201
|
The human histone H3 complement anno 2011. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2011; 1809:577-86. [DOI: 10.1016/j.bbagrm.2011.07.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 07/05/2011] [Accepted: 07/06/2011] [Indexed: 11/17/2022]
|
202
|
Tang Q, Chen Y, Meyer C, Geistlinger T, Lupien M, Wang Q, Liu T, Zhang Y, Brown M, Liu XS. A comprehensive view of nuclear receptor cancer cistromes. Cancer Res 2011; 71:6940-7. [PMID: 21940749 DOI: 10.1158/0008-5472.can-11-2091] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nuclear receptors comprise a superfamily of ligand-activated transcription factors that play important roles in both physiology and diseases including cancer. The technologies of chromatin immunoprecipitation followed by array hybridization (ChIP-chip) or massively parallel sequencing (ChIP-seq) has been used to map, at an unprecedented rate, the in vivo genome-wide binding (cistrome) of nuclear receptors in both normal and cancer cells. We developed a curated database of 88 nuclear receptor cistrome data sets and other associated high-throughput data sets including 121 collaborating factor cistromes, 94 epigenomes, and 319 transcriptomes. Through integrative analysis of the curated nuclear receptor ChIP-chip/seq data sets, we discovered novel factor-specific noncanonical motifs that may have important regulatory roles. We also revealed a common feature of nuclear receptor pioneering factors to recognize relatively short and AT-rich motifs. Most nuclear receptors bind predominantly to introns and distal intergenetic regions, and binding sites closer to transcription start sites were found to be neither stronger nor more evolutionarily conserved. Interestingly, while most nuclear receptors appear to be predominantly transcriptional activators, our analysis suggests that the binding of ESR1, RARA, and RARG has both activating and repressive effects. Through meta-analysis of different omic data of the same cancer cell line model from multiple studies, we generated consensus cistrome and expression profiles. We further made probabilistic predictions of the nuclear receptor target genes by integrating cistrome and transcriptome data and validated the predictions using expression data from tumor samples. The final database, with comprehensive cistrome, epigenome, and transcriptome data sets and downstream analysis results, constitutes a valuable resource for the nuclear receptor and cancer community.
Collapse
Affiliation(s)
- Qianzi Tang
- Department of Bioinformatics, School of Life Science and Technology, Tongji University, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
203
|
Spicuglia S, Vincent-Fabert C, Benoukraf T, Tibéri G, Saurin AJ, Zacarias-Cabeza J, Grimwade D, Mills K, Calmels B, Bertucci F, Sieweke M, Ferrier P, Duprez E. Characterisation of genome-wide PLZF/RARA target genes. PLoS One 2011; 6:e24176. [PMID: 21949697 PMCID: PMC3176768 DOI: 10.1371/journal.pone.0024176] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 08/02/2011] [Indexed: 01/30/2023] Open
Abstract
The PLZF/RARA fusion protein generated by the t(11;17)(q23;q21) translocation in acute promyelocytic leukaemia (APL) is believed to act as an oncogenic transcriptional regulator recruiting epigenetic factors to genes important for its transforming potential. However, molecular mechanisms associated with PLZF/RARA-dependent leukaemogenesis still remain unclear. We searched for specific PLZF/RARA target genes by ChIP-on-chip in the haematopoietic cell line U937 conditionally expressing PLZF/RARA. By comparing bound regions found in U937 cells expressing endogenous PLZF with PLZF/RARA-induced U937 cells, we isolated specific PLZF/RARA target gene promoters. We next analysed gene expression profiles of our identified target genes in PLZF/RARA APL patients and analysed DNA sequences and epigenetic modification at PLZF/RARA binding sites. We identify 413 specific PLZF/RARA target genes including a number encoding transcription factors involved in the regulation of haematopoiesis. Among these genes, 22 were significantly down regulated in primary PLZF/RARA APL cells. In addition, repressed PLZF/RARA target genes were associated with increased levels of H3K27me3 and decreased levels of H3K9K14ac. Finally, sequence analysis of PLZF/RARA bound sequences reveals the presence of both consensus and degenerated RAREs as well as enrichment for tissue-specific transcription factor motifs, highlighting the complexity of targeting fusion protein to chromatin. Our study suggests that PLZF/RARA directly targets genes important for haematopoietic development and supports the notion that PLZF/RARA acts mainly as an epigenetic regulator of its direct target genes.
Collapse
MESH Headings
- Acetylation
- Binding Sites/genetics
- Chromatin Immunoprecipitation/methods
- Cluster Analysis
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic
- Genome-Wide Association Study
- Histones/metabolism
- Humans
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Leukemia, Promyelocytic, Acute/genetics
- Leukemia, Promyelocytic, Acute/metabolism
- Leukemia, Promyelocytic, Acute/pathology
- Methylation
- Oligonucleotide Array Sequence Analysis/methods
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Promoter Regions, Genetic/genetics
- Promyelocytic Leukemia Zinc Finger Protein
- Protein Binding
- Reverse Transcriptase Polymerase Chain Reaction
- Transcription Factors/genetics
- Transcription Factors/metabolism
- U937 Cells
Collapse
Affiliation(s)
- Salvatore Spicuglia
- Centre d'Immunologie de Marseille-Luminy (CIML), Université de la Méditerranée, Campus de Luminy, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U631, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR 6102, Marseille, France
| | - Christelle Vincent-Fabert
- Institut National de la Santé et de la Recherche Médicale (INSERM) U891, Centre de Recherche en Cancérologie de Marseille, Marseille, France
- Institut Paoli-Calmettes, Marseille, France
| | - Touati Benoukraf
- Centre d'Immunologie de Marseille-Luminy (CIML), Université de la Méditerranée, Campus de Luminy, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U631, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR 6102, Marseille, France
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Guillaume Tibéri
- Institut National de la Santé et de la Recherche Médicale (INSERM) U891, Centre de Recherche en Cancérologie de Marseille, Marseille, France
- Institut Paoli-Calmettes, Marseille, France
| | - Andrew J. Saurin
- Institut de Biologie du Développement de Marseille Luminy, Université de la Méditerranée, Campus de Luminy, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR 6216, Marseille, France
| | - Joaquin Zacarias-Cabeza
- Centre d'Immunologie de Marseille-Luminy (CIML), Université de la Méditerranée, Campus de Luminy, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U631, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR 6102, Marseille, France
| | - David Grimwade
- Department of Medical and Molecular Genetics, King's College London School of Medicine, London, United Kingdom
| | - Ken Mills
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom
| | - Boris Calmels
- Institut National de la Santé et de la Recherche Médicale (INSERM) U891, Centre de Recherche en Cancérologie de Marseille, Marseille, France
- Institut Paoli-Calmettes, Marseille, France
| | - François Bertucci
- Institut National de la Santé et de la Recherche Médicale (INSERM) U891, Centre de Recherche en Cancérologie de Marseille, Marseille, France
- Oncologie Moléculaire, Institut Paoli-Calmettes, Marseille, France
| | - Michael Sieweke
- Centre d'Immunologie de Marseille-Luminy (CIML), Université de la Méditerranée, Campus de Luminy, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U631, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR 6102, Marseille, France
| | - Pierre Ferrier
- Centre d'Immunologie de Marseille-Luminy (CIML), Université de la Méditerranée, Campus de Luminy, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U631, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR 6102, Marseille, France
| | - Estelle Duprez
- Centre d'Immunologie de Marseille-Luminy (CIML), Université de la Méditerranée, Campus de Luminy, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U631, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR 6102, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U891, Centre de Recherche en Cancérologie de Marseille, Marseille, France
- Institut Paoli-Calmettes, Marseille, France
- * E-mail:
| |
Collapse
|
204
|
Valen E, Sandelin A. Genomic and chromatin signals underlying transcription start-site selection. Trends Genet 2011; 27:475-85. [PMID: 21924514 DOI: 10.1016/j.tig.2011.08.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 08/08/2011] [Accepted: 08/08/2011] [Indexed: 01/03/2023]
Abstract
A central question in cellular biology is how the cell regulates transcription and discerns when and where to initiate it. Locating transcription start sites (TSSs), the signals that specify them, and ultimately elucidating the mechanisms of regulated initiation has therefore been a recurrent theme. In recent years substantial progress has been made towards this goal, spurred by the possibility of applying genome-wide, sequencing-based analysis. We now have a large collection of high-resolution datasets identifying locations of TSSs, protein-DNA interactions, and chromatin features over whole genomes; the field is now faced with the daunting challenge of translating these descriptive maps into quantitative and predictive models describing the underlying biology. We review here the genomic and chromatin features that underlie TSS selection and usage, focusing on the differences between the major classes of core promoters.
Collapse
Affiliation(s)
- Eivind Valen
- The Bioinformatics Centre, Department of Biology, Ole Maaløes Vej 5, Copenhagen University, DK-2200, Denmark.
| | | |
Collapse
|
205
|
Laursen KB, Wong PM, Gudas LJ. Epigenetic regulation by RARα maintains ligand-independent transcriptional activity. Nucleic Acids Res 2011; 40:102-15. [PMID: 21911359 PMCID: PMC3245912 DOI: 10.1093/nar/gkr637] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Retinoic acid receptors (RARs) α, β and γ are key regulators of embryonic development. Hematopoietic differentiation is regulated by RARα, and several types of leukemia show aberrant RARα activity. Through microarray expression analysis, we identified transcripts differentially expressed between F9 wild-type (Wt) and RARα knockout cells cultured in the absence or presence of the RAR-specific ligand all trans retinoic acid (RA). We validated the decreased Mest, Tex13, Gab1, Bcl11a, Tcfap2a and HMGcs1 transcript levels, and increased Slc38a4, Stmn2, RpL39l, Ref2L, Mobp and Rlf1 transcript levels in the RARa knockout cells. The decreased Mest and Tex13 transcript levels were associated with increased promoter CpG-island methylation and increased repressive histone modifications (H3K9me3) in RARα knockout cells. Increased Slc38a4 and Stmn2 transcript levels were associated with decreased promoter CpG-island methylation and increased permissive histone modifications (H3K9/K14ac, H3K4me3) in RARα knockout cells. We demonstrated specific association of RARα and RXRα with the Mest promoter. Importantly, stable expression of a dominant negative, oncogenic PML–RARα fusion protein in F9 Wt cells recapitulated the decreased Mest transcript levels observed in RARα knockout cells. We propose that RARα plays an important role in cellular memory and imprinting by regulating the CpG methylation status of specific promoter regions.
Collapse
Affiliation(s)
- Kristian B Laursen
- Pharmacology Department of Weill Cornell Medical College of Cornell University, NY 10065, USA
| | | | | |
Collapse
|
206
|
Bogdanović O, Veenstra GJC. Affinity-based enrichment strategies to assay methyl-CpG binding activity and DNA methylation in early Xenopus embryos. BMC Res Notes 2011; 4:300. [PMID: 21851637 PMCID: PMC3169474 DOI: 10.1186/1756-0500-4-300] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 08/18/2011] [Indexed: 01/12/2023] Open
Abstract
Background DNA methylation is a widespread epigenetic modification in vertebrate genomes. Genomic sites of DNA methylation can be bound by methyl-CpG-binding domain proteins (MBDs) and specific zinc finger proteins, which can recruit co-repressor complexes to silence transcription on targeted loci. The binding to methylated DNA may be regulated by post-translational MBD modifications. Findings A methylated DNA affinity precipitation method was implemented to assay binding of proteins to methylated DNA. Endogenous MeCP2 and MBD3 were precipitated from Xenopus oocyte extracts and conditions for methylation-specific binding were optimized. For a reverse experiment, DNA methylation in early Xenopus embryos was assessed by MBD affinity capture. Conclusions A methylated DNA affinity resin can be applied to probe for MBD activity in extracts. This assay has a broad application potential as it can be coupled to downstream procedures such as western blotting, fluorimetric HDAC assays and quantitative mass spectrometry. Methylated DNA affinity capture by methyl-CpG binding proteins produces fractions highly enriched for methylated DNA, suitable for coupling to next generation sequencing technologies. The two enrichment strategies allow probing of methyl-CpG protein interactions in early vertebrate oocytes and embryos.
Collapse
Affiliation(s)
- Ozren Bogdanović
- Department of Molecular Biology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen, Faculty of Science, (Geert Grooteplein 28), Nijmegen, (6525 GA), The Netherlands.
| | | |
Collapse
|
207
|
Shima Y, Kitabayashi I. Deregulated transcription factors in leukemia. Int J Hematol 2011; 94:134-141. [PMID: 21823042 DOI: 10.1007/s12185-011-0905-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Revised: 07/19/2011] [Accepted: 07/19/2011] [Indexed: 12/16/2022]
Abstract
Specific chromosomal translocations and other mutations associated with acute myeloblastic leukemia (AML) often involve transcription factors and transcriptional coactivators. Such target genes include AML1, C/EBPα, RARα, MOZ, p300/CBP, and MLL, all of which are important in the regulation of hematopoiesis. The resultant fusion or mutant proteins deregulate the transcription of the affected genes and disrupt their essential role in hematopoiesis, causing differentiation block and abnormal proliferation and/or survival. This review focuses on such transcription factors and coactivators, and describes their roles in leukemogenesis and hematopoiesis.
Collapse
Affiliation(s)
- Yutaka Shima
- Division of Hematological Malignancy, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Issay Kitabayashi
- Division of Hematological Malignancy, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| |
Collapse
|
208
|
Emerging roles for retinoids in regeneration and differentiation in normal and disease states. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1821:213-21. [PMID: 21855651 DOI: 10.1016/j.bbalip.2011.08.002] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 07/29/2011] [Accepted: 08/02/2011] [Indexed: 12/22/2022]
Abstract
The vitamin A (retinol) metabolite, all-trans retinoic acid (RA), is a signaling molecule that plays key roles in the development of the body plan and induces the differentiation of many types of cells. In this review the physiological and pathophysiological roles of retinoids (retinol and related metabolites) in mature animals are discussed. Both in the developing embryo and in the adult, RA signaling via combinatorial Hox gene expression is important for cell positional memory. The genes that require RA for the maturation/differentiation of T cells are only beginning to be cataloged, but it is clear that retinoids play a major role in expression of key genes in the immune system. An exciting, recent publication in regeneration research shows that ALDH1a2 (RALDH2), which is the rate-limiting enzyme in the production of RA from retinaldehyde, is highly induced shortly after amputation in the regenerating heart, adult fin, and larval fin in zebrafish. Thus, local generation of RA presumably plays a key role in fin formation during both embryogenesis and in fin regeneration. HIV transgenic mice and human patients with HIV-associated kidney disease exhibit a profound reduction in the level of RARβ protein in the glomeruli, and HIV transgenic mice show reduced retinol dehydrogenase levels, concomitant with a greater than 3-fold reduction in endogenous RA levels in the glomeruli. Levels of endogenous retinoids (those synthesized from retinol within cells) are altered in many different diseases in the lung, kidney, and central nervous system, contributing to pathophysiology. This article is part of a Special Issue entitled Retinoid and Lipid Metabolism.
Collapse
|
209
|
Duong V, Rochette-Egly C. The molecular physiology of nuclear retinoic acid receptors. From health to disease. Biochim Biophys Acta Mol Basis Dis 2011; 1812:1023-31. [DOI: 10.1016/j.bbadis.2010.10.007] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 10/15/2010] [Indexed: 12/20/2022]
|
210
|
Bogdanović O, Long SW, van Heeringen SJ, Brinkman AB, Gómez-Skarmeta JL, Stunnenberg HG, Jones PL, Veenstra GJC. Temporal uncoupling of the DNA methylome and transcriptional repression during embryogenesis. Genome Res 2011; 21:1313-27. [PMID: 21636662 PMCID: PMC3149498 DOI: 10.1101/gr.114843.110] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 04/14/2011] [Indexed: 12/18/2022]
Abstract
DNA methylation is a tightly regulated epigenetic mark associated with transcriptional repression. Next-generation sequencing of purified methylated DNA obtained from early Xenopus tropicalis embryos demonstrates that this genome is heavily methylated during blastula and gastrula stages. Although DNA methylation is largely absent from transcriptional start sites marked with histone H3 lysine 4 trimethylation (H3K4me3), we find both promoters and gene bodies of active genes robustly methylated. In contrast, DNA methylation is absent in large H3K27me3 domains, indicating that these two repression pathways have different roles. Comparison with chromatin state maps of human ES cells reveals strong conservation of epigenetic makeup and gene regulation between the two systems. Strikingly, genes that are highly expressed in pluripotent cells and in Xenopus embryos but not in differentiated cells exhibit relatively high DNA methylation. Therefore, we tested the repressive potential of DNA methylation using transient and transgenic approaches and show that methylated promoters are robustly transcribed in blastula- and gastrula-stage embryos, but not in oocytes or late embryos. These findings have implications for reprogramming and the epigenetic regulation of pluripotency and differentiation and suggest a relatively open, pliable chromatin state in early embryos followed by reestablished methylation-dependent transcriptional repression during organogenesis and differentiation.
Collapse
Affiliation(s)
- Ozren Bogdanović
- Department of Molecular Biology, Faculty of Science, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen, 6500 Nijmegen, The Netherlands
| | - Steven W. Long
- Department of Cell and Developmental Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois 61801, USA
| | - Simon J. van Heeringen
- Department of Molecular Biology, Faculty of Science, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen, 6500 Nijmegen, The Netherlands
| | - Arie B. Brinkman
- Department of Molecular Biology, Faculty of Science, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen, 6500 Nijmegen, The Netherlands
| | - Jose Luis Gómez-Skarmeta
- Centro Andaluz de Biología del Desarrollo, Consejo Superior de Investigaciones Científicas and Universidad Pablo de Olavide, Carretera de Utrera Km1, 41013 Sevilla, Spain
| | - Hendrik G. Stunnenberg
- Department of Molecular Biology, Faculty of Science, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen, 6500 Nijmegen, The Netherlands
| | - Peter L. Jones
- Department of Cell and Developmental Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois 61801, USA
| | - Gert Jan C. Veenstra
- Department of Molecular Biology, Faculty of Science, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen, 6500 Nijmegen, The Netherlands
| |
Collapse
|
211
|
Wilson NK, Tijssen MR, Göttgens B. Deciphering transcriptional control mechanisms in hematopoiesis:the impact of high-throughput sequencing technologies. Exp Hematol 2011; 39:961-8. [PMID: 21781948 DOI: 10.1016/j.exphem.2011.07.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Accepted: 07/07/2011] [Indexed: 12/18/2022]
Abstract
One of the key challenges facing biomedical research is to extract biologically meaningful information from the ever-increasing scale and complexity of datasets generated through high-throughput approaches. Hematopoiesis represents one of the most experimentally tractable mammalian organ systems and, therefore, has historically tended to be at the forefront of applying new technologies within biomedical research. The combination of massive parallel sequencing technologies with chromatin-immunoprecipitation (ChIP-Seq) permits genome-scale characterization of histone modification status and identification of the complete set of binding sites for transcription factors. Because transcription factors have long been recognized as essential regulators of cell fate choice in hematopoiesis, ChIP-Seq technology has rapidly entered the arena of modern experimental hematology. Here we review the biological insights gained from ChIP-Seq studies performed in the hematopoietic system since the earliest studies just 4 years ago. A surprisingly large number of different approaches have already been implemented to extract new biological knowledge from ChIP-Seq datasets. By focusing on successful insights from multiple different approaches, we hope to provide stimulating reading for anyone wanting to utilize ChIP-Seq technology within their particular research field.
Collapse
Affiliation(s)
- Nicola K Wilson
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research, UK
| | | | | |
Collapse
|
212
|
Abstract
PAX5 encodes a master regulator of B-cell development. It fuses to other genes associated with acute lymphoblastoid leukemia (ALL). These fusion products are potent dominant-negative (DN) inhibitors of wild-type PAX5 resulting in a blockade of B-cell differentiation. Here, we show that multimerization of PAX5 DNA-binding domain (DBD) is necessary and sufficient to cause extremely stable chromatin binding and DN-activity. ALL-associated PAX5-C20S results from fusion of the N-terminal region of PAX5 including its paired DBD, to the C-terminus of C20orf112, a protein of unknown function. We report that PAX5-C20S is a tetramer which interacts extraordinarily stably with chromatin as determined by fluorescence recovery after photobleaching (FRAP) in living cells. Tetramerization, stable chromatin-binding and DN-activity all require a putative five-turn amphipathic α-helix at the C-terminus of C20orf112, and does not require potential co-repressor binding peptides elsewhere in the sequence. In vitro, the monomeric PAX5 DBD and PAX5-C20S binds a PAX5-binding site with equal affinity when it is at the center of an oligonucleotide too short to bind to more than one PAX5 DBD. But PAX5-C20S binds the same sequence with tenfold higher affinity than the monomeric PAX5 DBD when it is in a long DNA molecule. We suggest that the increased affinity results from interactions of one or more of the additional DBDs with neighboring non-specific sites in a long DNA molecule, and that this can account for the increased stability of PAX5-C20S chromatin binding compared to wt PAX5, resulting in DN-activity by competition for binding to PAX5-target sites. Consistent with this model, the ALL-associated PAX5 fused to ETV6 or the multimerization domain of ETV6 SAM results in stable chromatin binding and DN-activity. In addition, PAX5 DBD fused to artificial dimerization, trimerization, and tetramerization domains result in parallel increases in the stability of chromatin binding and DN-activity. Our studies suggest that oncogenic fusion proteins that retain the DBD of the transcription factor and the multimerization sequence of the partner protein can act in a DN fashion by multimerizing and binding avidly to gene targets preventing the normal transcription factor from binding and inducing expression of its target genes. Inhibition of this multimeriztion may provide a novel therapeutic approach for cancers with this or similar fusion proteins.
Collapse
|
213
|
Rao NAS, McCalman MT, Moulos P, Francoijs KJ, Chatziioannou A, Kolisis FN, Alexis MN, Mitsiou DJ, Stunnenberg HG. Coactivation of GR and NFKB alters the repertoire of their binding sites and target genes. Genome Res 2011; 21:1404-16. [PMID: 21750107 DOI: 10.1101/gr.118042.110] [Citation(s) in RCA: 160] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Glucocorticoid receptor (GR) exerts anti-inflammatory action in part by antagonizing proinflammatory transcription factors such as the nuclear factor kappa-b (NFKB). Here, we assess the crosstalk of activated GR and RELA (p65, major NFKB component) by global identification of their binding sites and target genes. We show that coactivation of GR and p65 alters the repertoire of regulated genes and results in their association with novel sites in a mutually dependent manner. These novel sites predominantly cluster with p65 target genes that are antagonized by activated GR and vice versa. Our data show that coactivation of GR and NFKB alters signaling pathways that are regulated by each factor separately and provide insight into the networks underlying the GR and NFKB crosstalk.
Collapse
Affiliation(s)
- Nagesha A S Rao
- Department of Molecular Biology, Radboud University, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
214
|
Lamsoul I, Burande CF, Razinia Z, Houles TC, Menoret D, Baldassarre M, Erard M, Moog-Lutz C, Calderwood DA, Lutz PG. Functional and structural insights into ASB2alpha, a novel regulator of integrin-dependent adhesion of hematopoietic cells. J Biol Chem 2011; 286:30571-30581. [PMID: 21737450 DOI: 10.1074/jbc.m111.220921] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
By providing contacts between hematopoietic cells and the bone marrow microenvironment, integrins are implicated in cell adhesion and thereby in control of cell fate of normal and leukemia cells. The ASB2 gene, initially identified as a retinoic acid responsive gene and a target of the promyelocytic leukemia retinoic acid receptor α oncoprotein in acute promyelocytic leukemia cells, encodes two isoforms, a hematopoietic-type (ASB2α) and a muscle-type (ASB2β) that are involved in hematopoietic and myogenic differentiation, respectively. ASB2α is the specificity subunit of an E3 ubiquitin ligase complex that targets filamins to proteasomal degradation. To examine the relationship of the ASB2α structure to E3 ubiquitin ligase function, functional assays and molecular modeling were performed. We show that ASB2α, through filamin A degradation, enhances adhesion of hematopoietic cells to fibronectin, the main ligand of β1 integrins. Furthermore, we demonstrate that a short N-terminal region specific to ASB2α, together with ankyrin repeats 1 to 10, is necessary for association of ASB2α with filamin A. Importantly, the ASB2α N-terminal region comprises a 9-residue segment with predicted structural homology to the filamin-binding motifs of migfilin and β integrins. Together, these data provide new insights into the molecular mechanisms of ASB2α binding to filamin.
Collapse
Affiliation(s)
- Isabelle Lamsoul
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 205 route de Narbonne, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France
| | - Clara F Burande
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 205 route de Narbonne, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France
| | - Ziba Razinia
- Department of Pharmacology and Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Thibault C Houles
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 205 route de Narbonne, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France
| | - Delphine Menoret
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 205 route de Narbonne, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France
| | - Massimiliano Baldassarre
- Department of Pharmacology and Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Monique Erard
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 205 route de Narbonne, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France
| | - Christel Moog-Lutz
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 205 route de Narbonne, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France
| | - David A Calderwood
- Department of Pharmacology and Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Pierre G Lutz
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 205 route de Narbonne, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France.
| |
Collapse
|
215
|
Abstract
Abstract
As the result of intense clinical and basic research, acute promyelocytic leukemia (APL) has progressively evolved from a deadly to a curable disease. Historically, efforts aimed at understanding the molecular bases for therapy response have repeatedly illuminated APL pathogenesis. The classic model attributes this therapeutic success to the transcriptional reactivation elicited by retinoic acid and the resulting overcoming of the differentiation block characteristic of APL blasts. However, in clinical practice, retinoic acid by itself only rarely yields prolonged remissions, even though it induces massive differentiation. In contrast, as a single agent, arsenic trioxide neither directly activates transcription nor triggers terminal differentiation ex vivo, but cures many patients. Here we review the evidence from recent ex vivo and in vivo studies that allow a reassessment of the role of differentiation in APL cure. We discuss alternative models in which PML-RARA degradation and the subsequent loss of APL cell self-renewal play central roles. Rather than therapy aimed at inducing differentiation, targeting cancer cell self-renewal may represent a more effective goal, achievable by a broader range of therapeutic agents.
Collapse
|
216
|
Cheung N, So CWE. Transcriptional and epigenetic networks in haematological malignancy. FEBS Lett 2011; 585:2100-11. [DOI: 10.1016/j.febslet.2011.03.068] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 03/28/2011] [Accepted: 03/28/2011] [Indexed: 12/16/2022]
|
217
|
Cassinat B, Zassadowski F, Ferry C, Llopis L, Bruck N, Lainey E, Duong V, Cras A, Despouy G, Chourbagi O, Beinse G, Fenaux P, Rochette Egly C, Chomienne C. New role for granulocyte colony-stimulating factor-induced extracellular signal-regulated kinase 1/2 in histone modification and retinoic acid receptor α recruitment to gene promoters: relevance to acute promyelocytic leukemia cell differentiation. Mol Cell Biol 2011; 31:1409-18. [PMID: 21262770 PMCID: PMC3135284 DOI: 10.1128/mcb.00756-10] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The induction of the granulocytic differentiation of leukemic cells by all-trans retinoic acid (RA) has been a major breakthrough in terms of survival for acute promyelocytic leukemia (APL) patients. Here we highlight the synergism and the underlying novel mechanism between RA and the granulocyte colony-stimulating factor (G-CSF) to restore differentiation of RA-refractory APL blasts. First, we show that in RA-refractory APL cells (UF-1 cell line), PML-RA receptor alpha (RARα) is not released from target promoters in response to RA, resulting in the maintenance of chromatin repression. Consequently, RARα cannot be recruited, and the RA target genes are not activated. We then deciphered how the combination of G-CSF and RA successfully restored the activation of RA target genes to levels achieved in RA-sensitive APL cells. We demonstrate that G-CSF restores RARα recruitment to target gene promoters through the activation of the extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) pathway and the subsequent derepression of chromatin. Thus, combinatorial activation of cytokines and RARs potentiates transcriptional activity through epigenetic modifications induced by specific signaling pathways.
Collapse
MESH Headings
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Cell Line, Tumor
- Chromatin Assembly and Disassembly/drug effects
- Enzyme Activation/drug effects
- Enzyme Induction/drug effects
- Extracellular Signal-Regulated MAP Kinases/biosynthesis
- Gene Expression Regulation, Leukemic/drug effects
- Granulocyte Colony-Stimulating Factor/pharmacology
- Histones/metabolism
- Humans
- Leukemia, Promyelocytic, Acute/enzymology
- Leukemia, Promyelocytic, Acute/genetics
- Leukemia, Promyelocytic, Acute/pathology
- MAP Kinase Signaling System/drug effects
- Mitogen-Activated Protein Kinase 3/biosynthesis
- Mitogen-Activated Protein Kinase 6/biosynthesis
- Phosphorylation/drug effects
- Promoter Regions, Genetic/genetics
- Protein Binding/drug effects
- Protein Processing, Post-Translational/drug effects
- Receptors, Retinoic Acid/metabolism
- Retinoic Acid Receptor alpha
- Transcription, Genetic/drug effects
- Tretinoin/pharmacology
Collapse
Affiliation(s)
- B Cassinat
- INSERM UMR-S-940, Université Paris Diderot, Hopital Saint-Louis, 75010 Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
218
|
Karlic H, Varga F. Impact of vitamin D metabolism on clinical epigenetics. Clin Epigenetics 2011; 2:55-61. [PMID: 22704269 PMCID: PMC3365599 DOI: 10.1007/s13148-011-0021-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Accepted: 01/14/2011] [Indexed: 01/28/2023] Open
Abstract
The bioactive vitamin D (VD) metabolite, 1,25-dihydroxyvitamin D3 regulates essential pathways of cellular metabolism and differentiation via its nuclear receptor (VDR). Molecular mechanisms which are known to play key roles in aging and cancer are mediated by complex processes involving epigenetic mechanisms contributing to efficiency of VD-activating CYP27A1 and CYP27B1 or inactivating CYP24 enzymes as well as VDR which binds to specific genomic sequences (VD response elements or VDREs). Activity of VDR can be modulated epigenetically by histone acetylation. It co-operates with other nuclear receptors which are influenced by histone acetyl transferases (HATs) as well as several types of histone deacetylases (HDACs). HDAC inhibitors (HDACi) and/or demethylating drugs may contribute to normalization of VD metabolism. Studies link VD signaling through the VDR directly to distinct molecular mechanisms of both HAT activity and the sirtuin class of HDACs (SIRT1) as well as the forkhead transcription factors thus contributing to elucidate complex epigenetic mechanisms for cancer preventive actions of VD.
Collapse
Affiliation(s)
- Heidrun Karlic
- Ludwig Boltzmann Cluster Oncology and Ludwig Boltzmann Institute for Leukemia Research and Hematology, Hanusch Hospital, Heinrich Collinstrasse 30, 1140 Vienna, Austria
| | - Franz Varga
- Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of WGKK and AUVA Trauma Centre Meidling, 1st Medical Department, Hanusch Hospital, Vienna, Austria
| |
Collapse
|
219
|
Al-Azzawi H, Yacqub-Usman K, Richardson A, Hofland LJ, Clayton RN, Farrell WE. Reversal of endogenous dopamine receptor silencing in pituitary cells augments receptor-mediated apoptosis. Endocrinology 2011; 152:364-73. [PMID: 21177832 DOI: 10.1210/en.2010-0886] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dopamine (DA)-agonist targeting of the DA D(2) receptor (D2R) in prolactinomas is the first-line treatment choice for suppression of prolactin and induction of tumor shrinkage. Resistance to DA agonists seems to be related to receptor number. Using the MMQ and GH3 pituitary cell lines, that either do or do not express D2R, respectively, we explored the epigenetic profile associated with the presence or absence of D2R in these cells lines. These studies led us to explore pharmacological strategies designed to restore receptor expression and thereby potentially augment DA agonist-mediated apoptosis. We show in GH3 cells that the D2R harbors increased CpG island-associated methylation and enrichment for histone H3K27me3. Conversely, MMQ cells and normal pituitaries show enrichment for H3K9Ac and barely detectable H3K27me3. Coculture of GH3 cells with the demethylating agent zebularine and the histone deacetylase inhibitor trichostatin A was responsible for a decrease in CpG island methylation and enrichment for the histone H3K9Ac mark. In addition, challenge of GH3 cells with zebularine alone or coculture with both agents led to expression of endogenous D2R in these cells. Induced expression D2R in GH3 cells was associated with a significant increase in apoptosis indices to challenge with either DA or bromocriptine. Specificity of a receptor-mediated response was established in coincubations with specific D2R antagonist and siRNA approaches in GH3 cell and D2R expressing MMQ cell lines. These studies point to the potential efficacy of combined treatment with epigenetic drugs and DA agonists for the medical management of different pituitary tumor subtypes, resistant to conventional therapies.
Collapse
Affiliation(s)
- Haneen Al-Azzawi
- Human Disease and Genomics Group, Institute of Science and Technology in Medicine, School of Medicine, Keele University, Stoke on Trent, Staffordshire ST4 7QB United Kingdom
| | | | | | | | | | | |
Collapse
|
220
|
Quintás-Cardama A, Santos FPS, Garcia-Manero G. Histone deacetylase inhibitors for the treatment of myelodysplastic syndrome and acute myeloid leukemia. Leukemia 2011; 25:226-35. [PMID: 21116282 DOI: 10.1038/leu.2010.276] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Epigenetic changes have been identified in recent years as important factors in the pathogenesis of myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Histone deacetylase inhibitors (HDACIs) regulate the acetylation of histones as well as other non-histone protein targets. Treatment with HDACIs results in chromatin remodeling that permits re-expression of silenced tumor suppressor genes in cancer cells, which, in turn, can potentially result in cellular differentiation, inhibition of proliferation and/or apoptosis. Several classes of HDACIs are currently under development for the treatment of patients with MDS and AML. Although modest clinical activity has been reported with the use of HDACIs as single-agent therapy, marked responses have been observed in selected subsets of patients. More importantly, HDACIs appear to be synergistic in vitro and improve response rates in vivo when combined with other agents, such as hypomethylating agents. Furthermore, HDACIs are also being investigated in combination with non-epigenetic therapies. This article synthesizes the most recent results reported with HDACIs in clinical trials conducted in patients with MDS and other myeloid malignancies.
Collapse
Affiliation(s)
- A Quintás-Cardama
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | |
Collapse
|
221
|
Bonadies N, Foster SD, Chan WI, Kvinlaug BT, Spensberger D, Dawson MA, Spooncer E, Whetton AD, Bannister AJ, Huntly BJ, Göttgens B. Genome-wide analysis of transcriptional reprogramming in mouse models of acute myeloid leukaemia. PLoS One 2011; 6:e16330. [PMID: 21297973 PMCID: PMC3030562 DOI: 10.1371/journal.pone.0016330] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Accepted: 12/12/2010] [Indexed: 11/27/2022] Open
Abstract
Acute leukaemias are commonly caused by mutations that corrupt the transcriptional circuitry of haematopoietic stem/progenitor cells. However, the mechanisms underlying large-scale transcriptional reprogramming remain largely unknown. Here we investigated transcriptional reprogramming at genome-scale in mouse retroviral transplant models of acute myeloid leukaemia (AML) using both gene-expression profiling and ChIP-sequencing. We identified several thousand candidate regulatory regions with altered levels of histone acetylation that were characterised by differential distribution of consensus motifs for key haematopoietic transcription factors including Gata2, Gfi1 and Sfpi1/Pu.1. In particular, downregulation of Gata2 expression was mirrored by abundant GATA motifs in regions of reduced histone acetylation suggesting an important role in leukaemogenic transcriptional reprogramming. Forced re-expression of Gata2 was not compatible with sustained growth of leukaemic cells thus suggesting a previously unrecognised role for Gata2 in downregulation during the development of AML. Additionally, large scale human AML datasets revealed significantly higher expression of GATA2 in CD34+ cells from healthy controls compared with AML blast cells. The integrated genome-scale analysis applied in this study represents a valuable and widely applicable approach to study the transcriptional control of both normal and aberrant haematopoiesis and to identify critical factors responsible for transcriptional reprogramming in human cancer.
Collapse
Affiliation(s)
- Nicolas Bonadies
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Samuel D. Foster
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Wai-In Chan
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Brynn T. Kvinlaug
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Dominik Spensberger
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Mark A. Dawson
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Elaine Spooncer
- School of Cancer and Imaging Sciences, University of Manchester, Manchester, United Kingdom
| | - Anthony D. Whetton
- School of Cancer and Imaging Sciences, University of Manchester, Manchester, United Kingdom
| | - Andrew J. Bannister
- Gurdon Institute and Department of Pathology, Cambridge University, Cambridge, United Kingdom
| | - Brian J. Huntly
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Berthold Göttgens
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
222
|
Sukhai MA, Thomas M, Hamadanizadeh SA, Xuan Y, Wells RA, Kamel-Reid S. Correlation among nuclear localization of NuMA-RARα, deregulation of gene expression and leukemic phenotype of hCG-NuMA-RARα transgenic mice. Leuk Res 2011; 35:670-6. [PMID: 21255834 DOI: 10.1016/j.leukres.2010.12.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2010] [Revised: 12/13/2010] [Accepted: 12/13/2010] [Indexed: 12/11/2022]
Abstract
Acute promyelocytic leukemia (APL) is a model system of aberrant transcription in cancer. We sought to elucidate the mechanism of action of the variant fusion NuMA-RARα in APL, using the hCG-NuMA-RARα transgenic model. We report that subcellular localization of NuMA-RARα in transgenic mice is dependent upon its protein expression and transgene dosage. Subcellular localization of the fusion is inversely correlated with extent of gene deregulation at the mRNA level for Cebpα, Cebpɛ and Pu.1. Finally, we report that phenotype onset is correlated with NuMA-RARα copy number; mice with higher copy number developing disease later than those with lower copy number.
Collapse
Affiliation(s)
- Mahadeo A Sukhai
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | | | | | | | | | | |
Collapse
|
223
|
Abstract
PML—RAR (retinoic acid receptor)α is the hallmark protein of acute promyelocytic leukaemia, a highly malignant subtype of acute myeloid leukaemia that accounts for approximately 10% of all AML cases. Recently, several studies have been set out to obtain a comprehensive genome-wide view of the molecular actions of this chimeric protein. In this review, we highlight the new insights that arose from these studies, in particular focussing on newly identified PML–RARα target genes, its interplay with RXR and deregulation of epigenetic modifications.
Collapse
|
224
|
Abstract
Retinoids are ubiquitous signaling molecules that influence nearly every cell type, exert profound effects on development, and complement cancer chemotherapeutic regimens. All-trans retinoic acid (RA) and other active retinoids are generated from vitamin A (retinol), but key aspects of the signaling pathways required to produce active retinoids remain unclear. Retinoids generated by one cell type can affect nearby cells, so retinoids also function in intercellular communication. RA induces differentiation primarily by binding to RARs, transcription factors that associate with RXRs and bind RAREs in the nucleus. Binding of RA: (1) initiates changes in interactions of RAR/RXRs with co-repressor and co-activator proteins, activating transcription of primary target genes; (2) alters interactions with proteins that induce epigenetic changes; (3) induces transcription of genes encoding transcription factors and signaling proteins that further modify gene expression (e.g., FOX03A, Hoxa1, Sox9, TRAIL, UBE2D3); and (4) results in alterations in estrogen receptor α signaling. Proteins that bind at or near RAREs include Sin3a, N-CoR1, PRAME, Trim24, NRIP1, Ajuba, Zfp423, and MN1/TEL. Interactions among retinoids, RARs/RXRs, and these proteins explain in part the powerful effects of retinoids on stem cell differentiation. Studies of this retinol signaling cascade enhance our ability to understand and regulate stem cell differentiation for therapeutic and scientific purposes. In cancer chemotherapeutic regimens retinoids can promote tumor cell differentiation and/or induce proteins that sensitize tumors to drug combinations. Mechanistic studies of retinoid signaling continue to suggest novel drug targets and will improve therapeutic strategies for cancer and other diseases, such as immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Medical College of Cornell University, New York, New York 10065, USA.
| | | |
Collapse
|
225
|
Peters AHFM, Schwaller J. Epigenetic mechanisms in acute myeloid leukemia. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2011; 67:197-219. [PMID: 21141731 DOI: 10.1007/978-3-7643-8989-5_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Acute leukemia is characterized by clonal expansion of hematopoietic stem and progenitor cells with blocked differentiation. Clinical and experimental evidences suggest that acute myeloid leukemia (AML) is the product of several functionally cooperating genetic alterations including chromosomal translocations leading to expression of leukemogenic fusion proteins. Several AML-associated lesions target chromatin regulators like histone methyltransferases or histone acetyltransferases, including mixed-lineage leukemia 1 (MLL1) or CREB bindung protein/p300. Molecular and biochemical studies start to provide useful insights into the mechanisms of targeting and mode-of-action of such leukemogenic fusion proteins resulting in aberrant gene expression programs and AML. Chromatin modulating mechanisms are also mediating the transforming activity of key drivers of leukemogenesis by aberrant recruitment of corepressors. Recent large-scale screening efforts demonstrated that both aberrant DNA promoter methylation and aberrantly expressed microRNAs play an important role in the pathogenesis of AML as well. Current efforts to therapeutically exploit the potential reversibility of epigenetic mechanisms are focused on small molecules that inhibit DNA methyltransferases or histone deacetylases. Several phase I/II clinical trials using such compounds have reported promising, but mostly transient, clinical responses. This underscores the need to further dissect the molecular players of epigenetic mechanisms driving induction, maintenance, and potential reversibility of leukemic state to develop efficient and long-lasting targeted therapeutic strategies.
Collapse
Affiliation(s)
- Antoine H F M Peters
- Friedrich Miescher Institute for Biomedical Research, CH-4058 Basel, Switzerland.
| | | |
Collapse
|
226
|
Luesink M, Jansen JH. Advances in understanding the pulmonary infiltration in acute promyelocytic leukaemia. Br J Haematol 2010; 151:209-20. [PMID: 20735400 DOI: 10.1111/j.1365-2141.2010.08325.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In acute promyelocytic leukaemia (APL), differentiation therapy can be complicated by the development of a differentiation syndrome (DS). Pulmonary infiltration of differentiating leukaemic cells is a key event in the development of DS. Several mediators have been identified that may promote migration and extravasation of differentiating APL cells from the bloodstream into the tissue. Adhesion of APL cells to each other and to the endothelium is induced by upregulation of the expression of adhesion molecules and constitutively active β2-integrins during differentiation therapy. The expression of chemokines and their receptors is significantly upregulated as well. Pulmonary chemokine production can trigger transendothelial migration of differentiating APL cells from the bloodstream into the underlying tissue (initiation phase of DS). Massive production of chemokines by infiltrated APL cells can further enhance transendothelial migration of differentiating APL cells, causing an uncontrollable hyperinflammatory reaction in the lung (aggravation phase), which is not efficiently switched-off by corticosteroids.
Collapse
Affiliation(s)
- Maaike Luesink
- Radboud University Nijmegen Medical Centre, The Netherlands
| | | |
Collapse
|
227
|
Mengeling BJ, Phan TQ, Goodson ML, Privalsky ML. Aberrant corepressor interactions implicated in PML-RAR(alpha) and PLZF-RAR(alpha) leukemogenesis reflect an altered recruitment and release of specific NCoR and SMRT splice variants. J Biol Chem 2010; 286:4236-47. [PMID: 21131350 DOI: 10.1074/jbc.m110.200964] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human acute promyelocytic leukemia is causally linked to chromosomal translocations that generate chimeric retinoic acid receptor-α proteins (x-RARα fusions). Wild-type RARα is a transcription factor that binds to the SMRT/NCoR family of corepressors in the absence of hormone but releases from corepressor and binds coactivators in response to retinoic acid. In contrast, the x-RARα fusions are impaired for corepressor release and operate in acute promyelocytic leukemia as dominant-negative inhibitors of wild-type RARα. We report that the two most common x-RARα fusions, PML-RARα and PLZF-RARα, have gained the ability to recognize specific splice variants of SMRT and NCoR that are poorly recognized by RARα. These differences in corepressor specificity between the normal and oncogenic receptors are further magnified in the presence of a retinoid X receptor heteromeric partner. The ability of retinoids to fully release corepressor from PML-RARα differs for the different splice variants, a phenomenon relevant to the requirement for supraphysiological levels of this hormone in differentiation therapy of leukemic cells. We propose that this shift in the specificity of the x-RARα fusions to a novel repertoire of corepressors contributes to the dominant-negative and oncogenic properties of these oncoproteins and helps explain previously paradoxical aspects of their behavior.
Collapse
Affiliation(s)
- Brenda J Mengeling
- Department of Microbiology, College of Biological Sciences, University of California at Davis, Davis, California 95616, USA
| | | | | | | |
Collapse
|
228
|
Pancione M, Sabatino L, Fucci A, Carafa V, Nebbioso A, Forte N, Febbraro A, Parente D, Ambrosino C, Normanno N, Altucci L, Colantuoni V. Epigenetic silencing of peroxisome proliferator-activated receptor γ is a biomarker for colorectal cancer progression and adverse patients' outcome. PLoS One 2010; 5:e14229. [PMID: 21151932 PMCID: PMC2997072 DOI: 10.1371/journal.pone.0014229] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2010] [Accepted: 11/09/2010] [Indexed: 12/17/2022] Open
Abstract
The relationship between peroxisome proliferator-activated receptor γ (PPARG) expression and epigenetic changes occurring in colorectal-cancer pathogenesis is largely unknown. We investigated whether PPARG is epigenetically regulated in colorectal cancer (CRC) progression. PPARG expression was assessed in CRC tissues and paired normal mucosa by western blot and immunohistochemistry and related to patients' clinicopathological parameters and survival. PPARG promoter methylation was analyzed by methylation-specific-PCR and bisulphite sequencing. PPARG expression and promoter methylation were similarly examined also in CRC derived cell lines. Chromatin immunoprecipitation in basal conditions and after epigenetic treatment was performed along with knocking-down experiments of putative regulatory factors. Gene expression was monitored by immunoblotting and functional assays of cell proliferation and invasiveness. Methylation on a specific region of the promoter is strongly correlated with PPARG lack of expression in 30% of primary CRCs and with patients' poor prognosis. Remarkably, the same methylation pattern is found in PPARG-negative CRC cell lines. Epigenetic treatment with 5'-aza-2'-deoxycytidine can revert this condition and, in combination with trichostatin A, dramatically re-activates gene transcription and receptor activity. Transcriptional silencing is due to the recruitment of MeCP2, HDAC1 and EZH2 that impart repressive chromatin signatures determining an increased cell proliferative and invasive potential, features that can experimentally be reverted. Our findings provide a novel mechanistic insight into epigenetic silencing of PPARG in CRC that may be relevant as a prognostic marker of tumor progression.
Collapse
Affiliation(s)
- Massimo Pancione
- Department of Biological and Environmental Sciences, University of Sannio, Benevento, Italy
| | - Lina Sabatino
- Department of Biological and Environmental Sciences, University of Sannio, Benevento, Italy
| | - Alessandra Fucci
- Department of Biological and Environmental Sciences, University of Sannio, Benevento, Italy
| | - Vincenzo Carafa
- Department of General Pathology, Second University of Naples, Napoli, Italy
| | - Angela Nebbioso
- Department of General Pathology, Second University of Naples, Napoli, Italy
| | - Nicola Forte
- Departments of Medical Oncology and Clinical Pathology, Fatebenefratelli Hospital, Benevento, Italy
| | - Antonio Febbraro
- Departments of Medical Oncology and Clinical Pathology, Fatebenefratelli Hospital, Benevento, Italy
| | - Domenico Parente
- Departments of Medical Oncology and Clinical Pathology, Fatebenefratelli Hospital, Benevento, Italy
| | - Concetta Ambrosino
- Department of Biological and Environmental Sciences, University of Sannio, Benevento, Italy
- Biogem “G. Salvatore” Genetic Research Institute, Ariano Irpino, Italy
| | - Nicola Normanno
- Pharmacogenomic Laboratory, Center for Oncology Research, Mercogliano, Italy
| | - Lucia Altucci
- Department of General Pathology, Second University of Naples, Napoli, Italy
- CNR-IGB, Napoli, Italy
| | - Vittorio Colantuoni
- Department of Biological and Environmental Sciences, University of Sannio, Benevento, Italy
- Biogem “G. Salvatore” Genetic Research Institute, Ariano Irpino, Italy
- * E-mail:
| |
Collapse
|
229
|
Abstract
After the first draft sequence of the human genome was announced by the International Human Genome Sequencing Consortium and Celera Genomics in February 2001, this and subsequent sequences have been instrumental for the systematic analysis of various human genomes, including the cancer genome. Now we are moving into an era in which comprehensive sequence-based information on vast numbers of tumors can be obtained. Such information can provide novel and detailed perspectives on how individual tumors develop. As a corollary, this information can be used to generate novel diagnostic and prognostic markers and to design novel therapeutic strategies. Because these advances are moving rapidly, any state-of-the-art overview is likely to be outdated before it reaches publication. Nevertheless, data presented at the 12th European Workshop on Cytogenetics and Molecular Genetics of Solid Tumors, held June 3-6, 2010, at Nijmegen in The Netherlands, and of which the abstracts are included in this special issue of Cancer Genetics and Cytogenetics, may provide a basis for such an overview. Although one nowadays hears talk of the post-genome era, we are in fact still very much within the genome era; indeed, most of the work presented at the workshop dealt with the cancer genome. Other emerging areas were also addressed, however, including the cancer epigenome, the cancer transcriptome, and the cancer fragilome.
Collapse
|
230
|
Abstract
The fusion oncogene, promyelocytic leukaemia (PML)-retinoic acid receptor-α (RARA), initiates acute promyelocytic leukaemia (APL) through both a block to differentiation and increased self-renewal of leukaemic progenitor cells. The current standard of care is retinoic acid (RA) and chemotherapy, but arsenic trioxide also cures many patients with APL, and an RA plus arsenic trioxide combination cures most patients. This Review discusses the recent evidence that reveals surprising new insights into how RA and arsenic trioxide cure this leukaemia, by targeting PML-RARα for degradation. Drug-triggered oncoprotein degradation may be a strategy that is applicable to many cancers.
Collapse
Affiliation(s)
- Hugues de Thé
- Institut National de Santé et de Recherche Médicale, Centre National de Recherche Scientifique, Institut Universitaire d'Hématologie, Université Paris-Diderot UMR 944/7212, Equipe labellisée par Ligue contre Cancer, Service de Biochimie, Hôpital St. Louis, 2 avenue C. Vellefaux, 75475 Paris, CEDEX 10, France.
| | | |
Collapse
|
231
|
Genome-wide interplay of nuclear receptors with the epigenome. Biochim Biophys Acta Mol Basis Dis 2010; 1812:818-23. [PMID: 20970499 DOI: 10.1016/j.bbadis.2010.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 10/12/2010] [Accepted: 10/14/2010] [Indexed: 01/19/2023]
Abstract
The nuclear receptor superfamily consists of DNA binding transcription factors that are involved in regulating a wide variety of processes such as metabolism, development, reproduction, and immune responses. Upon binding, nuclear receptors modulate transcription through affecting the local chromatin environment via recruitment of various coregulatory proteins. The recent development of new high-throughput sequencing methods allowed for the first time the comprehensive examination of nuclear receptor action in the context of the epigenome. Here, we discuss how recent genome-wide analyses have provided important new insights on the interplay of nuclear receptors and the epigenome in health and disease. This article is part of a Special Issue entitled: Translating nuclear receptors from health to disease.
Collapse
|
232
|
Subramanyam D, Belair CD, Barry-Holson KQ, Lin H, Kogan SC, Passegué E, Blelloch R. PML-RAR{alpha} and Dnmt3a1 cooperate in vivo to promote acute promyelocytic leukemia. Cancer Res 2010; 70:8792-801. [PMID: 20861188 DOI: 10.1158/0008-5472.can-08-4481] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The PML-RARα oncogene is the central effector of acute promyelocytic leukemia (APL). PML-RARα physically interacts with epigenetic-modifying enzymes including DNA methyltransferases (Dnmt) to suppress critical downstream targets. Here, we show that increased expression of Dnmt3a1 cooperates with PML-RARα in vivo to promote early lethality secondary to myeloid expansion and dysfunction in primary mice. Bone marrow cells from these mice cause leukemogenesis with a shortened latency and a higher penetrance on transplantation into irradiated recipients. Furthermore, leukemic cells overexpressing PML-RARα and Dnmt3a1 display increased methylation at a target promoter compared with PML-RARα or Dnmt3a1 controls. Our findings show a cooperation between the PML-RARα oncogene and the Dnmt3a1 enzyme in vivo and that Dnmt levels can be rate limiting in APL progression.
Collapse
Affiliation(s)
- Deepa Subramanyam
- Institute for Regeneration Medicine, University of California, San Francisco, California 94143, USA
| | | | | | | | | | | | | |
Collapse
|
233
|
Jeanne M, Lallemand-Breitenbach V, Ferhi O, Koken M, Le Bras M, Duffort S, Peres L, Berthier C, Soilihi H, Raught B, de Thé H. PML/RARA oxidation and arsenic binding initiate the antileukemia response of As2O3. Cancer Cell 2010; 18:88-98. [PMID: 20609355 DOI: 10.1016/j.ccr.2010.06.003] [Citation(s) in RCA: 250] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Revised: 05/05/2010] [Accepted: 06/10/2010] [Indexed: 11/26/2022]
Abstract
As(2)O(3) cures acute promyelocytic leukemia (APL) by initiating PML/RARA oncoprotein degradation, through sumoylation of its PML moiety. However, how As(2)O(3) initiates PML sumoylation has remained largely unexplained. As(2)O(3) binds vicinal cysteines and increases reactive oxygen species (ROS) production. We demonstrate that upon As(2)O(3) exposure, PML undergoes ROS-initiated intermolecular disulfide formation and binds arsenic directly. Disulfide-linked PML or PML/RARA multimers form nuclear matrix-associated nuclear bodies (NBs), become sumoylated and are degraded. Hematopoietic progenitors transformed by an As(2)O(3)-binding PML/RARA mutant exhibit defective As(2)O(3) response. Conversely, nonarsenical oxidants elicit PML/RARA multimerization, NB-association, degradation, and leukemia response in vivo, but do not affect PLZF/RARA-driven APLs. Thus, PML oxidation regulates NB-biogenesis, while oxidation-enforced PML/RARA multimerization and direct arsenic-binding cooperate to enforce APL's exquisite As(2)O(3) sensitivity.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Arsenic Trioxide
- Arsenicals/pharmacology
- Blotting, Western
- CHO Cells
- COS Cells
- Chlorocebus aethiops
- Cricetinae
- Cricetulus
- Disulfides/metabolism
- Embryo, Mammalian/cytology
- Embryo, Mammalian/metabolism
- Fibroblasts/cytology
- Fibroblasts/metabolism
- Hematopoietic Stem Cells/cytology
- Hematopoietic Stem Cells/metabolism
- Humans
- Intranuclear Inclusion Bodies/metabolism
- Leukemia, Promyelocytic, Acute/drug therapy
- Leukemia, Promyelocytic, Acute/metabolism
- Leukemia, Promyelocytic, Acute/pathology
- Mice
- Mice, Knockout
- Mutation/genetics
- Nuclear Proteins/physiology
- Oncogene Proteins, Fusion/chemistry
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Oxides/pharmacology
- Promyelocytic Leukemia Protein
- Proteasome Endopeptidase Complex/metabolism
- Proteasome Inhibitors
- Protein Processing, Post-Translational
- Reactive Oxygen Species/metabolism
- Signal Transduction
- Small Ubiquitin-Related Modifier Proteins/metabolism
- Transcription Factors/physiology
- Tumor Suppressor Proteins/physiology
Collapse
Affiliation(s)
- Marion Jeanne
- Inserm/Centre National de la Recherche Scientifique (CNRS)/Université Paris Diderot/Institut Universitaire Hématologie U944/UMR7212, Laboratoire associé de la Ligue Nationale contre le Cancer, Hôpital St Louis, 1, Av. C. Vellefaux, 75475 Paris, Cedex 10, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
234
|
Chi P, Allis CD, Wang GG. Covalent histone modifications--miswritten, misinterpreted and mis-erased in human cancers. Nat Rev Cancer 2010; 10:457-69. [PMID: 20574448 PMCID: PMC3262678 DOI: 10.1038/nrc2876] [Citation(s) in RCA: 819] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Post-translational modification of histones provides an important regulatory platform for processes such as gene transcription and DNA damage repair. It has become increasingly apparent that the misregulation of histone modification, which is caused by the deregulation of factors that mediate the modification installation, removal and/or interpretation, actively contributes to human cancer. In this Review, we summarize recent advances in understanding the interpretation of certain histone methylations by plant homeodomain finger-containing proteins, and how misreading, miswriting and mis-erasing of histone methylation marks can be associated with oncogenesis and progression. These observations provide us with a greater mechanistic understanding of epigenetic alterations in human cancers and might also help direct new therapeutic interventions in the future.
Collapse
Affiliation(s)
- Ping Chi
- Laboratory of Chromatin Biology & Epigenetics, The Rockefeller University, New York, NY 10065, USA
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - C. David Allis
- Laboratory of Chromatin Biology & Epigenetics, The Rockefeller University, New York, NY 10065, USA
- Correspondence to C.D.A. () or G.G.W. (), Contact: 1230 York Avenue, The Rockefeller University, Box 78, New York, NY 10065, USA. Tel: (212) 327-7872; FAX: (212) 327-7849
| | - Gang Greg Wang
- Laboratory of Chromatin Biology & Epigenetics, The Rockefeller University, New York, NY 10065, USA
- Correspondence to C.D.A. () or G.G.W. (), Contact: 1230 York Avenue, The Rockefeller University, Box 78, New York, NY 10065, USA. Tel: (212) 327-7872; FAX: (212) 327-7849
| |
Collapse
|
235
|
Tuckermann J, Bourguet W, Mandrup S. Meeting report: nuclear receptors: transcription factors and drug targets connecting basic research with translational medicine. Mol Endocrinol 2010; 24:1311-21. [PMID: 20519330 DOI: 10.1210/me.2010-0083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The biannual European Molecular Biology Organization (EMBO) conference on nuclear receptors was organized by Beatrice Desvergne and Laszlo Nagy and took place in Cavtat near Dubrovnik on the Adriatic coast of Croatia September 25-29, 2009. The meeting brought together researchers from all over the world covering a wide spectrum from fundamental mechanistic studies to metabolism, clinical studies, and drug development. In this report, we summarize the recent and exciting findings presented by the speakers at the meeting.
Collapse
Affiliation(s)
- Jan Tuckermann
- Leibniz Institute for Age Research-Fritz Lipmann Institute, D-07745 Jena, Germany
| | | | | |
Collapse
|
236
|
Nasr R, de Thé H. Eradication of acute promyelocytic leukemia-initiating cells by PML/RARA-targeting. Int J Hematol 2010; 91:742-7. [PMID: 20455087 DOI: 10.1007/s12185-010-0582-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2010] [Accepted: 04/19/2010] [Indexed: 12/20/2022]
Abstract
Acute promyelocytic leukemia (APL) is characterized by a t(15;17) translocation that yields a PML/RARA fusion protein. Expression of PML/RARA, a potent transcriptional repressor, induces APL in mice. Both retinoic acid (RA) and arsenic trioxide directly target PML/RARA-mediated transcriptional repression and protein stability, inducing rapid differentiation of the promyelocytes and clinical remission in most APL patients. RA also triggers growth arrest and progressive clearance of leukemia initiating cells (LIC), both ex vivo and in vivo. Suboptimal RA concentrations or expression of the PLZF/RARA variant allows complete RA-induced differentiation, but neither LIC clearance nor disease remission. Thus, RA-induced differentiation and LIC clearance may be uncoupled. The RA/arsenic trioxide association, which dramatically synergizes for PML/RARA degradation but not for differentiation, rapidly clears LIC in a proteasome-dependent manner, resulting in APL eradication in murine models and patients. Collectively, these results demonstrate that LIC clearance, which mirrors PML/RARA degradation, is the primary basis for APL cure by the RA/arsenic trioxide association, rather than differentiation. Oncogene degradation could be a generally applicable therapeutic strategy to clear LICs in several types of tumors.
Collapse
Affiliation(s)
- Rihab Nasr
- Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | | |
Collapse
|
237
|
Martens JHA, Stunnenberg HG. The molecular signature of oncofusion proteins in acute myeloid leukemia. FEBS Lett 2010; 584:2662-9. [PMID: 20388510 DOI: 10.1016/j.febslet.2010.04.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Accepted: 04/03/2010] [Indexed: 02/02/2023]
Abstract
Acute myeloid leukemia (AML) associated translocations often cause gene fusions that encode oncofusion proteins. Although many of the breakpoints involved in chromosomal translocations have been cloned, in most cases the role of the chimeric proteins in tumorigenesis is not elucidated. Here we will discuss the fusion proteins of the 4 most common translocations associated with AML as well as the common molecular mechanisms that these four and other fusion proteins utilize to transform progenitor cells. Intriguingly, although the individual partners within the fusion proteins represent a wide variety of cellular functions, at the molecular level many commodities can be found.
Collapse
Affiliation(s)
- Joost H A Martens
- Radboud University, Department of Molecular Biology, Faculty of Science, Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | | |
Collapse
|
238
|
Mikesch JH, Gronemeyer H, So CWE. Discovery of novel transcriptional and epigenetic targets in APL by global ChIP analyses: Emerging opportunity and challenge. Cancer Cell 2010; 17:112-4. [PMID: 20159604 DOI: 10.1016/j.ccr.2010.01.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Identifying transcriptional program(s) deregulated by oncoproteins is key to understanding the molecular basis of the disease. In this issue of Cancer Cell, two studies by Martens et al. and Wang et al. provide global blueprints for transcriptional targets and epigenetic modifications mediated by PML-RARalpha in acute promyelocytic leukemia.
Collapse
|