Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
3.
Proc Natl Acad Sci U S A ; 106(23): 9280-5, 2009 Jun 09.
Article in English | MEDLINE | ID: mdl-19470456

ABSTRACT

A number of nuclear complexes modify chromatin structure and operate as functional units. However, the in vivo role of each component within the complexes is not known. ATP-dependent chromatin remodeling complexes form several types of protein complexes, which reorganize chromatin structure cooperatively with histone modifiers. Williams syndrome transcription factor (WSTF) was biochemically identified as a major subunit, along with 2 distinct complexes: WINAC, a SWI/SNF-type complex, and WICH, an ISWI-type complex. Here, WSTF(-/-) mice were generated to investigate its function in chromatin remodeling in vivo. Loss of WSTF expression resulted in neonatal lethality, and all WSTF(-/-) neonates and approximately 10% of WSTF(+/-) neonates suffered cardiovascular abnormalities resembling those found in autosomal-dominant Williams syndrome patients. Developmental analysis of WSTF(-/-) embryos revealed that Gja5 gene regulation is aberrant from E9.5, conceivably because of inappropriate chromatin reorganization around the promoter regions where essential cardiac transcription factors are recruited. In vitro analysis in WSTF(-/-) mouse embryonic fibroblast (MEF) cells also showed impaired transactivation functions of cardiac transcription activators on the Gja5 promoter, but the effects were reversed by overexpression of WINAC components. Likewise in WSTF(-/-) MEF cells, recruitment of Snf2h, an ISWI ATPase, to PCNA and cell survival after DNA damage were both defective, but were ameliorated by overexpression of WICH components. Thus, the present study provides evidence that WSTF is shared and is a functionally indispensable subunit of the WICH complex for DNA repair and the WINAC complex for transcriptional control.


Subject(s)
Chromatin Assembly and Disassembly , Transcription Factors/metabolism , Animals , Cardiovascular Abnormalities/genetics , Cardiovascular Abnormalities/metabolism , Cells, Cultured , DNA Repair , DNA Replication , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Gene Expression , Mice , Transcription Factors/genetics
4.
EMBO Rep ; 9(6): 563-8, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18451880

ABSTRACT

The female sex steroid hormone oestrogen stimulates both cell proliferation and cell differentiation in target tissues. These biological actions are mediated primarily through nuclear oestrogen receptors (ERs). The ligand-dependent transactivation of ERs requires several nuclear co-regulator complexes; however, the cell-cycle-dependent associations of these complexes are poorly understood. By using a synchronization system, we found that the transactivation function of ERalpha at G2/M was lowered. Biochemical approaches showed that ERalpha associated with two discrete classes of ATP-dependent chromatin-remodelling complex in a cell-cycle-dependent manner. The components of the NuRD-type complex were identified as G2/M-phase-specific ERalpha co-repressors. Thus, our results indicate that the transactivation function of ERalpha is cell-cycle dependent and is coupled with a cell-cycle-dependent association of chromatin-remodelling complexes.


Subject(s)
Cell Cycle/physiology , Chromatin Assembly and Disassembly , Chromatin/metabolism , Estrogen Receptor alpha/metabolism , Macromolecular Substances/metabolism , Cell Line , Estrogen Receptor alpha/genetics , Estrogens/metabolism , Female , Humans , Transcriptional Activation
5.
Mol Cell Biol ; 28(12): 3995-4003, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18391013

ABSTRACT

TLX is an orphan nuclear receptor (also called NR2E1) that regulates the expression of target genes by functioning as a constitutive transrepressor. The physiological significance of TLX in the cytodifferentiation of neural cells in the brain is known. However, the corepressors supporting the transrepressive function of TLX have yet to be identified. In this report, Y79 retinoblastoma cells were subjected to biochemical techniques to purify proteins that interact with TLX, and we identified LSD1 (also called KDM1), which appears to form a complex with CoREST and histone deacetylase 1. LSD1 interacted with TLX directly through its SWIRM and amine oxidase domains. LSD1 potentiated the transrepressive function of TLX through its histone demethylase activity as determined by a luciferase assay using a genomically integrated reporter gene. LSD1 and TLX were recruited to a TLX-binding site in the PTEN gene promoter, accompanied by the demethylation of H3K4me2 and deacetylation of H3. Knockdown of either TLX or LSD1 derepressed expression of the endogenous PTEN gene and inhibited cell proliferation of Y79 cells. Thus, the present study suggests that LSD1 is a prime corepressor for TLX.


Subject(s)
Gene Expression Regulation , Oxidoreductases, N-Demethylating/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation , Genes, Reporter , Histone Deacetylases/metabolism , Histone Demethylases , Histones/metabolism , Humans , Models, Biological , Orphan Nuclear Receptors , Oxidoreductases, N-Demethylating/metabolism , Promoter Regions, Genetic , Protein Structure, Tertiary
6.
J Biol Chem ; 283(18): 11947-53, 2008 May 02.
Article in English | MEDLINE | ID: mdl-18326047

ABSTRACT

Lipofuscin contains fluorophores, which represent a biomarker for cellular aging. Although it remains unsubstantiated clinically, experimental results support that the accumulation of lipofuscin is related to an increased risk of choroidal neovascularization due to age-related macular degeneration, a leading cause of legal blindness. Here, we report that a major lipofuscin component, A2E, activates the retinoic acid receptor (RAR). In vitro experiments using luciferase reporter assay, competitional binding assay, analysis of target genes, and chromatin immunoprecipitation (ChIP) assay strongly suggest that A2E is a bona fide ligand for RAR and induces sustained activation of RAR target genes. A2E-induced vascular endothelial growth factor (VEGF) expression in a human retinal pigment epithelial cell line (ARPE-19) and RAR antagonist blocked the up-regulation of VEGF. The conditioned medium of A2E-treated ARPE-19 cells induced tube formation in human umbilical vascular endothelial cells, which was blocked by the RAR antagonist and anti-VEGF antibody. These results suggest that A2E accumulation results in the phenotypic alteration of retinal pigment epithelial cells, predisposing the environment to choroidal neovascularization development. This is mediated through the agonistic function of A2E, at least in part. The results of this study provide a novel potential therapeutic target for this incurable condition.


Subject(s)
Epithelial Cells/metabolism , Lipofuscin/metabolism , Pigment Epithelium of Eye/cytology , Pigment Epithelium of Eye/metabolism , Pyridinium Compounds/metabolism , Receptors, Retinoic Acid/metabolism , Retinal Pigments/metabolism , Retinoids/metabolism , Cell Line , Epithelial Cells/drug effects , Gene Expression Regulation/drug effects , Humans , Ligands , Neovascularization, Physiologic/drug effects , Pigment Epithelium of Eye/drug effects , Pyridinium Compounds/pharmacology , Retinal Pigments/pharmacology , Retinoids/pharmacology , Vascular Endothelial Growth Factor A/genetics
7.
Nat Cell Biol ; 9(11): 1273-85, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17952062

ABSTRACT

Histone modifications induced by activated signalling cascades are crucial to cell-lineage decisions. Osteoblast and adipocyte differentiation from common mesenchymal stem cells is under transcriptional control by numerous factors. Although PPAR-gamma (peroxisome proliferator activated receptor-gamma) has been established as a prime inducer of adipogenesis, cellular signalling factors that determine cell lineage in bone marrow remain generally unknown. Here, we show that the non-canonical Wnt pathway through CaMKII-TAK1-TAB2-NLK transcriptionally represses PPAR-gamma transactivation and induces Runx2 expression, promoting osteoblastogenesis in preference to adipogenesis in bone marrow mesenchymal progenitors. Wnt-5a activates NLK (Nemo-like kinase), which in turn phosphorylates a histone methyltransferase, SETDB1 (SET domain bifurcated 1), leading to the formation of a co-repressor complex that inactivates PPAR-gamma function through histone H3-K9 methylation. These findings suggest that the non-canonical Wnt signalling pathway suppresses PPAR-gamma function through chromatin inactivation triggered by recruitment of a repressing histone methyltransferase, thus leading to an osteoblastic cell lineage from mesenchymal stem cells.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , PPAR gamma/metabolism , Signal Transduction/physiology , Transcriptional Activation/physiology , Wnt Proteins/physiology , Adipogenesis , Animals , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Down-Regulation , Genetic Vectors , Histone-Lysine N-Methyltransferase/drug effects , Mice , Mice, Transgenic , Mutation , Osteogenesis , PPAR gamma/drug effects , PPAR gamma/genetics , Phosphorylation , Plasmids , Signal Transduction/drug effects , Wnt Proteins/genetics , Wnt Proteins/pharmacology , Wnt-5a Protein
8.
Nat Cell Biol ; 9(5): 604-11, 2007 May.
Article in English | MEDLINE | ID: mdl-17435748

ABSTRACT

MicroRNAs (miRNAs) control cell proliferation, differentiation and fate through modulation of gene expression by partially base-pairing with target mRNA sequences. Drosha is an RNase III enzyme that is the catalytic subunit of a large complex that cleaves pri-miRNAs with distinct structures into pre-miRNAs. Here, we show that both the p68 and p72 DEAD-box RNA helicase subunits in the mouse Drosha complex are indispensable for survival in mice, and both are required for primary miRNA and rRNA processing. Gene disruption of either p68 or p72 in mice resulted in early lethality, and in both p68(-/-) and p72(-/-) embryos, expression levels of a set of, but not all, miRNAs and 5.8S rRNA were significantly lowered. In p72(-/-) MEF cells, expression of p72, but not a mutant lacking ATPase activity, restored the impaired expression of miRNAs and 5.8S rRNA. Furthermore, we purified the large complex of mouse Drosha and showed it could generate pre-miRNA and 5.8S rRNA in vitro. Thus, we suggest that DEAD-box RNA helicase subunits are required for recognition of a subset of primary miRNAs in mDrosha-mediated processing.


Subject(s)
DEAD-box RNA Helicases/metabolism , Embryo, Mammalian/metabolism , MicroRNAs/metabolism , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA, Ribosomal/metabolism , Ribonuclease III/metabolism , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Chromatin Immunoprecipitation , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/enzymology , Fibroblasts/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Isoenzymes/metabolism , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , RNA Interference , RNA, Ribosomal, 5.8S/metabolism
9.
EMBO J ; 26(3): 764-74, 2007 Feb 07.
Article in English | MEDLINE | ID: mdl-17255935

ABSTRACT

Photoreceptor cell-specific nuclear receptor (PNR) (NR2E3) acts as a sequence-specific repressor that controls neuronal differentiation in the developing retina. We identified a novel PNR co-repressor, Ret-CoR, that is expressed in the developing retina and brain. Biochemical purification of Ret-CoR identified a multiprotein complex that included E2F/Myb-associated proteins, histone deacetylases (HDACs) and NCoR/HDAC complex-related components. Ret-CoR appeared to function as a platform protein for the complex, and interacted with PNR via two CoRNR motifs. Purified Ret-CoR complex exhibited HDAC activity, co-repressed PNR transrepression function in vitro, and co-repressed PNR function in PNR target gene promoters, presumably in the retinal progenitor cells. Notably, the appearance of Ret-CoR protein was cell-cycle-stage-dependent (from G1 to S). Therefore, Ret-CoR appears to act as a component of an HDAC co-repressor complex that supports PNR repression function in the developing retina, and may represent a co-regulator class that supports transcriptional regulator function via cell-cycle-dependent expression.


Subject(s)
Cell Cycle/genetics , Gene Expression Regulation, Developmental , Multiprotein Complexes/metabolism , Photoreceptor Cells/embryology , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Cell Proliferation , Histone Deacetylases/metabolism , Mice , Multiprotein Complexes/genetics , Orphan Nuclear Receptors , RNA Helicases , Receptors, Cytoplasmic and Nuclear/genetics , Repressor Proteins/genetics , Repressor Proteins/isolation & purification
10.
Endocr J ; 53(2): 157-72, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16618973

ABSTRACT

Nuclear steroid/thyroid vitamin A/D receptor genes form a gene superfamily and encode DNA-binding transcription factors that control the transcription of target genes in a ligand-dependent manner. It has become clear that chromatin remodeling and the modification of histones, the main components of chromatin, play crucial roles in gene transcription, and many distinct classes of NR-interacting co-regulators have been identified that perform significant roles in gene transcription. Since NR dysfunction can lead to the onset or progression of endocrine disease, elucidation of the mechanisms of gene regulation mediated by NRs, as well as the identification and characterization of co-regulator complexes (especially chromatin remodeling and histone-modifying complexes), is essential not only for better understanding of NR ligand function, but also for pathophysiological studies and the development of therapeutic interventions in humans.


Subject(s)
Chromatin Assembly and Disassembly , Gene Expression Regulation , Histones/metabolism , Protein Processing, Post-Translational , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Humans , Models, Biological , Multiprotein Complexes , Protein Structure, Tertiary , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction
11.
J Biol Chem ; 281(1): 20-6, 2006 Jan 06.
Article in English | MEDLINE | ID: mdl-16260778

ABSTRACT

It is well established that genetic mutations that impair BRCA1 function predispose women to early onset of breast and ovarian cancer. However, the co-regulatory factors that support normal BRCA1 functions remain to be identified. Using a biochemical approach to search for such co-regulatory factors, we identified hGCN5, TRRAP, and hMSH2/6 as BRCA1-interacting proteins. Genetic mutations in the C-terminal transactivation domain of BRCA1, as found in breast cancer patients (Chapman, M. S., and Verma, I. M. (1996) Nature 382, 678-679), caused the loss of physical interaction between BRCA1 and TRRAP and significantly reduced the co-activation of BRCA1 transactivation function by hGCN5/TRRAP. The reported transcriptional squelching between BRCA1 and estrogen receptor alpha (Fan, S., Wang, J., Yuan, R., Ma, Y., Meng, Q., Erdos, M. R., Pestell, R. G., Yuan, F., Auborn, K. J., Goldberg, I. D., and Rosen, E. M. (1999) Science 284, 1354-1356) was rescued by the overexpression of TRRAP or hGCN5. Histone acetyltransferase hGCN5 activity appeared to be indispensable for coregulator complex function in both BRCA1-mediated gene regulation and DNA repair. Biochemical purification of the hGCN5/TRRAP-containing complex suggested that hGCN5/TRRAP formed a complex with hMSH2/hMSH6, presumably as a novel subclass of hGCN5/TRRAP-containing known TFTC (TBP-free TAF-containing)-type histone acetyltransferase complex (hTFTC, hPCAF, and hSTAGA) (Yanagisawa, J., Kitagawa, H., Yanagida, M., Wada, O., Ogawa, S., Nakagomi, M., Oishi, H., Yamamoto, Y., Nagasawa, H., McMahon, S. B., Cole, M. D., Tora, L., Takahashi, N., and Kato, S. (2002) Mol. Cell 9, 553-562). Unlike other subclasses, the isolated complex harbored a previously unknown combination of components including hMSH2 and hMSH6, major components of the BRCA1 genome surveillance repair complex (BASC). Thus, our results suggested that the multiple BRCA1 functions require a novel hGCN5/TRRAP histone acetyltransferase complex subclass.


Subject(s)
BRCA1 Protein/metabolism , Breast Neoplasms/metabolism , Cell Cycle Proteins/metabolism , Histone Acetyltransferases/metabolism , Histones/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Histone Acetyltransferases/genetics , Humans , Multienzyme Complexes/metabolism , Nuclear Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic/physiology , p300-CBP Transcription Factors
12.
Biochem Biophys Res Commun ; 327(3): 933-8, 2005 Feb 18.
Article in English | MEDLINE | ID: mdl-15649435

ABSTRACT

TBP-free TAF II-containing-type HAT complex subclasses, which contain hGCN5 HAT and TRRAP, appear to act as common coactivator complexes for nuclear receptors. However, their physiological significance with respect to each nuclear receptor remains to be established. To address this issue, we used hepatic cell lines (HepG2) with reduced endogenous TRRAP expression through antisense RNA expression or with overexpressed TRRAP or other major coactivators. The ligand-induced transactivation function of liver X receptor alpha (LXRalpha) and farnesoid X receptor/bile acid receptor reflected TRRAP expression levels, while that of PPARgamma did not. A GST pull-down assay indicated that TRRAP contains two potential LXRalpha-interacting domains in the C-terminal and central domains. Expression of antisense TRRAP RNA in HepG2 cells abolished the ligand-induced expression of LXRalpha target genes. These results suggested that TRRAP plays an important role as a coactivator, presumably part of a complex, in lipid metabolism through regulation of the LXRalpha-mediated gene cascade in hepatic cells.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , Liver/metabolism , Nuclear Proteins/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Bile Acids and Salts/metabolism , DNA-Binding Proteins/genetics , Ligands , Lipid Metabolism , Liver X Receptors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Orphan Nuclear Receptors , PPAR gamma/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Transcriptional Activation
13.
Invest Ophthalmol Vis Sci ; 43(11): 3489-94, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12407160

ABSTRACT

PURPOSE: To clarify the functions of a specific subtype of thyroid hormone receptor (TR), TRbeta2, and photoreceptor cell-specific nuclear receptor (PNR) in the development of cone photoreceptors. METHODS: The expression of short (S)- and medium (M)-wavelength cone opsins was analyzed by reverse transcription polymerase chain reaction (RT-PCR) and Northern blot analysis in mice without a functional PNR (rd7/rd7 mice), and levels of plasma thyroid hormones and expression of TRbeta2 were also examined. Concomitantly, by means of reporter assays, the roles of PNR and TRbeta2 in the S- and M-cone opsin expression were explored at the transcriptional level. RESULTS: In rd7/rd7 mice, an abnormal increase in cone photoreceptors was observed immediately before retinal maturation normally occurs. Although an increase in S-cone opsin in the retina was observed during and after retinal development, the expression of M-cone opsin expression was not perturbed during retinal maturation. Plasma concentrations of thyroid hormone and levels of TRbeta2 expression in the rd7/rd7 mouse retina over the developmental period were normal. Transcriptional studies demonstrated that TRbeta2, but not PNR, activated the M-cone opsin gene promoter function, while suppressing the S-cone opsin promoter function enhanced by CRX in a thyroid hormone-dependent manner. CONCLUSIONS: The results indicate that PNR may suppress proliferation of cone photoreceptor progenitor cells and that the regulation of S- and M-cone opsin gene expression is mediated by TRbeta2 and CRX, but not by PNR. Thus, our results partly disclosed the molecular mechanism of cone photoreceptor development, highlighting the distinct functions of PNR and TRbeta2.


Subject(s)
Homeodomain Proteins/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Retinal Cone Photoreceptor Cells/embryology , Retinal Degeneration/metabolism , Thyroid Hormone Receptors beta/physiology , Trans-Activators/physiology , Transcription Factors/physiology , Animals , Blotting, Northern , Mice , Mice, Inbred C57BL , Mice, Knockout , Orphan Nuclear Receptors , RNA/isolation & purification , RNA, Messenger/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/pathology , Reverse Transcriptase Polymerase Chain Reaction , Rod Opsins/genetics , Rod Opsins/metabolism , Thyroid Hormones/blood , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...