regulation of adipocyte differentiation

6
556 + SPECIAL SECTION: MOLECULAR MECHANISMS IN OBESITY AND VISCERAL OBESITY + Regulation of adipocyte differentiation Hana Koutnikova and Johan Auwerx Once multipotent mesechymal cells become committed to the adipoblast lineage, adipogenesis, the process o f preadipocytes differentiation into adipocytes is initiated. This process starts with a phase of exponential growth o f adipoblasts. Following confluence o f these adipoblasts, the cells enter into a cell cycle arrest, they re-enter the cell cycle and pass through a limited number o f cell divisions, and finally differentiate into fully mature adipocytes. Adipogenesis is controlled by a complex cross-talk between positive and negative regulators, such as hormonal and nutritional stimuli, that change the activity of a selected set of transcription factors. Regulation o f adipogenesis is crucial to keep the body energy balance because a limited amount o f adipose tissue, lipodystrophy, or an excess o f adipose tissue, such as occurs in obesity, lead to profound metabolic dysfunctions and disease. Keywords: adipocyte; differentiation; regulation; transcription. Ann Med 2001; 33: 556-561. Introduction Mesenchymal stem cells are the precursors for three distinct cell lineages: adipocytes, myocytes and chondrocytes. The process in which such pluripotent mesenchymal cells give rise to adipoblasts is known as lineage determination, while the process of transition from preadipocytes to adipocytes is referred to as adipocyte differentiation. Following exponential growth of precursors, and at the moment of con- fluence, preadipocytes enter into a cell cycle arrest. To start the terminal phase of adipocyte differentiation, cells re-enter the cell cycle and undergo a limited number of cell divisions known as the clonal expan- sion of preadipocytes. Terminal differentiation is then ~~ From the lnstitut de Genetique et de Biologie Molkculaire et Cellulaire (IGBMC), CNRSIINSERM, Universite Louis Pasteur, Illkirch, CU de Strasbourg, France. Correspondence: Johan Auwerx, MD, IGBMC, PO Box 163, 67404 Illkirch, France. E-mail: [email protected], Fax: +33 388 653201. characterized by rounding up of the fibroblast-like preadipocytes and accumulation of lipid vacuoles. Our current understanding of transcriptional regu- lation of adipogenesis is primarily based on in vitro studies with different white preadipocyte cell lines such as the mouse cell lines 3T3-L1, 3T3-F442A and Ob17. These preadipocytes, when cultured in the presence of methylisobutylxanthine, dexamethasone, insulin and fetal serum, differentiate into mature adipocytes. These cell lines induced to differentiate in vitro do not, however, recapitulate the complexity of adipose tissue development because this tissue is composed not only of white and brown adipocytes, but also of vascular endothelial cells, smooth muscle cells, fibroblasts, local mast cells and macrophages. In addition, adipose tissue differs in its adipogenic potential and lipolytic response depending on its regional distribution. Transcriptional regulation of early phases of adipogenesis The mechanisms that regulate the switch between cell growth and differentiation in the adipogenic lineage are not yet completely elucidated; however, some molecular factors have been identified recently. Cell cycle regulators The appropriate cell cycle regulation is critical for the mitotic clonal expansion phase and is assured by the coordinated action of the E2F transcription factors and the family of retinoblastoma proteins. The E2F family of transcription factors are DNA-binding proteins that bind as a heterodimeric complex com- posed of an E2F protein and a DP protein (1, 2). E2F proteins are not only important to drive the re-entry into the cell cycle, but they also induce peroxisome proliferator activated receptor (PPAR)y expression early during the clonal expansion phase of adipocyte differentiation. Recently, our laboratory identified an E2F response element in the promotor of PPARyl gene, which is a key player in regulating downstream www.annmed.org 0 The Finnish Medical Society Duodecim, Ann Med 2001; 33: 556-561 Ann Med Downloaded from informahealthcare.com by Michigan University on 10/29/14 For personal use only.

Upload: johan

Post on 06-Mar-2017

213 views

Category:

Documents


0 download

TRANSCRIPT

556

+ SPECIAL SECTION: MOLECULAR MECHANISMS IN OBESITY AND VISCERAL OBESITY +

Regulation of adipocyte differentiation Hana Koutnikova and Johan Auwerx

Once multipotent mesechymal cells become committed t o the adipoblast lineage, adipogenesis, the process o f preadipocytes differentiation into adipocytes is initiated. This process starts with a phase o f exponential growth o f adipoblasts. Following confluence o f these adipoblasts, the cells enter into a cell cycle arrest, they re-enter the cell cycle and pass through a limited number o f cell divisions, and finally differentiate into ful ly mature adipocytes. Adipogenesis is controlled by a complex cross-talk between positive and negative regulators, such as hormonal and nutritional stimuli, that change the activity o f a selected set o f transcription factors. Regulation o f adipogenesis is crucial t o keep the body energy balance because a limited amount o f adipose tissue, lipodystrophy, or an excess o f adipose tissue, such as occurs in obesity, lead t o profound metabolic dysfunctions and disease.

Keywords: adipocyte; differentiation; regulation; transcription.

Ann Med 2001; 33: 556-561.

Introduction

Mesenchymal stem cells are the precursors for three distinct cell lineages: adipocytes, myocytes and chondrocytes. The process in which such pluripotent mesenchymal cells give rise to adipoblasts is known as lineage determination, while the process of transition from preadipocytes to adipocytes is referred to as adipocyte differentiation. Following exponential growth of precursors, and at the moment of con- fluence, preadipocytes enter into a cell cycle arrest. To start the terminal phase of adipocyte differentiation, cells re-enter the cell cycle and undergo a limited number of cell divisions known as the clonal expan- sion of preadipocytes. Terminal differentiation is then

~~

From the lnstitut de Genetique et de Biologie Molkculaire et Cellulaire (IGBMC), CNRSIINSERM, Universite Louis Pasteur, Illkirch, CU de Strasbourg, France.

Correspondence: Johan Auwerx, MD, IGBMC, PO Box 163, 67404 Illkirch, France. E-mail: [email protected], Fax: +33 388 653201.

characterized by rounding up of the fibroblast-like preadipocytes and accumulation of lipid vacuoles.

Our current understanding of transcriptional regu- lation of adipogenesis is primarily based on in vitro studies with different white preadipocyte cell lines such as the mouse cell lines 3T3-L1, 3T3-F442A and Ob17. These preadipocytes, when cultured in the presence of methylisobutylxanthine, dexamethasone, insulin and fetal serum, differentiate into mature adipocytes. These cell lines induced to differentiate in vitro do not, however, recapitulate the complexity of adipose tissue development because this tissue is composed not only of white and brown adipocytes, but also of vascular endothelial cells, smooth muscle cells, fibroblasts, local mast cells and macrophages. In addition, adipose tissue differs in its adipogenic potential and lipolytic response depending on its regional distribution.

Transcriptional regulation of early phases of adipogenesis

The mechanisms that regulate the switch between cell growth and differentiation in the adipogenic lineage are not yet completely elucidated; however, some molecular factors have been identified recently.

Cell cycle regulators

The appropriate cell cycle regulation is critical for the mitotic clonal expansion phase and is assured by the coordinated action of the E2F transcription factors and the family of retinoblastoma proteins. The E2F family of transcription factors are DNA-binding proteins that bind as a heterodimeric complex com- posed of an E2F protein and a DP protein (1, 2). E2F proteins are not only important to drive the re-entry into the cell cycle, but they also induce peroxisome proliferator activated receptor (PPAR)y expression early during the clonal expansion phase of adipocyte differentiation. Recently, our laboratory identified an E2F response element in the promotor of PPARyl gene, which is a key player in regulating downstream

www.annmed.org 0 The Finnish Medical Society Duodecim, Ann Med 2001; 33: 556-561

Ann

Med

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Mic

higa

n U

nive

rsity

on

10/2

9/14

For

pers

onal

use

onl

y.

REGULATION OF ADIPOCYTE DIFFERENTIATION 557

events in adipocyte differentiation (L Fajas, J Auwerx, unpublished observation, IGBMC, 2001). Interaction with hypophosphorylated retinoblastoma protein (Rb) or other pocket proteins (p107, p130) inhibits the action of E2F-DP heterodimers in the G, phase of cell cycle. This inhibitory effect is relieved once the pocket proteins are phosphorylated by cyclin-dependent kinase (cdk) ( 3 ) , which occurs early during adipocyte differentiation (4). The active E2F-DP heterodimer is inhibited by a feedback loop by ligand-activated PPARy through a decrease in gene expression of the serine-threonine phosphatase PP2A which increases the phosphorylation of DP-1 (5).

Interestingly, the Rb protein plays a dual role in cell cycle withdrawal and differentiation (6). First, Rb inactivation initially enables clonal expansion by stimulating E2F activity, whereas subsequent growth arrest after this expansion phase requires active Rb positively influencing adipocyte differentiation (6). In addition to these effects on the cell cycle, Rb binds to and stimulates the activity of the CCAATIenhancer binding proteins a and p (CIEBPa and p), which are the key downstream transcription factors implicated in terminal adipocyte differentiation (7).

The permanent exit from the cell cycle establishes the irreversible commitment of cells to undergo terminal adipocyte differentiation, and this transition is characterized by changes in gene expression of cdk inhibitors (cdkI) p18, p21 and p27 (8) . In the case of p21, PPARy may be responsible for this direct regulation (8) .

Antiadipogenic regulators

Induction of the early phases of adipogenesis requires a decreased activity of a number of antiadipogenic factors, which in general seem to block the induction of the transcriptional activity of PPARy and CIEBPa. The Wnt family of paracrine and autocrine factors, which signal through Frizzled seven-transmembrane receptor, have a critical role during adipogenesis. Activation of the Wnt signalling pathway blocks p- catenin degradation, resulting in p-catenin translocation into the nucleus and its association with the T-cell factor/lymphoid-enhancer factor (TCFILEF) tran- scription factors, which then regulate the expression of Wnt target genes. In preadipocytes, Wnt signalling maintains the cells in undifferentiated state through inhibition of CIEBPa and PPARy expression (9) . A decrease in Wnt signalling allows initiation of adipo- genesis, and this can even cause a transdifferentiation of myoblasts into adipocytes (9) . The GATA-2 and GATA-3 are other transcription factors the down- regulation of which releases a direct suppression of PPARy and is also a prerequisite for the terminal differentiation (10). Retinoic acid (RA) also inhibits adipogenesis by blocking the induction of C/EBPa

Key message

Regulation o f adipogenesis is crucial to keep the body energy balance because a limited amount of adipose tissue, lipodystrophy, or an excess o f adipose tissue, such as occurs in obesity, both lead to profound metabolic dys- functions and disease.

and PPARy (11, 12). This negative regulation of C/ EBPa and PPARy expression is mediated by the retinoic acid receptor (RAR) as a consequence of interference with CIEBPP action. In addition, RA can inhibit PPARy target gene induction by favouring the formation of the nonpermissive RARhetinoid X receptor (RXR) heterodimer over the PPAR/RXR dimer (13). Another player is Pref-1 (preadipocyte factor-1), a plasma membrane six epidermal growth factor repeats domain-containing inhibitor of adipocyte differentiation that is highly expressed in 3T3-Ll preadipocytes (14). Its gene expression is repressed by a synthetic glucocorticoid, dexamethasone, which is an adipogenetic agent (15). Pref-1 is proteolytically processed to generate an inhibitory soluble form with a molecular weight of 50 kDa (16). Finally, the tumour necrosis factor alpha (TNF-a) reverses adipocyte differ- entiation and inhibits adipogenesis by down-regulating the expression of PPARy and CIEBPa (17, 18).

The adipocyte enhancer binding protein (AEBP)l is a transcriptional repressor with carboxypeptidase activity (19) that is down-regulated during adi- pogenesis. AEBPl binds the y5 subunit of a hetero- trimeric G protein, and this interaction leads to a decrease in AEBPl transcriptional repression (20). In addition, AEBPl binds to mitogen-activated protein kinase (MAPK) while probably serving as a nuclear anchor for the MAPK nuclear accumulation (21). As a consequence, the nuclear accumulation of MAPK may modify phosphorylation status of different tran- scription factors and define the genes to be switched on, thereby influencing cell growth and differentiation in the adipogenic lineage. The effects of MAPK on adipogenesis are, however, more complex. The insulin- like growth factor (1GF)-I stimulates mitogenesis in proliferating preadipocytes, but when cells reach confluence and become growth arrested, IGF-I stimu- lates differentiation into adipocytes. IGF-I signals through the IGF-I receptor-mediated tyrosine phos- phorylation of Shc leading to MAPK activation, and this activation is lost as preadipocytes begin to differentiate (22). Thus the down-regulation of MAPK activity is in this case linked to the switch leading to adipocyte differentiation.

www.annmed.org 0 The Finnish Medical Society Duodecim, Ann Med 2001; 33: 556-561

Ann

Med

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Mic

higa

n U

nive

rsity

on

10/2

9/14

For

pers

onal

use

onl

y.

558 KOUTNIKOVA AUWERX

Proadipogenic factors in the early phase of adipocyte differentiation

In addition to insulin or glucocorticoids, few pro- adipogenic factors are known. Notch-1 is one of these positive regulators of adipogenesis. It is a trans- membrane receptor for epidermal growth factor-like proteins that plays a role in cell-to-cell signalling during adipocyte differentiation. An inhibition of its expression leads to a decrease in the PPARy and 6 expression (23). Also, the extracellular leukaemia inhibitory factor (LIF) and its receptor play a positive role during adipocyte differentiation of embryonic stem cells as LIF stimulates expression of the early adipogenic transcription factors C/EBPP and C/EBP6 via the MAPK cascade (24).

Transcriptional regulation of terminal differentiation

In contrast to these early phases of adipogenesis, the terminal phase of adipogenesis is relatively well understood, and the key players in this process are the family of C/EBP transcription factors, PPARy, and the basic helix-loop-helix factor adipocyte determination and differentiation factor l/sterol regulatory element- binding protein l c (ADDUSREBPlc). The C/EBPs belongs to the basic-leucine zipper class of tran- scription factors, which are composed of C-terminal leucine zipper dimerization domain and a basic DNA- binding domain. Four members of the family, C/ EBPa, p, 6 and CHOP-10 (C/EBP homologous protein-lo), are expressed during adipogenesis, and each plays a distinct role (25-27). CEBP p and 6 are expressed early in adipogenesis and play critical roles in initiating the terminal phase (28). In vzvo, in double C/EBPP and 6 knock-out embryos a defect in differ- entiation of preadipocytes to adipocytes is observed. Only about one-fourth of these animals survive to adulthood having impaired adipogenesis and by con- sequence reduced fat mass (29). Two important downstream genes of C/EBPP and 6, ie CIEBPa and PPARy, however, are still normally expressed in these animal models. The transcriptional activity of C/EBPP is positively regulated by a direct interaction with high-mobility group I nonhistone chromosomal protein HMGI(Y), an architectural factor with a role in transcription (30). The importance of this interaction for the regulation of adipocyte differentiation is confirmed in vivo as a targeted disruption of another family member, the HMGIC, leads to a severe deficiency in adipose tissue (31). The transcriptional activity of CEBPP (also a) is negatively controlled by CHOP-10, a dominant-negative CEBP family member (32). CHOP-10 can transiently sequester C/EBPP by

heterodimerization. Once CHOP-1 0 expression is down-regulated, C/EBPP is released from the re- pressive effect and is able to induce adipogenesis (33).

The expression of CEBPP and 6 lose importance during the later phases of adipocyte differentiation, which is mainly coordinated by CIEBPa and PPARy that cooperate to establish the phenotype of mature adipocytes (28, 34). PPARy belongs to the nuclear hormone receptor family of ligand-activated tran- scription factors that act together with its hetero- dimerization partner, the retinoid X receptor. The two additional forms of PPAR, ie PPARcl and WARP (6) , are also expressed in adipose tissue, albeit to a lower extent. Although it has been reported that PPARa, but not PPARG, can drive adipogenesis, it is much less effective than PPARy (35). Ectopic expression of PPARy in NIH-3T3 fibroblasts leads to adipogenesis in the presence of its natural or synthetic ligands, such as prostaglandin derivatives, polyunsaturated fatty acids or thiazolidinediones, respectively (36-38). In vivo, PPARy deficiency is lethal because of a defect in development of the placenta and the heart (39, 40). When the PPARyI- embryos are rescued by the use of tetraploid strategy, animals suffer from severe lipodys- trophy, confirming the important role of PPARy in adipogenesis. Additional evidence for a role of PPARy in adipogenesis was obtained by the lack of contri- bution of WART’- embryonic stem cells to white adipose tissue formation in chimeric animals (40). Furthermore, the heterozygote PPARy+’- animals are protected from adipocyte hypertrophy and insulin resistance induced by high-fat diet (41). In addition, in human, several mutations in PPARy were described. The most prevalent P12A mutation in the adipose tissue-specific PPARy2 isoform, which attenuates PPARy activity, is associated with a decreased body mass index and improved insulin sensitivity (42, 43). In contrast, two mutations in the ligand-binding domain of PPARy, the P467L and V290M, lead to a severe insulin resistance, type 2 diabetes and hyper- tension (44). In combination, all these data thus suggest that PPARy serves as a thrifty gene by enabling energy storage in times of plenty and by safeguarding energy stores at the time of carence (45).

CIEBPa regulates terminal adipocyte differentiation by turning on fat-specific genes required for the synthesis, uptake, and storage of long-chain fatty acids. CIEBPa is antimitotic, and its premature expression would block the mitotic clonal expansion required for differentiation. Therefore, C/EBPa is expressed relatively late during adipogenesis. This rather late induction is controlled by the nuclear factor C/EBP undifferentiated protein (CUP), an isoform of activator protein-2a (AP-2a), which re- presses the CEBPa gene promoter leading to its silencing until late in the differentiation process (46). The CUP regulatory element overlaps the binding site

www.annmed.org 0 The Finnish Medical Society Duodecim, Ann Med 2001; 33: 556-561

Ann

Med

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Mic

higa

n U

nive

rsity

on

10/2

9/14

For

pers

onal

use

onl

y.

REGULATION OF ADIPOCYTE DIFFERENTIATION 559

for Spl or Sp3, strong transcriptional activators of the CEBPa, and only one of these factors can bind the Sp element at a time. Following down-regulation of CUP during adipocyte differentiation, the Sp proteins that are stably expressed throughout adipogenesis can bind and induce CEBPa expression.

The in vivo role of CIEBPa was confirmed by using knock-out mice. C/EBPa-I- mice do not store hepatic glycogen, fail to accumulate lipids and die from hypoglycaemia on postnatal day 1 (47). The mRNA expression of glycogen synthase and uncoupling protein is decreased in these animals while the gene expression of two gluconeogenic enzymes, phospho- enolpyruvate carboxykinase and glucose-6-phosphatase, is delayed. Further experiments with fibroblasts from C/EBPa4- mice have shown that the differentiation of adipose tissue can be achieved by a positive cross- regulation between virally provided C/EBPa and PPARy (48). These CEBPa-’- cells also show a complete absence of insulin-stimulated glucose trans- port, secondary to reduced gene expression and tyrosine phosphorylation of the insulin receptor and

ADD-l/SREBPlc also plays a role in terminal adipocyte differentiation (49, 50). ADD-1hREBPlc is a membrane-bound basic helix-loop-helix leucine zip-

IRS-I.

per transcription factor, which is activated through a two-step intramolecular proteolytic cleavage facili- tated by the SREBP cleavage activation protein, the SCAP (51). This proteolysis is regulated by the sterol content within the cell through the sterol-sensing domain at the C-terminus of SREBP. There are at least two mechanisms by which SREBP functions during the phase of terminal differentiation. First, ADD1/ SREBPlc controls the production of endogenous ligand(s) for PPARy regulating thus its transcriptional activity (52). Second, ADDUSREBPlc induces directly PPARy activity (53). The activity of ADDUSREBPlc is negatively regulated by the inhibitor of DNA binding Id2 and Id3, two dominant negative members of bHLH family that are down-regulated during adipogenesis (54, 55). These proteins interact with ADDUSREBPl and decrease its DNA binding in preadipocytes. The in vivo role of ADDUSREBPlc was analysed in mice models that over-express SREBP- l c in adipose tissue. These animals have smaller depots of fully differentiated white adipose tissue and hypertrophic brown adipose tissue while showing, unexpectedly, the features of congenital generalized lipodystrophy (56). This finding shows the complexity of adipogenesis and limitation of a single-gene transgenic animal model.

Regulators

Target genes

GLUT4

SCDl UCP TNFa aP2 IR Resistin LPL IRS-1 ACS

GS PAR K l F c K IL

Flgure 1. Schematic representation of the regulation of adipocyte differentiation by different transcriptional or hormonal factors. The balance between positive and negative regulatory players will define which target genes will be switched on or off and whether the cell will undergo differentiation or remain in the nondifferentiated state. ACS, acyl-coenzyme A synthase; AEBPl , adipocyte enhancer binding protein 1 ; P-AR, beta adrenergic receptor; C/EBPa,P,G, CCAAT/enhancer-binding protein alpha, beta and delta; CHOP1 0, C/EBP homologous protein-1 0; CUP, C/EBP undifferentiated protein; GLUT4, glucose transporter-4; GS, glycogen synthase; GH, growth hormone; HMGIM, high-mobility group I nonhistone chromosomal protein; IL, interleukin; ld2,3, inhibitor of DNA binding 2 and 3; IR, insulin receptor; IRS-1, insulin receptor substrate 1 ; LIF, leukemia inhibitory factor; LPL, lipoprotein lipase; PEPCK, phosphoenolpyruvate carboxykinase; PPARy, peroxisome proliferator-activated receptor gamma; Pref-1 , preadipocyte factor-1 ; RAR, retinoic acid receptor; Rb, Retinoblastoma protein; RXR, retinoid X receptor; SCDl , stearoyl-coenzyme A desaturase 1 ; SREBP1 c, sterol regulatory element binding protein 1 c; TNF-a, tumour necrosis factor alpha; UCP, uncoupling protein.

www.annmed.org 0 The Finnish Medical Society Duodecim, Ann Med 2001; 33: 556-561

Ann

Med

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Mic

higa

n U

nive

rsity

on

10/2

9/14

For

pers

onal

use

onl

y.

560 KOUTNIKOVA AUWERX

Conclusion

During the last 10 years our understanding of the process leading to adipocyte determination and differ- entiation has improved significantly (summarised in Fig 1). Adipocyte differentiation is dependent on a balance between factors that influence the adipocyte differentiation process either positively or negatively. The nuclear transcription factors of the C/EBP family and PPARy have emerged as the key coordinating factors of this differentiation process to which many more factors contribute. This new insight into the

References

1. Krek W, Livingston DM, Shirodkar S. Binding to DNA and the retinoblastoma gene product promoted by complex formation of different E2F family members. Science 1993;

2. Girling R, Partridge JF, Bandara LR, Burden N, Totty NF, Hsuan JJ, et al. A new component of the transcription factor DRTFllE2F. Nature 1993; 365: 468.

3. Jacks T, Weinberg RA. The expanding role of cell cycle regulators. Science 1998; 280: 1035-6.

4. Richon V, Lyle RE, McGehee REJ. Regulation and expression of retinoblastoma proteins p107 and p130 during 3T3-Ll adipocyte differentiation. J Biol Chem 1997; 272: 10117- 24.

5. Altiok S, Xu M, Spiegelman B. PPARgamma induces cell cycle withdrawal: inhibition of E2F/DP DNA-binding activity via down-regulation of PP2A. Genes Deu 1997; 11: 1987- 98.

6. Classon M, Kennedy BK, Mulloy R, Harlow E. Opposing roles of pRB and p107 in adipocyte differentiation. Proc Natl Acad Sci U S A 2000; 97: 10826-31.

7. Chen P, Riley DJ, Chen Y, Lee W. Retinoblastoma protein positively regulates terminal adipocyte differentiation through direct interaction with CEBPs. Genes Deu 1996; 10: 2794- 804.

8. Morrison RF, Farmer SR. Role of PPARgamma in regulating a cascade expression of cyclin-dependent kinase inhibitors, plS(INK4c) and pZl(Wafl/Cipl), during adipogenesis. J Biol Chem 1999; 274: 17088-97.

9 . Ross SE, Hemati N, Longo KA, Bennett CN, Lucas PC, Erickson RL, et al. Inhibition of adipogenesis by Wnt signaling. Science 2000; 289: 950-3.

10. Tong Q, Dalgin G, Xu H, Ting CN, Leiden JM, Hotamisligil GS. Function of GATA transcription factors in preadipocyte- adipocyte transition. Science 2000; 290: 134-8.

11. Schwarz EJ, Reginato MJ, Shao D, Krakow SL, Lazar MA. Retinoic acid blocks adipogenesis by inhibiting CEBPbeta- mediated transcription. Mol Cell Biol 1997; 17: 1552-61.

12. Shao D, Lazar MA. Peroxisome proliferator activated receptor gamma, CCAATknhancer-binding protein alpha, and cell cycle status regulate the commitment to adipocyte differentiation. J Biol Chem 1997; 272: 21473-8.

13. DiRenzo J, Soderstrom M, Kurokawa R, Ogliastro MH, Ricote M, Ingrey S, et al. Peroxisome proliferator-activated receptors and retinoic acid receptors differentially control the interactions of retinoid X receptor heterodimers with ligands, coactivators and corepressors. Mol Cell Biol 1997;

14. Smas CM, Sul HS. Pref-1, a protein containing EGF-like

262: 1557-60.

17: 2166-76.

adipocyte differentiation process has an immediate impact on human health as it will provide potential new ways to treat diseases such as obesity, lipo- dystrophy, waisting syndrome AIDS, insulin resistance or type 2 diabetes, which are all associated with adipocyte dysfunction.

This work was supported by grants of CNRS, INSERM, CHU of Strasbourg, ARC (contract no 9943), the Juvenile Diabetes Foundation (1-1999-819), the European community RTD pro- gramme (QLGl-CT-1999-00674), Ligue Nationale Contre le Cancer, and the Human Frontier Science Programme (RG00411 1999-M).

repeats, inhibits adipocyte differentiation. Cell 1993; 73:

15. Smas CM, Chen L, Zhao L, Latasa MJ, Sul HS. Tran- scriptional repression of pref-1 by glucocorticoids promotes 3T3-Ll adipocyte differentiation. J Biol Chem 1999; 274: 12632-41.

16. Smas CM, Chen L, Sul HS. Cleavage of membrane- associated pref-1 generates a soluble inhibitor of adipocyte differentiation. Mol Cell Biol 1997; 17: 977-88.

17. Xing H, Northrop JP, Grove JR, Kilpatrick KE, Su JL, Ringold GM. TNFalpha-mediated inhibition and reversal of adipocyte differentiation is accompanied by suppressed expression of PPARgamma without effects on Pref-1 expression. Endocrinology 1997; 138: 2776-83.

18. Kurebayashi S, Sumitani S, Kasayama S, Jetten AM, Hirose T. TNF-alpha inhibits 3T3-Ll adipocyte differentiation without downregulating the expression of ClEBPbeta and delta. Endocrin J 2001; 48: 249-53.

19. He GP, Muise A, Li AW, Ro HS. A eucaryotic tran- scriptional repressor with carboxypeptidase activity. Nature

20. Park J-G, Muise A, He G-P, Kim S-W, Ro H-S. Tran- scriptional regulation by the gamma5 subunit of a hetero- trimeric G protein during adipogenesis. EMBO J 1999; 18:

21. Kim SW, Muise AM, Lyons PJ, Ro HS. Regulation of adipogenesis by a transcriptional repressor that modulates MAPK activation. J Biol Chem 2001; 276: 10199-206.

22. Boney CM, Gruppuso PA, Faris RA, Frackelton AR Jr. The critical role of Shc in insulin-like growth factor-I-mediated mitogenesis and differentiation in 3T3-Ll preadipocytes. Mol Endocrinol2000; 14: 805-13.

23. Garces C , Ruiz-Hidalgo MJ, de Mora JF, Park C, Miele L, Goldstein J, et al. Notch-1 controls the expression of fatty acid-activated transcription factors and is required for adipogenesis. J Biol Chem 1997; 272: 29729-34.

24. Auben J, Dessolin S, Belmonte N, Li M, McKenzie FR, Staccini L, et al. Leukemia inhibitory factor and its receptor promote adipocyte differentiation via the mitogen-activated protein kinase cascade. J Biol Chem 1999; 274: 24965-72.

25. Christy RJ, Yang W, Ntambi JM, Geiman DE, Landschulz WH, Friedman AD, et al. Differentiation-induced gene expression in 3T3-Ll preadipocytes: CCAATknhancer binding protein interacts with and activates the promoters of two adipocyte-specific genes. Genes Dev 1989; 3: 1323-35.

26. Herrera R, Ro HS, Robinson GS, Xanthopoulos KG, Spiegelman BM. A direct role for ClEBP and the AP-I- binding site in gene expression linked to adipocyte differ-

725-34.

1995; 378: 92-6.

4004-12.

www.annmed.org 0 The Finnish Medical Society Duodecim, Ann Med 2001; 33: 556-561

Ann

Med

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Mic

higa

n U

nive

rsity

on

10/2

9/14

For

pers

onal

use

onl

y.

REGULATION OF ADIPOCYTE DIFFERENTIATION 561

entiation. Mol Cell Biol 1989; 9: 5331-9. 27. Cao Z, Umek RM, McKnight SL. Regulated expression of

three CEBP isoforms during adipose conversion of 3T3-Ll cells. Genes Dev 1991; 5: 1538-52.

28. Yeh WC, Cao Z, Classon M, McKnight S. Cascade regu- lation of terminal adipocyte differentiation by three members of the C/EBP family of leucine zipper proteins. Genes Dev

29. Tanaka T, Yoshida N, Kishimoto T, Akira S. Defective adipocyte differentiation in mice lacking the CEBPbeta and/ or C/EBPdelta gene. Embo J 1997; 16: 7432-43.

30. Melillo RM, Pierantoni GM, Scala S, Battista S, Fedele M, Stella A, et al. Critical role of the hmgi(y) proteins in adipocytic cell growth and differentiation. Mol Cell Biol

31. Anand A, Chada K. In vivo modulation of Hmgic reduces obesity. Nat Genet 2000; 24: 377-80.

32. Batchvarova N, Wang X-Z, Ron D. Inhibition of adipo- genesis by the stress-induced protein CHOP (Gadd153).

33. Tang QQ, Lane MD. Role of C/EBP homologous protein (CHOP-10) in the programmed activation of CCAAT/ enhancer-binding protein-beta during adipogenesis. Proc Natl Acad Sci U S A 2000; 97: 12446-50.

34. Wu Z, Xie Y, Bucher NLR, Fanner SR. Conditional ectopic expression of C/EBPb in NIH-3T3 cells induces PPARgamma and stimulates adipogenesis. Genes Dev 1995; 9: 2350-63.

35. Castillo G , Brun RP, Rosenfield JK, Hauser S, Park CW, Troy AE, et al. An adipogenic cofactor bound by the differentiation domain of PPARgamma. Embo J 1999; 18:

36. Forman BM, Tontonoz P, Chen J, Brun RP, Spiegelman BM, Evans RM. 15-Deoxy-Deltal2,14 prostaglandin 52 is a ligand for the adipocyte determination factor PPARgamma. Cell 1995; 83: 803-12.

37. Tontonoz P, Hu E, Spiegelman BM. Stimulation of adipo- genesis in fibroblasts by PPARgamma2, a lipid-activated transcription factor. Cell 1994; 79: 1147-56.

38. Kliewer SA, Lenhard JM, Willson TM, Patel I, Morris DC, Lehman JM. A prostaglandin 52 metabolite binds peroxi- some proliferator-activated receptor gamma and promotes adipocyte differentiation. Cell 1995; 83: 813-9.

39. Barak Y, Nelson MC, Ong ES, Jones YZ, Ruiz-Lozano P, Chien KR, et al. PPARgamma is required for placental, cardiac, and adipose tissue development. M o l Cell 1999; 4:

40. Rosen ED, Sarraf P, Troy AE, Bradwin G , Moore K, Milstone DS, et al. PPARgamma is required for the differentiation of adipose tissue in vivo and in vitro. M o l Cell 1999; 4: 611-7.

41. Kubota N, Terauchi Y, Miki H, Tamemoto H, Yamauchi T, Komeda K, et al. PPARgamma mediates high-fat diet- induced adipocyte hypertrophy and insulin resistance. Mol Cell 1999; 4: 597-609.

42. Deeb S , Fajas L, Nemoto M, Laakso M, Fujimoto W, Auwerx J. A Pro 12 Ala substitution in the human peroxisome proliferator-activated receptor gamma2 is associ-

1995; 9: 168-81.

2001; 21: 2485-95.

E M B O J 1995; 14: 4654-61.

3676-87.

585-95.

ated with decreased receptor activity, improved insulin sensitivity, and lowered body mass index. Nat Genet 1998;

43. Altshuler D, Hirschhorn JN, Klannemark M, Lindgren CM, Vohl MC, Nemesh J, et al. The common PPARgamma Prol2AIa polymorphism is associated with decreased risk of type 2 diabetes. Nat Genet 2000; 26: 76-80.

44. Barroso I, Gurnell M, Crowley VE, Agostini M, Schwabe JW, Soos MA, et al. Dominant negative mutations in human PPARgamma associated with severe insulin resistance, diabetes mellitus and hypertension. Nature 1999; 402: 880-3.

45. Auwerx J. PPARgamma, the ultimate thrifty gene. Dia- betologia 1999; 42: 103349.

46. Jiang MS, Lane MD. Sequential repression and activation of the CCAAT enhancer-binding protein-alpha (CEBPalpha) gene during adipogenesis. Proc Natl Acad Sci U S A 2000;

47. Wang ND, Finegold MJ, Bradley A, Ou CN, Abdelsayed SV, '

Wilde MD, et al. Impaired energy homeostasis in CEBPalpha knockout mice. Science 1995; 269: 1108-12.

48. Wu Z, Rosen ED, Brun R, Hauser S, Adelmant G , Troy AE, et al. Cross-regulation of C/EBPalpha and PPARgamma controls the transcriptional pathway of adipogenesis and insulin sensitivity. Mol Cell 1999; 3: 143-50.

49. 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

50. Kim JB, Spiegelman BM. ADDl/SREBPl promotes adi- pocyte differentiation and gene expression linked to fatty acid metabolism. Genes Dev 1996; 10: 1096-107.

51. Brown MS, Goldstein JL. The SREBP pathway: regulation of cholesterol metabolism by proteolysis of a membrane- bound transcription factor. Cell 1997; 89: 331-40.

52. Kim JB, Wright HM, Wright M, Spiegelman BM. ADDlI SREBPl activates PPARgamma through the production of endogeneous ligand. Proc Natl Acad Sci U S A 1998; 95: 4333-7.

53. Fajas L, Schoonjans K, Gelman L, Kim JB, Najib J, Martin G , et al. Regulation of PPARgamma expression by ADD-1/ SREBP-1: implications for adipocyte differentiation and metabolism. Mol Cell Biol 1999; 19: 5495-503.

54. Moldes M, Lasnier F, Feve B, Pairault J, Djian P. Id3 prevents differentiation of preadipose cells. Mol Cell Biol

55. Moldes M, Boizard M, Liepvre XL, Feve B, Dugail I, Pairault J. Functional antagonism between inhibitor of DNA binding (Id) and adipocyte determination and differentiation factor llsterol regulatory element-binding protein-lc (ADDl/ SREBP-lc) trans-factors for the regulation of fatty acid synthase promoter in adipocytes. Biochem J 1999; 344:

56. Shimomura I, Hammer RE, Richardson JA, Ikemoto S, Bashmakov Y, Goldstein JL, et al. Insulin resistance and diabetes mellitus in transgenic mice expressing nuclear SREBP-lc in adipose tissue: a model for congenital general- ized lipodystrophy. Genes Dev 1998; 12: 3182-94.

20: 284-7.

97: 12519-23.

1993; 13: 4753-9.

1997; 17: 1796-804.

873-80.

www.annmed.org 0 The Finnish Medical Society Duodecim, Ann Med 2001; 33: 556-561

Ann

Med

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Mic

higa

n U

nive

rsity

on

10/2

9/14

For

pers

onal

use

onl

y.