Skip to main content
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems
  • Log in
  • My alerts
  • My Cart

Main menu

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • Archive
    • Minireviews
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About MCB
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems

User menu

  • Log in
  • My alerts
  • My Cart

Search

  • Advanced search
Molecular and Cellular Biology
publisher-logosite-logo

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • Archive
    • Minireviews
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About MCB
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
GENE EXPRESSION

RIP140-Targeted Repression of Gene Expression in Adipocytes

Mark Christian, Evangelos Kiskinis, Darja Debevec, Göran Leonardsson, Roger White, Malcolm G. Parker
Mark Christian
1Institute of Reproductive and Developmental Biology, Imperial College London, Faculty of Medicine, Du Cane Road, London W12 0NN, United Kingdom
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Evangelos Kiskinis
1Institute of Reproductive and Developmental Biology, Imperial College London, Faculty of Medicine, Du Cane Road, London W12 0NN, United Kingdom
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Darja Debevec
1Institute of Reproductive and Developmental Biology, Imperial College London, Faculty of Medicine, Du Cane Road, London W12 0NN, United Kingdom
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Göran Leonardsson
2Arexis AB, Arvid Wallgrens Backe, 20 SE-413 46 Göteborg, Sweden
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Roger White
1Institute of Reproductive and Developmental Biology, Imperial College London, Faculty of Medicine, Du Cane Road, London W12 0NN, United Kingdom
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Malcolm G. Parker
1Institute of Reproductive and Developmental Biology, Imperial College London, Faculty of Medicine, Du Cane Road, London W12 0NN, United Kingdom
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: m.parker@imperial.ac.uk
DOI: 10.1128/MCB.25.21.9383-9391.2005
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

ABSTRACT

Ligand-dependent repression of nuclear receptor activity forms a novel mechanism for regulating gene expression. To investigate the intrinsic role of the corepressor RIP140, we have monitored gene expression profiles in cells that express or lack the RIP140 gene and that can be induced to undergo adipogenesis in vitro. In contrast to normal white adipose tissue and in vitro-differentiated wild-type adipocytes, RIP140-null cells show elevated energy expenditure and express high levels of the uncoupling protein 1 gene (Ucp1), carnitine palmitoyltransferase 1b, and the cell-death-inducing DFF45-like effector A. Conversely, all these changes are abrogated by the reexpression of RIP140. Analysis of the Ucp1 promoter showed RIP140 recruitment to a key enhancer element, demonstrating a direct role in repressing gene expression. Therefore, reduction in the levels of RIP140 or prevention of its recruitment to nuclear receptors may provide novel mechanisms for the control of energy expenditure in adipose cells.

The adipocyte plays an active and fundamental role in energy homeostasis, and adipose tissue is central in many of the pathologies associated with obesity (10, 22, 36). Adipocyte functions include the processes of lipid metabolism, glucose metabolism, and endocrine secretion of hormones and cytokines. Two types of adipose tissue, white adipose tissue (WAT) and brown adipose tissue (BAT), have been described. This classification is based on morphological and histological factors as well as a number of characteristic biochemical processes that are controlled in part by the activation of the sympathetic nervous system (2, 7, 25). In addition, genetic factors play a major role in the control of energy balance, and genes that regulate adipose tissue mass have been identified and may be implicated in the differentiation of white and brown adipose cells (5, 11, 24, 26, 38).

One of the key genes in BAT (Ucp1) encodes uncoupling protein 1, a member of the mitochondrial carrier family of proteins. Upon Ucp1 activation, respiration is uncoupled from ATP synthesis, resulting in an increased metabolic rate and the release of chemical energy from the brown fat in the form of heat (8). Functionally defective BAT has been associated with obesity (9, 12), and transgenic mice with reduced BAT are obese, with symptoms of hyperglycemia and insulin resistance (21). In addition, ectopic expression of Ucp1 in WAT in transgenic mice results in resistance to diet-induced obesity and diabetes (15). Thus, the amount of BAT and the expression of Ucp1 correlate with protection from obesity.

Adipose tissue is controlled by humoral factors, by para- and intracrine factors, and by neural regulation. The differentiation of direct effects from systemic effects following genetic manipulation requires the analysis of in vitro and in vivo systems. Cell culture systems provide the opportunity to study the cell-autonomous action of genes. Many transcriptional events that mediate adipogenesis have been elucidated in cell lines, particularly in 3T3-L1 cells (22, 24). The primary regulators include the CCAAT/enhancer binding proteins (C/EBPβ, C/EBPδ, and C/EBPα) and peroxisome proliferator-activated receptor γ2 (PPARγ2), a member of the nuclear receptor (NR) family of transcription factors (22). These factors seem to have similar roles in both white and brown adipocyte differentiation. Little is known about the potential interconversion of white and brown adipose tissue in terms of whether specific white or brown preadipocytes give rise to each type of adipose tissue or whether a single preadipocyte may be able to differentiate into either form of adipose cell. However, a number of transcription factors, coactivators, and cell cycle regulators, including NRs and NR cofactors, have been implicated in the control of white and brown adipocyte differentiation (30). One key regulator of many metabolic processes is the PPARγ coactivator 1α (PGC1α), which is required for adaptation to metabolic and physiologic stimuli (18, 20, 27). PGC1α coordinates a number of signaling pathways in adipocytes, including those involving additional coactivators for NRs, such as the p160 family members SRC-1 and TIF2. The relative expression levels of these coactivators regulate the development and functions of WAT and BAT (26).

The role of NR corepressors in adipogenesis and adipocyte function is less well understood. The corepressor RIP140 binds to the ligand binding domain of NRs in the presence of agonists, and lack of RIP140 results in reduced fat accumulation in vivo (17). The aim of this study was to determine the intrinsic role of RIP140 in adipose biology by using differentiated wild-type and RIP140-null primary adipogenic cultures, a RIP140-null cell line that retains its ability to undergo adipogenesis, and cells into which RIP140 has been reintroduced. Comparison of gene expression profiles in undifferentiated cells and adipocytes lacking and expressing RIP140 identifies derepressed and repressed genes that contribute to adipocyte function.

MATERIALS AND METHODS

Animals.The generation of RIP140-null mice has previously been described (39). Mice used in this study were backcrossed six generations to a C57BL/6J background, maintained under standard conditions with controlled light and temperature, and fed a chow diet ad libitum. All experiments were performed according to United Kingdom Home Office guidelines.

Morphological and immunohistochemical analysis.Tissue sections prepared as described previously (17) were incubated with polyclonal goat anti-mouse primary antibody to Cidea (sc-8732; Santa Cruz Biotechnology), diluted to a ratio of 1:200 in phosphate-buffered saline (PBS), and detected using a Vectastain Elite ABC kit (Vector Laboratories). Sections were counterstained with hematoxylin.

Cell culturing.Primary WAT cultures were prepared from inguinal fat depots. The tissue was finely minced and digested with collagenase (500 μg/ml) and DNase (100 μg/ml), followed by centrifugation at 1,000 rpm for 5 min. The cells were passed through a 70-μm filter and cultured in Dulbecco's modified Eagle's medium (DMEM)-F12 medium supplemented with 10% fetal bovine serum (Invitrogen). Mouse embryonic fibroblasts (MEFs) from embryos at embryonic day 11.5 were isolated and cultured in DMEM-F12 medium supplemented with 10% fetal bovine serum (Invitrogen). The RIPKO-1 cell line was generated by continuous culturing of MEFs. Differentiation of the cells was performed as previously described (35) in the absence or presence of 2.5 μM rosiglitazone (Ro) as indicated below. Differentiated cells were visualized with oil red O staining.

Lentiviral transduction of RIPKO-1 cells.The PCR-amplified coding sequence for human RIP140 was cloned into pLenti6/V5-D-TOPO (Invitrogen). This vector was cotransfected into 293FT cells with ViraPower packaging mix to generate the lentivirus. RIPKO-1 cells were transduced with the lentivirus and stable cell lines (RIPKO-L) generated by selecting with blasticidin.

Transient transfection. Ucp1 promoter reporter constructs were generated by cloning a 4-kb fragment or 220-bp enhancer element (bp −2530 to −2310 relative to the transcriptional start site) of the 5′-flanking region of the murine Ucp1 gene into pGL3/basic vector (Promega). A 4-kb promoter construct lacking the enhancer region (ΔEnh; deletion of bp −2615 to −2228) was generated by PCR. RIPKO-1 cells were transfected in 24-well plates by using Fugene6 (Roche) with 1 μg of reporter gene, 250 ng of pRL-CMV, and/or 500 ng of pCI-RIP. Cells were harvested for luciferase assay 1, 3, and 5 days following the addition of the differentiation cocktail. Renilla luciferase activity was used to correct for differences in transfection efficiencies.

Expression analysis.RNA extraction from cell lines and tissue and cDNA preparation were performed as described previously (17). RIP140, L19, and UCP1 gene expression levels were determined using specific primers and TaqMan probes. Expression levels for all other genes were determined with SYBR green reagent by using specific primers. Expression levels for all genes were correlated to that for the L19 ribosomal coding gene. Primer sequences may be obtained on request.

Quantitation of mitochondrial DNA.DNA was extracted from untreated and differentiated primary MEFs using a DNeasy kit (QIAGEN). Real-time PCR with SYBR green reagent and specific primers was used to monitor levels of the mitochondrial cytochrome c oxidase subunit II gene and normalized to levels of the nucleus-encoded Ucp1 promoter.

Western blots.Sodium dodecyl sulfate-polyacrylamide gel electrophoresis, Western blotting, and immunodetection were performed as described previously (6) using rabbit polyclonal anti-mouse UCP1 (AB1426; Chemicon International) at a ratio of 1/1,000 and mouse monoclonal anti-β-actin (ab6276; Abcam Ltd., Cambridge, United Kingdom) at 1/5,000. Bands were visualized using secondary peroxidase-conjugated antibody and enhanced chemiluminescence.

Immunocytochemical staining.RIPKO-1 cells cultured on chamber slides were fixed in methanol and permeabilized in 0.2% Triton. UCP1 was detected using a rabbit polyclonal anti-mouse UCP1 antibody (AB3038; Chemicon International) diluted 1/100, a procedure followed by incubation with fluorescein isothiocyanate-conjugated swine anti-rabbit immunoglobulin G (IgG) (DAKO) diluted 1/50. Cell numbers were assessed by counting nuclei stained in the presence of a DAPI (4′,6′diamidino-2-phenylindole)-containing Vectashield mountant (Vector Labs).

Mitochondrial stain.Cells were incubated for 30 min in MitoTracker Green (Molecular Probes) at 100 nM, washed in PBS, and mounted in Vectashield medium (Vector Labs).

[3H]palmitate oxidation assay.Following differentiation, primary MEFs and RIPKO-1, RIPKO-L, and 3T3-L1 cells were assayed for 3H2O production from excess [3H]palmitic acid as described previously (38).

Chromatin immunoprecipitation (ChIP) assay.Cells were incubated in the protein-protein cross-linking reagent dimethyl adipimidate · 2HCl (Pierce) at 10 mM in PBS for 30 min, followed by incubation in 1% formaldehyde in DMEM for 5 min at 37°C. The cross-linked cells were lysed, sonicated, and immunoprecipitated with protein A/G PLUS-agarose (Santa Cruz) according to the manufacturer's instructions using mouse anti-V5 antibody (Invitrogen) or control normal mouse IgG (Santa Cruz). DNA fragments were purified with a QIAquick PCR purification kit (QIAGEN) and used for templates in PCRs.

Affymetrix array hybridization and data analysis.Total RNA was isolated from three samples each of RIPKO-1 and RIPKO-L cells, both undifferentiated and after adipocyte differentiation (10 days) in the presence of the PPARγ ligand GW1929 (2.5 μm) (Sigma). Two RIP140-lentivirus-transduced cell lines were used for profiling in order to avoid variation due to differences in the sites of incorporation of the virus. Affymetrix array hybridization and scanning were performed by the CSC/IC Microarray Centre, Imperial College London, Hammersmith Campus, using murine 430 2.0 chips. Array data were analyzed with d-CHIP software (19). The microarray data are available at http://www.ebi.ac.uk/arrayexpress/ under accession number E-MIMR-42 .

RESULTS

Adipogenesis of wild-type and RIP140-null cells.To investigate the intrinsic role of RIP140 in adipocytes, primary cultures of WAT and MEFs were induced to differentiate with a standard hormone cocktail in the absence or presence of the PPARγ agonist rosiglitazone. In these primary cell systems, both wild-type and RIP140-null cells underwent adipogenesis, as judged morphologically by the accumulation of cytosolic fat droplets (data not shown). In addition to these changes, the induction of the adipocyte marker aP2 gene and the adipocyte transcription factor PPARγ2 was detected in all cell types following differentiation (Fig. 1A and B).

FIG. 1.
  • Open in new tab
  • Download powerpoint
FIG. 1.

Gene expression in wild-type and RIP140-null cells. Results from real-time PCR analysis of mRNAs for factors required for adipocyte function and energy dissipation are shown. (A and B) Pri-mary cultures of WAT and MEFs were untreated (Co) or differentiated in the absence (D) or presence (D+Ro) of rosiglitazone. Cells were harvested after 9 days (WAT) or 6 days (MEFs) of treatment. (C) RIPKO-1 cells preconfluent (PC) and differentiated in the presence of Ro up to day 10. Data are expressed relative to results for untreated RIP140-null cells.

The absence of RIP140 in mice results in altered expression in adipose tissue of key genes involved in energy metabolism, including Ucp1 and the carnitine palmitoyltransferase 1b gene (Cpt1b); however, the signaling mechanisms in vivo are complex, involving intrinsic and systemic factors. We therefore analyzed the expression levels of these genes in the cell culture systems. Ucp1 and Cpt1b were more highly expressed in primary cultures of WAT and MEFs devoid of RIP140, with expression levels being dependent on differentiation (Fig. 1A and B).

We generated a MEF line lacking the RIP140 gene (termed RIPKO-1) by continuous culturing of the primary MEFs. This cell line retained the potential to differentiate into adipocytes, as judged morphologically by cytosolic fat droplet accumulation (data not shown). Consistent with these changes, aP2 and PPARγ were induced in the cells, with maximal expression being reached by day 6 of treatment (Fig. 1C, upper panel). Ucp1 and Cpt1b displayed progressive increases in expression in the RIPKO-1 cells (Fig. 1C, lower panel). The expression of Ucp1 in BAT is normally associated with the transcription factor PGC1α. However, expression levels of PGC1α or of the related gene PGC1β were similar in wild-type and RIP140-null primary WAT adipocytes (data not shown).

Identification of RIP140 target genes following reexpression of RIP140 in RIPKO-1 cells.To verify an essential role for RIP140 in gene regulation, it was reexpressed in the RIPKO-1 cells using a lentiviral vector. Adipocyte differentiation of stably expressing cell lines (termed RIPKO-L cells) was unaltered compared to that of the parental null cells, as determined by oil red O staining (Fig. 2A). Furthermore, both aP2 and PPARγ2 gene expression levels were progressively increased during differentiation, as observed in both RIPKO-1 and 3T3-L1 cells (data not shown). The expression levels of RIP140 in the RIPKO-L and 3T3-L1 cells were similar at days 5 and 14 following differentiation (Fig. 2B). However, the patterns of expression differed between the cell lines; expression increased in 3T3-L1 cells following differentiation, whereas in RIPKO-L cells, expression was reduced. This difference is due to the reintroduced RIP140 being under the control of the cytomegalovirus promoter rather than that of its endogenous regulatory mechanisms.

FIG.2.
  • Open in new tab
  • Download powerpoint
FIG.2.

Identification of RIP140 target genes following reexpression of RIP140 in RIPKO-1 adipocytes. (A) Oil red O staining of undifferentiated and differentiated RIPKO-1 and RIPKO-L cells. (B) Real-time PCR analysis of RIP140 expression in RIPKO-1, RIPKO-L, and 3T3-L1 cells harvested untreated (day 0) or differentiated in the presence of rosiglitazone (days 5 and 14). Data are expressed relative to results for untreated 3T3-L1 cells. (C) Cluster analysis of gene expression in undifferentiated and differentiated RIPKO-1 and RIPKO-L cells. Adipogenesis-related genes are listed in the upper panel. The middle panel lists genes that are upregulated by at least twofold in RIPKO-1 cells upon differentiation yet repressed fivefold upon RIP140 reexpression. Genes associated with BAT are listed in the lower panel. S. pombe, Schizosaccharomyces pombe.

To identify genes that, in adipocytes, are targeted for regulation by RIP140, we performed expression profiling using Affymetrix microarrays. Gene expression in RIPKO-1 cells was compared to that in cells in which RIP140 had been reintroduced with a lentiviral vector. The transcription factors C/EBPα, PPARγ, and SREBP1, which are essential for normal adipocyte differentiation and function, were induced following differentiation in both RIPKO-1 and RIPKO-L cells (Fig. 2C, upper panel). Genes that are associated with adipocyte function, such as those for SCD1, aP2, and adipsin, were also induced.

The microarray data were analyzed to identify genes that are induced in mature adipocytes and may be targeted for repression by RIP140. We selected genes that are upregulated by at least twofold in RIPKO-1 cells upon differentiation yet are repressed fivefold upon RIP140 reexpression (Fig. 2C, middle panel). The Ucp1 gene was identified within this gene cluster, showing that the reexpression of RIP140 in RIPKO-1 cells ablates the adipocyte-dependent expression of this gene. This analysis identified a number of additional genes repressed by the reexpression of RIP140 (Fig. 2C, middle panel). These include genes implicated in adipocyte function, namely, the Cidea (cell-death-inducing DFF45-like effector A) (41), Aquaporin 7 (AQPap) (13), β3-adrenergic receptor (2), carboxylesterase 3 (34), acetyl coenzyme A synthetase 2 (33), and membrane metalloendopeptidase (31) genes. In addition, a number of other genes are similarly repressed by RIP140 expression (Fig. 2C, middle panel). In contrast, the expression levels of key regulators and markers of BAT function (37) are unaltered or only moderately affected (Fig. 2C, lower panel). For example, PGC1α and FOXC2 were expressed at lower levels in RIP140-null cells.

Cidea expression is altered in RIP140−/− cells in vitro and in vivo.We next investigated the regulation of the Cidea gene, identified by the microarray study as highly expressed only in differentiated RIPKO-1 cells, which has been implicated in thermogenesis through the direct modulation of Ucp1 activity (41). Cidea mRNA expression was undetectable in RIPKO-1, RIPKO-L, and 3T3-L1 adipocytes prior to differentiation and, following adipogenesis, increased only in RIP140-null cells (Fig. 3A, left panel); the expression profile closely resembled that of Ucp1 (Fig. 4A).

FIG. 3.
  • Open in new tab
  • Download powerpoint
FIG. 3.

RIP140 represses transcription of the Cidea gene. Results from real-time PCR analysis of Cidea expression are shown. (A) Cell lines, as indicated, were harvested untreated (day 0), or differentiated in the presence of rosiglitazone (days 5 and 14). Primary MEFs that were untreated (Co) or differentiated in the absence (D) or presence of rosiglitazone (D+Ro) were harvested after 6 days of treatment. (B) Expression in WAT from wild-type (n = 4) and RIP140-null (n = 4) mice. Real-time PCR data are expressed relative to maximal expression. Detection of Cidea in WAT from wild-type and RIP140-null mice by immunohistochemistry is shown (right panel).

FIG. 4.
  • Open in new tab
  • Download powerpoint
FIG. 4.

RIP140 targets the Ucp1 promoter to repress transcription. (A) Real-time PCR analysis of Ucp1 expression in cell lines that were either untreated (day 0) or differentiated in the presence of rosiglitazone (days 5 and 14), as indicated. Data are expressed relative to results for untreated RIPKO-1 cells. (B) Immunoblot for UCP1 and β-actin (loading control) in RIPKO-1 and RIPKO-L cells untreated and differentiated in the presence of rosiglitazone (D+Ro) (day 14) (left panel), and immunocytochemical staining for UCP1 (right panel) in untreated cells (day 0) and cells differentiated in the presence of Ro (days 7, 9, and 14). (C) Transient transfection of RIPKO-1 cells with reporter constructs pGL3-Basic, Ucp1(4kb)/luc, or Ucp1(4kbΔEnh)/luc in the absence and presence of RIP140, differentiated for 5 days in the presence of rosiglitazone. (D) Transient transfection of RIPKO-1 cells with full-length 4-kb or 220-bp enhancer element Ucp1 promoter-luciferase reporter genes in the absence and presence of RIP140. Cells were harvested 1, 3, and 5 days following control or differentiation-plus Ro treatment (D+Ro). (E) Chromatin immunoprecipitation of the Ucp1 enhancer or upstream control region in RIPKO-1 and RIPKO-L16 cells with control (IgG) or anti-V5 antibody.

To confirm these changes, we determined the expression patterns of Cidea in primary adipocyte culture systems and in vivo. Cidea was undetectable in undifferentiated primary MEFs and induced only in the RIP140-null cells (Fig. 3A, right panel). A similar pattern was also observed in primary WAT cultures (data not shown). Quantitative analysis of Cidea expression showed a 15-fold increase in WAT in RIP140-null mice relative to wild-type mice (Fig. 3B). Immunohistochemical staining revealed that the expression of Cidea in unilocular adipocytes in WAT of RIP140−/− mice (Fig. 3B) localized predominantly in the cytoplasmic area near the plasma membrane, similarly to Ucp1.

RIP140 targets the Ucp1 promoter to repress transcription.To investigate further the mechanism of action of RIP140, we analyzed its ability to repress transcription from the Ucp1 promoter. Real-time PCR showed that Ucp1 expression was reduced in RIPKO-1 cells upon exogenous RIP140 expression to generate the RIPKO-L cells (Fig. 4A). Western blot analysis showed an induction of a 32-kDa immunoreactive band corresponding to UCP1 in fully differentiated RIP140-null cells (Fig. 4B). Immunocytochemical staining of RIPKO-1 cells showed a progressive increase in UCP1 as differentiation proceeded (Fig. 4B). This staining verified that UCP1 was confined to the lipid-containing differentiated cells and localized to the cytoplasm in accordance with its role as a protein confined to the mitochondrial compartment.

In mouse brown adipocytes, the transcription from the Ucp1 gene promoter is under the control of a region up to 4 kb upstream of the transcriptional start site (4, 16). Subsequent studies with transgenic mice have identified a 220-bp enhancer from kb −2.5 to −2.3 that controls tissue-specific expression (32). RIPKO-1 cells were transiently transfected with a luciferase reporter gene under the control of either the wild-type 4-kb Ucp1 promoter or a construct with the 220-bp enhancer region deleted (4kbΔEnh). Reporter activity was reduced by 50% with the deletion of the enhancer element (Fig. 4C). Exogenous expression of RIP140 inhibited luciferase activity from both the wild-type and 4kbΔEnh constructs. Following the demonstration that the enhancer region was necessary for the full induction of Ucp1 promoter activity, RIPKO-1 cells were transiently transfected with a luciferase reporter gene under the control of either the 4-kb Ucp1 promoter or the 220-bp enhancer element linked to the thymidine kinase promoter from −105 to +50. There was a progressive increase in promoter activity as the cells differentiated. Coexpression of RIP140 inhibited the Ucp1 promoter activity from both the full-length promoter and the 220-bp enhancer element, thus identifying this specific region as a target for RIP140 suppression of Ucp1 gene transcription (Fig. 4D).

Direct association of RIP140 with the Ucp1 enhancer element was determined using ChIP assays. The expression of RIP140-V5 in the RIPKO-L cells does not exceed that of the endogenous RIP140 in differentiated 3T3-L1 adipocytes (Fig. 2B). An antibody specific for V5 precipitated the Ucp1 enhancer element in differentiated RIPKO-L cells but not a control region 15 kb upstream of the mouse Ucp1 gene (Fig. 4E). Enrichment of the Ucp1 enhancer element was not observed with the V5 antibody in RIPKO-1 cells devoid of RIP140. Thus, RIP140 is recruited to the Ucp1 enhancer element in differentiated RIPKO-L cells.

Increased β-oxidation in RIP140-null adipocytes.Following the identification of the Ucp1 gene as a target for RIP140, we investigated the functional consequences of increased Ucp1 expression. The induction of factors that facilitate fatty acid oxidation and uncouple respiration leads to an increased capacity for β-oxidation. Measurement of the rate of 3H2O production derived from a [3H]palmitic acid substrate showed that RIP140-null adipocytes derived from primary MEFs exhibited a total fatty acid oxidation 1.5-fold higher than that of wild-type adipocytes (Fig. 5A). The cells had differentiated to similar extents, as judged by the expression of aP2; however, Ucp1 levels were elevated in the RIP140-null adipocytes (Fig. 5A). Measurement of the ratio of mitochondrial DNA to nuclear DNA in primary MEFs showed that mitochondrial biogenesis occurred in both wild-type and RIP140-null cells following differentiation (Fig. 5A). However, the absence of RIP140 did not significantly affect the level of mitochondrial DNA in the differentiated cells. The specific uptake of a MitoTracker mitochondrion-selective probe revealed that both RIPKO-1 and RIPKO-L differentiated cells contained more mitochondria than undifferentiated cells did (Fig. 5B).

FIG. 5.
  • Open in new tab
  • Download powerpoint
FIG. 5.

Mitochondrial biogenesis and β-oxidation in RIP140-null adipocytes. (A) Wild-type (WT) and RIP140−/− (KO) primary MEFs differentiated for 9 days were assayed for β-oxidation of [3H]palmitic acid (left panel; data are expressed as cpm/cell relative to results for the wild type), and expression levels of aP2 and Ucp1 were assayed by real-time PCR (center panel) and by measurement of the ratio of mitochondrial DNA to nuclear DNA (right panel). (B) MitoTracker staining of RIPKO-1 and RIPKO-L cells. (C) β-Oxidation of [3H]palmitic acid in differentiated cells (right panel) and aP2 expression in untreated cells (Co) and in cells differentiated in the presence of Ro (D+Ro) (left panel).

The rate of 3H2O production derived from [3H]palmitic acid was determined in RIPKO-1, RIPKO-L, and 3T3-L1 cells that were differentiated to similar extents, as judged by aP2 expression (Fig. 5C). RIP140-null adipocytes exhibited total fatty acid oxidation levels twofold higher than those of wild-type 3T3-L1 adipocytes, and this level was repressed by the reexpression of RIP140 (Fig. 5C). The potential to prevent the induction of the expression of genes involved in energy dissipation demonstrates a primary role for RIP140 in this process and provides a mechanism for regulating β-oxidation within adipose cells.

DISCUSSION

Adipogenesis and adipocyte functions in vivo are regulated by multiple mechanisms that involve combinations of cell-autonomous and systemic factors. To study the intrinsic role of RIP140 in adipocytes, we differentiated primary WAT cells, MEFs, and a RIP140-null cell line. In particular, the RIP140-null cells (RIPKO-1) and cell lines derived from it in which RIP140 is reexpressed (RIPKO-L) provide systems for elucidating the cell-autonomous mechanisms by which this corepressor regulates the expression of target genes involved in energy dissipation. In microarray analysis of both cell types, key regulators and markers of the adipogenic cascade, including PPARγ, C/EBPα, SCD1, aP2, and adipsin, are induced. Thus, the adipogenic program is not perturbed either by the absence of or by the exogenous expression of RIP140.

Affymetrix microarray analysis was used to identify genes that are targeted for repression by RIP140 in mature adipocytes. These studies confirm the in vivo observations that Ucp1, a key factor in energy expenditure in BAT which is upregulated in the WAT in RIP140-null mice, is also derepressed following the loss of RIP140 in isolated adipocytes. The Cpt1b gene, which was also shown to be elevated in expression in vivo, is similarly increased in null cells, but to a lesser extent than Ucp1 is. In addition to Ucp1, we identified a number of genes regulated with similar expression profiles. These include the gene encoding Cidea, a protein reported to interact directly with Ucp1 to modulate the process of thermogenesis (41), which is downregulated by obesity in human WAT (23). We hypothesize therefore that RIP140 may be directly involved in the prevention of the expression of genes such as those encoding UCP1, CPT1b, and Cidea. In particular, the demonstration that novel RIP140 targets such as Cidea are derepressed in primary cells, mouse embryo fibroblasts, and in vivo WAT provides evidence to validate the expression profile differences determined by the microarray studies.

The increased Ucp1, Cpt1b, and Cidea expression in the absence of RIP140 occurs progressively during differentiation, suggesting that it is dependent on factors generated during adipogenesis. This progressive increase was also observed when transcription from a transfected Ucp1 gene promoter was analyzed for RIPKO-1 cells. The upregulation seems to be mediated predominantly by a previously characterized 220-bp enhancer element in the promoter containing binding sites for PPARs and TR/RXR (1, 29, 32). Importantly, we found that this element was a target for repression by RIP140 in transfected RIPKO-1 cells. This was further confirmed using ChIP experiments, demonstrating that RIP140 is recruited directly to this regulatory element in differentiated RIPKO-L cells where Ucp1 expression is abrogated. In brown fat cells, PPARs, together with PGC-1α, have been shown to stimulate transcription from the Ucp1 promoter (1, 28, 32). Ucp1 expression is also regulated by neural signals under the control of the sympathetic nervous system that result in the activation of transcription factors such as ATF2 (2, 3). The levels of PGC-1α and PGC-1β did not differ between wild-type and RIP140-null adipocytes, suggesting that altered expression of these coactivators is not essential for Ucp1 expression in the absence of RIP140.

In addition to the Ucp1 gene, a number of other genes are regulated with similar expression profiles, many of which are expressed in adipocytes and may also be nuclear receptor targets, including the AQPap gene, encoding a protein forming adipose-specific glycerol channels (14). Whether such genes are also direct targets of RIP140 or are regulated indirectly by additional factors is still to be determined. In contrast, other genes implicated in BAT function, such as the FOXC2 gene, were not increased in cells lacking RIP140. It is apparent that the absence of RIP140 in adipose cells results in increased levels of a subset of genes normally restricted to BAT, some of which regulate metabolic uncoupling processes and hence energy expenditure.

It is noteworthy that the abilities of primary MEFs and 3T3-L1, RIPKO-1, and RIPKO-L cells to metabolize palmitate correlated with the expression levels of Ucp1, Cpt1b, and Cidea, with the highest rates observed in RIP140-null cells. Mitochondrial biogenesis is a fundamental aspect of white adipose cell differentiation (40); however, the degrees of mitochondrial biogenesis did not differ significantly between wild-type adipocytes and those null for the RIP140 gene. The increased number of mitochondria in differentiated cells, along with the elevation of Ucp1, provides increased capacity for the enhanced β-oxidation in cells lacking RIP140. These data demonstrating altered gene expression and mitochondrial respiration in RIP140-null adipocytes are in agreement with both expression-profiling studies and the increased O2 consumption in vivo found in RIP140-null mice (17).

Our studies demonstrate that processes that control metabolism and energy homeostasis in the adipocyte involve the direct action of a ligand-dependent NR corepressor and are independent of systemic effects. In summary, it has been demonstrated by a number of studies that NRs and their coactivators, by integrating different hormonal signals to regulate gene expression, play a fundamental role in the regulation of both the differentiation and the metabolic function of adipocytes. Here we describe a major role in adipocytes for a ligand-dependent NR corepressor in the prevention of the expression of genes that are associated with energy dissipation. Therefore, the prevention of RIP140 recruitment to receptors or a reduction in the levels of RIP140 itself may provide novel mechanisms for the control of energy use in adipose cells and assist in the treatment of obesity-related disorders.

ACKNOWLEDGMENTS

We thank J. Steel and the Molecular Endocrinology group for technical assistance.

This work was supported by the Wellcome Trust (M.C., E.K., and G.L.) grant no. 061930 and for a PhD studentship (D.D.) grant no. 069361.

FOOTNOTES

    • Received 4 March 2005.
    • Returned for modification 8 April 2005.
    • Accepted 5 August 2005.
  • Copyright © 2005 American Society for Microbiology

REFERENCES

  1. 1.↵
    Barbera, M. J., A. Schluter, N. Pedraza, R. Iglesias, F. Villarroya, and M. Giralt. 2001. Peroxisome proliferator-activated receptor alpha activates transcription of the brown fat uncoupling protein-1 gene. A link between regulation of the thermogenic and lipid oxidation pathways in the brown fat cell. J. Biol. Chem. 276 : 1486-1493.
    OpenUrlAbstract/FREE Full Text
  2. 2.↵
    Cannon, B., and J. Nedergaard. 2004. Brown adipose tissue: function and physiological significance. Physiol. Rev. 84 : 277-359.
    OpenUrlCrossRefPubMedWeb of Science
  3. 3.↵
    Cao, W., K. W. Daniel, J. Robidoux, P. Puigserver, A. V. Medvedev, X. Bai, L. M. Floering, B. M. Spiegelman, and S. Collins. 2004. p38 mitogen-activated protein kinase is the central regulator of cyclic AMP-dependent transcription of the brown fat uncoupling protein 1 gene. Mol. Cell. Biol. 24 : 3057-3067.
    OpenUrlAbstract/FREE Full Text
  4. 4.↵
    Cassard-Doulcier, A. M., C. Gelly, N. Fox, J. Schrementi, S. Raimbault, S. Klaus, C. Forest, F. Bouillaud, and D. Ricquier. 1993. Tissue-specific and beta-adrenergic regulation of the mitochondrial uncoupling protein gene: control by cis-acting elements in the 5′-flanking region. Mol. Endocrinol. 7 : 497-506.
    OpenUrlCrossRefPubMedWeb of Science
  5. 5.↵
    Cederberg, A., L. M. Gronning, B. Ahren, K. Tasken, P. Carlsson, and S. Enerback. 2001. FOXC2 is a winged helix gene that counteracts obesity, hypertriglyceridemia, and diet-induced insulin resistance. Cell 106 : 563-573.
    OpenUrlCrossRefPubMedWeb of Science
  6. 6.↵
    Christian, M., X. Zhang, T. Schneider-Merck, T. G. Unterman, B. Gellersen, J. O. White, and J. J. Brosens. 2002. Cyclic AMP-induced forkhead transcription factor, FKHR, cooperates with CCAAT/enhancer-binding protein beta in differentiating human endometrial stromal cells. J. Biol. Chem. 277 : 20825-20832.
    OpenUrlAbstract/FREE Full Text
  7. 7.↵
    Cinti, S. 2001. The adipose organ: morphological perspectives of adipose tissues. Proc. Nutr. Soc. 60 : 319-328.
    OpenUrlCrossRefPubMedWeb of Science
  8. 8.↵
    Del Mar Gonzalez-Barroso, M., D. Ricquier, and A. M. Cassard-Doulcier. 2000. The human uncoupling protein-1 gene (UCP1): present status and perspectives in obesity research. Obes. Rev. 1 : 61-72.
    OpenUrlCrossRefPubMed
  9. 9.↵
    Dulloo, A. G., and D. S. Miller. 1984. Energy balance following sympathetic denervation of brown adipose tissue. Can. J. Physiol. Pharmacol. 62 : 235-240.
    OpenUrlCrossRefPubMedWeb of Science
  10. 10.↵
    Flier, J. S. 2004. Obesity wars: molecular progress confronts an expanding epidemic. Cell 116 : 337-350.
    OpenUrlCrossRefPubMedWeb of Science
  11. 11.↵
    Hansen, J. B., C. Jorgensen, R. K. Petersen, P. Hallenborg, R. De Matteis, H. A. Boye, N. Petrovic, S. Enerback, J. Nedergaard, S. Cinti, H. te Riele, and K. Kristiansen. 2004. Retinoblastoma protein functions as a molecular switch determining white versus brown adipocyte differentiation. Proc. Natl. Acad. Sci. USA 101 : 4112-4117.
    OpenUrlAbstract/FREE Full Text
  12. 12.↵
    Himms-Hagen, J. 1990. Brown adipose tissue thermogenesis: interdisciplinary studies. FASEB J. 4 : 2890-2898.
    OpenUrlCrossRefPubMedWeb of Science
  13. 13.↵
    Kishida, K., I. Shimomura, H. Nishizawa, N. Maeda, H. Kuriyama, H. Kondo, M. Matsuda, H. Nagaretani, N. Ouchi, K. Hotta, S. Kihara, T. Kadowaki, T. Funahashi, and Y. Matsuzawa. 2001. Enhancement of the aquaporin adipose gene expression by a peroxisome proliferator-activated receptor gamma. J. Biol. Chem. 276 : 48572-48579.
    OpenUrlAbstract/FREE Full Text
  14. 14.↵
    Kondo, H., I. Shimomura, K. Kishida, H. Kuriyama, Y. Makino, H. Nishizawa, M. Matsuda, N. Maeda, H. Nagaretani, S. Kihara, Y. Kurachi, T. Nakamura, T. Funahashi, and Y. Matsuzawa. 2002. Human aquaporin adipose (AQPap) gene. Genomic structure, promoter analysis and functional mutation. Eur. J. Biochem. 269 : 1814-1826.
    OpenUrlPubMedWeb of Science
  15. 15.↵
    Kopecky, J., G. Clarke, S. Enerback, B. Spiegelman, and L. P. Kozak. 1995. Expression of the mitochondrial uncoupling protein gene from the aP2 gene promoter prevents genetic obesity. J. Clin. Investig. 96 : 2914-2923.
    OpenUrlCrossRefPubMedWeb of Science
  16. 16.↵
    Kozak, U. C., J. Kopecky, J. Teisinger, S. Enerback, B. Boyer, and L. P. Kozak. 1994. An upstream enhancer regulating brown-fat-specific expression of the mitochondrial uncoupling protein gene. Mol. Cell. Biol. 14 : 59-67.
    OpenUrlAbstract/FREE Full Text
  17. 17.↵
    Leonardsson, G., J. H. Steel, M. Christian, V. Pocock, S. Milligan, J. Bell, P. W. So, G. Medina-Gomez, A. Vidal-Puig, R. White, and M. G. Parker. 2004. Nuclear receptor corepressor RIP140 regulates fat accumulation. Proc. Natl. Acad. Sci. USA 101 : 8437-8442.
    OpenUrlAbstract/FREE Full Text
  18. 18.↵
    Leone, T. C., J. J. Lehman, B. N. Finck, P. J. Schaeffer, A. R. Wende, S. Boudina, M. Courtois, D. F. Wozniak, N. Sambandam, C. Bernal-Mizrachi, Z. Chen, J. O. Holloszy, D. M. Medeiros, R. E. Schmidt, J. E. Saffitz, E. D. Abel, C. F. Semenkovich, and D. P. Kelly. 2005. PGC-1alpha deficiency causes multi-system energy metabolic derangements: muscle dysfunction, abnormal weight control and hepatic steatosis. PLoS Biol. 3 : e101.
    OpenUrlCrossRefPubMed
  19. 19.↵
    Li, C., and W. H. Wong. 2001. Model-based analysis of oligonucleotide arrays: expression index computation and outlier detection. Proc. Natl. Acad. Sci. USA 98 : 31-36.
    OpenUrlAbstract/FREE Full Text
  20. 20.↵
    Lin, J., P. H. Wu, P. T. Tarr, K. S. Lindenberg, J. St-Pierre, C. Y. Zhang, V. K. Mootha, S. Jager, C. R. Vianna, R. M. Reznick, L. Cui, M. Manieri, M. X. Donovan, Z. Wu, M. P. Cooper, M. C. Fan, L. M. Rohas, A. M. Zavacki, S. Cinti, G. I. Shulman, B. B. Lowell, D. Krainc, and B. M. Spiegelman. 2004. Defects in adaptive energy metabolism with CNS-linked hyperactivity in PGC-1alpha null mice. Cell 119 : 121-135.
    OpenUrlCrossRefPubMedWeb of Science
  21. 21.↵
    Lowell, B. B., V. S.-Susulic, A. Hamann, J. A. Lawitts, J. Himms-Hagen, B. B. Boyer, L. P. Kozak, and J. S. Flier. 1993. Development of obesity in transgenic mice after genetic ablation of brown adipose tissue. Nature 366 : 740-742.
    OpenUrlCrossRefPubMedWeb of Science
  22. 22.↵
    Morrison, R. F., and S. R. Farmer. 2000. Hormonal signaling and transcriptional control of adipocyte differentiation. J. Nutr. 130 : 3116S-3121S.
    OpenUrlPubMedWeb of Science
  23. 23.↵
    Nordstrom, E. A., M. Ryden, E. C. Backlund, I. Dahlman, M. Kaaman, L. Blomqvist, B. Cannon, J. Nedergaard, and P. Arner. 2005. A human-specific role of cell death-inducing DFFA (DNA fragmentation factor-alpha)-like effector A (CIDEA) in adipocyte lipolysis and obesity. Diabetes 54 : 1726-1734.
    OpenUrlAbstract/FREE Full Text
  24. 24.↵
    Ntambi, J. M., and K. Young-Cheul. 2000. Adipocyte differentiation and gene expression. J. Nutr. 130 : 3122S-3126S.
    OpenUrlPubMedWeb of Science
  25. 25.↵
    Penicaud, L., B. Cousin, C. Leloup, A. Lorsignol, and L. Casteilla. 2000. The autonomic nervous system, adipose tissue plasticity, and energy balance. Nutrition 16 : 903-908.
    OpenUrlCrossRefPubMed
  26. 26.↵
    Picard, F., M. Gehin, J. Annicotte, S. Rocchi, M. F. Champy, B. W. O'Malley, P. Chambon, and J. Auwerx. 2002. SRC-1 and TIF2 control energy balance between white and brown adipose tissues. Cell 111 : 931-941.
    OpenUrlCrossRefPubMedWeb of Science
  27. 27.↵
    Puigserver, P., and B. M. Spiegelman. 2003. Peroxisome proliferator-activated receptor-gamma coactivator 1alpha (PGC-1alpha): transcriptional coactivator and metabolic regulator. Endocr. Rev. 24 : 78-90.
    OpenUrlCrossRefPubMedWeb of Science
  28. 28.↵
    Puigserver, P., Z. Wu, C. W. Park, R. Graves, M. Wright, and B. M. Spiegelman. 1998. A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell 92 : 829-839.
    OpenUrlCrossRefPubMedWeb of Science
  29. 29.↵
    Rabelo, R., C. Reyes, A. Schifman, and J. E. Silva. 1996. Interactions among receptors, thyroid hormone response elements, and ligands in the regulation of the rat uncoupling protein gene expression by thyroid hormone. Endocrinology 137 : 3478-3487.
    OpenUrlCrossRefPubMedWeb of Science
  30. 30.↵
    Rosen, E. D., C. J. Walkey, P. Puigserver, and B. M. Spiegelman. 2000. Transcriptional regulation of adipogenesis. Genes Dev. 14 : 1293-1307.
    OpenUrlFREE Full Text
  31. 31.↵
    Schling, P., and T. Schafer. 2002. Human adipose tissue cells keep tight control on the angiotensin II levels in their vicinity. J. Biol. Chem. 277 : 48066-48075.
    OpenUrlAbstract/FREE Full Text
  32. 32.↵
    Sears, I. B., M. A. MacGinnitie, L. G. Kovacs, and R. A. Graves. 1996. Differentiation-dependent expression of the brown adipocyte uncoupling protein gene: regulation by peroxisome proliferator-activated receptor gamma. Mol. Cell. Biol. 16 : 3410-3419.
    OpenUrlAbstract/FREE Full Text
  33. 33.↵
    Sone, H., H. Shimano, Y. Sakakura, N. Inoue, M. Amemiya-Kudo, N. Yahagi, M. Osawa, H. Suzuki, T. Yokoo, A. Takahashi, K. Iida, H. Toyoshima, A. Iwama, and N. Yamada. 2002. Acetyl-coenzyme A synthetase is a lipogenic enzyme controlled by SREBP-1 and energy status. Am. J. Physiol. Endocrinol. Metab. 282 : E222-E230.
    OpenUrlCrossRefPubMedWeb of Science
  34. 34.↵
    Soni, K. G., R. Lehner, P. Metalnikov, P. O'Donnell, M. Semache, W. Gao, K. Ashman, A. V. Pshezhetsky, and G. A. Mitchell. 2004. Carboxylesterase 3 (EC 3.1.1.1) is a major adipocyte lipase. J. Biol. Chem. 279 : 40683-40689.
    OpenUrlAbstract/FREE Full Text
  35. 35.↵
    Soukas, A., N. D. Socci, B. D. Saatkamp, S. Novelli, and J. M. Friedman. 2001. Distinct transcriptional profiles of adipogenesis in vivo and in vitro. J. Biol. Chem. 276 : 34167-34174.
    OpenUrlAbstract/FREE Full Text
  36. 36.↵
    Spiegelman, B. M., and J. S. Flier. 2001. Obesity and the regulation of energy balance. Cell 104 : 531-543.
    OpenUrlCrossRefPubMedWeb of Science
  37. 37.↵
    Unami, A., Y. Shinohara, K. Kajimoto, and Y. Baba. 2004. Comparison of gene expression profiles between white and brown adipose tissues of rat by microarray analysis. Biochem. Pharmacol. 67 : 555-564.
    OpenUrlCrossRefPubMed
  38. 38.↵
    Wang, Y. X., C. H. Lee, S. Tiep, R. T. Yu, J. Ham, H. Kang, and R. M. Evans. 2003. Peroxisome-proliferator-activated receptor delta activates fat metabolism to prevent obesity. Cell 113 : 159-170.
    OpenUrlCrossRefPubMedWeb of Science
  39. 39.↵
    White, R., G. Leonardsson, I. Rosewell, M. A. Jacobs, S. Milligan, and M. Parker. 2000. The nuclear receptor co-repressor nrip1 (RIP140) is essential for female fertility. Nat. Med. 6 : 1368-1374.
    OpenUrlCrossRefPubMedWeb of Science
  40. 40.↵
    Wilson-Fritch, L., A. Burkart, G. Bell, K. Mendelson, J. Leszyk, S. Nicoloro, M. Czech, and S. Corvera. 2003. Mitochondrial biogenesis and remodeling during adipogenesis and in response to the insulin sensitizer rosiglitazone. Mol. Cell. Biol. 23 : 1085-1094.
    OpenUrlAbstract/FREE Full Text
  41. 41.↵
    Zhou, Z., S. Y. Toh, Z. Chen, K. Guo, C. P. Ng, S. Ponniah, S. C. Lin, W. Hong, and P. Li. 2003. Cidea-deficient mice have lean phenotype and are resistant to obesity. Nat. Genet. 35 : 49-56.
    OpenUrl
PreviousNext
Back to top
Download PDF
Citation Tools
RIP140-Targeted Repression of Gene Expression in Adipocytes
Mark Christian, Evangelos Kiskinis, Darja Debevec, Göran Leonardsson, Roger White, Malcolm G. Parker
Molecular and Cellular Biology Oct 2005, 25 (21) 9383-9391; DOI: 10.1128/MCB.25.21.9383-9391.2005

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Print

Alerts
Sign In to Email Alerts with your Email Address
Email

Thank you for sharing this Molecular and Cellular Biology article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
RIP140-Targeted Repression of Gene Expression in Adipocytes
(Your Name) has forwarded a page to you from Molecular and Cellular Biology
(Your Name) thought you would be interested in this article in Molecular and Cellular Biology.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
RIP140-Targeted Repression of Gene Expression in Adipocytes
Mark Christian, Evangelos Kiskinis, Darja Debevec, Göran Leonardsson, Roger White, Malcolm G. Parker
Molecular and Cellular Biology Oct 2005, 25 (21) 9383-9391; DOI: 10.1128/MCB.25.21.9383-9391.2005
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Top
  • Article
    • ABSTRACT
    • MATERIALS AND METHODS
    • RESULTS
    • DISCUSSION
    • ACKNOWLEDGMENTS
    • FOOTNOTES
    • REFERENCES
  • Figures & Data
  • Info & Metrics
  • PDF

KEYWORDS

adipocytes
Gene Expression Profiling
Gene Expression Regulation
Nuclear Proteins

Related Articles

Cited By...

About

  • About MCB
  • Editor in Chief
  • Editorial Board
  • Policies
  • For Reviewers
  • For the Media
  • For Librarians
  • For Advertisers
  • Alerts
  • RSS
  • FAQ
  • Permissions
  • Journal Announcements

Authors

  • ASM Author Center
  • Submit a Manuscript
  • Article Types
  • Ethics
  • Contact Us

Follow #MCBJournal

@ASMicrobiology

       

ASM Journals

ASM journals are the most prominent publications in the field, delivering up-to-date and authoritative coverage of both basic and clinical microbiology.

About ASM | Contact Us | Press Room

 

ASM is a member of

Scientific Society Publisher Alliance

 

American Society for Microbiology
1752 N St. NW
Washington, DC 20036
Phone: (202) 737-3600

Copyright © 2021 American Society for Microbiology | Privacy Policy | Website feedback

Print ISSN: 0270-7306; Online ISSN: 1098-5549