101
|
New insights into transcriptional and leukemogenic mechanisms of AML1-ETO and E2A fusion proteins. ACTA ACUST UNITED AC 2016; 11:285-304. [PMID: 28261265 DOI: 10.1007/s11515-016-1415-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Nearly 15% of acute myeloid leukemia (AML) cases are caused by aberrant expression of AML1-ETO, a fusion protein generated by the t(8;21) chromosomal translocation. Since its discovery, AML1-ETO has served as a prototype to understand how leukemia fusion proteins deregulate transcription to promote leukemogenesis. Another leukemia fusion protein, E2A-Pbx1, generated by the t(1;19) translocation, is involved in acute lymphoblastic leukemias (ALLs). While AML1-ETO and E2A-Pbx1 are structurally unrelated fusion proteins, we have recently shown that a common axis, the ETO/E-protein interaction, is involved in the regulation of both fusion proteins, underscoring the importance of studying protein-protein interactions in elucidating the mechanisms of leukemia fusion proteins. OBJECTIVE In this review, we aim to summarize these new developments while also providing a historic overview of the related early studies. METHODS A total of 218 publications were reviewed in this article, a majority of which were published after 2004.We also downloaded 3D structures of AML1-ETO domains from Protein Data Bank and provided a systematic summary of their structures. RESULTS By reviewing the literature, we summarized early and recent findings on AML1-ETO, including its protein-protein interactions, transcriptional and leukemogenic mechanisms, as well as the recently reported involvement of ETO family corepressors in regulating the function of E2A-Pbx1. CONCLUSION While the recent development in genomic and structural studies has clearly demonstrated that the fusion proteins function by directly regulating transcription, a further understanding of the underlying mechanisms, including crosstalk with other transcription factors and cofactors, and the protein-protein interactions in the context of native proteins, may be necessary for the development of highly targeted drugs for leukemia therapy.
Collapse
|
102
|
Stacey W, Bhave S, Uht RM. Mechanisms by Which 17β-Estradiol (E2) Suppress Neuronal cox-2 Gene Expression. PLoS One 2016; 11:e0161430. [PMID: 27588681 PMCID: PMC5010190 DOI: 10.1371/journal.pone.0161430] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 08/05/2016] [Indexed: 11/19/2022] Open
Abstract
E2 attenuates inflammatory responses by suppressing expression of pro-inflammatory genes. Given that inflammation is increasingly being associated with neurodegenerative and psychiatric processes, we sought to elucidate mechanisms by which E2 down-regulates a component of an inflammatory response, cyclooxygenase- 2 (COX-2) expression. Although inflammatory processes in the brain are usually associated with microglia and astrocytes, we found that the COX-2 gene (cox-2) was expressed in a neuronal context, specifically in an amygdalar cell line (AR-5). Given that COX-2 has been reported to be in neurons in the brain, and that the amygdala is a site involved in neurodegenerative and neuropsychiatric processes, we investigated mechanisms by which E2 could down-regulate cox-2 expression in the AR-5 line. These cells express estrogen receptors alpha (ERα) and beta (ERβ), and as shown here cox-2. At the level of RNA, E2 and the ERβ selective ligand diarylpropionitrile (DPN) both attenuated gene expression, whereas the ERα selective ligand propyl pyrazole triol (PPT) had no effect. Neither ligand increased ERβ at the cox-2 promoter. Rather, DPN decreased promoter occupancy of NF-κB p65 and histone 4 (H4) acetylation. Treatment with the non-specific HDAC inhibitor Trichostatin A (TSA) counteracted DPN's repressive effects on cox-2 expression. In keeping with the TSA effect, E2 and DPN increased histone deacetylase one (HDAC1) and switch-independent 3A (Sin3A) promoter occupancy. Lastly, even though E2 increased CpG methylation, DPN did not. Taken together, the pharmacological data indicate that ERβ contributes to neuronal cox-2 expression, as measured by RNA levels. Furthermore, ER ligands lead to increased recruitment of HDAC1, Sin3A and a concomitant reduction of p65 occupancy and Ac-H4 levels. None of the events, however, are associated with a significant recruitment of ERβ at the promoter. Thus, ERβ directs recruitment to the cox-2 promoter, but does so in the absence of being recruited itself.
Collapse
Affiliation(s)
- Winfred Stacey
- Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Institute for Healthy Aging, Center for Alzheimer’s and Neurodegenerative Disease Research, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Shreyas Bhave
- Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Institute for Healthy Aging, Center for Alzheimer’s and Neurodegenerative Disease Research, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Rosalie M. Uht
- Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Institute for Healthy Aging, Center for Alzheimer’s and Neurodegenerative Disease Research, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- * E-mail:
| |
Collapse
|
103
|
Histone Deacetylases with Antagonistic Roles in Saccharomyces cerevisiae Heterochromatin Formation. Genetics 2016; 204:177-90. [PMID: 27489001 DOI: 10.1534/genetics.116.190835] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/14/2016] [Indexed: 12/18/2022] Open
Abstract
As the only catalytic member of the Sir-protein gene-silencing complex, Sir2's catalytic activity is necessary for silencing. The only known role for Sir2's catalytic activity in Saccharomyces cerevisiae silencing is to deacetylate N-terminal tails of histones H3 and H4, creating high-affinity binding sites for the Sir-protein complex, resulting in association of Sir proteins across the silenced domain. This histone deacetylation model makes the simple prediction that preemptively removing Sir2's H3 and H4 acetyl substrates, by mutating these lysines to unacetylatable arginines, or removing the acetyl transferase responsible for their acetylation, should restore silencing in the Sir2 catalytic mutant. However, this was not the case. We conducted a genetic screen to explore what aspect of Sir2's catalytic activity has not been accounted for in silencing. Mutation of a nonsirtuin histone deacetylase, Rpd3, restored Sir-protein-based silencing in the absence of Sir2's catalytic activity. Moreover, this antagonism could be mediated by either the large or the small Rpd3-containing complex. Interestingly, this restoration of silencing appeared independent of any known histone H3 or H4 substrates of Rpd3 Investigation of Sir-protein association in the Rpd3 mutant revealed that the restoration of silencing was correlated with an increased association of Sir proteins at the silencers, suggesting that Rpd3 was an antagonist of Sir2's function in nucleation of Sir proteins to the silencer. Additionally, restoration of silencing by Rpd3 was dependent on another sirtuin family member, Hst3, indicating multiple antagonistic roles for deacetylases in S. cerevisiae silencing.
Collapse
|
104
|
Priyanka A, Solanki V, Parkesh R, Thakur KG. Crystal structure of the N-terminal domain of human SIRT7 reveals a three-helical domain architecture. Proteins 2016; 84:1558-63. [DOI: 10.1002/prot.25085] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 06/02/2016] [Accepted: 06/02/2016] [Indexed: 11/10/2022]
Affiliation(s)
- Anu Priyanka
- Structural Biology Laboratory; G. N. Ramachandran Protein Centre; CSIR-Institute of Microbial Technology; Chandigarh 160036 India
| | - Vipul Solanki
- Structural Biology Laboratory; G. N. Ramachandran Protein Centre; CSIR-Institute of Microbial Technology; Chandigarh 160036 India
| | - Raman Parkesh
- Computational Medicinal Chemistry and Fluorescent Chemosensors Laboratory; G. N. Ramachandran Protein Centre; CSIR-Institute of Microbial Technology; Chandigarh 160036 India
| | - Krishan Gopal Thakur
- Structural Biology Laboratory; G. N. Ramachandran Protein Centre; CSIR-Institute of Microbial Technology; Chandigarh 160036 India
| |
Collapse
|
105
|
Lewis KA, Tollefsbol TO. Regulation of the Telomerase Reverse Transcriptase Subunit through Epigenetic Mechanisms. Front Genet 2016; 7:83. [PMID: 27242892 PMCID: PMC4860561 DOI: 10.3389/fgene.2016.00083] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 04/22/2016] [Indexed: 12/21/2022] Open
Abstract
Chromosome-shortening is characteristic of normal cells, and is known as the end replication problem. Telomerase is the enzyme responsible for extending the ends of the chromosomes in de novo synthesis, and occurs in germ cells as well as most malignant cancers. There are three subunits of telomerase: human telomerase RNA (hTERC), human telomerase associated protein (hTEP1), or dyskerin, and human telomerase reverse transcriptase (hTERT). hTERC and hTEP1 are constitutively expressed, so the enzymatic activity of telomerase is dependent on the transcription of hTERT. DNA methylation, histone methylation, and histone acetylation are basic epigenetic regulations involved in the expression of hTERT. Non-coding RNA can also serve as a form of epigenetic control of hTERT. This epigenetic-based regulation of hTERT is important in providing a mechanism for reversibility of hTERT control in various biological states. These include embryonic down-regulation of hTERT contributing to aging and the upregulation of hTERT playing a critical role in over 90% of cancers. Normal human somatic cells have a non-methylated/hypomethylated CpG island within the hTERT promoter region, while telomerase-positive cells paradoxically have at least a partially methylated promoter region that is opposite to the normal roles of DNA methylation. Histone acetylation of H3K9 within the promoter region is associated with an open chromatin state such that transcription machinery has the space to form. Histone methylation of hTERT has varied control of the gene, however. Mono- and dimethylation of H3K9 within the promoter region indicate silent euchromatin, while a trimethylated H3K9 enhances gene transcription. Non-coding RNAs can target epigenetic-modifying enzymes, as well as transcription factors involved in the control of hTERT. An epigenetics diet that can affect the epigenome of cancer cells is a recent fascination that has received much attention. By combining portions of this diet with epigenome-altering treatments, it is possible to selectively regulate the epigenetic control of hTERT and its expression.
Collapse
Affiliation(s)
- Kayla A Lewis
- Department of Biology, University of Alabama at Birmingham, Birmingham AL, USA
| | - Trygve O Tollefsbol
- Department of Biology, University of Alabama at Birmingham, BirminghamAL, USA; Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, BirminghamAL, USA; Comprehensive Cancer Center, University of Alabama at Birmingham, BirminghamAL, USA; Nutrition Obesity Research Center, University of Alabama at Birmingham, BirminghamAL, USA; Comprehensive Diabetes Center, University of Alabama at Birmingham, BirminghamAL, USA
| |
Collapse
|
106
|
Millard CJ, Varma N, Saleh A, Morris K, Watson PJ, Bottrill AR, Fairall L, Smith CJ, Schwabe JWR. The structure of the core NuRD repression complex provides insights into its interaction with chromatin. eLife 2016; 5:e13941. [PMID: 27098840 PMCID: PMC4841774 DOI: 10.7554/elife.13941] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 03/24/2016] [Indexed: 12/14/2022] Open
Abstract
The NuRD complex is a multi-protein transcriptional corepressor that couples histone deacetylase and ATP-dependent chromatin remodelling activities. The complex regulates the higher-order structure of chromatin, and has important roles in the regulation of gene expression, DNA damage repair and cell differentiation. HDACs 1 and 2 are recruited by the MTA1 corepressor to form the catalytic core of the complex. The histone chaperone protein RBBP4, has previously been shown to bind to the carboxy-terminal tail of MTA1. We show that MTA1 recruits a second copy of RBBP4. The crystal structure reveals an extensive interface between MTA1 and RBBP4. An EM structure, supported by SAXS and crosslinking, reveals the architecture of the dimeric HDAC1:MTA1:RBBP4 assembly which forms the core of the NuRD complex. We find evidence that in this complex RBBP4 mediates interaction with histone H3 tails, but not histone H4, suggesting a mechanism for recruitment of the NuRD complex to chromatin.
Collapse
Affiliation(s)
- Christopher J Millard
- Henry Wellcome Laboratories of Structural Biology, Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - Niranjan Varma
- Henry Wellcome Laboratories of Structural Biology, Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - Almutasem Saleh
- Henry Wellcome Laboratories of Structural Biology, Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - Kyle Morris
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Peter J Watson
- Henry Wellcome Laboratories of Structural Biology, Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - Andrew R Bottrill
- Protein and Nucleic Acid Chemistry Laboratory, Core Biotechnology Services, University of Leicester, Leicester, United Kingdom
| | - Louise Fairall
- Henry Wellcome Laboratories of Structural Biology, Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - Corinne J Smith
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - John WR Schwabe
- Henry Wellcome Laboratories of Structural Biology, Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
107
|
Bansal N, David G, Farias E, Waxman S. Emerging Roles of Epigenetic Regulator Sin3 in Cancer. Adv Cancer Res 2016; 130:113-35. [PMID: 27037752 DOI: 10.1016/bs.acr.2016.01.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Revolutionizing treatment strategies is an urgent clinical need in the fight against cancer. Recently the scientific community has recognized chromatin-associated proteins as promising therapeutic candidates. However, there is a need to develop more targeted epigenetic inhibitors with less toxicity. Sin3 family is one such target which consists of evolutionary conserved proteins with two paralogues Sin3A and Sin3B. Sin3A/B are global transcription regulators that provide a versatile platform for diverse chromatin-modifying activities. Sin3 proteins regulate key cellular functions that include cell cycle, proliferation, and differentiation, and have recently been implicated in cancer pathogenesis. In this chapter, we summarize the key concepts of Sin3 biology and elaborate the recent advancements in the role of Sin3 proteins in cancer with specific examples in multiple endocrine neoplasia type 2, pancreatic ductal adenocarcinoma, and triple negative breast cancer. Finally, a program to create an integrative approach for screening antitumor agents that target chromatin-associated factors like Sin3 is presented.
Collapse
Affiliation(s)
- N Bansal
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - G David
- New York University School of Medicine, New York, NY, United States
| | - E Farias
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - S Waxman
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
108
|
Lakisic G, Lebreton A, Pourpre R, Wendling O, Libertini E, Radford EJ, Le Guillou M, Champy MF, Wattenhofer-Donzé M, Soubigou G, Ait-Si-Ali S, Feunteun J, Sorg T, Coppée JY, Ferguson-Smith AC, Cossart P, Bierne H. Role of the BAHD1 Chromatin-Repressive Complex in Placental Development and Regulation of Steroid Metabolism. PLoS Genet 2016; 12:e1005898. [PMID: 26938916 PMCID: PMC4777444 DOI: 10.1371/journal.pgen.1005898] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 02/04/2016] [Indexed: 11/18/2022] Open
Abstract
BAHD1 is a vertebrate protein that promotes heterochromatin formation and gene repression in association with several epigenetic regulators. However, its physiological roles remain unknown. Here, we demonstrate that ablation of the Bahd1 gene results in hypocholesterolemia, hypoglycemia and decreased body fat in mice. It also causes placental growth restriction with a drop of trophoblast glycogen cells, a reduction of fetal weight and a high neonatal mortality rate. By intersecting transcriptome data from murine Bahd1 knockout (KO) placentas at stages E16.5 and E18.5 of gestation, Bahd1-KO embryonic fibroblasts, and human cells stably expressing BAHD1, we also show that changes in BAHD1 levels alter expression of steroid/lipid metabolism genes. Biochemical analysis of the BAHD1-associated multiprotein complex identifies MIER proteins as novel partners of BAHD1 and suggests that BAHD1-MIER interaction forms a hub for histone deacetylases and methyltransferases, chromatin readers and transcription factors. We further show that overexpression of BAHD1 leads to an increase of MIER1 enrichment on the inactive X chromosome (Xi). In addition, BAHD1 and MIER1/3 repress expression of the steroid hormone receptor genes ESR1 and PGR, both playing important roles in placental development and energy metabolism. Moreover, modulation of BAHD1 expression in HEK293 cells triggers epigenetic changes at the ESR1 locus. Together, these results identify BAHD1 as a core component of a chromatin-repressive complex regulating placental morphogenesis and body fat storage and suggest that its dysfunction may contribute to several human diseases.
Collapse
Affiliation(s)
- Goran Lakisic
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Équipe Microbiologie Cellulaire et Epigénétique, Jouy-en-Josas, France
| | - Alice Lebreton
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, Paris, France
- INSERM U604, Paris, France
- INRA USC2020, Paris, France
| | - Renaud Pourpre
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Équipe Microbiologie Cellulaire et Epigénétique, Jouy-en-Josas, France
| | - Olivia Wendling
- Institut Clinique de la Souris-ICS-MCI, PHENOMIN, CNRS UMR7104, INSERM U964, Université de Strasbourg, Illkirch, France
| | - Emanuele Libertini
- Plateforme Transcriptome et Epigénome, Département Génomes et Génétique, Institut Pasteur, Paris, France
| | - Elizabeth J. Radford
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
- Cambridge University Hospitals, NHS Foundation Trust, Cambridge, United Kingdom
| | - Morwenna Le Guillou
- CNRS UMR8200 Stabilité génétique et oncogenèse, Université Paris-Saclay, Villejuif, France
| | - Marie-France Champy
- Institut Clinique de la Souris-ICS-MCI, PHENOMIN, CNRS UMR7104, INSERM U964, Université de Strasbourg, Illkirch, France
| | - Marie Wattenhofer-Donzé
- Institut Clinique de la Souris-ICS-MCI, PHENOMIN, CNRS UMR7104, INSERM U964, Université de Strasbourg, Illkirch, France
| | - Guillaume Soubigou
- Plateforme Transcriptome et Epigénome, Département Génomes et Génétique, Institut Pasteur, Paris, France
| | | | - Jean Feunteun
- CNRS UMR8200 Stabilité génétique et oncogenèse, Université Paris-Saclay, Villejuif, France
| | - Tania Sorg
- Institut Clinique de la Souris-ICS-MCI, PHENOMIN, CNRS UMR7104, INSERM U964, Université de Strasbourg, Illkirch, France
| | - Jean-Yves Coppée
- Plateforme Transcriptome et Epigénome, Département Génomes et Génétique, Institut Pasteur, Paris, France
| | | | - Pascale Cossart
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, Paris, France
- INSERM U604, Paris, France
- INRA USC2020, Paris, France
| | - Hélène Bierne
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Équipe Microbiologie Cellulaire et Epigénétique, Jouy-en-Josas, France
| |
Collapse
|
109
|
Wen X, Klionsky DJ. An overview of macroautophagy in yeast. J Mol Biol 2016; 428:1681-99. [PMID: 26908221 DOI: 10.1016/j.jmb.2016.02.021] [Citation(s) in RCA: 185] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 02/15/2016] [Accepted: 02/16/2016] [Indexed: 12/19/2022]
Abstract
Macroautophagy is an evolutionarily conserved dynamic pathway that functions primarily in a degradative manner. A basal level of macroautophagy occurs constitutively, but this process can be further induced in response to various types of stress including starvation, hypoxia and hormonal stimuli. The general principle behind macroautophagy is that cytoplasmic contents can be sequestered within a transient double-membrane organelle, an autophagosome, which subsequently fuses with a lysosome or vacuole (in mammals, or yeast and plants, respectively), allowing for degradation of the cargo followed by recycling of the resulting macromolecules. Through this basic mechanism, macroautophagy has a critical role in cellular homeostasis; however, either insufficient or excessive macroautophagy can seriously compromise cell physiology, and thus, it needs to be properly regulated. In fact, a wide range of diseases are associated with dysregulation of macroautophagy. There has been substantial progress in understanding the regulation and molecular mechanisms of macroautophagy in different organisms; however, many questions concerning some of the most fundamental aspects of macroautophagy remain unresolved. In this review, we summarize current knowledge about macroautophagy mainly in yeast, including the mechanism of autophagosome biogenesis, the function of the core macroautophagic machinery, the regulation of macroautophagy and the process of cargo recognition in selective macroautophagy, with the goal of providing insights into some of the key unanswered questions in this field.
Collapse
Affiliation(s)
- Xin Wen
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
110
|
Eom GH, Kook H. Role of histone deacetylase 2 and its posttranslational modifications in cardiac hypertrophy. BMB Rep 2015; 48:131-8. [PMID: 25388210 PMCID: PMC4453031 DOI: 10.5483/bmbrep.2015.48.3.242] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Indexed: 11/20/2022] Open
Abstract
Cardiac hypertrophy is a form of global remodeling, although the initial step seems to be an adaptation to increased hemodynamic demands. The characteristics of cardiac hypertrophy include the functional reactivation of the arrested fetal gene program, where histone deacetylases (HDACs) are closely linked in the development of the process. To date, mammalian HDACs are divided into four classes: I, II, III, and IV. By structural similarities, class II HDACs are then subdivided into IIa and IIb. Among class I and II HDACs, HDAC2, 4, 5, and 9 have been reported to be involved in hypertrophic responses; HDAC4, 5, and 9 are negative regulators, whereas HDAC2 is a pro-hypertrophic mediator. The molecular function and regulation of class IIa HDACs depend largely on the phosphorylation-mediated cytosolic redistribution, whereas those of HDAC2 take place primarily in the nucleus. In response to stresses, posttranslational modification (PTM) processes, dynamic modifications after the translation of proteins, are involved in the regulation of the activities of those hypertrophy-related HDACs. In this article, we briefly review 1) the activation of HDAC2 in the development of cardiac hypertrophy and 2) the PTM of HDAC2 and its implications in the regulation of HDAC2 activity.
Collapse
Affiliation(s)
- Gwang Hyeon Eom
- Department of Pharmacology and Medical Research Center for Gene Regulation, Chonnam National University Medical School, Gwangju 501-746, Korea
| | - Hyun Kook
- Department of Pharmacology and Medical Research Center for Gene Regulation, Chonnam National University Medical School, Gwangju 501-746, Korea
| |
Collapse
|
111
|
Kim E, Beon J, Lee S, Park J, Kim S. IPMK: A versatile regulator of nuclear signaling events. Adv Biol Regul 2015; 61:25-32. [PMID: 26682649 DOI: 10.1016/j.jbior.2015.11.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/17/2015] [Accepted: 11/17/2015] [Indexed: 12/11/2022]
Abstract
Inositol-derived metabolites (e.g., phosphoinositides and inositol polyphosphates) are key second messengers that are essential for controlling a wide range of cellular events. Inositol polyphosphate multikinase (IPMK) exhibits complex catalytic activities that eventually yield water-soluble inositol polyphosphates (e.g., IP4 and IP5) and lipid-bound phosphatidylinositol 3,4,5-trisphosphate. A series of recent studies have suggested that IPMK may be a multifunctional regulator in the nucleus of mammalian cells. In this review, we highlight the novel modes of action of IPMK in transcriptional and epigenetic regulation, and discuss its roles in physiology and disease.
Collapse
Affiliation(s)
- Eunha Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Jiyoon Beon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Seulgi Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Jina Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Seyun Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
112
|
Essential Nonredundant Function of the Catalytic Activity of Histone Deacetylase 2 in Mouse Development. Mol Cell Biol 2015; 36:462-74. [PMID: 26598605 PMCID: PMC4719423 DOI: 10.1128/mcb.00639-15] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 11/16/2015] [Indexed: 12/16/2022] Open
Abstract
The class I histone deacetylases (HDACs) HDAC1 and HDAC2 play partially redundant roles in the regulation of gene expression and mouse development. As part of multisubunit corepressor complexes, these two deacetylases exhibit both enzymatic and nonenzymatic functions. To examine the impact of the catalytic activities of HDAC1 and HDAC2, we generated knock-in mice expressing catalytically inactive isoforms, which are still incorporated into the HDAC1/HDAC2 corepressor complexes. Surprisingly, heterozygous mice expressing catalytically inactive HDAC2 die within a few hours after birth, while heterozygous HDAC1 mutant mice are indistinguishable from wild-type littermates. Heterozygous HDAC2 mutant mice show an unaltered composition but reduced associated deacetylase activity of corepressor complexes and exhibit a more severe phenotype than HDAC2-null mice. They display changes in brain architecture accompanied by premature expression of the key regulator protein kinase C delta. Our study reveals a dominant negative effect of catalytically inactive HDAC2 on specific corepressor complexes resulting in histone hyperacetylation, transcriptional derepression, and, ultimately, perinatal lethality.
Collapse
|
113
|
Clark MD, Zhang Y, Radhakrishnan I. Solution NMR Studies of an Alternative Mode of Sin3 Engagement by the Sds3 Subunit in the Histone Deacetylase-Associated Sin3L/Rpd3L Corepressor Complex. J Mol Biol 2015; 427:3817-23. [PMID: 26522936 DOI: 10.1016/j.jmb.2015.10.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 10/05/2015] [Accepted: 10/22/2015] [Indexed: 11/19/2022]
Abstract
The Sds3 transcriptional corepressor facilitates the assembly of the 1- to 2-MDa histone deacetylase-associated Sin3L/Rpd3L complex by providing a crucial homodimerization activity. Sds3 engages the scaffolding protein Sin3A, via a bipartite motif within the Sin3 interaction domain (SID) comprising a helix and an extended segment. Here, we show that the SID samples two discrete, substantially populated conformations with lifetimes in the tens of milliseconds range. The two conformations differ via a translation of the main chain and the corresponding side chains in the 5- to 7-Å range. Given the close proximity of the SID to other functional motifs in Sds3 at the sequence level, the conformational exchange has the potential to regulate these activities.
Collapse
Affiliation(s)
- Michael David Clark
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Yongbo Zhang
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
| | - Ishwar Radhakrishnan
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
114
|
Histone Deacetylase 3 Is Required for Efficient T Cell Development. Mol Cell Biol 2015; 35:3854-65. [PMID: 26324326 DOI: 10.1128/mcb.00706-15] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 08/19/2015] [Indexed: 12/12/2022] Open
Abstract
Hdac3 is a key target for Hdac inhibitors that are efficacious in cutaneous T cell lymphoma. Moreover, the regulation of chromatin structure is critical as thymocytes transition from an immature cell with open chromatin to a mature T cell with tightly condensed chromatin. To define the phenotypes controlled by Hdac3 during T cell development, we conditionally deleted Hdac3 using the Lck-Cre transgene. This strategy inactivated Hdac3 in the double-negative stages of thymocyte development and caused a significant impairment at the CD8 immature single-positive (ISP) stage and the CD4/CD8 double-positive stage, with few mature CD4(+) or CD8(+) single-positive cells being produced. When Hdac3(-/-) mice were crossed with Bcl-xL-, Bcl2-, or TCRβ-expressing transgenic mice, a modest level of complementation was found. However, when the null mice were crossed with mice expressing a fully rearranged T cell receptor αβ transgene, normal levels of CD4 single-positive cells were produced. Thus, Hdac3 is required for the efficient transit from double-negative stage 4 through positive selection.
Collapse
|
115
|
Perspective on unraveling the versatility of ‘co-repressor’ complexes. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2015; 1849:1051-6. [DOI: 10.1016/j.bbagrm.2015.06.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/23/2015] [Accepted: 06/26/2015] [Indexed: 01/01/2023]
|
116
|
Presnell JS, Schnitzler CE, Browne WE. KLF/SP Transcription Factor Family Evolution: Expansion, Diversification, and Innovation in Eukaryotes. Genome Biol Evol 2015; 7:2289-309. [PMID: 26232396 PMCID: PMC4558859 DOI: 10.1093/gbe/evv141] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2015] [Indexed: 11/13/2022] Open
Abstract
The Krüppel-like factor and specificity protein (KLF/SP) genes play key roles in critical biological processes including stem cell maintenance, cell proliferation, embryonic development, tissue differentiation, and metabolism and their dysregulation has been implicated in a number of human diseases and cancers. Although many KLF/SP genes have been characterized in a handful of bilaterian lineages, little is known about the KLF/SP gene family in nonbilaterians and virtually nothing is known outside the metazoans. Here, we analyze and discuss the origins and evolutionary history of the KLF/SP transcription factor family and associated transactivation/repression domains. We have identified and characterized the complete KLF/SP gene complement from the genomes of 48 species spanning the Eukarya. We have also examined the phylogenetic distribution of transactivation/repression domains associated with this gene family. We report that the origin of the KLF/SP gene family predates the divergence of the Metazoa. Furthermore, the expansion of the KLF/SP gene family is paralleled by diversification of transactivation domains via both acquisitions of pre-existing ancient domains as well as by the appearance of novel domains exclusive to this gene family and is strongly associated with the expansion of cell type complexity.
Collapse
Affiliation(s)
| | - Christine E Schnitzler
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health
| | | |
Collapse
|
117
|
Structural insights into the assembly of the histone deacetylase-associated Sin3L/Rpd3L corepressor complex. Proc Natl Acad Sci U S A 2015; 112:E3669-78. [PMID: 26124119 DOI: 10.1073/pnas.1504021112] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Acetylation is correlated with chromatin decondensation and transcriptional activation, but its regulation by histone deacetylase (HDAC)-bearing corepressor complexes is poorly understood. Here, we describe the mechanism of assembly of the mammalian Sin3L/Rpd3L complex facilitated by Sds3, a conserved subunit deemed critical for proper assembly. Sds3 engages a globular, helical region of the HDAC interaction domain (HID) of the scaffolding protein Sin3A through a bipartite motif comprising a helix and an adjacent extended segment. Sds3 dimerizes through not only one of the predicted coiled-coil motifs but also, the segment preceding it, forming an ∼ 150-Å-long antiparallel dimer. Contrary to previous findings in yeast, Sin3A rather than Sds3 functions in recruiting HDAC1 into the complex by engaging the latter through a highly conserved segment adjacent to the helical HID subdomain. In the resulting model for the ternary complex, the two copies of the HDACs are situated distally and dynamically because of a natively unstructured linker connecting the dimerization domain and the Sin3A interaction domain of Sds3; these features contrast with the static organization described previously for the NuRD (nucleosome remodeling and deacetylase) complex. The Sds3 linker features several conserved basic residues that could potentially maintain the complex on chromatin by nonspecific interactions with DNA after initial recruitment by sequence-specific DNA-binding repressors.
Collapse
|
118
|
Kadamb R, Mittal S, Bansal N, Saluja D. Stress-mediated Sin3B activation leads to negative regulation of subset of p53 target genes. Biosci Rep 2015; 35:e00234. [PMID: 26181367 PMCID: PMC4613689 DOI: 10.1042/bsr20150122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 06/03/2015] [Accepted: 06/19/2015] [Indexed: 11/28/2022] Open
Abstract
The multiprotein SWI-independent 3 (Sin3)-HDAC (histone deacetylase) corepressor complex mediates gene repression through its interaction with DNA-binding factors and recruitment of chromatin-modifying proteins on to the promoters of target gene. Previously, an increased expression of Sin3B and tumour suppressor protein, p53 has been established upon adriamycin treatment. We, now provide evidence that Sin3B expression is significantly up-regulated under variety of stress conditions and this response is not stress-type specific. We observed that Sin3B expression is significantly up-regulated both at transcript and at protein level upon DNA damage induced by bleomycin drug, a radiomimetic agent. This increase in Sin3B expression upon stress is found to be p53-dependent and is associated with enhanced interaction of Sin3B with Ser(15) phosphorylated p53. Binding of Sin3-HDAC repressor complex on to the promoters of p53 target genes influences gene regulation by altering histone modifications (H3K9me3 and H3K27me3) at target genes. Furthermore, knockdown of Sin3B by shRNA severely compromises p53-mediated gene repression under stress conditions. Taken together, these results suggest that stress-induced Sin3B activation is p53-dependent and is essential for p53-mediated repression of its selective target genes. The present study has an implication in understanding the transrepression mechanism of p53 under DNA damaging conditions.
Collapse
Affiliation(s)
- Rama Kadamb
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi-110007, India
| | - Shilpi Mittal
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi-110007, India
| | - Nidhi Bansal
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi-110007, India
| | - Daman Saluja
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi-110007, India
| |
Collapse
|
119
|
Diolaiti D, McFerrin L, Carroll PA, Eisenman RN. Functional interactions among members of the MAX and MLX transcriptional network during oncogenesis. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1849:484-500. [PMID: 24857747 PMCID: PMC4241192 DOI: 10.1016/j.bbagrm.2014.05.016] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 04/23/2014] [Accepted: 05/14/2014] [Indexed: 01/27/2023]
Abstract
The transcription factor MYC and its related family members MYCN and MYCL have been implicated in the etiology of a wide spectrum of human cancers. Compared to other oncoproteins, such as RAS or SRC, MYC is unique because its protein coding region is rarely mutated. Instead, MYC's oncogenic properties are unleashed by regulatory mutations leading to unconstrained high levels of expression. Under both normal and pathological conditions MYC regulates multiple aspects of cellular physiology including proliferation, differentiation, apoptosis, growth and metabolism by controlling the expression of thousands of genes. How a single transcription factor exerts such broad effects remains a fascinating puzzle. Notably, MYC is part of a network of bHLHLZ proteins centered on the MYC heterodimeric partner MAX and its counterpart, the MAX-like protein MLX. This network includes MXD1-4, MNT, MGA, MONDOA and MONDOB proteins. With some exceptions, MXD proteins have been functionally linked to cell cycle arrest and differentiation, while MONDO proteins control cellular metabolism. Although the temporal expression patterns of many of these proteins can differ markedly they are frequently expressed simultaneously in the same cellular context, and potentially bind to the same, or similar DNA consensus sequence. Here we review the activities and interactions among these proteins and propose that the broad spectrum of phenotypes elicited by MYC deregulation is intimately connected to the functions and regulation of the other network members. Furthermore, we provide a meta-analysis of TCGA data suggesting that the coordinate regulation of the network is important in MYC driven tumorigenesis. This article is part of a Special Issue entitled: Myc proteins in cell biology and pathology.
Collapse
Affiliation(s)
- Daniel Diolaiti
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, USA
| | - Lisa McFerrin
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, USA
| | - Patrick A Carroll
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, USA
| | - Robert N Eisenman
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, USA.
| |
Collapse
|
120
|
Itoh T, Fairall L, Muskett FW, Milano CP, Watson PJ, Arnaudo N, Saleh A, Millard CJ, El-Mezgueldi M, Martino F, Schwabe JWR. Structural and functional characterization of a cell cycle associated HDAC1/2 complex reveals the structural basis for complex assembly and nucleosome targeting. Nucleic Acids Res 2015; 43:2033-44. [PMID: 25653165 PMCID: PMC4344507 DOI: 10.1093/nar/gkv068] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Recent proteomic studies have identified a novel histone deacetylase complex that is upregulated during mitosis and is associated with cyclin A. This complex is conserved from nematodes to man and contains histone deacetylases 1 and 2, the MIDEAS corepressor protein and a protein called DNTTIP1 whose function was hitherto poorly understood. Here, we report the structures of two domains from DNTTIP1. The amino-terminal region forms a tight dimerization domain with a novel structural fold that interacts with and mediates assembly of the HDAC1:MIDEAS complex. The carboxy-terminal domain of DNTTIP1 has a structure related to the SKI/SNO/DAC domain, despite lacking obvious sequence homology. We show that this domain in DNTTIP1 mediates interaction with both DNA and nucleosomes. Thus, DNTTIP1 acts as a dimeric chromatin binding module in the HDAC1:MIDEAS corepressor complex.
Collapse
Affiliation(s)
- Toshimasa Itoh
- Henry Wellcome Laboratories of Structural Biology, Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Louise Fairall
- Henry Wellcome Laboratories of Structural Biology, Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Frederick W Muskett
- Henry Wellcome Laboratories of Structural Biology, Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Charles P Milano
- Henry Wellcome Laboratories of Structural Biology, Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Peter J Watson
- Henry Wellcome Laboratories of Structural Biology, Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Nadia Arnaudo
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Almutasem Saleh
- Henry Wellcome Laboratories of Structural Biology, Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Christopher J Millard
- Henry Wellcome Laboratories of Structural Biology, Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Mohammed El-Mezgueldi
- Henry Wellcome Laboratories of Structural Biology, Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Fabrizio Martino
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - John W R Schwabe
- Henry Wellcome Laboratories of Structural Biology, Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| |
Collapse
|
121
|
Epigenetic basis of opiate suppression of Bdnf gene expression in the ventral tegmental area. Nat Neurosci 2015; 18:415-22. [PMID: 25643298 PMCID: PMC4340719 DOI: 10.1038/nn.3932] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 12/22/2014] [Indexed: 12/15/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) plays a crucial role in modulating neural and behavioral plasticity to drugs of abuse. Here, we demonstrate a persistent down-regulation of exon-specific Bdnf expression in the ventral tegmental area (VTA) in response to chronic opiate exposure, which is mediated by specific epigenetic modifications at the corresponding Bdnf gene promoters. Exposure to chronic morphine increases stalling of RNA polymerase II at these Bdnf promoters in VTA and alters permissive and repressive histone modifications and occupancy of their regulatory proteins at the specific promoters. Furthermore, we show that morphine suppresses binding of phospho-CREB (cAMP response element binding protein) to Bdnf promoters in VTA, which results from enrichment of trimethylated H3K27 at the promoters, and that decreased NURR1 (nuclear receptor related-1) expression also contributes to Bdnf repression and associated behavioral plasticity to morphine. These studies reveal novel epigenetic mechanisms of morphine-induced molecular and behavioral neuroadaptations.
Collapse
|
122
|
Spruijt CG, Vermeulen M. DNA methylation: old dog, new tricks? Nat Struct Mol Biol 2015; 21:949-54. [PMID: 25372310 DOI: 10.1038/nsmb.2910] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 09/30/2014] [Indexed: 12/17/2022]
Abstract
DNA methylation is an epigenetic modification that is generally associated with repression of transcription initiation at CpG-island promoters. Here we argue that, on the basis of recent high-throughput genomic and proteomic screenings, DNA methylation can also have different outcomes, including activation of transcription. This is evidenced by the fact that transcription factors can interact with methylated DNA sequences. Furthermore, in certain cellular contexts, genes containing methylated promoters are highly transcribed. Interestingly, this uncoupling between methylated DNA and repression of transcription seems to be particularly evident in germ cells and pluripotent cells. Thus, contrary to previous assumptions, DNA methylation is not exclusively associated with repression of transcription initiation.
Collapse
Affiliation(s)
- Cornelia G Spruijt
- Department of Molecular Cancer Research, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Michiel Vermeulen
- 1] Department of Molecular Cancer Research, University Medical Center Utrecht, Utrecht, the Netherlands. [2] Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, the Netherlands. [3] Cancer Genomics Netherlands, the Netherlands
| |
Collapse
|
123
|
50 years of protein acetylation: from gene regulation to epigenetics, metabolism and beyond. Nat Rev Mol Cell Biol 2014; 16:258-64. [PMID: 25549891 DOI: 10.1038/nrm3931] [Citation(s) in RCA: 589] [Impact Index Per Article: 58.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In 1964, Vincent Allfrey and colleagues reported the identification of histone acetylation and with deep insight proposed a regulatory role for this protein modification in transcription regulation. Subsequently, histone acetyltransferases (HATs), histone deacetylases (HDACs) and acetyl-Lys-binding proteins were identified as transcription regulators, thereby providing compelling evidence for his daring hypothesis. During the past 15 years, reversible protein acetylation and its modifying enzymes have been implicated in many cellular functions beyond transcription regulation. Here, we review the progress accomplished during the past 50 years and discuss the future of protein acetylation.
Collapse
|
124
|
Abstract
The cardiac conduction system coordinates electrical activation through a series of interconnected structures, including the atrioventricular node (AVN), the central connection point that delays impulse propagation to optimize cardiac performance. Although recent studies have uncovered important molecular details of AVN formation, relatively little is known about the transcriptional mechanisms that regulate AV delay, the primary function of the mature AVN. We identify here MyoR as a novel transcription factor expressed in Cx30.2(+) cells of the AVN. We show that MyoR specifically inhibits a Cx30.2 enhancer required for AVN-specific gene expression. Furthermore, we demonstrate that MyoR interacts directly with Gata4 to mediate transcriptional repression. Our studies reveal that MyoR contains two nonequivalent repression domains. While the MyoR C-terminal repression domain inhibits transcription in a context-dependent manner, the N-terminal repression domain can function in a heterologous context to convert the Hand2 activator into a repressor. In addition, we show that genetic deletion of MyoR in mice increases Cx30.2 expression by 50% and prolongs AV delay by 13%. Taken together, we conclude that MyoR modulates a Gata4-dependent regulatory circuit that establishes proper AV delay, and these findings may have wider implications for the variability of cardiac rhythm observed in the general population.
Collapse
|
125
|
Peter S, Bultinck J, Myant K, Jaenicke LA, Walz S, Müller J, Gmachl M, Treu M, Boehmelt G, Ade CP, Schmitz W, Wiegering A, Otto C, Popov N, Sansom O, Kraut N, Eilers M. Tumor cell-specific inhibition of MYC function using small molecule inhibitors of the HUWE1 ubiquitin ligase. EMBO Mol Med 2014; 6:1525-41. [PMID: 25253726 PMCID: PMC4287973 DOI: 10.15252/emmm.201403927] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Revised: 07/30/2014] [Accepted: 08/26/2014] [Indexed: 12/14/2022] Open
Abstract
Deregulated expression of MYC is a driver of colorectal carcinogenesis, necessitating novel strategies to inhibit MYC function. The ubiquitin ligase HUWE1 (HECTH9, ARF-BP1, MULE) associates with both MYC and the MYC-associated protein MIZ1. We show here that HUWE1 is required for growth of colorectal cancer cells in culture and in orthotopic xenograft models. Using high-throughput screening, we identify small molecule inhibitors of HUWE1, which inhibit MYC-dependent transactivation in colorectal cancer cells, but not in stem and normal colon epithelial cells. Inhibition of HUWE1 stabilizes MIZ1. MIZ1 globally accumulates on MYC target genes and contributes to repression of MYC-activated target genes upon HUWE1 inhibition. Our data show that transcriptional activation by MYC in colon cancer cells requires the continuous degradation of MIZ1 and identify a novel principle that allows for inhibition of MYC function in tumor cells.
Collapse
Affiliation(s)
- Stefanie Peter
- Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Jennyfer Bultinck
- Cytokine Receptor Lab, Department of Biochemistry, Ghent University, Ghent, Belgium
| | | | - Laura A Jaenicke
- Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Susanne Walz
- Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Judith Müller
- Department of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Michael Gmachl
- Department Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Matthias Treu
- Department Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Guido Boehmelt
- Department Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Carsten P Ade
- Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Werner Schmitz
- Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Armin Wiegering
- Department of General, Visceral, Vascular and Paediatric Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Christoph Otto
- Department of General, Visceral, Vascular and Paediatric Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Nikita Popov
- Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| | | | - Norbert Kraut
- Department Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Martin Eilers
- Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| |
Collapse
|
126
|
Abstract
Gene expression is controlled through the recruitment of large coregulator complexes to specific gene loci to regulate chromatin structure by modifying epigenetic marks on DNA and histones. Metastasis-associated protein 1 (MTA1) is an essential component of the nucleosome remodelling and deacetylase (NuRD) complex that acts as a scaffold protein to assemble enzymatic activity and nucleosome targeting proteins. MTA1 consists of four characterised domains, a number of interaction motifs, and regions that are predicted to be intrinsically disordered. The ELM2-SANT domain is one of the best-characterised regions of MTA1, which recruits histone deacetylase 1 (HDAC1) and activates the enzyme in the presence of inositol phosphate. MTA1 is highly upregulated in several types of aggressive tumours and is therefore a possible target for cancer therapy. In this review, we summarise the structure and function of the four domains of MTA1 and discuss the possible functions of less well-characterised regions of the protein.
Collapse
Affiliation(s)
- Christopher J. Millard
- Henry Wellcome Laboratories of Structural Biology, Department of Biochemistry, University of Leicester, Leicester, LE1 9HN UK
| | - Louise Fairall
- Henry Wellcome Laboratories of Structural Biology, Department of Biochemistry, University of Leicester, Leicester, LE1 9HN UK
| | - John W. R. Schwabe
- Henry Wellcome Laboratories of Structural Biology, Department of Biochemistry, University of Leicester, Leicester, LE1 9HN UK
| |
Collapse
|
127
|
Teperek M, Miyamoto K, Simeone A, Feret R, Deery MJ, Gurdon JB, Jullien J. Sperm and spermatids contain different proteins and bind distinct egg factors. Int J Mol Sci 2014; 15:16719-40. [PMID: 25244019 PMCID: PMC4200797 DOI: 10.3390/ijms150916719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 07/21/2014] [Accepted: 09/09/2014] [Indexed: 01/14/2023] Open
Abstract
Spermatozoa are more efficient at supporting normal embryonic development than spermatids, their immature, immediate precursors. This suggests that the sperm acquires the ability to support embryonic development during spermiogenesis (spermatid to sperm maturation). Here, using Xenopus laevis as a model organism, we performed 2-D Fluorescence Difference Gel Electrophoresis (2D-DIGE) and mass spectrometry analysis of differentially expressed proteins between sperm and spermatids in order to identify factors that could be responsible for the efficiency of the sperm to support embryonic development. Furthermore, benefiting from the availability of egg extracts in Xenopus, we also tested whether the chromatin of sperm could attract different egg factors compared to the chromatin of spermatids. Our analysis identified: (1) several proteins which were present exclusively in sperm; but not in spermatid nuclei and (2) numerous egg proteins binding to the sperm (but not to the spermatid chromatin) after incubation in egg extracts. Amongst these factors we identified many chromatin-associated proteins and transcriptional repressors. Presence of transcriptional repressors binding specifically to sperm chromatin could suggest its preparation for the early embryonic cell cycles, during which no transcription is observed and suggests that sperm chromatin has a unique protein composition, which facilitates the recruitment of egg chromatin remodelling factors. It is therefore likely that the acquisition of these sperm-specific factors during spermiogenesis makes the sperm chromatin suitable to interact with the maternal factors and, as a consequence, to support efficient embryonic development.
Collapse
Affiliation(s)
- Marta Teperek
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK.
| | - Kei Miyamoto
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK.
| | - Angela Simeone
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK.
| | - Renata Feret
- Cambridge Centre for Proteomics, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK.
| | - Michael J Deery
- Cambridge Centre for Proteomics, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK.
| | - John B Gurdon
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK.
| | - Jerome Jullien
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK.
| |
Collapse
|
128
|
Gonzalez-Zuñiga M, Contreras PS, Estrada LD, Chamorro D, Villagra A, Zanlungo S, Seto E, Alvarez AR. c-Abl stabilizes HDAC2 levels by tyrosine phosphorylation repressing neuronal gene expression in Alzheimer's disease. Mol Cell 2014; 56:163-73. [PMID: 25219501 DOI: 10.1016/j.molcel.2014.08.013] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 07/02/2014] [Accepted: 08/07/2014] [Indexed: 10/24/2022]
Abstract
In Alzheimer's disease (AD), there is a decrease in neuronal gene expression induced by HDAC2 increase; however, the mechanisms involved are not fully elucidated. Here, we described how the tyrosine kinase c-Abl increases HDAC2 levels, inducing transcriptional repression of synaptic genes. Our data demonstrate that (1) in neurons, c-Abl inhibition with Imatinib prevents the AβO-induced increase in HDAC2 levels; (2) c-Abl knockdown cells show a decrease in HDAC2 levels, while c-Abl overexpression increases them; (3) c-Abl inhibition reduces HDAC2-dependent repression activity and HDAC2 recruitment to the promoter of several synaptic genes, increasing their expression; (4) c-Abl induces tyrosine phosphorylation of HDAC2, a posttranslational modification, affecting both its stability and repression activity; and (5) treatment with Imatinib decreases HDAC2 levels in a transgenic mice model of AD. Our results support the participation of the c-Abl/HDAC2 signaling pathway in the epigenetic blockade of gene expression in AD pathology.
Collapse
Affiliation(s)
- Marcelo Gonzalez-Zuñiga
- Department of Cell & Molecular Biology, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile; Biological and Chemistry Sciences Department, Universidad Bernardo O'Higgins, Santiago 8370993, Chile
| | - Pablo S Contreras
- Department of Cell & Molecular Biology, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile; Department of Gastroenterology, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Lisbell D Estrada
- Department of Cell & Molecular Biology, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile; Biological and Chemistry Sciences Department, Universidad Bernardo O'Higgins, Santiago 8370993, Chile
| | - David Chamorro
- Department of Cell & Molecular Biology, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Alejandro Villagra
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Silvana Zanlungo
- Department of Gastroenterology, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Edward Seto
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Alejandra R Alvarez
- Department of Cell & Molecular Biology, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile.
| |
Collapse
|
129
|
Liberman AC, Antunica-Noguerol M, Arzt E. Modulation of the Glucocorticoid Receptor Activity by Post-Translational Modifications. NUCLEAR RECEPTOR RESEARCH 2014. [DOI: 10.11131/2014/101086] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Ana Clara Liberman
- Instituto de Investigación en Biomedicina de Buenos Aires - CONICET - Partner Institute of the Max Planck Society
| | - María Antunica-Noguerol
- Instituto de Investigación en Biomedicina de Buenos Aires - CONICET - Partner Institute of the Max Planck Society
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires
| | - Eduardo Arzt
- Instituto de Investigación en Biomedicina de Buenos Aires - CONICET - Partner Institute of the Max Planck Society
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires
| |
Collapse
|
130
|
N-Myc differentially regulates expression of MXI1 isoforms in neuroblastoma. Neoplasia 2014; 15:1363-70. [PMID: 24403858 DOI: 10.1593/neo.11606] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 11/18/2013] [Accepted: 11/18/2013] [Indexed: 12/15/2022] Open
Abstract
Amplification of the MYCN proto-oncogene is associated with a poor prognosis in patients with metastatic neuroblastoma (NB). MYCN encodes the N-Myc protein, a transcriptional regulator that dimerizes with the Max transcription factor, binds to E-box DNA sequences, and regulates genes involved in cell growth and apoptosis. Overexpression of N-Myc leads to transcriptional activation and an increase in NB cell proliferation. Mxi1, a member of the Myc family of transcriptional regulators, also binds to Max. However, Mxi1 is a transcriptional repressor and inhibits proliferation of NB cells, suggesting that Mxi1 functions as an N-Myc antagonist. Our laboratory previously identified Mxi1-0, an alternatively transcribed Mxi1 isoform. Mxi1-0 has properties distinct from those of Mxi1; in contrast to Mxi1, Mxi1-0 is unable to suppress c-Myc-dependent transcription. We now show that Mxi1-0 expression increases in response to MYCN overexpression in NB cells, with a positive correlation between MYCN and MXI1-0 RNA levels. We also show that N-Myc expression differentially regulates the MXI1 and MXI1-0 promoters: Increased MYCN expression suppresses MXI1 promoter activity while enhancing transcription through the MXI1-0 promoter. Finally, induction of Mxi1-0 leads to increased proliferation, whereas expression of Mxi1 inhibits cell growth, indicating differential roles for these two proteins. These data suggest that N-Myc differentially regulates the expression of MXI1 and MXI1-0 and can alter the balance between the two transcription factors. Furthermore, MXI1-0 appears to be a downstream target of MYCN-dependent signaling pathways and may contribute to N-Myc-dependent cell growth and proliferation.
Collapse
|
131
|
Ji Q, Hu H, Yang F, Yuan J, Yang Y, Jiang L, Qian Y, Jiang B, Zou Y, Wang Y, Shao C, Gong Y. CRL4B interacts with and coordinates the SIN3A-HDAC complex to repress CDKN1A and drive cell cycle progression. J Cell Sci 2014; 127:4679-91. [PMID: 25189618 DOI: 10.1242/jcs.154245] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
CUL4B, a scaffold protein that assembles the CRL4B ubiquitin ligase complex, participates in the regulation of a broad spectrum of biological processes. Here, we demonstrate a crucial role of CUL4B in driving cell cycle progression. We show that loss of CUL4B results in a significant reduction in cell proliferation and causes G1 cell cycle arrest, accompanied by the upregulation of the cyclin-dependent kinase (CDK) inhibitors (CKIs) p21 and p57 (encoded by CDKN1A and CDKN1C, respectively). Strikingly, CUL4B was found to negatively regulate the function of p21 through transcriptional repression, but not through proteolysis. Furthermore, we demonstrate that CRL4B and SIN3A-HDAC complexes interact with each other and co-occupy the CDKN1A and CDKN1C promoters. Lack of CUL4B led to a decreased retention of SIN3A-HDAC components and increased levels of acetylated H3 and H4. Interestingly, the ubiquitylation function of CRL4B is not required for the stable retention of SIN3A-HDAC on the promoters of target genes. Thus, in addition to directly contributing to epigenetic silencing by catalyzing H2AK119 monoubiquitylation, CRL4B also facilitates the deacetylation function of SIN3A-HDAC. Our findings reveal a coordinated action between CRL4B and SIN3A-HDAC complexes in transcriptional repression.
Collapse
Affiliation(s)
- Qinghong Ji
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, 250012, China
| | - Huili Hu
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, 250012, China
| | - Fan Yang
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, 250012, China
| | - Jupeng Yuan
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, 250012, China
| | - Yang Yang
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, 250012, China
| | - Liangqian Jiang
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, 250012, China
| | - Yanyan Qian
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, 250012, China
| | - Baichun Jiang
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, 250012, China
| | - Yongxin Zou
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, 250012, China
| | - Yan Wang
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, 250012, China
| | - Changshun Shao
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, 250012, China
| | - Yaoqin Gong
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, 250012, China
| |
Collapse
|
132
|
Rinaldi L, Benitah SA. Epigenetic regulation of adult stem cell function. FEBS J 2014; 282:1589-604. [PMID: 25060320 DOI: 10.1111/febs.12946] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 07/17/2014] [Accepted: 07/22/2014] [Indexed: 01/09/2023]
Abstract
Understanding the cellular and molecular mechanisms that specify cell lineages throughout development, and that maintain tissue homeostasis during adulthood, is paramount towards our understanding of why we age or develop pathologies such as cancer. Epigenetic mechanisms ensure that genetically identical cells acquire different fates during embryonic development and are therefore essential for the proper progression of development. How they do so is still a matter of intense investigation, but there is sufficient evidence indicating that they act in a concerted manner with inductive signals and tissue-specific transcription factors to promote and stabilize fate changes along the three germ layers during development. In consequence, it is generally hypothesized that epigenetic mechanisms are also required for the continuous maintenance of cell fate during adulthood. However, in vivo models in which different epigenetic factors have been depleted in different tissues do not show overt changes in cell lineage, thus not strongly supporting this view. Instead, the function of some of these factors appears to be primarily associated with tissue functionality, and a strong causal relationship has been established between their misregulation and a diseased state. In this review, we summarize our current knowledge of the role of epigenetic factors in adult stem cell function and tissue homeostasis.
Collapse
Affiliation(s)
- Lorenzo Rinaldi
- Centre for Genomic Regulation, Barcelona, Spain; Universitat Pompeu Fabra, Barcelona, Spain; Institute for Research in Biomedicine, Barcelona, Spain
| | | |
Collapse
|
133
|
Tan KL, Ali A, Du Y, Fu H, Jin HX, Chin TM, Khan M, Go ML. Synthesis and evaluation of bisbenzylidenedioxotetrahydrothiopranones as activators of endoplasmic reticulum (ER) stress signaling pathways and apoptotic cell death in acute promyelocytic leukemic cells. J Med Chem 2014; 57:5904-18. [PMID: 24960549 PMCID: PMC4216202 DOI: 10.1021/jm401352a] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Curcumin is known to trigger ER-stress induced cell death of acute promyelocytic leukemic (APL) cells by intercepting the degradation of nuclear co-repressor (N-CoR) protein which has a key role in the pathogenesis of APL. Replacing the heptadienedione moiety of curcumin with a monocarbonyl cross-conjugated dienone embedded in a tetrahydrothiopyranone dioxide ring resulted in thiopyranone dioxides that were more resilient to hydrolysis and had greater growth inhibitory activities than curcumin on APL cells. Several members intercepted the degradation of misfolded N-CoR and triggered the signaling cascade in the unfolded protein response (UPR) which led to apoptotic cell death. Microarray analysis showed that genes involved in protein processing pathways that were germane to the activation of the UPR were preferentially up-regulated in treated APL cells, supporting the notion that the UPR was a consequential mechanistic pathway affected by thiopyranone dioxides. The Michael acceptor reactivity of the scaffold may have a role in exacerbating ER stress in APL cells.
Collapse
Affiliation(s)
- Kheng-Lin Tan
- Department of Pharmacy, National University of Singapore , 18 Science Drive 4, 117543, Republic of Singapore
| | | | | | | | | | | | | | | |
Collapse
|
134
|
Jamaladdin S, Kelly RDW, O'Regan L, Dovey OM, Hodson GE, Millard CJ, Portolano N, Fry AM, Schwabe JWR, Cowley SM. Histone deacetylase (HDAC) 1 and 2 are essential for accurate cell division and the pluripotency of embryonic stem cells. Proc Natl Acad Sci U S A 2014; 111:9840-5. [PMID: 24958871 PMCID: PMC4103379 DOI: 10.1073/pnas.1321330111] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Histone deacetylases 1 and 2 (HDAC1/2) form the core catalytic components of corepressor complexes that modulate gene expression. In most cell types, deletion of both Hdac1 and Hdac2 is required to generate a discernible phenotype, suggesting their activity is largely redundant. We have therefore generated an ES cell line in which Hdac1 and Hdac2 can be inactivated simultaneously. Loss of HDAC1/2 resulted in a 60% reduction in total HDAC activity and a loss of cell viability. Cell death is dependent upon cell cycle progression, because differentiated, nonproliferating cells retain their viability. Furthermore, we observe increased mitotic defects, chromatin bridges, and micronuclei, suggesting HDAC1/2 are necessary for accurate chromosome segregation. Consistent with a critical role in the regulation of gene expression, microarray analysis of Hdac1/2-deleted cells reveals 1,708 differentially expressed genes. Significantly for the maintenance of stem cell self-renewal, we detected a reduction in the expression of the pluripotent transcription factors, Oct4, Nanog, Esrrb, and Rex1. HDAC1/2 activity is regulated through binding of an inositol tetraphosphate molecule (IP4) sandwiched between the HDAC and its cognate corepressor. This raises the important question of whether IP4 regulates the activity of the complex in cells. By rescuing the viability of double-knockout cells, we demonstrate for the first time (to our knowledge) that mutations that abolish IP4 binding reduce the activity of HDAC1/2 in vivo. Our data indicate that HDAC1/2 have essential and pleiotropic roles in cellular proliferation and regulate stem cell self-renewal by maintaining expression of key pluripotent transcription factors.
Collapse
Affiliation(s)
- Shereen Jamaladdin
- Department of Biochemistry, University of Leicester, Leicester LE1 9HN, United Kingdom; and
| | - Richard D W Kelly
- Department of Biochemistry, University of Leicester, Leicester LE1 9HN, United Kingdom; and
| | - Laura O'Regan
- Department of Biochemistry, University of Leicester, Leicester LE1 9HN, United Kingdom; and
| | - Oliver M Dovey
- Wellcome Trust Sanger Institute, Hinxton, Cambs CB10 1SA, United Kingdom
| | - Grace E Hodson
- Department of Biochemistry, University of Leicester, Leicester LE1 9HN, United Kingdom; and
| | - Christopher J Millard
- Department of Biochemistry, University of Leicester, Leicester LE1 9HN, United Kingdom; and
| | - Nicola Portolano
- Department of Biochemistry, University of Leicester, Leicester LE1 9HN, United Kingdom; and
| | - Andrew M Fry
- Department of Biochemistry, University of Leicester, Leicester LE1 9HN, United Kingdom; and
| | - John W R Schwabe
- Department of Biochemistry, University of Leicester, Leicester LE1 9HN, United Kingdom; and
| | - Shaun M Cowley
- Department of Biochemistry, University of Leicester, Leicester LE1 9HN, United Kingdom; and
| |
Collapse
|
135
|
Garcia-Sanz P, Quintanilla A, Lafita MC, Moreno-Bueno G, García-Gutierrez L, Tabor V, Varela I, Shiio Y, Larsson LG, Portillo F, Leon J. Sin3b interacts with Myc and decreases Myc levels. J Biol Chem 2014; 289:22221-36. [PMID: 24951594 DOI: 10.1074/jbc.m113.538744] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Myc expression is deregulated in many human cancers. A yeast two-hybrid screen has revealed that the transcriptional repressor Sin3b interacts with Myc protein. Endogenous Myc and Sin3b co-localize and interact in the nuclei of human and rat cells, as assessed by co-immunoprecipitation, immunofluorescence, and proximity ligation assay. The interaction is Max-independent. A conserved Myc region (amino acids 186-203) is required for the interaction with Sin3 proteins. Histone deacetylase 1 is recruited to Myc-Sin3b complexes, and its deacetylase activity is required for the effects of Sin3b on Myc. Myc and Sin3a/b co-occupied many sites on the chromatin of human leukemia cells, although the presence of Sin3 was not associated with gene down-regulation. In leukemia cells and fibroblasts, Sin3b silencing led to Myc up-regulation, whereas Sin3b overexpression induced Myc deacetylation and degradation. An analysis of Sin3b expression in breast tumors revealed an association between low Sin3b expression and disease progression. The data suggest that Sin3b decreases Myc protein levels upon Myc deacetylation. As Sin3b is also required for transcriptional repression by Mxd-Max complexes, our results suggest that, at least in some cell types, Sin3b limits Myc activity through two complementary activities: Mxd-dependent gene repression and reduction of Myc levels.
Collapse
Affiliation(s)
- Pablo Garcia-Sanz
- From the Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Consejo Superior de Investigaciones Científicas, Universidad de Cantabria, Sociedad para el Desarrollo de Cantabria and the Departamento de Biología Molecular, Universidad de Cantabria, Santander 39011, Spain, the Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Facultad de Medicina, Universidad Autónoma de Madrid, 28046 Madrid, Spain, the Fundación M. D. Anderson Internacional, Madrid, Spain
| | - Andrea Quintanilla
- From the Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Consejo Superior de Investigaciones Científicas, Universidad de Cantabria, Sociedad para el Desarrollo de Cantabria and the Departamento de Biología Molecular, Universidad de Cantabria, Santander 39011, Spain
| | - M Carmen Lafita
- From the Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Consejo Superior de Investigaciones Científicas, Universidad de Cantabria, Sociedad para el Desarrollo de Cantabria and the Departamento de Biología Molecular, Universidad de Cantabria, Santander 39011, Spain
| | - Gema Moreno-Bueno
- the Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Facultad de Medicina, Universidad Autónoma de Madrid, 28046 Madrid, Spain, the Fundación M. D. Anderson Internacional, Madrid, Spain
| | - Lucia García-Gutierrez
- From the Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Consejo Superior de Investigaciones Científicas, Universidad de Cantabria, Sociedad para el Desarrollo de Cantabria and the Departamento de Biología Molecular, Universidad de Cantabria, Santander 39011, Spain
| | - Vedrana Tabor
- the Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm SE-17177, Sweden, and
| | - Ignacio Varela
- From the Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Consejo Superior de Investigaciones Científicas, Universidad de Cantabria, Sociedad para el Desarrollo de Cantabria and the Departamento de Biología Molecular, Universidad de Cantabria, Santander 39011, Spain
| | - Yuzuru Shiio
- the Greehey Children's Cancer Research Institute, The University of Texas Health Science Center, San Antonio, Texas 78229-3900
| | - Lars-Gunnar Larsson
- the Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm SE-17177, Sweden, and
| | - Francisco Portillo
- the Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Facultad de Medicina, Universidad Autónoma de Madrid, 28046 Madrid, Spain,
| | - Javier Leon
- From the Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Consejo Superior de Investigaciones Científicas, Universidad de Cantabria, Sociedad para el Desarrollo de Cantabria and the Departamento de Biología Molecular, Universidad de Cantabria, Santander 39011, Spain,
| |
Collapse
|
136
|
Weng X, Cheng X, Wu X, Xu H, Fang M, Xu Y. Sin3B mediates collagen type I gene repression by interferon gamma in vascular smooth muscle cells. Biochem Biophys Res Commun 2014; 447:263-70. [DOI: 10.1016/j.bbrc.2014.03.140] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 03/25/2014] [Indexed: 11/26/2022]
|
137
|
Link JM, Hurlin PJ. The activities of MYC, MNT and the MAX-interactome in lymphocyte proliferation and oncogenesis. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:554-62. [PMID: 24731854 DOI: 10.1016/j.bbagrm.2014.04.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 03/25/2014] [Accepted: 04/04/2014] [Indexed: 12/29/2022]
Abstract
The MYC family of proteins plays essential roles in embryonic development and in oncogenesis. Efforts over the past 30 years to define the transcriptional activities of MYC and how MYC functions to promote proliferation have produced evolving models of MYC function. One picture that has emerged of MYC and its partner protein MAX is of a transcription factor complex with a seemingly unique ability to stimulate the transcription of genes that are epigenetically poised for transcription and to amplify the transcription of actively transcribed genes. During lymphocyte activation, MYC is upregulated and stimulates a pro-proliferative program in part through the upregulation of a wide variety of metabolic effector genes that facilitate cell growth and cell cycle progression. MYC upregulation simultaneously sensitizes cells to apoptosis and activated lymphocytes and lymphoma cells have pro-survival attributes that allow MYC-driven proliferation to prevail. For example, the MAX-interacting protein MNT is upregulated in activated lymphocytes and was found to protect lymphocytes from MYC-dependent apoptosis. Here we review the activities of MYC, MNT and other MAX interacting proteins in the setting of T and B cell activation and oncogenesis. This article is part of a Special Issue entitled: Myc proteins in cell biology and pathology.
Collapse
Affiliation(s)
- Jason M Link
- Shriners Hospitals for Children Portland, 3101 SW Sam Jackson Park Road, Portland, OR 97239, USA; Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| | - Peter J Hurlin
- Shriners Hospitals for Children Portland, 3101 SW Sam Jackson Park Road, Portland, OR 97239, USA; Department of Cell and Developmental Biology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| |
Collapse
|
138
|
Shen W, Gao Y, Lu B, Zhang Q, Hu Y, Chen Y. Negatively regulating TLR4/NF-κB signaling via PPARα in endotoxin-induced uveitis. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1109-20. [PMID: 24717912 DOI: 10.1016/j.bbadis.2014.03.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 03/27/2014] [Accepted: 03/31/2014] [Indexed: 02/06/2023]
Abstract
Toll-like receptor (TLR) signaling plays a fundamental role in the induction and progression of autoimmune disease. In the present study, we showed that lipopolysaccharide (LPS), a TLR4 ligand, functions as an antagonist of peroxisome proliferator-activated receptor alpha (PPARα), a nuclear transcription factor. Using endotoxin induced uveitis (EIU) as a model, we found that TLR was negatively regulated by PPARα. Our data revealed that treatment with the PPARα agonist fenofibrate dramatically prevented LPS-induced uveitis and inhibited TLR/ Nuclear factor-kappaB (NF-κB) signaling during inflammation. Evaluation of the severity of anterior uveitis further showed that PPARα agonist treatment significantly decreased inflammatory cell infiltration, total protein concentration, vessel density, inflammatory cytokine production, and clinical scores in the anterior section of the eye during EIU. Moreover, fenofibrate administration recovered retinal function and decreased the production of inflammatory cytokines, retinal vascular leukostasis, and inflammatory cell infiltration into the posterior section of the eyes during EIU. In vitro studies further showed that down-regulation or deletion of PPARα led to increased TLR4 levels and the activation of NF-κB signaling in RPE cells and also blocked the anti-inflammatory effects of fenofibrate. Furthermore, activation or up-regulation of PPARα decreased TLR4 levels and inhibited the NF-κB signaling pathway induced by LPS in RPE cells. In TLR4-expressing reporter cells, activation or up-regulation of PPARα partially inhibited the activation of NF-κB and also decreased TLR4 transcriptional activity. In conclusion, the activation of PPARα represents a novel therapeutic strategy for human uveitis, as PPARα negatively regulates TLR4 activity and therefore exerts anti-inflammatory actions.
Collapse
Affiliation(s)
- Wei Shen
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Yang Gao
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, PR China
| | - Boyu Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, PR China
| | - Qingjiong Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, PR China
| | - Yang Hu
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
| | - Ying Chen
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
139
|
Cloning and characterization of neoplasia-related genes in flat oyster Ostrea edulis. INFECTION GENETICS AND EVOLUTION 2014; 23:138-49. [DOI: 10.1016/j.meegid.2014.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 02/04/2014] [Accepted: 02/11/2014] [Indexed: 12/23/2022]
|
140
|
Lehalle D, Sanlaville D, Guimier A, Plouvier E, Leblanc T, Galmiche L, Radford I, Romana S, Colleaux L, de Pontual L, Lyonnet S, Amiel J. Multiple congenital anomalies-intellectual disability (MCA-ID) and neuroblastoma in a patient harboring a de novo 14q23.1q23.3 deletion. Am J Med Genet A 2014; 164A:1310-7. [DOI: 10.1002/ajmg.a.36452] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 12/15/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Daphné Lehalle
- Département de Génétique Histologie-Embryologie-Cytogénétique; Hôpital Necker-Enfants Malades; Paris France
- INSERM U781; Université Sorbonne Paris Cité, Institut IMAGINE; Paris France
| | - Damien Sanlaville
- Hospices Civils de Lyon; Service de Génétique and CRNL; CNRS UMR 5292; INSERM U1028, Université Claude Bernard Lyon I; Lyon France
| | - Anne Guimier
- Département de Génétique Histologie-Embryologie-Cytogénétique; Hôpital Necker-Enfants Malades; Paris France
- INSERM U781; Université Sorbonne Paris Cité, Institut IMAGINE; Paris France
| | - Emmanuel Plouvier
- Service d'Onco-Hématologie Pédiatrique; Centre Hospitalo-Universitaire de Besançon; Paris France
| | - Thierry Leblanc
- Département d'Hématologie Pédiatrique; Hôpitaux Robert Debré et Université Paris Diderot; Paris France
| | - Louise Galmiche
- Département d'Anatomo-Pathologie; Hôpital Necker-Enfants Malades; Paris France
| | - Isabelle Radford
- Département de Génétique Histologie-Embryologie-Cytogénétique; Hôpital Necker-Enfants Malades; Paris France
| | - Serge Romana
- Département de Génétique Histologie-Embryologie-Cytogénétique; Hôpital Necker-Enfants Malades; Paris France
| | - Laurence Colleaux
- INSERM U781; Université Sorbonne Paris Cité, Institut IMAGINE; Paris France
| | - Loïc de Pontual
- INSERM U781; Université Sorbonne Paris Cité, Institut IMAGINE; Paris France
| | - Stanislas Lyonnet
- Département de Génétique Histologie-Embryologie-Cytogénétique; Hôpital Necker-Enfants Malades; Paris France
- INSERM U781; Université Sorbonne Paris Cité, Institut IMAGINE; Paris France
| | - Jeanne Amiel
- Département de Génétique Histologie-Embryologie-Cytogénétique; Hôpital Necker-Enfants Malades; Paris France
- INSERM U781; Université Sorbonne Paris Cité, Institut IMAGINE; Paris France
| |
Collapse
|
141
|
Hagelkruys A, Lagger S, Krahmer J, Leopoldi A, Artaker M, Pusch O, Zezula J, Weissmann S, Xie Y, Schöfer C, Schlederer M, Brosch G, Matthias P, Selfridge J, Lassmann H, Knoblich JA, Seiser C. A single allele of Hdac2 but not Hdac1 is sufficient for normal mouse brain development in the absence of its paralog. Development 2014; 141:604-616. [PMID: 24449838 PMCID: PMC4773893 DOI: 10.1242/dev.100487] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The histone deacetylases HDAC1 and HDAC2 are crucial regulators of chromatin structure and gene expression, thereby controlling important developmental processes. In the mouse brain, HDAC1 and HDAC2 exhibit different developmental stage- and lineage-specific expression patterns. To examine the individual contribution of these deacetylases during brain development, we deleted different combinations of Hdac1 and Hdac2 alleles in neural cells. Ablation of Hdac1 or Hdac2 by Nestin-Cre had no obvious consequences on brain development and architecture owing to compensation by the paralog. By contrast, combined deletion of Hdac1 and Hdac2 resulted in impaired chromatin structure, DNA damage, apoptosis and embryonic lethality. To dissect the individual roles of HDAC1 and HDAC2, we expressed single alleles of either Hdac1 or Hdac2 in the absence of the respective paralog in neural cells. The DNA-damage phenotype observed in double knockout brains was prevented by expression of a single allele of either Hdac1 or Hdac2. Strikingly, Hdac1−/−Hdac2+/− brains showed normal development and no obvious phenotype, whereas Hdac1+/−Hdac2−/− mice displayed impaired brain development and perinatal lethality. Hdac1+/−Hdac2−/− neural precursor cells showed reduced proliferation and premature differentiation mediated by overexpression of protein kinase C, delta, which is a direct target of HDAC2. Importantly, chemical inhibition or knockdown of protein kinase C delta was sufficient to rescue the phenotype of neural progenitor cells in vitro. Our data indicate that HDAC1 and HDAC2 have a common function in maintaining proper chromatin structures and show that HDAC2 has a unique role by controlling the fate of neural progenitors during normal brain development.
Collapse
Affiliation(s)
- Astrid Hagelkruys
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna 1030, Austria
| | - Sabine Lagger
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna 1030, Austria
| | - Julia Krahmer
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna 1030, Austria
| | - Alexandra Leopoldi
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna 1030, Austria
| | - Matthias Artaker
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna 1030, Austria
| | - Oliver Pusch
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Jürgen Zezula
- Institute of Pharmacology, Medical University of Vienna, Vienna 1090, Austria
| | - Simon Weissmann
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna 1030, Austria
| | - Yunli Xie
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna 1030, Austria
| | - Christian Schöfer
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Michaela Schlederer
- Ludwig Boltzmann Institute for Cancer Research (LBICR), Vienna 1090, Austria
| | - Gerald Brosch
- Division of Molecular Biology, Biocenter Innsbruck, Medical University, Innsbruck 6020, Austria
| | - Patrick Matthias
- Friedrich Miescher Institute for Biomedical Research, Novartis Research Foundation, Basel 4058, Switzerland
| | - Jim Selfridge
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh EH9 3QR, UK
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna 1090, Austria
| | - Jürgen A Knoblich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna 1030, Austria
| | - Christian Seiser
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna 1030, Austria
| |
Collapse
|
142
|
Skelton MM, Kampira EE, Wonkam AA, Mhandire KK, Kumwenda JJ, Duri KK, Dandara CC. Frequency variation among sub-Saharan populations in virus restriction gene, BST-2 proximal promoter polymorphisms: implications for HIV-1 prevalence differences among African countries. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2014; 18:461-71. [PMID: 24601767 DOI: 10.1089/omi.2013.0127] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The present study reports promoter variants in four sub-Saharan African populations that may affect BST-2 gene regulation. Recently, an in/del within the BST-2 promoter has been associated with HIV-1 disease progression in a Spanish cohort. Hence, we sequenced the proximal promoter region of the BST-2 gene in 581 individuals from South Africa, Zimbabwe, Malawi, and Cameroon. Seven SNPs were identified: rs28413176 (+26i6/Δ6); rs28413175 (-160i1/Δ1), -187A>G (nucleotide position -17516614); rs28413174 (-193G>A); rs73921425 (-199G>A); rs12609479 (-201C>T); and rs112492472 (-225C>T). The -199A and -225T alleles showed interesting trends across the sub-Saharan continent. Using predictive bioinformatics tools, we show that allelic variation at -199 and -201 potentially affect key transcription factor binding sites including bHLH, c-Myb, and E47. Importantly, data available from the ENCODE study gave further credence to our hypothesis of transcriptional regulation of BST-2 by a bHLH TF such as Mxi1. The possible repressive transcriptional effect of Mxi1 combined with the allelic frequency trend seen at -199 between African populations overlays well with current HIV-1 prevalence data, and may be a contributing factor to this phenomenon. The differences in HIV-1 prevalence in African countries could be, in part, due to distribution of genetic variants that affect susceptibility to HIV-1. Our findings therefore have substantive value for the design of future diagnostics for global health oriented diagnostics for HIV-1 susceptibility, and rational therapeutics on the critical path to personalized medicine in the African continent. As HIV-1 epidemiology vastly impacts human populations around the world, the population genomics strategy we have utilized herein can have value for other global regions as well.
Collapse
Affiliation(s)
- Michelle M Skelton
- 1 Division of Human Genetics, Faculty of Health Sciences, University of Cape Town , Observatory, Cape Town, South Africa
| | | | | | | | | | | | | |
Collapse
|
143
|
Moser MA, Hagelkruys A, Seiser C. Transcription and beyond: the role of mammalian class I lysine deacetylases. Chromosoma 2014; 123:67-78. [PMID: 24170248 PMCID: PMC3967066 DOI: 10.1007/s00412-013-0441-x] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 10/07/2013] [Accepted: 10/14/2013] [Indexed: 11/25/2022]
Abstract
The Rpd3-like members of the class I lysine deacetylase family are important regulators of chromatin structure and gene expression and have pivotal functions in the control of proliferation, differentiation and development. The highly related class I deacetylases HDAC1 and HDAC2 have partially overlapping but also isoform-specific roles in diverse biological processes, whereas HDAC3 and HDAC8 have unique functions. This review describes the role of class I KDACs in the regulation of transcription as well as their non-transcriptional functions, in particular their contributions to splicing, mitosis/meiosis, replication and DNA repair. During the past years, a number of mouse loss-of-function studies provided new insights into the individual roles of class I deacetylases in cell cycle control, differentiation and tumorigenesis. Simultaneous ablation of HDAC1 and HDAC2 or single deletion of Hdac3 severely impairs cell cycle progression in all proliferating cell types indicating that these class I deacetylases are promising targets for small molecule inhibitors as anti-tumor drugs.
Collapse
Affiliation(s)
- Mirjam Andrea Moser
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria
| | - Astrid Hagelkruys
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria
| | - Christian Seiser
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria
| |
Collapse
|
144
|
Wambua MK, Nalawansha DA, Negmeldin AT, Pflum MKH. Mutagenesis studies of the 14 Å internal cavity of histone deacetylase 1: insights toward the acetate-escape hypothesis and selective inhibitor design. J Med Chem 2014; 57:642-50. [PMID: 24405391 PMCID: PMC3983352 DOI: 10.1021/jm401837e] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
![]()
Histone
deacetylase (HDAC) proteins are promising targets for cancer
treatment, as shown by the approval of two HDAC inhibitors for the
treatment of cutaneous T-cell lymphoma. HDAC1 in particular has been
linked to cell growth and cell cycle regulation and is therefore an
attractive target for anticancer drugs. The HDAC1 active site contains
a hydrophobic 11 Å active-site channel, with a 14 Å internal
cavity at the bottom of the active site. Several computational and
biochemical studies have proposed an acetate-escape hypothesis where
the acetate byproduct of the deacetylation reaction escapes via the
14 Å internal cavity. Selective HDAC inhibitors that bind to
the 14 Å cavity have also been created. To understand the influence
of amino acids lining the HDAC1 14 Å cavity in acetate escape
and inhibitor binding, we used mutagenesis coupled with acetate competition
assays. The results indicate that amino acids lining the 14 Å
cavity are critical for catalytic activity and acetate competition,
confirming the role of the cavity in acetate escape. In addition,
these mutagenesis studies will aid in HDAC1-inhibitor design that
exploits the 14 Å cavity.
Collapse
Affiliation(s)
- Magdalene K Wambua
- Department of Chemistry, Wayne State University , 5101 Cass Avenue, Detroit, Michigan 48202, United States
| | | | | | | |
Collapse
|
145
|
Marchion D, Münster P. Development of histone deacetylase inhibitors for cancer treatment. Expert Rev Anticancer Ther 2014; 7:583-98. [PMID: 17428177 DOI: 10.1586/14737140.7.4.583] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Histone deacetylase (HDAC) inhibitors are an exciting new addition to the arsenal of cancer therapeutics. The inhibition of HDAC enzymes by HDAC inhibitors shifts the balance between the deacetylation activity of HDAC enzymes and the acetylation activity of histone acetyltransferases, resulting in hyperacetylation of core histones. Exposure of cancer cells to HDAC inhibitors has been associated with a multitude of molecular and biological effects, ranging from transcriptional control, chromatin plasticity, protein-DNA interaction to cellular differentiation, growth arrest and apoptosis. In addition to the antitumor effects seen with HDAC inhibitors alone, these compounds may also potentiate cytotoxic agents or synergize with other targeted anticancer agents. The exact mechanism by which HDAC inhibitors cause cell death is still unclear and the specific roles of individual HDAC enzymes as therapeutic targets has not been established. However, emerging evidence suggests that the effects of HDAC inhibitors on tumor cells may not only depend on the specificity and selectivity of the HDAC inhibitor, but also on the expression patterns of HDAC enzymes in the tumor tissue. In this review, the recent advances in the understanding and clinical development of HDAC inhibitors, as well as their current role in cancer therapy, will be discussed.
Collapse
Affiliation(s)
- Douglas Marchion
- H Lee Moffitt Cancer Center, Experimental Therapeutics Program, Department of Interdisciplinary Oncology, Tampa, FL 33612, USA
| | | |
Collapse
|
146
|
Conacci-Sorrell M, McFerrin L, Eisenman RN. An overview of MYC and its interactome. Cold Spring Harb Perspect Med 2014; 4:a014357. [PMID: 24384812 DOI: 10.1101/cshperspect.a014357] [Citation(s) in RCA: 304] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This review is intended to provide a broad outline of the biological and molecular functions of MYC as well as of the larger protein network within which MYC operates. We present a view of MYC as a sensor that integrates multiple cellular signals to mediate a broad transcriptional response controlling many aspects of cell behavior. We also describe the larger transcriptional network linked to MYC with emphasis on the MXD family of MYC antagonists. Last, we discuss evidence that the network has evolved for millions of years, dating back to the emergence of animals.
Collapse
|
147
|
The physiological roles of histone deacetylase (HDAC) 1 and 2: complex co-stars with multiple leading parts. Biochem Soc Trans 2013; 41:741-9. [PMID: 23697933 DOI: 10.1042/bst20130010] [Citation(s) in RCA: 235] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
HDACs (histone deacetylases) 1 and 2 are ubiquitous long-lived proteins, which are often found together in three major multiprotein co-repressor complexes: Sin3, NuRD (nucleosome remodelling and deacetylation) and CoREST (co-repressor for element-1-silencing transcription factor). Although there is a burgeoning number of non-histone proteins within the acetylome, these complexes contain multiple DNA/chromatin-recognition motifs, which, in combination with transcription factors, target HDAC1/2 to chromatin. Their physiological roles should therefore be viewed within the framework of chromatin manipulation. Classically, HDACs were thought to be recruited predominantly by transcriptional repressors to facilitate local histone deacetylation and transcriptional repression. More recently, genome-wide assays have mapped HDAC1/2 and their associated proteins to transcriptionally active loci and have provided alternative context-specific functions, whereby their repressive functions are subtly exerted to balance transcriptional activation and repression. With a few significant exceptions (early embryogenesis, brain development), HDAC1 and HDAC2 are functionally redundant. In most mouse knockout studies, deletion of both enzymes is required in order to produce a substantial phenotype. HDAC1/2 activity has been implicated in the development of numerous tissue and cell types, including heart, skin, brain, B-cells and T-cells. A common feature in all HDAC1/2-knockout, -knockdown and small-molecule inhibitor studies is a reduction in cell proliferation. A generic role in cell cycle progression could be exploited in cancer cells, by blocking HDAC1/2 activity with small-molecule inhibitors, making them potentially useful drug targets.
Collapse
|
148
|
Zhou J, Wang W, Gao Z, Peng X, Chen X, Chen W, Xu W, Xu H, Lin MC, Jiang S. MicroRNA-155 promotes glioma cell proliferation via the regulation of MXI1. PLoS One 2013; 8:e83055. [PMID: 24376632 PMCID: PMC3871643 DOI: 10.1371/journal.pone.0083055] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 10/30/2013] [Indexed: 01/06/2023] Open
Abstract
Gliomas are the most common and aggressive primary tumors in the central nervous system. Recently, Max interactor-1 (MXI1), an antagonist of c-Myc that is involved in brain tumor progression, has been reported to be deregulated in a variety of tumors including glioma. However, the mechanism of MXI1 deregulation in gliomas remains unclear. In this study, we show that the relative expression level of MXI1 is markedly down-regulated in glioma cell lines. Using integrated bioinformatic analysis and experimental confirmation, we identified several miRNAs by screening a panel of predicted miRNAs that may regulate the MXI1 3′UTR. The strongest inhibitory miRNA, miR-155, can attenuate the activity of a luciferase reporter gene that is fused with the MXI1 3′UTR and decrease the expression levels of MXI1 mRNA and protein in U87 glioma cells. The potential role of miR-155 in promoting glioma cell proliferation by targeting MXI1 was confirmed in various glioma cell lines by rescue experiments using MTT assays, EdU incorporation assay, and cell counting experiments. In addition, we determined that the level of MXI1 mRNA was inversely correlated with the expression of miR-155 in 18 sets of glioblastoma multiforme specimens. These findings reveal for the first time that the targeting of MXI1 by miR-155 may result in a reduction in MXI1 expression and promote glioma cell proliferation; this result suggests a novel function of miR-155 in targeting MXI1 in glioma-genesis.
Collapse
Affiliation(s)
- Jianwen Zhou
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Department of Pathology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Wang
- Neurosurgery Department, Epilepsy Centre, Guangzhou General Hospital, Guangzhou Command, PLA, Guangzhou, China
- HuaBo Bio-Pharmaceutic Institute of Guangzhou, Guangzhou, China
| | - Zhenhua Gao
- Department of Radiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xueling Peng
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Shenzhen State High-Tech Industrial Innovation Centre, Shenzhen, China
| | - Xulin Chen
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Wei Chen
- Department of Gynecology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Weiyi Xu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Haixiong Xu
- Department of Neurosurgery, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, China
| | - Marie C. Lin
- Biomedical Eng. Res. Center, Kunming Medical University, Kunming, China
| | - Songshan Jiang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- * E-mail:
| |
Collapse
|
149
|
Epigenetic silencing of the proapoptotic gene BIM in anaplastic large cell lymphoma through an MeCP2/SIN3a deacetylating complex. Neoplasia 2013; 15:511-22. [PMID: 23633923 DOI: 10.1593/neo.121784] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 02/12/2013] [Accepted: 02/15/2013] [Indexed: 01/29/2023] Open
Abstract
BIM is a proapoptotic member of the Bcl-2 family. Here, we investigated the epigenetic status of the BIM locus in NPM/ALK+ anaplastic large cell lymphoma (ALCL) cell lines and in lymph node biopsies from NPM/ALK+ ALCL patients. We show that BIM is epigenetically silenced in cell lines and lymph node specimens and that treatment with the deacetylase inhibitor trichostatin A restores the histone acetylation, strongly upregulates BIM expression, and induces cell death. BIM silencing occurs through recruitment of MeCP2 and the SIN3a/histone deacetylase 1/2 (HDAC1/2) corepressor complex. This event requires BIM CpG methylation/demethylation with 5-azacytidine that leads to detachment of the MeCP2 corepressor complex and reacetylation of the histone tails. Treatment with the ALK inhibitor PF2341066 or with an inducible shRNA targeting NPM/ALK does not restore BIM locus reacetylation; however, enforced expression of NPM/ALK in an NPM/ALK-negative cell line significantly increases the methylation at the BIM locus. This study demonstrates that BIM is epigenetically silenced in NPM/ALK-positive cells through recruitment of the SIN3a/HDAC1/2 corepressor complex and that NPM/ALK is dispensable to maintain BIM epigenetic silencing but is able to act as an inducer of BIM methylation.
Collapse
|
150
|
Kadamb R, Mittal S, Bansal N, Batra H, Saluja D. Sin3: insight into its transcription regulatory functions. Eur J Cell Biol 2013; 92:237-46. [PMID: 24189169 DOI: 10.1016/j.ejcb.2013.09.001] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 08/27/2013] [Accepted: 09/11/2013] [Indexed: 10/26/2022] Open
Abstract
Sin3, a large acidic protein, shares structural similarity with the helix-loop-helix dimerization domain of proteins of the Myc family of transcription factors. Sin3/HDAC corepressor complex functions in transcriptional regulation of several genes and is therefore implicated in the regulation of key biological processes. Knockdown studies have confirmed the role of Sin3 in cellular proliferation, differentiation, apoptosis and cell cycle regulation, emphasizing Sin3 as an essential regulator of critical cellular events in normal and pathological processes. The present review covers the diverse functions of this master transcriptional regulator as well as illustrates the redundant and distinct functions of its two mammalian isoforms.
Collapse
Affiliation(s)
- Rama Kadamb
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi 110007, India.
| | | | | | | | | |
Collapse
|