201
|
Bovio P, Roidl D, Heidrich S, Vogel T, Franz H. Isolation and Cultivation of Neural Progenitors Followed by Chromatin-Immunoprecipitation of Histone 3 Lysine 79 Dimethylation Mark. J Vis Exp 2018:56631. [PMID: 29443015 PMCID: PMC5908698 DOI: 10.3791/56631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Brain development is a complex process, which is controlled in a temporo-spatial manner by gradients of morphogens and different transcriptional programs. Additionally, epigenetic chromatin modifications, like histone methylation, have an important role for establishing and maintaining specific cell fates within this process. The vast majority of histone methylation occurs on the flexible histone tail, which is accessible to histone modifiers, erasers, and histone reader proteins. In contrast, H3K79 methylation is located in the globular domain of histone 3 and is implicated in different developmental functions. H3K79 methylation is evolutionarily conserved and can be found in a wide range of species from Homo sapiens to Saccharomyces cerevisiae. The modification occurs in different cell populations within organisms, including neural progenitors. The location of H3K79 methylation in the globular domain of histone 3 makes it difficult to assess. Here, we present methods to isolate and culture cortical progenitor cells (CPCs) from embryonic cortical brain tissue (E11.5-E14.5) or cerebellar granular neuron progenitors (CGNPs) from postnatal tissue (P5-P7), and to efficiently immunoprecipitate H3K79me2 for quantitative PCR (qPCR) and genome-wide sequencing.
Collapse
Affiliation(s)
- Patrick Bovio
- Institute for Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg; Faculty of Biology, University of Freiburg
| | - Deborah Roidl
- Institute for Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg
| | - Stefanie Heidrich
- Institute for Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg
| | - Tanja Vogel
- Institute for Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg;
| | - Henriette Franz
- Institute for Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg;
| |
Collapse
|
202
|
The histone demethylase LSD1 regulates inner ear progenitor differentiation through interactions with Pax2 and the NuRD repressor complex. PLoS One 2018; 13:e0191689. [PMID: 29370269 PMCID: PMC5784988 DOI: 10.1371/journal.pone.0191689] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/09/2018] [Indexed: 01/07/2023] Open
Abstract
The histone demethylase LSD1 plays a pivotal role in cellular differentiation, particularly in silencing lineage-specific genes. However, little is known about how LSD1 regulates neurosensory differentiation in the inner ear. Here we show that LSD1 interacts directly with the transcription factor Pax2 to form the NuRD co-repressor complex at the Pax2 target gene loci in a mouse otic neuronal progenitor cell line (VOT-N33). VOT-N33 cells expressing a Pax2-response element reporter were GFP-negative when untreated, but became GFP positive after forced differentiation or treatment with a potent LSD inhibitor. Pharmacological inhibition of LSD1 activity resulted in the enrichment of mono- and di-methylation of H3K4, upregulation of sensory neuronal genes and an increase in the number of sensory neurons in mouse inner ear organoids. Together, these results identify the LSD1/NuRD complex as a previously unrecognized modulator for Pax2-mediated neuronal differentiation in the inner ear.
Collapse
|
203
|
Janardhan A, Kathera C, Darsi A, Ali W, He L, Yang Y, Luo L, Guo Z. Prominent role of histone lysine demethylases in cancer epigenetics and therapy. Oncotarget 2018; 9:34429-34448. [PMID: 30344952 PMCID: PMC6188137 DOI: 10.18632/oncotarget.24319] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 12/04/2017] [Indexed: 12/14/2022] Open
Abstract
Protein methylation has an important role in the regulation of chromatin, gene expression and regulation. The protein methyl transferases are genetically altered in various human cancers. The enzymes that remove histone methylation have led to increased awareness of protein interactions as potential drug targets. Specifically, Lysine Specific Demethylases (LSD) removes methylated histone H3 lysine 4 (H3K4) and H3 lysine 9 (H3K9) through formaldehyde-generating oxidation. It has been reported that LSD1 and its downstream targets are involved in tumor-cell growth and metastasis. Functional studies of LSD1 indicate that it regulates activation and inhibition of gene transcription in the nucleus. Here we made a discussion about the summary of histone lysine demethylase and their functions in various human cancers.
Collapse
Affiliation(s)
- Avilala Janardhan
- The No. 7 People's Hospital of Changzhou, Changzhou, China.,Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Chandrasekhar Kathera
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Amrutha Darsi
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Wajid Ali
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Lingfeng He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yanhua Yang
- The No. 7 People's Hospital of Changzhou, Changzhou, China
| | - Libo Luo
- The No. 7 People's Hospital of Changzhou, Changzhou, China
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
204
|
Liu Q, Qu X, Xie X, He P, Huang S. Repression of Akt3 gene transcription by the tumor suppressor RIZ1. Sci Rep 2018; 8:1528. [PMID: 29367689 PMCID: PMC5784169 DOI: 10.1038/s41598-018-19943-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/10/2018] [Indexed: 12/04/2022] Open
Abstract
RIZ1 has been studied as a tumor suppressor and may play a role in metabolic diseases related to the Western style diet, such as cancer and obesity. The Akt pathway is known to play a role in both cancer and obesity, and a link between Akt and RIZ1 has also been found. To better understand the role of RIZ1 in obesity and cancer, we investigated how RIZ1 regulates the expression of Akt3. We found that overexpression of RIZ1 in HEK293 cells reduced the expression of Akt3 protein. Luciferase reporter activity of Akt3 gene promoter was significantly reduced in cells co-transfected with RIZ1. Recombinant proteins of RIZ1 was able to bind the Akt3 promoter in vitro, and chromatin immunoprecipitation assay also demonstrated the ability of RIZ1 binding to the Akt3 promoter in vivo. Overexpression of RIZ1 increased H3K9 methylation on the Akt3 promoter. These results identify Akt3 as a target of RIZ1 regulation and expand our understanding of the Akt pathway in cancer and obesity.
Collapse
Affiliation(s)
- Qingnan Liu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.,Department of Pathology, YiYang Medical College, Yiyang, Hunan, China
| | - Xiaotian Qu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Xiaolei Xie
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Pei He
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Shi Huang
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.
| |
Collapse
|
205
|
Zhu B, Chen S, Wang H, Yin C, Han C, Peng C, Liu Z, Wan L, Zhang X, Zhang J, Lian CG, Ma P, Xu ZX, Prince S, Wang T, Gao X, Shi Y, Liu D, Liu M, Wei W, Wei Z, Pan J, Wang Y, Xuan Z, Hess J, Hayward NK, Goding CR, Chen X, Zhou J, Cui R. The protective role of DOT1L in UV-induced melanomagenesis. Nat Commun 2018; 9:259. [PMID: 29343685 PMCID: PMC5772495 DOI: 10.1038/s41467-017-02687-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 12/13/2017] [Indexed: 11/09/2022] Open
Abstract
The DOT1L histone H3 lysine 79 (H3K79) methyltransferase plays an oncogenic role in MLL-rearranged leukemogenesis. Here, we demonstrate that, in contrast to MLL-rearranged leukemia, DOT1L plays a protective role in ultraviolet radiation (UVR)-induced melanoma development. Specifically, the DOT1L gene is located in a frequently deleted region and undergoes somatic mutation in human melanoma. Specific mutations functionally compromise DOT1L methyltransferase enzyme activity leading to reduced H3K79 methylation. Importantly, in the absence of DOT1L, UVR-induced DNA damage is inefficiently repaired, so that DOT1L loss promotes melanoma development in mice after exposure to UVR. Mechanistically, DOT1L facilitates DNA damage repair, with DOT1L-methylated H3K79 involvement in binding and recruiting XPC to the DNA damage site for nucleotide excision repair (NER). This study indicates that DOT1L plays a protective role in UVR-induced melanomagenesis.
Collapse
Affiliation(s)
- Bo Zhu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA.,Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Shuyang Chen
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA.,Department of Dermatology & China Hunan key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Hongshen Wang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA.,Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, China
| | - Chengqian Yin
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA.,Institute of Life Science, Jiangsu University, 212013, Zhenjiang, China
| | - Changpeng Han
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA.,Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, China
| | - Cong Peng
- Department of Dermatology & China Hunan key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Zhaoqian Liu
- Department of Dermatology & China Hunan key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Lixin Wan
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Xiaoyang Zhang
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Jie Zhang
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Christine G Lian
- Department of Pathology, The Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Ave, Boston, MA, 02115, USA
| | - Peilin Ma
- Department of Pathology, Indiana University School of Medicine, 340 West 10th Street, Fairbanks 6200, Indianapolis, IN, 46202, USA
| | - Zhi-Xiang Xu
- Division of Hematology/Oncology, Department of Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, AL, 35233, USA
| | - Sharon Prince
- Department of Human Biology, University of Cape Town, Rondebosch, Cape Town, 7700, South Africa
| | - Tao Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 300193, Tianjin, China
| | - Xiumei Gao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 300193, Tianjin, China
| | - Yujiang Shi
- Department of Medicine, Endocrinology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Dali Liu
- Department of Chemistry and Biochemistry, Loyola University Chicago, Chicago, IL, 60660, USA
| | - Min Liu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Zhi Wei
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Jingxuan Pan
- Cancer Pharmacology Research Institute, Jinan University, 510632, Guangzhou, China
| | - Yongjun Wang
- Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, China
| | - Zhenyu Xuan
- Department of Biological Sciences, Center for Systems Biology, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Jay Hess
- Department of Pathology, Indiana University School of Medicine, 340 West 10th Street, Fairbanks 6200, Indianapolis, IN, 46202, USA
| | - Nicholas K Hayward
- QIMR Berghofer Medical Research Institute, Brisbane City, QLD, 4006, Australia
| | - Colin R Goding
- Ludwig Institute for Cancer Research, University of Oxford, Headington, Oxford, OX3 7DQ, UK
| | - Xiang Chen
- Department of Dermatology & China Hunan key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, 410008, Changsha, China.
| | - Jun Zhou
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, 250014, Jinan, China.
| | - Rutao Cui
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA.
| |
Collapse
|
206
|
Skiles WM, Kester A, Pryor JH, Westhusin ME, Golding MC, Long CR. Oxygen-induced alterations in the expression of chromatin modifying enzymes and the transcriptional regulation of imprinted genes. Gene Expr Patterns 2018; 28:1-11. [PMID: 29339137 PMCID: PMC6094953 DOI: 10.1016/j.gep.2018.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/03/2017] [Accepted: 01/11/2018] [Indexed: 02/07/2023]
Abstract
Embryo culture and assisted reproductive technologies have been associated with a disproportionately high number of epigenetic abnormalities in the resulting offspring. However, the mechanisms by which these techniques influence the epigenome remain poorly defined. In this study, we evaluated the capacity of oxygen concentration to influence the transcriptional control of a selection of key enzymes regulating chromatin structure. In mouse embryonic stem cells, oxygen concentrations modulated the transcriptional regulation of the TET family of enzymes, as well as the de novo methyltransferase Dnmt3a. These transcriptional changes were associated with alterations in the control of multiple imprinted genes, including H19, Igf2, Igf2r, and Peg3. Similarly, exposure of in vitro produced bovine embryos to atmospheric oxygen concentrations was associated with disruptions in the transcriptional regulation of TET1, TET3, and DNMT3a, along with the DNA methyltransferase co-factor HELLS. In addition, exposure to high oxygen was associated with alterations in the abundance of transcripts encoding members of the Polycomb repressor complex (EED and EZH2), the histone methyltransferase SETDB1 and multiple histone demethylases (KDM1A, KDM4B, and KDM4C). These disruptions were accompanied by a reduction in embryo viability and suppression of the pluripotency genes NANOG and SOX2. These experiments demonstrate that oxygen has the capacity to modulate the transcriptional control of chromatin modifying genes involved in the establishment and maintenance of both pluripotency and genomic imprinting.
Collapse
Affiliation(s)
- William M Skiles
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843-4466, USA
| | - Avery Kester
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843-4466, USA
| | - Jane H Pryor
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843-4466, USA
| | - Mark E Westhusin
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843-4466, USA
| | - Michael C Golding
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843-4466, USA.
| | - Charles R Long
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843-4466, USA
| |
Collapse
|
207
|
Ateia IM, Sutthiboonyapan P, Kamarajan P, Jin T, Godovikova V, Kapila YL, Fenno JC. Treponema denticola increases MMP-2 expression and activation in the periodontium via reversible DNA and histone modifications. Cell Microbiol 2018; 20. [PMID: 29205773 DOI: 10.1111/cmi.12815] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/03/2017] [Accepted: 11/28/2017] [Indexed: 12/21/2022]
Abstract
Host-derived matrix metalloproteinases (MMPs) and bacterial proteases mediate destruction of extracellular matrices and supporting alveolar bone in periodontitis. The Treponema denticola dentilisin protease induces MMP-2 expression and activation in periodontal ligament (PDL) cells, and dentilisin-mediated activation of pro-MMP-2 is required for cellular fibronectin degradation. Here, we report that T. denticola regulates MMP-2 expression through epigenetic modifications in the periodontium. PDL cells were treated with epigenetic enzyme inhibitors before or after T. denticola challenge. Fibronectin fragmentation, MMP-2 expression, and activation were assessed by immunoblot, zymography, and qRT-PCR, respectively. Chromatin modification enzyme expression in T. denticola-challenged PDL cells and periodontal tissues were evaluated using gene arrays. Several classes of epigenetic enzymes showed significant alterations in transcription in diseased tissue and T. denticola-challenged PDL cells. T. denticola-mediated MMP-2 expression and activation were significantly reduced in PDL cells treated with inhibitors of aurora kinases and histone deacetylases. In contrast, DNA methyltransferase inhibitors had little effect, and inhibitors of histone acetyltransferases, methyltransferases, and demethylases exacerbated T. denticola-mediated MMP-2 expression and activation. Chronic epigenetic changes in periodontal tissues mediated by T. denticola or other oral microbes may contribute to the limited success of conventional treatment of chronic periodontitis and may be amenable to therapeutic reversal.
Collapse
Affiliation(s)
- Islam M Ateia
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Department of Periodontics and Oral Medicine, University of Mansoura Faculty of Dentistry, Mansoura, Egypt
| | - Pimchanok Sutthiboonyapan
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Department of Periodontology, Chulalongkorn University Faculty of Dentistry, Bangkok, Thailand
| | - Pachiyappan Kamarajan
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Department of Orofacial Sciences, University of California San Francisco School of Dentistry, San Francisco, CA, USA
| | - Taocong Jin
- Office of Research, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Valentina Godovikova
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Yvonne L Kapila
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Department of Orofacial Sciences, University of California San Francisco School of Dentistry, San Francisco, CA, USA
| | - J Christopher Fenno
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| |
Collapse
|
208
|
Suzuki S, Minami N. CHD1 Controls Cell Lineage Specification Through Zygotic Genome Activation. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2018; 229:15-30. [PMID: 29177762 DOI: 10.1007/978-3-319-63187-5_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In mammals, the processes spanning from fertilization to the generation of a new organism are very complex and are controlled by multiple genes. Life begins with the encounter of eggs and spermatozoa, in which gene expression is inactive prior to fertilization. After several cell divisions, cells arise that are specialized in implantation, a developmental process unique to mammals. Cells involved in the establishment and maintenance of implantation differentiate from totipotent embryos, and the remaining cells generate the embryo proper. Although this process of differentiation, termed cell lineage specification, is supported by various gene expression networks, many components have yet to be identified. Moreover, despite extensive research it remains unclear which genes are controlled by each of the factors involved. Although it has become clear that epigenetic factors regulate gene expression, elucidation of the underlying mechanisms remains challenging. In this chapter, we propose that the chromatin remodeling factor CHD1, together with epigenetic factors, is involved in a subset of gene expression networks involved in processes spanning from zygotic genome activation to cell lineage specification.
Collapse
Affiliation(s)
- Shinnosuke Suzuki
- Technology and Development Team for Mammalian Genome Dynamics, RIKEN BioResource Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Naojiro Minami
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8052, Japan.
| |
Collapse
|
209
|
Hosseini A, Minucci S. Alterations of Histone Modifications in Cancer. EPIGENETICS IN HUMAN DISEASE 2018:141-217. [DOI: 10.1016/b978-0-12-812215-0.00006-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
210
|
FIH Is an Oxygen Sensor in Ovarian Cancer for G9a/GLP-Driven Epigenetic Regulation of Metastasis-Related Genes. Cancer Res 2017; 78:1184-1199. [DOI: 10.1158/0008-5472.can-17-2506] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/01/2017] [Accepted: 12/15/2017] [Indexed: 11/16/2022]
|
211
|
Piao L, Nakakido M, Suzuki T, Dohmae N, Nakamura Y, Hamamoto R. Automethylation of SUV39H2, an oncogenic histone lysine methyltransferase, regulates its binding affinity to substrate proteins. Oncotarget 2017; 7:22846-56. [PMID: 26988914 PMCID: PMC5008405 DOI: 10.18632/oncotarget.8072] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 02/25/2016] [Indexed: 12/14/2022] Open
Abstract
We previously reported that the histone lysine methyltransferase SUV39H2, which is overexpressed in various types of human cancer, plays a critical role in the DNA repair after double strand breakage, and possesses oncogenic activity. Although its biological significance in tumorigenesis has been elucidated, the regulatory mechanism of SUV39H2 activity through post-translational modification is not well known. In this study, we demonstrate in vitro and in vivo automethylation of SUV39H2 at lysine 392. Automethylation of SUV39H2 led to impairment of its binding affinity to substrate proteins such as histone H3 and LSD1. Furthermore, we observed that hyper-automethylated SUV39H2 reduced methylation activities to substrates through affecting the binding affinity to substrate proteins. Our finding unveils a novel autoregulatory mechanism of SUV39H2 through lysine automethylation.
Collapse
Affiliation(s)
- Lianhua Piao
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Makoto Nakakido
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Takehiro Suzuki
- Biomolecular Characterization Unit, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Naoshi Dohmae
- Biomolecular Characterization Unit, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Yusuke Nakamura
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Ryuji Hamamoto
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA.,Division of Molecular Modification and Cancer Biology, National Cancer Center, Chuo-ku, Tokyo 104-0045, Japan
| |
Collapse
|
212
|
An K, Du F, Meng H, Li G, Zhang M, Liu Z, Zhao Z, Zhang Z, Yu D, Wang D, Yang C, Ma W, Yuan L, Zhou M, Duan L, Jin L, Li H, Zhang Y, Su J, Qiao J, Sun Y. Transgenerational analysis of H3K4me3 and H3K27me3 by ChIP-Seq links epigenetic inheritance to metabolism. J Genet Genomics 2017; 45:169-172. [PMID: 29574113 DOI: 10.1016/j.jgg.2017.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 11/16/2017] [Accepted: 11/28/2017] [Indexed: 01/20/2023]
Affiliation(s)
- Ke An
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fengxia Du
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hao Meng
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guochao Li
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Minjie Zhang
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zongzhi Liu
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zitong Zhao
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zilong Zhang
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Di Yu
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dong Wang
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Caiyun Yang
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Wencui Ma
- Heze Third People's Hospital, Heze 274000, China
| | - Lin Yuan
- Department of Nuclear Medicine, Fengxian Central Hospital, Shanghai 116044, China
| | | | - Lili Duan
- Weihai Municipal Hospital, Weihai 264200, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Hui Li
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Yan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Jianzhong Su
- College of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325035, China.
| | - Jie Qiao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Key Laboratory of Assisted, Beijing 100191, China.
| | - Yingli Sun
- Key Laboratory of Genomic and Precision Medicine, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
213
|
Morita Y, Ohno M, Nishi K, Hiraoka Y, Saijo S, Matsuda S, Kita T, Kimura T, Nishi E. Genome-wide profiling of nardilysin target genes reveals its role in epigenetic regulation and cell cycle progression. Sci Rep 2017; 7:14801. [PMID: 29093577 PMCID: PMC5665917 DOI: 10.1038/s41598-017-14942-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/18/2017] [Indexed: 11/28/2022] Open
Abstract
Post-translational histone modifications, such as acetylation and methylation, are prerequisites for transcriptional regulation. The metalloendopeptidase nardilysin (Nrdc) is a H3K4me2-binding protein that controls thermoregulation and β-cell functions through its transcriptional coregulator function. We herein combined high-throughput ChIP-seq and RNA-seq to achieve the first genome-wide identification of Nrdc target genes. A ChIP-seq analysis of immortalized mouse embryo fibroblasts (iMEF) identified 4053 Nrdc-binding sites, most of which were located in proximal promoter sites (2587 Nrdc-binding genes). Global H3K4me2 levels at Nrdc-binding promoters slightly increased, while H3K9ac levels decreased in the absence of Nrdc. Among Nrdc-binding genes, a comparative RNA-seq analysis identified 448 candidates for Nrdc target genes, among which cell cycle-related genes were significantly enriched. We confirmed decreased mRNA and H3K9ac levels at the promoters of individual genes in Nrdc-deficient iMEF, which were restored by the ectopic introduction of Nrdc. Reduced mRNA levels, but not H3K9ac levels were fully restored by the reintroduction of the peptidase-dead mutant of Nrdc. Furthermore, Nrdc promoted cell cycle progression at multiple stages, which enhanced cell proliferation in vivo. Collectively, our integrative studies emphasize the importance of Nrdc for maintaining a proper epigenetic status and cell growth.
Collapse
Affiliation(s)
- Yusuke Morita
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Mikiko Ohno
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.,Department of Pharmacology, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, 520-2192, Japan
| | - Kiyoto Nishi
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yoshinori Hiraoka
- Division of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, 650-8586, Japan
| | - Sayaka Saijo
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Shintaro Matsuda
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Toru Kita
- Kobe Home Medical and Nursing Care Promotion Foundation, 14-1 Naka Ichiriyama, Kami Aza, Shimotani, Yamada-cho, Kita-ku, Kobe, 651-1102, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Eiichiro Nishi
- Department of Pharmacology, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, 520-2192, Japan.
| |
Collapse
|
214
|
Shi X, Tasdogan A, Huang F, Hu Z, Morrison SJ, DeBerardinis RJ. The abundance of metabolites related to protein methylation correlates with the metastatic capacity of human melanoma xenografts. SCIENCE ADVANCES 2017; 3:eaao5268. [PMID: 29109980 PMCID: PMC5665593 DOI: 10.1126/sciadv.aao5268] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/06/2017] [Indexed: 05/27/2023]
Abstract
Metabolic reprogramming is a major factor in transformation, and particular metabolic phenotypes correlate with oncogenotype, tumor progression, and metastasis. By profiling metabolites in 17 patient-derived xenograft melanoma models, we identified durable metabolomic signatures that correlate with biological features of the tumors. BRAF mutant tumors had metabolomic and metabolic flux features of enhanced glycolysis compared to BRAF wild-type tumors. Tumors that metastasized efficiently from their primary sites had elevated levels of metabolites related to protein methylation, including trimethyllysine (TML). TML levels correlated with histone H3 trimethylation at Lys9 and Lys27, and methylation at these sites was also enhanced in efficiently metastasizing tumors. Erasing either of these marks by genetically or pharmacologically silencing the histone methyltransferase SETDB1 or EZH2 had no effect on primary tumor growth but reduced cellular invasiveness and metastatic spread. Thus, metabolite profiling can uncover targetable epigenetic requirements for the metastasis of human melanoma cells.
Collapse
Affiliation(s)
- Xiaolei Shi
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Alpaslan Tasdogan
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Fang Huang
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zeping Hu
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sean J. Morrison
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ralph J. DeBerardinis
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pediatrics and Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
215
|
Tvardovskiy A, Schwämmle V, Kempf SJ, Rogowska-Wrzesinska A, Jensen ON. Accumulation of histone variant H3.3 with age is associated with profound changes in the histone methylation landscape. Nucleic Acids Res 2017; 45:9272-9289. [PMID: 28934504 PMCID: PMC5766163 DOI: 10.1093/nar/gkx696] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 07/26/2017] [Indexed: 12/20/2022] Open
Abstract
Deposition of replication-independent histone variant H3.3 into chromatin is essential for many biological processes, including development and reproduction. Unlike replication-dependent H3.1/2 isoforms, H3.3 is expressed throughout the cell cycle and becomes enriched in postmitotic cells with age. However, lifelong dynamics of H3 variant replacement and the impact of this process on chromatin organization remain largely undefined. Using quantitative middle-down proteomics we demonstrate that H3.3 accumulates to near saturation levels in the chromatin of various mouse somatic tissues by late adulthood. Accumulation of H3.3 is associated with profound changes in global levels of both individual and combinatorial H3 methyl modifications. A subset of these modifications exhibit distinct relative abundances on H3 variants and remain stably enriched on H3.3 throughout the lifespan, suggesting a causal relationship between H3 variant replacement and age-dependent changes in H3 methylation. Furthermore, the H3.3 level is drastically reduced in human hepatocarcinoma cells as compared to nontumoral hepatocytes, suggesting the potential utility of the H3.3 relative abundance as a biomarker of abnormal cell proliferation activity. Overall, our study provides the first quantitative characterization of dynamic changes in H3 proteoforms throughout lifespan in mammals and suggests a role for H3 variant replacement in modulating H3 methylation landscape with age.
Collapse
Affiliation(s)
- Andrey Tvardovskiy
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark.,Center for Epigenetics, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Veit Schwämmle
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark.,Center for Epigenetics, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Stefan J Kempf
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Adelina Rogowska-Wrzesinska
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark.,Center for Epigenetics, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Ole N Jensen
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark.,Center for Epigenetics, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| |
Collapse
|
216
|
Analysis of the meiotic transcriptome reveals the genes related to the regulation of pollen abortion in cytoplasmic male-sterile pepper (Capsicum annuum L.). Gene 2017; 641:8-17. [PMID: 29031775 DOI: 10.1016/j.gene.2017.10.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 10/04/2017] [Accepted: 10/10/2017] [Indexed: 01/23/2023]
Abstract
CMS, which refers to the inability to generate functional pollen grains while still producing a normal gynoecium, has been widely used for pepper hybrid seed production. Pepper line 8214A is an excellent CMS line exhibiting 100% male sterility and superior economic characteristics. A TUNEL assay revealed the nuclear DNA is damaged in 8214A PMCs during meiosis. TEM images indicated that the 8214A PMCs exhibited asynchronous meiosis after prophase I, and some PMCs degraded prematurely with morphological features typical of PCD. Additionally, at the end of meiosis, the 8214A PMCs formed abnormal non-tetrahedral tetrads that degraded in situ. To identify the genes involved in the pollen abortion of line 8214A, the transcriptional profiles of the 8214A and the 8214B anthers (i.e., from the fertile maintainer line) during meiosis were analyzed using an RNA-seq approach. A total of 1355 genes were determined to be differentially expressed, including 424 and 931 up- and down- regulated genes, respectively, in the 8214A anthers during meiosis relative to the expression levels in the 8214B. The expression levels of ubiquitin ligase and cell cycle-related genes were apparently down-regulated, while the expression of methyltransferase genes was up-regulated in the 8214A anthers during meiosis, which likely contributed to the PCD of these PMCs during meiosis. Thus, our results may be useful for revealing the molecular mechanism regulating the pollen abortion of CMS pepper.
Collapse
|
217
|
Fang D, Gan H, Wang H, Zhou H, Zhang Z. Probe the function of histone lysine 36 methylation using histone H3 lysine 36 to methionine mutant transgene in mammalian cells. Cell Cycle 2017; 16:1781-1789. [PMID: 28129023 PMCID: PMC5628648 DOI: 10.1080/15384101.2017.1281483] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/20/2016] [Accepted: 01/05/2017] [Indexed: 12/12/2022] Open
Abstract
Chondroblastoma is a cartilaginous tumor that typically arises under 25 y of age (80%). Recent studies have identified a somatic and heterozygous mutation at the H3F3B gene in over 90% chondroblastoma cases, leading to a lysine 36 to methionine replacement (H3.3K36M). In human cells, H3F3B gene is one of 2 genes that encode identical H3.3 proteins. It is not known how H3.3K36M mutant proteins promote tumorigenesis. We and others have shown that, the levels of H3K36 di- and tri-methylation (H3K36me2/me3) are reduced dramatically in chondroblastomas and chondrocytes bearing the H3.3K36M mutation. Mechanistically, H3.3K36M mutant proteins inhibit enzymatic activity of some, but not all H3K36 methyltransferases. Chondrocytes harboring the same H3F3B mutation exhibited the cancer cell associated phenotypes. Here, we discuss the potential effects of H3.3K36M mutation on epigenomes including H3K36 and H3K27 methylation and cellular phenotypes. We suggest that H3.3K36M mutant proteins alter epigenomes of specific progenitor cells, which in turn lead to cellular transformation and tumorigenesis.
Collapse
Affiliation(s)
- Dong Fang
- Department of Pediatrics and Department of Genetics and Development, Institute for Cancer Genetics, Irving Cancer Research Center, Columbia University, New York, NY, USA
| | - Haiyun Gan
- Department of Pediatrics and Department of Genetics and Development, Institute for Cancer Genetics, Irving Cancer Research Center, Columbia University, New York, NY, USA
| | - Heping Wang
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Hankou, Wuhan, P.R. China
| | - Hui Zhou
- Department of Pediatrics and Department of Genetics and Development, Institute for Cancer Genetics, Irving Cancer Research Center, Columbia University, New York, NY, USA
| | - Zhiguo Zhang
- Department of Pediatrics and Department of Genetics and Development, Institute for Cancer Genetics, Irving Cancer Research Center, Columbia University, New York, NY, USA
| |
Collapse
|
218
|
Tidwell TR, Søreide K, Hagland HR. Aging, Metabolism, and Cancer Development: from Peto's Paradox to the Warburg Effect. Aging Dis 2017; 8:662-676. [PMID: 28966808 PMCID: PMC5614328 DOI: 10.14336/ad.2017.0713] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 06/13/2017] [Indexed: 12/15/2022] Open
Abstract
Medical advances made over the last century have increased our lifespan, but age-related diseases are a fundamental health burden worldwide. Aging is therefore a major risk factor for cardiovascular disease, cancer, diabetes, obesity, and neurodegenerative diseases, all increasing in prevalence. However, huge inter-individual variations in aging and disease risk exist, which cannot be explained by chronological age, but rather physiological age decline initiated even at young age due to lifestyle. At the heart of this lies the metabolic system and how this is regulated in each individual. Metabolic turnover of food to energy leads to accumulation of co-factors, byproducts, and certain proteins, which all influence gene expression through epigenetic regulation. How these epigenetic markers accumulate over time is now being investigated as the possible link between aging and many diseases, such as cancer. The relationship between metabolism and cancer was described as early as the late 1950s by Dr. Otto Warburg, before the identification of DNA and much earlier than our knowledge of epigenetics. However, when the stepwise gene mutation theory of cancer was presented, Warburg's theories garnered little attention. Only in the last decade, with epigenetic discoveries, have Warburg's data on the metabolic shift in cancers been brought back to life. The stepwise gene mutation theory fails to explain why large animals with more cells, do not have a greater cancer incidence than humans, known as Peto's paradox. The resurgence of research into the Warburg effect has given us insight to what may explain Peto's paradox. In this review, we discuss these connections and how age-related changes in metabolism are tightly linked to cancer development, which is further affected by lifestyle choices modulating the risk of aging and cancer through epigenetic control.
Collapse
Affiliation(s)
- Tia R. Tidwell
- Department of Mathematics and Natural Sciences, Centre for Organelle Research, University of Stavanger, Stavanger, Norway
- Gastrointestinal Translational Research Unit, Molecular Laboratory, Hillevaåg, Stavanger University Hospital, Stavanger, Norway
| | - Kjetil Søreide
- Gastrointestinal Translational Research Unit, Molecular Laboratory, Hillevaåg, Stavanger University Hospital, Stavanger, Norway
- Department of Gastrointestinal Surgery, Stavanger University Hospital, Stavanger, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Hanne R. Hagland
- Department of Mathematics and Natural Sciences, Centre for Organelle Research, University of Stavanger, Stavanger, Norway
- Gastrointestinal Translational Research Unit, Molecular Laboratory, Hillevaåg, Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|
219
|
Epigenetics in multiple myeloma: From mechanisms to therapy. Semin Cancer Biol 2017; 51:101-115. [PMID: 28962927 DOI: 10.1016/j.semcancer.2017.09.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 08/25/2017] [Accepted: 09/25/2017] [Indexed: 12/22/2022]
Abstract
Multiple myeloma (MM) is a tumor of antibody producing plasmablasts/plasma cells that resides within the bone marrow (BM). In addition to the well-established role of genetic lesions and tumor-microenvironment interactions in the development of MM, deregulated epigenetic mechanisms are emerging as important in MM pathogenesis. Recently, MM sequencing and expression projects have revealed that mutations and copy number variations as well as deregulation in the expression of epigenetic modifiers are characteristic features of MM. In the past decade, several studies have suggested epigenetic mechanisms via DNA methylation, histone modifications and non-coding RNAs as important contributing factors in MM with impacts on disease initiation, progression, clonal heterogeneity and response to treatment. Herein we review the present view and knowledge that has accumulated over the past decades on the role of epigenetics in MM, with focus on the interplay between epigenetic mechanisms and the potential use of epigenetic inhibitors as future treatment modalities for MM.
Collapse
|
220
|
Abstract
As an important epigenetic mark, lysine methylations play critical roles in the regulation of both chromatin and non-chromatin proteins. There are three levels of lysine methylation, mono-, di-, and trimethylation. Each one has turned out to be biologically distinctive. For the biochemical characterization of proteins with lysine methylation, multiple chemical biology methods have been developed. This concept article will highlight these developments and their applications in epigenetic investigation of protein functions.
Collapse
Affiliation(s)
- Zhipeng A. Wang
- Chemistry Department, Texas A&M University, College Station, TX, 77843, USA
| | - Wenshe R. Liu
- Chemistry Department, Texas A&M University, College Station, TX, 77843, USA
| |
Collapse
|
221
|
Bhan A, Deb P, Shihabeddin N, Ansari KI, Brotto M, Mandal SS. Histone methylase MLL1 coordinates with HIF and regulate lncRNA HOTAIR expression under hypoxia. Gene 2017; 629:16-28. [DOI: 10.1016/j.gene.2017.07.069] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/11/2017] [Accepted: 07/25/2017] [Indexed: 12/17/2022]
|
222
|
Baranek M, Belter A, Naskręt-Barciszewska MZ, Stobiecki M, Markiewicz WT, Barciszewski J. Effect of small molecules on cell reprogramming. MOLECULAR BIOSYSTEMS 2017; 13:277-313. [PMID: 27918060 DOI: 10.1039/c6mb00595k] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The essential idea of regenerative medicine is to fix or replace tissues or organs with alive and patient-specific implants. Pluripotent stem cells are able to indefinitely self-renew and differentiate into all cell types of the body which makes them a potent substantial player in regenerative medicine. The easily accessible source of induced pluripotent stem cells may allow obtaining and cultivating tissues in vitro. Reprogramming refers to regression of mature cells to its initial pluripotent state. One of the approaches affecting pluripotency is the usage of low molecular mass compounds that can modulate enzymes and receptors leading to the formation of pluripotent stem cells (iPSCs). It would be great to assess the general character of such compounds and reveal their new derivatives or modifications to increase the cell reprogramming efficiency. Many improvements in the methods of pluripotency induction have been made by various groups in order to limit the immunogenicity and tumorigenesis, increase the efficiency and accelerate the kinetics. Understanding the epigenetic changes during the cellular reprogramming process will extend the comprehension of stem cell biology and lead to potential therapeutic approaches. There are compounds which have been already proven to be or for now only putative inducers of the pluripotent state that may substitute for the classic reprogramming factors (Oct3/4, Sox2, Klf4, c-Myc) in order to improve the time and efficiency of pluripotency induction. The effect of small molecules on gene expression is dosage-dependent and their application concentration needs to be strictly determined. In this review we analysed the role of small molecules in modulations leading to pluripotency induction, thereby contributing to our understanding of stem cell biology and uncovering the major mechanisms involved in that process.
Collapse
Affiliation(s)
- M Baranek
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego str. 12/14, 61-704 Poznań, Poland.
| | - A Belter
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego str. 12/14, 61-704 Poznań, Poland.
| | - M Z Naskręt-Barciszewska
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego str. 12/14, 61-704 Poznań, Poland.
| | - M Stobiecki
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego str. 12/14, 61-704 Poznań, Poland.
| | - W T Markiewicz
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego str. 12/14, 61-704 Poznań, Poland.
| | - J Barciszewski
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego str. 12/14, 61-704 Poznań, Poland.
| |
Collapse
|
223
|
Meyer C, Burmeister T, Gröger D, Tsaur G, Fechina L, Renneville A, Sutton R, Venn NC, Emerenciano M, Pombo-de-Oliveira MS, Barbieri Blunck C, Almeida Lopes B, Zuna J, Trka J, Ballerini P, Lapillonne H, De Braekeleer M, Cazzaniga G, Corral Abascal L, van der Velden VHJ, Delabesse E, Park TS, Oh SH, Silva MLM, Lund-Aho T, Juvonen V, Moore AS, Heidenreich O, Vormoor J, Zerkalenkova E, Olshanskaya Y, Bueno C, Menendez P, Teigler-Schlegel A, Zur Stadt U, Lentes J, Göhring G, Kustanovich A, Aleinikova O, Schäfer BW, Kubetzko S, Madsen HO, Gruhn B, Duarte X, Gameiro P, Lippert E, Bidet A, Cayuela JM, Clappier E, Alonso CN, Zwaan CM, van den Heuvel-Eibrink MM, Izraeli S, Trakhtenbrot L, Archer P, Hancock J, Möricke A, Alten J, Schrappe M, Stanulla M, Strehl S, Attarbaschi A, Dworzak M, Haas OA, Panzer-Grümayer R, Sedék L, Szczepański T, Caye A, Suarez L, Cavé H, Marschalek R. The MLL recombinome of acute leukemias in 2017. Leukemia 2017; 32:273-284. [PMID: 28701730 PMCID: PMC5808070 DOI: 10.1038/leu.2017.213] [Citation(s) in RCA: 506] [Impact Index Per Article: 63.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/25/2017] [Accepted: 06/21/2017] [Indexed: 12/16/2022]
Abstract
Chromosomal rearrangements of the human MLL/KMT2A gene are associated with infant, pediatric, adult and therapy-induced acute leukemias. Here we present the data obtained from 2345 acute leukemia patients. Genomic breakpoints within the MLL gene and the involved translocation partner genes (TPGs) were determined and 11 novel TPGs were identified. Thus, a total of 135 different MLL rearrangements have been identified so far, of which 94 TPGs are now characterized at the molecular level. In all, 35 out of these 94 TPGs occur recurrently, but only 9 specific gene fusions account for more than 90% of all illegitimate recombinations of the MLL gene. We observed an age-dependent breakpoint shift with breakpoints localizing within MLL intron 11 associated with acute lymphoblastic leukemia and younger patients, while breakpoints in MLL intron 9 predominate in AML or older patients. The molecular characterization of MLL breakpoints suggests different etiologies in the different age groups and allows the correlation of functional domains of the MLL gene with clinical outcome. This study provides a comprehensive analysis of the MLL recombinome in acute leukemia and demonstrates that the establishment of patient-specific chromosomal fusion sites allows the design of specific PCR primers for minimal residual disease analyses for all patients.
Collapse
Affiliation(s)
- C Meyer
- Institute of Pharmaceutical Biology/Diagnostic Center of Acute Leukemia (DCAL), Goethe-University, Frankfurt/Main, Germany
| | - T Burmeister
- Charité-Department of Hematology, Oncology and Tumorimmunology, Berlin, Germany
| | - D Gröger
- Charité-Department of Hematology, Oncology and Tumorimmunology, Berlin, Germany
| | - G Tsaur
- Regional Children Hospital 1, Research Institute of Medical Cell Technologies, Pediatric Oncology and Hematology Center, Ural Federal University, Ekaterinburg, Russia
| | - L Fechina
- Regional Children Hospital 1, Research Institute of Medical Cell Technologies, Pediatric Oncology and Hematology Center, Ural Federal University, Ekaterinburg, Russia
| | - A Renneville
- Laboratory of Hematology, Biology and Pathology Center, CHRU of Lille; INSERM, UMR-S 1172, Cancer Research Institute of Lille, Lille, France
| | - R Sutton
- Children's Cancer Institute Australia, Uinversity of NSW Sydney, Sydney, New South Wales, Australia
| | - N C Venn
- Children's Cancer Institute Australia, Uinversity of NSW Sydney, Sydney, New South Wales, Australia
| | - M Emerenciano
- Pediatric Hematology-Oncology Program-Research Center, Instituto Nacional de Cancer Rio de Janeiro, Rio de Janeiro, Brazil
| | - M S Pombo-de-Oliveira
- Pediatric Hematology-Oncology Program-Research Center, Instituto Nacional de Cancer Rio de Janeiro, Rio de Janeiro, Brazil
| | - C Barbieri Blunck
- Pediatric Hematology-Oncology Program-Research Center, Instituto Nacional de Cancer Rio de Janeiro, Rio de Janeiro, Brazil
| | - B Almeida Lopes
- Pediatric Hematology-Oncology Program-Research Center, Instituto Nacional de Cancer Rio de Janeiro, Rio de Janeiro, Brazil
| | - J Zuna
- CLIP, Department of Paediatric Haematology/Oncology, Charles University Prague, 2nd Faculty of Medicine, Prague, Czech Republic
| | - J Trka
- CLIP, Department of Paediatric Haematology/Oncology, Charles University Prague, 2nd Faculty of Medicine, Prague, Czech Republic
| | - P Ballerini
- Biological Hematology, AP-HP A. Trousseau, Pierre et Marie Curie University, Paris, France
| | - H Lapillonne
- Biological Hematology, AP-HP A. Trousseau, Pierre et Marie Curie University, Paris, France
| | - M De Braekeleer
- Université de Bretagne Occidentale, Faculté de Médecine et des Sciences de la Santé, Laboratoire d'Histologie, Embryologie et Cytogénétique & INSERM-U1078, Brest, France
| | - G Cazzaniga
- Centro Ricerca Tettamanti, Clinica Pediatrica Univ. Milano Bicocca, Monza, Italy
| | - L Corral Abascal
- Centro Ricerca Tettamanti, Clinica Pediatrica Univ. Milano Bicocca, Monza, Italy
| | | | - E Delabesse
- CHU Purpan, Laboratoire d'Hématologie, Toulouse, France
| | - T S Park
- Department of Laboratory Medicine, School of Medicine, Kyung Hee University, Seoul, Korea
| | - S H Oh
- Department of Laboratory Medicine, Inje University College of Medicine, Busan, Korea
| | - M L M Silva
- Cytogenetics Department, Bone Marrow Transplantation Unit, National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - T Lund-Aho
- Laboratory of Clinical Genetics, Fimlab Laboratories, Tampere, Finland
| | - V Juvonen
- Department of Clinical Chemistry and TYKSLAB, University of Turku and Turku University Central Hospital, Turku, Finland
| | - A S Moore
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - O Heidenreich
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - J Vormoor
- The Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - E Zerkalenkova
- Dmitry Rogachev National Scientific and Practical Center of Pediatric Hematology, Oncology and Immunology, Moscow
| | - Y Olshanskaya
- Dmitry Rogachev National Scientific and Practical Center of Pediatric Hematology, Oncology and Immunology, Moscow
| | - C Bueno
- Josep Carreras Leukemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain.,CIBER de Cancer (CIBERONC), ISCIII, Madrid, Spain.,Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - P Menendez
- Josep Carreras Leukemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain.,CIBER de Cancer (CIBERONC), ISCIII, Madrid, Spain.,Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - A Teigler-Schlegel
- Department of Experimental Pathology and Cytology, Institute of Pathology, Giessen, Germany
| | - U Zur Stadt
- Center for Diagnostic, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - J Lentes
- Department of Human Genetics, Hannover Medical School, Hanover, Germany
| | - G Göhring
- Department of Human Genetics, Hannover Medical School, Hanover, Germany
| | - A Kustanovich
- Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk, Republic of Belarus
| | - O Aleinikova
- Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk, Republic of Belarus
| | - B W Schäfer
- Department of Oncology, University Children's Hospital Zurich, Zurich, Switzerland
| | - S Kubetzko
- Department of Oncology, University Children's Hospital Zurich, Zurich, Switzerland
| | - H O Madsen
- Department of Clinical Immunology, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - B Gruhn
- Department of Pediatrics, Jena University Hospital, Jena, Germany
| | - X Duarte
- Department of Pediatrics, Portuguese Institute of Oncology of Lisbon, Lisbon, Portugal
| | - P Gameiro
- Hemato-Oncology Laboratory, UIPM, Portuguese Institute of Oncology of Lisbon, Lisbon, Portugal
| | - E Lippert
- Hématologie Biologique, CHU de Brest and INSERM U1078, Université de Bretagne Occidentale, Brest, France
| | - A Bidet
- Hématologie Biologique, CHU de Brest and INSERM U1078, Université de Bretagne Occidentale, Brest, France
| | - J M Cayuela
- Laboratoire d'hématologie, AP-HP Saint-Louis, Paris Diderot University, Paris, France
| | - E Clappier
- Laboratoire d'hématologie, AP-HP Saint-Louis, Paris Diderot University, Paris, France
| | - C N Alonso
- Hospital Nacional de Pediatría Prof Dr J. P. Garrahan, Servcio de Hemato-Oncología, Buenos Aires, Argentina
| | - C M Zwaan
- Department of Pediatric Oncology/Hematology, Erasmus MC, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - M M van den Heuvel-Eibrink
- Department of Pediatric Oncology/Hematology, Erasmus MC, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - S Izraeli
- The Chaim Sheba Medical Center, Department of Pediatric Hemato-Oncology and the Cancer Research Center, Tel Aviv, Israel.,Sackler Medical School Tel Aviv University, Tel Aviv, Israel
| | - L Trakhtenbrot
- The Chaim Sheba Medical Center, Department of Pediatric Hemato-Oncology and the Cancer Research Center, Tel Aviv, Israel.,Sackler Medical School Tel Aviv University, Tel Aviv, Israel
| | - P Archer
- Bristol Genetics Laboratory, Pathology Sciences, Southmead Hospital, North Bristol NHS Trust, Bristol, UK
| | - J Hancock
- Bristol Genetics Laboratory, Pathology Sciences, Southmead Hospital, North Bristol NHS Trust, Bristol, UK
| | - A Möricke
- Department of Pediatrics, University Medical Centre Schleswig-Holstein, Kiel, Germany
| | - J Alten
- Department of Pediatrics, University Medical Centre Schleswig-Holstein, Kiel, Germany
| | - M Schrappe
- Department of Pediatrics, University Medical Centre Schleswig-Holstein, Kiel, Germany
| | - M Stanulla
- Department of Pediatrics, MHH, Hanover, Germany
| | - S Strehl
- Children's Cancer Research Institute and St Anna Children's Hospital, Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - A Attarbaschi
- Children's Cancer Research Institute and St Anna Children's Hospital, Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - M Dworzak
- Children's Cancer Research Institute and St Anna Children's Hospital, Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - O A Haas
- Children's Cancer Research Institute and St Anna Children's Hospital, Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - R Panzer-Grümayer
- Children's Cancer Research Institute and St Anna Children's Hospital, Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - L Sedék
- Department of Microbiology and Immunology, Medical University of Silesia, Zabrze, Poland
| | - T Szczepański
- Department of Pediatric Hematology and Oncology, Medical University of Silesia, Zabrze, Poland
| | - A Caye
- Department of Genetics, AP-HP Robert Debré, Paris Diderot University, Paris, France
| | - L Suarez
- Department of Genetics, AP-HP Robert Debré, Paris Diderot University, Paris, France
| | - H Cavé
- Department of Genetics, AP-HP Robert Debré, Paris Diderot University, Paris, France
| | - R Marschalek
- Institute of Pharmaceutical Biology/Diagnostic Center of Acute Leukemia (DCAL), Goethe-University, Frankfurt/Main, Germany
| |
Collapse
|
224
|
He Y, Bao B, Li H. Using zebrafish as a model to study the role of epigenetics in hearing loss. Expert Opin Drug Discov 2017; 12:967-975. [PMID: 28682135 DOI: 10.1080/17460441.2017.1340270] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The rapid progress of bioinformatics and high-throughput screening techniques in recent years has led to the identification of many candidate genes and small-molecule drugs that have the potential to make significant contributions to our understanding of the developmental and pathological processes of hearing, but it remains unclear how these genes and regulatory factors are coordinated. Increasing evidence suggests that epigenetic mechanisms are essential for establishing gene expression profiles and likely play an important role in the development of inner ear and in the pathology of hearing-associated diseases. Zebrafish are a valuable and tractable in vivo model organism for monitoring changes in the epigenome and for identifying new epigenetic processes and drug molecules that can influence vertebrate development. Areas covered: In this review, the authors focus on zebrafish as a model to summarize recent findings concerning the roles of epigenetics in the development, regeneration, and protection of hair cells. Expert opinion: Using the zebrafish model in combination with high-throughput screening and genome-editing technologies to investigate the function of epigenetics in hearing is crucial to help us better understand the molecular and genetic mechanisms of auditory development and function. It will also contribute to the development of new strategies to restore hearing loss.
Collapse
Affiliation(s)
- Yingzi He
- a ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology , Fudan University , Shanghai , China.,c Key Laboratory of Hearing Medicine of NHFPC , Shanghai , China
| | - Beier Bao
- a ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology , Fudan University , Shanghai , China
| | - Huawei Li
- a ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology , Fudan University , Shanghai , China.,b Institutes of Biomedical Sciences , Fudan University , Shanghai , China.,c Key Laboratory of Hearing Medicine of NHFPC , Shanghai , China.,d Shanghai Engineering Research Centre of Cochlear Implant , Shanghai , China.,e The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science , Fudan University , Shanghai , China
| |
Collapse
|
225
|
Mori S, Hirano T, Takaguchi A, Fujiwara T, Okazaki Y, Kagechika H. Selective Reagent for Detection ofN-ε-Monomethylation of a Peptide Lysine Residue through SNAr Reaction. European J Org Chem 2017. [DOI: 10.1002/ejoc.201700488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Shuichi Mori
- Institute of Biomaterials and Bioengineering; Tokyo Medical and Dental University (TMDU); 2-3-10 Kanda-Surugadai, Chiyoda-ku 101-0062 Tokyo Japan
| | - Tomoya Hirano
- Institute of Biomaterials and Bioengineering; Tokyo Medical and Dental University (TMDU); 2-3-10 Kanda-Surugadai, Chiyoda-ku 101-0062 Tokyo Japan
| | - Asuka Takaguchi
- Institute of Biomaterials and Bioengineering; Tokyo Medical and Dental University (TMDU); 2-3-10 Kanda-Surugadai, Chiyoda-ku 101-0062 Tokyo Japan
| | - Takashi Fujiwara
- Institute of Biomaterials and Bioengineering; Tokyo Medical and Dental University (TMDU); 2-3-10 Kanda-Surugadai, Chiyoda-ku 101-0062 Tokyo Japan
| | - Yusuke Okazaki
- Institute of Biomaterials and Bioengineering; Tokyo Medical and Dental University (TMDU); 2-3-10 Kanda-Surugadai, Chiyoda-ku 101-0062 Tokyo Japan
| | - Hiroyuki Kagechika
- Institute of Biomaterials and Bioengineering; Tokyo Medical and Dental University (TMDU); 2-3-10 Kanda-Surugadai, Chiyoda-ku 101-0062 Tokyo Japan
| |
Collapse
|
226
|
Histone methylation in the freeze-tolerant wood frog (Rana sylvatica). J Comp Physiol B 2017; 188:113-125. [DOI: 10.1007/s00360-017-1112-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 05/23/2017] [Accepted: 06/06/2017] [Indexed: 12/28/2022]
|
227
|
BMI-1 is a potential therapeutic target in diffuse intrinsic pontine glioma. Oncotarget 2017; 8:62962-62975. [PMID: 28968963 PMCID: PMC5609895 DOI: 10.18632/oncotarget.18002] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 04/26/2017] [Indexed: 12/14/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a poor-prognosis pediatric brain tumor. No effective curative therapy is currently available and no therapeutic advances have been made in several decades. BMI-1 is a member of the multimeric protein complex Polycomb repressor complex 1. It is highly expressed in a number of diseases and malignancies and has been implicated in self-renewal of normal and cancer cells, and in DNA damage signaling. The role of BMI-1 in DIPG is largely unknown. Here, we show that BMI-1 is highly expressed in tumor tissue samples of DIPG patients and in patient-derived cancer stem-like cells. BMI-1 downregulation leads to the inhibition of DIPG patient-derived neurosphere cell proliferation, cell cycle signaling, self-renewal, telomerase expression and activity, and suppresses DIPG cell migration. Moreover, targeted inhibition of BMI-1 sensitizes DIPG cells to radiomimetic drug-induced DNA damage. Together, our data validate BMI-1 as a potential therapeutic target to treat children with DIPG.
Collapse
|
228
|
Wang W, Zheng K, Pei Y, Zhang X. Histone Demethylase JARID1B Is Overexpressed in Osteosarcoma and Upregulates Cyclin D1 Expression via Demethylation of H3K27me3. Oncol Res 2017; 26:373-384. [PMID: 28492139 PMCID: PMC7844678 DOI: 10.3727/096504017x14939809845080] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
JARID1B has been proven to be upregulated in many human malignancies and is correlated with tumor progression. However, its expression and clinical significance in osteosarcoma are still unclear. Thus, the aim of this study was to explore the effects of JARID1B in osteosarcoma tumorigenesis and development. In this study, we found that the expression levels of JARID1B in osteosarcoma tissues were significantly higher than those in corresponding noncancerous bone tissues. In addition, JARID1B upregulation occurred more frequently in osteosarcoma specimens from patients with a poor prognosis. After JARID1B transfection in osteosarcoma cells, cell proliferation was significantly promoted in vitro and in vivo. On the contrary, knockdown of JARID1B inhibited cell proliferation in vitro and tumor growth in vivo. JARID1B can also decrease the G0/G1 phase cell numbers and increase the S and G2/M phase cell numbers. We further demonstrated that JARID1B regulates cyclin D1 expression through H3K27me3. These findings indicate that JARID1B may act not only as a novel diagnostic and prognostic marker but also as a potential target for molecular therapy in osteosarcoma.
Collapse
Affiliation(s)
- Wei Wang
- Department of Bone and Soft-Tissue Tumor Surgery, Cancer Hospital of China Medical UniversityShenyang, Liaoning ProvinceP.R. China
| | - Ke Zheng
- Department of Bone and Soft-Tissue Tumor Surgery, Cancer Hospital of China Medical UniversityShenyang, Liaoning ProvinceP.R. China
| | - Yi Pei
- Department of Bone and Soft-Tissue Tumor Surgery, Cancer Hospital of China Medical UniversityShenyang, Liaoning ProvinceP.R. China
| | - XiaoJing Zhang
- Department of Bone and Soft-Tissue Tumor Surgery, Cancer Hospital of China Medical UniversityShenyang, Liaoning ProvinceP.R. China
| |
Collapse
|
229
|
Matrix Metalloproteinase Gene Activation Resulting from Disordred Epigenetic Mechanisms in Rheumatoid Arthritis. Int J Mol Sci 2017; 18:ijms18050905. [PMID: 28441353 PMCID: PMC5454818 DOI: 10.3390/ijms18050905] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/18/2017] [Accepted: 04/19/2017] [Indexed: 12/29/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are implicated in the degradation of extracellular matrix (ECM). Rheumatoid arthritis (RA) synovial fibroblasts (SFs) produce matrix-degrading enzymes, including MMPs, which facilitate cartilage destruction in the affected joints in RA. Epigenetic mechanisms contribute to change in the chromatin state, resulting in an alteration of gene transcription. Recently, MMP gene activation has been shown to be caused in RASFs by the dysregulation of epigenetic changes, such as histone modifications, DNA methylation, and microRNA (miRNA) signaling. In this paper, we review the role of MMPs in the pathogenesis of RA as well as the disordered epigenetic mechanisms regulating MMP gene activation in RASFs.
Collapse
|
230
|
Goyama S, Kitamura T. Epigenetics in normal and malignant hematopoiesis: An overview and update 2017. Cancer Sci 2017; 108:553-562. [PMID: 28100030 PMCID: PMC5406607 DOI: 10.1111/cas.13168] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 01/05/2017] [Accepted: 01/11/2017] [Indexed: 12/11/2022] Open
Abstract
Epigenetic regulation in hematopoiesis has been a field of rapid expansion. Genome‐wide analyses have revealed, and will continue to identify genetic alterations in epigenetic genes that are present in various types of hematopoietic neoplasms. Development of new mouse models for individual epigenetic modifiers has revealed their novel, sometimes unexpected, functions. In this review, we provide an overview of genetic alterations within epigenetic genes in various types of hematopoietic neoplasms. We then summarize the physiologic roles of these epigenetic modifiers during hematopoiesis, and describe therapeutic approaches targeting the epigenetic modifications. Interestingly, the mutational spectrum of epigenetic genes indicates that myeloid neoplasms are similar to T‐cell neoplasms, whereas B‐cell lymphomas have distinct features. Furthermore, it appears that the epigenetic mutations related to active transcription are more associated with myeloid/T‐cell neoplasms, whereas those that repress transcription are associated with B‐cell lymphomas. These observations may imply that the global low‐level or high‐level transcriptional activity underlies the development of myeloid/T‐cell tumors or B‐cell tumors, respectively.
Collapse
Affiliation(s)
- Susumu Goyama
- Division of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Toshio Kitamura
- Division of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
231
|
How does chromatin package DNA within nucleus and regulate gene expression? Int J Biol Macromol 2017; 101:862-881. [PMID: 28366861 DOI: 10.1016/j.ijbiomac.2017.03.165] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/28/2017] [Accepted: 03/28/2017] [Indexed: 01/26/2023]
Abstract
The human body is made up of 60 trillion cells, each cell containing 2 millions of genomic DNA in its nucleus. How is this genomic deoxyribonucleic acid [DNA] organised into nuclei? Around 1880, W. Flemming discovered a nuclear substance that was clearly visible on staining under primitive light microscopes and named it 'chromatin'; this is now thought to be the basic unit of genomic DNA organization. Since long before DNA was known to carry genetic information, chromatin has fascinated biologists. DNA has a negatively charged phosphate backbone that produces electrostatic repulsion between adjacent DNA regions, making it difficult for DNA to fold upon itself. In this article, we will try to shed light on how does chromatin package DNA within nucleus and regulate gene expression?
Collapse
|
232
|
Effects of histone deacetylase inhibitory prodrugs on epigenetic changes and DNA damage response in tumor and heart of glioblastoma xenograft. Invest New Drugs 2017; 35:412-426. [DOI: 10.1007/s10637-017-0448-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 03/01/2017] [Indexed: 12/22/2022]
|
233
|
Zhou ZH, Yang J, Kong AN. Phytochemicals in Traditional Chinese Herbal Medicine: Cancer Prevention and Epigenetics Mechanisms. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/s40495-017-0086-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
234
|
SETD4 Regulates Cell Quiescence and Catalyzes the Trimethylation of H4K20 during Diapause Formation in Artemia. Mol Cell Biol 2017; 37:MCB.00453-16. [PMID: 28031330 DOI: 10.1128/mcb.00453-16] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 12/02/2016] [Indexed: 01/19/2023] Open
Abstract
As a prominent characteristic of cell life, the regulation of cell quiescence is important for proper development, regeneration, and stress resistance and may play a role in certain degenerative diseases. However, the mechanism underlying quiescence remains largely unknown. Encysted embryos of Artemia are useful for studying the regulation of this state because they remain quiescent for prolonged periods during diapause, a state of obligate dormancy. In the present study, SET domain-containing protein 4, a histone lysine methyltransferase from Artemia, was identified, characterized, and named Ar-SETD4. We found that Ar-SETD4 was expressed abundantly in Artemia diapause embryos, in which cells were in a quiescent state. Meanwhile, trimethylated histone H4K20 (H4K20me3) was enriched in diapause embryos. The knockdown of Ar-SETD4 reduced the level of H4K20me3 significantly and prevented the formation of diapause embryos in which neither the cell cycle nor embryogenesis ceased. The catalytic activity of Ar-SETD4 on H4K20me3 was confirmed by an in vitro histone methyltransferase (HMT) assay and overexpression in cell lines. This study provides insights into the function of SETD4 and the mechanism of cell quiescence regulation.
Collapse
|
235
|
Zhou HR, Fu HY, Wu DS, Zhang YY, Huang SH, Chen CJ, Yan JG, Huang JL, Shen JZ. Relationship between epigenetic changes in Wnt antagonists and acute leukemia. Oncol Rep 2017; 37:2663-2671. [PMID: 28440495 PMCID: PMC5428301 DOI: 10.3892/or.2017.5509] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 12/16/2016] [Indexed: 11/05/2022] Open
Abstract
The present study was designed to investigate the relationship among epigenetic changes in Wnt antagonists, histone H4K20me1 and the expression of tumor-suppressor genes in acute leukemia (AL) to better understand the pathogenesis of leukemia. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was performed to detect messenger RNA (mRNA) expression levels of Wnt antagonists (Wnt5a, HDPR1, DKK1 and DKK3) in patients with AL and in normal controls; pyrophosphate sequencing was performed to detect the methylation status of the Wnt5a promoter; and western blotting was performed to detect the overall expression levels of Wnt5a protein and histone H4K20me1 in patients with acute myeloid leukemia (AML) and in normal controls. The relationship between Wnt5a protein expression and histone H4K20me1 was analyzed. Chromatin immunoprecipitation-qPCR (ChIP-qPCR) was performed to investigate the recruitment of H4K20me1 and SET8 to the Wnt5a promoter and coding regions. Our results demonstrated that the expression levels of Wnt antagonists were generally low in AML, but showed differential expression in acute lymphocytic leukemia (ALL). In most cases of AML, methylation of the Wnt5a promoter was observed and Wnt5a protein expression was low. In some cases of AML, the overall level of H4K20me1 protein was higher than that in normal controls. In addition, Wnt5a expression was positively correlated with H4K20me1 expression and was unrelated to the methylation status of its promoter. Moreover, H4K20me1 and SET8 were enriched in the Wnt5a promoter region and coding region. By contrast, Wnt5a expression was unrelated to H4K20me1 expression in normal controls. Moreover, we observed that the methylation of Wnt antagonists was often found in patients with AL, particularly those with AML, whereas the extent of methylation was variable in ALL patients. Wnt5a expression was positively correlated with the enrichment of H4K20me1 and SET8 at the Wnt5a promoter and coding regions. H4K20me1 increased Wnt5a expression by promoting transcription initiation and elongation.
Collapse
Affiliation(s)
- Hua-Rong Zhou
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Hai-Ying Fu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Dan-Sen Wu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Yuan-Yuan Zhang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Si-Han Huang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Cong-Jie Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Jian-Guo Yan
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Jin-Long Huang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Jian-Zhen Shen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
236
|
Kowluru RA, Mishra M. Regulation of Matrix Metalloproteinase in the Pathogenesis of Diabetic Retinopathy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 148:67-85. [PMID: 28662829 DOI: 10.1016/bs.pmbts.2017.02.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Diabetic retinopathy, a progressive disease, is the major cause of acquired blindness in the developed countries. Despite cutting-edge research in the field, the exact mechanism of this multifactorial disease remains elusive. Matrix metalloproteinases (MMPs) degrade extracellular matrix and play significant role in regulating intracellular homeostasis. In the pathogenesis of diabetic retinopathy, activation of gelatinase MMPs (MMP-2 and MMP-9) in the retina is an early event, and activated MMPs damage the mitochondria and augment retinal capillary cell apoptosis, a phenomenon which is observed before histopathology characteristic of diabetic retinopathy can be seen. MMPs are regulated by a number of different mechanisms including cleavage of their zymogens, regulation of their tissue inhibitors, and their gene expressions by transcriptional factors and epigenetic modifications. This chapter reviews the current literature about the role of MMPs in the development of diabetic retinopathy, and describes different mechanisms to regulate their activation. With evolving research implicating MMPs in both preneovascularization and neovascularization stages of diabetic retinopathy, they could be an attractive target to inhibit the development/progression of diabetic retinopathy, a disease which has potential to rob vision during the most productive years of a diabetic patient's life.
Collapse
Affiliation(s)
- Renu A Kowluru
- Kresge Eye Institute, Wayne State University, Detroit, MI, United States.
| | - Manish Mishra
- Kresge Eye Institute, Wayne State University, Detroit, MI, United States
| |
Collapse
|
237
|
Tsukamoto D, Ito M, Takamatsu N. HNF-4 participates in the hibernation-associated transcriptional regulation of the chipmunk hibernation-related protein gene. Sci Rep 2017; 7:44279. [PMID: 28281641 PMCID: PMC5345028 DOI: 10.1038/srep44279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 02/06/2017] [Indexed: 11/17/2022] Open
Abstract
The chipmunk hibernation-related protein 25 (HP-25) is involved in the circannual control of hibernation in the brain. The liver-specific expression of the HP-25 gene is repressed in hibernating chipmunks under the control of endogenous circannual rhythms. However, the molecular mechanisms that differentially regulate the HP-25 gene during the nonhibernation and hibernation seasons are unknown. Here, we show that the hibernation-associated HP-25 expression is regulated epigenetically. Chromatin immunoprecipitation analyses revealed that significantly less hepatocyte nuclear receptor HNF-4 bound to the HP-25 gene promoter in the liver of hibernating chipmunks compared to nonhibernating chipmunks. Concurrently in the hibernating chipmunks, coactivators were dissociated from the promoter, and active transcription histone marks on the HP-25 gene promoter were lost. On the other hand, small heterodimer partner (SHP) expression was upregulated in the liver of hibernating chipmunks. Overexpressing SHP in primary hepatocytes prepared from nonhibernating chipmunks caused HNF-4 to dissociate from the HP-25 gene promoter, and reduced the HP-25 mRNA level. These results suggest that hibernation-related HP-25 expression is epigenetically regulated by the binding of HNF-4 to the HP-25 promoter, and that this binding might be modulated by SHP in hibernating chipmunks.
Collapse
Affiliation(s)
| | - Michihiko Ito
- Kitasato University School of Science, Kanagawa 252-0373, Japan
| | | |
Collapse
|
238
|
Fan L, Zong M, Gong R, He D, Li N, Sun LS, Ye Q, Yu S. PADI4 Epigenetically Suppresses p21 Transcription and Inhibits Cell Apoptosis in Fibroblast-like Synoviocytes from Rheumatoid Arthritis Patients. Int J Biol Sci 2017; 13:358-366. [PMID: 28367100 PMCID: PMC5370443 DOI: 10.7150/ijbs.16879] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 12/27/2016] [Indexed: 01/28/2023] Open
Abstract
Rheumatoid arthritis (RA) is characterized by synovial lining hyperplasia, which involves abnormal growth of fibroblast-like synoviocytes (FLSs). This study aimed to investigate the function and molecular mechanism of peptidylarginine deiminase type 4 (PADI4) in FLSs isolated from RA patients (RA-FLSs). FLSs were isolated from RA patients and transfected with small interfering RNAs (siRNAs) or PADI4 overexpression plasmid. FLSs were treated by Adriamycin (ADR) to induce apoptosis, and apoptotic cells were detected by flow cytometry. The expression of PADI4, p53 and p21 was detected by qRT-PCR and Western blot analysis. The recruitment of PADI4 and histone H3 arginine modifications to p21 promoter was measured by chromatin immunoprecipitation. The results showed that knockdown of PADI4 promoted the apoptosis of RA-FLSs and the expression of p53 and p21. Ectopic expression of PADI4 inhibited ADR-induced apoptosis of RA-FLSs, and down-regulated the expression of p53 and p21. In RA-FLSs, global H3 citrullination (CitH3) and H3 arginine 17 methylation levels were dynamically changed by PADI4 and ADR treatment. PADI4 and H3 could bind p21 promoter region to regulate p21 expression. In conclusion, PADI4 contributes to the pathogenesis of RA by protecting FLSs from apoptosis. PADI4 suppresses p21 transcription through altering histone H3 arginine modifications on p21 promoter region. Our study provides new insight into the anti-apoptotic role of PADI4 in RA development.
Collapse
Affiliation(s)
- Lieying Fan
- Department of Clinical Laboratory, Shanghai East Hospital, School of Medicine, Tong Ji University, 150 Ji Mo Road, Shanghai 200120, PR China
| | - Ming Zong
- Department of Clinical Laboratory, Shanghai East Hospital, School of Medicine, Tong Ji University, 150 Ji Mo Road, Shanghai 200120, PR China
| | - Ruhan Gong
- Department of Clinical Laboratory, Shanghai East Hospital, School of Medicine, Tong Ji University, 150 Ji Mo Road, Shanghai 200120, PR China
| | - Dongyi He
- Department of Rheumatology, Guanghua Integrative Medicine Hospital, 540 Xin Hua Road, Shanghai 200052, PR China
| | - Niu Li
- Department of Clinical Laboratory, Shanghai East Hospital, School of Medicine, Tong Ji University, 150 Ji Mo Road, Shanghai 200120, PR China
| | - Li Shan Sun
- Department of Clinical Laboratory, Shanghai East Hospital, School of Medicine, Tong Ji University, 150 Ji Mo Road, Shanghai 200120, PR China
| | - Qin Ye
- Department of Clinical Laboratory, Shanghai East Hospital, School of Medicine, Tong Ji University, 150 Ji Mo Road, Shanghai 200120, PR China
| | - Shanshan Yu
- Department of Clinical Laboratory, Shanghai East Hospital, School of Medicine, Tong Ji University, 150 Ji Mo Road, Shanghai 200120, PR China
| |
Collapse
|
239
|
Wu M, Wei W, Chen J, Cong R, Shi T, Bouvet P, Li J, Wong J, Du JX. Acidic domains differentially read histone H3 lysine 4 methylation status and are widely present in chromatin-associated proteins. SCIENCE CHINA. LIFE SCIENCES 2017; 60:138-151. [PMID: 28194553 DOI: 10.1007/s11427-016-0413-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 01/09/2017] [Indexed: 02/03/2023]
Abstract
Histone methylation is believed to provide binding sites for specific reader proteins, which translate histone code into biological function. Here we show that a family of acidic domain-containing proteins including nucleophosmin (NPM1), pp32, SET/TAF1β, nucleolin (NCL) and upstream binding factor (UBF) are novel H3K4me2-binding proteins. These proteins exhibit a unique pattern of interaction with methylated H3K4, as their binding is stimulated by H3K4me2 and inhibited by H3K4me1 and H3K4me3. These proteins contain one or more acidic domains consisting mainly of aspartic and/or glutamic residues that are necessary for preferential binding of H3K4me2. Furthermore, we demonstrate that the acidic domain with sufficient length alone is capable of binding H3K4me2 in vitro and in vivo. NPM1, NCL and UBF require their acidic domains for association with and transcriptional activation of rDNA genes. Interestingly, by defining acidic domain as a sequence with at least 20 acidic residues in 50 continuous amino acids, we identified 655 acidic domain-containing protein coding genes in the human genome and Gene Ontology (GO) analysis showed that many of the acidic domain proteins have chromatin-related functions. Our data suggest that acidic domain is a novel histone binding motif that can differentially read the status of H3K4 methylation and is broadly present in chromatin-associated proteins.
Collapse
Affiliation(s)
- Meng Wu
- Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Wei Wei
- Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jiwei Chen
- Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Rong Cong
- Université de Lyon, Ecole Normale Supérieure de Lyon, CNRS USR 3010, Laboratoire Joliot-Curie, Lyon, 69364, France
| | - Tieliu Shi
- Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Philippe Bouvet
- Université de Lyon, Ecole Normale Supérieure de Lyon, CNRS USR 3010, Laboratoire Joliot-Curie, Lyon, 69364, France
- Université de Lyon, Centre de Recherche en Cancérologie de Lyon, Cancer Cell Plasticity Department, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, Lyon, France
| | - Jiwen Li
- Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jiemin Wong
- Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - James X Du
- Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
240
|
Chitsazian F, Sadeghi M, Elahi E. Confident gene activity prediction based on single histone modification H2BK5ac in human cell lines. BMC Bioinformatics 2017; 18:67. [PMID: 28122488 PMCID: PMC5264486 DOI: 10.1186/s12859-016-1418-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 12/10/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The histones in the core of nucleosomes may be subject to covalent post-transcriptional modifications. These modifications are thought to correlate with and possibly affect various genomic functions, including transcription. Each modification may alone or in combination with other modifications influence or be influenced by transcription. We aimed to identify correlations between single modifications or combinations of modifications at specific nucleosome sized gene regions with transcription activity based on global histone modification and transcription data of human CD4+ T cells and three other human cell lines. Transcription activity was defined in a binary fashion as either on or off. The analysis was done using the Classification and Regression Tree (CART) data mining protocol, and the Multifactorial Dimensionality Reduction (MDR) method was performed to confirm the CART results. These powerful methods have not previously been used for analysis of histone modification data. RESULTS We showed that analysis of the single histone modification H2BK5ac at only four gene regions correctly predicted transcription activity status of over 75% of genes in CD4+ T-cells. The H2BK5ac modification status also had high power for prediction of gene transcription activity in the three other cell lines studied. The informative gene regions with the H2BK5ac modification were all positioned proximal to transcription initiation sites. The CART and MDR methods were appropriate tools for the analysis performed. In the study, we also developed a non-arbitrary protocol for binary classification of genes as transcriptionally active or inactive. CONCLUSIONS The importance of H2BK5ac modification with regards to transcription control has not previously been emphasized. Analysis of this single modification at only four nucleosome sized gene regions, all of which are at or proximal to transcription initiation, has high power for prediction of gene transcription activity.
Collapse
Affiliation(s)
| | - Mehdi Sadeghi
- National Institute for Genetic Engineering and Biotechnology, Tehran, Iran
| | - Elahe Elahi
- School of Biology, College of Science, University of Tehran, Tehran, Iran. .,Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran.
| |
Collapse
|
241
|
Abstract
Progression of cells through distinct phases of the cell cycle, and transition into out-of-cycling states, such as terminal differentiation and senescence, is accompanied by specific patterns of gene expression. These cell fate decisions are mediated not only by distinct transcription factors, but also chromatin modifiers that establish heritable epigenetic patterns. Lysine methyltransferases (KMTs) that mediate methylation marks on histone and non-histone proteins are now recognized as important regulators of gene expression in cycling and non-cycling cells. Among these, the SUV39 sub-family of KMTs, which includes SUV39H1, SUV39H2, G9a, GLP, SETDB1, and SETDB2, play a prominent role. In this review, we discuss their biochemical properties, sub-cellular localization and function in cell cycle, differentiation programs, and cellular senescence. We also discuss their aberrant expression in cancers, which exhibit de-regulation of cell cycle and differentiation.
Collapse
Affiliation(s)
- Vinay Kumar Rao
- a Department of Physiology , Yong Loo Lin School of Medicine, National University of Singapore , Singapore
| | - Ananya Pal
- a Department of Physiology , Yong Loo Lin School of Medicine, National University of Singapore , Singapore
| | - Reshma Taneja
- a Department of Physiology , Yong Loo Lin School of Medicine, National University of Singapore , Singapore
| |
Collapse
|
242
|
Interplay between epigenetics and metabolism in oncogenesis: mechanisms and therapeutic approaches. Oncogene 2017; 36:3359-3374. [PMID: 28092669 PMCID: PMC5485177 DOI: 10.1038/onc.2016.485] [Citation(s) in RCA: 185] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/07/2016] [Accepted: 11/07/2016] [Indexed: 02/06/2023]
Abstract
Epigenetic and metabolic alterations in cancer cells are highly intertwined. Oncogene-driven metabolic rewiring modifies the epigenetic landscape via modulating the activities of DNA and histone modification enzymes at the metabolite level. Conversely, epigenetic mechanisms regulate the expression of metabolic genes, thereby altering the metabolome. Epigenetic-metabolomic interplay has a critical role in tumourigenesis by coordinately sustaining cell proliferation, metastasis and pluripotency. Understanding the link between epigenetics and metabolism could unravel novel molecular targets, whose intervention may lead to improvements in cancer treatment. In this review, we summarized the recent discoveries linking epigenetics and metabolism and their underlying roles in tumorigenesis; and highlighted the promising molecular targets, with an update on the development of small molecule or biologic inhibitors against these abnormalities in cancer.
Collapse
|
243
|
The Histone Modification Code in the Pathogenesis of Autoimmune Diseases. Mediators Inflamm 2017; 2017:2608605. [PMID: 28127155 PMCID: PMC5239974 DOI: 10.1155/2017/2608605] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/08/2016] [Indexed: 12/19/2022] Open
Abstract
Autoimmune diseases are chronic inflammatory disorders caused by a loss of self-tolerance, which is characterized by the appearance of autoantibodies and/or autoreactive lymphocytes and the impaired suppressive function of regulatory T cells. The pathogenesis of autoimmune diseases is extremely complex and remains largely unknown. Recent advances indicate that environmental factors trigger autoimmune diseases in genetically predisposed individuals. In addition, accumulating results have indicated a potential role of epigenetic mechanisms, such as histone modifications, in the development of autoimmune diseases. Histone modifications regulate the chromatin states and gene transcription without any change in the DNA sequence, possibly resulting in phenotype alteration in several different cell types. In this paper, we discuss the significant roles of histone modifications involved in the pathogenesis of autoimmune diseases, including rheumatoid arthritis, systemic lupus erythematosus, systemic sclerosis, primary biliary cirrhosis, and type 1 diabetes.
Collapse
|
244
|
Peng M, Ying P, Liu X, Li C, Xia R, Li J, Zhao M. Genome-Wide Identification of Histone Modifiers and Their Expression Patterns during Fruit Abscission in Litchi. FRONTIERS IN PLANT SCIENCE 2017; 8:639. [PMID: 28496451 PMCID: PMC5406457 DOI: 10.3389/fpls.2017.00639] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 04/10/2017] [Indexed: 05/07/2023]
Abstract
Modifications to histones, including acetylation and methylation processes, play crucial roles in the regulation of gene expression in plant development as well as in stress responses. However, limited information on the enzymes catalyzing histone acetylation and methylation in non-model plants is currently available. In this study, several histone modifier (HM) types, including six histone acetyltransferases (HATs), 11 histone deacetylases (HDACs), 48 histone methyltransferases (HMTs), and 22 histone demethylases (HDMs), are identified in litchi (Litchi chinensis Sonn. cv. Feizixiao) based on similarities in their sequences to homologs in Arabidopsis (A. thaliana), tomato (Solanum lycopersicum), and rice (Oryza sativa). Phylogenetic analyses reveal that HM enzymes can be grouped into four HAT, two HDAC, two HMT, and two HDM subfamilies, respectively, while further expression profile analyses demonstrate that 17 HMs were significantly altered during fruit abscission in two field treatments. Analyses reveal that these genes exhibit four distinct patterns of expression in response to fruit abscission, while an in vitro assay was used to confirm the HDAC activity of LcHDA2, LcHDA6, and LcSRT2. Our findings are the first in-depth analysis of HMs in the litchi genome, and imply that some are likely to play important roles in fruit abscission in this commercially important plant.
Collapse
Affiliation(s)
- Manjun Peng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, China Litchi Research Center, South China Agricultural UniversityGuangzhou, China
- Guangdong Litchi Engineering Research Center, College of Horticulture, South China Agricultural UniversityGuangzhou, China
| | - Peiyuan Ying
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, China Litchi Research Center, South China Agricultural UniversityGuangzhou, China
- Guangdong Litchi Engineering Research Center, College of Horticulture, South China Agricultural UniversityGuangzhou, China
| | - Xuncheng Liu
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, South China Botanical Garden, Chinese Academy of SciencesGuangzhou, China
| | - Caiqin Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, China Litchi Research Center, South China Agricultural UniversityGuangzhou, China
- Guangdong Litchi Engineering Research Center, College of Horticulture, South China Agricultural UniversityGuangzhou, China
| | - Rui Xia
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, China Litchi Research Center, South China Agricultural UniversityGuangzhou, China
- Guangdong Litchi Engineering Research Center, College of Horticulture, South China Agricultural UniversityGuangzhou, China
| | - Jianguo Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, China Litchi Research Center, South China Agricultural UniversityGuangzhou, China
- Guangdong Litchi Engineering Research Center, College of Horticulture, South China Agricultural UniversityGuangzhou, China
- *Correspondence: Jianguo Li
| | - Minglei Zhao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, China Litchi Research Center, South China Agricultural UniversityGuangzhou, China
- Guangdong Litchi Engineering Research Center, College of Horticulture, South China Agricultural UniversityGuangzhou, China
- Minglei Zhao
| |
Collapse
|
245
|
Abstract
Recent studies from a number of model organisms have indicated chromatin structure and its remodeling as a major contributory agent for aging. Few recent experiments also demonstrate that modulation in the chromatin modifying agents also affect the life span of an organism and even in some cases the change is inherited epigenetically to subsequent generations. Hence, in the present report we discuss the chromatin organization and its changes during aging.
Collapse
Affiliation(s)
- Pramod C. Rath
- School of Life Sciences, Molecular Biology Laboratory, Jawaharlal Nehru University, New Delhi, Delhi India
| | - Ramesh Sharma
- Department of Biochemistry, North Eastern Hill University, Shillong, Megalaya India
| | - S. Prasad
- Biochemistry & Molecular Biology Lab, Department of Zoology, Banaras Hindu University, Varanasi, Uttar Pradesh India
| |
Collapse
|
246
|
Sgarbieri VC, Pacheco MTB. Human development: from conception to maturity. BRAZILIAN JOURNAL OF FOOD TECHNOLOGY 2017. [DOI: 10.1590/1981-6723.16116] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Abstract The main objective of this review was to describe and emphasize the care that a woman must have in the period prior to pregnancy, as well as throughout pregnancy and after the birth of the baby, cares and duties that should continue to be followed by mother and child throughout the first years of the child’s life. Such cares are of nutritional, behavioral and lifestyle natures, and also involve the father and the whole family. Human development, from conception to maturity, consists of a critical and important period due to the multitude of intrinsic genetic and environmental factors that influence, positively or negatively, the person's entire life. The human being, who originated and passed his/her first phase of development in the womb, receives influence from different factors: a) of parental origin (father and mother), including health and lifestyle of the father and mother, genetic inheritance, nutrition of the mother prior to and during pregnancy; b) events that affected the mother and hence the child under development in intrauterine life, at birth (delivery), during perinatal period, and throughout the early years of life. The fragility of development continues throughout the preschool, school and adolescent periods during which proper nutrition with a balanced lifestyle is essential and depends on guidance from the parents, caregivers and teachers.
Collapse
|
247
|
Zawadzka M, Jagodziński PP. Exercise-induced epigenetic regulations in inflammatory related cells. J Appl Biomed 2017. [DOI: 10.1016/j.jab.2016.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
248
|
Zhang C, Liu Y. Retrieving Quantitative Information of Histone PTMs by Mass Spectrometry. Methods Enzymol 2016; 586:165-191. [PMID: 28137562 DOI: 10.1016/bs.mie.2016.10.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Posttranslational modifications (PTMs) of histones are one of the main research interests in the rapidly growing field of epigenetics. Accurate and precise quantification of these highly complex histone PTMs is critical for understanding the histone code and the biological significance behind it. It nonetheless remains a major analytical challenge. Mass spectrometry (MS) has been proven as a robust tool in retrieving quantitative information of histone PTMs, and a variety of MS-based quantitative strategies have been successfully developed and employed in basic research as well as clinical studies. In this chapter, we provide an overview for quantitative analysis of histone PTMs, often highly flexible and case dependent, as a primer for future experimental designs.
Collapse
Affiliation(s)
- C Zhang
- Baylor College of Medicine, Houston, TX, United States.
| | - Y Liu
- University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
249
|
Wang C, Kubiak LJ, Du LM, Li WJ, Jian ZY, Tang C, Fan ZX, Zhang XY, Yue BS. Comparison of microsatellite distribution in genomes of Centruroides exilicauda and Mesobuthus martensii. Gene 2016; 594:41-46. [DOI: 10.1016/j.gene.2016.08.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 08/13/2016] [Accepted: 08/28/2016] [Indexed: 10/21/2022]
|
250
|
Perrella G, Kaiserli E. Light behind the curtain: photoregulation of nuclear architecture and chromatin dynamics in plants. THE NEW PHYTOLOGIST 2016; 212:908-919. [PMID: 27813089 PMCID: PMC5111779 DOI: 10.1111/nph.14269] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 09/14/2016] [Indexed: 05/24/2023]
Abstract
Light is a powerful stimulus regulating many aspects of plant development and phenotypic plasticity. Plants sense light through the action of specialized photoreceptor protein families that absorb different wavelengths and intensities of light. Recent discoveries in the area of photobiology have uncovered photoreversible changes in nuclear organization correlated with transcriptional regulation patterns that lead to de-etiolation and photoacclimation. Novel signalling components bridging photoreceptor activation with chromatin remodelling and regulation of gene expression have been discovered. Moreover, coregulated gene loci have been shown to relocate to the nuclear periphery in response to light. The study of photoinduced changes in nuclear architecture is a flourishing area leading to major discoveries that will allow us to better understand how highly conserved mechanisms underlying genomic reprogramming are triggered by environmental and endogenous stimuli. This review aims to discuss fundamental and innovative reports demonstrating how light triggers changes in chromatin and nuclear architecture during photomorphogenesis.
Collapse
Affiliation(s)
- Giorgio Perrella
- Institute of Molecular, Cell and Systems BiologyCollege of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowG12 8QQUK
| | - Eirini Kaiserli
- Institute of Molecular, Cell and Systems BiologyCollege of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowG12 8QQUK
| |
Collapse
|