227
Views
0
CrossRef citations to date
0
Altmetric
Review

Transcription Factor Interactions in Genomic Nuclear Receptor Function

, &
Pages 471-485 | Published online: 18 Aug 2011

Bibliography

  • Lee W , HaslingerA, KarinM, TjianR. Activation of transcription by two factors that bind promoter and enhancer sequences of the human metallothionein gene and SV40. Nature325(6102) , 368–372 (1987).
  • Renkawitz R , SchützG, von der Ahe D, Beato M. Sequences in the promoter region of the chicken lysozyme gene required for steroid regulation and receptor binding. Cell37(2) , 503–510 (1984).
  • Schüle R , MullerM, KaltschmidtC, RenkawitzR. Many transcription factors interact synergistically with steroid-receptors. Science242(4884) , 1418–1420 (1988).
  • Carroll JS , MeyerCA, SongJet al. Genome-wide analysis of estrogen receptor binding sites. Nat. Genet. 38(11) , 1289–1297 (2006).
  • Krum SA , Miranda-CarboniGA, LupienM, EeckhouteJ, CarrollJS, BrownM. Unique ERα cistromes control cell type-specific gene regulation. Mol. Endocrinol.22(11) , 2393–2406 (2008).
  • Lefterova MI , StegerDJ, ZhuoDet al. Cell-specific determinants of peroxisome proliferator-activated receptor γ function in adipocytes and macrophages. Mol. Cell. Biol. 30(9) , 2078–2089 (2010).
  • Ravasi T , SuzukiH, CannistraciCVet al. An atlas of combinatorial transcriptional regulation in mouse and man. Cell 140(5) , 744–752 (2010).
  • Archer TK , CordingleyMG, WolfordRG, HagerGL. Transcription factor access is mediated by accurately positioned nucleosomes on the mouse mammary tumor virus promoter. Mol. Cell. Biol.11(2) , 688–698 (1991).
  • John S , SaboPJ, JohnsonTAet al. Interaction of the glucocorticoid receptor with the chromatin landscape. Mol. Cell 29(5) , 611–624 (2008).
  • Fletcher T , XiaoN, MautinoGet al. ATP-dependent mobilization of the glucocorticoid receptor during chromatin remodeling. Mol. Cell. Biol. 22(10) , 3255–3263 (2002).
  • Richard-Foy H , HagerGL. Sequence-specific positioning of nucleosomes over the steroid-inducible MMTV promoter. EMBO J.6(8) , 2321–2328 (1987).
  • Green CD , HanJJ. Epigenetic regulation by nuclear receptors. Epigenomics3(1) , 59–72 (2011).
  • Klokk TI , KurysP, ElbiCet al. Ligand-specific dynamics of the androgen receptor at its response element in living cells. Mol. Cell. Biol. 27(5) , 1823–1843 (2007).
  • Nagaich AK , WalkerD, WolfordR, HagerGL. Rapid periodic binding and displacement of the glucocorticoid receptor during chromatin remodeling. Mol. Cell14(2) , 163–174 (2004).
  • Zaret K , YamamotoKR. Reversible and persistent changes in chromatin structure accompany activation of a gluocorticoid-dependent enhancer element. Cell38(1) , 29–38 (1984).
  • McNally JG , MüllerWG, WalkerD, WolfordR, HagerGL. The glucocorticoid receptor: rapid exchange with regulatory sites in living cells. Science287(5456) , 1262–1265 (2000).
  • Fletcher T , RyuB, BaumannCet al. Structure and dynamic properties of a glucocorticoid receptor-induced chromatin transition. Mol. Cell. Biol. 20(17) , 6466–6475 (2000).
  • Östlund Farrants AK , BlomquistP, KwonH, WrangeO. Glucocorticoid receptor-glucocorticoid response element binding stimulates nucleosome disruption by the SWI/SNF complex. Mol. Cell. Biol.17(2) , 895–905 (1997).
  • Brüggemeier U , RoggeL, WinnackerEL, BeatoM. Nuclear factor I acts as a transcription factor on the MMTV promoter but competes with steroid hormone receptors for DNA binding. EMBO J.9(7) , 2233–2239 (1990).
  • Cordingley MG , RiegelAT, HagerGL. Steroid-dependent interaction of transcription factors with the inducible promoter of mouse mammary tumor virus in vivo. Cell48(2) , 261–270 (1987).
  • Stenoien DL , PatelK, ManciniMet al. FRAP reveals that mobility of oestrogen receptor-α is ligand- and proteasome-dependent. Nat. Cell Biol. 3(1) , 15–23 (2001).
  • Métivier R , PenotG, HübnerMRet al. Estrogen receptor-α directs ordered, cyclical, and combinatorial recruitment of cofactors on a natural target promoter. Cell 115(6) , 751–763 (2003).
  • Conway-Campbell BL , GeorgeCL, PooleyJRet al. The HSP90 molecular chaperone cycle regulates cyclical transcriptional dynamics of the glucocorticoid receptor and its co-regulatory molecules CBP/P300 during ultradian ligand treatment. Mol. Endocrinol. 26(6) , 944–954 (2011).
  • Kangaspeska S , StrideB, MétivierRet al. Transient cyclical methylation of promoter DNA. Nature 452(7183) , 112–115 (2008).
  • Métivier R , GallaisR, TiffocheCet al. Cyclical DNA methylation of a transcriptionally active promoter. Nature 452(7183) , 45–50 (2008).
  • Biddie SC , JohnS, HagerGL. Genome-wide mechanisms of nuclear receptor action. Trends in endocrinology and metabolism. Trends Endocrinol. Metabol.21(1) , 3–9 (2010).
  • Delacroix L , MoutierE, AltobelliGet al. Cell-specific interaction of retinoic acid receptors with target genes in mouse embryonic fibroblasts and embryonic stem cells. Mol. Cell. Biol. 30(1) , 231–244 (2010).
  • John S , SaboPJ, ThurmanREet al. Chromatin accessibility pre-determines glucocorticoid receptor binding patterns. Nat. Genet. 43(3) , 264–268 (2011).
  • Mikkelsen TS , XuZ, ZhangXet al. Comparative epigenomic analysis of murine and human adipogenesis. Cell 143 , 156–169 (2010).
  • Welboren W -J, van Driel MA, Janssen-Megens EM et al. ChIP-seq of ERα and RNA polymerase II defines genes differentially responding to ligands. EMBO J.28(10) , 1418–1428 (2009).
  • Rigaud G , RouxJ, PictetR, GrangeT. In vivo footprinting of rat tat gene: dynamic interplay between the glucocorticoid receptor and a liver-specific factor. Cell67(5) , 977–986 (1991).
  • Lefterova MI , ZhangY, StegerDJet al. PPARγ and C/EBP factors orchestrate adipocyte biology via adjacent binding on a genome-wide scale. Genes Dev. 22(21) , 2941–2952 (2008).
  • Lupien M , EeckhouteJ, MeyerCAet al. FoxA1 translates epigenetic signatures into enhancer-driven lineage-specific transcription. Cell 132(6) , 958–970 (2008).
  • Heinz S , BennerC, SpannNet al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B-cell identities. Mol. Cell 38(4) , 576–589 (2010).
  • Boyle AP , DavisS, ShulhaHPet al. High-resolution mapping and characterization of open chromatin across the genome. Cell 132(2) , 311–322 (2008).
  • He HH , MeyerCA, ShinHet al. Nucleosome dynamics define transcriptional enhancers. Nat. Genet. 42 , 343–347 (2010).
  • Jin C , FelsenfeldG. Nucleosome stability mediated by histone variants H3.3 and H2A.Z. Genes Dev.21(12) , 1519–1529 (2007).
  • Jin C , ZangC, WeiGet al. H3.3/H2A.Z double variant-containing nucleosomes mark nucleosome-free regions of active promoters and other regulatory regions. Nat. Genet. 41(8) , 941–945 (2009).
  • Carroll JS , LiuXS, BrodskyASet al. Chromosome-wide mapping of estrogen receptor binding reveals long-range regulation requiring the forkhead protein FoxA1. Cell 122(1) , 33–43 (2005).
  • Kininis M , ChenBS, DiehlAGet al. Genomic analyses of transcription factor binding, histone acetylation, and gene expression reveal mechanistically distinct classes of estrogen-regulated promoters. Mol. Cell. Biol. 27(14) , 5090–5104 (2007).
  • So AY -L, Chaivorapol C, Bolton EC, Li H, Yamamoto KR. Determinants of cell- and gene-specific transcriptional regulation by the glucocorticoid receptor. PLoS Genet.3(6) , e94 (2007).
  • Stender JD , KimK, CharnTHet al. Genome-wide analysis of estrogen receptor α DNA binding and tethering mechanisms identifies Runx1 as a novel tethering factor in receptor-mediated transcriptional activation. Mol. Cell. Biol. 30(16) , 3943–3955 (2010).
  • Wang Q , LiW, LiuXSet al. A hierarchical network of transcription factors governs androgen receptor-dependent prostate cancer growth. Mol. Cell 27(3) , 380–392 (2007).
  • Reddy TE , PauliF, SprouseROet al. Genomic determination of the glucocorticoid response reveals unexpected mechanisms of gene regulation. Genome Res. 19(12) , 2163–2171 (2009).
  • Hakim O , JohnS, LingJQ, BiddieSC, HoffmanAR, HagerGL. Glucocorticoid receptor activation of the Ciz1-Lcn2 locus by long range interactions. J. Biol. Chem.284(10) , 6048–6052 (2009).
  • Tolhuis B , PalstraR, SplinterE, GrosveldF, de Laat W. Looping and interaction between hypersensitive sites in the active β-globin locus. Mol. Cell10(6) , 1453–1465 (2002).
  • Hakim O , SungM, VossTCet al. Diverse gene reprogramming events occur in the same spatial clusters of distal regulatory elements. Genome Res. 21(5) , 697–706 (2011).
  • Fullwood MJ , LiuMH, PanYFet al. An oestrogen-receptor-α-bound human chromatin interactome. Nature 462(7269) , 58–64 (2009).
  • Schoenfelder S , ClayI, FraserP. The transcriptional interactome: gene expression in 3D. Curr. Opin. Genet. Develop.20 , 127–133 (2010).
  • Kim K , DoiA, WenBet al. Epigenetic memory in induced pluripotent stem cells. Nature 467(7313) , 285–290 (2010).
  • Lieberman-Aiden E , Van Berkum NL, Williams L et al. Comprehensive mapping of long-range interactions reveals folding principals of the human genome. Science326(5950) , 289–293 (2010).
  • Lupien M , MeyerCA, BaileySTet al. Growth factor stimulation induces a distinct ER(α) cistrome underlying breast cancer endocrine resistance. Genes Dev. 24(19) , 2219–2227 (2010).
  • Hurtado A , HolmesKA, Ross-InnesCS, SchmidtD, CarrollJS. FOXA1 is a key determinant of estrogen receptor function and endocrine response. Nat. Genet.43(1) , 27–33 (2011).
  • Cirillo LA , LinFR, CuestaI, FriedmanD, JarnikM, ZaretKS. Opening of compacted chromatin by early developmental transcription factors HNF3 (FoxA) and GATA-4. Mol. Cell9(2) , 279–289 (2002).
  • Eeckhoute J , LupienM, MeyerCAet al. Cell-type selective chromatin remodeling defines the active subset of FOXA1-bound enhancers. Genome Res. 19(3) , 372–380 (2009).
  • Polo JM , LiuS, FigueroaMEet al. Cell type of origin influences the molecular and functional properties of mouse induced pluripotent stem cells. Nat. Biotechnol. 28(8) , 848–855 (2010).
  • Hua S , KittlerR, WhiteKP. Genomic antagonism between retinoic acid and estrogen signaling in breast cancer. Cell137(7) , 1259–1271 (2009).
  • Ross-Innes CS , StarkR, HolmesKAet al. Cooperative interaction between retinoic acid receptor-α and estrogen receptor in breast cancer. Genes Dev. 24(2) , 171–182 (2010).
  • Jonat C , RahmsdorfHJ, ParkKKet al. Antitumor promotion and antiinflammation: down-modulation of AP-1 (Fos/Jun) activity by glucocorticoid hormone. Cell 62(6) , 1189–1204 (1990).
  • Schüle R , RangarajanP, KliewerSet al. Functional antagonism between oncoprotein c-Jun and the glucocorticoid receptor. Cell 62(6) , 1217–1226 (1990).
  • Gaub MP , BellardM, ScheuerI, ChambonP, Sassone-CorsiP. Activation of the ovalbumin gene by the estrogen receptor involves the fos-jun complex. Cell63(6) , 1267–1276 (1990).
  • Nielsen R , PedersenTA, HagenbeekDet al. Genome-wide profiling of PPARγ:RXR and RNA polymerase II occupancy reveals temporal activation of distinct metabolic pathways and changes in RXR dimer composition during adipogenesis. Genes Dev. 22(21) , 2953–2967 (2008).
  • Diamond M , MinerJ, YoshinagaSK, YamamotoKR. Transcription factor interactions – selectors of positive or negative regulation from a single DNA element. Science249(4974) , 1266–1272 (1990).
  • Nissen RM , YamamotoKR. The glucocorticoid receptor inhibits NFκB by interfering with serine-2 phosphorylation of the RNA polymerase II carboxy-terminal domain. Genes Dev.14(18) , 2314–2329 (2000).
  • Norris JD , ChangC, WittmannBMet al. The homeodomain protein HOXB13 regulates the cellular response to androgens. Mol. Cell 36(3) , 405–416 (2009).
  • Heldring N , IssacsGD, DiehlAGet al. Multiple sequence-specific DNA-binding proteins mediate estrogen receptor signaling through a tethering pathway. Mol. Endocrinol. 25(4) , 564–574 (2011).
  • Schmidt D , WilsonMD, BallesterBet al. Five-vertebrate ChIP-seq reveals the evolutionary dynamics of transcription factor binding. Science 328(5981) , 1036–1040 (2010).
  • Wilson MD , Barbosa-MoraisNL, SchmidtDet al. Species-specific transcription in mice carrying human chromosome 21. Science 322(5900) , 434–438 (2008).
  • Nechushtan H , BenvenistyN, BrandeisR, ReshefL. Glucocorticoids control phosphoenolpyruvate carboxykinase gene expression in a tissue specific manner. Nucleic Acids Res.15 , 6405–6417 (1987).
  • Odom DT , ZizlzpergerN, GordonDBet al. Control of pancreas and liver gene expression by HNF transcription factors. Science 303(5662) , 1378–1381 (2004).
  • Odom DT , DowellRD, JacobsenESet al. Tissue-specific transcriptional regulation has diverged significantly between human and mouse. Nat. Genet. 39 , 730–732 (2007).
  • Wakabayashi K -I, Okamura M, Tsutsuni S et al. The peroxisome proliferator-activated receptor γ/retinoid X receptor α heterodimer targets the histone modification enzyme PR-Set7/Setd8 gene and regulates adipogenesis through a positive feedback loop. Mol. Cell. Biol.29(13) , 3544–3555 (2009).
  • Siersbæk R , NielsenR, JohnSet al. Extensive chromatin remodeling and establishment of transcription factor hotspots during early adipogenesis. EMBO J. 30(8) , 1459–1472 (2011).
  • Steger DJ , GrantGR, SchuppMet al. Propagation of adipogenic signals through an epigenomic transition state. Genes Dev. 24(10) , 1035–1044 (2010).
  • Becker PB , RuppertS, SchützG. Genomic footprinting reveals cell type-specific DNA binding of ubiquitous factors. Cell51(3) , 435–443 (1987).
  • Heintzman ND , HonGC, HawkinsRDet al. Histone modifications at human enhancers reflect global cell-type-specific gene expression. Nature 459(7243) , 108–112 (2009).
  • Wang D , Garcia-BassetsI, BennerCet al. Reprogramming transcription by distinct classes of enhancers functionally defined by eRNA. Nature 474(7351) , 390–394 (2011).
  • Kagey MH , NewmanJJ, BilodeauSet al. Mediator and cohesin connect gene expression and chromatin architecture. Nature 467(7314) , 430–435 (2010).
  • Guelen L , PagieL, BrassetEet al. Domain organization of human chromosomes revealed by mapping of nuclear lamina interactions. Nature 453(7197) , 948–951 (2008).
  • Peric-Hupkes D , MeulemanW, PagieLet al. Molecular maps of the reorganization of genome-nuclear lamina interactions during differentiation. Mol. Cell 38(4) , 603–613 (2010).
  • Nader N , ChrousosGP, KinoT. Circadian rhythm transcription factor CLOCK regulates the transcriptional activity of the glucocorticoid receptor by acetylating its hinge region lysine cluster: potential physiological implications. FASEB J.23(5) , 1572–1583 (2009).
  • Fu M , RaoM, WangCet al. Acetylation of androgen receptor enhances coactivator binding and promotes prostate cancer cell growth. Mol. Cell. Biol. 23(23) , 8563–8575 (2003).
  • Ghisletti S , HuangW, OgawaSet al. Parallel SUMOylation-dependent pathways mediate gene- and signal-specific transrepression by LXRs and PPARγ. Mol. Cell 25(1) , 57–70 (2007).
  • Ieda M , FuJ, Delgado-OlguinPet al. Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell 142(3) , 375–386 (2010).
  • Takahashi K , YamanakaS. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell126(4) , 663–676 (2006).
  • Vierbuchen T , OstermeierA, PangZP, KokubuY, SüdhofTC, WernigM. Direct conversion of fibroblasts to functional neurons by defined factors. Nature463(7284) , 1035–1041 (2010).
  • Okamoto K , IwaiY, Oh-HoraMet al. IkappaBzeta regulates regulates T(H)17 development by cooperating with ROR nuclear receptors. Nature 464(7293) , 1381–1385 (2010).
  • Grimaldi B , BelletMM, KatadaSet al. PER2 controls lipid metabolism by direct regulation of PPARγ. Cell Metabol. 12(5) , 509–520 (2010).
  • Schmutz I , RippergerJA, Baeriswyl-AebischerS, AlbrechtU. The mammalian clock component PERIOD2 coordinates circadian output by interaction with nuclear receptors. Genes Dev.24(4) , 345–357 (2010).
  • Climent J , Perez-LosadaJ, QuigleyDAet al. Deletion of the PER3 gene on chromosome 1p36 in recurrent ER-positive breast cancer. J. Clin. Oncol. 28(23) , 3770–3778 (2010).
  • Cermakian N , Waddington Lamont E, Boudreau P, Boivin DB. Circadian clock gene expression in brain regions of Alzheimer‘s disease patients and control subjects. J. Biol. Rhythms26(2) , 160–170 (2011).
  • Asher G , SchiblerU. Crosstalk between components of circadian and metabolic cycles in mammals. Cell Metab.13(2) , 125–137 (2011).
  • Patel R , PatelM, TsaiRet al. LXRβ is required for glucocorticoid-induced hyperglycemia and hepatosteatosis in mice. J. Clin. Invest. 121(1) , 431–441 (2011).
  • Langlais D , CoutureC, BalsalobreA, DrouinJ. Regulatory network analyses reveal genome-wide potentiation of LIF signaling by glucocorticoids and define an innate cell defense response. PLoS Genet.4(10) , e1000224 (2008).
  • Szanto A , BalintBL, NagyZSet al. STAT6 transcription factor is a facilitator of the nuclear receptor PPARγ-regulated gene expression in macrophages and dendritic cells. Immunity 33(5) , 699–712 (2010).
  • Stavreva DA , WienchM, JohnSet al. Ultradian hormone stimulation induces glucocorticoid receptor-mediated pulses of gene transcription. Nat. Cell. Biol. 11(9) , 1093–1102 (2009).
  • Lightman SL , Conway-CampbellBL. The crucial role of pulsatile activity of the HPA axis for continuous dynamic equilibration. Nat. Rev. Neurosci.11(10) , 710–718 (2010).
  • Rauch A , SeitzS, BaschantUet al. Glucocorticoids suppress bone formation by attenuating osteoblast differentiation via the monomeric glucocorticoid receptor. Cell Metab. 11(6) , 517–531 (2010).
  • Guiducci C , GongM, XuZet al. TLR recognition of self nucleic acids hampers glucocorticoid activity in lupus. Nature 465(7300) , 937–941 (2010).
  • Tomlins SA , RhodesDR, PernerSet al. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 310(5748) , 644–648 (2005).
  • Mani RS , TomlinsSA, CallahanKet al. Induced chromosomal proximity and gene fusions in prostate cancer. Science 326(5957) , 1230 (2009).
  • Lin C , YangL, TanasaBet al. Nuclear receptor-induced chromosomal proximity and DNA breaks underlie specific translocations in cancer. Cell 139(6) , 1069–1083 (2009).
  • Yu J , YuJ, CaoQet al. An integrated network of androgen receptor, polycomb, and TMPRSS2–ERG gene fusions in prostate cancer progression. Cancer Cell 17(5) , 443–454 (2010).
  • Gévry N , HardyS, JacquesPEet al. Histone H2A.Z is essential for estrogen receptor signaling. Genes Dev. 23(13) , 1522–1533 (2009).
  • Phuc Le P , FriedmanJR, SchugJet al. Glucocorticoid receptor-dependent gene regulatory networks. PLoS Genet. 1(2) , e16 (2005).
  • Gao H , FältS, SandelinAet al. Genome-wide identification of estrogen receptor α-binding sites in mouse liver. Mol. Endocrinol. 22(1) , 10–22 (2007).
  • Hu S , YaoG, GuanXet al. Research resource: genome-wide mapping of in vivo androgen receptor binding sites in mouse epididymis. Mol. Endocrinol. 24(12) , 2392–2405 (2010).
  • Zhang C , FriasMA, MeleAet al. Integrative modeling defines the Nova splicing-regulatory network and its combinatorial controls. Science 329(5990) , 439–443 (2010).
  • Bandyopadhyay S , MehtaM, KuoDet al. Rewiring of genetic networks in response to DNA damage. Science 330(2009) , 1385–1389 (2010).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.