1
|
Pan Y, Peng M, Tong M, He Y, Hao M, Gao HL, Lao Y, Xue J, Liu M, Zhong Q, Liu X, Li B. The globular domain of extracellular histones mediates cytotoxicity via membrane disruption mechanism. J Biol Chem 2025; 301:108038. [PMID: 39615681 PMCID: PMC11732447 DOI: 10.1016/j.jbc.2024.108038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/19/2024] [Accepted: 11/23/2024] [Indexed: 12/28/2024] Open
Abstract
Histones are traditionally recognized for structuring nuclear architecture and regulating gene expression. Recent advances have revealed their roles in inflammation, coagulation, and immune responses, where they act as damage-associated molecular patterns. The mechanisms by which histones induce membrane leakage are not well understood, and certain cells, including endothelial cells and peritoneal macrophages, show resistance to histone-mediated pore formation. We utilized liposome leakage assays to explore the pore-forming capabilities of different histone configurations, including individual histones, tail regions, and globular domains. Our results demonstrate that globular domains primarily drive pore formation. Using cytotoxicity assays, we further demonstrate that the globular domain of extracellular histones is primarily implicated in inducing lytic cell death. This study provides insights into the pathological roles of histones and suggests potential therapeutic targets to mitigate their harmful effects.
Collapse
Affiliation(s)
- Yixuan Pan
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengyuan Peng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mindan Tong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue He
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Hao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - He Lilian Gao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yimin Lao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingdong Xue
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meiyang Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Zhong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxia Liu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Bing Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Hendrickson-Rebizant T, Sudhakar SRN, Rowley MJ, Frankel A, Davie JR, Lakowski TM. Structure, Function, and Activity of Small Molecule and Peptide Inhibitors of Protein Arginine Methyltransferase 1. J Med Chem 2024; 67:15931-15946. [PMID: 39250434 PMCID: PMC11440505 DOI: 10.1021/acs.jmedchem.4c00490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/12/2024] [Accepted: 07/25/2024] [Indexed: 09/11/2024]
Abstract
Protein arginine N-methyltransferases (PRMT) are a family of S-adenosyl-l-methionine (SAM)-dependent enzymes that transfer methyl-groups to the ω-N of arginyl residues in proteins. PRMTs are involved in regulating gene expression, RNA splicing, and other activities. PRMT1 is responsible for most cellular arginine methylation, and its dysregulation is involved in many cancers. Accordingly, many groups have targeted PRMT1 using small molecules and peptide inhibitors. In this Perspective, we discuss the structure and function of selected peptide and small molecule inhibitors of PRMT1. We examine inhibitors that target the substrate arginyl peptide, SAM, or both binding sites, and the type of inhibition that results. Small molecules, and peptides that are bisubstrate, and/or PRMT transition state mimic inhibitors as well as inhibitors that alkylate PRMTs will be discussed. We define a structure-activity relationship for the aromatic/heteroaromatic N-methylethylenediamine inhibitors of PRMT1 and review current progress of PRMT1 inhibitors in clinical trials.
Collapse
Affiliation(s)
- Thordur Hendrickson-Rebizant
- Pharmaceutical
analysis Laboratory, College of Pharmacy, University of Manitoba, 750 McDermot Avenue West, Winnipeg, Manitoba R3E 0T5, Canada
- Paul
Albrechtsen Research Institute, CancerCare
Manitoba, Winnipeg, MB R3E 0 V9, Canada
| | - Sadhana R. N. Sudhakar
- Paul
Albrechtsen Research Institute, CancerCare
Manitoba, Winnipeg, MB R3E 0 V9, Canada
- Department
of Biochemistry and Medical Genetics, University
of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Michael J. Rowley
- Faculty
of Pharmaceutical Sciences, The University
of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3, Canada
| | - Adam Frankel
- Faculty
of Pharmaceutical Sciences, The University
of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3, Canada
| | - James R. Davie
- Paul
Albrechtsen Research Institute, CancerCare
Manitoba, Winnipeg, MB R3E 0 V9, Canada
- Department
of Biochemistry and Medical Genetics, University
of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Ted M. Lakowski
- Pharmaceutical
analysis Laboratory, College of Pharmacy, University of Manitoba, 750 McDermot Avenue West, Winnipeg, Manitoba R3E 0T5, Canada
- Paul
Albrechtsen Research Institute, CancerCare
Manitoba, Winnipeg, MB R3E 0 V9, Canada
| |
Collapse
|
3
|
Xu Q, Zhu F, Pan Y, Ren Y, Li J, Huang N, Liu K, Wang Y. HIV Tat-Conjugated Histone H3 Peptides Induce Tumor Cell Death Via Cellular Stress Responses. Hum Gene Ther 2023; 34:42-55. [PMID: 36373826 DOI: 10.1089/hum.2022.165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Histone H3 is a nucleosome scaffold protein that is involved in a variety of intracellular processes. Aberrant modification of H3 is important in carcinogenesis. In contrast, free histones in cells can act as stimuli to trigger cellular immune responses and cell death. In this study, we linked cell-penetrating peptide HIV Tat to a histone H3 fragment to achieve intracellular delivery in tumor cells. We found that Tat-conjugated histone polypeptides localized to nuclei of lung and breast cancer cells and caused cell death. A trans-configured Tat sequence displayed dramatically improved peptide half-life and cytotoxicity. Mechanistic studies demonstrated that treatment with the peptides significantly elevated mitogen-activated protein kinase (MAPK) signaling, reactive oxygen species (ROS) production, as well as levels of stress-inducible transcription factor ATF3 (activating transcription factor 3) and AP-1 (activating protein-1). Cytotoxicity of the peptide was significantly reduced by inhibition of AP-1 activity and ROS production. These results suggest the potential of Tat-conjugated H3 peptides as antitumor agents to induce cell death via increased cellular stress response by activating p38-MAPK signaling and intracellular ROS production.
Collapse
Affiliation(s)
- Qian Xu
- Department of Pathophysiology, West China College of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Feimei Zhu
- Department of Pathophysiology, West China College of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yixuan Pan
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanlin Ren
- Department of Pathophysiology, West China College of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Jingyu Li
- Department of Pathophysiology, West China College of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Ning Huang
- Department of Pathophysiology, West China College of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Keyun Liu
- Department of Physiology, School of Medicine, Hubei Minzu University, Enshi, China
| | - Yi Wang
- Department of Pathophysiology, West China College of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Nakatsuka T, Tateishi K, Kato H, Fujiwara H, Yamamoto K, Kudo Y, Nakagawa H, Tanaka Y, Ijichi H, Ikenoue T, Ishizawa T, Hasegawa K, Tachibana M, Shinkai Y, Koike K. Inhibition of histone methyltransferase G9a attenuates liver cancer initiation by sensitizing DNA-damaged hepatocytes to p53-induced apoptosis. Cell Death Dis 2021; 12:99. [PMID: 33468997 PMCID: PMC7815717 DOI: 10.1038/s41419-020-03381-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023]
Abstract
While the significance of acquired genetic abnormalities in the initiation of hepatocellular carcinoma (HCC) has been established, the role of epigenetic modification remains unknown. Here we identified the pivotal role of histone methyltransferase G9a in the DNA damage-triggered initiation of HCC. Using liver-specific G9a-deficient (G9aΔHep) mice, we revealed that loss of G9a significantly attenuated liver tumor initiation caused by diethylnitrosamine (DEN). In addition, pharmacological inhibition of G9a attenuated the DEN-induced initiation of HCC. After treatment with DEN, while the induction of γH2AX and p53 were comparable in the G9aΔHep and wild-type livers, more apoptotic hepatocytes were detected in the G9aΔHep liver. Transcriptome analysis identified Bcl-G, a pro-apoptotic Bcl-2 family member, to be markedly upregulated in the G9aΔHep liver. In human cultured hepatoma cells, a G9a inhibitor, UNC0638, upregulated BCL-G expression and enhanced the apoptotic response after treatment with hydrogen peroxide or irradiation, suggesting an essential role of the G9a-Bcl-G axis in DNA damage response in hepatocytes. The proposed mechanism was that DNA damage stimuli recruited G9a to the p53-responsive element of the Bcl-G gene, resulting in the impaired enrichment of p53 to the region and the attenuation of Bcl-G expression. G9a deletion allowed the recruitment of p53 and upregulated Bcl-G expression. These results demonstrate that G9a allows DNA-damaged hepatocytes to escape p53-induced apoptosis by silencing Bcl-G, which may contribute to the tumor initiation. Therefore, G9a inhibition can be a novel preventive strategy for HCC.
Collapse
Affiliation(s)
- Takuma Nakatsuka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Keisuke Tateishi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Hiroyuki Kato
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hiroaki Fujiwara
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Division of Gastroenterology, The Institute for Adult Diseases, Asahi Life Foundation, 2-2-6 Bakurocho, Chuo-ku, Tokyo, 103-0002, Japan
| | - Keisuke Yamamoto
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yotaro Kudo
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hayato Nakagawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yasuo Tanaka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hideaki Ijichi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tsuneo Ikenoue
- Division of Clinical Genome Research, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Takeaki Ishizawa
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kiyoshi Hasegawa
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Makoto Tachibana
- Laboratory of Epigenome Dynamics, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, 565-0871, Japan
| | - Yoichi Shinkai
- Cellular Memory Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
5
|
Teles K, Fernandes V, Silva I, Leite M, Grisolia C, Lobbia VR, van Ingen H, Honorato R, Lopes-de-Oliveira P, Treptow W, Santos G. Nucleosome binding peptide presents laudable biophysical and in vivo effects. Biomed Pharmacother 2019; 121:109678. [PMID: 31810135 DOI: 10.1016/j.biopha.2019.109678] [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] [Received: 09/27/2019] [Revised: 10/30/2019] [Accepted: 11/13/2019] [Indexed: 10/25/2022] Open
Abstract
Chromatin state is highly dependent on the nucleosome binding proteins. Herein, we used a multipronged approach employing biophysical and in vivo experiments to characterize the effects of Nucleosome Binding Peptides (NBPeps) on nucleosome and cell activity. We performed a series of structure-based calculations on the nucleosome surface interaction with GMIP1 (a novel NBPep generated in silico), and HMGN2 (nucleosome binding motif of HMGN2), which contains sites that bind DNA and the acid patch, and also LANA and H4pep (nucleosome binding motif of H4 histone tail) that only bind to the acidic patch. Biochemical assays shows that H4pep, but not HMGN2, GMIP1 and LANA, is highly specific for targeting the nucleosome, with important effects on the final nucleosome structure and robust in vivo effects. These findings suggest that NBPeps might have important therapeutic implications and relevance as tools for chromatin investigation.
Collapse
Affiliation(s)
- Kaian Teles
- Laboratório de Farmacologia Molecular, Departamento de Farmácia, Universidade de Brasília, Brasília, 70919-970, Brazil
| | - Vinicius Fernandes
- Laboratório de Farmacologia Molecular, Departamento de Farmácia, Universidade de Brasília, Brasília, 70919-970, Brazil; Laboratório de Biologia Teórica e Computacional, Departamento de Biologia Celular, Universidade de Brasília, DF, 70910-900, Brasília, Brazil
| | - Isabel Silva
- Laboratório de Farmacologia Molecular, Departamento de Farmácia, Universidade de Brasília, Brasília, 70919-970, Brazil
| | - Manuela Leite
- Laboratório de Farmacologia Molecular, Departamento de Farmácia, Universidade de Brasília, Brasília, 70919-970, Brazil
| | - Cesar Grisolia
- Laboratório de Genética Toxicológica, Departamento de Genética e Morfologia, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| | - Vincenzo R Lobbia
- NMR Spectroscopy Group, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH, Utrecht, the Netherlands
| | - Hugo van Ingen
- NMR Spectroscopy Group, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH, Utrecht, the Netherlands
| | - Rodrigo Honorato
- Laboratório Nacional de Biociências (LNBio), Campinas, SP, Brazil
| | | | - Werner Treptow
- Laboratório de Biologia Teórica e Computacional, Departamento de Biologia Celular, Universidade de Brasília, DF, 70910-900, Brasília, Brazil
| | - Guilherme Santos
- Laboratório de Farmacologia Molecular, Departamento de Farmácia, Universidade de Brasília, Brasília, 70919-970, Brazil.
| |
Collapse
|
6
|
Chen G, Yu X, Zhang M, Zheng A, Wang Z, Zuo Y, Liang Q, Jiang D, Chen Y, Zhao L, Jiang L, Li D, Liao S. Inhibition of Euchromatic Histone Lysine Methyltransferase 2 (EHMT2) Suppresses the Proliferation and Invasion of Cervical Cancer Cells. Cytogenet Genome Res 2019; 158:205-212. [DOI: 10.1159/000502072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2019] [Indexed: 12/18/2022] Open
Abstract
EHMT2 (euchromatic histone lysine methyltransferase 2), a histone methyltransferase, has been shown to be involved in multiple human cancers. In this study, we determined mRNA and protein expression of EHMT2 in cervical cancer cells and normal cervical epithelial cells. EHMT2 was inhibited with short hairpin RNA (shEHMT2) in cervical cancer cells. Cell viability, colony proliferation, apoptosis, adhesion, and invasion assays and Western blot were performed to assess the function of EHMT2. As a result, EHMT2 was upregulated in human cervical cancer cells compared to normal cervical epithelial cells. Suppression of EHMT2 expression impairs cell proliferation and induces apoptosis. Furthermore, EHMT2 silencing inhibited cell adhesion and invasion. Finally, knockdown of EHMT2 resulted in a reduction of the expression of the tumorigenic proteins Bcl-2, Mcl-1, and Survivin and in an increase in the expression of the anti-malignant protein E-cadherin. In conclusion, our data suggest that EHMT2 plays a key role in cell proliferation and metastatic capacity in cervical cancer cells and could serve as a potential therapeutic target.
Collapse
|
7
|
Lee PY, Park BC, Chi SW, Bae KH, Kim S, Cho S, Kang S, Kim JH, Park SG. Histone H4 is cleaved by granzyme A during staurosporine-induced cell death in B-lymphoid Raji cells. BMB Rep 2017; 49:560-565. [PMID: 27439606 PMCID: PMC5227298 DOI: 10.5483/bmbrep.2016.49.10.105] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Indexed: 01/06/2023] Open
Abstract
Granzyme A (GzmA) was first identified as a cytotoxic T lymphocyte protease protein with limited tissue expression. A number of cellular proteins are known to be cleaved by GzmA, and its function is to induce apoptosis. Histones H1, H2B, and H3 were identified as GzmA substrates during apoptotic cell death. Here, we demonstrated that histone H4 was cleaved by GzmA during staurosporine-induced cell death; however, in the presence of caspase inhibitors, staurosporine-treated Raji cells underwent necroptosis instead of apoptosis. Furthermore, histone H4 cleavage was blocked by the GzmA inhibitor nafamostat mesylate and by GzmA knockdown using siRNA. These results suggest that histone H4 is a novel substrate for GzmA in staurosporine-induced cells. [BMB Reports 2016; 49(10): 560-565]
Collapse
Affiliation(s)
- Phil Young Lee
- Disease Target Structure Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141; Division of Life Sciences, Korea University, Seoul 02841, Korea
| | - Byoung Chul Park
- Disease Target Structure Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Seung Wook Chi
- Disease Target Structure Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Sunhong Kim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Sayeon Cho
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea University, Seoul 02841, Korea
| | - Seongman Kang
- Division of Life Sciences, Korea University, Seoul 02841, Korea
| | - Jeong-Hoon Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Sung Goo Park
- Disease Target Structure Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Korea
| |
Collapse
|
8
|
Li M, Liu C, Yang L, Zhang L, Chen C, He M, Lu Y, Feng W, Pi H, Zhang Y, Zhong M, Yu Z, Zhou Z. G9a-mediated histone methylation regulates cadmium-induced male fertility damage in pubertal mice. Toxicol Lett 2016; 252:11-21. [DOI: 10.1016/j.toxlet.2016.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 03/18/2016] [Accepted: 04/05/2016] [Indexed: 10/22/2022]
|
9
|
Zhao R, Chen K, Cao J, Yu H, Tian L, Liu M. A correlation analysis between HDAC1 over-expression and clinical features of laryngeal squamous cell carcinoma. Acta Otolaryngol 2015; 136:172-6. [PMID: 26588579 DOI: 10.3109/00016489.2015.1101781] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
CONCLUSION HDAC1 may be a prognostic biomarker for LSCC malignant potency and a potent factor resulting in decreased sensitivity of LSCC in radiotherapy. OBJECTIVE The aim of this study was to evaluate the correlation between histone deacetylase 1 (HDAC1) over-expression and clinical features in laryngeal squamous cell carcinoma (LSCC). METHODS This study assessed the HDAC1 expressions in 90 formalin-fixed paraffin-embedded LSCC samples, 30 adjacent non-neoplastic tissues, and 30 laryngeal polyp tissues by immunohistochemistry (IHC). In addition, another 40 LSCC samples were collected that were divided into two groups after 3-month radiotherapy: the high radio-sensitive group (HRS) and low radio-sensitive group (LRS). Overall survival curves of all the LSCC patients were constructed by Kaplan-Meier method and long-rank test. RESULTS All ninety samples were positively immunostained for HDAC1. The expression of HDAC1 was up-regulated and significantly associated with T classification, lymph node metastases, tumor location and clinical stage. HDAC1 was mainly labeled in the epithelial cells of laryngeal polyp tissues and adjacent non-neoplastic tissues. In addition, the expression of HDAC1 was significantly higher in LRS than that in HRS. The positive rates for stage III-IV tumor were significantly higher than those for stage II. LSCC patients with HDAC1 over-expression and LRS presented a shorter 5-year overall survival rate.
Collapse
Affiliation(s)
- Rui Zhao
- a Department of Otorhinolaryngology, Head and Neck Surgery , The Second Affiliated Hospital of Harbin Medical University , Harbin , PR China
| | - Kexin Chen
- a Department of Otorhinolaryngology, Head and Neck Surgery , The Second Affiliated Hospital of Harbin Medical University , Harbin , PR China
| | - Jing Cao
- a Department of Otorhinolaryngology, Head and Neck Surgery , The Second Affiliated Hospital of Harbin Medical University , Harbin , PR China
| | - Hao Yu
- a Department of Otorhinolaryngology, Head and Neck Surgery , The Second Affiliated Hospital of Harbin Medical University , Harbin , PR China
| | - Linli Tian
- a Department of Otorhinolaryngology, Head and Neck Surgery , The Second Affiliated Hospital of Harbin Medical University , Harbin , PR China
| | - Ming Liu
- a Department of Otorhinolaryngology, Head and Neck Surgery , The Second Affiliated Hospital of Harbin Medical University , Harbin , PR China
| |
Collapse
|
10
|
Howard CJ, Yu RR, Gardner ML, Shimko JC, Ottesen JJ. Chemical and biological tools for the preparation of modified histone proteins. Top Curr Chem (Cham) 2015; 363:193-226. [PMID: 25863817 DOI: 10.1007/128_2015_629] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Eukaryotic chromatin is a complex and dynamic system in which the DNA double helix is organized and protected by interactions with histone proteins. This system is regulated through a large network of dynamic post-translational modifications (PTMs) which ensure proper gene transcription, DNA repair, and other processes involving DNA. Homogenous protein samples with precisely characterized modification sites are necessary to understand better the functions of modified histone proteins. Here, we discuss sets of chemical and biological tools developed for the preparation of modified histones, with a focus on the appropriate choice of tool for a given target. We start with genetic approaches for the creation of modified histones, including the incorporation of genetic mimics of histone modifications, chemical installation of modification analogs, and the use of the expanded genetic code to incorporate modified amino acids. We also cover the chemical ligation techniques which have been invaluable in the generation of complex modified histones indistinguishable from their natural counterparts. We end with a prospectus on future directions.
Collapse
Affiliation(s)
- Cecil J Howard
- Department of Chemistry and Biochemistry and The Ohio State Biochemistry Program, The Ohio State University, Columbus, OH, 43210, USA
| | | | | | | | | |
Collapse
|
11
|
Paul S, Giri AK. Epimutagenesis: A prospective mechanism to remediate arsenic-induced toxicity. ENVIRONMENT INTERNATIONAL 2015; 81:8-17. [PMID: 25898228 DOI: 10.1016/j.envint.2015.04.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 03/30/2015] [Accepted: 04/10/2015] [Indexed: 06/04/2023]
Abstract
Arsenic toxicity is a global issue, addressed by the World Health Organization as one of the major natural calamities faced by humans. More than 137 million individuals in 70 nations are affected by arsenic mainly through drinking water and also through diet. Chronic arsenic exposure leads to various types of patho-physiological end points in humans including cancers. Arsenic, a xenobiotic substance, is biotransformed in the body to its methylated species by using the physiological S-adenosyl methionine (SAM). SAM dictates methylation status of the genome and arsenic metabolism leads to depletion of SAM leading to an epigenetic disequilibrium. Since epigenetics is one of the major phenomenon at the interface between the environment and human health impact, its disequilibrium by arsenic inflicts upon the chromatin compaction, gene expression, genomic stability and a host of biomolecular interactions, the interactome within the cell. Since arsenic is not mutagenic but is carcinogenic in nature, arsenic induced epimutagenesis has come to the forefront since it determines the transcriptional and genomic integrity of the cell. Arsenic toxicity brings forth several pathophysiological manifestations like dermatological non-cancerous, pre-cancerous and cancerous lesions, peripheral neuropathy, DNA damage, respiratory disorders and cancers of several internal organs. Recently, several diseases of similar manifestations have been explained with the relevant epigenetic perspectives regarding the possible molecular mechanism for their onset. Hence, in the current review, we comprehensively try to intercalate the information on arsenic-induced epigenetic alterations of DNA, histones and microRNA so as to understand whether the arsenic-induced toxic manifestations are brought about by the epigenetic changes. We highlight the need to understand the aspect of epimutagenesis and subsequent alterations in the cellular interactome due to arsenic-induced molecular changes, which may be utilized to develop putative therapeutic strategies targeting both oxidative potential and epimutagenesis in humans.
Collapse
Affiliation(s)
- Somnath Paul
- Molecular and Human Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Ashok K Giri
- Molecular and Human Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India.
| |
Collapse
|
12
|
Silva ITGD, Oliveira PSLD, Santos GM. Featuring the nucleosome surface as a therapeutic target. Trends Pharmacol Sci 2015; 36:263-9. [DOI: 10.1016/j.tips.2015.02.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/20/2015] [Accepted: 02/27/2015] [Indexed: 01/18/2023]
|
13
|
de Figueiredo IR, Freire JM, Flores L, Veiga AS, Castanho MARB. Cell-penetrating peptides: A tool for effective delivery in gene-targeted therapies. IUBMB Life 2014; 66:182-194. [PMID: 24659560 DOI: 10.1002/iub.1257] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 03/06/2014] [Indexed: 12/24/2022]
Abstract
The current landscapes of novel therapeutic approaches rely mostly on gene-targeted technologies, enabling to fight rare genomic diseases, from infections to cancer and hereditary diseases. Although, reaching the action-site for this novel treatments requires to deliver nucleic acids, or other macromolecules into cells, which may pose difficult tasks to pharmaceutical companies. To overcome this technological limitation, a wide variety of vectors have been developed in the past decades and have proven to be successful in delivering various therapeutics. Cell-penetrating peptides (CPP) have been one of the technologies widely studied and have been increasingly used to transport small RNA/DNA, plasmids, antibodies, and nanoparticles into cells. Despite the already proved huge potential that these peptide-based approaches may suggest, few advances have been put to pharmacological or clinical use. This review will describe the origin, development, and usage of CPP to deliver therapeutic agents into cells, with special emphasis on their current application to gene-therapies. Specifically, we will describe the current trials being conducted to treat cancer, gene disorders, and autoimmune diseases using CPP-based therapies. © 2014 IUBMB Life, 66(3):182-194, 2014.
Collapse
Affiliation(s)
- Inês Rego de Figueiredo
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - João Miguel Freire
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Luís Flores
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Salomé Veiga
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Miguel A R B Castanho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
14
|
Kim K, Kim JM, Kim JS, Choi J, Lee YS, Neamati N, Song JS, Heo K, An W. VprBP has intrinsic kinase activity targeting histone H2A and represses gene transcription. Mol Cell 2013; 52:459-67. [PMID: 24140421 DOI: 10.1016/j.molcel.2013.09.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 06/26/2013] [Accepted: 09/11/2013] [Indexed: 10/26/2022]
Abstract
Histone modifications play important roles in the regulation of gene expression and chromatin organization. VprBP has been implicated in transcriptionally silent chromatin formation and cell-cycle regulation, but the molecular basis underlying such effects remains unclear. Here we report that VprBP possesses an intrinsic protein kinase activity and is capable of phosphorylating histone H2A on threonine 120 (H2AT120p) in a nucleosomal context. VprBP is localized to a large set of tumor suppressor genes and blocks their transcription, in a manner that is dependent on its kinase activity toward H2AT120. The functional significance of VprBP-mediated H2AT120p is further underscored by the fact that RNAi knockdown and small-molecule inhibition of VprBP reactivate growth regulatory genes and impede tumor growth. Our findings establish VprBP as a major kinase responsible for H2AT120p in cancer cells and suggest that VprBP inhibition could be a new strategy for the development of anticancer therapeutics.
Collapse
Affiliation(s)
- Kyunghwan Kim
- Department of Biochemistry and Molecular Biology, University of Southern California Keck School of Medicine, Los Angeles, CA 90089, USA; Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA 90089, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Kim K, Punj V, Choi J, Heo K, Kim JM, Laird PW, An W. Gene dysregulation by histone variant H2A.Z in bladder cancer. Epigenetics Chromatin 2013; 6:34. [PMID: 24279307 PMCID: PMC3853418 DOI: 10.1186/1756-8935-6-34] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 09/27/2013] [Indexed: 12/20/2022] Open
Abstract
Background The incorporation of histone variants into nucleosomes is one of the main strategies that the cell uses to regulate the structure and function of chromatin. Histone H2A.Z is an evolutionarily conserved histone H2A variant that is preferentially localized within nucleosomes at the transcriptional start site (TSS). H2A.Z reorganizes the local chromatin structure and recruits the transcriptional machinery for gene activation. High expression of H2A.Z has been reported in several types of cancers and is causally linked to genomic instability and tumorigenesis. However, it is not entirely clear how H2A.Z overexpression in cancer cells establishes aberrant chromatin states and promotes gene expression. Results Through integration of genome-wide H2A.Z ChIP-seq data with microarray data, we demonstrate that H2A.Z is enriched around the TSS of cell cycle regulatory genes in bladder cancer cells, and this enrichment is correlated with the elevated expression of cancer-promoting genes. RNAi-mediated knockdown of H2A.Z in the cancer cells causes transcriptional suppression of multiple cell cycle regulatory genes with a distinct decrease in cell proliferation. H2A.Z nucleosomes around the TSS have higher levels of H3K4me2/me3, which coincides with the recruitment of two chromatin factors, WDR5 and BPTF. The observed recruitment is functional, as the active states of H2A.Z target genes are largely erased by suppressing the expression of WDR5 or BPTF, effects resembling H2A.Z knockdown. Conclusions We conclude that H2A.Z is overexpressed in bladder cancer cells and contributes to cancer-related transcription pathways. We also provide evidence in support of the engagement of H3K4me2/me3 and WDR5/BPTF in H2A.Z-induced cancer pathogenesis. Further studies are warranted to understand how H2A.Z overexpression contributes to the recruitment of the full repertoire of transcription machinery to target genes in bladder cancer cells.
Collapse
|
16
|
Pirouz M, Pilarski S, Kessel M. A critical function of Mad2l2 in primordial germ cell development of mice. PLoS Genet 2013; 9:e1003712. [PMID: 24009519 PMCID: PMC3757036 DOI: 10.1371/journal.pgen.1003712] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 06/25/2013] [Indexed: 01/13/2023] Open
Abstract
The development of primordial germ cells (PGCs) involves several waves of epigenetic reprogramming. A major step is following specification and involves the transition from the stably suppressive histone modification H3K9me2 to the more flexible, still repressive H3K27me3, while PGCs are arrested in G2 phase of their cycle. The significance and underlying molecular mechanism of this transition were so far unknown. Here, we generated mutant mice for the Mad2l2 (Mad2B, Rev7) gene product, and found that they are infertile in both males and females. We demonstrated that Mad2l2 is essential for PGC, but not somatic development. PGCs were specified normally in Mad2l2−/− embryos, but became eliminated by apoptosis during the subsequent phase of epigenetic reprogramming. A majority of knockout PGCs failed to arrest in the G2 phase, and did not switch from a H3K9me2 to a H3K27me3 configuration. By the analysis of transfected fibroblasts we found that the interaction of Mad2l2 with the histone methyltransferases G9a and GLP lead to a downregulation of H3K9me2. The inhibitory binding of Mad2l2 to Cyclin dependent kinase 1 (Cdk1) could arrest the cell cycle in the G2 phase, and also allowed another histone methyltransferase, Ezh2, to upregulate H3K27me3. Together, these results demonstrate the potential of Mad2l2 in the regulation of both cell cycle and the epigenetic status. The function of Mad2l2 is essential in PGCs, and thus of high relevance for fertility. Primordial germ cells (PGCs) are the origin of sperm and oocytes, and are responsible for transferring genetic information to the next generation faithfully. PGCs are first specified from pluripotent epiblast cells early in embryonic development. Second, they reprogram their epigenetic signature by changing histone modifications. This developmental event is specific to germ cells but not somatic cells. Although many players in the specification of PGCs are identified, only little is known about the genes essential for the regulation of the second phase. Here, we report that the Mad2l2 gene product plays an important role in the epigenetic reprogramming of PGCs. In wild type PGCs the cell cycle is arrested, and the methylation of histone 3 on residue K9 is replaced by methylation on K27. Our findings indicate that Mad2l2 is involved in this coordination of cell cycle and epigenetic reprogramming. The elucidation of this mechanism would help to identify the genetic basis of infertility.
Collapse
Affiliation(s)
- Mehdi Pirouz
- Department of Molecular Cell Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.
| | | | | |
Collapse
|