Sie sind auf Seite 1von 13

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 281, NO. 10, pp.

6608 –6615, March 10, 2006


© 2006 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.

Down-regulation of Histone Deacetylases Stimulates


Adipocyte Differentiation*□ S

Received for publication, August 15, 2005, and in revised form, November 3, 2005 Published, JBC Papers in Press, January 5, 2006, DOI 10.1074/jbc.M508982200
Eung Jae Yoo1,2, Jun-Jae Chung1,2, Sung Sik Choe2, Kang Ho Kim2, and Jae Bum Kim2,3
From the Department of Biological Sciences and Research Center for Functional Cellulomics, Seoul National University,
Seoul 151-742, South Korea

Specific cell type differentiation is driven by programmed regu- lating histones and other proteins, such as transcription factors (10, 11).
lation of gene expression, which is the result of coordinated modu- Until now, five yeast and eleven human HDACs have been identified,
lation of the transcription machinery and chromatin-remodeling which are classified into three classes (12). Class I HDACs consist of
factors. We present evidence here that the down-regulation of his- HDAC1, -2, -3, and -8. HDAC 4, -5, -6, -7, -9, and -10 belong to the class
tone deacetylases is an important process during adipocyte differ- II HDACs. Class III HDACs are members of the sirtuin family of
entiation. In 3T3-L1 cells, histone hyperacetylation was selectively HDACs (13–17). In addition, a new member of the HDAC family,
induced at the promoter regions of adipogenic genes during adipo- HDAC11, has been recently identified (18). Class I HDACs are
cyte differentiation. Interestingly, this was accompanied by a dra- expressed ubiquitously, whereas class II HDACs are abundantly
matic decrease in the expression level of several histone deacety- expressed in heart, skeletal muscle, and brain (12). The major function
lases including HDAC1, -2, and -5 and a reduction in overall histone

Downloaded from http://www.jbc.org/ by guest on May 7, 2017


of HDACs is to remove acetyl groups from histones, which results in
deacetylase enzyme activity. Inhibition of histone deacetylase activ- condensation of the chromatin structure. This, in turn, diminishes the
ity using sodium butyrate resulted in stimulation of adipogenic gene access of transcription factors to the target DNA and ultimately leads to
expression and adipocyte differentiation. Consistently, HDAC1 transcriptional repression.
knock-down promoted adipogenesis whereas HDAC1 overexpres- Chromatin modifications, notably histone acetylation and deacetyla-
sion attenuated adipocyte differentiation in 3T3-L1 cells. Together, tion, are crucial for the regulation of gene expression and development
these results suggest that the regulation of not only adipogenic tran-
in eukaryotes (19, 20). During tissue differentiation, early inductive pro-
scription factors, but also chromatin-modifying enzymes is crucial
cesses that determine cell fate leave traces at particular genes in the form
for the execution of bona fide adipogenesis.
of histone modifications. HATs and HDACs have been shown to
respond to signals that regulate cell differentiation and directly modu-
late tissue-specific gene expression. For example, studies of myogenesis
The architecture of the eukaryotic chromatin is dynamically modu-
have suggested a role for histone-modifying enzymes in coordinating
lated by post-translational modifications of the histones, including
the activation and repression of genes involved in differentiation proc-
acetylation, methylation, phosphorylation, and ubiquitination (1). The
ess (21–23). HDAC1 and members of the class II HDACs including
changes in nucleosome structure influence gene expression by modu-
HDAC4, -5, and -6 inhibit MyoD and Mef2, respectively, to block mus-
lating the accessibility of the promoter regions to specific transcription
factors. In particular, acetylation of histones H3 (Lys9 and Lys14) and H4 cle cell differentiation (24 –27).
(Lys8 and Lys12), mediated by histone acetyltransferases (HATs)4 such Adipogenesis is the process by which undifferentiated precursor cells
as p300, CBP, and P/CAF, is associated with transcriptional activation differentiate into adipocytes. In vitro studies using 3T3-L1 and 3T3-
(2, 3). Methylation of H3 Lys4, H4 Arg3, and phosphorylation of H3 Ser10 F442A cells have elucidated the complex transcriptional cascades that
also results in gene activation (4 –7). In contrast, methylation of histone are essential for the execution of adipocyte differentiation (28, 29).
H3 Lys9 generally correlates with gene repression (8, 9). CCAAT/enhancer-binding proteins (C/EBPs), peroxisome prolifera-
Histone deacetylases (HDACs) repress gene expression by deacety- tor-activated receptor ␥ (PPAR␥), and adipocyte determination- and
differentiation-dependent factor 1/sterol response element-binding
protein 1c (ADD1/SREBP1c) have been identified as the key transcrip-
* This work was supported in part by grants from Stem Cell Research Center of the 21st
Century Frontier Research Program (SC13150), Research Center for Functional Cellu- tion factors involved in adipogenesis (30, 31). When preadipocytes are
lomics of Science Research Center Program, and the National Research Laboratory exposed to differentiation conditions, a complex signaling cascade
Program of Korea Science and Engineering Foundation. The costs of publication of
this article were defrayed in part by the payment of page charges. This article must induces the expression of previously silent adipogenic genes. Although
therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section it is not clear how these genes are maintained quiescent in preadipo-
1734 solely to indicate this fact.
□S
The on-line version of this article (available at http://www.jbc.org) contains supple- cytes, it is possible to speculate that HDACs are involved.
mental Figs. S1–S3. Past research on tissue differentiation has been mainly focused on
1
Both authors contributed equally to this work.
2
Supported by BK21 Research Fellowships from the Ministry of Education and Human
understanding the regulatory roles of specific transcription factors. On
Resources Development. the contrary, little is known about the functional roles of coregulators
3
To whom correspondence should be addressed: Dept. of Biological Sciences, Research such as HATs and HDACs during differentiation processes. In this
Center for Functional Cellulomics, Seoul National University, San 56-1, Sillim-Dong,
Gwanak-Gu, Seoul, South Korea 151-742. Tel.: 82-2-880-5852; Fax: 82-2-878-5852; study we have discovered a novel role of HDACs during adipogenesis.
E-mail: jaebkim@snu.ac.kr. Our results suggest that the down-regulation of HDACs at the early
4
The abbreviations used are: HATs, histone acetyltransferases; HDAC, histone deacety-
lase; C/EBP, CCAAT/enhancer-binding protein; PPAR␥, peroxisome proliferator-acti- stages adipogenesis is a rate-limiting step during adipogenesis. These
vated receptor ␥; ADD1/SREBP1c, adipocyte determination- and differentiation-de- findings add to the list of the functional roles of coregulators in regulat-
pendent factor 1/sterol response element-binding protein 1c; NaB, sodium butyrate;
siRNA, small interference RNA; ChIP, chromatin immunoprecipitation, Pref-1, preadi-
ing cell differentiation and may provide a new target for discovering
pocyte factor 1; GST, glutathione S-transferase; HEK, human embryonic kidney cells. novel therapies for the treatment of fat-related disorders.

6608 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 • NUMBER 10 • MARCH 10, 2006
HDAC and Adipogenesis

FIGURE 1. Histone modifications at the pro-


moter regions of adipogenic genes during adi-
pocyte differentiation. ChIP assays were per-
formed using 3T3-L1 cells at different stages of
adipogenesis. Isolated DNA was immunoprecipi-
tated with anti-acetylated histone H3 (K9, A), anti-
acetylated H4 (K8, B), or anti-dimethylated histone
H3 (K9, C) antibodies, and amplified using primers
specific for the promoter regions of the indicated
genes (see “Materials and Methods” for details).

MATERIALS AND METHODS extracts from 3T3-L1 and 3T3-F442A preadipocytes and adipocytes
Cell Culture—3T3-L1 and 3T3-F442A mouse fibroblasts were main- were incubated with a fluorometric substrate in HDAC assay buffer for
tained in Dulbecco’s modified Eagle’s medium supplemented with 10% 45 min at 30 °C. An activator solution was then added to release the
fluorophore from the deacetylated substrates, and the fluorescence was

Downloaded from http://www.jbc.org/ by guest on May 7, 2017


bovine calf serum (BCS, Invitrogen), with a change of medium every 2–3
days. 3T3-L1 cells were differentiated into adipocytes as previously measured in a fluorescence plate reader.
described (32). 3T3-F442A cells were differentiated into adipocytes by mRNA Analysis—Total RNA was isolated with TRIzol reagent
replacing the medium with Dulbecco’s modified Eagle’s medium con- (Invitrogen) according to the manufacturer’s protocol. For Northern
taining 10% fetal bovine serum (FBS, Invitrogen) and 5 ␮g/ml insulin blotting, 20 ␮g of RNA was denatured in formamide and formaldehyde,
upon reaching confluence. BOSC and H29D retrovirus packaging cells and separated by electrophoresis on formaldehyde-containing agarose
were maintained in Dulbecco’s modified Eagle’s medium supplemented gels. RNA was transferred to Nytran membranes (Schleicher and
with 10% FBS. Schuell), and the membranes were cross-linked, hybridized, and washed
Chromatin Immunoprecipitation (ChIP) Assays—Chromatin was as described by the manufacturers. Probes were labeled by the random
immunoprecipitated as described previously (33). Conditions for PCR priming method using the Klenow fragment of DNA polymerase I (Pro-
reactions were as follows: 0.25 ␮M of each primer, 0.1 mM of each dNTP, mega) and [␣-32P]dCTP (Amersham Biosciences). PPAR␥, aP2,
1⫻ PCR buffer, 1 unit Nova Taq polymerase (Genenmed), and 0.06 C/EBP␣, and FAS cDNAs were used as probes. For real-time PCR anal-
mCi/ml [␣-32P]dCTP in 20 ␮l of reaction volume. PCR primers used ysis, cDNA was synthesized by reverse transcription using oligodT and
were as follows: ADD1-P5, 5⬘-GGT TGG TAC CAC AGT GAC CG-3⬘; subjected to PCR amplification with gene-specific primers in the pres-
ADD1-P3, 5⬘-AAT GTG CAA TCC ATG GCT CCG TGG T-3⬘; Adi- ence of Cybergreen (Bio-Rad). Relative abundance of mRNA was calcu-
ponectin-P5, 5⬘-GGT GCT GGG AAT TGA ACT CA-3⬘; Adiponectin- lated after normalization to 18 S ribosomal RNA. Primer sequences for
P3, 5⬘-CCT GTT TCC AGG CTT TGG CC-3⬘; aP2-P5, 5⬘-TTC CAG real-time PCR analyses are available upon request.
GTG AGA AAG GCT GC-3⬘; aP2-P3, 5⬘-GTC CAG TGA TCA TTG Retroviral Infection—BOSC or H29D cells were transfected with
CCA GG-3⬘; C/EBP␣-P5, 5⬘-CCC GGG CTC CCT AGT GTT-3⬘; either pBABE, pBABE-HDAC1, pSUPERretro (Oligoengine), or pSUPER-
C/EBP␣-P3, 5⬘-CTG GCT CGC GCC CGC GCA-3⬘; PPAR␥2-P5, retro-HDAC1 RNAi (5⬘-GCA GCG TCT CTT TGA GAA C-3⬘) using
5⬘-CTG TAC AGT TCA CGC CCC TC-3⬘; PPAR␥2-P3, 5⬘-TCA CAC Lipofectamine (Invitrogen). 48 h after transfection, the medium con-
TGG TGT TTT GTC TAT G-3⬘; Pref-1-P5, 5⬘-CTT TTC GTG GTG taining retroviruses was harvested, filtered, and transferred to 3T3-L1
GTT TTC GT-3⬘; Pref-1-P3, 5⬘-GCA AGT CTC AGG AAC CAA target cells with polybrene (1 ␮g/ml). Infected cells were selected with 5
GC-3⬘; GAPDH-f, 5⬘-GTG TTC CTA CCC CCA ATG TG-3⬘; ␮g/ml puromycin (Sigma) for 7 days.
GAPDH-r, 5⬘-CTT GCT CAG TGT CCT TGC TG-3⬘. Transient Transfections and Luciferase Assays—HEK293 cells were
Acid Extraction of Histones and Western Blotting—Histone proteins grown in Dulbecco’s modified Eagle’s medium containing 10% bovine
were isolated from 3T3-L1 preadipocytes and adipocytes by acid extrac- calf serum. A luciferase reporter driven by multiple PPREs (DR1-luc)
tion according to the protocol provided by Upstate Biotechnology. The was cotransfected with PPAR␥ and HDAC1 expression vectors using
proteins were separated by electrophoresis on SDS-polyacrylamide gels the calcium phosphate method (32). The cells were lysed and assayed for
and transferred to polyvinylidene difluoride membranes (Millipore). luciferase activity.
Following transfer, the membranes were blocked with milk and probed Protein Interaction Analyses—GST-pulldown assays were performed
with antibodies against HDAC1, -2, -3, -4, -5, -6, -7, -8, PPAR␥, C/EBP␣, as previously described (34). Briefly, GST and GST-PPAR␥ fusion pro-
CBP, p300 (Santa Cruz Biotechnology), acetylated histone H3 (Lys9), teins were bound to glutathione beads and incubated with 35S- labeled
dimethylated histone H3 (Lys9), histone H3, acetylated histone H4 (Lys8 HDAC1 protein. The beads were washed with TBS and analyzed by
and Lys12), and histone H4 (Upstate Biotechnology). Results were visu- autoradiography after SDS-PAGE. Protein-protein interaction in cells
alized with horseradish peroxidase-conjugated secondary antibodies was also analyzed by co-immunoprecipitation experiments. HDAC1
(Sigma Aldrich) and enhanced chemiluminescence. and PPAR␥ expression vectors were transfected in HEK293 cells using
HDAC Enzyme Activity Assays—Total cellular HDAC enzymatic the calcium phosphate method. After transfection, whole cell extracts
activity was measured using an HDAC assay kit (Upstate Biotechnol- were prepared and incubated with HDAC1 antibody overnight at 4 °C.
ogy) according to the manufacturer’s protocol. Briefly, 40 ␮g of total cell The immunocomplexes were isolated with protein A-Sepharose beads

MARCH 10, 2006 • VOLUME 281 • NUMBER 10 JOURNAL OF BIOLOGICAL CHEMISTRY 6609
HDAC and Adipogenesis

FIGURE 2. Western blot analysis of histone-modifying enzymes in 3T3-L1 (A) and


3T3-F442A (B) cells during adipocyte differentiation. PPAR␥ and C/EBP␣ are markers

Downloaded from http://www.jbc.org/ by guest on May 7, 2017


of differentiated adipocytes.

(Amersham Biosciences) and washed three times with TBS. The immu-
noprecipitates were separated by SDS-PAGE and immunoblotted using
antibodies against PPAR␥.

RESULTS
Histone Modification at the Promoters of Adipogenic Genes during FIGURE 3. Changes in overall histone acetylation and HDAC activity during adipo-
cyte differentiation. A, changes of histone modification during adipocyte differentia-
Adipocyte Differentiation—The dramatic changes in gene expression tion. B, total cellular HDAC enzyme activity of 3T3-L1 and 3T3-F442A preadipocytes/
profiles during adipocyte differentiation have been previously described adipocytes. Representative results from three independent experiments carried out in
duplicate are shown.
(35, 36). Notably, the expression of adipogenic genes such as PPAR␥,
C/EBP␣, and ADD1/SREBP1c is increased (28), whereas that of preadi-
pocyte marker genes including preadipocyte factor-1 (Pref-1) and HATs, such as CBP or p300, were shown to be unchanged or slightly
GATA2 is conversely decreased (37, 38). Although many transcription increased, respectively (Fig. 2A). A similar reduction in HDAC1, -2, -3,
factors that participate in adipocyte differentiation have been identified, -5, -7, and -8 was observed in 3T3-F442A cells, another adipogenic cell
the roles of histone-modifying enzymes and chromatin-remodeling fac- line (Fig. 2B). C/EBP␣ and PPAR␥ were used as control to confirm
tors during this process have not been thoroughly investigated. proper differentiation.
To determine if histone modification is involved in adipogenesis, we The finding that the expression of several HDACs is severely
examined the level of histone acetylation and methylation at the pro- decreased prompted us to examine whether the overall balance would
moter regions of adipocyte marker genes. The degree of histone H3 Lys9 be shifted in favor of histone acetylation. Indeed, the degree of overall
acetylation at the promoters of ADD1/SREBP1c, adiponectin, aP2, histone acetylation was increased in adipocytes compared with undif-
C/EBP␣, and PPAR␥ was increased upon differentiation (Fig. 1A). Sim- ferentiated preadipocytes (Fig. 3A). Methylation of histone H3 lysine 9,
ilarly, histone H4 Lys8 acetylation was increased at the promoters of which usually displays an opposite pattern to acetylation, was decreased
ADD1/SREBP1c and adiponectin (Fig. 1B). In contrast, histone acetyla- in adipocytes. To determine whether histone deacetylase activity is
tion gradually decreased at the promoter of Pref-1, consistent with the actually regulated during differentiation, we directly measured HDAC
fact that it is down-regulated during adipogenesis (37). On the other enzyme activity using the same total cell extracts. As shown in Fig. 3B,
hand, the level of histone H3 Lys9 methylation displayed opposite pat- total cellular HDAC activity was reduced to ⬃50⬃60% in both 3T3-L1
terns, decreasing at the promoters of ADD1/SREBP1c and C/EBP␣ and and 3T3-F442A cells upon differentiation (Fig. 3B). On the contrary,
marginally increasing at the Pref-1 promoter (Fig. 1C). These results total cellular HAT activity was slightly increased in differentiated adi-
indicate that the up-regulation of adipogenic genes during adipogenesis pocytes (data not shown). Thus, it is likely that the increase of histone
is tightly associated with the selective induction of histone hyperacety- acetylation during adipogenesis is the result of a combination of
lation at the promoter regions of these genes. decreased HDAC activity, and to some extent, increased HAT activity.
Changes of Histone Modification and Expression of HDACs during Such modification of histone tails would probably contribute to the
Adipogenesis—Because the steady state of histone acetylation is deter- overall increase of gene expression in adipocytes, which have been
mined by the balance between HATs and HDACs, we next examined reported to express a larger number of genes than preadipocytes (36).
the protein levels of several histone-modifying enzymes before and after Effects of HDAC Inhibition on Adipocyte Differentiation—Next, to
differentiation. In 3T3-L1 cells, the expression levels of several HDACs determine whether the down-regulation of HDAC activity indeed
including HDAC1, -2, -5, and -6 were drastically reduced during differ- affects adipogenesis, we treated 3T3-L1 cells with an inhibitor of
entiation (Fig. 2A) whereas HDAC3, -4, -7, and -8 were either slightly HDACs, sodium butyrate (NaB). Butyrate is a short chain fatty acid that
increased or unchanged (supplemental Fig. S1). The protein levels of noncompetitively inhibits the activity of most HDAC isoforms (39, 40).

6610 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 • NUMBER 10 • MARCH 10, 2006
HDAC and Adipogenesis

Downloaded from http://www.jbc.org/ by guest on May 7, 2017


FIGURE 4. Induction of adipocyte differentiation and adipogenic gene expression by the HDAC inhibitor, NaB. A, Oil Red-O staining of differentiated 3T3-L1 cells with or without
NaB treatment. B, Northern blot analysis. Total RNA from 3T3-L1 cells incubated in the absence (left) or presence (right) of NaB (500 ␮M) was isolated at the indicated time points after
induction of differentiation. C, differentiating 3T3-L1 cells were treated with NaB for various time periods and stained with Oil Red-O. Bars indicate the duration of NaB treatment.
Representative data from three separate experiments are shown.

As shown in Fig. 4A chronic treatment of NaB accelerated differentia- overexpressing cells, suggesting that HDAC1 suppresses adipogenesis
tion and increased lipid accumulation assessed by Oil Red-O staining. by down-regulating the expression of adipogenic genes (Fig. 5D).
Northern blot analysis revealed that the expression of adipogenic genes In a reciprocal experiment, we examined the effect of HDAC1 down-
was induced at an earlier time point (Fig. 4B). A similar effect was regulation via siRNA. To test whether the HDAC1-siRNA effectively
observed in C3H10T1/2 cells, a murine mesenchymal fibroblast cell line reduced HDAC1 expression, HEK293 cells were co-transfected with
that can also be induced to differentiate into adipocytes (supplemental FLAG-tagged HDAC1 and HDAC1-siRNA vectors. As expected, over-
Fig. S2). expression of HDAC1-siRNA efficiently repressed HDAC1 expression
To investigate the time periods at which HDAC inhibition exhibits (Fig. 6A). Next, 3T3-L1 cells overexpressing the retroviral HDAC1-
the most critical effect on adipogenesis, we treated the cells with NaB for siRNA were tested for their ability to differentiate into adipocytes. Con-
different periods during adipocyte differentiation. Cells treated with sistent with the above observations, HDAC1 knock-down 3T3-L1 cells
NaB throughout the entire differentiation period clearly exhibited stim- clearly displayed accelerated differentiation (Fig. 6B) and stimulated
ulated adipocyte differentiation compared with control cells (Fig. 4C, expression of ADD1/SREBP1c, aP2, and PPAR␥ (Fig. 6C). Furthermore,
panels II and VIII). Notably, inhibition of the HDACs at the early stage HDAC1 knockdown led to the increase of histone acetylation at H3
of adipogenesis (the first 1 or 2 days) seemed to be sufficient to stimulate Lys9, H4 Lys8, and H4 Lys12 residues. Conversely, the extent of histone
adipogenesis (Fig. 4C, panels IV and V), while treatment of NaB at later H3 Lys9 methylation decreased (Fig. 6D). These results provide strong
periods had little or no effect (Fig. 4C, panels VI and VII). Taken evidence that HDAC1 controls adipocyte differentiation by regulating
together, these results indicate that suppression of HDAC activity at the the acetylation of histones and implicate that a threshold level of HDAC
early stage of adipogenesis may be crucial for adipocyte differentiation. activity is probably required to maintain preadipocytes by silencing adi-
The Role of HDAC1 during Adipogenesis—Among many HDAC iso- pogenic genes in the undifferentiated state.
forms, HDAC1 was most consistently and dramatically down-regulated Regulation of PPAR␥ Activity by HDAC1—To understand how
during adipogenesis (Fig. 2, A and B). To determine if HDAC1 plays an HDAC1 affects adipocyte differentiation, we examined whether
important role in adipogenic gene expression, we determined the HDAC1 could directly regulate the transcriptional activity of PPAR␥, a
recruitment of HDAC1 to the promoter regions of adipogenic genes master adipogenic transcription factor. In transient transfection assays,
using ChIP assays. As expected, recruitment of HDAC1 decreased upon HDAC1 repressed the transcriptional activity PPAR␥ in a dose-depend-
differentiation in most adipogenic genes including adiponectin, ADD1/ ent manner (Fig. 7A). On the other hand, the activity of ADD1/SREBP1c
SREBP1c, aP2, C/EBP␣, and PPAR␥, indicating that HDAC1 may be was not altered by HDAC1 (data not shown). To see if HDAC1 physi-
involved in regulating the expression of these genes (Fig. 5A). cally associates with PPAR␥, we performed GST-pulldown assays using
To directly determine whether HDAC1 is specifically involved in recombinant GST-PPAR␥ fusion proteins and in vitro translated
adipocyte differentiation, we generated 3T3-L1 preadipocytes retrovi- HDAC1. HDAC1 bound specifically with purified GST-PPAR␥ but not
rally overexpressing HDAC1. These cells expressed higher amounts of GST, suggesting that HDAC1 may directly interact with PPAR␥ (Fig.
HDAC1 (⬃2-fold) compared with mock-infected cells (Fig. 5B). Under 7B). To investigate whether this interaction occurs in cells, we isolated
differentiation conditions, the HDAC1-overexpressing cells exhibited whole cell lysates of HEK293 cells after transfection with plasmids
substantially attenuated adipocyte differentiation (Fig. 5C). In addition, encoding PPAR␥ and HDAC1. Immunoprecipitation with anti-HDAC1
the expression of several adipogenic genes is also decreased in HDAC1- antibody followed by Western blot assays using anti-PPAR␥ antibody

MARCH 10, 2006 • VOLUME 281 • NUMBER 10 JOURNAL OF BIOLOGICAL CHEMISTRY 6611
HDAC and Adipogenesis

FIGURE 5. Recruitment of HDAC1 to the promot-


ers of adipocyte marker genes and the effect of
HDAC1 overexpression on 3T3-L1 cell differen-
tiation. A, ChIP assays were performed with
␣-HDAC1 antibodies. Band intensities were deter-
mined with LabworksTM (Media Cybernetics). B,
Western blot of pBABE-mock and pBABE-HDAC1

Downloaded from http://www.jbc.org/ by guest on May 7, 2017


infected 3T3-L1 cells. C, HDAC1 overexpression
inhibits adipocyte differentiation. Oil Red O stain-
ing of HDAC1 overexpressing 3T3-L1 cells differ-
entiated for 6 days. D, real-time qPCR analysis of
adipocyte marker gene expression. *, p ⬍ 0.05.

demonstrates that HDAC1 indeed interacts with PPAR␥ in vivo (Fig. HDACs significantly decreased upon differentiation (Fig. 2, A and B).
7C). It is therefore possible to speculate that HDAC1 could modulate Indeed, the endogenous HDAC enzyme activity measured in adipocytes
adipocyte differentiation by repressing PPAR␥ activity. was shown to be only half that of preadipocytes (Fig. 3B). Inversely, total
HAT activity of adipocytes was slightly elevated upon differentiation
DISCUSSION (data not shown). Therefore, it seems that histone hyperacetylation in
Recent studies have demonstrated the involvement of chromatin adipocytes, as a result of decreased HDAC activity and increased HAT
remodeling factors in the differentiation of specific cell types including activity, leads to enhanced adipogenic gene expression. This increase of
muscle, neurons, and T cells (41– 43). Although it is well known that histone acetylation is most likely preferentially targeted to adipogenic
certain transcription factors or cofactors interact with HATs or HDACs genes by tissue-specific transcription factors and cofactors. This idea is
to regulate their target gene expression, there are few reports that the strongly supported by the fact that PPAR␥, a master adipogenic tran-
overall level of histone modifications or expression of histone-modify- scription factor, interacts with several coactivators such as p300, CBP,
ing enzymes such as HDACs is changed during bona fide differentiation. TRAP, p160, and PGC1␣ and corepressors like HDAC3 to selectively
Furthermore, little is known about the regulatory role of chromatin regulate the expression of its target genes during adipogenesis (44 – 48).
remodeling such as histone modifications in adipocyte differentiation. Because these cofactors are able to interact with specific transcription
In the present study, we sought to identify the regulatory function of factors in a highly specific manner, decondensation of chromatin is
histone modifications in adipocyte differentiation. ChIP assays revealed tightly controlled so that it only takes place in a subset of genes in the
that histones are dynamically modified at the promoter regions of adi- appropriate tissues and at the appropriate times.
pocyte marker genes indicating its involvement in adipogenesis. More Stimulation of adipogenesis and adipogenic gene expression by NaB
importantly, we discovered that the protein level of class I and class II suggests that the down-regulation of histone deacetylases might be one

6612 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 • NUMBER 10 • MARCH 10, 2006
HDAC and Adipogenesis

FIGURE 6. Effect of HDAC1 knock-down by


siRNA on 3T3-L1 cell differentiation. A, HDAC1-
specific siRNA inhibits HDAC1 expression. Western
blot analysis of cells cotransfected with
pcDNA-HDAC1-FLAG and pSuper-retro-HDAC1-
siRNA was performed with FLAG antibody. B,
HDAC1 siRNA stimulates adipocyte differentia-
tion. 3T3-L1 cells were infected with retroviruses
containing either pSuper-retro-mock or pSuper-
retro-HDAC1-siRNA. After selection with puromy-
cin, the stably infected cells were differentiated
into adipocytes. C, real-time qPCR analysis of adi-
pocyte marker genes in mock and pSuper-retro-
HDAC1-siRNA infected 3T3-L1 cells. *, p ⬍ 0.05. D,

Downloaded from http://www.jbc.org/ by guest on May 7, 2017


acid extracts from mock or pSuper-retro-HDAC1-
siRNA-infected cells were subjected to Western
blot analysis with the indicated antibodies.

of the rate-limiting steps during adipocyte differentiation (Fig. 4, A and is also known that HD3 is a component of a corepressor complex with
B). Interestingly, cells treated with NaB for the first 2 or 3 days, but not NCoR/SMRT that represses PPAR␥. Interestingly, HDAC1 interacted
at later stages of differentiation, showed a remarkable stimulation of with PPAR␥ and repressed its transcriptional activity (Fig. 7). Even
adipogenesis (Fig. 3C). These results implicate that a critical determina- though many HDACs are known to bind to transcription factors as part
tion step for adipogenesis occurs through histone modification at an of a corepressor complex, results from GST-pulldown and co-immuno-
early phase of the differentiation process. This crucial event is probably precipitation assays suggest that HDAC1 may bind directly to PPAR␥.
associated with tissue- or developmentally specific transcription factors In addition, it should be noted that PPAR␥ and HDAC1 display recip-
and/or cofactors responsible for triggering tissue-specific gene expres- rocal expression patterns during adipogenesis (Fig. 2A). Thus, it appears
sion with down-regulation of HDAC activity. Similar results have been that activation of PPAR␥ might be regulated by two steps during adipo-
obtained with Swiss 3T3 cells (49) and was also observed in C3H 10T1/2 cyte differentiation. First, PPAR␥ activity is stimulated by increases in
cells (supplemental Fig. S2). These cell lines are less determined to the protein levels and its endogenous ligands. Second, the down-regulation
adipogenic lineage, suggesting that this phenomenon is not restricted to of HDAC1 would further promote PPAR␥ activity by relieving it from
a specific cell line. repression. Therefore, in addition to the previously described role as a
A remarkable drop in the expression of HDAC1 suggested that this modulator of C/EBP␤, HDAC1 would also regulate adipocyte differen-
specific isoform may play a major role in adipogenesis. This idea was tiation by controlling PPAR␥.
supported by the observation that the amounts of HDAC1 localized to In addition to these findings, which suggest that HDACs interact with
the promoters of adipogenic genes were clearly decreased after differ- several adipogenic transcription factors to modulate their activities, our
entiation (Fig. 5A). Gain-of-function and loss-of function studies in results implicate the importance of the regulation of HDAC expression
3T3-L1 cells also confirmed the critical role of HDAC1 in adipogenesis. during adipocyte differentiation. Although the exact mechanism by
While we were preparing this article, Wiper-Bergeron et al. (50) dem- which adipogenic stimuli decrease the level of HDACs is yet to be deter-
onstrated that HDAC1, which forms a complex with mSin3A and mined, it appears that the lowering of HDAC expression results in
C/EBP␤, is able to attenuate differentiation by repressing C/EBP␣ decondensation of the chromatin structure around the promoter
expression. In another recent report, Fajas et al. (48) demonstrated that regions of adipogenic genes, thereby facilitating the binding of tran-
HDAC3 can suppress PPAR␥ by forming a PPAR␥䡠Rb䡠HD3 complex. It scription factors to their binding sites (supplemental Fig. S3).

MARCH 10, 2006 • VOLUME 281 • NUMBER 10 JOURNAL OF BIOLOGICAL CHEMISTRY 6613
HDAC and Adipogenesis
of colon cancer by altering gene expression and arresting cell division or
to induce apoptosis in human leukemic lymphoblasts (53). Further-
more, HDAC inhibitors induce the expression of cell cycle kinase inhib-
itor p21, causing growth arrest in transformed cells (54, 55). It is inter-
esting to note that expression of p21 is induced by HDAC inhibitors
since p21 is induced at a very early stage of adipocyte differentiation to
modulate clonal expansion (56). Although the precise mechanisms by
which HDAC inhibitors induce cell cycle arrest via p21 expression dur-
ing adipogenesis remain to be elucidated, it might shed light in under-
standing how histone acetylation and deacetylation are possibly linked
to adipocyte differentiation.
In summary, our results show that the down-regulation of HDAC
expression and the resulting increase of histone acetylation are impor-
tant for adipogenesis. The substantial decrease in HDAC activity results
in preferential histone hyperacetylation at the promoter regions of adi-
pocyte marker genes. It is therefore likely that regulation of HDAC
expression would be an essential process in adipocyte differentiation
and that preadipocytes could initiate the differentiation process only
when cellular level of HDACs falls below a certain threshold level and
also that HDACs are involved in maintaining the preadipocyte pheno-
type. This is a novel viewpoint concerning the regulation of adipogen-

Downloaded from http://www.jbc.org/ by guest on May 7, 2017


esis since previous studies have mainly focused on adipogenic transcrip-
tion factors, such as PPAR␥ and C/EBP␣ and their ability to recruit
coregulators to adipogenic gene promoters. Our findings could also
provide a novel means of treating obesity-related diseases. Further stud-
ies will reveal whether this is a general mechanism controlling the dif-
ferentiation of diverse cell types.

Acknowledgments—We thank Dr. W-M. Yang for providing the


pcDNA-HDAC1-FLAG vector. We are grateful to Dr. K. U. Lee and Y. S. Lee for
critically reading the manuscript.
FIGURE 7. Repression of PPAR␥ activity by HDAC1. A, HEK293 cells were transfected
with DR1-luciferase reporter construct and expression vectors for PPAR␥ and HDAC1.
The cells were lysed and assayed for luciferase activity after transfection. B, in vitro trans- REFERENCES
lated 35S-labeled HDAC1 was incubated with GST or GST-PPAR␥ fusion proteins and
analyzed by autoradiography. Gels were stained with Coomassie Brilliant Blue to confirm 1. Jenuwein, T., and Allis, C. D. (2001) Science 293, 1074 –1080
equal loading. C, whole cell extracts of HEK293 cells cotransfected with HDAC1 and 2. Ogryzko, V. V., Schiltz, R. L., Russanova, V., Howard, B. H., and Nakatani, Y. (1996)
PPAR␥ were immunoprecipitated with HDAC1 antibody. The immunocomplexes were Cell 87, 953–959
subjected to SDS-PAGE and immunoblotted with PPAR␥ and HDAC1 antibodies. 3. Schiltz, R. L., Mizzen, C. A., Vassilev, A., Cook, R. G., Allis, C. D., and Nakatani, Y.
(1999) J. Biol. Chem. 274, 1189 –1192
Our group has also discovered that a very similar phenomenon 4. Spencer, V. A., and Davie, J. R. (1999) Gene (Amst.) 240, 1–12
occurs during osteoblast differentiation, suggesting that the modulation 5. Chen, H., Lin, R. J., Xie, W., Wilpitz, D., and Evans, R. M. (1999) Cell 98, 675– 686
6. Wang, H., Huang, Z. Q., Xia, L., Feng, Q., Erdjument-Bromage, H., Strahl, B. D.,
of HDAC expression and activity may be a general means of regulating Briggs, S. D., Allis, C. D., Wong, J., Tempst, P., and Zhang, Y. (2001) Science 293,
cell differentiation.5 The fact that NaB accelerated the differentiation of 853– 857
C3H10T1/2 cells into both adipocytes (supplemental Fig. S1) and osteo- 7. Cheung, P., Tanner, K. G., Cheung, W. L., Sassone-Corsi, P., Denu, J. M., and Allis,
blasts5 depending on the differentiation condition implies that the C. D. (2000) Mol. Cell 5, 905–915
8. Rea, S., Eisenhaber, F., O’Carroll, D., Strahl, B. D., Sun, Z. W., Schmid, M., Opravil, S.,
down-regulation of HDACs is a prerequisite process to convert the cells Mechtler, K., Ponting, C. P., Allis, C. D., and Jenuwein, T. (2000) Nature 406, 593–599
into a state susceptible to induction of differentiation. However, it does 9. Nakayama, J., Rice, J. C., Strahl, B. D., Allis, C. D., and Grewal, S. I. (2001) Science 292,
not seem that the down-regulation of HDAC1 specifies the cell-type 110 –113
into which the precursor cells will differentiate. Instead, this event 10. Ayer, D. E. (1999) Trends Cell Biol. 9, 193–198
11. Johnstone, R. W. (2002) Nat. Rev. Drug Discov. 1, 287–299
seems be a part of a transition of the undifferentiated cells into a state in 12. de Ruijter, A. J., van Gennip, A. H., Caron, H. N., Kemp, S., and van Kuilenburg, A. B.
which cell type-specific transcription factors can bind to the chromatin (2003) Biochem. J. 370, 737–749
and activate the expression of cell type-specific marker genes. Further 13. Fischle, W., Emiliani, S., Hendzel, M. J., Nagase, T., Nomura, N., Voelter, W., and
studies will reveal whether this event is common during the differenti- Verdin, E. (1999) J. Biol. Chem. 274, 11713–11720
14. Grozinger, C. M., Hassig, C. A., and Schreiber, S. L. (1999) Proc. Natl. Acad. Sci.
ation from mesenchymal stem cells. U. S. A. 96, 4868 – 4873
The effects of HDAC inhibitors appear to be selective since the 15. Fischle, W., Kiermer, V., Dequiedt, F., and Verdin, E. (2001) Biochem. Cell Biol. 79,
expression of only 2⬃5% of mammalian genes is affected when HDAC 337–348
activity is inhibited (51, 52). As an HDAC inhibitor, NaB has many 16. Guardiola, A. R., and Yao, T. P. (2002) J. Biol. Chem. 277, 3350 –3356
17. Kao, H. Y., Lee, C. H., Komarov, A., Han, C. C., and Evans, R. M. (2002) J. Biol. Chem.
effects on cultured mammalian cells including inhibition of prolifera- 277, 187–193
tion and induction of differentiation and gene expression. For example, 18. Gao, L., Cueto, M. A., Asselbergs, F., and Atadja, P. (2002) J. Biol. Chem. 277,
HDAC inhibition with NaB has been shown to inhibit the development 25748 –25755
19. Berger, S. L. (2002) Curr. Opin. Genet. Dev. 12, 142–148
20. Kadam, S., and Emerson, B. M. (2002) Curr. Opin. Cell Biol. 14, 262–268
5
H. W. Lee, J. H. Suh, Y. S. Lee, S. Y. Park, and J. B. Kim, manuscript in preparation. 21. Puri, P. L., Sartorelli, V., Yang, X. J., Hamamori, Y., Ogryzko, V. V., Howard, B. H.,

6614 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 • NUMBER 10 • MARCH 10, 2006
HDAC and Adipogenesis
Kedes, L., Wang, J. Y., Graessmann, A., Nakatani, Y., and Levrero, M. (1997) Mol. Cell 40. Sealy, L., and Chalkley, R. (1978) Cell 14, 115–121
1, 35– 45 41. de la Serna, I. L., Carlson, K. A., and Imbalzano, A. N. (2001) Nat. Genet. 27, 187–190
22. McKinsey, T. A., Zhang, C. L., and Olson, E. N. (2001) Curr. Opin. Genet. Dev. 11, 42. Machida, Y., Murai, K., Miyake, K., and Iijima, S. (2001) J. Biochem. (Tokyo) 129,
497–504 43– 49
23. Mal, A., and Harter, M. L. (2003) Proc. Natl. Acad. Sci. U. S. A. 100, 1735–1739 43. Avni, O., Lee, D., Macian, F., Szabo, S. J., Glimcher, L. H., and Rao, A. (2002) Nat.
24. Ng, H. H., and Bird, A. (2000) Trends Biochem. Sci 25, 121–126 Immunol. 3, 643– 651
25. Mal, A., Sturniolo, M., Schiltz, R. L., Ghosh, M. K., and Harter, M. L. (2001) EMBO J. 44. Puigserver, P., Wu, Z., Park, C. W., Graves, R., Wright, M., and Spiegelman, B. M.
20, 1739 –1753 (1998) Cell 92, 829 – 839
26. Lemercier, C., Verdel, A., Galloo, B., Curtet, S., Brocard, M. P., and Khochbin, S. 45. Yang, W., Rachez, C., and Freedman, L. P. (2000) Mol. Cell Biol. 20, 8008 – 8017
(2000) J. Biol. Chem. 275, 15594 –15599 46. Takahashi, N., Kawada, T., Yamamoto, T., Goto, T., Taimatsu, A., Aoki, N., Kawasaki,
27. McKinsey, T. A., Zhang, C. L., Lu, J., and Olson, E. N. (2000) Nature 408, 106 –111 H., Taira, K., Yokoyama, K. K., Kamei, Y., and Fushiki, T. (2002) J. Biol. Chem. 277,
28. Rosen, E. D., Walkey, C. J., Puigserver, P., and Spiegelman, B. M. (2000) Genes Dev. 14, 16906 –16912
1293–1307
47. Wallberg, A. E., Yamamura, S., Malik, S., Spiegelman, B. M., and Roeder, R. G. (2003)
29. Morrison, R. F., and Farmer, S. R. (2000) J. Nutr. 130, 3116S-3121S
Mol. Cell 12, 1137–1149
30. Wu, Z., Bucher, N. L., and Farmer, S. R. (1996) Mol. Cell Biol. 16, 4128 – 4136
48. Fajas, L., Egler, V., Reiter, R., Hansen, J., Kristiansen, K., Debril, M. B., Miard, S., and
31. Hamm, J. K., Park, B. H., and Farmer, S. R. (2001) J. Biol. Chem. 276, 18464 –18471
Auwerx, J. (2002) Dev. Cell 3, 903–910
32. Seo, J. B., Moon, H. M., Kim, W. S., Lee, Y. S., Jeong, H. W., Yoo, E. J., Ham, J., Kang,
49. Toscani, A., Soprano, D. R., and Soprano, K. J. (1990) J. Biol. Chem. 265, 5722–5730
H., Park, M. G., Steffensen, K. R., Stulnig, T. M., Gustafsson, J. A., Park, S. D., and Kim,
50. Wiper-Bergeron, N., Wu, D., Pope, L., Schild-Poulter, C., and Hache, R. J. (2003)
J. B. (2004) Mol. Cell Biol. 24, 3430 –3444
33. Seo, J. B., Noh, M. J., Yoo, E. J., Park, S. Y., Park, J., Lee, I. K., Park, S. D., and Kim, J. B. EMBO J. 22, 2135–2145
(2003) Mol. Endocrinol. 17, 1522–1533 51. Van Lint, C., Emiliani, S., and Verdin, E. (1996) Gene Expr. 5, 245–253
34. Lee, Y. S., Lee, H. H., Park, J., Yoo, E. J., Glackin, C. A., Choi, Y. I., Jeon, S. H., Seong, 52. Mariadason, J. M., Corner, G. A., and Augenlicht, L. H. (2000) Cancer Res. 60,
R. H., Park, S. D., and Kim, J. B. (2003) Nucleic Acids Res. 31, 7165–7174 4561– 4572
35. Soukas, A., Socci, N. D., Saatkamp, B. D., Novelli, S., and Friedman, J. M. (2001) J. Biol. 53. Rahmani, M., Yu, C., Dai, Y., Reese, E., Ahmed, W., Dent, P., and Grant, S. (2003)
Chem. 276, 34167–34174 Cancer Res. 63, 8420 – 8427
36. Sadowski, H. B., Wheeler, T. T., and Young, D. A. (1992) J. Biol. Chem. 267, 54. Richon, V. M., Webb, Y., Merger, R., Sheppard, T., Jursic, B., Ngo, L., Civoli, F.,

Downloaded from http://www.jbc.org/ by guest on May 7, 2017


4722– 4731 Breslow, R., Rifkind, R. A., and Marks, P. A. (1996) Proc. Natl. Acad. Sci. U. S. A. 93,
37. Smas, C. M., and Sul, H. S. (1993) Cell 73, 725–734 5705–5708
38. Tong, Q., Dalgin, G., Xu, H., Ting, C. N., Leiden, J. M., and Hotamisligil, G. S. (2000) 55. Vrana, J. A., Decker, R. H., Johnson, C. R., Wang, Z., Jarvis, W. D., Richon, V. M.,
Science 290, 134 –138 Ehinger, M., Fisher, P. B., and Grant, S. (1999) Oncogene 18, 7016 –7025
39. Candido, E. P., Reeves, R., and Davie, J. R. (1978) Cell 14, 105–113 56. Morrison, R. F., and Farmer, S. R. (1999) J. Biol. Chem. 274, 17088 –17097

MARCH 10, 2006 • VOLUME 281 • NUMBER 10 JOURNAL OF BIOLOGICAL CHEMISTRY 6615
` Yoo et al.

Sup Fig. 1. Western blot analysis of histone modifying enzymes in 3T3-L1 cells during adipocyte
differentiation.

Sup Fig. 2. Stimulation of C3H10T1/2 cell differentiation by NaB. C3H10T1/2 cells were
differentiated into adipocytes in the absence or presence of NaB (500 µM) and stained with Oil Red O.

Sup Fig. 3. Schematic model. The expression of adipocyte marker genes is kept silent by histone
deacetylases. Induction of adipocyte differentiation by hormonal signaling reduces the expression and
activity of histone deacetylases, leading to loosening of the chromatin. This facilitates the binding of
adipogenic transcription factors to their binding sites.

16
Supplementary Figure 1

e
ytc

e
po

yt
c
di

po
ea

i
Ad
Pr
α-HDAC3

α-HDAC4

α-HDAC7

α-HDAC8

3T3-L1
Supplementary Figure 2

-NaB +NaB
Supplementary Figure 3

Preadipocytes

Ac Ac Ac

HDAC Me HDAC Me HDAC

Adipogenic
Me HAT genes
HDAC HDAC

Ac Ac

Adipogenesis
Adipocytes
HAT HAT
Ac Ac Ac Ac Ac Ac

TF TF TF Adipogenic
Me
Ac Ac Ac Ac Ac
genes
HDAC
HAT
Down-regulation of Histone Deacetylases Stimulates Adipocyte Differentiation
Eung Jae Yoo, Jun-Jae Chung, Sung Sik Choe, Kang Ho Kim and Jae Bum Kim
J. Biol. Chem. 2006, 281:6608-6615.
doi: 10.1074/jbc.M508982200 originally published online January 5, 2006

Access the most updated version of this article at doi: 10.1074/jbc.M508982200

Alerts:
• When this article is cited
• When a correction for this article is posted

Click here to choose from all of JBC's e-mail alerts

Supplemental material:

Downloaded from http://www.jbc.org/ by guest on May 7, 2017


http://www.jbc.org/content/suppl/2006/01/06/M508982200.DC1

This article cites 56 references, 29 of which can be accessed free at


http://www.jbc.org/content/281/10/6608.full.html#ref-list-1

Das könnte Ihnen auch gefallen