1
|
Lai PM, Chan KM. Roles of Histone H2A Variants in Cancer Development, Prognosis, and Treatment. Int J Mol Sci 2024; 25:3144. [PMID: 38542118 PMCID: PMC10969971 DOI: 10.3390/ijms25063144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/09/2024] [Accepted: 02/09/2024] [Indexed: 07/16/2024] Open
Abstract
Histones are nuclear proteins essential for packaging genomic DNA and epigenetic gene regulation. Paralogs that can substitute core histones (H2A, H2B, H3, and H4), named histone variants, are constitutively expressed in a replication-independent manner throughout the cell cycle. With specific chaperones, they can be incorporated to chromatin to modify nucleosome stability by modulating interactions with nucleosomal DNA. This allows the regulation of essential fundamental cellular processes for instance, DNA damage repair, chromosomal segregation, and transcriptional regulation. Among all the histone families, histone H2A family has the largest number of histone variants reported to date. Each H2A variant has multiple functions apart from their primary role and some, even be further specialized to perform additional tasks in distinct lineages, such as testis specific shortH2A (sH2A). In the past decades, the discoveries of genetic alterations and mutations in genes encoding H2A variants in cancer had revealed variants' potentiality in driving carcinogenesis. In addition, there is growing evidence that H2A variants may act as novel prognostic indicators or biomarkers for both early cancer detection and therapeutic treatments. Nevertheless, no studies have ever concluded all identified variants in a single report. Here, in this review, we summarize the respective functions for all the 19 mammalian H2A variants and their roles in cancer biology whilst potentiality being used in clinical setting.
Collapse
Affiliation(s)
| | - Kui Ming Chan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China;
| |
Collapse
|
2
|
Hsu KF, Wilkins SE, Hopkinson RJ, Sekirnik R, Flashman E, Kawamura A, McCullagh JS, Walport LJ, Schofield CJ. Hypoxia and hypoxia mimetics differentially modulate histone post-translational modifications. Epigenetics 2021; 16:14-27. [PMID: 32609604 PMCID: PMC7889154 DOI: 10.1080/15592294.2020.1786305] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/07/2020] [Accepted: 05/22/2020] [Indexed: 12/13/2022] Open
Abstract
Post-translational modifications (PTMs) to the tails of the core histone proteins are critically involved in epigenetic regulation. Hypoxia affects histone modifications by altering the activities of histone-modifying enzymes and the levels of hypoxia-inducible factor (HIF) isoforms. Synthetic hypoxia mimetics promote a similar response, but how accurately the hypoxia mimetics replicate the effects of limited oxygen availability on the levels of histone PTMs is uncertain. Here we report studies on the profiling of the global changes to PTMs on intact histones in response to hypoxia/hypoxia-related stresses using liquid chromatography-mass spectrometry (LC-MS). We demonstrate that intact protein LC-MS profiling is a relatively simple and robust method for investigating potential effects of drugs on histone modifications. The results provide insights into the profiles of PTMs associated with hypoxia and inform on the extent to which hypoxia and hypoxia mimetics cause similar changes to histones. These findings imply chemically-induced hypoxia does not completely replicate the substantial effects of physiological hypoxia on histone PTMs, highlighting that caution should be used in interpreting data from their use.
Collapse
Affiliation(s)
- Kuo-Feng Hsu
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
- Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Sarah E. Wilkins
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
| | - Richard J. Hopkinson
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
- Leicester Institute of Structural and Chemical Biology and School of Chemistry, University of Leicester, Leicester, UK
| | - Rok Sekirnik
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
| | - Emily Flashman
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
| | - Akane Kawamura
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, BHF Centre of Research Excellence, Wellcome Trust Centre for Human Genetics, Oxford, UK
- Chemistry - School of Natural and Environmental Sciences, Newcastle University, UK
| | - James S.O. McCullagh
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
| | - Louise J. Walport
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
- Protein-Protein Interaction Laboratory, The Francis Crick Institute, London, UK
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | | |
Collapse
|
3
|
Indole Derivative Interacts with Estrogen Receptor Beta and Inhibits Human Ovarian Cancer Cell Growth. Molecules 2020; 25:molecules25194438. [PMID: 32992652 PMCID: PMC7582771 DOI: 10.3390/molecules25194438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/16/2020] [Accepted: 09/25/2020] [Indexed: 01/08/2023] Open
Abstract
Ovarian cancer remains the leading cause of mortality among gynecological tumors. Estrogen receptor beta (ERβ) expression has been suggested to act as a tumor suppressor in epithelial ovarian cancer by reducing both tumor growth and metastasis. ERβ expression abnormalities represent a critical step in the development and progression of ovarian cancer: for these reasons, its re-expression by genetic engineering, as well as the use of targeted ERβ therapies, still constitute an important therapeutic approach. 3-{[2-chloro-1-(4-chlorobenzyl)-5-methoxy-6-methyl-1H-indol-3-yl]methylene}-5-hydroxy-6-methyl-1,3-dihydro-2H-indol-2-one, referred to here as compound 3, has been shown to have cytostatic as well cytotoxic effects on various hormone-dependent cancer cell lines. However, the mechanism of its anti-carcinogenic activity is not well understood. Here, we offer a possible explanation of such an effect in the human ovarian cancer cell line IGROV1. Chromatin binding protein assay and liquid chromatography mass spectrometry were exploited to localize and quantify compound 3 in cells. Molecular docking was used to prove compound 3 binding to ERβ. Mass spectrometry-based approaches were used to analyze histone post-translational modifications. Finally, gene expression analyses revealed a set of genes regulated by the ERβ/3 complex, namely CCND1, MYC, CDKN2A, and ESR2, providing possible molecular mechanisms that underline the observed antiproliferative effects.
Collapse
|
4
|
Leroy G, Dimaggio PA, Chan EY, Zee BM, Blanco MA, Bryant B, Flaniken IZ, Liu S, Kang Y, Trojer P, Garcia BA. A quantitative atlas of histone modification signatures from human cancer cells. Epigenetics Chromatin 2013; 6:20. [PMID: 23826629 PMCID: PMC3710262 DOI: 10.1186/1756-8935-6-20] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 06/18/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND An integral component of cancer biology is the understanding of molecular properties uniquely distinguishing one cancer type from another. One class of such properties is histone post-translational modifications (PTMs). Many histone PTMs are linked to the same diverse nuclear functions implicated in cancer development, including transcriptional activation and epigenetic regulation, which are often indirectly assayed with standard genomic technologies. Thus, there is a need for a comprehensive and quantitative profiling of cancer lines focused on their chromatin modification states. RESULTS To complement genomic expression profiles of cancer lines, we report the proteomic classification of 24 different lines, the majority of which are cancer cells, by quantifying the abundances of a large panel of single and combinatorial histone H3 and H4 PTMs, and histone variants. Concurrent to the proteomic analysis, we performed transcriptomic analysis on histone modifying enzyme abundances as a proxy for quantifying their activity levels. While the transcriptomic and proteomic results were generally consistent in terms of predicting histone PTM abundance from enzyme abundances, several PTMs were regulated independently of the modifying enzyme expression. In addition, combinatorial PTMs containing H3K27 methylation were especially enriched in breast cell lines. Knockdown of the predominant H3K27 methyltransferase, enhancer of zeste 2 (EZH2), in a mouse mammary xenograft model significantly reduced tumor burden in these animals and demonstrated the predictive utility of proteomic techniques. CONCLUSIONS Our proteomic and genomic characterizations of the histone modification states provide a resource for future investigations of the epigenetic and non-epigenetic determinants for classifying and analyzing cancer cells.
Collapse
Affiliation(s)
- Gary Leroy
- Epigenetics Program, Perelman School of Medicine, University of Pennsylvania, Smilow Center for Translational Research, 3400 Civic Center Blvd,, Bldg 421, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Furey A, Moriarty M, Bane V, Kinsella B, Lehane M. Ion suppression; a critical review on causes, evaluation, prevention and applications. Talanta 2013; 115:104-22. [PMID: 24054567 DOI: 10.1016/j.talanta.2013.03.048] [Citation(s) in RCA: 342] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 03/17/2013] [Accepted: 03/20/2013] [Indexed: 11/30/2022]
Abstract
The consequences of matrix effects in mass spectrometry analysis are a major issue of concern to analytical chemists. The identification of any ion suppressing (or enhancing) agents caused by sample matrix, solvent or LC-MS system components should be quantified and measures should be taken to eliminate or reduce the problem. Taking account of ion suppression should form part of the optimisation and validation of any quantitative LC-MS method. For example the US Food and Drug Administration has included the evaluation of matrix effects in its "Guidance for Industry on Bioanalytical Method Validation" (F.D.A. Department of Health and Human Services, Guidance for industry on bioanalytical method validation, Fed. Regist. 66 (100) 2001). If ion suppression is not assessed and corrected in an analytical method, the sensitivity of the LC-MS method can be seriously undermined, and it is possible that the target analyte may be undetected even when using very sensitive instrumentation. Sample analysis may be further complicated in cases where there are large sample-to-sample matrix variations (e.g. blood samples from different people can sometimes vary in certain matrix components, shellfish tissue samples sourced from different regions where different phytoplankton food sources are present, etc) and therefore exhibit varying ion-suppression effects. Although it is widely agreed that there is no generic method to overcome ion suppression, the purpose of this review is to: provide an overview of how ion suppression occurs, outline the methodologies used to assess and quantify the impact of ion suppression, discuss the various corrective actions that have been used to eliminate ion suppression in sample analysis, that is to say the deployment of techniques that eliminate or reduce the components in the sample matrix that cause ion suppression. This review article aims to collect together the latest information on the causes of ion suppression in LC-MS analysis and to consider the efficacy of common approaches to eliminate or reduce the problem using relevant examples published in the literature.
Collapse
Affiliation(s)
- Ambrose Furey
- Mass Spectrometry Research Centre (MSRC), Department of Chemistry, Cork Institute of Technology, Cork, Ireland; Team Elucidate/Mass Spectrometry Centre for Proteomic and Biotoxin Research (PROTEOBIO), Department of Chemistry, Cork Institute of Technology, Cork, Ireland.
| | | | | | | | | |
Collapse
|
6
|
Kakimoto Y, Tsuruyama T, Yamamoto T, Furuta M, Kotani H, Ozeki M, Yoshizawa A, Haga H, Tamaki K. Novel in situ pretreatment method for significantly enhancing the signal in MALDI-TOF MS of formalin-fixed paraffin-embedded tissue sections. PLoS One 2012; 7:e41607. [PMID: 22899997 PMCID: PMC3416821 DOI: 10.1371/journal.pone.0041607] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 06/22/2012] [Indexed: 01/03/2023] Open
Abstract
The application of matrix-assisted laser desorption/ionization (MALDI)-based mass spectrometry (MS) to the proteomic analysis of formalin-fixed paraffin-embedded (FFPE) tissue presents significant technical challenges. In situ enzymatic digestion is frequently used to unlock formalin-fixed tissues for analysis, but the results are often unsatisfactory. Here, we report a new, simplified in situ pretreatment method for preparing tissue sections for MS that involves heating with vapor containing acetonitrile in a small airtight pressurized space. The utility of the novel method is shown using FFPE tissue of human colon carcinoma. The number and intensity of MALDI peaks obtained from analysis of pretreated tissue was significantly higher than control tissue not subjected to pretreatment. A prominent peak (m/z 850) apparently specific to cancerous tissue was identified as a fragment of histone H2A in FFPE tissue pretreated using our method. This highly sensitive treatment may enable MALDI-MS analysis of archived pathological FFPE samples, thus leading to the identification of new biomarkers.
Collapse
Affiliation(s)
- Yu Kakimoto
- Department of Forensic Medicine and Molecular Pathology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tatsuaki Tsuruyama
- Department of Forensic Medicine and Molecular Pathology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- * E-mail:
| | - Takushi Yamamoto
- Analytical and Measuring Instruments Division, Kyoto Applications Development Center, Shimadzu Corporation, Kyoto, Japan
| | - Masaru Furuta
- Analytical and Measuring Instruments Division, Kyoto Applications Development Center, Shimadzu Corporation, Kyoto, Japan
| | - Hirokazu Kotani
- Department of Forensic Medicine and Molecular Pathology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Munetaka Ozeki
- Department of Forensic Medicine and Molecular Pathology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akihiko Yoshizawa
- Department of Diagnostic Pathology, Graduate School of Medicine, Shinshu University Hospital, Matsumoto, Japan
| | - Hironori Haga
- Department of Diagnostic Pathology, Graduate School of Medicine, Kyoto University Hospital, Kyoto, Japan
| | - Keiji Tamaki
- Department of Forensic Medicine and Molecular Pathology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
7
|
Kakimoto Y, Tsuruyama T, Yamamoto T, Furuta M, Kotani H, Ozeki M, Yoshizawa A, Haga H, Tamaki K. Novel in situ pretreatment method for significantly enhancing the signal in MALDI-TOF MS of formalin-fixed paraffin-embedded tissue sections. PLoS One 2012. [PMID: 22899997 DOI: 10.1371/journal.pone.0041607pone-d-12-07910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The application of matrix-assisted laser desorption/ionization (MALDI)-based mass spectrometry (MS) to the proteomic analysis of formalin-fixed paraffin-embedded (FFPE) tissue presents significant technical challenges. In situ enzymatic digestion is frequently used to unlock formalin-fixed tissues for analysis, but the results are often unsatisfactory. Here, we report a new, simplified in situ pretreatment method for preparing tissue sections for MS that involves heating with vapor containing acetonitrile in a small airtight pressurized space. The utility of the novel method is shown using FFPE tissue of human colon carcinoma. The number and intensity of MALDI peaks obtained from analysis of pretreated tissue was significantly higher than control tissue not subjected to pretreatment. A prominent peak (m/z 850) apparently specific to cancerous tissue was identified as a fragment of histone H2A in FFPE tissue pretreated using our method. This highly sensitive treatment may enable MALDI-MS analysis of archived pathological FFPE samples, thus leading to the identification of new biomarkers.
Collapse
Affiliation(s)
- Yu Kakimoto
- Department of Forensic Medicine and Molecular Pathology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Parolin C, Calonghi N, Presta E, Boga C, Caruana P, Naldi M, Andrisano V, Masotti L, Sartor G. Mechanism and stereoselectivity of HDAC I inhibition by (R)-9-hydroxystearic acid in colon cancer. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1821:1334-40. [PMID: 22814230 DOI: 10.1016/j.bbalip.2012.07.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 06/29/2012] [Accepted: 07/09/2012] [Indexed: 12/21/2022]
Abstract
9-Hydroxystearic acid (9-HSA) belongs to the endogenous lipid peroxidation by-products that decrease in tumors, causing as a consequence the loss of one of the control mechanisms on cell division. It acts as a histone deacetylase (HDAC, E.C 3.5.1.98) inhibitor, and the interaction of the two enantiomers of 9-HSA with the catalytic site of the enzyme, investigated by using a molecular modelling approach, has been reported to be different. In this work we tested out this prediction by synthesizing the two enantiomers (R)-9-HSA (R-9) and (S)-9-HSA (S-9) starting from the natural source methyl dimorphecolate obtained from Dimorphotheca sinuata seeds and investigating their biological activity in HT29 cells. Both enantiomers inhibit the enzymatic activity of HDAC1, HDAC2 and HDAC3, R-9 being more active; R-9 and S-9 inhibitory effect induces an increase in histone H4 acetylation. We also demonstrate that the antiproliferative effect brought about by R-9 is more pronounced as well as we observe increase of p21 transcription and protein content, while the expression of cyclin D1 is decreased. Starting from these observations it can be hypothesized that the interaction of R-9 with HDAC1 induce conformational changes in the enzyme causing loss of its interaction with other proteins, like cyclin D1 itself.
Collapse
Affiliation(s)
- Carola Parolin
- Department of Biochemistry "G. Moruzzi", Alma Mater Studiorum, University of Bologna, Via Irnerio 48, 40126, Bologna, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Činčárová L, Lochmanová G, Nováková K, Šultesová P, Konečná H, Fajkusová L, Fajkus J, Zdráhal Z. A combined approach for the study of histone deacetylase inhibitors. MOLECULAR BIOSYSTEMS 2012; 8:2937-45. [DOI: 10.1039/c2mb25136a] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
10
|
Badaloni E, Barbarino M, Cabri W, D’Acquarica I, Forte M, Gasparrini F, Giorgi F, Pierini M, Simone P, Ursini O, Villani C. Efficient organic monoliths prepared by γ-radiation induced polymerization in the evaluation of histone deacetylase inhibitors by capillary(nano)-high performance liquid chromatography and ion trap mass spectrometry. J Chromatogr A 2011; 1218:3862-75. [DOI: 10.1016/j.chroma.2011.04.048] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 04/14/2011] [Accepted: 04/16/2011] [Indexed: 11/25/2022]
|
11
|
Contrepois K, Ezan E, Mann C, Fenaille F. Ultra-High Performance Liquid Chromatography−Mass Spectrometry for the Fast Profiling of Histone Post-Translational Modifications. J Proteome Res 2010; 9:5501-9. [DOI: 10.1021/pr100497a] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Kévin Contrepois
- CEA, iBiTecS, Service de Biologie Intégrative et Génétique Moléculaire (SBIGeM) and Service de Pharmacologie et d’Immunoanalyse (SPI), F-91191 Gif Sur Yvette, France
| | - Eric Ezan
- CEA, iBiTecS, Service de Biologie Intégrative et Génétique Moléculaire (SBIGeM) and Service de Pharmacologie et d’Immunoanalyse (SPI), F-91191 Gif Sur Yvette, France
| | - Carl Mann
- CEA, iBiTecS, Service de Biologie Intégrative et Génétique Moléculaire (SBIGeM) and Service de Pharmacologie et d’Immunoanalyse (SPI), F-91191 Gif Sur Yvette, France
| | - François Fenaille
- CEA, iBiTecS, Service de Biologie Intégrative et Génétique Moléculaire (SBIGeM) and Service de Pharmacologie et d’Immunoanalyse (SPI), F-91191 Gif Sur Yvette, France
| |
Collapse
|
12
|
Dal Piaz F, Tosco A, Eletto D, Piccinelli AL, Moltedo O, Franceschelli S, Sbardella G, Remondelli P, Rastrelli L, Vesci L, Pisano C, De Tommasi N. The identification of a novel natural activator of p300 histone acetyltranferase provides new insights into the modulation mechanism of this enzyme. Chembiochem 2010; 11:818-27. [PMID: 20373302 DOI: 10.1002/cbic.200900721] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Many severe human pathologies are related to alterations of the fine balance between histone acetylation and deacetylation; because not all such diseases involve hypoacetylation, but also hyperacetylation, compounds able to enhance or repress the activities of histone acetyltransferases (HATs) could be promising therapeutic agents. We evaluated in vitro and in cell the ability of eleven natural polyisoprenylated benzophenone derivatives to modulate the HAT activity of p300/CBP, an enzyme that plays a pivotal role in a variety of cellular processes. Some of the tested compounds bound efficiently to the p300/CBP protein: in particular, guttiferone A, guttiferone E and clusianone inhibit its HAT activity, whereas nemorosone showed a surprising ability to activate the enzyme. The ability of nemorosone to penetrate cell membranes and modulate histone acetylation into the cell together with its high affinity for the p300/CBP enzyme made this compound a suitable lead for the design of optimized anticancer drugs. Besides, the studies performed at a cellular and molecular level on both the inhibitors and the activator provided new insights into the modulation mechanism of p300/CBP by small molecules.
Collapse
Affiliation(s)
- Fabrizio Dal Piaz
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Salerno, Via Ponte Don Melillo 1, 84084 Fisciano, SA, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Tobisawa Y, Imai Y, Fukuda M, Kawashima H. Sulfation of colonic mucins by N-acetylglucosamine 6-O-sulfotransferase-2 and its protective function in experimental colitis in mice. J Biol Chem 2010; 285:6750-60. [PMID: 20018871 PMCID: PMC2825469 DOI: 10.1074/jbc.m109.067082] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Revised: 12/04/2009] [Indexed: 12/20/2022] Open
Abstract
N-Acetylglucosamine 6-O-sulfotransferase-2 (GlcNAc6ST-2) catalyzes the sulfation of mucin-like glycoproteins, which function as ligands for a lymphocyte homing receptor, L-selectin, in the lymph node high endothelial venules (HEVs). We previously showed that GlcNAc6ST-2 is expressed not only in lymph node HEVs but also in the colonic epithelial cells in mice. Here we investigated the regulatory mechanism and physiological significance of colonic expression of GlcNAc6ST-2 in mice. Treatment of a mouse colonic epithelial cell line with butyrate, a short-chain fatty acid produced by anaerobic bacteria, induced GlcNAc6ST-2 expression in the presence of epidermal growth factor. Administration of butyrate in the drinking water stimulated GlcNAc6ST-2 expression in the mouse intestine, indicating that butyrate could serve as a regulatory molecule for the GlcNAc6ST-2 expression in vivo. Immunohistochemical analysis indicated that the sulfation of colonic mucins was greatly diminished in GlcNAc6ST-2-deficient mice. Liquid chromatography coupled to electrospray ionization tandem mass spectrometry of the colonic-mucin O-glycans from wild-type and GlcNAc6ST-2-deficient mice showed that GlcNAc-6-O-sulfation was the predominant sulfate modification of these mucins, and it was exclusively mediated by GlcNAc6ST-2. After colitis induction by dextran sulfate sodium, significantly more leukocyte infiltration was observed in the colon of GlcNAc6ST-2-deficient mice than in that of wild-type mice, indicating that the sulfation of colonic mucins by GlcNAc6ST-2 has a protective function in experimental colitis. These findings indicate that GlcNAc6ST-2, whose expression is regulated by butyrate, is a major sulfotransferase in the biosynthesis of sulfomucins in the mouse colon, where they serve as a mucosal barrier against colonic inflammation.
Collapse
Affiliation(s)
- Yuki Tobisawa
- From the Laboratory of Microbiology and Immunology and the Global COE Program, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Yasuyuki Imai
- From the Laboratory of Microbiology and Immunology and the Global COE Program, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Minoru Fukuda
- the Tumor Microenvironment Program, Cancer Research Center, Burnham Institute for Medical Research, La Jolla, California 92037, and
| | - Hiroto Kawashima
- From the Laboratory of Microbiology and Immunology and the Global COE Program, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
- PRESTO, Japan Science and Technology Agency, Kawaguchi 332-0012, Japan
| |
Collapse
|
14
|
Naldi M, Calonghi N, Masotti L, Parolin C, Valente S, Mai A, Andrisano V. Histone post-translational modifications by HPLC-ESI-MS after HT29 cell treatment with histone deacetylase inhibitors. Proteomics 2010; 9:5437-45. [PMID: 19834889 DOI: 10.1002/pmic.200800866] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The goal of the present work is to establish a correlation between the degree of histone post-translational modifications and the effects caused by treatment of HT29 colon cancer cells with class I-selective (MS-275 and MC1855), class II-selective (MC1568), and non-selective (suberoylanilide hydroxamic acid (SAHA) histone deacetylase inhibitors (HDACi). This correlation could afford a mean to better understand the mechanism of action of new, more potent, and selective HDACi directly on the cells. To this end, LC coupled to MS was applied in studies of time and concentration-dependent treatment with HDACi in HT29 cells. The results were correlated to their potency of histone deacetylase inhibition and to their effects on the cell cycle. The results indicate that the four tested inhibitors show a different pattern of time- and concentration-dependent modification after treatment of HT29 cells. At the selected concentrations, they cause different histone hyperacetylation and different cell cycle effects. In particular, SAHA (non-selective HDACi) affected hyperacetylation of all histones and caused massive cell death. MC1855 (class I-selective HDACi, hydroxamate) proved to be more potent and less toxic (cell arrest in G2/M phase) than SAHA. MS-275 (class I-selective HDACi, benzamide) exhibited a higher degree of hyperacetylation of H4 and a lower degree of H2A, H2B, and H3 acetylation, causing a cell arrest in G0/G1 phase. On the contrary, MC1568 (class II-selective HDACi) produced only a modest hyperacetylation of H4, was ineffective on the other histones, and showed no effect on cell cycle in HT29 cells.
Collapse
Affiliation(s)
- Marina Naldi
- Dipartimento di Scienze Farmaceutiche, Alma Mater Studiorum Università degli Studi di Bologna, Bologna, Italy
| | | | | | | | | | | | | |
Collapse
|
15
|
McLean CA, Wang Z, Babu K, Edwards A, Kasinathan P, Robl J, Sheppard AM. Normal development following chromatin transfer correlates with donor cell initial epigenetic state. Anim Reprod Sci 2009; 118:388-93. [PMID: 19632072 DOI: 10.1016/j.anireprosci.2009.06.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Revised: 06/10/2009] [Accepted: 06/23/2009] [Indexed: 10/20/2022]
Abstract
If the full potential of chromatin transfer (CT) technology is to be realized for both animal production and biomedical applications it is imperative that the efficiency of the reprogramming process be improved, and the potential for deleterious development be eliminated. Generation of the first cloned animals from adult somatic cells demonstrated that development is substantially an epigenetic process (Wilmut I, Schnieke AE, McWhir J, Kind AJ, Campbell KH, 1997. Viable offspring derived from fetal and adult mammalian cells. Nature. 385(6619): 810-813.). In this study, we provide preliminary evidence that the epigenetic state of the donor cell, may be valuable in assessing potential cloning success. We have measured key indicators of cellular epigenetic state in both serially derived cell populations of the same genetic origin, but differing in epigenomic status, and in a distinct cohort of donor cell populations with diverse genetic origins and epigenomic status. Specifically, the relative abundance of particular histone modifications in donor populations prior to manipulation has been correlated with the measurable variance in reprogramming efficiencies observed following CT, as defined by the number of resulting live births and healthy progeny, and the concomitant incidence of deleterious growth measures (notably the appearance of large offspring syndrome (LOS)). Thus, we suggest that the likely outcome and relative success of cloning may be predictable based on the expression of discriminating histone marks present in the donor cell population before CT. This approach may provide the basis of a prognostic signature for the future evaluation and risk assessment of putative donor cells prior to CT, and thus increase future cloning success and alleviate the incidence of abnormal development.
Collapse
Affiliation(s)
- Cameron A McLean
- AgResearch Ltd., Ruakura Research Centre, Hamilton, Private Bag 3123, New Zealand.
| | | | | | | | | | | | | |
Collapse
|
16
|
Su X, Jacob NK, Amunugama R, Hsu PH, Fishel R, Freitas MA. Enrichment and characterization of histones by two-dimensional hydroxyapatite/reversed-phase liquid chromatography-mass spectrometry. Anal Biochem 2009; 388:47-55. [PMID: 19454230 PMCID: PMC2848711 DOI: 10.1016/j.ab.2009.01.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Revised: 01/20/2009] [Accepted: 01/28/2009] [Indexed: 01/19/2023]
Abstract
Here we report a novel two-dimensional liquid chromatography-mass spectrometry (2D LC-MS) method that combines offline hydroxyapatite (HA) chromatography with online reversed-phase liquid chromatography-mass spectrometry (HA/RP LC-MS). The 2D LC-MS method was used to enrich and characterize histones and their posttranslational modifications. The 2D HA/RP LC-MS approach separates histones based on their relative binding affinity to DNA and relative hydrophobicity. HA/RP separations showed improvement in the number of histone isoforms detected as compared with one-dimensional RP LC-MS of acid-extracted histones. The improved histone fractionation resulted in better detection of lower abundant histone variants as well as their posttranslationally modified isoforms. Histones eluted from the HA/RP in the following order: H1, H2A/H2B heterodimers followed by H3/H4 heterotetramers, as predicted from their spatial organization in nucleosomes for binding affinity to DNA. Comparison between HA-purified and acid-extracted histones revealed similar histone profiles with the exception that the HA fractions showed a greater number of H1 isoforms. Two elution conditions were also examined: batch elution and salt gradient elution. Although both elution techniques were able to fractionate the histones sufficiently, the salt gradient approach has the most potential for purification of selected histone isoforms.
Collapse
Affiliation(s)
- Xiaodan Su
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University Medical Center, Columbus, OH-43210
| | - Naduparambil K. Jacob
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University Medical Center, Columbus, OH-43210
| | - Ravindra Amunugama
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University Medical Center, Columbus, OH-43210
| | - Pang-Hung Hsu
- The Genomics Research Center, Academia Sinica, Taiwan
| | - Richard Fishel
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University Medical Center, Columbus, OH-43210
| | - Michael A. Freitas
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University Medical Center, Columbus, OH-43210
| |
Collapse
|
17
|
Mackay CL, Ramsahoye B, Burgess K, Cook K, Weidt S, Creanor J, Harrison D, Langridge-Smith P, Hupp T, Hayward L. Sensitive, specific, and quantitative FTICR mass spectrometry of combinatorial post-translational modifications in intact histone H4. Anal Chem 2008; 80:4147-53. [PMID: 18457408 DOI: 10.1021/ac702452d] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We describe a quantitative Fourier transform ion cyclotron resonance mass spectrometric (FTICR MS) analysis of the relative proportions of post-translational modification states (PTMs) of core histones in cultured cells and tissues. A novel preseparation process using a monolithic column interfaced to a 12 T FTICR MS equipped with electron capture dissociation (ECD) yields very high mass accuracy spectra, allowing direct assignment of the PTMs present in the dominant modification states of intact H4, resolving a well recognized ambiguity between trimethylation and acetylation states. By eliminating preseparation, we also obtain a highly quantitative analysis of the distribution of H4 PTMs. Rapid, extensive, and reversible effects on PTMs induced by a histone deacetylase inhibitor indicate that H4 and other core histones are accessible to modification throughout the chromatin, not just in regions of active transcription. These methods provide tools for analysis of the histone code and its role in chromatin function.
Collapse
Affiliation(s)
- C Logan Mackay
- SIRCAMS, School of Chemistry, University of Edinburgh, Edinburgh, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Analysis of human histone H4 by capillary electrophoresis in a pullulan-coated capillary, LC-ESI-MS and MALDI-TOF-MS. Anal Bioanal Chem 2008; 390:1881-8. [DOI: 10.1007/s00216-008-1903-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Revised: 12/18/2007] [Accepted: 01/21/2008] [Indexed: 10/22/2022]
|
19
|
Arab S, Liu PP, Emili A. Lost in translation: five grand challenges for proteomic biomarker discovery. EXPERT OPINION ON MEDICAL DIAGNOSTICS 2007; 1:325-336. [PMID: 23489353 DOI: 10.1517/17530059.1.3.325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The biomedical community has the imperative to develop reliable, clinically relevant and generalizable bioassays that can be used to accurately recognize those individuals with early-stage disease or those patients who will respond to therapy, with the ultimate aim of achieving individualized medicine. In recent years, increasingly sophisticated proteomic screening technologies have been introduced, providing the biomedical community with a valuable new approach for the systematic discovery and validation of novel diagnostic, prognostic and therapeutic tools. Nevertheless, the complexity of the cellular milieu wherein a variety of macromolecules interact in dynamic fashion, combined with the complex clinical manifestation of chronic pathologies and widespread diversity of patient populations, mean that universal biomarkers will not be easily developed. In this review, the five key challenges that must be surmounted in order to advance the clinical impact of this nascent field are described, and plausible solutions based on the authors' own ongoing proteomic profiling of cardiovascular disease is outlined.
Collapse
Affiliation(s)
- Sara Arab
- University of Toronto, Toronto General Hospital, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | | | | |
Collapse
|
20
|
Su X, Jacob NK, Amunugama R, Lucas DM, Knapp AR, Ren C, Davis ME, Marcucci G, Parthun MR, Byrd JC, Fishel RA, Freitas MA. Liquid chromatography mass spectrometry profiling of histones. J Chromatogr B Analyt Technol Biomed Life Sci 2007; 850:440-54. [PMID: 17254850 PMCID: PMC2694509 DOI: 10.1016/j.jchromb.2006.12.037] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2006] [Revised: 12/08/2006] [Accepted: 12/17/2006] [Indexed: 11/25/2022]
Abstract
Here we describe the use of reverse-phase liquid chromatography mass spectrometry (RPLC-MS) to simultaneously characterize variants and post-translationally modified isoforms for each histone. The analysis of intact proteins significantly reduces the time of sample preparation and simplifies data interpretation. LC-MS analysis and peptide mass mapping have previously been applied to identify histone proteins and to characterize their post-translational modifications. However, these studies provided limited characterization of both linker histones and core histones. The current LC-MS analysis allows for the simultaneous observation of all histone PTMs and variants (both replacement and bulk histones) without further enrichment, which will be valuable in comparative studies. Protein identities were verified by the analysis of histone H2A species using RPLC fractionation, AU-PAGE separation and nano-LC-MS/MS.
Collapse
Affiliation(s)
- Xiaodan Su
- Department of Chemistry, Human Cancer Genetics, College of Medicine and Public Health The Ohio State University Columbus, OH
| | - Naduparambil K. Jacob
- Department of Molecular Virology, Immunology, and Medical Genetics; Human Cancer Genetics, College of Medicine and Public Health The Ohio State University Columbus, OH
| | - Ravindra Amunugama
- Department of Molecular Virology, Immunology, and Medical Genetics; Human Cancer Genetics, College of Medicine and Public Health The Ohio State University Columbus, OH
| | - David M. Lucas
- Department of Internal Medicine, Human Cancer Genetics, College of Medicine and Public Health The Ohio State University Columbus, OH
| | - Amy R. Knapp
- Department of Molecular and Cellular Biochemistry, Human Cancer Genetics, College of Medicine and Public Health The Ohio State University Columbus, OH
| | - Chen Ren
- Department of Chemistry, Human Cancer Genetics, College of Medicine and Public Health The Ohio State University Columbus, OH
| | - Melanie E. Davis
- Department of Internal Medicine, Human Cancer Genetics, College of Medicine and Public Health The Ohio State University Columbus, OH
| | - Guido Marcucci
- Department of Internal Medicine, Human Cancer Genetics, College of Medicine and Public Health The Ohio State University Columbus, OH
| | - Mark R. Parthun
- Department of Molecular and Cellular Biochemistry, Human Cancer Genetics, College of Medicine and Public Health The Ohio State University Columbus, OH
| | - John C. Byrd
- Department of Internal Medicine, Human Cancer Genetics, College of Medicine and Public Health The Ohio State University Columbus, OH
| | - Richard A. Fishel
- Department of Molecular Virology, Immunology, and Medical Genetics; Human Cancer Genetics, College of Medicine and Public Health The Ohio State University Columbus, OH
| | - Michael A. Freitas
- Department of Molecular Virology, Immunology, and Medical Genetics; Human Cancer Genetics, College of Medicine and Public Health The Ohio State University Columbus, OH
| |
Collapse
|
21
|
Ouvry-Patat SA, Schey KL. Characterization of antimicrobial histone sequences and posttranslational modifications by mass spectrometry. JOURNAL OF MASS SPECTROMETRY : JMS 2007; 42:664-74. [PMID: 17405180 DOI: 10.1002/jms.1200] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Histones typically play a role in DNA packaging and transcription regulation. These proteins are heavily modified by acetylation, methylation, phosphorylation and/or ubiquitination, and various combinations of these modifications alter histone functions and form the basis of the histone code. Furthermore, histones, including those found in shrimp, have recently been found to possess antimicrobial properties; however, the sequences and posttranslational modifications of shrimp histones are largely unknown. In this study mass spectrometry was used to characterize the primary structure of the shrimp antimicrobial histone. A combination of in-solution digestion and in-gel propionylation/digestion followed by LC-MS-MS and MALDI-TOF-TOF analysis was used. Over 80% of each histone sequence was obtained by in-solution digestion; however, none of the N-terminal domains was sequenced with this method. An in-gel propionylation method was optimized to recover and sequence the extremely hydrophilic histone N-termini. This method was then applied to shrimp hemocyte lysates separated on a 1-D SDS-PAGE gel. Overall, 95% coverage was obtained for the histone sequences as well as the identification of posttranslational sites such as acetylation, methylation and phosphorylation.
Collapse
Affiliation(s)
- Séverine A Ouvry-Patat
- Medical University of South Carolina, Department of Cell and Molecular Pharmacology, Charleston, SC 29425, USA
| | | |
Collapse
|
22
|
Weston AD, Ozolins TRS, Brown NA. Thoracic skeletal defects and cardiac malformations: a common epigenetic link? ACTA ACUST UNITED AC 2007; 78:354-70. [PMID: 17315248 DOI: 10.1002/bdrc.20084] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Congenital heart defects (CHDs) are the most common birth defects in humans. In addition, cardiac malformations represent the most frequently identified anomaly in teratogenicity experiments with laboratory animals. To explore the mechanisms of these drug-induced defects, we developed a model in which pregnant rats are treated with dimethadione, resulting in a high incidence of heart malformations. Interestingly, these heart defects were accompanied by thoracic skeletal malformations (cleft sternum, fused ribs, extra or missing ribs, and/or wavy ribs), which are characteristic of anterior-posterior (A/P) homeotic transformations and/or disruptions at one or more stages in somite development. A review of other teratogenicity studies suggests that the co-occurrence of these two disparate malformations is not unique to dimethadione, rather it may be a more general phenomenon caused by various structurally unrelated agents. The coexistence of cardiac and thoracic skeletal malformations has also presented clinically, suggesting a mechanistic link between cardiogenesis and skeletal development. Evidence from genetically modified mice reveals that several genes are common to heart development and to formation of the axial skeleton. Some of these genes are important in regulating chromatin architecture, while others are tightly controlled by chromatin-modifying proteins. This review focuses on the role of these epigenetic factors in development of the heart and axial skeleton, and examines the hypothesis that posttranslational modifications of core histones may be altered by some developmental toxicants.
Collapse
MESH Headings
- Abnormalities, Drug-Induced/etiology
- Abnormalities, Drug-Induced/genetics
- Abnormalities, Drug-Induced/metabolism
- Abnormalities, Multiple/etiology
- Abnormalities, Multiple/genetics
- Abnormalities, Multiple/metabolism
- Animals
- Bone and Bones/abnormalities
- Chromosomal Proteins, Non-Histone
- Epigenesis, Genetic
- Female
- Heart Defects, Congenital/etiology
- Heart Defects, Congenital/genetics
- Heart Defects, Congenital/metabolism
- Histones/metabolism
- Humans
- MicroRNAs/genetics
- Models, Biological
- Pregnancy
- Protein Processing, Post-Translational
- Ribs/abnormalities
- Sternum/abnormalities
- Teratogens/toxicity
- Transcription Factors/genetics
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Andrea D Weston
- Developmental and Reproductive Toxicology Center of Emphasis, Drug Safety Research, and Development, Pfizer Global Research and Development, Groton, Connecticut 06340, USA
| | | | | |
Collapse
|
23
|
Su X, Ren C, Freitas MA. Mass spectrometry-based strategies for characterization of histones and their post-translational modifications. Expert Rev Proteomics 2007; 4:211-25. [PMID: 17425457 PMCID: PMC2572816 DOI: 10.1586/14789450.4.2.211] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Due to the intimate interactions between histones and DNA, the characterization of histones has become the focus of great attention. A series of mass spectrometry-based technologies have been dedicated to the characterization and quantitation of different histone forms. This review focuses on the discussion of mass spectrometry-based strategies used for the characterization of histones and their post-translational modifications.
Collapse
Affiliation(s)
- Xiaodan Su
- The Ohio State Unviersity, Department of Molecular Virology Immunology & Medical Genetics, Human Cancer Genetics, Columbus, OH, USA
| | | | | |
Collapse
|