Acessibilidade / Reportar erro

Modulation of thyroid hormone receptor transactivation by the early region 1A (E1A)-like inhibitor of differentiation 1 (EID1)

Abstract

Transcriptional activation (TA) mediated by the effect of thyroid hormones on target genes requires co-activator proteins such as the early region 1A (E1A) associated 300 kDa binding protein (p300) and the cAMP response element binding protein (CREB) binding protein (CBP), known as the p300/CBP complex, which acetylate histones 3 and 4 to allow transcriptional machinery access to the target gene promoter. Little is known on the role of p300 in thyroid hormone receptor (TR) mediated TA but the E1A-like inhibitor of differentiation 1 (EID1), an inhibitor of p300 histone acetyltransferase (HAT), is a functional homolog of E1A and may inhibit myogenic differentiation factor D (MyoD) transcriptional activity and reduces muscle cell differentiation. We evaluated the influence of EID1 on TR-mediated transcriptional activity using transfection and mammalian two-hybrid studies to show that EID1 may partially reduces TA activity of the TR receptor, probably due to p300 blockage since EID1 mutants cannot reduce TR-mediated TA. The EID1 does not affect the function of p160 co-activator proteins (160 kDa proteins of steroid receptor co-activators) and is functionally independent of co-repressor proteins or TR binding. Summarizing, EID1 reduces TR-mediated transcriptional activity by blocking p300 and may play an important role in thyroid receptor activity in muscle and other tissues.

co-activator; EID1; p300; thyroid receptors; transcription


HUMAN AND MEDICAL GENETICS

RESEARCH ARTICLE

Modulation of thyroid hormone receptor transactivation by the early region 1A (E1A)-like inhibitor of differentiation 1 (EID1)

Diana Vargas; Luis Celis; Carolina Romero; Fernando Lizcano

School of Medicine, Center of Biomedical Research, La Sabana University, Chía, Colombia

Send correspondence to Send correspondence to: Fernando Lizcano School of Medicine, La Sabana University Autopista Norte de Bogotá km 21, Chía, Colombia E-mail: fernando.lizcano@unisabana.edu.co

ABSTRACT

Transcriptional activation (TA) mediated by the effect of thyroid hormones on target genes requires co-activator proteins such as the early region 1A (E1A) associated 300 kDa binding protein (p300) and the cAMP response element binding protein (CREB) binding protein (CBP), known as the p300/CBP complex, which acetylate histones 3 and 4 to allow transcriptional machinery access to the target gene promoter. Little is known on the role of p300 in thyroid hormone receptor (TR) mediated TA but the E1A-like inhibitor of differentiation 1 (EID1), an inhibitor of p300 histone acetyltransferase (HAT), is a functional homolog of E1A and may inhibit myogenic differentiation factor D (MyoD) transcriptional activity and reduces muscle cell differentiation. We evaluated the influence of EID1 on TR-mediated transcriptional activity using transfection and mammalian two-hybrid studies to show that EID1 may partially reduces TA activity of the TR receptor, probably due to p300 blockage since EID1 mutants cannot reduce TR-mediated TA. The EID1 does not affect the function of p160 co-activator proteins (160 kDa proteins of steroid receptor co-activators) and is functionally independent of co-repressor proteins or TR binding. Summarizing, EID1 reduces TR-mediated transcriptional activity by blocking p300 and may play an important role in thyroid receptor activity in muscle and other tissues.

Key words: co-activator, EID1, p300, thyroid receptors, transcription.

Introduction

The thyroid hormone receptor (TR) is a member of the nuclear hormone receptor (NHR) family of transcription factors, which control growth, development and homeostasis. There are four known TR isotypes (TRa1, TRb1, TRb2 and TRb3) which bind the thyroid hormone 3,5,3’-L-triiodothyronine (T3) (Williams, 2000). As a hormone-regulated, DNA-binding transcription factor, the TR can either enhance or repress transcription by binding to thyroid hormone response elements (TREs) in the promoters of target genes. The TRs can bind TREs as monomers, homodimers or heterodimers, and there can be variation in the primary nucleotide sequences of TREs as well as their number, spacing and orientation. In particular, TRs can bind to TREs in which half the sites are arranged as palindromes (TREpal), direct repeats (DRs) or inverted palindromes (IPs), the optimal spacing for these half-site arrangements being zero, four (DR4) and six nucleotides respectively. Most of the natural TREs described have DR4 motifs on which TRs can form heterodimers with retinoid X receptors (RXRs), which also are members of the nuclear hormone receptor superfamily (Yen, 2001).

The activating and repressive regulatory properties of TRs are regulated by crucial protein-protein contacts with basal transcription factors and transcriptional co-repressor or co-activator proteins. In the absence of thyroid hormone (TH), TRs typically bind to TREs in the promoters of regulated genes and reduce transcription by binding with the co-repressors nuclear receptor co-repressor (N-CoR) and silencing mediator for retinoid receptors and TRs (SMRT) (Lizcano et al., 2001). The binding of TH induces conformational changes in the TR that promote dissociation of TR and co-repressors and the simultaneous recruitment of several co-activator protein complexes. These co-activators induce changes in nucleosomes, which enable interaction with general transcription factors (Smith and O’Malley, 2004). Some co-activators that interact with TR have histone acetyltransferase (HAT) activity, required for the chromatin unfolding which allows access to transcriptional activator complexes, these co-activators include the 160 kDa proteins of the steroid receptor co-activator (p160), the early region 1A (E1A) associated 300 kDa binding protein (p300), the cAMP response element binding protein (CREB) binding protein (CREBBP, or CBP) and the p300/CBP complex associated factor (p/CAF) (Glass and Rosenfeld, 2000; McKenna and O’Malley, 2002). Although the p300/CBP complex may bind directly to TRs, it can be recruited to the promoters through p160 co-activators (Sharma and Fondell, 2002). Furthermore, p300/CBP can bind to switching/sucrose nonfermenting (SWI/SNF) ATP-dependent chromatin remodeling complexes (Becker and Horz, 2002) and the thyroid hormone receptor-associated protein (TRAP) complex (Ito and Roeder, 2002), acting as a bridge between general transcription factors and TR (Huang et al., 2003). This suggests that besides having potent HAT activity, essential for hormone-dependent activation by TR, p300 can also act as an integrator between TR and the general transcriptional machinery (Ogryzko et al., 1996; Korzus et al., 1998).

The E1A-like inhibitor of differentiation 1 (EID1, also known as p/CAF inhibitor 1 or CRI-1) shares functional homology with E1A (Miyake et al., 2000) and is an inhibitor of p300 (De Luca et al., 2003) that has been shown to reduce the activity of other transcription factors (Bavner et al., 2005) and has been described as a functional homolog of the adenovirus E1A protein and may decrease muscle cell differentiation by inhibiting the activation of myogenic differentiation factor D (MyoD) by blocking p300 (MacLellan et al., 2000). Interestingly, while E1A reduces retinoic acid receptor (RAR) transactivation (Chakvarti et al., 1999; Hamamori et al., 1999; Meng et al., 2005), it has also shown activity as a potent TR co-activator in mammalian and yeast cells (Wahlstrom et al., 1999; Meng et al., 2003), probably as a result of direct interaction with TRs.

To investigate the role of p300 in TR activation we evaluated a possible role of the EID1 protein as a functional co-activator of ligand-bound TR transcriptional activity. Our results showed that EID1 reduced TR transactivation, even though it did not bind to TR in mammalian two-hybrid studies. This reduction was probably secondary to blocked p300 activity, because EID1 mutants that could not bind to p300 did not decrease TR-mediated transcriptional activation. We also found that EID1 did not affect the function of p160 co-activator proteins and was functionally independent of binding to co-repressor proteins.

Materials and Methods

Plasmids

Expression vector plasmids containing rat thyroid receptor b1 (TRb1) and b2 (TRb2) protein DNA sequences were generated by inserting the Eco RI fragment of rat TRb1 and TRb2 into the expression vector plasmid pcDNAI/AMP (Invitrogen, USA) as previously described (Koibuchi et al., 1999). An in vitro mutagenesis kit (Promega, Madison, WI) was employed to create the TRb1 mutant (TRAHTm) using a mutagenesis primer that altered codons 223 from A to G, 224 from H to G and 227 from T to A and inserted this into pcDNAI/Amp (Lizcano et al., 2001). These changes in the hinge region of TRb1 reduce its interaction with the co-repressor complex. Oligonucleotides containing the 5xUAS and DR4 reporter fragments were cloned into the BamHI and HindIII restriction sites of the pT109 vector, which contains the herpes simplex thymidine kinase (TK) minimum promoter coupled to a luciferase (LUC) coding sequence (Hodin et al., 1989; Koibuchi et al., 1999). The pcDNA3-EID1 and pcDNA3-EID1 mutant plasmids (pcDNA-DEID1) have been described previously (Miyake et al., 2000), pcDNA-DEID1 (157-D53D92) lacking the acidic regions created by residues 53-63 and 92-115 that have been reported to be necessary for p300 binding. This mutant also has a deletion of residues 158-187 in the C-terminal region, which contains a retinoblastoma protein (pRB) binding motif (LXCXE, where X equals any amino acid residue) (Miyake et al., 2000). The cMVb-NHAp300 plasmid (Eckner et al., 1996) was a kind donation from the Livingstong’s Lababoratory (Dana Farber Institute, Boston, USA) The following plasmids were kindly donated by A. Takeshita (Gunma, Japan): the steroid receptor-1, also referred to as nuclear receptor co-activator 1/NCoA, pcDNA-SRC-1 plasmid (Onate et al., 1995; Kamei et al., 1996); the thyroid hormone receptor activator molecule, also referred to as amplified in breast cancer 1 (AIB1), 1pBK-CMV-TRAM1 plasmid; the activator of transcription of nuclear receptors (ACTR) plasmid; (Takeshita et al., 1997; Anzick et al., 1997; Chen et al., 1997; The thyroid receptor binding protein (also referred as peroxisome proliferator receptor interacting protein (PRIP or PPPAR), receptor associated protein (RAP250/250-kDa), nuclear receptor co-regulator (NRC) or activating signal co-integrator-2 (ASC-2) plasmid pcDNA3-TRBP has been described previously (Ko et al., 2000; Zhu et al., 2000; Caira et., 2000; Mahajan and Samuels, 2000; Lee et al., 2000.

Transient transfection

The U2OS and CV-1 cell lines were obtained from American Tissue Culture Collection (ATCC) and grown under a 5% (v/v) CO2 atmosphere in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% (v/v) fetal bovine serum (FBS), 100 units mL-1 penicillin and 100 µg mL-1 streptomycin. The serum was stripped of thyroid hormones by constant mixing with 5% (w/v) AGI-X8 resin (Mallinckrodt, Raleigh, USA) and subjected to ultra-filtration using a 0.22 µm membrane (Nalgene, Rochester, USA). The cells were seeded at 103 cells per well of a twelve multiwell cell culture plates (Falcon, San Jose, USA) 48 h prior to transfection. When cells reached 70% confluence, they were transiently transfected using a calcium-phosphate method (Sambrook et al., 1989). Reporter plasmids (0.85 µg/well) containing DR4 or a constitutive active thymidine kinase-luciferase (TK-Luc) reporter containing five GAL4-binding sites (5x UAS-TK-Luc) were co-transfected with 0.1 µg per well of the Gal4-TRb1-LBD, VP16-EID-1, Gal4-NCoR, VP16-TRb1-LBD, EID1, DEID1 (1-157D53D92) plasmids and 0.1 µg per well of a co-activator protein (p300, SRC-1, TRAM1 or TRBP). The total amount of transfected DNA in each well was balanced by adding the pcDNA3.1 plasmid. For both cells types, control wells were transfected with the appropriated amount of empty expression vector. Cells were grown in the presence or absence of 1 x 10-6 M T3, and after 24 h cells were harvested in triton X-100 buffer (USB, Cleveland,USA). Luciferase activity, introduced into the cells using the 5xUAS and DR4 plasmids, was measured with the luminometer (Lumat 9507, Berthold technologies, Bad Wildbad, Germany), beta-galactosidase (b-gal) activity, introduced using the CMV-b-gal plasmid, was measured photospectrometrically at 410 nm (Variant, USA). The fold-change in luciferase or b-gal expression in the cells containing the experimental vectors was calculated relative to expression in the presence of only the control expression vector pcDNA3.1. The data was expressed as the mean ± the standard error of the mean (SEM) and represent a minimum of three independent experiments, with each data point run in triplicate for each experiment.

Mammalian two-hybrid system

The Gal4-blank and Gal4-N-CoR (aa1552-2453) plasmids have been previously described (Miyazaki et al., 2004), while the VP16-EID1 plasmid was generated by recovering EID1 from the pcDNA 3.1 plasmid by double digestion with Eco RI and Hind III and subcloning into the VP-16 plasmid which was then digested with the same restriction enzymes. The Gal4-TRb1-LBD plasmid (Miyazaki et al., 2004) was obtained from A. N. Hollenberg (Brigham and Women’s Hospital, Boston, USA) and the Gal4-pRB and Gal4-p300 (Miyake et al., 2000) plasmids were obtained from W. Kaelin Jr. (Dana Farber Institute, Boston, USA). The VP16-TRb-1-ligand binding domain (LBD) was constructed by inserting a PCR-generated in-frame fragment downstream of the VP-16 activation domain in the AASV-VP16 plasmid (Miyazaki et al., 2004). We transiently transfected U2OS cell in twelve-well plates, in duplicate, with 500 ng of CMV-b-Gal plasmid, 1 µg of 5x UAS-TK-Luc reporter plasmid and 1 µg each of the Gal4-Blank, Gal4-pRB, Gal4-TRb1-LBD, Gal4-NCoR and Gal4-p300 plasmids and 1 µg or 2 µg each of the VP16-blank, VP16-EID1 and VP16-TRb1 plasmids. The luciferase and b-Gal assays were carried out as described above.

Results and Discussion

We first performed transient transfection studies using the natural (DR-4) reporter plasmid and detected a ligand-independent reduction in the transcriptional activation of the TRb1 plasmid. After addition of the T3, there was a significant increase in TRb1-mediated transcription. Co-transfection of EID1 did not alter the ligand-independent repression, but we observed a 50% reduction in T3-induced TRb1 transcriptional transactivation (Figure 1A). In order to evaluate whether a co-repressor complex might be responsible for the reduction in transcriptional activation caused by EID1, transfection studies were performed using TRb2 and TRAHT mutants, which are thyroid receptors with a lower binding capacity for the co-repressor complex. We found that EID1 was able to significantly reduce the T3-mediated activation of both TRb2 and TRAHT receptors (Figure 1B), indicating that perhaps the co-repressor complex was not necessary for the EID1-mediated reduction in thyroid receptor-mediated transcription.


To investigate whether the reduction in TRb1-mediated transcription caused by EID1 was a consequence of p300 inhibition, CV-1 and U2OS cells were co-transfected with a plasmid containing the Gal4 DNA-binding domain fused to the TRb1-ligand binding domain (TRb1-LBD) and a plasmid with the EID1 mutant (DEID1-D1-157D53D92), but which lacked the p300 binding sites, together with a reporter plasmid that contained multiple Gal4 biding sites fused to TK-Luc (UAS-TK-Luc). The Gal4-TRb1-LBD-mediated transcriptional activation following administration of T3 was not modifying when the DEID1 mutant was co-transfected, indicating that the DEID1 mutant lacked the necessary sites to cause the reduction in TRb1-mediated transcription which was observed with EID1 (Figure 2A).


The reduction in transcriptional activation mediated by EID1 was not accompanied by a significant change in the characteristic ligand-independent repression mediated by TRb1 and was, instead, possibly due to a reduction in p300 activity. This prompted us to study the influence of EID1 on other co-activator proteins with HAT activities, which are recruited to the promoters of genes influenced by TR. It is known that mutual interactions among these co-activator proteins produces complete co-activation due to interactions with general transcription factors and enzymatic changes in the nucleosomes (Ito and Roeder, 2002). Therefore, in order to understand the role of EID1 on co-activators that collaborate with ligand-bound TR, we co-transfected EID1 with plasmid expression vectors containing SRC-1 and TRAM1. In both cases, we observed that the co-transfection of EID1 with p160 proteins did not significantly reduce the strong activation mediated by these co-activators. However, EID1 significantly reduced the remarkable activation mediated by p300 and TRBP co-activator proteins (Figure 2B).

Considering that the EID1-mediated reduction in TRb1 activation might be a consequence of a direct interaction with the thyroid receptor that obstructs key sites for co-activator interaction, we evaluated a possible interaction using mammalian two-hybrid studies. There was a significant increase in Luciferase activity when Gal4-N-CoR was co-transfected with VP16-TRb1-LBD in the absence of T3 and also when Gal4-p300 and Gal-4-pRB were co-transfected with VP16-EID1 (Figure 3) but there was not increase in luciferase activity when Gal4-TR TRb1-LBD was co-transfected with VP16-EID1, indicating that these proteins probably do not have a direct interaction.


In this study, we have established the effect of EID1 protein on TRb1-mediated transcriptional activation. Our results show that EID1 reduces T3 activation of thyroid hormone receptors without affecting TRb1-mediated ligand-independent transcriptional repression. An important question in transcriptional regulation by transcription factors, which are regulated by a ligand, is how they can either activate or silence the promoters of target genes. Binding of T3 results in a conformational change in the TR (Glass and Rosenfeld, 2000), which facilitates the dissociation of TR/co-repressor complexes such as N-CoR and SMRT and the recruitment of co-activating proteins with intrinsic HAT activity such as p160 co-activators and p300/CBP. The most probable effect of EID1 on TRb1 is a reduction in the activity of co-activators, since we did not observe any effect of EID1 in the absence of T3. This is consistent with our results and those of previous studies (Bavner et al., 2005). To determine if the EID1-mediated reduction in TRb1 activation might be due to inhibition caused by the co-repressor complex, we performed studies with thyroid receptors that could not interact with the co-repressor complex. The TRb2 protein is a thyroid hormone receptor isoform derived from the same gene as TRb1, but with an alternative splicing pattern that produces additional N-terminal amino acids (Yen, 2001). These receptors are expressed primarily in the hypothalamus and pituitary gland, and the additional amino terminus of TRb2 may block the recruitment of components of the co-repressor complex (Yang et al., 1999). The TRb1 mutant (TRAHT) contains three amino acid substitutions in the hinge region of TRb1 and has a decreased affinity for the co-repressor complex (Lizcano et al., 2001). However, EID1 still reduced ligand-dependent TR activity when co-transfection studies were performed with TRb2 and TRAHT receptors (Fig. 1B). These results preclude the possibility of a dual inhibitory mechanism, which would include the recruitment of co-repressor complex and might explain the effect of EID1 on TRs.

The results of transfection studies with EID1 showed only a partial reduction in TRb1 activation, which was probably a consequence of interference with a specific co-activator. This effect was not seen after co-transfection with the DEID1 mutant, which cannot interact with p300, suggesting that EID1 might block the activity of this co-activator. The p300 protein has been characterized as a binding partner of E1A viral protein, and subsequently, as a histone acetylase (Chan and La Thangue, 2001). Functionally, p300 may also act as a bridge between the TR transcriptional complex and general transcription factors (Vo and Goodman, 2001). The EID1 protein has been recognized as a functional homolog of the viral protein E1A, with both proteins having the capacity to block p300 co-activation activity (Chakvarti et al., 1999). However, while E1A is a potent co-activator of TR-mediated transcription (Wahlstrom et al., 1999), we observed that EID1 acted as an inhibitor of T3-dependent transactivation. The co-activation mediated by E1A requires a direct interaction with the nuclear receptor but in our mammalian two-hybrid studies we did not observe an interaction between EID1 and TRb1 (Figure 3).

Our data shows that EID1 reduces the activity of p300 and TRBP but has practically no effect on the function of p160 co-activator proteins (Figure 2B), which can bind directly to TR and have HAT activity. Given that the TRBP co-activator shows no HAT activity, its activation requires the presence of p300 as an integrator protein (Mahajan and Samuels, 2005). Therefore, we suggest that EID1 not only acts as an inhibitor of HAT activity but also blocks the presence of p300 in the activation complex. Still, it is possible that histone acetylation is necessary for TRBP-mediated contributions to transcription (Huang et al., 2003). Some studies have shown that EID1 may bind to the small heterodimer partner (SHP), an atypical orphan nuclear receptor that lacks a DNA-binding domain and is responsible for reducing the activation of some nuclear receptors (Bavner et al., 2002; Park et al., 2004). The SHP protein binds to the C-terminal regions that harbor activation function 2 (AF2) in ligand-bound TRs and blocks the recruitment of co-activator proteins in the presence of ligand. Because we observed that the reduction in TR function occurred mainly through inhibition of CBP/p300 activity, without a major effect on p160 co-activator proteins, SHP probably did not play a major role in EID1-mediated inhibition of ligand-bound TR transactivation activity.

In conclusion, EID1 may reduce the activity of ligand-bound thyroid hormone receptors through inhibition of p300 activity. It is possible that EID1 block p300 co-activators from binding to the promoter regions of target genes and may play an important role in thyroid receptor activity in muscle an other tissues. EID1 might have and important role in the differentiation of tissues associate with energy expenditure and a target for overweight and obesity drugs.

Acknowledgments

We are grateful to Dr. William Kaelin Jr. for the EID1 and DEID1 plasmids, Dr. Akira Takeshita for SRC-1 and TRAM1 and Dr. William W. Chin for TRBP. This work was made possible by support from Fondo Patrimonial Especial 2003 La Sabana University, Chía, Colombia.

Received: April 27, 2007; Accepted: September 18, 2007.

Associate Editor: Carlos F.M. Menck

  • Anzick SL, Kononen J, Walker RL, Azorsa DO, Tanner MM, Guan XY, Sauter G, Kallionimei OP, Trent JM and Meltzer PS (1997) AIB1, asteroid receptor coactivator amplified in breast and ovarian cancer. Science 277:965-968.
  • Bavner A, Johansson L, Toresson G, Gustafsson JA and Treuter E (2002) A transcriptional inhibitor targeted by the atypical orphan nuclear receptor SHP. EMBO Rep 3:478-484.
  • Bavner A, Matthews J, Sanyal S, Gustafsson J and Treuter E (2005) EID3 is a novel EID family member and an inhibitor of CBP-dependent co-activation. Nucleic Acids Res 33:3561-3569.
  • Becker PB and Horz W (2002) ATP-dependent nucleosome remodeling. Annu Rev Biochem 71:247-273.
  • Caira F, Antonson P, Pelto-Huikko M, Treuter E and Gustafsson JA (2000) Cloning and characterization of RAP250, a novel nuclear receptor coactivator. J Biol Chem 275:5308-5317.
  • Chan HM and La Thangue NB (2001) p300/CBP proteins: HATs for transcriptional bridges and scaffolds. J Cell Sci 114:2363-2373.
  • Chakvarti D, Orgyzko V, Kao H-Y, Nash A, Chen H, Nakatani Y and Evans R (1999) A viral mechanism for inhibition of p300 and PCAF acetyltransferase activity. Cell 96:393-403.
  • Chen H, Li RJ, Chiltz RL, Chakeavarti D, Nash A, Nagy L, Privalsky NL, Nakatani Y and Evans RM (1997) Nuclear receptor co-activator ACTR is a novel histone acetyltransferease and forms multimeric activation complex with P/CAF and CBP6p300. Cell 90:569-580.
  • De Luca A, Severino A, De Paolis P, Cottone G, De Luca L, De Falco M, Porcellini A, Volpe M and Condorelli G (2003) p300/cAMP-response-element-binding-protein('CREB)-binding protein (CBP) modulates co-operation between myocyte enhancer factor 2A (MEF2A) and thyroid hormone receptor-retinoid X receptor. Biochem J 369:477-484.
  • Eckner R, Ewen M, Newsome D, Gerdes M, DeCaprio JA, Lawrence JB and Livingston DM (1996) Molecular cloning and functional analysis of the adenovirus E1A- Associated 300-kD protein (p300) reveals a protein with properties of a transcriptional adaptor. Genes Dev 8:869-884.
  • Glass CK and Rosenfeld MG (2000) The coregulator exchange in transcriptional function of nuclear receptors. Genes Dev 14:121-141.
  • Hamamori Y, Sartorelli V, Orgyzko V, Puri PL, Wu H-Y, Wang JYJ, Nakatani Y and Kedes L (1999) Regulation of histone acetyltransferase p300 and PCAF by the bHLH protein twist and adenoviral E1A. Cell 96:405-413.
  • Hodin R, Lazar MA, Wintman B, Darling DS, Koening RJ, Larsen PR, Moore DD and Chin WW (1989) Identification of a thyroid hormone receptor that is pituitary-specific. Science 244:76-79.
  • Huang Z-Q, Li J, Sachs L, Cole P and Wong J (2003) A role for cofactor-cofactor and cofactor-histone interactions in targeting p300, SWI/SNF and Mediator for transcription. EMBO J 22:2146-2155.
  • Ito M and Roeder RG (2002) The TRAP/SMCC/Mediator complex and thyroid hormone receptor function. Trends Endocrinol Metab 12:127-134.
  • Kamei Y, Xu L, Heinzel T, Torchia J, Kurokawa R, Gloss B, Lin S-C, Heyman RA, Rose DW, Glass CK et al. (1996) A CBP integrator complex mediates transcriptional activation and AP-1inhibition by nuclear receptors. Cell 85:403-414.
  • Ko L, Cardona G and Chin W (2000) Thyroid hormone receptor-binding protein, an LXXLL motif-containing protein, functions as a general coactivator. Proc Natl Acad Sci USA 97:6212-6217.
  • Koibuchi N, Liu Y, Fukuda H, Takeshita A, Yen PM and Chin WW (1999) RORa augments thyroid hormone receptor-mediated transcriptional activation. Endocrinology 140:1356-1364
  • Korzus E, Torchia J, Rose D, Xu L, Kurokawa R, Mcinerney EM, Mullen TM, Glass CK and Rosenfeld MG (1998) Transcription factor-specific requirements for coactivators and their acetyltransferase functions. Science 279:703-707.
  • Lee SK, Anzick SL, Choi JE, Bubendorf L, Guan XY, Jung YK, Kalioniemi OP, Kononen L, Trent JM, Azorsa D et al. (1999) A nuclear factor, ASC-2, is a cancer-amplified transcriptional coactivator essential for ligand-dependent transactivation by nuclear receptors in vivo J Biol Chem 274:34283-34293.
  • Lizcano F, Koibuchi N, Fukuda H, Dangond F and Chin WW (2001) Cell type-specific roles of histone deacetylase in TR ligand-independent transcriptional repression. Mol Cell Endocrinol 172:13-20.
  • Mahajan MA and Samuels HH (2000) A new family of nuclear receptor coregulators that integrate nuclear receptor sinaling through CREB-binding protein. Mol Cell Biol 20:5048-5063.
  • Mahajan MA and Samuels HH (2005) Nuclear hormone receptor coregulator: Role in hormone action, metabolism, growth, and development. Endocrine Rev 26:583-597.
  • McKenna NJ and OMalley BW (2002) Minireview: Nuclear receptor coactivators An update. Endocrinology 143:2461-2465.
  • MacLellan WR, Xiao G, Abdellatif M and Schneider MD (2000) A novel Rb- and p300-binding protein inhibits transactivation by MyoD. Mol Cell Biol 20:8903-8915.
  • Meng X, Yang Y-F, Cao X, Govidan MJ, Shuen M, Hollenberg AN, Mymrk JS and Walfish PG (2003) Cellular context of co-regulator and adaptor proteins regulates human adenovirus 5 E1A dependent gene activation by the thyroid hormone receptor. Mol Endocrinol 17:1095-1105.
  • Meng X, Webb P, Yang YF, Shuen M, Yousef AF, Baxter JD, Mymryk JS and Walfish PG (2005) E1A and a nuclear receptor corepressor splice variant (N-CoRI) are thyroid hormone receptor coactivators that bind in the corepressor mode. Proc Natl Acad Sci USA 102:6267-6272.
  • Miyake S, Sellers WR, Safran M, Li X, Zhao W, Grossman SR, Gan J, DeCaprio JA, Adams PD and Kaelin WG (2000) Cells degrade a novel inhibitor of differentiation with E1A-like properties upon exiting the cell cycle. Mol Cell Biol 20:8889-8902.
  • Miyazaki W, Iwasaki T, Takeshita A, Kuroda Y and Koibuchi N (2004) Polychlorinated biphenyls suppress thyroid hormone receptor-mediated transcription through a novel mechanism. J Biol Chem 279:18195-18202.
  • Onate SA, Tsai SY, Tsai M-J and OMalley BW (1995) Sequence and characterization of a coactivator of the steroid hormone receptor superfamily. Science 270:1354-1357.
  • Ogryzko V, Shiltz R, Russanova V, Howard B and Nakatani Y (1996) The trasncriptional coactivators p300 and CBP are histone acetyl-transferases. Cell 87:953-959.
  • Park YY, Kim H-J, Kim J-Y, Kim M-Y, Song K-H, Park KC, Yu K-Y, Shong M, Kim K-H and Choi H-J (2004) Differential role of the loop region between helices H6 and H7 within the orphan nuclear receptors small heterodimer partner and DAX-1. Mol Endocrinol 18:1082-1095.
  • Sambrook J, Fritsch E and Maniatis T (1989). Molecular Cloning: A Laboratory Manual. Cold Spring Harbor, NY, pp 1632-1636.
  • Sharma D and Fondell JD (2002) Ordered recruitment of histone acetyltransferases and the TRAP/Mediator complex to thyroid hormone-responsive promoters in vivo Proc Natl Acad. Sci USA 99:7934-7939.
  • Smith CL and OMalley BW (2004) Coregulator function: A key to understanding tissue specificity of selective receptor modulators. Endocrine Rev 25:45-71.
  • Takeshita A, Cardona G, Koibuchi N, Suen C and Chin WW (1997) TRAM-1, a novel 160-kDa thyroid hormone receptor activator molecule, exhibits distinct properties from steroid receptor coactivator-1. J Biol Chem 272:27629-27634.
  • Yang Z, Hong SH and Privalsky ML (1999) Transcriptional anti-repression. Thyroid hormone with subsequent assembly of a functional corepressor-complex. J Biol Chem 274:37131-37138.
  • Yen PM (2001) Physiological and molecular basis of thyroid hormone action. Physiol Rev 81:1097-1142.
  • Vo N and Goodman RH (2001) CREB-binding protein and p300 in transcriptional regulation. J Biol Chem 276:13505-13508.
  • Wahlstrom G, Vennstrom B and Bolin M (1999) The adenovirus E1A protein is a potent coactivator for thyroid hormone receptors. Mol Endocrinol13:1119-1129.
  • Williams GR (2000) Cloning and characterization of two novel thyroid hormone receptor beta isoforms. Mol Cell Biol 20:8329-8342.
  • Zhu Y, Kan L, Qi C, Kanwar YS, Yeldandi AV, Rao MS and Reddy JK 2000 Isolation and characteriztion of Peroxisome proliferator-activated receptor (PPAR) interacting protein (PRIP) as a coactivator for PPAR. J Biol Chem 275:13510-13516.
  • Send correspondence to:

    Fernando Lizcano
    School of Medicine, La Sabana University
    Autopista Norte de Bogotá km 21, Chía, Colombia
    E-mail:
  • Publication Dates

    • Publication in this collection
      24 June 2008
    • Date of issue
      2008

    History

    • Accepted
      18 Sept 2007
    • Received
      27 Apr 2007
    Sociedade Brasileira de Genética Rua Cap. Adelmio Norberto da Silva, 736, 14025-670 Ribeirão Preto SP Brazil, Tel.: (55 16) 3911-4130 / Fax.: (55 16) 3621-3552 - Ribeirão Preto - SP - Brazil
    E-mail: editor@gmb.org.br