. J Cell Sci 128: 2881‐2890, 2015.
 324.Zamani N and Brown CW. Emerging roles for the transforming growth factor‐{beta} superfamily in regulating adiposity and energy expenditure. Endocr Rev 32: 387‐403, 2011.
 325.Zhang B, Berger J, Zhou G, Elbrecht A, Biswas S, White‐Carrington S, Szalkowski D, Moller DE. Insulin‐ and mitogen‐activated protein kinase‐mediated phosphorylation and activation of peroxisome proliferator‐activated receptor gamma. J Biol Chem 271: 31771‐31774, 1996.
 326.Zhang J, Bakheet R, Parhar RS, Huang CH, Hussain MM, Pan X, Siddiqui SS, Hashmi S. Regulation of fat storage and reproduction by Kruppel‐like transcription factor KLF3 and fat‐associated genes in Caenorhabditis elegans. J Mol Biol 411: 537‐553, 2011.
 327.Zhang K, Guo W, Yang Y, Wu J. JAK2/STAT3 pathway is involved in the early stage of adipogenesis through regulating C/EBPbeta transcription. J Cell Biochem 112: 488‐497, 2011.
 328.Zhang P, O'Loughlin L, Brindley DN, Reue K. Regulation of lipin‐1 gene expression by glucocorticoids during adipogenesis. J Lipid Res 49: 1519‐1528, 2008.
 329.Zhang W, Schmull S, Du M, Liu J, Lu Z, Zhu H, Xue S, Lian F. Estrogen receptor alpha and beta in mouse: Adipose‐derived stem cell proliferation, migration, and brown adipogenesis in vitro. Cell Physiol Biochem 38: 2285‐2299, 2016.
 330.Zhang YY, Li X, Qian SW, Guo L, Huang HY, He Q, Liu Y, Ma CG, Tang QQ. Down‐regulation of type I Runx2 mediated by dexamethasone is required for 3T3‐L1 adipogenesis. Mol Endocrinol 26: 798‐808, 2012.
 331.Zhang Z, Wang H, Sun Y, Li H, Wang N. Klf7 modulates the differentiation and proliferation of chicken preadipocyte. Acta Biochim Biophys Sin (Shanghai) 45: 280‐288, 2013.
 332.Zhang ZW, Wu CY, Li H, Wang N. Expression and functional analyses of Kruppel‐like factor 3 in chicken adipose tissue. Biosci Biotechnol Biochem 78: 614‐623, 2014.
 333.Zingaretti MC, Crosta F, Vitali A, Guerrieri M, Frontini A, Cannon B, Nedergaard J, Cinti S. The presence of UCP1 demonstrates that metabolically active adipose tissue in the neck of adult humans truly represents brown adipose tissue. FASEB J 23: 3113‐3120, 2009.
 334.Zubiria MG, Alzamendi A, Moreno G, Portales A, Castrogiovanni D, Spinedi E, Giovambattista A. Relationship between the Balance of Hypertrophic/Hyperplastic Adipose Tissue Expansion and the Metabolic Profile in a High Glucocorticoids Model. Nutrients 8: 410, 2016.

Related Articles:

Adiposity
Hormonal Regulation of Adipogenesis
Adipose Organ Development and Remodeling
Sex Differences

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Paula Mota de Sá, Allison J. Richard, Hardy Hang, Jacqueline M. Stephens. Transcriptional Regulation of Adipogenesis. Compr Physiol 2017, 7: 635-674. doi: 10.1002/cphy.c160022