251
|
Segré CV, Chiocca S. Regulating the regulators: the post-translational code of class I HDAC1 and HDAC2. J Biomed Biotechnol 2010; 2011:690848. [PMID: 21197454 PMCID: PMC3004424 DOI: 10.1155/2011/690848] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Accepted: 10/15/2010] [Indexed: 11/18/2022] Open
Abstract
Class I histone deacetylases (HDACs) are cellular enzymes expressed in many tissues and play crucial roles in differentiation, proliferation, and cancer. HDAC1 and HDAC2 in particular are highly homologous proteins that show redundant or specific roles in different cell types or in response to different stimuli and signaling pathways. The molecular details of this dual regulation are largely unknown. HDAC1 and HDAC2 are not only protein modifiers, but are in turn regulated by post-translational modifications (PTMs): phosphorylation, acetylation, ubiquitination, SUMOylation, nitrosylation, and carbonylation. Some of these PTMs occur and crosstalk specifically on HDAC1 or HDAC2, creating a rational "code" for a differential, context-related regulation. The global comprehension of this PTM code is central for dissecting the role of single HDAC1 and HDAC2 in physiology and pathology.
Collapse
Affiliation(s)
- Chiara V. Segré
- Department of Experimental Oncology, European Institute of Oncology, IFOM-IEO Campus, Via Adamello 16, 20139 Milan, Italy
| | - Susanna Chiocca
- Department of Experimental Oncology, European Institute of Oncology, IFOM-IEO Campus, Via Adamello 16, 20139 Milan, Italy
| |
Collapse
|
252
|
Bhaskara S, Knutson SK, Jiang G, Chandrasekharan MB, Wilson AJ, Zheng S, Yenamandra A, Locke K, Yuan JL, Bonine-Summers AR, Wells CE, Kaiser JF, Washington MK, Zhao Z, Wagner FF, Sun ZW, Xia F, Holson EB, Khabele D, Hiebert SW. Hdac3 is essential for the maintenance of chromatin structure and genome stability. Cancer Cell 2010; 18:436-47. [PMID: 21075309 PMCID: PMC3004468 DOI: 10.1016/j.ccr.2010.10.022] [Citation(s) in RCA: 291] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Revised: 06/16/2010] [Accepted: 08/23/2010] [Indexed: 01/10/2023]
Abstract
Hdac3 is essential for efficient DNA replication and DNA damage control. Deletion of Hdac3 impaired DNA repair and greatly reduced chromatin compaction and heterochromatin content. These defects corresponded to increases in histone H3K9,K14ac; H4K5ac; and H4K12ac in late S phase of the cell cycle, and histone deposition marks were retained in quiescent Hdac3-null cells. Liver-specific deletion of Hdac3 culminated in hepatocellular carcinoma. Whereas HDAC3 expression was downregulated in only a small number of human liver cancers, the mRNA levels of the HDAC3 cofactor NCOR1 were reduced in one-third of these cases. siRNA targeting of NCOR1 and SMRT (NCOR2) increased H4K5ac and caused DNA damage, indicating that the HDAC3/NCOR/SMRT axis is critical for maintaining chromatin structure and genomic stability.
Collapse
Affiliation(s)
- Srividya Bhaskara
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232
| | - Sarah K. Knutson
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232
| | - Guochun Jiang
- Department of Radiation Oncology Vanderbilt University Medical Center, Nashville, TN 37212
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37212
| | | | - Andrew J. Wilson
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Vanderbilt University Medical Center, Nashville, TN 37212
| | - Siyuan Zheng
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232
- Bioinformatics Resource Center, Vanderbilt University School of Medicine, Nashville, TN 37232
| | | | | | - Jia-ling Yuan
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232
| | | | | | | | - M. Kay Washington
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Zhongming Zhao
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37212
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232
- Bioinformatics Resource Center, Vanderbilt University School of Medicine, Nashville, TN 37232
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Florence F. Wagner
- The Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA 02142
| | - Zu-Wen Sun
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Fen Xia
- Department of Radiation Oncology Vanderbilt University Medical Center, Nashville, TN 37212
| | - Edward B. Holson
- The Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA 02142
| | - Dineo Khabele
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37212
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Vanderbilt University Medical Center, Nashville, TN 37212
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Scott W. Hiebert
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232
- To whom correspondence should be sent: Department of Biochemistry, 512 Preston Research Building, Vanderbilt University School of Medicine, 23rd and Pierce Ave., Nashville Tennessee, 37232, Phone: (615) 936-3582; Fax: (615) 936-1790;
| |
Collapse
|
253
|
Fischer A, Sananbenesi F, Mungenast A, Tsai LH. Targeting the correct HDAC(s) to treat cognitive disorders. Trends Pharmacol Sci 2010; 31:605-17. [PMID: 20980063 DOI: 10.1016/j.tips.2010.09.003] [Citation(s) in RCA: 286] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Revised: 09/15/2010] [Accepted: 09/16/2010] [Indexed: 01/18/2023]
Abstract
Changes in gene expression in the brain may underlie cognitive deficits inherent to normal aging and neurodegenerative disease. However, the mechanisms underlying pathological alterations in the brain transcriptome are incompletely understood. Epigenetic mechanisms such as DNA methylation and histone acetylation have been shown to be important for memory processes in the adult brain. There is accumulating evidence that altered chromatin plasticity and histone acetylation are also involved in cognitive aging, neurodegeneration, and neuropsychiatric diseases. Inhibitors of histone deacetylase (HDAC) exhibit neuroprotective and neuroregenerative properties in animal models of various brain diseases. As such, targeting of HDACs seems to be a promising therapeutic strategy. In this review, we discuss the specific roles of each HDAC protein and the possible function of distinct histone modifications. We hope that this knowledge will aid in the development of diagnostic tools and in designing more potent and specific treatment for neurological disorders targeting selective HDAC proteins.
Collapse
Affiliation(s)
- André Fischer
- Laboratory for Aging and Cognitive Diseases, European Neuroscience Institute, Grisebach Str. 5, D-37077 Goettingen, Germany.
| | | | | | | |
Collapse
|
254
|
Rossetto D, Truman AW, Kron SJ, Côté J. Epigenetic modifications in double-strand break DNA damage signaling and repair. Clin Cancer Res 2010; 16:4543-52. [PMID: 20823147 DOI: 10.1158/1078-0432.ccr-10-0513] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Factors involved in the cellular response to double-strand break (DSB) DNA damage have been identified as potential therapeutic targets that would greatly sensitize cancer cells to radiotherapy and genotoxic chemotherapy. These targets could disable the repair machinery and/or reinstate normal cell-cycle checkpoint leading to growth arrest, senescence, and apoptosis. It is now clear that a major aspect of the DNA damage response occurs through specific interactions with chromatin structure and its modulation. It implicates highly dynamic posttranslational modifications of histones that are critical for DNA damage recognition and/or signaling, repair of the lesion, and release of cell-cycle arrest. Therefore, drugs that target the enzymes responsible for these modifications, or the protein modules reading them, have very high therapeutic potential. This review presents the current state of knowledge on the different chromatin modifications and their roles in each step of eukaryotic DSB DNA damage response.
Collapse
Affiliation(s)
- Dorine Rossetto
- Laval University Cancer Research Center, Hôtel-Dieu de Québec, Quebec City, Canada
| | | | | | | |
Collapse
|
255
|
Chia K, Beamish H, Jafferi K, Gabrielli B. The histone deacetylase inhibitor MGCD0103 has both deacetylase and microtubule inhibitory activity. Mol Pharmacol 2010; 78:436-43. [PMID: 20538840 DOI: 10.1124/mol.110.065169] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Histone deacetylase inhibitors (HDACis) are currently in trial or are in clinical use for the treatment of a number of tumor types. The clinical efficacy of HDACis can be partly attributed to the modulation of the cell cycle by the HDACis. Here, we have examined the effects of N-(2-aminophenyl)-4-((4-pyridin-3-ylpyrimidin-2-ylamino)methyl)benzamide (MGCD0103), a class I-selective histone deacetylase inhibitor, on the cell cycle and cell killing. Surprisingly, MGCD0103 treatment failed to initiate a G(1)-phase arrest but caused marked accumulation of cells in G(2)/M at 6 and 12 h after treatment and was cytotoxic 24 h after treatment. These cell cycle effects were considerably distinct from the effects of suberic bishydroxamic acid, a representative of the pan-isoform HDACi used in this study. MGCD0103 shared the ability of the pan-isoform HDACi to trigger defective mitosis and promote mitotic slippage. Likewise, it also specifically targeted tumor cells and was nontoxic to normal nontransformed cells. However, MGDC0103 also seemed to disrupt normal microtubule spindle formation, whereas HDACis generally have only a minor effect on spindle formation. The effect of MGCD0103 on spindle formation was shown to be a consequence of microtubule destabilization. This is the first example of an HDACi with microtubule destabilizing activity, and the combined effects of this drug have advantages for its therapeutic use.
Collapse
Affiliation(s)
- Keeming Chia
- University of Queensland Diamantina Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | | | | | | |
Collapse
|
256
|
Razidlo DF, Whitney TJ, Casper ME, McGee-Lawrence ME, Stensgard BA, Li X, Secreto FJ, Knutson SK, Hiebert SW, Westendorf JJ. Histone deacetylase 3 depletion in osteo/chondroprogenitor cells decreases bone density and increases marrow fat. PLoS One 2010; 5:e11492. [PMID: 20628553 PMCID: PMC2901996 DOI: 10.1371/journal.pone.0011492] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 06/11/2010] [Indexed: 01/04/2023] Open
Abstract
Histone deacetylase (Hdac)3 is a nuclear enzyme that contributes to epigenetic programming and is required for embryonic development. To determine the role of Hdac3 in bone formation, we crossed mice harboring loxP sites around exon 7 of Hdac3 with mice expressing Cre recombinase under the control of the osterix promoter. The resulting Hdac3 conditional knockout (CKO) mice were runted and had severe deficits in intramembranous and endochondral bone formation. Calvarial bones were significantly thinner and trabecular bone volume in the distal femur was decreased 75% in the Hdac3 CKO mice due to a substantial reduction in trabecular number. Hdac3-CKO mice had fewer osteoblasts and more bone marrow adipocytes as a proportion of tissue area than their wildtype or heterozygous littermates. Bone formation rates were depressed in both the cortical and trabecular regions of Hdac3 CKO femurs. Microarray analyses revealed that numerous developmental signaling pathways were affected by Hdac3-deficiency. Thus, Hdac3 depletion in osterix-expressing progenitor cells interferes with bone formation and promotes bone marrow adipocyte differentiation. These results demonstrate that Hdac3 inhibition is detrimental to skeletal health.
Collapse
Affiliation(s)
- David F Razidlo
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | | | | | | | | | | | | | | | | | | |
Collapse
|
257
|
Flak MB, Connell CM, Chelala C, Archibald K, Salako MA, Pirlo KJ, Lockley M, Wheatley SP, Balkwill FR, McNeish IA. p21 Promotes oncolytic adenoviral activity in ovarian cancer and is a potential biomarker. Mol Cancer 2010; 9:175. [PMID: 20598155 PMCID: PMC2904726 DOI: 10.1186/1476-4598-9-175] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 07/03/2010] [Indexed: 11/22/2022] Open
Abstract
The oncolytic adenovirus dl922-947 replicates selectively within and lyses cells with a dysregulated Rb pathway, a finding seen in > 90% human cancers. dl922-947 is more potent than wild type adenovirus and the E1B-deletion mutant dl1520 (Onyx-015). We wished to determine which host cell factors influence cytotoxicity. SV40 large T-transformed MRC5-VA cells are 3-logs more sensitive to dl922-947 than isogenic parental MRC5 cells, confirming that an abnormal G1/S checkpoint increases viral efficacy. The sensitivity of ovarian cancer cells to dl922-947 varied widely: IC50 values ranged from 51 (SKOV3ip1) to 0.03 pfu/cell (TOV21G). Cells sensitive to dl922-947 had higher S phase populations and supported earlier E1A expression. Cytotoxicity correlated poorly with both infectivity and replication, but well with expression of p21 by microarray and western blot analyses. Matched p21+/+ and -/- Hct116 cells confirmed that p21 influences dl922-947 activity in vitro and in vivo. siRNA-mediated p21 knockdown in sensitive TOV21G cells decreases E1A expression and viral cytotoxicity, whilst expression of p21 in resistant A2780CP cells increases virus activity in vitro and in intraperitoneal xenografts. These results highlight that host cell factors beyond simple infectivity can influence the efficacy of oncolytic adenoviruses. p21 expression may be an important biomarker of response in clinical trials.
Collapse
Affiliation(s)
- Magdalena B Flak
- Centre for Molecular Oncology and Imaging, Institute of Cancer, Barts and the London School of Medicine, Queen Mary University of London, London EC1 M 6BQ, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
258
|
Khan SN, Khan AU. Role of histone acetylation in cell physiology and diseases: An update. Clin Chim Acta 2010; 411:1401-11. [PMID: 20598676 DOI: 10.1016/j.cca.2010.06.020] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 06/04/2010] [Accepted: 06/16/2010] [Indexed: 01/06/2023]
Abstract
Although the role of histone acetylation in gene regulation has been the subject of many reviews, their impact on cell physiology and pathological states of proliferation, differentiation and genome stability in eukaryotic cells remain to be elucidated. Therefore, this review will discuss the molecular, physiological and biochemical aspects of histone acetylation and focus on the interplay of histone acetyltransferases (HATs) and histone deacetylases (HDACs) in different disease states. Current treatment strategies are mostly limited to enzyme inhibitors, though potential lies in targeting other imperative chromatin remodeling factors involved in gene regulation.
Collapse
Affiliation(s)
- Shahper N Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | | |
Collapse
|
259
|
Wilting RH, Yanover E, Heideman MR, Jacobs H, Horner J, van der Torre J, DePinho RA, Dannenberg JH. Overlapping functions of Hdac1 and Hdac2 in cell cycle regulation and haematopoiesis. EMBO J 2010; 29:2586-97. [PMID: 20571512 DOI: 10.1038/emboj.2010.136] [Citation(s) in RCA: 186] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Accepted: 05/20/2010] [Indexed: 11/09/2022] Open
Abstract
Histone deacetylases (HDACs) counterbalance acetylation of lysine residues, a protein modification involved in numerous biological processes. Here, Hdac1 and Hdac2 conditional knock-out alleles were used to study the function of class I Hdac1 and Hdac2 in cell cycle progression and haematopoietic differentiation. Combined deletion of Hdac1 and Hdac2, or inactivation of their deacetylase activity in primary or oncogenic-transformed fibroblasts, results in a senescence-like G(1) cell cycle arrest, accompanied by up-regulation of the cyclin-dependent kinase inhibitor p21(Cip). Notably, concomitant genetic inactivation of p53 or p21(Cip) indicates that Hdac1 and Hdac2 regulate p53-p21(Cip)-independent pathways critical for maintaining cell cycle progression. In vivo, we show that Hdac1 and Hdac2 are not essential for liver homeostasis. In contrast, total levels of Hdac1 and Hdac2 in the haematopoietic system are critical for erythrocyte-megakaryocyte differentiation. Dual inactivation of Hdac1 and Hdac2 results in apoptosis of megakaryocytes and thrombocytopenia. Together, these data indicate that Hdac1 and Hdac2 have overlapping functions in cell cycle regulation and haematopoiesis. In addition, this work provides insights into mechanism-based toxicities observed in patients treated with HDAC inhibitors.
Collapse
Affiliation(s)
- Roel H Wilting
- Division of Molecular Genetics, Plesmanlaan 121, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
260
|
Marks PA. Histone deacetylase inhibitors: a chemical genetics approach to understanding cellular functions. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2010; 1799:717-25. [PMID: 20594930 DOI: 10.1016/j.bbagrm.2010.05.008] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Accepted: 05/28/2010] [Indexed: 01/29/2023]
Abstract
There are eleven zinc dependent histone deacetylases (HDAC) in humans which have histones and many non-histone substrates. The substrates of these enzymes include proteins that have a role in regulation of gene expression, cell proliferation, cell migration, cell death, immune pathways and angiogenesis. Inhibitors of HDACs (HDACi) have been developed which alter the structure and function of these proteins, causing molecular and cellular changes that induce transformed cell death. The HDACi are being developed as anti-cancer drugs and have therapeutic potential for many non-oncologic diseases.
Collapse
Affiliation(s)
- Paul A Marks
- Cell Biology and Genetics Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
261
|
Abstract
Endothelial dysfunction/death is an initial event for the development of atherosclerosis occurring in the areas of arteries where blood flow is disturbed. Recent data indicated that rapid turnover of endothelial cells in atherosclerosis-prone areas is followed by endothelial repair. The mechanisms of endothelial death/proliferation in these areas related to altered blood flow involve different signal pathways, for example, endoplasmic reticulum stress-X-box binding protein 1-caspase for apoptosis and vascular endothelial growth factor receptor-histone deacetylase 3-Akt for the survival. The present review aims to update the progress in endothelial turnover in response to disturbed flow and to discuss the underlining mechanisms in the development of atherosclerosis.
Collapse
Affiliation(s)
- Qingbo Xu
- Cardiovascular Division, King's College London BHF Center, London, United Kingdom.
| |
Collapse
|
262
|
Conti C, Leo E, Eichler GS, Sordet O, Martin MM, Fan A, Aladjem MI, Pommier Y. Inhibition of histone deacetylase in cancer cells slows down replication forks, activates dormant origins, and induces DNA damage. Cancer Res 2010; 70:4470-80. [PMID: 20460513 DOI: 10.1158/0008-5472.can-09-3028] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Protein acetylation is a reversible process regulated by histone deacetylases (HDAC) that is often altered in human cancers. Suberoylanilide hydroxamic acid (SAHA) is the first HDAC inhibitor to be approved for clinical use as an anticancer agent. Given that histone acetylation is a key determinant of chromatin structure, we investigated how SAHA may affect DNA replication and integrity to gain deeper insights into the basis for its anticancer activity. Nuclear replication factories were visualized with confocal immunofluorescence microscopy and single-replicon analyses were conducted by genome-wide molecular combing after pulse labeling with two thymidine analogues. We found that pharmacologic concentrations of SAHA induce replication-mediated DNA damage with activation of histone gammaH2AX. Single DNA molecule analyses indicated slowdown in replication speed along with activation of dormant replication origins in response to SAHA. Similar results were obtained using siRNA-mediated depletion of HDAC3 expression, implicating this HDAC member as a likely target in the SAHA response. Activation of dormant origins was confirmed by molecular analyses of the beta-globin locus control region. Our findings demonstrate that SAHA produces profound alterations in DNA replication that cause DNA damage, establishing a critical link between robust chromatin acetylation and DNA replication in human cancer cells.
Collapse
Affiliation(s)
- Chiara Conti
- Laboratory of Molecular Pharmacology, National Cancer Institute, NIH, Bethesda, Maryland 20892-4255, USA
| | | | | | | | | | | | | | | |
Collapse
|
263
|
Regulation of Epstein-Barr virus origin of plasmid replication (OriP) by the S-phase checkpoint kinase Chk2. J Virol 2010; 84:4979-87. [PMID: 20200249 DOI: 10.1128/jvi.01300-09] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The Epstein-Barr virus (EBV) origin of plasmid replication (OriP) is required for episome stability during latent infection. Telomere repeat factor 2 (TRF2) binds directly to OriP and facilitates DNA replication and plasmid maintenance. Recent studies have found that TRF2 interacts with the DNA damage checkpoint protein Chk2. We show here that Chk2 plays an important role in regulating OriP plasmid stability, chromatin modifications, and replication timing. The depletion of Chk2 by small interfering RNA (siRNA) leads to a reduction in DNA replication efficiency and a loss of OriP-dependent plasmid maintenance. This corresponds to a change in OriP replication timing and an increase in constitutive histone H3 acetylation. We show that Chk2 interacts with TRF2 in the early G(1)/S phase of the cell cycle. We also show that Chk2 can phosphorylate TRF2 in vitro at a consensus acceptor site in the amino-terminal basic domain of TRF2. TRF2 mutants with a serine-to-aspartic acid phosphomimetic substitution mutation were reduced in their ability to recruit the origin recognition complex (ORC) and stimulate OriP replication. We suggest that the Chk2 phosphorylation of TRF2 is important for coordinating ORC binding with chromatin remodeling during the early S phase and that a failure to execute these events leads to replication defects and plasmid instability.
Collapse
|
264
|
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
|
265
|
Abstract
NeuroD, a basic helix-loop-helix transcription factor, is capable of converting embryonic epidermal cells into neuronal cells. However, whether histone deacetylases (HDACs) are involved in the autoregulation of neuroD or not is unclear. In this study, neuroD expression was found to be significantly increased in the all-trans retinoid acid-treated P19 cells. Meanwhile, neuroD could itself enhance its promoter activity and mRNA expression. By using specific inhibitors to histone modification enzymes, HDAC3 was identified to specifically augment the autoactivation of neuroD in P19 cells. The data suggest that the elevation of HDAC3 and neuroD in all-trans retinoid acid-treated cells exponentially increases the neuroD expression and mediates an early commitment of P19 cells for neuronal differentiation.
Collapse
|
266
|
Oehme I, Deubzer HE, Lodrini M, Milde T, Witt O. Targeting of HDAC8 and investigational inhibitors in neuroblastoma. Expert Opin Investig Drugs 2010; 18:1605-17. [PMID: 19780707 DOI: 10.1517/14728220903241658] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Histone deacetylase (HDAC) inhibitors are an emerging class of promising novel anticancer drugs. However, little is known which one of the 11 classical HDAC family members is the most relevant drug target for therapy. The first Phase I/II trials show that unselective inhibition of HDACs causes a variety of side effects. Therefore, identification and selective targeting of the most critical tumor entity-relevant HDAC family member may reduce unspecific effects and increase antitumor efficacy in the future. Here, we review the clinical relevance of a particular HDAC family member, HDAC8, in neuroblastoma biology, a highly malignant embryonal childhood cancer. HDAC8 expression correlates with poor outcome in neuroblastoma and selective HDAC8 inhibition induces differentiation. In contrast, the targeting of other HDAC family members results in a completely different phenotype. Because HDAC8-selective inhibitors are available, HDAC8 may be a potential drug target for neuroblastoma differentiation therapy using selective inhibitors, avoiding unspecific side effects.
Collapse
Affiliation(s)
- Ina Oehme
- German Cancer Research Center, CCU Pediatric Oncology, INF 280, D-69120 Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
267
|
Li Y, Li X, Guo B. Chemopreventive agent 3,3'-diindolylmethane selectively induces proteasomal degradation of class I histone deacetylases. Cancer Res 2010; 70:646-54. [PMID: 20068155 DOI: 10.1158/0008-5472.can-09-1924] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
3,3'-Diindolylmethane (DIM) is an anticancer agent that induces cell cycle arrest and apoptosis through unknown mechanisms. Here, we report that DIM can selectively induce proteasome-mediated degradation of class I histone deacetylases (HDAC1, HDAC2, HDAC3, and HDAC8) without affecting the class II HDAC proteins. DIM induced downregulation of class I HDACs in human colon cancer cells in vitro and in vivo in tumor xenografts. HDAC depletion relieved HDAC-mediated transcriptional inhibition of the cyclin-dependent kinase inhibitors p21WAF1 and p27KIP2, significantly increasing their expression and triggering cell cycle arrest in the G(2) phase of the cell cycle. Additionally, HDAC depletion was associated with an induction of DNA damage that triggered apoptosis. Our findings indicate that DIM acts to selectively target the degradation of class I HDACs.
Collapse
Affiliation(s)
- Yongming Li
- Department of Pharmaceutical Sciences, College of Pharmacy, North Dakota State University, Fargo, North Dakota 58108, USA
| | | | | |
Collapse
|
268
|
Rosato RR, Kolla SS, Hock SK, Almenara JA, Patel A, Amin S, Atadja P, Fisher PB, Dent P, Grant S. Histone deacetylase inhibitors activate NF-kappaB in human leukemia cells through an ATM/NEMO-related pathway. J Biol Chem 2010; 285:10064-10077. [PMID: 20065354 DOI: 10.1074/jbc.m109.095208] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Mechanisms underlying histone deacetylase inhibitor (HDACI)-mediated NF-kappaB activation were investigated in human leukemia cells. Exposure of U937 and other leukemia cells to LBH-589 induced reactive oxygen species (ROS) followed by single strand (XRCC1) and double strand (gamma-H2AX) DNA breaks. Notably, LBH-589 lethality was markedly attenuated by small interfering RNA (siRNA) knockdown of the DNA damage-linked histone, H1.2. LBH-589 triggered p65/RelA activation, NF-kappaB-dependent induction of Mn-SOD2, and ROS elimination. Interference with LBH-589-mediated NF-kappaB activation (e.g. in I kappaB alpha super-repressor transfected cells) diminished HDACI-mediated Mn-SOD2 induction and increased ROS accumulation, DNA damage, and apoptosis. The Mn-SOD2 mimetic TBAP (manganese(III)-tetrakis 4-benzoic acid porphyrin) prevented HDACI-induced ROS and NF-kappaB activation while dramatically attenuating DNA damage and cell death. In contrast, TRAF2 siRNA knockdown, targeting receptor-mediated NF-kappaB activation, blocked TNFalpha- but not HDACI-mediated NF-kappaB activation and lethality. Consistent with ROS-mediated DNA damage, LBH-589 exposure activated ATM (on serine 1981) and increased its association with NEMO. Significantly, siRNA NEMO or ATM knockdown blocked HDACI-mediated NF-kappaB activation, resulting in diminished MnSOD2 induction and enhanced oxidative DNA damage and cell death. In accord with the recently described DNA damage/ATM/NEMO pathway, SUMOylation site mutant NEMO (K277A or K309A) cells exposed to LBH-589 displayed diminished ATM/NEMO association, NEMO and p65/RelA nuclear localization/activation, and MnSOD2 up-regulation. These events were accompanied by increased ROS production, gamma-H2AX formation, and cell death. Together, these findings indicate that in human leukemia cells, HDACIs activate the cytoprotective NF-kappaB pathway through an ATM/NEMO/SUMOylation-dependent process involving the induction of ROS and DNA damage and suggest that blocking NF-kappaB activation via the atypical ATM/NEMO nuclear pathway can enhance HDACI antileukemic activity.
Collapse
Affiliation(s)
| | | | | | | | - Ankita Patel
- Departments of Medicine, Richmond, Virginia 23298
| | - Sanjay Amin
- Departments of Medicine, Richmond, Virginia 23298
| | - Peter Atadja
- Department of Oncology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139
| | - Paul B Fisher
- Human and Molecular Genetics, Richmond, Virginia 23298; Virginia Commonwealth University Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Paul Dent
- Biochemistry, the Massey Cancer Center, Richmond, Virginia 23298; Virginia Commonwealth University Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Steven Grant
- Departments of Medicine, Richmond, Virginia 23298; Biochemistry, the Massey Cancer Center, Richmond, Virginia 23298; Virginia Commonwealth University Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, Virginia 23298.
| |
Collapse
|
269
|
Zampetaki A, Zeng L, Margariti A, Xiao Q, Li H, Zhang Z, Pepe AE, Wang G, Habi O, deFalco E, Cockerill G, Mason JC, Hu Y, Xu Q. Histone Deacetylase 3 Is Critical in Endothelial Survival and Atherosclerosis Development in Response to Disturbed Flow. Circulation 2010; 121:132-42. [DOI: 10.1161/circulationaha.109.890491] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Anna Zampetaki
- From the Cardiovascular Division, King’s College London BHF Centre (A.Z., L.Z., A.M., Q.X., H.L., Z.Z., A.E.P., G.W., O.H., E.d.F., Y.H., Q.X.); Department of Cardiovascular Medicine, St George’s University of London (G.C.); and Bywaters Center for Vascular Inflammation, Imperial College, Hammersmith Hospital (J.C.M.), London, UK
| | - Lingfang Zeng
- From the Cardiovascular Division, King’s College London BHF Centre (A.Z., L.Z., A.M., Q.X., H.L., Z.Z., A.E.P., G.W., O.H., E.d.F., Y.H., Q.X.); Department of Cardiovascular Medicine, St George’s University of London (G.C.); and Bywaters Center for Vascular Inflammation, Imperial College, Hammersmith Hospital (J.C.M.), London, UK
| | - Andriana Margariti
- From the Cardiovascular Division, King’s College London BHF Centre (A.Z., L.Z., A.M., Q.X., H.L., Z.Z., A.E.P., G.W., O.H., E.d.F., Y.H., Q.X.); Department of Cardiovascular Medicine, St George’s University of London (G.C.); and Bywaters Center for Vascular Inflammation, Imperial College, Hammersmith Hospital (J.C.M.), London, UK
| | - Qingzhong Xiao
- From the Cardiovascular Division, King’s College London BHF Centre (A.Z., L.Z., A.M., Q.X., H.L., Z.Z., A.E.P., G.W., O.H., E.d.F., Y.H., Q.X.); Department of Cardiovascular Medicine, St George’s University of London (G.C.); and Bywaters Center for Vascular Inflammation, Imperial College, Hammersmith Hospital (J.C.M.), London, UK
| | - Hongling Li
- From the Cardiovascular Division, King’s College London BHF Centre (A.Z., L.Z., A.M., Q.X., H.L., Z.Z., A.E.P., G.W., O.H., E.d.F., Y.H., Q.X.); Department of Cardiovascular Medicine, St George’s University of London (G.C.); and Bywaters Center for Vascular Inflammation, Imperial College, Hammersmith Hospital (J.C.M.), London, UK
| | - Zhongyi Zhang
- From the Cardiovascular Division, King’s College London BHF Centre (A.Z., L.Z., A.M., Q.X., H.L., Z.Z., A.E.P., G.W., O.H., E.d.F., Y.H., Q.X.); Department of Cardiovascular Medicine, St George’s University of London (G.C.); and Bywaters Center for Vascular Inflammation, Imperial College, Hammersmith Hospital (J.C.M.), London, UK
| | - Anna Elena Pepe
- From the Cardiovascular Division, King’s College London BHF Centre (A.Z., L.Z., A.M., Q.X., H.L., Z.Z., A.E.P., G.W., O.H., E.d.F., Y.H., Q.X.); Department of Cardiovascular Medicine, St George’s University of London (G.C.); and Bywaters Center for Vascular Inflammation, Imperial College, Hammersmith Hospital (J.C.M.), London, UK
| | - Gang Wang
- From the Cardiovascular Division, King’s College London BHF Centre (A.Z., L.Z., A.M., Q.X., H.L., Z.Z., A.E.P., G.W., O.H., E.d.F., Y.H., Q.X.); Department of Cardiovascular Medicine, St George’s University of London (G.C.); and Bywaters Center for Vascular Inflammation, Imperial College, Hammersmith Hospital (J.C.M.), London, UK
| | - Ouassila Habi
- From the Cardiovascular Division, King’s College London BHF Centre (A.Z., L.Z., A.M., Q.X., H.L., Z.Z., A.E.P., G.W., O.H., E.d.F., Y.H., Q.X.); Department of Cardiovascular Medicine, St George’s University of London (G.C.); and Bywaters Center for Vascular Inflammation, Imperial College, Hammersmith Hospital (J.C.M.), London, UK
| | - Elena deFalco
- From the Cardiovascular Division, King’s College London BHF Centre (A.Z., L.Z., A.M., Q.X., H.L., Z.Z., A.E.P., G.W., O.H., E.d.F., Y.H., Q.X.); Department of Cardiovascular Medicine, St George’s University of London (G.C.); and Bywaters Center for Vascular Inflammation, Imperial College, Hammersmith Hospital (J.C.M.), London, UK
| | - Gillian Cockerill
- From the Cardiovascular Division, King’s College London BHF Centre (A.Z., L.Z., A.M., Q.X., H.L., Z.Z., A.E.P., G.W., O.H., E.d.F., Y.H., Q.X.); Department of Cardiovascular Medicine, St George’s University of London (G.C.); and Bywaters Center for Vascular Inflammation, Imperial College, Hammersmith Hospital (J.C.M.), London, UK
| | - Justin C. Mason
- From the Cardiovascular Division, King’s College London BHF Centre (A.Z., L.Z., A.M., Q.X., H.L., Z.Z., A.E.P., G.W., O.H., E.d.F., Y.H., Q.X.); Department of Cardiovascular Medicine, St George’s University of London (G.C.); and Bywaters Center for Vascular Inflammation, Imperial College, Hammersmith Hospital (J.C.M.), London, UK
| | - Yanhua Hu
- From the Cardiovascular Division, King’s College London BHF Centre (A.Z., L.Z., A.M., Q.X., H.L., Z.Z., A.E.P., G.W., O.H., E.d.F., Y.H., Q.X.); Department of Cardiovascular Medicine, St George’s University of London (G.C.); and Bywaters Center for Vascular Inflammation, Imperial College, Hammersmith Hospital (J.C.M.), London, UK
| | - Qingbo Xu
- From the Cardiovascular Division, King’s College London BHF Centre (A.Z., L.Z., A.M., Q.X., H.L., Z.Z., A.E.P., G.W., O.H., E.d.F., Y.H., Q.X.); Department of Cardiovascular Medicine, St George’s University of London (G.C.); and Bywaters Center for Vascular Inflammation, Imperial College, Hammersmith Hospital (J.C.M.), London, UK
| |
Collapse
|
270
|
The cyclin-dependent kinase inhibitor p21 is a crucial target for histone deacetylase 1 as a regulator of cellular proliferation. Mol Cell Biol 2009; 30:1171-81. [PMID: 20028735 DOI: 10.1128/mcb.01500-09] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Histone deacetylases (HDACs) are chromatin-modifying enzymes that are involved in the regulation of proliferation, differentiation and development. HDAC inhibitors induce cell cycle arrest, differentiation, or apoptosis in tumor cells and are therefore promising antitumor agents. Numerous genes were found to be deregulated upon HDAC inhibitor treatment; however, the relevant target enzymes are still unidentified. HDAC1 is required for mouse development and unrestricted proliferation of embryonic stem cells. We show here that HDAC1 reversibly regulates cellular proliferation and represses the cyclin-dependent kinase inhibitor p21 in embryonic stem cells. Disruption of the p21 gene rescues the proliferation phenotype of HDAC1(-/-) embryonic stem cells but not the embryonic lethality of HDAC1(-/-) mice. In the absence of HDAC1, mouse embryonic fibroblasts scarcely undergo spontaneous immortalization and display increased p21 expression. Chromatin immunoprecipitation assays demonstrate a direct regulation of the p21 gene by HDAC1 in mouse embryonic fibroblasts. Transformation with simian virus 40 large T antigen or ablation of p21 restores normal immortalization of primary HDAC1(-/-) fibroblasts. Our data demonstrate that repression of the p21 gene is crucial for HDAC1-mediated control of proliferation and immortalization. HDAC1 might therefore be one of the relevant targets for HDAC inhibitors as anticancer drugs.
Collapse
|
271
|
Godmann M, Lambrot R, Kimmins S. The dynamic epigenetic program in male germ cells: Its role in spermatogenesis, testis cancer, and its response to the environment. Microsc Res Tech 2009; 72:603-19. [PMID: 19319879 DOI: 10.1002/jemt.20715] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Spermatogenesis is a truly remarkable process that requires exquisite control and synchronization of germ cell development. It is prone to frequent error, as paternal infertility contributes to 30-50% of all infertility cases; yet, in many cases, the mechanisms underlying its causes are unknown. Strikingly, aberrant epigenetic profiles, in the form of anomalous DNA and histone modifications, are characteristic of cancerous testis cells. Germ cell development is a critical period during which epigenetic patterns are established and maintained. The progression from diploid spermatogonia to haploid spermatozoa involves stage- and testis-specific gene expression, mitotic and meiotic division, and the histone-protamine transition. All are postulated to engender unique epigenetic controls. In support of this idea are the findings that mouse models with gene deletions for epigenetic modifiers have severely compromised fertility. Underscoring the importance of understanding how epigenetic marks are set and interpreted is evidence that abnormal epigenetic programming of gametes and embryos contributes to heritable instabilities in subsequent generations. Numerous studies have documented the existence of transgenerational consequences of maternal nutrition, or other environmental exposures, but it is only now recognized that there are sex-specific male-line transgenerational responses in humans and other species. Epigenetic events in the testis have just begun to be studied. New work on the function of specific histone modifications, chromatin modifiers, DNA methylation, and the impact of the environment on developing sperm suggests that the correct setting of the epigenome is required for male reproductive health and the prevention of paternal disease transmission.
Collapse
Affiliation(s)
- Maren Godmann
- Department of Animal Science, McGill University, Montreal H9X3V9 Canada
| | | | | |
Collapse
|
272
|
Bandyopadhyay K, Banères JL, Martin A, Blonski C, Parello J, Gjerset RA. Spermidinyl-CoA-based HAT inhibitors block DNA repair and provide cancer-specific chemo- and radiosensitization. Cell Cycle 2009; 8:2779-88. [PMID: 19652528 DOI: 10.4161/cc.8.17.9416] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Acetyl group turnover on specific lysine epsilon-amino groups of the core chromosomal histones regulates DNA accessibility function, and the acetylating and deacetylating enzymes that govern the turnover provide important targets for the development of anti-cancer drugs. Histone deacetylase (HDAC) inhibitors have been developed and evaluated extensively in clinical trials, while the development of inhibitors of histone acetyltransferase (HAT) has proceeded more slowly. Here we have examined the cellular effects of an S-substituted coenzyme A (CoA) inhibitor of histone acetylation, consisting of spermidine (Spd) linked to the S-terminus of CoA through a thioglycolic acid linkage (adduct abbreviated as Spd-CoA), as well as the effects of a truncated Spd-CoA derivative lacking the negatively charged portion of the CoA moiety. While exposure of cancer cells to Spd-CoA has little effect on cell viability, it causes a rapid inhibition of histone acetylation that correlates with a transient arrest of DNA synthesis, a transient delay in S-phase progression, and an inhibition of nucleotide excision repair and DNA double strand break repair. These effects correlate with increased cellular sensitivity to the DNA-targeted chemotherapeutic drugs, cisplatin (Platinol()) and 5-fluorouracil, to the DNA damaging drug, camptothecin, and to UV-C irradiation. The sensitization effects of Spd-CoA are not observed in normal cells due to a barrier to uptake. The truncated Spd-CoA derivative displays similar but enhanced chemosensitization effects, suggesting that further modifications of the Spd-CoA structure could further improve potency. The results demonstrate that Spd-CoA and its truncated version are efficiently and selectively internalized into cancer cells, and suggest that the resulting inhibition of acetylation-dependent DNA repair enhances cellular sensitivity to DNA damage. These and related inhibitors of histone acetylation could therefore constitute a novel class of potent therapy sensitizers applicable to a broad range of conventional cancer treatments.
Collapse
|
273
|
Horiuchi M, Morinobu A, Chin T, Sakai Y, Kurosaka M, Kumagai S. Expression and function of histone deacetylases in rheumatoid arthritis synovial fibroblasts. J Rheumatol 2009; 36:1580-9. [PMID: 19531758 DOI: 10.3899/jrheum.081115] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE To explore the effects of histone deacetylases (HDAC) on rheumatoid arthritis synovial fibroblasts (RA-SF). METHODS The expression of mRNA encoding HDAC1 through HDAC11 in RA-SF and osteoarthritis-SF (OA-SF) was determined using real-time polymerase chain reactions. The functions of HDAC1 and HDAC2 in RA-SF were assessed using small interfering RNA (siRNA) technology. Cell counts and proliferation were examined by MTT assays and BrDU ELISA, respectively, and apoptosis was determined using the TUNEL assay and annexin V staining. Levels of cell cycle-related molecules and matrix metalloproteinases (MMP) were tested by Western blotting and ELISA, respectively. RESULTS Messenger RNA expression of HDAC1 was significantly higher in RA-SF than in OA-SF. Knockdown of HDAC1 and HDAC2 by siRNA resulted in decreased cell counts and cell proliferation, and increased apoptosis in RA-SF. Expression of p16, p21, and p53 was increased by knockdown of both HDAC1 and HDAC2. On the other hand, knockdown of HDAC1, but not of HDAC2, upregulated tumor necrosis factor-alpha-induced MMP-1 production by RA-SF. CONCLUSION HDAC1 is overexpressed in RA-SF compared to OA-SF. HDAC1 supports cell proliferation and survival of RA-SF, but suppresses MMP-1 production. HDAC2 also plays an important role in cell proliferation and apoptosis of RA-SF. Our study provides useful information to develop new HDAC inhibitors for the treatment of RA.
Collapse
Affiliation(s)
- Marika Horiuchi
- Department of Clinical Pathology and Immunology, Kobe University School of Medicine, Kobe, Japan
| | | | | | | | | | | |
Collapse
|
274
|
Schrump DS. Cytotoxicity mediated by histone deacetylase inhibitors in cancer cells: mechanisms and potential clinical implications. Clin Cancer Res 2009; 15:3947-57. [PMID: 19509170 DOI: 10.1158/1078-0432.ccr-08-2787] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Aberrant expression of epigenetic regulators of gene expression contributes to initiation and progression of cancer. During recent years, considerable research efforts have focused on the role of histone acetyltransferases (HATs) and histone deacetylases (HDACs) in cancer cells, and the identification of pharmacologic agents that modulate gene expression via inhibition of HDACs. The following review highlights recent studies pertaining to HDAC expression in cancer cells, the plieotropic mechanisms by which HDAC inhibitors (HDACi) mediate antitumor activity, and the potential clinical implications of HDAC inhibition as a strategy for cancer therapy.
Collapse
Affiliation(s)
- David S Schrump
- Thoracic Oncology Section, Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892-1201, USA.
| |
Collapse
|
275
|
Ellis L, Atadja PW, Johnstone RW. Epigenetics in cancer: targeting chromatin modifications. Mol Cancer Ther 2009; 8:1409-20. [PMID: 19509247 DOI: 10.1158/1535-7163.mct-08-0860] [Citation(s) in RCA: 330] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Posttranslational modifications to histones affect chromatin structure and function resulting in altered gene expression and changes in cell behavior. Aberrant gene expression and altered epigenomic patterns are major features of cancer. Epigenetic changes including histone acetylation, histone methylation, and DNA methylation are now thought to play important roles in the onset and progression of cancer in numerous tumor types. Indeed dysregulated epigenetic modifications, especially in early neoplastic development, may be just as significant as genetic mutations in driving cancer development and growth. The reversal of aberrant epigenetic changes has therefore emerged as a potential strategy for the treatment of cancer. A number of compounds targeting enzymes that regulate histone acetylation, histone methylation, and DNA methylation have been developed as epigenetic therapies, with some demonstrating efficacy in hematological malignancies and solid tumors. This review highlights the roles of epigenetic modifications to histones and DNA in tumorigenesis and emerging epigenetic therapies being developed for the treatment of cancer.
Collapse
Affiliation(s)
- Leigh Ellis
- Peter MacCallum Cancer Center, St. Andrews Place, East Melbourne 3002, Australia
| | | | | |
Collapse
|
276
|
Haberland M, Johnson A, Mokalled MH, Montgomery RL, Olson EN. Genetic dissection of histone deacetylase requirement in tumor cells. Proc Natl Acad Sci U S A 2009; 106:7751-5. [PMID: 19416910 PMCID: PMC2683118 DOI: 10.1073/pnas.0903139106] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Indexed: 11/18/2022] Open
Abstract
Histone deacetylase inhibitors (HDACi) represent a new group of drugs currently being tested in a wide variety of clinical applications. They are especially effective in preclinical models of cancer where they show antiproliferative action in many different types of cancer cells. Recently, the first HDACi was approved for the treatment of cutaneous T cell lymphomas. Most HDACi currently in clinical development act by unspecifically interfering with the enzymatic activity of all class I HDACs (HDAC1, 2, 3, and 8), and it is widely believed that the development of isoform-specific HDACi could lead to better therapeutic efficacy. The contribution of the individual class I HDACs to different disease states, however, has so far not been fully elucidated. Here, we use a genetic approach to dissect the involvement of the different class I HDACs in tumor cells. We show that deletion of a single HDAC is not sufficient to induce cell death, but that HDAC1 and 2 play redundant and essential roles in tumor cell survival. Their deletion leads to nuclear bridging, nuclear fragmentation, and mitotic catastrophe, mirroring the effects of HDACi on cancer cells. These findings suggest that pharmacological inhibition of HDAC1 and 2 may be sufficient for anticancer activity, providing an experimental framework for the development of isoform-specific HDAC inhibitors.
Collapse
Affiliation(s)
- Michael Haberland
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9148
| | - Aaron Johnson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9148
| | - Mayssa H. Mokalled
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9148
| | - Rusty L. Montgomery
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9148
| | - Eric N. Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9148
| |
Collapse
|
277
|
New insights into the functions and regulation of the transcriptional corepressors SMRT and N-CoR. Cell Div 2009; 4:7. [PMID: 19383165 PMCID: PMC2678994 DOI: 10.1186/1747-1028-4-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Accepted: 04/21/2009] [Indexed: 11/10/2022] Open
Abstract
Corepressors are large proteins that facilitate transcriptional repression through recruitment of histone-modifying enzymes. Two major corepressors, SMRT (silencing mediator for retinoid and thyroid hormone receptors) and N-CoR (nuclear receptor corepressor), have been shown to mediate repression associated with nuclear receptors and a myriad of other transcription factors. This review will focus on recent studies on these proteins, including newly discovered physiological roles of the corepressors, their modes of regulation, their roles in antiestrogen-resistant breast cancer and their functions during the cell cycle.
Collapse
|
278
|
The many roles of histone deacetylases in development and physiology: implications for disease and therapy. Nat Rev Genet 2009; 10:32-42. [PMID: 19065135 DOI: 10.1038/nrg2485] [Citation(s) in RCA: 1961] [Impact Index Per Article: 122.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Histone deacetylases (HDACs) are part of a vast family of enzymes that have crucial roles in numerous biological processes, largely through their repressive influence on transcription. The expression of many HDAC isoforms in eukaryotic cells raises questions about their possible specificity or redundancy, and whether they control global or specific programmes of gene expression. Recent analyses of HDAC knockout mice have revealed highly specific functions of individual HDACs in development and disease. Mutant mice lacking individual HDACs are a powerful tool for defining the functions of HDACs in vivo and the molecular targets of HDAC inhibitors in disease.
Collapse
|
279
|
Mastroeni D, Grover A, Delvaux E, Whiteside C, Coleman PD, Rogers J. Epigenetic changes in Alzheimer's disease: decrements in DNA methylation. Neurobiol Aging 2008; 31:2025-37. [PMID: 19117641 DOI: 10.1016/j.neurobiolaging.2008.12.005] [Citation(s) in RCA: 265] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2008] [Revised: 12/10/2008] [Accepted: 12/10/2008] [Indexed: 12/27/2022]
Abstract
DNA methylation is a vital component of the epigenetic machinery that orchestrates changes in multiple genes and helps regulate gene expression in all known vertebrates. We evaluated immunoreactivity for two markers of DNA methylation and eight methylation maintenance factors in entorhinal cortex layer II, a region exhibiting substantial Alzheimer's disease (AD) pathology in which expression changes have been reported for a wide variety of genes. We show, for the first time, neuronal immunoreactivity for all 10 of the epigenetic markers and factors, with highly significant decrements in AD cases. These decrements were particularly marked in PHF1/PS396 immunoreactive, neurofibrillary tangle-bearing neurons. In addition, two of the DNA methylation maintenance factors, DNMT1 and MBD2, have been reported also to interact with ribosomal RNAs and ribosome synthesis. Consistent with these findings, DNMT1 and MBD2, as well as p66α, exhibited punctate cytoplasmic immunoreactivity that co-localized with the ribosome markers RPL26 and 5.8s rRNA in ND neurons. By contrast, AD neurons generally lacked such staining, and there was a qualitative decrease in RPL26 and 5.8s rRNA immunoreactivity. Collectively, these findings suggest epigenetic dysfunction in AD-vulnerable neurons.
Collapse
Affiliation(s)
- Diego Mastroeni
- L.J. Roberts Center for Alzheimer's Research, Sun Health Research Institute, P.O. Box 1278, Sun City, AZ 85372, USA
| | | | | | | | | | | |
Collapse
|
280
|
Wardell SE, Ilkayeva OR, Wieman HL, Frigo DE, Rathmell JC, Newgard CB, McDonnell DP. Glucose metabolism as a target of histone deacetylase inhibitors. Mol Endocrinol 2008; 23:388-401. [PMID: 19106193 DOI: 10.1210/me.2008-0179] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The therapeutic efficacy of histone deacetylase inhibitors (HDACI) is generally attributed to their ability to alter gene expression secondary to their effects on the acetylation status of transcription factors and histones. However, because HDACIs exhibit similar transcriptional effects in most cells, the molecular basis for their therapeutic selectivity toward malignant cells is largely unknown. In this study, we report that HDACI, of distinct chemotypes, quantitatively inhibit glucose transporter 1 (GLUT1)-mediated glucose transport into multiple myeloma cells through both down-regulation of GLUT1 and inhibition of hexokinase 1 (HXK1) enzymatic activity. Unexpectedly, however, this inhibition of glucose utilization is accompanied by an increase in amino acid catabolism with no increase in fatty acid oxidation. Our findings suggest that an HDACI-induced change in carbon source preference could contribute to the therapeutic efficacy of these drugs by creating a pattern of fuel utilization that is incompatible with rapid tumor growth and survival. Furthermore, these results, which implicate glucose metabolism as a target of HDACI, suggest that caution should be exercised in attributing effects of this class of drug to primary alterations in gene transcription.
Collapse
Affiliation(s)
- Suzanne E Wardell
- Duke University Medical Center, Pharmacology and Cancer Biology, Box 3813, Durham, North Carolina 27710, USA
| | | | | | | | | | | | | |
Collapse
|
281
|
SIRT1 redistribution on chromatin promotes genomic stability but alters gene expression during aging. Cell 2008; 135:907-18. [PMID: 19041753 DOI: 10.1016/j.cell.2008.10.025] [Citation(s) in RCA: 638] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Revised: 08/19/2008] [Accepted: 10/10/2008] [Indexed: 11/22/2022]
Abstract
Genomic instability and alterations in gene expression are hallmarks of eukaryotic aging. The yeast histone deacetylase Sir2 silences transcription and stabilizes repetitive DNA, but during aging or in response to a DNA break, the Sir complex relocalizes to sites of genomic instability, resulting in the desilencing of genes that cause sterility, a characteristic of yeast aging. Using embryonic stem cells, we show that mammalian Sir2, SIRT1, represses repetitive DNA and a functionally diverse set of genes across the mouse genome. In response to DNA damage, SIRT1 dissociates from these loci and relocalizes to DNA breaks to promote repair, resulting in transcriptional changes that parallel those in the aging mouse brain. Increased SIRT1 expression promotes survival in a mouse model of genomic instability and suppresses age-dependent transcriptional changes. Thus, DNA damage-induced redistribution of SIRT1 and other chromatin-modifying proteins may be a conserved mechanism of aging in eukaryotes.
Collapse
|
282
|
Alenghat T, Meyers K, Mullican SE, Leitner K, Adeniji-Adele A, Avila J, Bućan M, Ahima RS, Kaestner KH, Lazar MA. Nuclear receptor corepressor and histone deacetylase 3 govern circadian metabolic physiology. Nature 2008; 456:997-1000. [PMID: 19037247 PMCID: PMC2742159 DOI: 10.1038/nature07541] [Citation(s) in RCA: 226] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Accepted: 10/14/2008] [Indexed: 11/08/2022]
Abstract
Rhythmic changes in histone acetylation at circadian clock genes suggest that temporal modulation of gene expression is regulated by chromatin modifications. Furthermore, recent studies demonstrate a critical relationship between circadian and metabolic physiology. The nuclear receptor corepressor 1 (Ncor1) functions as an activating subunit for the chromatin modifying enzyme histone deacetylase 3 (Hdac3). Lack of Ncor1 is incompatible with life, and hence it is unknown whether Ncor1, and particularly its regulation of Hdac3, is critical for adult mammalian physiology. Here we show that specific, genetic disruption of the Ncor1-Hdac3 interaction in mice causes aberrant regulation of clock genes and results in abnormal circadian behaviour. These mice are also leaner and more insulin-sensitive owing to increased energy expenditure. Unexpectedly, loss of a functional Ncor1-Hdac3 complex in vivo does not lead to sustained increases in known catabolic genes, but instead significantly alters the oscillatory patterns of several metabolic genes, demonstrating that circadian regulation of metabolism is critical for normal energy balance. These findings indicate that activation of Hdac3 by Ncor1 is a nodal point in the epigenetic regulation of circadian and metabolic physiology.
Collapse
Affiliation(s)
- Theresa Alenghat
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 USA
- The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 USA
| | - Katherine Meyers
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 USA
- The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 USA
| | - Stannon E. Mullican
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 USA
- The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 USA
| | - Kirstin Leitner
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 USA
- The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 USA
| | - Adetoun Adeniji-Adele
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 USA
| | - Jacqueline Avila
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 USA
- The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 USA
| | - Maja Bućan
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 USA
| | - Rexford S. Ahima
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 USA
- The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 USA
| | - Klaus H. Kaestner
- The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 USA
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 USA
| | - Mitchell A. Lazar
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 USA
- The Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104 USA
| |
Collapse
|
283
|
Abstract
The temporal regulation of DNA replication is thought to be important for chromosome organization and genome stability. We show here that Epstein-Barr virus (EBV) genomes replicate in mid- to late S phase and that agents that accelerate replication timing of EBV reduce viral genome stability. Hydroxyurea (HU) treatment, which is known to eliminate EBV episomes, shifted EBV replication to earlier times in the cell cycle. HU treatment correlated with hyperacetylation of histone H3 and loss of telomere repeat factor 2 (TRF2) binding at the EBV origin of plasmid replication (OriP). Deletion of TRF2 binding sites within OriP or short hairpin RNA depletion of TRF2 advanced the replication timing of OriP-containing plasmids. Inhibitors of class I histone deacetylases (HDACs) increased histone acetylation at OriP, advanced the replication timing of EBV, and reduced EBV genome copy number. We also show that HDAC1 and -2 form a stable complex with TRF2 at OriP and that HU treatment inhibits HDAC activity. We propose that the TRF2-HDAC complex enhances EBV episome stability by providing a checkpoint that delays replication initiation at OriP.
Collapse
|
284
|
Making copies of chromatin: the challenge of nucleosomal organization and epigenetic information. Trends Cell Biol 2008; 19:29-41. [PMID: 19027300 DOI: 10.1016/j.tcb.2008.10.002] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2008] [Revised: 10/22/2008] [Accepted: 10/23/2008] [Indexed: 12/18/2022]
Abstract
Understanding the basic mechanisms underlying chromatin dynamics during DNA replication in eukaryotic cells is of fundamental importance. Beyond DNA compaction, chromatin organization represents a means to regulate genome function. Thus, the inheritance and maintenance of the DNA sequence, along with its organization into chromatin, is central for eukaryotic life. To orchestrate DNA replication in the context of chromatin is a challenge, both in terms of accessibility to the compact structures and maintenance of chromatin organization. To meet the challenge of maintenance, cells have evolved efficient nucleosome dynamics involving assembly pathways and chromatin maturation mechanisms that restore chromatin organization in the wake of DNA replication. In this review, we describe our current knowledge concerning how these pathways operate at the nucleosomal level and highlight the key players, such as histone chaperones, chromatin remodelers or modifiers, involved in the process of chromatin duplication. Major advances have been made recently concerning de novo nucleosome assembly and our understanding of its coordination with recycling of parental histones is progressing. Insights into the transmission of chromatin-based information during replication have important implications in the field of epigenetics to fully comprehend how the epigenetic landscape might, or at times might not, be stably maintained in the face of dramatic changes in chromatin structure.
Collapse
|
285
|
Blanco-García N, Asensio-Juan E, de la Cruz X, Martínez-Balbás MA. Autoacetylation regulates P/CAF nuclear localization. J Biol Chem 2008; 284:1343-52. [PMID: 19015268 DOI: 10.1074/jbc.m806075200] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Acetylation is a posttranslational modification that alters the biological activities of proteins by affecting their association with other proteins or DNA, their catalytic activities, or their subcellular distribution. The acetyltransferase P/CAF is autoacetylated and acetylated by p300 in vivo. P/CAF autoacetylation is an intramolecular or intermolecular event. Intramolecular acetylation targets five lysines within the nuclear localization signal at the P/CAF C terminus. We analyzed how the subcellular distribution of P/CAF is regulated by intramolecular autoacetylation and found that a P/CAF mutant lacking histone acetyltransferase activity accumulated primarily in the cytoplasm. This cytoplasmic fraction of P/CAF is enriched for nonautoacetylated P/CAF. In addition, P/CAF deacetylation by HDAC3 and in a minor degree by HDAC1, HDAC2, or HDAC4 leads to cytoplasmic accumulation of P/CAF. Importantly, our data show that P/CAF accumulates in the cytoplasm during apoptosis. These results reveal the molecular mechanism of autoacetylation control of P/CAF nuclear translocation and suggest a novel pathway by which P/CAF activity is controlled in vivo.
Collapse
Affiliation(s)
- Noemí Blanco-García
- Instituto de Biología Molecular de Barcelona, Consejo Superior de Investigaciones Científicas, Baldiri i Reixac 15-21, Parc Cientific de Barcelona, E-08028 Barcelona, Spain
| | | | | | | |
Collapse
|
286
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide an overview of recent advances in the development of histone deacetylase inhibitors (HDACi) for the treatment of cancer. RECENT FINDINGS Recently, there has been a dramatic expansion of HDACi clinical investigation. There are now 11 HDACi in clinical trial, including inhibitors with a broad spectrum of HDAC isoform inhibitory activity as well as drugs with isoform selectivity. Over 70 combination therapy trials are in progress. Major areas of progress covered include the entry of new HDAC inhibitors into clinical development, recent progress in understanding of molecular mechanisms of HDACi anticancer activity, and a preclinical and clinical update on HDACi in combination. SUMMARY In the period under review there have been advances in understanding of HDACi mechanisms of action, identification of rational combinations that address increased efficacy and overcoming resistance, and greatly expanded clinical development of pan-HDAC-inhibitory and isoform-selective inhibitors in monotherapy and combination therapy protocols.
Collapse
Affiliation(s)
- Min-Jung Lee
- Medical Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|
287
|
Affiliation(s)
- John M Mariadason
- Albert Einstein College of Medicine, Department of Oncology, Montefiore Medical Center, Bronx, NY 10467, USA.
| |
Collapse
|
288
|
Abstract
Histone deacetylase inhibitors represent a promising new class of compounds for the treatment of cancer. Inhibitors of this kind currently under clinical evaluation mainly target the classical (Rpd3/Hda1) family of histone deacetylases. Of particular note, the U.S. Food and Drug Administration recently approved the first histone deacetylase inhibitor (Zolinza: Merck and Co., Whitehouse Station, NJ, U.S.A.) for the treatment of cutaneous T-cell lymphoma. Dozens of such inhibitors are now in phase ii-iii clinical trials, sometimes in combination with other chemotherapy drugs, for diverse cancer types, including both hematologic and solid tumours. In this mini-review, we provide an overview of the histone deacetylase superfamily, highlight the positive results of deacetylase inhibitors in cancer clinical trials, and comment on the prospects for the next generation of such inhibitors.
Collapse
Affiliation(s)
- D.R. Walkinshaw
- Rosalind and Morris Goodman Cancer Centre (formerly the McGill Cancer Centre, renamed in September 2008), McGill University, and Department of Medicine, McGill University Health Centre, Montreal, QC
| | - X.J. Yang
- Rosalind and Morris Goodman Cancer Centre (formerly the McGill Cancer Centre, renamed in September 2008), McGill University, and Department of Medicine, McGill University Health Centre, Montreal, QC
| |
Collapse
|
289
|
Eot-Houllier G, Fulcrand G, Watanabe Y, Magnaghi-Jaulin L, Jaulin C. Histone deacetylase 3 is required for centromeric H3K4 deacetylation and sister chromatid cohesion. Genes Dev 2008; 22:2639-44. [PMID: 18832068 PMCID: PMC2559902 DOI: 10.1101/gad.484108] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Accepted: 08/11/2008] [Indexed: 11/24/2022]
Abstract
We describe here the role of histone deacetylase 3 (HDAC3) in sister chromatid cohesion and the deacetylation of histone H3 Lys 4 (H3K4) at the centromere. HDAC3 knockdown induced spindle assembly checkpoint activation and sister chromatid dissociation. The depletion of Polo-like kinase 1 (Plk1) or Aurora B restored cohesion in HDAC3-depleted cells. HDAC3 was also required for Shugoshin localization at centromeres. Finally, we show that HDAC3 depletion results in the acetylation of centromeric H3K4, correlated with a loss of dimethylation at the same position. These findings provide a functional link between sister chromatid cohesion and the mitotic "histone code".
Collapse
Affiliation(s)
- Grégory Eot-Houllier
- INSERM, U860, Montpellier, F-34298 France
- CRLC Val d’Aurelle-Paul Lamarque, Montpellier, F-34298 France
- Université Montpellier1, Montpellier, F-34298 France
| | - Géraldine Fulcrand
- INSERM, U860, Montpellier, F-34298 France
- CRLC Val d’Aurelle-Paul Lamarque, Montpellier, F-34298 France
- Université Montpellier1, Montpellier, F-34298 France
| | - Yoshinori Watanabe
- Laboratory of Chromosome Dynamics, Institute of Molecular and Cellular Biosciences, University of Tokyo, Yayoi, Tokyo 113-0032, Japan
| | - Laura Magnaghi-Jaulin
- INSERM, U860, Montpellier, F-34298 France
- CRLC Val d’Aurelle-Paul Lamarque, Montpellier, F-34298 France
- Université Montpellier1, Montpellier, F-34298 France
| | - Christian Jaulin
- INSERM, U860, Montpellier, F-34298 France
- CRLC Val d’Aurelle-Paul Lamarque, Montpellier, F-34298 France
- Université Montpellier1, Montpellier, F-34298 France
| |
Collapse
|
290
|
Witt O, Deubzer HE, Milde T, Oehme I. HDAC family: What are the cancer relevant targets? Cancer Lett 2008; 277:8-21. [PMID: 18824292 DOI: 10.1016/j.canlet.2008.08.016] [Citation(s) in RCA: 806] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2008] [Revised: 08/08/2008] [Accepted: 08/11/2008] [Indexed: 12/31/2022]
Abstract
Histone deacetylases comprise a family of 18 genes, which are grouped into classes I-IV based on their homology to their respective yeast orthologues. Classes I, II, and IV consist of 11 family members, which are referred to as "classical" HDACs, whereas the 7 class III members are called sirtuins. Classical HDACs are a promising novel class of anti-cancer drug targets. First HDAC inhibitors have been evaluated in clinical trials and show activity against several cancer diseases. However, these compounds act unselectively against several or all 11 HDAC family members. As a consequence, clinical phase I trials document a wide range of side effects. Therefore, the current challenge in the field is to define the cancer relevant HDAC family member(s) in a given tumor type and to design selective inhibitors, which target cancer cells but leave out normal cells. Knockout of single HDAC family members in mice produces a variety of phenotypes ranging from early embryonic death to viable animals with only discrete alterations, indicating that potential side effects of HDAC inhibitors depend on the selectivity of the compounds. Recently, several studies have shown that certain HDAC family members are aberrantly expressed in several tumors and have non-redundant function in controlling hallmarks of cancer cells. The aim of this review is to discuss individual HDAC family members as drug targets in cancer taking into consideration their function under physiological conditions and their oncogenic potential in malignant disease.
Collapse
Affiliation(s)
- Olaf Witt
- CCU Pediatric Oncology (G340), German Cancer Research Center, Im Neuenheimer Feld 280, Heidelberg, Germany.
| | | | | | | |
Collapse
|
291
|
Epping MT, Bernards R. Molecular basis of the anti-cancer effects of histone deacetylase inhibitors. Int J Biochem Cell Biol 2008; 41:16-20. [PMID: 18765293 DOI: 10.1016/j.biocel.2008.07.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2008] [Revised: 07/25/2008] [Accepted: 07/28/2008] [Indexed: 12/11/2022]
Abstract
Histone deacetylase inhibitors comprise a variety of natural and synthetic compounds, which have in common that they inhibit enzymes that mediate the removal of acetyl groups from a range of proteins, including nucleosomal histones. Histone deacetylase inhibitors have anti-cancer activities in vitro and in vivo and are used in the clinic for the treatment of advanced cutaneous T cell lymphoma. The molecular pathways targeted by these compounds are discussed with an emphasis on the effects of these compounds on retinoic acid signaling.
Collapse
Affiliation(s)
- Mirjam T Epping
- Center for Biomedical Genetics and Cancer Genomics Centre, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | |
Collapse
|
292
|
Eot-Houllier G, Fulcrand G, Magnaghi-Jaulin L, Jaulin C. Histone deacetylase inhibitors and genomic instability. Cancer Lett 2008; 274:169-76. [PMID: 18635312 DOI: 10.1016/j.canlet.2008.06.005] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2008] [Revised: 06/03/2008] [Accepted: 06/04/2008] [Indexed: 12/15/2022]
Abstract
Histone deacetylase inhibitors (HDACIs) are a promising new class of anticancer drugs. However, their mechanism of action has not been fully elucidated. Most studies have investigated the effect of HDACIs on the regulation of gene transcription. HDAC inhibition also leads to genomic instability by a variety of mechanisms. This phenomenon, which has been largely overlooked, may contribute to the cytotoxic effects of these drugs. Indeed, HDACIs sensitize DNA to exogenous genotoxic damage and induce the generation of reactive oxygen species. Moreover, HDACIs target mitosis resulting in chromosome segregation defects. Here, we review the effects of HDACI treatment on DNA damage and repair, and chromosome segregation control.
Collapse
Affiliation(s)
- Grégory Eot-Houllier
- Groupe Microtubules et Cycle Cellulaire, Institut de Génétique Humaine, CNRS UPR 1142, rue de la cardonille, 34396 Montpellier cedex 5, France
| | | | | | | |
Collapse
|
293
|
Trivedi CM, Lu MM, Wang Q, Epstein JA. Transgenic overexpression of Hdac3 in the heart produces increased postnatal cardiac myocyte proliferation but does not induce hypertrophy. J Biol Chem 2008; 283:26484-9. [PMID: 18625706 DOI: 10.1074/jbc.m803686200] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Class I and II histone deacetylases (HDACs) play vital roles in regulating cardiac development, morphogenesis, and hypertrophic responses. Although the roles of Hdac1 and Hdac2, class I HDACs, in cardiac hyperplasia, growth, and hypertrophic responsiveness have been reported, the role in the heart of Hdac3, another class I HDAC, has been less well explored. Here we report that myocyte-specific overexpression of Hdac3 in mice results in cardiac abnormalities at birth. Hdac3 overexpression produces thickening of ventricular myocardium, especially the interventricular septum, and reduction of both ventricular cavities in newborn hearts. Our data suggest that increased thickness of myocardium in Hdac3-transgenic (Hdac3-Tg) mice is due to increased cardiomyocyte hyperplasia without hypertrophy. Hdac3 overexpression inhibits several cyclin-dependent kinase inhibitors, including Cdkn1a, Cdkn1b, Cdkn1c, Cdkn2b, and Cdkn2c. Hdac3-Tg mice did not develop cardiac hypertrophy at 3 months of age, unlike previously reported Hdac2-Tg mice. Further, Hdac3 overexpression did not augment isoproterenol-induced cardiac hypertrophy when compared with wild-type littermates. These findings identify Hdac3 as a novel regulator of cardiac myocyte proliferation during cardiac development.
Collapse
Affiliation(s)
- Chinmay M Trivedi
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | |
Collapse
|
294
|
Research highlights. Nat Cell Biol 2008. [DOI: 10.1038/ncb0608-642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
295
|
Knutson SK, Chyla BJ, Amann JM, Bhaskara S, Huppert SS, Hiebert SW. Liver-specific deletion of histone deacetylase 3 disrupts metabolic transcriptional networks. EMBO J 2008; 27:1017-28. [PMID: 18354499 PMCID: PMC2323257 DOI: 10.1038/emboj.2008.51] [Citation(s) in RCA: 218] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2008] [Accepted: 02/25/2008] [Indexed: 01/04/2023] Open
Abstract
Histone deacetylase 3 (Hdac3) is an enzymatic component of transcriptional repression complexes recruited by the nuclear hormone receptors. Inactivation of Hdac3 in cancer cell lines triggered apoptosis, and removal of Hdac3 in the germ line of mice caused embryonic lethality. Therefore, we deleted Hdac3 in the postnatal mouse liver. These mice developed hepatomegaly, which was the result of hepatocyte hypertrophy, and these morphological changes coincided with significant imbalances between carbohydrate and lipid metabolism. Loss of Hdac3 triggered changes in gene expression consistent with inactivation of repression mediated by nuclear hormone receptors. Loss of Hdac3 also increased the levels of Ppar gamma2, and treatment of these mice with a Ppar gamma antagonist partially reversed the lipid accumulation in the liver. In addition, gene expression analysis identified mammalian target of rapamycin signalling as being activated after deletion of Hdac3, and inhibition by rapamycin affected the accumulation of neutral lipids in Hdac3-null livers. Thus, Hdac3 regulates metabolism through multiple signalling pathways in the liver, and deletion of Hdac3 disrupts normal metabolic homeostasis.
Collapse
Affiliation(s)
- Sarah K Knutson
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Brenda J Chyla
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Joseph M Amann
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Srividya Bhaskara
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Stacey S Huppert
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Scott W Hiebert
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Biochemistry, Vanderbilt University School of Medicine, 512 Preston Research Building, 23rd and Pierce Avenue, Nashville, TN 37232, USA. Tel.: +1 615 936 3582; Fax: +1 615 936 1790; E-mail:
| |
Collapse
|