101
|
Bojkowska K, Kapopoulou A, Santoni de Sio F, Aloisio F, Cassano M, Zangger N, Offner S, Cartoni C, Thomas C, Quenneville S, Johnsson K, Trono D. Liver-specific ablation of Krüppel-associated box-associated protein 1 in mice leads to male-predominant hepatosteatosis and development of liver adenoma. Hepatology 2012; 56:1279-90. [PMID: 22684873 PMCID: PMC4894457 DOI: 10.1002/hep.25767] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 04/01/2012] [Indexed: 02/06/2023]
Abstract
UNLABELLED The liver is characterized by sexually dimorphic gene expression translating into sex-specific differences in lipid, drug, steroid hormone, and xenobiotic metabolism, with distinct responses of males and females to environmental challenges. Here, we investigated the role of the Krüppel-associated box (KRAB)-associated protein 1 (KAP1) epigenetic regulator in this process. Liver-specific KAP1 knockout (KO) led to strikingly sexually dimorphic phenotypic disturbances, including male-predominant steatosis and hepatic tumors with up-regulation of protein kinase B and extracellular signal-related kinases 1/2 mitogen-activated protein kinase signaling. This correlated with the sex-specific transcriptional dysregulation of a wide range of metabolic genes, notably those involved in retinol and sex hormone processing as well as in detoxification. Furthermore, chromatin immunoprecipitation followed by deep sequencing indicated that a number of dysregulated genes are direct targets of the KRAB/KAP1 repression system. Those genes include sexually dimorphic cytochrome P 450 Cyp2d9, glutathione S-transferase π, Cyp2a, Cyp2b, and Cyp3a gene clusters. Additionally, we identified a male-restricted KAP1-binding site in the fat-specific protein 27 gene, correlating with its male-predominant up-regulation upon Kap1 deletion, suggesting that the latter might be an important trigger in the development of male-specific hepatosteatosis and secondary tumorigenesis. CONCLUSION This work reveals KRAB/KAP1-mediated transcriptional regulation as a central event in metabolic control hormones, drugs, and xenobiotics in the liver and further links disturbances in these processes with hepatic carcinogenesis.
Collapse
Affiliation(s)
- Karolina Bojkowska
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland,“Frontiers in Genetics” National Program, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Adamandia Kapopoulou
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland,Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Francesca Santoni de Sio
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland,“Frontiers in Genetics” National Program, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Fabio Aloisio
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland,School of Veterinary Medicine, University of Bern, Switzerland
| | - Marco Cassano
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland,“Frontiers in Genetics” National Program, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Nadine Zangger
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland,“Frontiers in Genetics” National Program, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Sandra Offner
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland,“Frontiers in Genetics” National Program, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Cristina Cartoni
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Charles Thomas
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Simon Quenneville
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland,“Frontiers in Genetics” National Program, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Kai Johnsson
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland,School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland,“Chemical Biology” National Program, Ecole Polytechnique Fédérale de Lausanne
| | - Didier Trono
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland,“Frontiers in Genetics” National Program, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
102
|
Bastie JN, Aucagne R, Droin N, Solary E, Delva L. Heterogeneity of molecular markers in chronic myelomonocytic leukemia: a disease associated with several gene alterations. Cell Mol Life Sci 2012; 69:2853-61. [PMID: 22415325 PMCID: PMC11114957 DOI: 10.1007/s00018-012-0956-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 02/29/2012] [Accepted: 03/01/2012] [Indexed: 12/21/2022]
Abstract
The relatively homogenous clinical features and poor prognosis of chronic myelomonocytic leukemia (CMML) are associated with a molecular heterogeneity, with various mutations impacting several convergent pathways. Due to the restricted understanding of the mechanism involved in leukemogenesis, CMML still appears as a diagnostic and therapeutic undertaking, and poor prognosis of leukemia. Contrary to chronic myelogenous leukemia, BCR-ABL1-positive, cytogenetic, and molecular abnormalities of CMML are not specific and not pathognomonic, confirming the different levels of heterogeneity of this disease. Various mutations can be associated with a common phenotype not distinct at the clinical level, further demonstrating that molecular probings are needed for choosing individual targeted therapies.
Collapse
Affiliation(s)
- Jean-Noël Bastie
- Faculté de Médecine, Inserm UMR 866, Université de Bourgogne, 7 bd Jeanne d’Arc, 21000 Dijon, France
- Service d’Hématologie Clinique, Centre Hospitalo-Universitaire, 21000 Dijon, France
| | - Romain Aucagne
- Faculté de Médecine, Inserm UMR 866, Université de Bourgogne, 7 bd Jeanne d’Arc, 21000 Dijon, France
- Laboratoire de Génétique Moléculaire des Cellules Souches, Institut de Recherche en Immunologie et en Cancérologie, Université de Montréal, Montréal, QC H3C 3J7 Canada
| | - Nathalie Droin
- Inserm UMR 1009, IRCIV, Institut Gustave Roussy, 114 rue Edouard Vaillant, 94805 Villejuif, France
| | - Eric Solary
- Inserm UMR 1009, IRCIV, Institut Gustave Roussy, 114 rue Edouard Vaillant, 94805 Villejuif, France
| | - Laurent Delva
- Faculté de Médecine, Inserm UMR 866, Université de Bourgogne, 7 bd Jeanne d’Arc, 21000 Dijon, France
| |
Collapse
|
103
|
Messerschmidt DM. Should I stay or should I go: protection and maintenance of DNA methylation at imprinted genes. Epigenetics 2012; 7:969-75. [PMID: 22869105 PMCID: PMC3515016 DOI: 10.4161/epi.21337] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Recent findings shed light on the coordination of two fundamental, yet mechanistically opposing, processes in the early mammalian embryo. During the oocyte-to-embryo transition and early preimplantation development nuclear reprogramming occurs. This resetting of the epigenome in maternal and paternal pronuclei to a ground state is the essential step ensuring totipotency in the zygote, the first embryonic stage. Radical, global DNA demethylation, which occurs actively in the paternal and passively in the maternal genome, is a prominent feature of nuclear reprogramming; yet, this process poses a danger to a subset of methylated sequences that must be preserved for their germline to soma inheritance. Genomic imprinting and its importance were demonstrated three decades ago by a series of experiments generating non-viable mammalian uniparental embryos. Indeed, imprinted loci, gene clusters with parent-of-origin specific gene expression patterns, must retain their differential methylation status acquired during gametogenesis throughout embryogenesis and in adult tissues. It is just recently that the molecular players that protect/maintain imprinting marks during reprogramming in preimplantation embryos have been identified, in particular, an epigenetic modifier complex formed by ZFP57 and TRIM28/KAP1. The interaction of these and other molecules with the newly formed embryonic chromatin and imprinted genes is discussed and highlighted herein.
Collapse
|
104
|
Santoni de Sio FR, Barde I, Offner S, Kapopoulou A, Corsinotti A, Bojkowska K, Genolet R, Thomas JH, Luescher IF, Pinschewer D, Harris N, Trono D. KAP1 regulates gene networks controlling T-cell development and responsiveness. FASEB J 2012; 26:4561-75. [PMID: 22872677 DOI: 10.1096/fj.12-206177] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Chromatin remodeling at specific genomic loci controls lymphoid differentiation. Here, we investigated the role played in this process by Kruppel-associated box (KRAB)-associated protein 1 (KAP1), the universal cofactor of KRAB-zinc finger proteins (ZFPs), a tetrapod-restricted family of transcriptional repressors. T-cell-specific Kap1-deleted mice displayed a significant expansion of immature thymocytes, imbalances in CD4(+)/CD8(+) cell ratios, and altered responses to TCR and TGFβ stimulation when compared to littermate KAP1 control mice. Transcriptome and chromatin studies revealed that KAP1 binds T-cell-specific cis-acting regulatory elements marked by the H3K9me3 repressive mark and enriched in Ikaros/NuRD complexes. Also, KAP1 directly controls the expression of several genes involved in TCR and cytokine signaling. Among these, regulation of FoxO1 seems to play a major role in this system. Likely responsible for tethering KAP1 to at least part of its genomic targets, a small number of KRAB-ZFPs are selectively expressed in T-lymphoid cells. These results reveal the so far unsuspected yet important role of KAP1-mediated epigenetic regulation in T-lymphocyte differentiation and activation.
Collapse
|
105
|
Embryonic Stem Cell Interactomics: The Beginning of a Long Road to Biological Function. Stem Cell Rev Rep 2012; 8:1138-54. [PMID: 22847281 DOI: 10.1007/s12015-012-9400-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
106
|
Herquel B, Ouararhni K, Davidson I. The TIF1α-related TRIM cofactors couple chromatin modifications to transcriptional regulation, signaling and tumor suppression. Transcription 2012; 2:231-6. [PMID: 22231120 DOI: 10.4161/trns.2.5.17725] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
TRIM24 (TIF1α), TRIM28 (TIF1β) and TRIM33 (TIF1γ) are related cofactors defining a subgroup of the tripartite motif (TRIM) superfamily comprising an N-terminal RING finger E3 ligase and a C-terminal PHD-Bromodomain chromatin interacting module. Increasing evidence highlights the important roles of these proteins as modulators of multiple signaling pathways during normal development and as tumor suppressors. The finding that they interact to form a multiprotein complex suggests new mechanisms to integrate multiple signaling pathways for tumor suppression.
Collapse
Affiliation(s)
- Benjamin Herquel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch Cédex, France
| | | | | |
Collapse
|
107
|
de Sio FRS, Massacand J, Barde I, Offner S, Corsinotti A, Kapopoulou A, Bojkowska K, Dagklis A, Fernandez M, Ghia P, Thomas JH, Pinschewer D, Harris N, Trono D. KAP1 regulates gene networks controlling mouse B-lymphoid cell differentiation and function. Blood 2012; 119:4675-85. [PMID: 22452978 PMCID: PMC3683646 DOI: 10.1182/blood-2011-12-401117] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Chromatin remodeling is fundamental for B-cell differentiation. In the present study, we explored the role of KAP1, the cofactor of KRAB-ZFP transcriptional repressors, in this process. B-lymphoid-specific Kap1-KO mice displayed reduced numbers of mature B cells, lower steady-state levels of Abs, and accelerated rates of decay of neutralizing Abs after viral immunization. Transcriptome analyses of Kap1-deleted B splenocytes revealed an up-regulation of PTEN, the enzymatic counteractor of PIK3 signaling, and of genes encoding DNA-damage response factors, cell-cycle regulators, and chemokine receptors. ChIP/seq studies established that KAP1 bound at or close to several of these genes and controlled chromatin status at their promoters. Genome wide, KAP1 binding sites lacked active B cell-specific enhancers and were enriched in repressive histone marks, further supporting a role for this molecule in gene silencing in vivo. Likely responsible for tethering KAP1 to at least some of these targets, a discrete subset of KRAB-ZFPs is enriched in B lymphocytes. Our results therefore reveal the role of KRAB/KAP1-mediated epigenetic regulation in B-cell development and homeostasis.
Collapse
Affiliation(s)
- Francesca R. Santoni de Sio
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Frontiers in Genetics Program, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Joanna Massacand
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Swiss Vaccine Research Institute, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Isabelle Barde
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Frontiers in Genetics Program, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Sandra Offner
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Frontiers in Genetics Program, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Andrea Corsinotti
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Frontiers in Genetics Program, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Adamandia Kapopoulou
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Frontiers in Genetics Program, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), 1015 Lausanne, Switzerland
| | - Karolina Bojkowska
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Frontiers in Genetics Program, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Antonis Dagklis
- Laboratory of B cell Neoplasia, Division of Molecular Oncology and Unit of Lymphoid Malignancies, Department of Onco-Hematology, Università Vita-Salute and Istituto Scientifico San Raffaele, Milan, Italy
| | | | - Paolo Ghia
- Laboratory of B cell Neoplasia, Division of Molecular Oncology and Unit of Lymphoid Malignancies, Department of Onco-Hematology, Università Vita-Salute and Istituto Scientifico San Raffaele, Milan, Italy
| | - James H. Thomas
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | | | - Nicola Harris
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Swiss Vaccine Research Institute, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Didier Trono
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Frontiers in Genetics Program, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
108
|
TRIM28 prevents autoinflammatory T cell development in vivo. Nat Immunol 2012; 13:596-603. [PMID: 22544392 DOI: 10.1038/ni.2293] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 03/26/2012] [Indexed: 12/15/2022]
Abstract
TRIM28 is a component of heterochromatin complexes whose function in the immune system is unknown. By studying mice with conditional T cell-specific deletion of TRIM28 (CKO mice), we found that TRIM28 was phosphorylated after stimulation via the T cell antigen receptor (TCR) and was involved in the global regulation of CD4(+) T cells. The CKO mice had a spontaneous autoimmune phenotype that was due in part to early lymphopenia associated with a defect in the production of interleukin 2 (IL-2) as well as incomplete cell-cycle progression of their T cells. In addition, CKO T cells showed derepression of the cytokine TGF-β3, which resulted in an altered cytokine balance; this caused the accumulation of autoreactive cells of the T(H)17 subset of helper T cells and of Foxp3(+) T cells. Notably, CKO Foxp3(+) T cells were unable to prevent the autoimmune phenotype in vivo. Our results show critical roles for TRIM28 in both T cell activation and T cell tolerance.
Collapse
|
109
|
Hu C, Zhang S, Gao X, Gao X, Xu X, Lv Y, Zhang Y, Zhu Z, Zhang C, Li Q, Wong J, Cui Y, Zhang W, Ma L, Wang C. Roles of Kruppel-associated Box (KRAB)-associated Co-repressor KAP1 Ser-473 Phosphorylation in DNA Damage Response. J Biol Chem 2012; 287:18937-52. [PMID: 22496453 DOI: 10.1074/jbc.m111.313262] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The Kruppel-associated box (KRAB)-associated co-repressor KAP1 is an essential nuclear co-repressor for the KRAB zinc finger protein superfamily of transcriptional factors. Ataxia telangiectasia mutated (ATM)-Chk2 and ATM- and Rad3-related (ATR)-Chk1 are two primary kinase signaling cascades activated in response to DNA damage. A growing body of evidence suggests that ATM and ATR phosphorylate KAP1 at Ser-824 in response to DNA damage and regulate KAP1-dependent chromatin condensation, DNA repair, and gene expression. Here, we show that, depending on the type of DNA damage that occurs, KAP1 Ser-473 can be phosphorylated by ATM-Chk2 or ATR-Chk1 kinases. Phosphorylation of KAP1 at Ser-473 attenuated its binding to the heterochromatin protein 1 family proteins and inhibited its transcriptional repression of KRAB-zinc finger protein (KRAB-ZFP) target genes. Moreover, KAP1 Ser-473 phosphorylation induced by DNA damage stimulated KAP1-E2F1 binding. Overexpression of heterochromatin protein 1 significantly inhibited E2F1-KAP1 binding. Elimination of KAP1 Ser-473 phosphorylation increased E2F1-targeted proapoptotic gene expression and E2F1-induced apoptosis in response to DNA damage. Furthermore, loss of phosphorylation of KAP1 Ser-473 led to less BRCA1 focus formation and slower kinetics of loss of γH2AX foci after DNA damage. KAP1 Ser-473 phosphorylation was required for efficient DNA repair and cell survival in response to DNA damage. Our studies reveal novel functions of KAP1 Ser-473 phosphorylation under stress.
Collapse
Affiliation(s)
- Chen Hu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and College of Life Science and
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
110
|
Messerschmidt DM, de Vries W, Ito M, Solter D, Ferguson-Smith A, Knowles BB. Trim28 is required for epigenetic stability during mouse oocyte to embryo transition. Science 2012; 335:1499-502. [PMID: 22442485 DOI: 10.1126/science.1216154] [Citation(s) in RCA: 246] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Phenotypic variability in genetic disease is usually attributed to genetic background variation or environmental influence. Here, we show that deletion of a single gene, Trim28 (Kap1 or Tif1β), from the maternal germ line alone, on an otherwise identical genetic background, results in severe phenotypic and epigenetic variability that leads to embryonic lethality. We identify early and minute epigenetic variations in blastomeres of the preimplantation embryo of these animals, suggesting that the embryonic lethality may result from the misregulation of genomic imprinting in mice lacking maternal Trim28. Our results reveal the long-range effects of a maternal gene deletion on epigenetic memory and illustrate the delicate equilibrium of maternal and zygotic factors during nuclear reprogramming.
Collapse
|
111
|
Shibata M, Blauvelt KE, Liem KF, García-García MJ. TRIM28 is required by the mouse KRAB domain protein ZFP568 to control convergent extension and morphogenesis of extra-embryonic tissues. Development 2012; 138:5333-43. [PMID: 22110054 DOI: 10.1242/dev.072546] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
TRIM28 is a transcriptional regulator that is essential for embryonic development and is implicated in a variety of human diseases. The roles of TRIM28 in distinct biological processes are thought to depend on its interaction with factors that determine its DNA target specificity. However, functional evidence linking TRIM28 to specific co-factors is scarce. chatwo, a hypomorphic allele of Trim28, causes embryonic lethality and defects in convergent extension and morphogenesis of extra-embryonic tissues. These phenotypes are remarkably similar to those of mutants in the Krüppel-associated box (KRAB) zinc finger protein ZFP568, providing strong genetic evidence that ZFP568 and TRIM28 control morphogenesis through a common molecular mechanism. We determined that chatwo mutations decrease TRIM28 protein stability and repressive activity, disrupting both ZFP568-dependent and ZFP568-independent roles of TRIM28. These results, together with the analysis of embryos bearing a conditional inactivation of Trim28 in embryonic-derived tissues, revealed that TRIM28 is differentially required by ZFP568 and other factors during the early stages of mouse embryogenesis. In addition to uncovering novel roles of TRIM28 in convergent extension and morphogenesis of extra-embryonic tissues, our characterization of chatwo mutants demonstrates that KRAB domain proteins are essential to determine some of the biological functions of TRIM28.
Collapse
Affiliation(s)
- Maho Shibata
- Department of Molecular Biology and Genetics, Cornell University, 526 Campus Road, Ithaca, NY 14853, USA
| | | | | | | |
Collapse
|
112
|
TRIM involvement in transcriptional regulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 770:59-76. [PMID: 23631000 DOI: 10.1007/978-1-4614-5398-7_5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Members of the tripartite motif (TRIM) protein family are found in all multicellular eukaryotes and function in a wide range of cellular processes such as cell cycle regulation, differentiation, development, oncogenesis and viral response. Over the past few years, several TRIM proteins have been reported to control gene expression through regulation of the transcriptional activity of numerous sequence-specific transcription factors. These proteins include the transcriptional intermediary factor 1 (TIF1) regulators, the promyelocytic leukemia tumor suppressor PML and the RET finger protein (RFP). In this chapter, we will consider the molecular interactions made by these TRIM proteins and will attempt to clarify some of the molecular mechanisms underlying their regulatory effect on transcription.
Collapse
|
113
|
Jeevan-Raj BP, Robert I, Heyer V, Page A, Wang JH, Cammas F, Alt FW, Losson R, Reina-San-Martin B. Epigenetic tethering of AID to the donor switch region during immunoglobulin class switch recombination. ACTA ACUST UNITED AC 2011; 208:1649-60. [PMID: 21746811 PMCID: PMC3149220 DOI: 10.1084/jem.20110118] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Immunoglobulin class switch recombination (CSR) is initiated by double-stranded DNA breaks (DSBs) in switch regions triggered by activation-induced cytidine deaminase (AID). Although CSR correlates with epigenetic modifications at the IgH locus, the relationship between these modifications and AID remains unknown. In this study, we show that during CSR, AID forms a complex with KAP1 (KRAB domain-associated protein 1) and HP1 (heterochromatin protein 1) that is tethered to the donor switch region (Sμ) bearing H3K9me3 (trimethylated histone H3 at lysine 9) in vivo. Furthermore, in vivo disruption of this complex results in impaired AID recruitment to Sμ, inefficient DSB formation, and a concomitant defect in CSR but not in somatic hypermutation. We propose that KAP1 and HP1 tether AID to H3K9me3 residues at the donor switch region, thus providing a mechanism linking AID to epigenetic modifications during CSR.
Collapse
Affiliation(s)
- Beena Patricia Jeevan-Raj
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Institut National de Santé et de Recherche Médicale Unité 964/Centre National de Recherche Scientifique Unité Mixte de Recherche 7104, Université de Strasbourg, 67404 Illkirch, France
| | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Abstract
In mammalian cells, multiple cellular processes, including gene silencing, cell growth and differentiation, pluripotency, neoplastic transformation, apoptosis, DNA repair, and maintenance of genomic integrity, converge on the evolutionarily conserved protein KAP1, which is thought to regulate the dynamic organization of chromatin structure via its ability to influence epigenetic patterns and chromatin compaction. In this minireview, we discuss how KAP1 might execute such pleiotropic effects, focusing on genomic targeting mechanisms, protein-protein interactions, specific post-translational modifications of both KAP1 and associated histones, and transcriptome analyses of cells deficient in KAP1.
Collapse
Affiliation(s)
- Sushma Iyengar
- From the Genetics Graduate Group, University of California, Davis, California 95616, USA
| | | |
Collapse
|
115
|
Aucagne R, Droin N, Paggetti J, Lagrange B, Largeot A, Hammann A, Bataille A, Martin L, Yan KP, Fenaux P, Losson R, Solary E, Bastie JN, Delva L. Transcription intermediary factor 1γ is a tumor suppressor in mouse and human chronic myelomonocytic leukemia. J Clin Invest 2011; 121:2361-70. [PMID: 21537084 PMCID: PMC3104753 DOI: 10.1172/jci45213] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Accepted: 03/08/2011] [Indexed: 12/27/2022] Open
Abstract
Transcription intermediary factor 1γ (TIF1γ) was suggested to play a role in erythropoiesis. However, how TIF1γ regulates the development of different blood cell lineages and whether TIF1γ is involved in human hematological malignancies remain to be determined. Here we have shown that TIF1γ was a tumor suppressor in mouse and human chronic myelomonocytic leukemia (CMML). Loss of Tif1g in mouse HSCs favored the expansion of the granulo-monocytic progenitor compartment. Furthermore, Tif1g deletion induced the age-dependent appearance of a cell-autonomous myeloproliferative disorder in mice that recapitulated essential characteristics of human CMML. TIF1γ was almost undetectable in leukemic cells of 35% of CMML patients. This downregulation was related to the hypermethylation of CpG sequences and specific histone modifications in the gene promoter. A demethylating agent restored the normal epigenetic status of the TIF1G promoter in human cells, which correlated with a reestablishment of TIF1γ expression. Together, these results demonstrate that TIF1G is an epigenetically regulated tumor suppressor gene in hematopoietic cells and suggest that changes in TIF1γ expression may be a biomarker of response to demethylating agents in CMML.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Aging/genetics
- Animals
- Antimetabolites, Antineoplastic/pharmacology
- Antimetabolites, Antineoplastic/therapeutic use
- Azacitidine/analogs & derivatives
- Azacitidine/pharmacology
- Azacitidine/therapeutic use
- Base Sequence
- Cell Differentiation
- DNA Methylation
- Decitabine
- Female
- Gene Expression Regulation, Leukemic
- Genes, Tumor Suppressor
- Hematopoiesis/genetics
- Hematopoiesis/physiology
- Hematopoietic Stem Cells/pathology
- Humans
- Leukemia, Myelomonocytic, Chronic/drug therapy
- Leukemia, Myelomonocytic, Chronic/genetics
- Leukemia, Myelomonocytic, Chronic/pathology
- Male
- Mice
- Mice, Knockout
- Middle Aged
- Molecular Sequence Data
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- Promoter Regions, Genetic
- Receptor, Macrophage Colony-Stimulating Factor/biosynthesis
- Receptor, Macrophage Colony-Stimulating Factor/genetics
- Specific Pathogen-Free Organisms
- Transcription Factors/deficiency
- Transcription Factors/genetics
- Transcription Factors/physiology
Collapse
Affiliation(s)
- Romain Aucagne
- Inserm UMR 866, University of Burgundy, Dijon, France.
IFR “Santé-STIC,” University of Burgundy, Dijon, France.
Inserm UMR 1009, Integrated Research Cancer Institute Villejuif (IRCIV), Institut Gustave Roussy, Villejuif, France.
Flow Cytometry Facility,
Cellular Imagery Facility, and
Department of Pathology, University Hospital, Dijon, France.
Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics, CNRS UMR 7104, Inserm U964, Louis Pasteur University, Collège de France, Illkirch, France.
University Hospital, Assistance Publique–Hôpitaux de Paris (AP-HP) and University of Paris 13, Bobigny, France.
University Hospital, Clinical Hematology Department, Dijon, France
| | - Nathalie Droin
- Inserm UMR 866, University of Burgundy, Dijon, France.
IFR “Santé-STIC,” University of Burgundy, Dijon, France.
Inserm UMR 1009, Integrated Research Cancer Institute Villejuif (IRCIV), Institut Gustave Roussy, Villejuif, France.
Flow Cytometry Facility,
Cellular Imagery Facility, and
Department of Pathology, University Hospital, Dijon, France.
Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics, CNRS UMR 7104, Inserm U964, Louis Pasteur University, Collège de France, Illkirch, France.
University Hospital, Assistance Publique–Hôpitaux de Paris (AP-HP) and University of Paris 13, Bobigny, France.
University Hospital, Clinical Hematology Department, Dijon, France
| | - Jérôme Paggetti
- Inserm UMR 866, University of Burgundy, Dijon, France.
IFR “Santé-STIC,” University of Burgundy, Dijon, France.
Inserm UMR 1009, Integrated Research Cancer Institute Villejuif (IRCIV), Institut Gustave Roussy, Villejuif, France.
Flow Cytometry Facility,
Cellular Imagery Facility, and
Department of Pathology, University Hospital, Dijon, France.
Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics, CNRS UMR 7104, Inserm U964, Louis Pasteur University, Collège de France, Illkirch, France.
University Hospital, Assistance Publique–Hôpitaux de Paris (AP-HP) and University of Paris 13, Bobigny, France.
University Hospital, Clinical Hematology Department, Dijon, France
| | - Brice Lagrange
- Inserm UMR 866, University of Burgundy, Dijon, France.
IFR “Santé-STIC,” University of Burgundy, Dijon, France.
Inserm UMR 1009, Integrated Research Cancer Institute Villejuif (IRCIV), Institut Gustave Roussy, Villejuif, France.
Flow Cytometry Facility,
Cellular Imagery Facility, and
Department of Pathology, University Hospital, Dijon, France.
Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics, CNRS UMR 7104, Inserm U964, Louis Pasteur University, Collège de France, Illkirch, France.
University Hospital, Assistance Publique–Hôpitaux de Paris (AP-HP) and University of Paris 13, Bobigny, France.
University Hospital, Clinical Hematology Department, Dijon, France
| | - Anne Largeot
- Inserm UMR 866, University of Burgundy, Dijon, France.
IFR “Santé-STIC,” University of Burgundy, Dijon, France.
Inserm UMR 1009, Integrated Research Cancer Institute Villejuif (IRCIV), Institut Gustave Roussy, Villejuif, France.
Flow Cytometry Facility,
Cellular Imagery Facility, and
Department of Pathology, University Hospital, Dijon, France.
Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics, CNRS UMR 7104, Inserm U964, Louis Pasteur University, Collège de France, Illkirch, France.
University Hospital, Assistance Publique–Hôpitaux de Paris (AP-HP) and University of Paris 13, Bobigny, France.
University Hospital, Clinical Hematology Department, Dijon, France
| | - Arlette Hammann
- Inserm UMR 866, University of Burgundy, Dijon, France.
IFR “Santé-STIC,” University of Burgundy, Dijon, France.
Inserm UMR 1009, Integrated Research Cancer Institute Villejuif (IRCIV), Institut Gustave Roussy, Villejuif, France.
Flow Cytometry Facility,
Cellular Imagery Facility, and
Department of Pathology, University Hospital, Dijon, France.
Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics, CNRS UMR 7104, Inserm U964, Louis Pasteur University, Collège de France, Illkirch, France.
University Hospital, Assistance Publique–Hôpitaux de Paris (AP-HP) and University of Paris 13, Bobigny, France.
University Hospital, Clinical Hematology Department, Dijon, France
| | - Amandine Bataille
- Inserm UMR 866, University of Burgundy, Dijon, France.
IFR “Santé-STIC,” University of Burgundy, Dijon, France.
Inserm UMR 1009, Integrated Research Cancer Institute Villejuif (IRCIV), Institut Gustave Roussy, Villejuif, France.
Flow Cytometry Facility,
Cellular Imagery Facility, and
Department of Pathology, University Hospital, Dijon, France.
Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics, CNRS UMR 7104, Inserm U964, Louis Pasteur University, Collège de France, Illkirch, France.
University Hospital, Assistance Publique–Hôpitaux de Paris (AP-HP) and University of Paris 13, Bobigny, France.
University Hospital, Clinical Hematology Department, Dijon, France
| | - Laurent Martin
- Inserm UMR 866, University of Burgundy, Dijon, France.
IFR “Santé-STIC,” University of Burgundy, Dijon, France.
Inserm UMR 1009, Integrated Research Cancer Institute Villejuif (IRCIV), Institut Gustave Roussy, Villejuif, France.
Flow Cytometry Facility,
Cellular Imagery Facility, and
Department of Pathology, University Hospital, Dijon, France.
Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics, CNRS UMR 7104, Inserm U964, Louis Pasteur University, Collège de France, Illkirch, France.
University Hospital, Assistance Publique–Hôpitaux de Paris (AP-HP) and University of Paris 13, Bobigny, France.
University Hospital, Clinical Hematology Department, Dijon, France
| | - Kai-Ping Yan
- Inserm UMR 866, University of Burgundy, Dijon, France.
IFR “Santé-STIC,” University of Burgundy, Dijon, France.
Inserm UMR 1009, Integrated Research Cancer Institute Villejuif (IRCIV), Institut Gustave Roussy, Villejuif, France.
Flow Cytometry Facility,
Cellular Imagery Facility, and
Department of Pathology, University Hospital, Dijon, France.
Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics, CNRS UMR 7104, Inserm U964, Louis Pasteur University, Collège de France, Illkirch, France.
University Hospital, Assistance Publique–Hôpitaux de Paris (AP-HP) and University of Paris 13, Bobigny, France.
University Hospital, Clinical Hematology Department, Dijon, France
| | - Pierre Fenaux
- Inserm UMR 866, University of Burgundy, Dijon, France.
IFR “Santé-STIC,” University of Burgundy, Dijon, France.
Inserm UMR 1009, Integrated Research Cancer Institute Villejuif (IRCIV), Institut Gustave Roussy, Villejuif, France.
Flow Cytometry Facility,
Cellular Imagery Facility, and
Department of Pathology, University Hospital, Dijon, France.
Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics, CNRS UMR 7104, Inserm U964, Louis Pasteur University, Collège de France, Illkirch, France.
University Hospital, Assistance Publique–Hôpitaux de Paris (AP-HP) and University of Paris 13, Bobigny, France.
University Hospital, Clinical Hematology Department, Dijon, France
| | - Régine Losson
- Inserm UMR 866, University of Burgundy, Dijon, France.
IFR “Santé-STIC,” University of Burgundy, Dijon, France.
Inserm UMR 1009, Integrated Research Cancer Institute Villejuif (IRCIV), Institut Gustave Roussy, Villejuif, France.
Flow Cytometry Facility,
Cellular Imagery Facility, and
Department of Pathology, University Hospital, Dijon, France.
Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics, CNRS UMR 7104, Inserm U964, Louis Pasteur University, Collège de France, Illkirch, France.
University Hospital, Assistance Publique–Hôpitaux de Paris (AP-HP) and University of Paris 13, Bobigny, France.
University Hospital, Clinical Hematology Department, Dijon, France
| | - Eric Solary
- Inserm UMR 866, University of Burgundy, Dijon, France.
IFR “Santé-STIC,” University of Burgundy, Dijon, France.
Inserm UMR 1009, Integrated Research Cancer Institute Villejuif (IRCIV), Institut Gustave Roussy, Villejuif, France.
Flow Cytometry Facility,
Cellular Imagery Facility, and
Department of Pathology, University Hospital, Dijon, France.
Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics, CNRS UMR 7104, Inserm U964, Louis Pasteur University, Collège de France, Illkirch, France.
University Hospital, Assistance Publique–Hôpitaux de Paris (AP-HP) and University of Paris 13, Bobigny, France.
University Hospital, Clinical Hematology Department, Dijon, France
| | - Jean-Noël Bastie
- Inserm UMR 866, University of Burgundy, Dijon, France.
IFR “Santé-STIC,” University of Burgundy, Dijon, France.
Inserm UMR 1009, Integrated Research Cancer Institute Villejuif (IRCIV), Institut Gustave Roussy, Villejuif, France.
Flow Cytometry Facility,
Cellular Imagery Facility, and
Department of Pathology, University Hospital, Dijon, France.
Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics, CNRS UMR 7104, Inserm U964, Louis Pasteur University, Collège de France, Illkirch, France.
University Hospital, Assistance Publique–Hôpitaux de Paris (AP-HP) and University of Paris 13, Bobigny, France.
University Hospital, Clinical Hematology Department, Dijon, France
| | - Laurent Delva
- Inserm UMR 866, University of Burgundy, Dijon, France.
IFR “Santé-STIC,” University of Burgundy, Dijon, France.
Inserm UMR 1009, Integrated Research Cancer Institute Villejuif (IRCIV), Institut Gustave Roussy, Villejuif, France.
Flow Cytometry Facility,
Cellular Imagery Facility, and
Department of Pathology, University Hospital, Dijon, France.
Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics, CNRS UMR 7104, Inserm U964, Louis Pasteur University, Collège de France, Illkirch, France.
University Hospital, Assistance Publique–Hôpitaux de Paris (AP-HP) and University of Paris 13, Bobigny, France.
University Hospital, Clinical Hematology Department, Dijon, France
| |
Collapse
|
116
|
Transcription cofactors TRIM24, TRIM28, and TRIM33 associate to form regulatory complexes that suppress murine hepatocellular carcinoma. Proc Natl Acad Sci U S A 2011; 108:8212-7. [PMID: 21531907 DOI: 10.1073/pnas.1101544108] [Citation(s) in RCA: 172] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
TRIM24 (TIF1α), TRIM28 (TIF1β), and TRIM33 (TIF1γ) are three related cofactors belonging to the tripartite motif superfamily that interact with distinct transcription factors. TRIM24 interacts with the liganded retinoic acid (RA) receptor to repress its transcriptional activity. Germ line inactivation of TRIM24 in mice deregulates RA-signaling in hepatocytes leading to the development of hepatocellular carcinoma (HCC). Here we show that TRIM24 can be purified as at least two macromolecular complexes comprising either TRIM33 or TRIM33 and TRIM28. Somatic hepatocyte-specific inactivation of TRIM24, TRIM28, or TRIM33 all promote HCC in a cell-autonomous manner in mice. Moreover, HCC formation upon TRIM24 inactivation is strongly potentiated by further loss of TRIM33. These results demonstrate that the TIF1-related subfamily of TRIM proteins interact both physically and functionally to modulate HCC formation in mice.
Collapse
|
117
|
Bhatia N, Yang B, Xiao TZ, Peters N, Hoffmann MF, Longley BJ. Identification of novel small molecules that inhibit protein-protein interactions between MAGE and KAP-1. Arch Biochem Biophys 2011; 508:217-21. [PMID: 21277283 DOI: 10.1016/j.abb.2011.01.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Revised: 01/08/2011] [Accepted: 01/10/2011] [Indexed: 01/06/2023]
Abstract
The Class I MAGE proteins are normally expressed only in developing germ cells but are often aberrantly expressed in malignancies, particularly melanoma, making them good therapeutic targets. MAGE proteins promote tumor survival by binding to the RBCC region of KAP-1 and suppressing p53. Although, suppression of MAGE expression, by RNA interference, relieves p53 suppression and inhibits tumor growth, its therapeutic uses are limited by lack of methods for systemic delivery of small interfering RNA. To overcome this barrier, we sought to discover chemical compounds that inhibit binding between MAGE and KAP-1 proteins. Based on previously published effects of MAGE suppression, we developed a strategy for screening a small molecule library based on selective death of MAGE positive cells, activation of p53 and lack of caspase activity. We screened the Maybridge HitFinder library of compounds and eight compounds fulfilled these criteria. Seven of these compounds interfered with co-precipitation of MAGE and KAP-1, and three interfered with binding of MAGE and KAP-1 in a mammalian two hybrid assay. We now report identification of three potential compounds that interfere with MAGE/KAP-1 binding and can be developed as novel chemo-therapeutic agents for treatment of advanced melanoma and other cancers.
Collapse
Affiliation(s)
- Neehar Bhatia
- Department of Dermatology, The University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, USA.
| | | | | | | | | | | |
Collapse
|
118
|
Rowe HM, Trono D. Dynamic control of endogenous retroviruses during development. Virology 2011; 411:273-87. [PMID: 21251689 DOI: 10.1016/j.virol.2010.12.007] [Citation(s) in RCA: 210] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Accepted: 12/06/2010] [Indexed: 02/07/2023]
Abstract
Close to half of the human genome encompasses mobile genetic elements, most of which are retrotransposons. These genetic invaders are formidable evolutionary forces that have shaped the architecture of the genomes of higher organisms, with some conserving the ability to induce new integrants within their hosts' genome. Expectedly, the control of endogenous retroviruses is tight and multi-pronged. It is most crucially established in the germ line and during the first steps of embryogenesis, primarily through transcriptional mechanisms that have likely evolved under their very pressure, but are now engaged in controlling gene expression at large, notably during early development.
Collapse
Affiliation(s)
- Helen M Rowe
- National Program, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | | |
Collapse
|
119
|
Herzog M, Wendling O, Guillou F, Chambon P, Mark M, Losson R, Cammas F. TIF1β association with HP1 is essential for post-gastrulation development, but not for Sertoli cell functions during spermatogenesis. Dev Biol 2010; 350:548-58. [PMID: 21163256 DOI: 10.1016/j.ydbio.2010.12.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 11/17/2010] [Accepted: 12/06/2010] [Indexed: 12/30/2022]
Abstract
TIF1β is an essential mammalian transcriptional corepressor. It interacts with the heterochromatin proteins HP1 through a highly conserved motif, the HP1box, and we have previously shown that this interaction is essential for the differentiation of F9 cells to occur. Here we address the in vivo functions of the TIF1β-HP1 interaction, by generating mice in which the TIF1β HP1box is mutated, leading to the loss of TIF1β interaction with HP1. The effects of the mutation were monitored in two instances, where TIF1β is known to play key roles: early embryonic development and spermatogenesis. We find that mutating the HP1box of TIF1β disrupts embryonic development soon after gastrulation. This effect is likely caused by the misexpression of TIF1β targets that regulate mitotic progression and pluripotency. In contrast, in Sertoli cells, we found that the absence of TIF1β but not its mutation in the HP1box leads to a clear defect of spermatogenesis characterized by a failure of spermatid release and a testicular degeneration. These data show that the interaction between TIF1β and HP1 is essential for some but not all TIF1β functions in vivo. Furthermore, we observed that TIF1β is dispersed through the nucleoplasm of E7.0 embryos, whereas it is mainly associated with pericentromeric heterochromatin of E8.5 embryos and of Sertoli cells, an association that is lost upon TIF1β HP1box mutation. Altogether, these data provide strong evidence that nuclear organization plays key roles during early embryonic development.
Collapse
Affiliation(s)
- Marielle Herzog
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch-Cedex, France
| | | | | | | | | | | | | |
Collapse
|
120
|
Whitelaw NC, Chong S, Morgan DK, Nestor C, Bruxner TJ, Ashe A, Lambley E, Meehan R, Whitelaw E. Reduced levels of two modifiers of epigenetic gene silencing, Dnmt3a and Trim28, cause increased phenotypic noise. Genome Biol 2010; 11:R111. [PMID: 21092094 PMCID: PMC3156950 DOI: 10.1186/gb-2010-11-11-r111] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 09/30/2010] [Accepted: 11/19/2010] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Inbred individuals reared in controlled environments display considerable variance in many complex traits but the underlying cause of this intangible variation has been an enigma. Here we show that two modifiers of epigenetic gene silencing play a critical role in the process. RESULTS Inbred mice heterozygous for a null mutation in DNA methyltransferase 3a (Dnmt3a) or tripartite motif protein 28 (Trim28) show greater coefficients of variance in body weight than their wild-type littermates. Trim28 mutants additionally develop metabolic syndrome and abnormal behavior with incomplete penetrance. Genome-wide gene expression analyses identified 284 significantly dysregulated genes in Trim28 heterozygote mutants compared to wild-type mice, with Mas1, which encodes a G-protein coupled receptor implicated in lipid metabolism, showing the greatest average change in expression (7.8-fold higher in mutants). This gene also showed highly variable expression between mutant individuals. CONCLUSIONS These studies provide a molecular explanation of developmental noise in whole organisms and suggest that faithful epigenetic control of transcription is central to suppressing deleterious levels of phenotypic variation. These findings have broad implications for understanding the mechanisms underlying sporadic and complex disease in humans.
Collapse
Affiliation(s)
- Nadia C Whitelaw
- Genetics and Population Health, Queensland Institute of Medical Research, 300 Herston Road, Brisbane, Queensland 4006, Australia
- School of Medicine, University of Queensland, 288 Herston Road, Brisbane, Queensland 4001, Australia
| | - Suyinn Chong
- Genetics and Population Health, Queensland Institute of Medical Research, 300 Herston Road, Brisbane, Queensland 4006, Australia
| | - Daniel K Morgan
- Genetics and Population Health, Queensland Institute of Medical Research, 300 Herston Road, Brisbane, Queensland 4006, Australia
- School of Medicine, University of Queensland, 288 Herston Road, Brisbane, Queensland 4001, Australia
| | - Colm Nestor
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Crewe Road, Edinburgh EH4 2XU, UK
- Breakthrough Research Unit, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Timothy J Bruxner
- Genetics and Population Health, Queensland Institute of Medical Research, 300 Herston Road, Brisbane, Queensland 4006, Australia
| | | | - Eleanore Lambley
- Genetics and Population Health, Queensland Institute of Medical Research, 300 Herston Road, Brisbane, Queensland 4006, Australia
| | - Richard Meehan
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Crewe Road, Edinburgh EH4 2XU, UK
- Breakthrough Research Unit, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Emma Whitelaw
- Genetics and Population Health, Queensland Institute of Medical Research, 300 Herston Road, Brisbane, Queensland 4006, Australia
| |
Collapse
|
121
|
Abstract
The dynamics of embryonic stem cell pluripotency is orchestrated by an interplay of transcriptional and epigenetic regulation in a systematic and modular manner. While the ES cell stage is marked by multiple loci with bivalent chromatin marks that prepare genes for imminent activation on differentiation, this open chromatin conformation is tempered by repressive machinery that prevent premature expression of key developmental genes. This review serves to highlight key ES transcription factors and their known links to the epigenetic machinery via known protein complexes.
Collapse
Affiliation(s)
- Clara Y Cheong
- Stem Cell and Developmental Biology, Genome Institute of Singapore, Singapore, Singapore
| | | |
Collapse
|
122
|
Morsut L, Yan KP, Enzo E, Aragona M, Soligo SM, Wendling O, Mark M, Khetchoumian K, Bressan G, Chambon P, Dupont S, Losson R, Piccolo S. Negative control of Smad activity by ectodermin/Tif1gamma patterns the mammalian embryo. Development 2010; 137:2571-8. [PMID: 20573697 DOI: 10.1242/dev.053801] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The definition of embryonic potency and induction of specific cell fates are intimately linked to the tight control over TGFbeta signaling. Although extracellular regulation of ligand availability has received considerable attention in recent years, surprisingly little is known about the intracellular factors that negatively control Smad activity in mammalian tissues. By means of genetic ablation, we show that the Smad4 inhibitor ectodermin (Ecto, also known as Trim33 or Tif1gamma) is required to limit Nodal responsiveness in vivo. New phenotypes, which are linked to excessive Nodal activity, emerge from such a modified landscape of Smad responsiveness in both embryonic and extra-embryonic territories. In extra-embryonic endoderm, Ecto is required to confine expression of Nodal antagonists to the anterior visceral endoderm. In trophoblast cells, Ecto precisely doses Nodal activity, balancing stem cell self-renewal and differentiation. Epiblast-specific Ecto deficiency shifts mesoderm fates towards node/organizer fates, revealing the requirement of Smad inhibition for the precise allocation of cells along the primitive streak. This study unveils that intracellular negative control of Smad function by ectodermin/Tif1gamma is a crucial element in the cellular response to TGFbeta signals in mammalian tissues.
Collapse
Affiliation(s)
- Leonardo Morsut
- Department of Medical Biotechnologies, Section of Histology and Embryology, University of Padua, viale Colombo 3, 35126 Padua, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
123
|
TIF1beta regulates the pluripotency of embryonic stem cells in a phosphorylation-dependent manner. Proc Natl Acad Sci U S A 2010; 107:10926-31. [PMID: 20508149 DOI: 10.1073/pnas.0907601107] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Transcription networks composed of various transcriptional factors specifically expressed in undifferentiated embryonic stem (ES) cells have been implicated in the regulation of pluripotency in ES cells. However, the molecular mechanisms responsible for self-renewal, maintenance of pluripotency, and lineage specification during differentiation of ES cells are still unclear. The results of this study demonstrate that a phosphorylation-dependent chromatin relaxation factor, transcriptional intermediary factor-1beta (TIF1beta), is a unique regulator of the pluripotency of ES cells and regulates Oct3/4-dependent transcription in a phosphorylation-dependent manner. TIF1beta is specifically phosphorylated in pluripotent mouse ES cells at the C-terminal serine 824, which has been previously shown to induce chromatin relaxation. Phosphorylated TIF1beta is partially colocalized at the activated chromatin markers, and forms a complex with the pluripotency-specific transcription factor Oct3/4 and subunits of the switching defective/sucrose nonfermenting, ATP-dependent chromatin remodeling complex, Smarcd1 [corrected], Brg-1, and BAF155, all of which are components of an ES-specific chromatin remodeling complex, esBAF. Phosphorylated TIF1beta specifically induces ES cell-specific genes and enables prolonged maintenance of an undifferentiated state in mouse ES cells. Moreover, TIF1beta regulates the reprogramming process of somatic cells in a phosphorylation-dependent manner. Our results suggest that TIF1beta provides a phosphorylation-dependent, bidirectional platform for specific transcriptional factors and chromatin remodeling enzymes that regulate the cell differentiation process and the pluripotency of stem cells.
Collapse
|
124
|
Lorenz P, Dietmann S, Wilhelm T, Koczan D, Autran S, Gad S, Wen G, Ding G, Li Y, Rousseau-Merck MF, Thiesen HJ. The ancient mammalian KRAB zinc finger gene cluster on human chromosome 8q24.3 illustrates principles of C2H2 zinc finger evolution associated with unique expression profiles in human tissues. BMC Genomics 2010; 11:206. [PMID: 20346131 PMCID: PMC2865497 DOI: 10.1186/1471-2164-11-206] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Accepted: 03/26/2010] [Indexed: 11/17/2022] Open
Abstract
Background Expansion of multi-C2H2 domain zinc finger (ZNF) genes, including the Krüppel-associated box (KRAB) subfamily, paralleled the evolution of tetrapodes, particularly in mammalian lineages. Advances in their cataloging and characterization suggest that the functions of the KRAB-ZNF gene family contributed to mammalian speciation. Results Here, we characterized the human 8q24.3 ZNF cluster on the genomic, the phylogenetic, the structural and the transcriptome level. Six (ZNF7, ZNF34, ZNF250, ZNF251, ZNF252, ZNF517) of the seven locus members contain exons encoding KRAB domains, one (ZNF16) does not. They form a paralog group in which the encoded KRAB and ZNF protein domains generally share more similarities with each other than with other members of the human ZNF superfamily. The closest relatives with respect to their DNA-binding domain were ZNF7 and ZNF251. The analysis of orthologs in therian mammalian species revealed strong conservation and purifying selection of the KRAB-A and zinc finger domains. These findings underscore structural/functional constraints during evolution. Gene losses in the murine lineage (ZNF16, ZNF34, ZNF252, ZNF517) and potential protein truncations in primates (ZNF252) illustrate ongoing speciation processes. Tissue expression profiling by quantitative real-time PCR showed similar but distinct patterns for all tested ZNF genes with the most prominent expression in fetal brain. Based on accompanying expression signatures in twenty-six other human tissues ZNF34 and ZNF250 revealed the closest expression profiles. Together, the 8q24.3 ZNF genes can be assigned to a cerebellum, a testis or a prostate/thyroid subgroup. These results are consistent with potential functions of the ZNF genes in morphogenesis and differentiation. Promoter regions of the seven 8q24.3 ZNF genes display common characteristics like missing TATA-box, CpG island-association and transcription factor binding site (TFBS) modules. Common TFBS modules partly explain the observed expression pattern similarities. Conclusions The ZNF genes at human 8q24.3 form a relatively old mammalian paralog group conserved in eutherian mammals for at least 130 million years. The members persisted after initial duplications by undergoing subfunctionalizations in their expression patterns and target site recognition. KRAB-ZNF mediated repression of transcription might have shaped organogenesis in mammalian ontogeny.
Collapse
Affiliation(s)
- Peter Lorenz
- Institute of Immunology, University of Rostock, Schillingallee 70, 18055 Rostock, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
125
|
Groner AC, Meylan S, Ciuffi A, Zangger N, Ambrosini G, Dénervaud N, Bucher P, Trono D. KRAB-zinc finger proteins and KAP1 can mediate long-range transcriptional repression through heterochromatin spreading. PLoS Genet 2010; 6:e1000869. [PMID: 20221260 PMCID: PMC2832679 DOI: 10.1371/journal.pgen.1000869] [Citation(s) in RCA: 264] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2009] [Accepted: 02/02/2010] [Indexed: 01/05/2023] Open
Abstract
Krüppel-associated box domain-zinc finger proteins (KRAB–ZFPs) are tetrapod-specific transcriptional repressors encoded in the hundreds by the human genome. In order to explore their as yet ill-defined impact on gene expression, we developed an ectopic repressor assay, allowing the study of KRAB–mediated transcriptional regulation at hundreds of different transcriptional units. By targeting a drug-controllable KRAB–containing repressor to gene-trapping lentiviral vectors, we demonstrate that KRAB and its corepressor KAP1 can silence promoters located several tens of kilobases (kb) away from their DNA binding sites, with an efficiency which is generally higher for promoters located within 15 kb or less. Silenced promoters exhibit a loss of histone H3-acetylation, an increase in H3 lysine 9 trimethylation (H3K9me3), and a drop in RNA Pol II recruitment, consistent with a block of transcriptional initiation following the establishment of silencing marks. Furthermore, we reveal that KRAB–mediated repression is established by the long-range spreading of H3K9me3 and heterochromatin protein 1 β (HP1β) between the repressor binding site and the promoter. We confirm the biological relevance of this phenomenon by documenting KAP1–dependent transcriptional repression at an endogenous KRAB–ZFP gene cluster, where KAP1 binds to the 3′ end of genes and mediates propagation of H3K9me3 and HP1β towards their 5′ end. Together, our data support a model in which KRAB/KAP1 recruitment induces long-range repression through the spread of heterochromatin. This finding not only suggests auto-regulatory mechanisms in the control of KRAB–ZFP gene clusters, but also provides important cues for interpreting future genome-wide DNA binding data of KRAB–ZFPs and KAP1. The regulation of gene activity by transcription factors is crucial to the function of all cells. Here, we studied the mechanisms of action of the largest family of gene regulators encoded by the human genome, the so-called KRAB–containing zinc finger proteins (KRAB–ZFPs), which in concert with their universal cofactor KAP1 act as transcriptional repressors. For this, we used two parallel approaches. First, by targeting an ectopic KRAB domain to hundreds of different genes, we found that KRAB/KAP1 can repress promoters located several tens of kilobases from the repressor DNA docking site. We further could show that KRAB induces such long-range effects by mediating the spread of repressive chromatin marks along the body of the gene, resulting in a block of transcriptional initiation at the promoter. In a second set of experiments, we analyzed an endogenous KRAB–ZFP gene cluster, where we could also document KAP1–dependent heterochromatin spreading and transcriptional repression. Together, these results support a model whereby KRAB–ZFPs and KAP1 can mediate long-range transcriptional repression through the spread of silencing chromatin marks. This study thus provides insight into KRAB/KAP1–induced gene regulation at KRAB–ZFP gene clusters, and will further help interpret genome-wide studies of KRAB–ZFPs and KAP1 DNA binding patterns.
Collapse
Affiliation(s)
- Anna C. Groner
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Frontiers-in-Genetics National Center of Competence in Research, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Sylvain Meylan
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Frontiers-in-Genetics National Center of Competence in Research, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Angela Ciuffi
- Institute of Microbiology, University Hospital Center and University of Lausanne, Lausanne, Switzerland
| | - Nadine Zangger
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Frontiers-in-Genetics National Center of Competence in Research, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Giovanna Ambrosini
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Nicolas Dénervaud
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Philipp Bucher
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Didier Trono
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Frontiers-in-Genetics National Center of Competence in Research, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
126
|
Perissi V, Jepsen K, Glass CK, Rosenfeld MG. Deconstructing repression: evolving models of co-repressor action. Nat Rev Genet 2010; 11:109-23. [PMID: 20084085 DOI: 10.1038/nrg2736] [Citation(s) in RCA: 414] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A crucial aspect of development, homeostasis and prevention of disease is the strict maintenance of patterns of gene repression. Gene repression is largely achieved by the combinatorial action of various enzymatic complexes - known as co-repressor complexes - that are recruited to DNA by transcription factors and often act through enzymatic modification of histone protein tails. Our understanding of how co-repressors act has begun to change over recent years owing to the increased availability of genome-scale data. Here, we consider specific strategies that underlie repression events - for example, those mediated by the nuclear receptor co-repressor (NCoR, also known as NCOR1) and silencing mediator of retinoic acid and thyroid hormone receptor (SMRT, also known as NCOR2) co-repressor complexes - and discuss emerging themes in gene repression.
Collapse
Affiliation(s)
- Valentina Perissi
- Department of Medicine, Howard Hughes Medical Institute, School of Medicine, University of California-San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
| | | | | | | |
Collapse
|
127
|
Shigematsu Y, Hanagiri T, Shiota H, Kuroda K, Baba T, Mizukami M, So T, Ichiki Y, Yasuda M, So T, Takenoyama M, Yasumoto K. Clinical significance of cancer/testis antigens expression in patients with non-small cell lung cancer. Lung Cancer 2009; 68:105-10. [PMID: 19545928 DOI: 10.1016/j.lungcan.2009.05.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Revised: 05/12/2009] [Accepted: 05/17/2009] [Indexed: 12/15/2022]
Abstract
Cancer/testis antigens (CT antigens) are thought to be suitable targets for antigen-specific immunotherapy, because of the cancer-specific expression except for the testis among various normal tissues and no-expression of HLA class I in the testis. In the present study, the expressions of CT antigens (MAGE-A3, MAGE-A4, NY-ESO-1 and KK-LC-1) in non-small cell lung cancer (NSCLC) were analyzed by RT-PCR. The subjects were 239 patients with NSCLC who underwent surgery from 2001 to 2005 in our department. The expression rates of MAGE-A3, MAGE-A4, NY-ESO-1 and KK-LC-1 were 23.8%, 20.1%, 10.5% and 32.6% in patients with NSCLC, respectively. MAGE-A4 was expressed more frequently in male (25.3%) than in female (10.6%) (p<0.01). The positive proportion of MAGE-A4 was higher in stages II-IV (30.6%) than in stage I (12.8%) (p<0.01). Both of MAGE-A3 and MAGE-A4 were expressed more frequently in squamous cell carcinoma than in adenocarcinoma (p<0.01). Such tendency was not observed among NY-ESO-1 and KK-LC-1 expression. KK-LC-1 was expressed in 32.1% of patients with adenocarcinoma and in 36.5% of patients with squamous cell carcinoma. Patients with positive MAGE-A4 expression showed significantly poorer overall survival than those without MAGE-A4 expression (p=0.013), and such effect on survival was also observed, when the analysis was limited to patients at stage I (p=0.0037). Expression of MAGE-A3, NY-ESO-1 or KK-LC-1 did not affect survival of patients with NSCLC significantly, however, expression of at least one of such CT antigens negatively affect survival of patients with NSCLC (p=0.045).
Collapse
Affiliation(s)
- Yoshiki Shigematsu
- Second Department of Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Rambaud J, Desroches J, Balsalobre A, Drouin J. TIF1beta/KAP-1 is a coactivator of the orphan nuclear receptor NGFI-B/Nur77. J Biol Chem 2009; 284:14147-56. [PMID: 19321449 PMCID: PMC2682863 DOI: 10.1074/jbc.m809023200] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Revised: 03/25/2009] [Indexed: 01/02/2023] Open
Abstract
In efforts to define mechanisms of transcriptional activation by the orphan nuclear receptor NGFI-B (Nur77), we identified TIF1beta by mass spectrometry within a nuclear protein complex containing NGFI-B. TIF1beta, also known as KAP-1 (KRAB domain-associated protein) or KRIP-1, acts as a transcriptional corepressor for many transcription factors, in particular for the Krüppel-associated box domain-containing zinc finger transcription factors. TIF1beta is also an intrinsic component of two chromatin remodeling and histone deacetylase complexes, the N-CoR1 and nucleosome remodeling and deacetylation complexes. In contrast to these activities, we report that TIF1beta is a coactivator of NGFI-B and that it is as potent as the SRC coactivators in this context. Using pull-down assays and immunoprecipitation, we showed that TIF1beta interacts directly with NGFI-B and with other Nur family members. NGFI-B is an important mediator of hypothalamic corticotropin-releasing hormone (CRH) activation of proopiomelanocortin (POMC) transcription, and TIF1beta enhances transcription mediated through the NGFI-B target, the Nur response element (NurRE). The NurRE binds Nur factor dimers and is responsive to signaling pathways. In keeping with the role of NGFI-B as mediator of CRH signaling, we found that TIF1beta is recruited to the POMC promoter following CRH stimulation and that TIF1beta potentiates CRH and protein kinase A signaling through the NurRE; it acts synergistically with the SRC2 coactivator. However, the actions of TIF1beta and SRC2 were mapped to different NGFI-B AF-1 subdomains. Taken together, these results indicate that TIF1beta is an important coactivator of NGFI-B-dependent transcription.
Collapse
Affiliation(s)
- Juliette Rambaud
- Laboratory of Molecular Genetics, Institut de Recherches Cliniques de Montréal, Montréal, Quebec H2W 1R7, Canada
| | | | | | | |
Collapse
|
129
|
Hu G, Kim J, Xu Q, Leng Y, Orkin SH, Elledge SJ. A genome-wide RNAi screen identifies a new transcriptional module required for self-renewal. Genes Dev 2009; 23:837-48. [PMID: 19339689 DOI: 10.1101/gad.1769609] [Citation(s) in RCA: 324] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We performed a genome-wide siRNA screen in mouse embryonic stem (ES) cells to identify genes essential for self-renewal, and found 148 genes whose down-regulation caused differentiation. Many of the identified genes function in gene regulation and/or development, and are highly expressed in ES cells and embryonic tissues. We further identified target genes of two transcription regulators Cnot3 and Trim28. We discovered that Cnot3 and Trim28 co-occupy many putative gene promoters with c-Myc and Zfx, but not other pluripotency-associated transcription factors. They form a unique module in the self-renewal transcription network, separate from the core module formed by Nanog, Oct4, and Sox2. The transcriptional targets of this module are enriched for genes involved in cell cycle, cell death, and cancer. This supports the idea that regulatory networks controlling self-renewal in stem cells may also be active in certain cancers and may represent novel anti-cancer targets. Our screen has implicated over 100 new genes in ES cell self-renewal, and illustrates the power of RNAi and forward genetics for the systematic study of self-renewal.
Collapse
Affiliation(s)
- Guang Hu
- Howard Hughes Medical Institute, Department of Genetics, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
130
|
Riclet R, Chendeb M, Vonesch JL, Koczan D, Thiesen HJ, Losson R, Cammas F. Disruption of the interaction between transcriptional intermediary factor 1{beta} and heterochromatin protein 1 leads to a switch from DNA hyper- to hypomethylation and H3K9 to H3K27 trimethylation on the MEST promoter correlating with gene reactivation. Mol Biol Cell 2008; 20:296-305. [PMID: 18923144 DOI: 10.1091/mbc.e08-05-0510] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Here, we identified the imprinted mesoderm-specific transcript (MEST) gene as an endogenous TIF1beta primary target gene and demonstrated that transcriptional intermediary factor (TIF) 1beta, through its interaction with heterochromatin protein (HP) 1, is essential in establishing and maintaining a local heterochromatin-like structure on MEST promoter region characterized by H3K9 trimethylation and hypoacetylation, H4K20 trimethylation, DNA hypermethylation, and enrichment in HP1 that correlates with preferential association to foci of pericentromeric heterochromatin and transcriptional repression. On disruption of the interaction between TIF1beta and HP1, TIF1beta is released from the promoter region, and there is a switch from DNA hypermethylation and histone H3K9 trimethylation to DNA hypomethylation and histone H3K27 trimethylation correlating with rapid reactivation of MEST expression. Interestingly, we provide evidence that the imprinted MEST allele DNA methylation is insensitive to TIF1beta loss of function, whereas the nonimprinted allele is regulated through a distinct TIF1beta-DNA methylation mechanism.
Collapse
Affiliation(s)
- Raphaël Riclet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique/Institut National de la Santé et de la Recherche Médicale/Université Louis Pasteur/Collège de France, Illkirch-Cedex, France
| | | | | | | | | | | | | |
Collapse
|
131
|
TRIM28 mediates primer binding site-targeted silencing of Lys1,2 tRNA-utilizing retroviruses in embryonic cells. Proc Natl Acad Sci U S A 2008; 105:12521-6. [PMID: 18713861 DOI: 10.1073/pnas.0805540105] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Murine leukemia viruses (MLVs) and related retroelements are potently restricted in embryonic cells by postintegration transcriptional silencing, likely to protect the germ line from insertional mutagenesis. This silencing is in large part attributable to the presence of a nuclear repression complex, which targets a sequence element of the proviral DNA, the repressor-binding site. The repressor-binding site closely overlaps the tRNA primer binding site, a highly conserved sequence essential for virus replication and defining the site of initiation of DNA synthesis during reverse transcription. We have recently demonstrated that the cellular corepressor TRIM28 is recruited to the proline tRNA primer-binding site used by many MLVs and is required to mediate this silencing. Here, we show that TRIM28 is also required for the restriction of retroviruses using a completely distinct tRNA for the priming of their DNA synthesis, namely Lys-1,2 tRNA. These results generalize the role of TRIM28 in retroviral restriction and suggest that this system has evolved to restrict multiple retroviruses.
Collapse
|
132
|
Abstract
Trim33 (Tif1gamma, ectodermin, moonshine), a member of the TIF1 family of transcriptional coactivators and corepressors, is a large nuclear protein that contains an N-terminal tripartite (Trim) domain composed of a RING domain, two B-box domains, and a coiled coil domain. It has been suggested that Trim33 (Ectodermin) mediates ectodermal induction in the Xenopus by functioning as a Smad4 ubiquitin ligase, while in the zebrafish Trim33 (moonshine) has been reported to act as a R-Smad binding protein in induction of erythroid differentiation. Since the developmental role of Trim33 in mammals is currently unknown, we generated mice carrying the conditional Trim33 (Trim33(FX)) allele by flanking exons 2-4 encoding most of the functionally critical N-terminal tripartite domain by loxP sites. We confirmed the null genotype by using the EIIa-Cre transgenic approach to create mice that lack exons 2-4. Embryos deficient in Trim33 die during early somitogenesis, demonstrating that Trim33 plays an important nonredundant role in mammalian embryonic development.
Collapse
Affiliation(s)
- Jieun Kim
- Developmental Biology Program, The Saban Research Institute of Childrens Hospital Los Angeles, Department of Pathology, The Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | | |
Collapse
|
133
|
Quénet D, Gasser V, Fouillen L, Cammas F, Sanglier‐Cianferani S, Losson R, Dantzer F. The histone subcode: poly(ADP‐ribose) polymerase‐1 (Parp‐1) and Parp‐2 control cell differentiation by regulating the transcriptional intermediary factor TIF1β and the heterochromatin protein HPlα. FASEB J 2008; 22:3853-65. [DOI: 10.1096/fj.08-113464] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Delphine Quénet
- Département Intégrité du GénomeUMR7175, Ecole Supérieure de Biotechnologie de StrasbourgIllkirchFrance
| | - Véronique Gasser
- Département Intégrité du GénomeUMR7175, Ecole Supérieure de Biotechnologie de StrasbourgIllkirchFrance
| | - Laetitia Fouillen
- Laboratoire de Spectrométrie de Masse Bio‐organiqueUMR7178, Ecole de Chimie, Polyméres et MatériauxStrasbourgFrance
| | - Florence Cammas
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
| | - Sarah Sanglier‐Cianferani
- Laboratoire de Spectrométrie de Masse Bio‐organiqueUMR7178, Ecole de Chimie, Polyméres et MatériauxStrasbourgFrance
| | - Régine Losson
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
| | - Francoise Dantzer
- Département Intégrité du GénomeUMR7175, Ecole Supérieure de Biotechnologie de StrasbourgIllkirchFrance
| |
Collapse
|
134
|
Yang B, O'Herrin SM, Wu J, Reagan-Shaw S, Ma Y, Bhat KMR, Gravekamp C, Setaluri V, Peters N, Hoffmann FM, Peng H, Ivanov AV, Simpson AJG, Longley BJ. MAGE-A, mMage-b, and MAGE-C proteins form complexes with KAP1 and suppress p53-dependent apoptosis in MAGE-positive cell lines. Cancer Res 2007; 67:9954-62. [PMID: 17942928 DOI: 10.1158/0008-5472.can-07-1478] [Citation(s) in RCA: 219] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The MAGE-A, MAGE-B, and MAGE-C protein families comprise the class-I MAGE/cancer testes antigens, a group of highly homologous proteins whose expression is suppressed in all normal tissues except developing sperm. Aberrant expression of class I MAGE proteins occurs in melanomas and many other malignancies, and MAGE proteins have long been recognized as tumor-specific targets; however, their functions have largely been unknown. Here, we show that suppression of class I MAGE proteins induces apoptosis in the Hs-294T, A375, and S91 MAGE-positive melanoma cell lines and that members of all three families of MAGE class I proteins form complexes with KAP1, a scaffolding protein that is known as a corepressor of p53 expression and function. In addition to inducing apoptosis, MAGE suppression decreases KAP1 complexing with p53, increases immunoreactive and acetylated p53, and activates a p53 responsive reporter gene. Suppression of class I MAGE proteins also induces apoptosis in MAGE-A-positive, p53wt/wt parental HCT 116 colon cancer cells but not in a MAGE-A-positive HCT 116 p53-/- variant, indicating that MAGE suppression of apoptosis requires p53. Finally, treatment with MAGE-specific small interfering RNA suppresses S91 melanoma growth in vivo, in syngenic DBA2 mice. Thus, class I MAGE protein expression may suppress apoptosis by suppressing p53 and may actively contribute to the development of malignancies and by promoting tumor survival. Because the expression of class I MAGE proteins is limited in normal tissues, inhibition of MAGE antigen expression or function represents a novel and specific treatment for melanoma and diverse malignancies.
Collapse
Affiliation(s)
- Bing Yang
- Department of Dermatology and Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin Medical School, Madison, Wisconsin 53706, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Wiznerowicz M, Jakobsson J, Szulc J, Liao S, Quazzola A, Beermann F, Aebischer P, Trono D. The Kruppel-associated box repressor domain can trigger de novo promoter methylation during mouse early embryogenesis. J Biol Chem 2007; 282:34535-41. [PMID: 17893143 DOI: 10.1074/jbc.m705898200] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The Krüppel-associated box (KRAB) domain is a transcriptional repression module responsible for the DNA binding-dependent gene silencing activity of hundreds of vertebrate zinc finger proteins. We previously exploited KRAB-mediated repression within the context of a tet repressor-KRAB fusion protein and of lentiviral vectors to create a method of external gene control. We demonstrated that with this system transcriptional silencing was fully reversible in cell culture as well as in vivo. Here we reveal that, in sharp contrast, KRAB-mediated repression results in irreversible gene silencing through promoter DNA methylation if it acts during the first few days of mouse development.
Collapse
Affiliation(s)
- Maciej Wiznerowicz
- School of Life Sciences, "Frontiers in Genetics" National Center for Competence in Research, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
136
|
Wang C, Rauscher FJ, Cress WD, Chen J. Regulation of E2F1 function by the nuclear corepressor KAP1. J Biol Chem 2007; 282:29902-9. [PMID: 17704056 DOI: 10.1074/jbc.m704757200] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
KAP1 is a nuclear corepressor with conserved domains for RING finger, B boxes, leucine zipper alpha helical coiled-coil region, plant homeo domain finger, and bromo domain. The plant homeo domain finger and bromo domain of KAP1 cooperatively function as a transcription repression domain by recruiting the histone deacetylase complex NuRD and histone H3 lysine 9-specific methyltransferase SETDB1. Here we report that KAP1 binds the E2F1 transcription factor in a retinoblastoma protein (pRb)-independent fashion and inhibits E2F1 activity. KAP1 stimulates formation of E2F1-HDAC1 complex and inhibits E2F1 acetylation. Ectopic expression of KAP1 represses E2F1 transcription and apoptosis functions independent of pRb. Depletion of endogenous KAP1 in pRb-deficient Saos2 cells by RNA interference increases E2F1 acetylation level, stimulates E2F1 transcriptional activity, and sensitizes apoptosis response to DNA damage. Therefore, KAP1 contributes to the negative regulation of E2F1 and may serve as a partial backup to prevent E2F1-mediated apoptosis in the absence of pRb.
Collapse
Affiliation(s)
- Chuangui Wang
- Molecular Oncology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| | | | | | | |
Collapse
|
137
|
Mascle XH, Germain-Desprez D, Huynh P, Estephan P, Aubry M. Sumoylation of the transcriptional intermediary factor 1beta (TIF1beta), the Co-repressor of the KRAB Multifinger proteins, is required for its transcriptional activity and is modulated by the KRAB domain. J Biol Chem 2007; 282:10190-202. [PMID: 17298944 DOI: 10.1074/jbc.m611429200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Small ubiquitin-related modifier (SUMO) has emerged as a key post-translational modulator of protein functions. Here we show that TIF1beta, a developmental regulator proposed to act as a universal co-repressor for the large family of KRAB domain-containing zinc finger proteins, is a heavily SUMO-modified substrate. A combined analysis of deletion and punctual mutants identified TIF1beta as a multilysine acceptor for SUMO which specifically targets six lysine residues (Lys(554), Lys(575), Lys(676), Lys(750), Lys(779), and Lys(804)) within the TIF1beta C-terminal repressive region. Reporter gene assays indicate that TIF1beta requires SUMO-modification for its repressive activity. Indeed, sumoylation-less mutants failed to recapitulate TIF1beta-dependent repression. TIF1beta homodimerization properties and interaction with the KRAB domain are preserved in the mutants with lysine to arginine substitutions as confirmed by in vivo bioluminescence resonance energy transfer (BRET). Using histone deacetylase (HDAC) inhibitors, we also demonstrate that TIF1beta sumoylation is a prerequisite for the recruitment of HDAC and that TIF1beta SUMO-dependent repressive activity involves both HDAC-dependent and HDAC-independent components. Finally, we report that, in addition to relying on the integrity of its PHD finger and on its self-oligomerization, TIF1beta sumoylation is positively regulated by its interaction with KRAB domain-containing proteins. Altogether, our results provide new mechanistic insights into TIF1beta transcriptional repression and suggest that KRAB multifinger proteins not only recruit TIF1beta co-repressor to target genes but also increase its repressive activity through enhancement of its sumoylation.
Collapse
Affiliation(s)
- Xavier H Mascle
- Department of Biochemistry, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | | | | | | | | |
Collapse
|
138
|
Meroni G, Diez-Roux G. TRIM/RBCC, a novel class of 'single protein RING finger' E3 ubiquitin ligases. Bioessays 2006; 27:1147-57. [PMID: 16237670 DOI: 10.1002/bies.20304] [Citation(s) in RCA: 569] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The TRIM/RBCC proteins are defined by the presence of the tripartite motif composed of a RING domain, one or two B-box motifs and a coiled-coil region. These proteins are involved in a plethora of cellular processes such as apoptosis, cell cycle regulation and viral response. Consistently, their alteration results in many diverse pathological conditions. The highly conserved modular structure of these proteins suggests that a common biochemical function may underlie their assorted cellular roles. Here, we review recent data indicating that some TRIM/RBCC proteins are implicated in ubiquitination and propose that this large protein family represents a novel class of 'single protein RING finger' ubiquitin E3 ligases.
Collapse
Affiliation(s)
- Germana Meroni
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy.
| | | |
Collapse
|
139
|
Torres-Padilla ME, Zernicka-Goetz M. Role of TIF1alpha as a modulator of embryonic transcription in the mouse zygote. ACTA ACUST UNITED AC 2006; 174:329-38. [PMID: 16880268 PMCID: PMC2064229 DOI: 10.1083/jcb.200603146] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The first events of the development of any embryo are under maternal control until the zygotic genome becomes activated. In the mouse embryo, the major wave of transcription activation occurs at the 2-cell stage, but transcription starts already at the zygote (1-cell) stage. Very little is known about the molecules involved in this process. We show that the transcription intermediary factor 1 α (TIF1α) is involved in modulating gene expression during the first wave of transcription activation. At the onset of genome activation, TIF1α translocates from the cytoplasm into the pronuclei to sites of active transcription. These sites are enriched with the chromatin remodelers BRG-1 and SNF2H. When we ablate TIF1α through either RNA interference (RNAi) or microinjection of specific antibodies into zygotes, most of the embryos arrest their development at the 2–4-cell stage transition. The ablation of TIF1α leads to mislocalization of RNA polymerase II and the chromatin remodelers SNF2H and BRG-1. Using a chromatin immunoprecipitation cloning approach, we identify genes that are regulated by TIF1α in the zygote and find that transcription of these genes is misregulated upon TIF1α ablation. We further show that the expression of some of these genes is dependent on SNF2H and that RNAi for SNF2H compromises development, suggesting that TIF1α mediates activation of gene expression in the zygote via SNF2H. These studies indicate that TIF1α is a factor that modulates the expression of a set of genes during the first wave of genome activation in the mouse embryo.
Collapse
Affiliation(s)
- Maria Elena Torres-Padilla
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QR, England, UK
| | | |
Collapse
|
140
|
Ziv Y, Bielopolski D, Galanty Y, Lukas C, Taya Y, Schultz DC, Lukas J, Bekker-Jensen S, Bartek J, Shiloh Y. Chromatin relaxation in response to DNA double-strand breaks is modulated by a novel ATM- and KAP-1 dependent pathway. Nat Cell Biol 2006; 8:870-6. [PMID: 16862143 DOI: 10.1038/ncb1446] [Citation(s) in RCA: 573] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2006] [Accepted: 07/04/2006] [Indexed: 12/13/2022]
Abstract
The cellular DNA-damage response is a signaling network that is vigorously activated by cytotoxic DNA lesions, such as double-strand breaks (DSBs). The DSB response is mobilized by the nuclear protein kinase ATM, which modulates this process by phosphorylating key players in these pathways. A long-standing question in this field is whether DSB formation affects chromatin condensation. Here, we show that DSB formation is followed by ATM-dependent chromatin relaxation. ATM's effector in this pathway is the protein KRAB-associated protein (KAP-1, also known as TIF1beta, KRIP-1 or TRIM28), previously known as a corepressor of gene transcription. In response to DSB induction, KAP-1 is phosphorylated in an ATM-dependent manner on Ser 824. KAP-1 is phosphorylated exclusively at the damage sites, from which phosphorylated KAP-1 spreads rapidly throughout the chromatin. Ablation of the phosphorylation site of KAP-1 leads to loss of DSB-induced chromatin decondensation and renders the cells hypersensitive to DSB-inducing agents. Knocking down KAP-1, or mimicking a constitutive phosphorylation of this protein, leads to constitutive chromatin relaxation. These results suggest that chromatin relaxation is a fundamental pathway in the DNA-damage response and identify its primary mediators.
Collapse
Affiliation(s)
- Yael Ziv
- The David and Inez Myers Laboratory for Genetic Research, Department of Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Liu J, Wang J, Huang Q, Higdon J, Magdaleno S, Curran T, Zuo J. Gene expression profiles of mouse retinas during the second and third postnatal weeks. Brain Res 2006; 1098:113-25. [PMID: 16777074 DOI: 10.1016/j.brainres.2006.04.086] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2005] [Revised: 04/20/2006] [Accepted: 04/21/2006] [Indexed: 10/24/2022]
Abstract
Mouse retina undergoes crucial changes during early postnatal development. By using Affymetrix microarrays, we analyzed gene expression profiles of wild-type 129SvEv/C57BL/6 mouse retinas at postnatal days (P) 7, 10, 14, 18, and 21 and found significantly altered expression of 355 genes. Characterization of these 355 genes provided insight into physiologic and pathologic processes of mouse retinal development during the second and third postnatal weeks, a period that corresponds to human embryogenesis between weeks 12 and 28. These genes formed 6 groups with similar change patterns. Among the genes, sixteen cause retinal diseases when mutated; most of these 16 genes were upregulated in retina during this period. Using the PathArt program, we identified the biological processes in which many of the 355 gene products function. Among the most active processes in the P7-P21 retina are those involved in neurogenesis, obesity, diabetes type II, apoptosis, growth and differentiation, and protein kinase activity. We examined the expression patterns of 58 genes in P7 and adult retinas by searching the Brain Gene Expression Map database. Although most genes were present in various cell types in retinas, many displayed high levels of expression specifically in the outer nuclear, inner nuclear, and/or ganglion cell layers. By combining our 3 analyses, we demonstrated that during this period of mouse retinal development, many genes play important roles in various cell types, multiple pathways are involved, and some genes in a pathway are expressed in coordinated patterns. Our results thus provide foundation for future detailed studies of specific genes and pathways in various genetic and environmental conditions during retinal development.
Collapse
Affiliation(s)
- Jiewu Liu
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | | | | | | | | | |
Collapse
|
142
|
Duval D, Trouillas M, Thibault C, Dembelé D, Diemunsch F, Reinhardt B, Mertz AL, Dierich A, Boeuf H. Apoptosis and differentiation commitment: novel insights revealed by gene profiling studies in mouse embryonic stem cells. Cell Death Differ 2006; 13:564-75. [PMID: 16311515 DOI: 10.1038/sj.cdd.4401789] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Mouse embryonic stem (ES) cells remain pluripotent in vitro when grown in the presence of leukemia inhibitory factor (LIF). LIF starvation leads to apoptosis of some of the ES-derived differentiated cells, together with p38alpha mitogen-activated protein kinase (MAPK) activation. Apoptosis, but not morphological cell differentiation, is blocked by a p38 inhibitor, PD169316. To further understand the mechanism of action of this compound, we have identified its specific targets by microarray studies. We report on the global expression profiles of genes expressed at 3 days upon LIF withdrawal (d3) compared to pluripotent cells and of genes whose expression is modulated at d3 under anti-apoptotic conditions. We showed that at d3 without LIF cells express, earlier than anticipated, specialized cell markers and that when the apoptotic process was impaired, expression of differentiation markers was altered. In addition, functional tests revealed properties of anti-apoptotic proteins not to alter cell pluripotency and a novel role for metallothionein 1 gene, which prevents apoptosis of early differentiated cells.
Collapse
Affiliation(s)
- D Duval
- UMR5096-CNRS/UP/IRD, Perpignan, France
| | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Bour G, Taneja R, Rochette‐Egly C. Mouse embryocarcinoma F9 cells and retinoic acid: A model to study the molecular mechanisms of endodermal differentiation. NUCLEAR RECEPTORS IN DEVELOPMENT 2006. [DOI: 10.1016/s1574-3349(06)16007-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
144
|
Wang C, Ivanov A, Chen L, Fredericks WJ, Seto E, Rauscher FJ, Chen J. MDM2 interaction with nuclear corepressor KAP1 contributes to p53 inactivation. EMBO J 2005; 24:3279-90. [PMID: 16107876 PMCID: PMC1224681 DOI: 10.1038/sj.emboj.7600791] [Citation(s) in RCA: 197] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2005] [Accepted: 07/29/2005] [Indexed: 01/08/2023] Open
Abstract
MDM2 is a RING domain ubiquitin E3 ligase and a major regulator of the p53 tumor suppressor. MDM2 binds to p53, inactivates p53 transcription function, inhibits p53 acetylation, and promotes p53 degradation. Here, we present evidence that MDM2 interacts with the nuclear corepressor KAP1. The binding is mediated by the N-terminal coiled-coil domain of KAP1 and the central acidic domain of MDM2. KAP1 stimulates formation of p53-HDAC1 complex and inhibits p53 acetylation by interacting with MDM2. Expression of KAP1 cooperates with MDM2 to promote p53 ubiquitination and degradation. The tumor suppressor ARF competes with KAP1 in MDM2 binding; oncogene induction of ARF expression reduces MDM2-KAP1 interaction. Depletion of endogenous KAP1 expression by RNAi stimulates p53 transcriptional activity, sensitizes p53 response to DNA damage, and increases apoptosis. Therefore, MDM2 interaction with KAP1 contributes to p53 functional regulation. ARF may regulate p53 acetylation and stability in part by inhibiting KAP1-MDM2 binding.
Collapse
Affiliation(s)
- Chuangui Wang
- Molecular Oncology Program, H Lee Moffitt Comprehensive Cancer Center and Research Institute, Tampa, FL, USA
| | | | - Lihong Chen
- Molecular Oncology Program, H Lee Moffitt Comprehensive Cancer Center and Research Institute, Tampa, FL, USA
| | | | - Ed Seto
- Molecular Oncology Program, H Lee Moffitt Comprehensive Cancer Center and Research Institute, Tampa, FL, USA
| | | | - Jiandong Chen
- Molecular Oncology Program, H Lee Moffitt Comprehensive Cancer Center and Research Institute, Tampa, FL, USA
- Molecular Oncology Program, H Lee Moffitt Cancer Center, MRC3057A, 12902 Magnolia Drive, Tampa, FL 33612, USA. Tel.: +1 813 903 6822; Fax: +1 813 903 6817; E-mail:
| |
Collapse
|
145
|
Schuler M, Dierich A, Chambon P, Metzger D. Efficient temporally controlled targeted somatic mutagenesis in hepatocytes of the mouse. Genesis 2005; 39:167-72. [PMID: 15282742 DOI: 10.1002/gene.20039] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
146
|
Beckstead RB, Ner SS, Hales KG, Grigliatti TA, Baker BS, Bellen HJ. Bonus, a Drosophila TIF1 homolog, is a chromatin-associated protein that acts as a modifier of position-effect variegation. Genetics 2005; 169:783-94. [PMID: 15545640 PMCID: PMC1449102 DOI: 10.1534/genetics.104.037085] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2004] [Accepted: 11/10/2004] [Indexed: 11/18/2022] Open
Abstract
Bonus, a Drosophila TIF1 homolog, is a nuclear receptor cofactor required for viability, molting, and numerous morphological events. Here we establish a role for Bonus in the modulation of chromatin structure. We show that weak loss-of-function alleles of bonus have a more deleterious effect on males than on females. This male-enhanced lethality is not due to a defect in dosage compensation or somatic sex differentiation, but to the presence of the Y chromosome. Additionally, we show that bonus acts as both an enhancer and a suppressor of position-effect variegation. By immunostaining, we demonstrate that Bonus is associated with both interphase and prophase chromosomes and through chromatin immunoprecipitation show that two of these sites correspond to the histone gene cluster and the Stellate locus.
Collapse
Affiliation(s)
- R B Beckstead
- Department of Molecular and Human Genetics, Howard Hughes Medical Institute, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
147
|
Cammas F, Herzog M, Lerouge T, Chambon P, Losson R. Association of the transcriptional corepressor TIF1beta with heterochromatin protein 1 (HP1): an essential role for progression through differentiation. Genes Dev 2004; 18:2147-60. [PMID: 15342492 PMCID: PMC515292 DOI: 10.1101/gad.302904] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The transcriptional intermediary factor 1beta (TIF1beta) is a corepressor for KRAB-domain-containing zinc finger proteins and is believed to play essential roles in cell physiology by regulating chromatin organization at specific loci through association with chromatin remodeling and histone-modifying activities and recruitment of heterochromatin protein 1 (HP1) proteins. In this study, we have engineered a modified embryonal carcinoma F9 cell line (TIF1beta(HP1box/-)) expressing a mutated TIF1beta protein (TIF1beta(HP1box)) unable to interact with HP1 proteins. Phenotypic analysis of TIF1beta(HP1box/-) and TIF1beta(+/-) cells shows that TIF1beta-HP1 interaction is not required for differentiation of F9 cells into primitive endoderm-like (PrE) cells on retinoic acid (RA) treatment but is essential for further differentiation into parietal endoderm-like (PE) cells on addition of cAMP and for differentiation into visceral endoderm-like cells on treatment of vesicles with RA. Complementation experiments reveal that TIF1beta-HP1 interaction is essential only during a short window of time within early differentiating PrE cells to establish a selective transmittable competence to terminally differentiate on further cAMP inducing signal. Moreover, the expression of three endoderm-specific genes, GATA6, HNF4, and Dab2, is down-regulated in TIF1beta(HP1box/-) cells compared with wild-type cells during PrE differentiation. Collectively, these data demonstrate that the interaction between TIF1beta and HP1 proteins is essential for progression through differentiation by regulating the expression of endoderm differentiation master players.
Collapse
Affiliation(s)
- Florence Cammas
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP/Collège de France, BP10142, 67404 Illkirch, France
| | | | | | | | | |
Collapse
|
148
|
Yan KP, Dollé P, Mark M, Lerouge T, Wendling O, Chambon P, Losson R. Molecular cloning, genomic structure, and expression analysis of the mouse transcriptional intermediary factor 1 gamma gene. Gene 2004; 334:3-13. [PMID: 15256250 DOI: 10.1016/j.gene.2004.02.056] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2003] [Revised: 02/19/2004] [Accepted: 02/27/2004] [Indexed: 12/31/2022]
Abstract
Human transcriptional intermediary factor 1 gamma (Tif1gamma), also known as Ret fused gene 7 (RFG7), is a member of a novel family of transcriptional coregulator-encoding genes which function in cell differentiation and development. Here, we report the structure and expression pattern of the mouse Tif1gamma gene. This gene comprises 20 coding exons spanning about 77 kb of genomic DNA on chromosome 3F2, and encodes a 1142-amino-acid protein with 96% identity to the human protein. The locations of exon/intron boundaries correlated well with those for the regions of conserved amino acid sequences (RBCC motif, PHD finger and bromodomain). In situ hybridization analysis of the TIF1gamma mRNA on sections from staged mouse embryos revealed a low level of ubiquitous expression at midgestation, and higher expression levels within the brain and spinal cord epithelium at later developmental stages. Prominent expression was also found in developing sensory epithelia (cochlea, retina, olfactory epithelium), and in several developing organs including the thymus, lung, stomach, intestine, liver, and kidney cortex. In the adult mouse, Tif1gamma mRNA was detected by Northern blot analysis in all tissues examined, with the highest expression level in testis. In situ hybridization and immunohistochemistry studies revealed that expression of the Tif1gamma mRNA and protein varied according to the stage of the seminiferous epithelium cycle. Taken together, these results indicate-and serve as a basis for investigating-a possible involvement of Tif1gamma in the control of embryonic development and spermatogenesis.
Collapse
Affiliation(s)
- Kai-Ping Yan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS/INSERM/ULP/Collège de France, BP 10142, 67 404 Illkirch Cedex, France
| | | | | | | | | | | | | |
Collapse
|
149
|
Khetchoumian K, Teletin M, Mark M, Lerouge T, Cerviño M, Oulad-Abdelghani M, Chambon P, Losson R. TIF1delta, a novel HP1-interacting member of the transcriptional intermediary factor 1 (TIF1) family expressed by elongating spermatids. J Biol Chem 2004; 279:48329-41. [PMID: 15322135 DOI: 10.1074/jbc.m404779200] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
TIF1 (transcriptional intermediary factor 1) proteins are encoded by an expanding family of developmental and physiological control genes that are conserved from flies to man. These proteins are characterized by an N-terminal RING-B box-coiled-coil (RBCC) motif and a C-terminal PHD finger/bromodomain unit, and have been implicated in epigenetic mechanisms of transcriptional repression involving histone modifiers and heterochromatin-binding proteins. We describe here the isolation and functional characterization of a fourth murine TIF1 gene, TIF1delta. The predicted TIF1delta protein displays all the structural hallmarks of a bona fide TIF1 family member and resembles the other TIF1s in that it can exert a deacetylase-dependent silencing effect when tethered to a promoter region. Moreover, like TIF1alpha and TIF1beta, TIF1delta can homodimerize and contains a PXVXL motif necessary and sufficient for HP1 (heterochromatin protein 1) binding. Although TIF1alpha and TIF1beta also bind nuclear receptors and Kruppel-associated boxes specifically and respectively, TIF1delta appears to lack nuclear receptor- and Kruppel-associated box binding activity. Furthermore, TIF1delta is unique among the TIF1 family proteins in that its expression is largely restricted to the testis and confined to haploid elongating spermatids, where it associates preferentially with HP1 isotype gamma (HP1gamma) and forms discrete foci dispersed within the centromeric chromocenter and the surrounding nucleoplasm. Collectively, these data are consistent with specific, nonredundant functions for the TIF1 family members in vivo and suggest a role for TIF1delta in heterochromatin-mediated gene silencing during postmeiotic phases of spermatogenesis.
Collapse
Affiliation(s)
- Konstantin Khetchoumian
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP/Collège de France and Institut Clinique de la Souris, BP 10142, 67 404 Illkirch-Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
150
|
Ransom DG, Bahary N, Niss K, Traver D, Burns C, Trede NS, Paffett-Lugassy N, Saganic WJ, Lim CA, Hersey C, Zhou Y, Barut BA, Lin S, Kingsley PD, Palis J, Orkin SH, Zon LI. The zebrafish moonshine gene encodes transcriptional intermediary factor 1gamma, an essential regulator of hematopoiesis. PLoS Biol 2004; 2:E237. [PMID: 15314655 PMCID: PMC509301 DOI: 10.1371/journal.pbio.0020237] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2004] [Accepted: 05/26/2004] [Indexed: 11/18/2022] Open
Abstract
Hematopoiesis is precisely orchestrated by lineage-specific DNA-binding proteins that regulate transcription in concert with coactivators and corepressors. Mutations in the zebrafish moonshine (mon) gene specifically disrupt both embryonic and adult hematopoiesis, resulting in severe red blood cell aplasia. We report that mon encodes the zebrafish ortholog of mammalian transcriptional intermediary factor 1gamma (TIF1gamma) (or TRIM33), a member of the TIF1 family of coactivators and corepressors. During development, hematopoietic progenitor cells in mon mutants fail to express normal levels of hematopoietic transcription factors, including gata1, and undergo apoptosis. Three different mon mutant alleles each encode premature stop codons, and enforced expression of wild-type tif1gamma mRNA rescues embryonic hematopoiesis in homozygous mon mutants. Surprisingly, a high level of zygotic tif1gamma mRNA expression delineates ventral mesoderm during hematopoietic stem cell and progenitor formation prior to gata1 expression. Transplantation studies reveal that tif1gamma functions in a cell-autonomous manner during the differentiation of erythroid precursors. Studies in murine erythroid cell lines demonstrate that Tif1gamma protein is localized within novel nuclear foci, and expression decreases during erythroid cell maturation. Our results establish a major role for this transcriptional intermediary factor in the differentiation of hematopoietic cells in vertebrates.
Collapse
Affiliation(s)
- David G Ransom
- Division of Hematology/Oncology, Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|