1
|
Behrends M, Engmann O. Linker histone H1.5 is an underestimated factor in differentiation and carcinogenesis. ENVIRONMENTAL EPIGENETICS 2020; 6:dvaa013. [PMID: 33214908 PMCID: PMC7660118 DOI: 10.1093/eep/dvaa013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/15/2020] [Accepted: 07/03/2020] [Indexed: 06/11/2023]
Abstract
Human histone H1.5, in mice called H1b, belongs to the family of linker histones (H1), which are key players in chromatin organization. These proteins sit on top of nucleosomes, in part to stabilize them, and recruit core histone modifying enzymes. Through subtype-specific deposition patterns and numerous post-translational modifications, they fine-tune gene expression and chromatin architecture, and help to control cell fate and homeostasis. However, even though it is increasingly implicated in mammalian development, H1.5 has not received as much research attention as its relatives. Recent studies have focused on its prognostic value in cancer patients and its contribution to tumorigenesis through specific molecular mechanisms. However, many functions of H1.5 are still poorly understood. In this review, we will summarize what is currently known about H1.5 and its function in cell differentiation and carcinogenesis. We will suggest key experiments that are required to understand the molecular network, in which H1.5 is embedded. These experiments will advance our understanding of the epigenetic reprogramming occurring in developmental and carcinogenic processes.
Collapse
Affiliation(s)
- Marthe Behrends
- Faculty of Medicine, Friedrich Schiller Universität, Jena, Thüringen 07747, Germany
| | - Olivia Engmann
- Institute for Human Genetics, Jena University Hospital, Am Klinikum 1, Thüringen 07747, Germany
| |
Collapse
|
2
|
Synergistic Anti Leukemia Effect of a Novel Hsp90 and a Pan Cyclin Dependent Kinase Inhibitors. Molecules 2020; 25:molecules25092220. [PMID: 32397330 PMCID: PMC7248782 DOI: 10.3390/molecules25092220] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML) is among the top four malignancies in Saudi nationals, and it is the top leukemia subtype worldwide. Resistance to available AML drugs requires the identification of new targets and agents. Hsp90 is one of the emerging important targets in AML, which has a central role in the regulation of apoptosis and cell proliferation through client proteins including the growth factor receptors and cyclin dependent kinases. The objective of the first part of this study is to investigate the putative Hsp90 inhibition activity of three novel previously synthesized quinazolines, which showed HL60 cytotoxicity and VEGFR2 and EGFR kinases inhibition activities. Using surface plasmon resonance, compound 1 (HAA2020) showed better Hsp90 inhibition compared to 17-AAG, and a docking study revealed that it fits nicely into the ATPase site. The objective of the second part is to maximize the anti-leukemic activity of HAA2020, which was combined with each of the eleven standard inhibitors. The best resulting synergistic effect in HL60 cells was with the pan cyclin-dependent kinases (CDK) inhibitor dinaciclib, using an MTT assay. Furthermore, the inhibiting effect of the Hsp90α gene by the combination of HAA2020 and dinaciclib was associated with increased caspase-7 and TNF-α, leading to apoptosis in HL60 cells. In addition, the combination upregulated p27 simultaneously with the inhibition of cyclinD3 and CDK2, leading to abolished HL60 proliferation and survival. The actions of HAA2020 propagated the apoptotic and cell cycle control properties of dinaciclib, showing the importance of co-targeting Hsp90 and CDK, which could lead to the better management of leukemia.
Collapse
|
3
|
Application of CE-MS for the analysis of histones and histone modifications. Methods 2020; 184:125-134. [PMID: 32014606 DOI: 10.1016/j.ymeth.2020.01.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/02/2019] [Accepted: 01/26/2020] [Indexed: 02/02/2023] Open
Abstract
The analysis, identification and quantification of histones and their post-translational modifications plays a central role in chromatin research and in studying epigenetic regulations during physiological processes. In the last decade analytical strategies based on mass spectrometry have been greatly improved for providing a global view of single modification abundances or to determine combinatorial patterns of modifications. Presented here is a newly developed strategy for histone protein analysis and a number of applications are illustrated with an emphasis on PTM characterization. Capillary electrophoresis is coupled to mass spectrometry (CE-MS) and has proven to be a very promising concept as it enables to study intact histones (top-down proteomics) as well as the analysis of enzymatically digested proteins (bottom-up proteomics). This technology combines highly efficient low-flow CE separations with ionization in a single device and offers an orthogonal separation principle to conventional LC-MS analysis, thus expanding the existing analytical repertoire in a perfect manner.
Collapse
|
4
|
Zheng H, Li X, Jia Q. Self-Assembling Glutamate-Functionalized Cyclodextrin Molecular Tube for Specific Enrichment of N-Linked Glycopeptides. ACS APPLIED MATERIALS & INTERFACES 2018; 10:19914-19921. [PMID: 29792669 DOI: 10.1021/acsami.8b01445] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Cyclodextrin molecular tube (CDMT), a new comer of cyclodextrin family, possesses large and hydrophilic outer area and stable structure. Its development and applications remain highly desired, especially in the field of separation and enrichment. Herein, we developed a CDMT-based enrichment platform focusing on the specific capture of glycopeptides. To enhance the hydrophilicity of CDMT, it was functionalized with glutamate (glu). The prepared gluCDMT exhibited large hydrophilic surface, high stability, and good acidic/alkalic resistance. A solid monolithic support was employed to immobilize gluCDMT by a host-guest self-assembly synthetic strategy, which did not occupy the surface hydrophilic sites. The gluCDMT-based monolith exhibited high binding capacity (∼50 mg g-1), good ability to capture glycopeptides (23 HRP glycopeptides and 28 IgG glycopeptides), and high selectivity (horseradish peroxidase/bovine serum albumin = 1:10 000). Moreover, the developed platform was successfully applied to analyze glycopetides in acute myelogenous leukemia cell lysate and human serum samples.
Collapse
Affiliation(s)
| | - Xiqian Li
- China-Japan Hospital of Jilin University , Changchun 130033 , China
| | | |
Collapse
|
5
|
Abbaoui B, Telu KH, Lucas CR, Thomas-Ahner JM, Schwartz SJ, Clinton SK, Freitas MA, Mortazavi A. The impact of cruciferous vegetable isothiocyanates on histone acetylation and histone phosphorylation in bladder cancer. J Proteomics 2017; 156:94-103. [PMID: 28132875 DOI: 10.1016/j.jprot.2017.01.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 12/07/2016] [Accepted: 01/25/2017] [Indexed: 12/17/2022]
Abstract
Cruciferous vegetable intake is associated with reduced risk of bladder cancer, yet mechanisms remain unclear. Cruciferous vegetable isothiocyanates (ITCs), namely sulforaphane (SFN) and erucin (ECN), significantly inhibit histone deacetylase (HDAC) activity in human bladder cancer cells representing superficial to invasive biology (59-83% inhibition with 20μM, 48h treatment), and in bladder cancer xenografts (59±3% ECN inhibition). Individual HDACs inhibited by SFN and ECN include HDACs 1, 2, 4 and 6. Interestingly, global acetylation status of histones H3 or H4 remain unaltered. The interplay between HDAC inhibition and modest modulation of AcH3 and AcH4 status is partially explained by decreased histone acetyl transferase activity (48.8±5.3%). In contrast, a significant decrease in phosphorylation status of all isoforms of histone H1 was observed, concomitant with increased phosphatase PP1β and PP2A activity. Together, these findings suggest that ITCs modulate histone status via HDAC inhibition and phosphatase enhancement. This allows for reduced levels of histone H1 phosphorylation, a marker correlated with human bladder cancer progression. Therefore, ITC-mediated inhibition of histone H1 phosphorylation presents a novel direction of research in elucidating epidemiological relationships and supports future food-based prevention strategies. SIGNIFICANCE Collectively, our findings suggest that the cruciferous vegetable isothiocyanates: sulforaphane (SFN) and erucin (ECN), impact histones status in bladder cancer cells by modulating specific HDACs and HATs, and enhancing phosphatase activity, resulting in reduction of histone H1 phosphorylation. These findings are significant due to the fact that our previous work positively correlated histone H1 phosphorylation with bladder cancer carcinogenesis and progression. Therefore, we propose that SFN and ECN may inhibit bladder carcinogenesis via epigenetic modulation of gene expression associated with histone H1 phosphorylation. These efforts may elucidate biomarkers useful in epidemiologic studies related to cruciferous vegetable intake and cancer risk or provide intermediate biomarkers for food-based clinical intervention studies in high-risk cohorts.
Collapse
Affiliation(s)
- Besma Abbaoui
- The Integrated Biomedical Science Graduate Program, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Kelly H Telu
- Department of Chemistry, College of Arts and Sciences, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Christopher R Lucas
- The Integrated Biomedical Science Graduate Program, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | | | - Steven J Schwartz
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; Department of Food Science and Technology, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Steven K Clinton
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; Division of Medical Oncology, Department of Internal Medicine, College of Medicine; The Ohio State University, Columbus, OH 43210, USA
| | - Michael A Freitas
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Amir Mortazavi
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; Division of Medical Oncology, Department of Internal Medicine, College of Medicine; The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
6
|
Singh R, Harshman SW, Ruppert AS, Mortazavi A, Lucas DM, Thomas-Ahner JM, Clinton SK, Byrd JC, Freitas MA, Parthun MR. Proteomic profiling identifies specific histone species associated with leukemic and cancer cells. Clin Proteomics 2015; 12:22. [PMID: 26321891 PMCID: PMC4551702 DOI: 10.1186/s12014-015-9095-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 08/12/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chromatin is an extraordinarily complex structure. Much of this complexity results from the presence of numerous histone post-translational modifications and histone variants. Alterations in the patterns of histone post-translational modifications are emerging as a feature of many types of cancer and have been shown to have prognostic value. RESULTS We have applied a liquid chromatography/mass spectrometry-based approach to comprehensively characterize the histone proteome in primary samples from chronic lymphocytic leukemia (CLL) patients, as well as bladder and breast cancer cell culture models. When compared to non-malignant CD19+ B cells from healthy donors, the CLL histone proteome showed a distinct signature of differentially expressed species, spanning all the histones studied and including both post-translationally modified species and unmodified, non-allelic replication-dependent histone isoforms. However, the large changes in histone H3 and H4 that are characteristic of many cancer types were not observed. One of species of H2A (mass = 14,063 Da) was the most strongly associated with time to treatment in CLL patients. CLL patient samples also demonstrated histone profiles that were distinct from those of the bladder and breast cancer cells. CONCLUSIONS Signatures of histone profiles are complex and can distinguish between healthy individuals and CLL patients and may provide prognostic markers. In addition, histone profiles may define tissue specific malignancies.
Collapse
Affiliation(s)
- Rajbir Singh
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210 USA
| | - Sean W Harshman
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, OH 43210 USA
| | - Amy S Ruppert
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210 USA
| | - Amir Mortazavi
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210 USA
| | - David M Lucas
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210 USA ; Division of Medicinal Chemistry in the College of Pharmacy, The Ohio State University, Columbus, OH 43210 USA
| | | | - Steven K Clinton
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210 USA
| | - John C Byrd
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210 USA
| | - Michael A Freitas
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, OH 43210 USA
| | - Mark R Parthun
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210 USA
| |
Collapse
|
7
|
Chen Y, Hoover ME, Dang X, Shomo AA, Guan X, Marshall AG, Freitas MA, Young NL. Quantitative Mass Spectrometry Reveals that Intact Histone H1 Phosphorylations are Variant Specific and Exhibit Single Molecule Hierarchical Dependence. Mol Cell Proteomics 2015. [PMID: 26209608 DOI: 10.1074/mcp.m114.046441] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Breast cancer was the second leading cause of cancer related mortality for females in 2014. Recent studies suggest histone H1 phosphorylation may be useful as a clinical biomarker of breast and other cancers because of its ability to recognize proliferative cell populations. Although monitoring a single phosphorylated H1 residue is adequate to stratify high-grade breast tumors, expanding our knowledge of how H1 is phosphorylated through the cell cycle is paramount to understanding its role in carcinogenesis. H1 analysis by bottom-up MS is challenging because of the presence of highly homologous sequence variants expressed by most cells. These highly basic proteins are difficult to analyze by LC-MS/MS because of the small, hydrophilic nature of peptides produced by tryptic digestion. Although bottom-up methods permit identification of several H1 phosphorylation events, these peptides are not useful for observing the combinatorial post-translational modification (PTM) patterns on the protein of interest. To complement the information provided by bottom-up MS, we utilized a top-down MS/MS workflow to permit identification and quantitation of H1 proteoforms related to the progression of breast cells through the cell cycle. Histones H1.2 and H1.4 were observed in MDA-MB-231 metastatic breast cells, whereas an additional histone variant, histone H1.3, was identified only in nonneoplastic MCF-10A cells. Progressive phosphorylation of histone H1.4 was identified in both cell lines at mitosis (M phase). Phosphorylation occurred first at S172 followed successively by S187, T18, T146, and T154. Notably, phosphorylation at S173 of histone H1.2 and S172, S187, T18, T146, and T154 of H1.4 significantly increases during M phase relative to S phase, suggesting that these events are cell cycle-dependent and may serve as markers for proliferation. Finally, we report the observation of the H1.2 SNP variant A18V in MCF-10A cells.
Collapse
Affiliation(s)
- Yu Chen
- From the ‡Ion Cyclotron Resonance Program, National High Magnetic Field Laboratory, Florida State University, Tallahassee, Florida, 32310
| | - Michael E Hoover
- §Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, Ohio, College of Medicine and Arthur G. James Comprehensive Cancer Center, Columbus, Ohio, 43210
| | - Xibei Dang
- ¶Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, 32306
| | - Alan A Shomo
- ¶Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, 32306
| | - Xiaoyan Guan
- From the ‡Ion Cyclotron Resonance Program, National High Magnetic Field Laboratory, Florida State University, Tallahassee, Florida, 32310
| | - Alan G Marshall
- From the ‡Ion Cyclotron Resonance Program, National High Magnetic Field Laboratory, Florida State University, Tallahassee, Florida, 32310; ¶Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, 32306
| | - Michael A Freitas
- §Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, Ohio, College of Medicine and Arthur G. James Comprehensive Cancer Center, Columbus, Ohio, 43210;
| | - Nicolas L Young
- From the ‡Ion Cyclotron Resonance Program, National High Magnetic Field Laboratory, Florida State University, Tallahassee, Florida, 32310;
| |
Collapse
|
8
|
Renusch SR, Harshman S, Pi H, Workman E, Wehr A, Li X, Prior TW, Elsheikh BH, Swoboda KJ, Simard LR, Kissel JT, Battle D, Parthun MR, Freitas MA, Kolb SJ. Spinal Muscular Atrophy Biomarker Measurements from Blood Samples in a Clinical Trial of Valproic Acid in Ambulatory Adults. J Neuromuscul Dis 2015; 2:119-130. [PMID: 27858735 PMCID: PMC5271431 DOI: 10.3233/jnd-150081] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background: Clinical trials of therapies for spinal muscular atrophy (SMA) that are designed to increase the expression the SMN protein ideally include careful assessment of relevant SMN biomarkers. Objective: In the SMA VALIANT trial, a recent double-blind placebo-controlled crossover study of valproic acid (VPA) in ambulatory adult subjects with SMA, we investigated relevant pharmacodynamic biomarkers in blood samples from SMA subjects by direct longitudinal measurement of histone acetylation and SMN mRNA and protein levels in the presence and absence of VPA treatment. Methods: Thirty-three subjects were randomized to either VPA or placebo for the first 6 months followed by crossover to the opposite arm for an additional 6 months. Outcome measures were compared between the two treatments (VPA and placebo) using a standard crossover analysis. Results: A significant increase in histone H4 acetylation was observed with VPA treatment (p = 0.005). There was insufficient evidence to suggest a treatment effect with either full length or truncated SMN mRNA transcript levels or SMN protein levels. Conclusions: These measures were consistent with the observed lack of change in the primary clinical outcome measure in the VALIANT trial. These results also highlight the added benefit of molecular and pharmacodynamic biomarker measurements in the interpretation of clinical trial outcomes.
Collapse
Affiliation(s)
- Samantha R Renusch
- Department of Molecular & Cellular Biochemistry, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Sean Harshman
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Hongyang Pi
- Department of Molecular & Cellular Biochemistry, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Eileen Workman
- Department of Molecular & Cellular Biochemistry, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Allison Wehr
- Center for Biostatistics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Xiaobai Li
- Center for Biostatistics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Thomas W Prior
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Bakri H Elsheikh
- Department of Neurology, Dhahran Medical Center, Dhahran, Saudi Arabia
| | - Kathryn J Swoboda
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Louise R Simard
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - John T Kissel
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Daniel Battle
- Department of Molecular & Cellular Biochemistry, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Mark R Parthun
- Department of Molecular & Cellular Biochemistry, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Michael A Freitas
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Stephen J Kolb
- Department of Molecular & Cellular Biochemistry, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
9
|
Harshman SW, Hoover ME, Huang C, Branson OE, Chaney S, Cheney CM, Rosol TJ, Shapiro CL, Wysocki VH, Huebner K, Freitas MA. Histone H1 phosphorylation in breast cancer. J Proteome Res 2014; 13:2453-67. [PMID: 24601643 PMCID: PMC4012839 DOI: 10.1021/pr401248f] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Indexed: 12/18/2022]
Abstract
Breast cancer is the second leading cause of cancer-related deaths in women. The need for new clinical biomarkers in breast cancer is necessary to further predict prognosis and therapeutic response. In this article, the LC-MS histone H1 phosphorylation profiles were established for three distinct breast cancer cell lines. The results show that the extent of H1 phosphorylation can distinguish between the different cell lines. The histone H1 from the metastatic cell line, MDA-MB-231, was subjected to chemical derivitization and LC-MS/MS analysis. The results suggest that the phosphorylation at threonine 146 is found on both histone H1.2 and histone H1.4. Cell lines were then treated with an extracellular stimulus, estradiol or kinase inhibitor LY294002, to monitor changes in histone H1 phosphorylation. The data show that histone H1 phosphorylation can increase and decrease in response to extracellular stimuli. Finally, primary breast tissues were stained for the histone H1 phosphorylation at threonine 146. Variable staining patterns across tumor grades and subtypes were observed with pT146 labeling correlating with tumor grade. These results establish the potential for histone H1 phosphorylation at threonine 146 as a clinical biomarker in breast cancer.
Collapse
Affiliation(s)
- Sean W. Harshman
- Department of Molecular Virology, Immunology and
Medical Genetics, Comprehensive Cancer
Center, Department
of Chemistry & Biochemistry, Veterinary Biosciences, College of Veterinary
Medicine, and Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Michael E. Hoover
- Department of Molecular Virology, Immunology and
Medical Genetics, Comprehensive Cancer
Center, Department
of Chemistry & Biochemistry, Veterinary Biosciences, College of Veterinary
Medicine, and Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Chengsi Huang
- Department of Molecular Virology, Immunology and
Medical Genetics, Comprehensive Cancer
Center, Department
of Chemistry & Biochemistry, Veterinary Biosciences, College of Veterinary
Medicine, and Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Owen E. Branson
- Department of Molecular Virology, Immunology and
Medical Genetics, Comprehensive Cancer
Center, Department
of Chemistry & Biochemistry, Veterinary Biosciences, College of Veterinary
Medicine, and Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Sarah
B. Chaney
- Department of Molecular Virology, Immunology and
Medical Genetics, Comprehensive Cancer
Center, Department
of Chemistry & Biochemistry, Veterinary Biosciences, College of Veterinary
Medicine, and Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Carolyn M. Cheney
- Department of Molecular Virology, Immunology and
Medical Genetics, Comprehensive Cancer
Center, Department
of Chemistry & Biochemistry, Veterinary Biosciences, College of Veterinary
Medicine, and Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Thomas J. Rosol
- Department of Molecular Virology, Immunology and
Medical Genetics, Comprehensive Cancer
Center, Department
of Chemistry & Biochemistry, Veterinary Biosciences, College of Veterinary
Medicine, and Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Charles L. Shapiro
- Department of Molecular Virology, Immunology and
Medical Genetics, Comprehensive Cancer
Center, Department
of Chemistry & Biochemistry, Veterinary Biosciences, College of Veterinary
Medicine, and Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Vicki H. Wysocki
- Department of Molecular Virology, Immunology and
Medical Genetics, Comprehensive Cancer
Center, Department
of Chemistry & Biochemistry, Veterinary Biosciences, College of Veterinary
Medicine, and Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Kay Huebner
- Department of Molecular Virology, Immunology and
Medical Genetics, Comprehensive Cancer
Center, Department
of Chemistry & Biochemistry, Veterinary Biosciences, College of Veterinary
Medicine, and Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Michael A. Freitas
- Department of Molecular Virology, Immunology and
Medical Genetics, Comprehensive Cancer
Center, Department
of Chemistry & Biochemistry, Veterinary Biosciences, College of Veterinary
Medicine, and Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
10
|
Harshman SW, Chen MM, Branson OE, Jacob NK, Johnson AJ, Byrd JC, Freitas MA. Isolation and analysis of linker histones across cellular compartments. J Proteomics 2013; 91:595-604. [PMID: 24013129 DOI: 10.1016/j.jprot.2013.08.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 07/23/2013] [Accepted: 08/29/2013] [Indexed: 01/09/2023]
Abstract
UNLABELLED Analysis of histones, especially histone H1, is severely limited by immunological reagent availability. This paper describes the application of cellular fractionation with LC-MS for profiling histones in the cytosol and upon chromatin. First, we show that linker histones enriched by cellular fractionation gives less nuclear contamination and higher histone content than when prepared by nuclei isolation. Second, we profiled the soluble linker histones throughout the cell cycle revealing phosphorylation increases as cells reach mitosis. Finally, we monitored histone H1.2-H1.5 translocation to the cytosol in response to the CDK inhibitor flavopiridol in primary CLL cells treated ex vivo. Data shows that all H1 variants translocate in response to drug treatment with no specific order to their cytosolic appearance. The results illustrate the utility of cellular fractionation in conjunction with LC-MS for the analysis of histone H1 throughout the cell. BIOLOGICAL SIGNIFICANCE This paper demonstrates the first time application of cellular fractionation to characterize cytosolic histone H1 by liquid chromatography mass spectrometry (LC-MS). Using the Ramos Burkitt's lymphoma cell line, cellular fractionation was shown to give less nuclear contamination and higher histone content than preparations by nuclei isolation. Further application of the cellular fractionation approach was shown by using primary chronic lymphocytic leukemia (CLL) cells to monitor the movement of histone H1 across cellular compartments in response to the cyclin dependent kinase inhibitor flavopiridol. Collectively, these data establish a mass spectrometric method for exploration into the function of cytosolic histone H1.
Collapse
Affiliation(s)
- Sean W Harshman
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, OH, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | | | | | | | | | | | | |
Collapse
|
11
|
Singh R, Mortazavi A, Telu KH, Nagarajan P, Lucas DM, Thomas-Ahner JM, Clinton SK, Byrd JC, Freitas MA, Parthun MR. Increasing the complexity of chromatin: functionally distinct roles for replication-dependent histone H2A isoforms in cell proliferation and carcinogenesis. Nucleic Acids Res 2013; 41:9284-95. [PMID: 23956221 PMCID: PMC3814372 DOI: 10.1093/nar/gkt736] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Replication-dependent histones are encoded by multigene families found in several large clusters in the human genome and are thought to be functionally redundant. However, the abundance of specific replication-dependent isoforms of histone H2A is altered in patients with chronic lymphocytic leukemia. Similar changes in the abundance of H2A isoforms are also associated with the proliferation and tumorigenicity of bladder cancer cells. To determine whether these H2A isoforms can perform distinct functions, expression of several H2A isoforms was reduced by siRNA knockdown. Reduced expression of the HIST1H2AC locus leads to increased rates of cell proliferation and tumorigenicity. We also observe that regulation of replication-dependent histone H2A expression can occur on a gene-specific level. Specific replication-dependent histone H2A genes are either up- or downregulated in chronic lymphocytic leukemia tumor tissue samples. In addition, discreet elements are identified in the 5′ untranslated region of the HIST1H2AC locus that confer translational repression. Taken together, these results indicate that replication-dependent histone isoforms can possess distinct cellular functions and that regulation of these isoforms may play a role in carcinogenesis.
Collapse
Affiliation(s)
- Rajbir Singh
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH 43210, USA, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA, Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Harshman SW, Young NL, Parthun MR, Freitas MA. H1 histones: current perspectives and challenges. Nucleic Acids Res 2013; 41:9593-609. [PMID: 23945933 PMCID: PMC3834806 DOI: 10.1093/nar/gkt700] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
H1 and related linker histones are important both for maintenance of higher-order chromatin structure and for the regulation of gene expression. The biology of the linker histones is complex, as they are evolutionarily variable, exist in multiple isoforms and undergo a large variety of posttranslational modifications in their long, unstructured, NH2- and COOH-terminal tails. We review recent progress in understanding the structure, genetics and posttranslational modifications of linker histones, with an emphasis on the dynamic interactions of these proteins with DNA and transcriptional regulators. We also discuss various experimental challenges to the study of H1 and related proteins, including limitations of immunological reagents and practical difficulties in the analysis of posttranslational modifications by mass spectrometry.
Collapse
Affiliation(s)
- Sean W Harshman
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, Ohio, USA, College of Medicine and Arthur G. James Comprehensive Cancer Center, Columbus, Ohio, USA, National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL, USA and Molecular and Cellular Biochemistry, The Ohio State University, Columbus, Ohio, USA
| | | | | | | |
Collapse
|
13
|
Telu KH, Abbaoui B, Thomas-Ahner JM, Zynger DL, Clinton SK, Freitas MA, Mortazavi A. Alterations of histone H1 phosphorylation during bladder carcinogenesis. J Proteome Res 2013; 12:3317-26. [PMID: 23675690 DOI: 10.1021/pr400143x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
There is a crucial need for development of prognostic and predictive biomarkers in human bladder carcinogenesis in order to personalize preventive and therapeutic strategies and improve outcomes. Epigenetic alterations, such as histone modifications, are implicated in the genetic dysregulation that is fundamental to carcinogenesis. Here we focus on profiling the histone modifications during the progression of bladder cancer. Histones were extracted from normal human bladder epithelial cells, an immortalized human bladder epithelial cell line (hTERT), and four human bladder cancer cell lines (RT4, J82, T24, and UMUC3) ranging from superficial low-grade to invasive high-grade cancers. Liquid chromatography-mass spectrometry (LC-MS) profiling revealed a statistically significant increase in phosphorylation of H1 linker histones from normal human bladder epithelial cells to low-grade superficial to high-grade invasive bladder cancer cells. This finding was further validated by immunohistochemical staining of the normal epithelium and transitional cell cancer from human bladders. Cell cycle analysis of histone H1 phosphorylation by Western blotting showed an increase of phosphorylation from G0/G1 phase to M phase, again supporting this as a proliferative marker. Changes in histone H1 phosphorylation status may further clarify epigenetic changes during bladder carcinogenesis and provide diagnostic and prognostic biomarkers or targets for future therapeutic interventions.
Collapse
Affiliation(s)
- Kelly H Telu
- Department of Chemistry, College of Arts and Sciences, The Ohio State University and the Comprehensive Cancer Center, Columbus, Ohio 43210, USA
| | | | | | | | | | | | | |
Collapse
|