1
|
Sánchez-Ramírez E, Ung TPL, Stringari C, Aguilar-Arnal L. Emerging Functional Connections Between Metabolism and Epigenetic Remodeling in Neural Differentiation. Mol Neurobiol 2024; 61:6688-6707. [PMID: 38340204 PMCID: PMC11339152 DOI: 10.1007/s12035-024-04006-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
Stem cells possess extraordinary capacities for self-renewal and differentiation, making them highly valuable in regenerative medicine. Among these, neural stem cells (NSCs) play a fundamental role in neural development and repair processes. NSC characteristics and fate are intricately regulated by the microenvironment and intracellular signaling. Interestingly, metabolism plays a pivotal role in orchestrating the epigenome dynamics during neural differentiation, facilitating the transition from undifferentiated NSC to specialized neuronal and glial cell types. This intricate interplay between metabolism and the epigenome is essential for precisely regulating gene expression patterns and ensuring proper neural development. This review highlights the mechanisms behind metabolic regulation of NSC fate and their connections with epigenetic regulation to shape transcriptional programs of stemness and neural differentiation. A comprehensive understanding of these molecular gears appears fundamental for translational applications in regenerative medicine and personalized therapies for neurological conditions.
Collapse
Affiliation(s)
- Edgar Sánchez-Ramírez
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Thi Phuong Lien Ung
- Laboratory for Optics and Biosciences, Ecole Polytechnique, CNRS, INSERM, Institut Polytechnique de Paris, Palaiseau, France
| | - Chiara Stringari
- Laboratory for Optics and Biosciences, Ecole Polytechnique, CNRS, INSERM, Institut Polytechnique de Paris, Palaiseau, France
| | - Lorena Aguilar-Arnal
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
2
|
Fan Y, Sun C, Yan K, Li P, Hein I, Gilroy EM, Kear P, Bi Z, Yao P, Liu Z, Liu Y, Bai J. Recent Advances in Studies of Genomic DNA Methylation and Its Involvement in Regulating Drought Stress Response in Crops. PLANTS (BASEL, SWITZERLAND) 2024; 13:1400. [PMID: 38794470 PMCID: PMC11125032 DOI: 10.3390/plants13101400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 05/10/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024]
Abstract
As global arid conditions worsen and groundwater resources diminish, drought stress has emerged as a critical impediment to plant growth and development globally, notably causing declines in crop yields and even the extinction of certain cultivated species. Numerous studies on drought resistance have demonstrated that DNA methylation dynamically interacts with plant responses to drought stress by modulating gene expression and developmental processes. However, the precise mechanisms underlying these interactions remain elusive. This article consolidates the latest research on the role of DNA methylation in plant responses to drought stress across various species, focusing on methods of methylation detection, mechanisms of methylation pattern alteration (including DNA de novo methylation, DNA maintenance methylation, and DNA demethylation), and overall responses to drought conditions. While many studies have observed significant shifts in genome-wide or gene promoter methylation levels in drought-stressed plants, the identification of specific genes and pathways involved remains limited. This review aims to furnish a reference for detailed research into plant responses to drought stress through epigenetic approaches, striving to identify drought resistance genes regulated by DNA methylation, specific signaling pathways, and their molecular mechanisms of action.
Collapse
Affiliation(s)
- Youfang Fan
- State Key Laboratory of Aridland Crop Science, College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China; (Y.F.); (P.L.); (Z.B.); (P.Y.); (Z.L.); (Y.L.)
| | - Chao Sun
- State Key Laboratory of Aridland Crop Science, College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China; (Y.F.); (P.L.); (Z.B.); (P.Y.); (Z.L.); (Y.L.)
| | - Kan Yan
- School of Biological and Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou 730070, China;
| | - Pengcheng Li
- State Key Laboratory of Aridland Crop Science, College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China; (Y.F.); (P.L.); (Z.B.); (P.Y.); (Z.L.); (Y.L.)
| | - Ingo Hein
- The James Hutton Institute, Dundee DD2 5DA, UK; (I.H.); (E.M.G.)
| | | | - Philip Kear
- International Potato Center (CIP), CIP China Center for Asia Pacific (CCCAP), Beijing 102199, China;
| | - Zhenzhen Bi
- State Key Laboratory of Aridland Crop Science, College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China; (Y.F.); (P.L.); (Z.B.); (P.Y.); (Z.L.); (Y.L.)
| | - Panfeng Yao
- State Key Laboratory of Aridland Crop Science, College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China; (Y.F.); (P.L.); (Z.B.); (P.Y.); (Z.L.); (Y.L.)
| | - Zhen Liu
- State Key Laboratory of Aridland Crop Science, College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China; (Y.F.); (P.L.); (Z.B.); (P.Y.); (Z.L.); (Y.L.)
| | - Yuhui Liu
- State Key Laboratory of Aridland Crop Science, College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China; (Y.F.); (P.L.); (Z.B.); (P.Y.); (Z.L.); (Y.L.)
| | - Jiangping Bai
- State Key Laboratory of Aridland Crop Science, College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China; (Y.F.); (P.L.); (Z.B.); (P.Y.); (Z.L.); (Y.L.)
| |
Collapse
|
3
|
Yano N, Fedulov AV. Targeted DNA Demethylation: Vectors, Effectors and Perspectives. Biomedicines 2023; 11:biomedicines11051334. [PMID: 37239005 DOI: 10.3390/biomedicines11051334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/21/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Aberrant DNA hypermethylation at regulatory cis-elements of particular genes is seen in a plethora of pathological conditions including cardiovascular, neurological, immunological, gastrointestinal and renal diseases, as well as in cancer, diabetes and others. Thus, approaches for experimental and therapeutic DNA demethylation have a great potential to demonstrate mechanistic importance, and even causality of epigenetic alterations, and may open novel avenues to epigenetic cures. However, existing methods based on DNA methyltransferase inhibitors that elicit genome-wide demethylation are not suitable for treatment of diseases with specific epimutations and provide a limited experimental value. Therefore, gene-specific epigenetic editing is a critical approach for epigenetic re-activation of silenced genes. Site-specific demethylation can be achieved by utilizing sequence-dependent DNA-binding molecules such as zinc finger protein array (ZFA), transcription activator-like effector (TALE) and clustered regularly interspaced short palindromic repeat-associated dead Cas9 (CRISPR/dCas9). Synthetic proteins, where these DNA-binding domains are fused with the DNA demethylases such as ten-eleven translocation (Tet) and thymine DNA glycosylase (TDG) enzymes, successfully induced or enhanced transcriptional responsiveness at targeted loci. However, a number of challenges, including the dependence on transgenesis for delivery of the fusion constructs, remain issues to be solved. In this review, we detail current and potential approaches to gene-specific DNA demethylation as a novel epigenetic editing-based therapeutic strategy.
Collapse
Affiliation(s)
- Naohiro Yano
- Department of Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
| | - Alexey V Fedulov
- Department of Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
| |
Collapse
|
4
|
Chandramouly G. Gadd45 in DNA Demethylation and DNA Repair. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1360:55-67. [PMID: 35505162 DOI: 10.1007/978-3-030-94804-7_4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Growth arrest and DNA damage 45 (Gadd45) family genes, Gadd45A, Gadd45B, and GADD45 G are implicated as stress sensors that are rapidly induced upon genotoxic/physiological stress. They are involved in regulation of various cellular functions such as DNA repair, senescence, and cell cycle control. Gadd45 family of genes serve as tumor suppressors in response to different stimuli and defects in Gadd45 pathway can give rise to oncogenesis. More recently, Gadd45 has been shown to promote gene activation by demethylation and this function is important for transcriptional regulation and differentiation during development. Gadd45 serves as an adaptor for DNA repair factors to promote removal of 5-methylcytosine from DNA at gene specific loci. Therefore, Gadd45 serves as a powerful link between DNA repair and epigenetic gene regulation.
Collapse
Affiliation(s)
- Gurushankar Chandramouly
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
5
|
In Response to Abiotic Stress, DNA Methylation Confers EpiGenetic Changes in Plants. PLANTS 2021; 10:plants10061096. [PMID: 34070712 PMCID: PMC8227271 DOI: 10.3390/plants10061096] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
Epigenetics involves the heritable changes in patterns of gene expression determined by developmental and abiotic stresses, i.e., drought, cold, salinity, trace metals, and heat. Gene expression is driven by changes in DNA bases, histone proteins, the biogenesis of ncRNA, and changes in the nucleotide sequence. To cope with abiotic stresses, plants adopt certain changes driven by a sophisticated biological system. DNA methylation is a primary mechanism for epigenetic variation, which can induce phenotypic alterations in plants under stress. Some of the stress-driven changes in plants are temporary, while some modifications may be stable and inheritable to the next generations to allow them to cope with such extreme stress challenges in the future. In this review, we discuss the pivotal role of epigenetically developed phenotypic characteristics in plants as an evolutionary process participating in adaptation and tolerance responses to abiotic and biotic stresses that alter their growth and development. We emphasize the molecular process underlying changes in DNA methylation, differential variation for different species, the roles of non-coding RNAs in epigenetic modification, techniques for studying DNA methylation, and its role in crop improvement in tolerance to abiotic stress (drought, salinity, and heat). We summarize DNA methylation as a significant future research priority for tailoring crops according to various challenging environmental issues.
Collapse
|
6
|
do Amaral MN, Auler PA, Rossatto T, Barros PM, Oliveira MM, Braga EJB. Long-term somatic memory of salinity unveiled from physiological, biochemical and epigenetic responses in two contrasting rice genotypes. PHYSIOLOGIA PLANTARUM 2020; 170:248-268. [PMID: 32515828 DOI: 10.1111/ppl.13149] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/30/2020] [Accepted: 06/04/2020] [Indexed: 06/11/2023]
Abstract
Plants are constantly exposed to environmental fluctuations, that may occur in a single day or over longer periods. In many cases, abiotic stresses are transient and recurrent, impacting how plants respond in subsequent adverse conditions. Adaptation mechanisms may occur at the physiological, biochemical and molecular level, modifying transcriptional response, regulatory proteins, epigenetic marks or metabolites. Here, we aimed to uncover the different strategies that rice uses to respond to recurrent stress. We tested varieties with contrasting behavior towards salinity (tolerance or sensitivity) and imposed salt stress (150 mM NaCl) during 48 h at vegetative and/or reproductive stages. After 48 h of stress in reproductive stage, leaves and roots were harvested separately or otherwise the plants were submitted to a 24 h recovery, prior to sample harvesting. Plants submitted to a recurrent stress responded differently from those suffering a single stress event. In the case of the sensitive genotype, recurrent stress led to lower Na/K ratio in roots and lower hydrogen peroxide accumulation and lipid peroxidation in leaves, but maintenance of global DNA methylation levels. In the tolerant genotype, recurrent stress did neither affect the Na/K ratio nor the stomatal conductance, although the levels of superoxide anion and hydrogen peroxide accumulation were lower, as also observed for global levels of DNA methylation. Our work shows that a short pre-exposure to salt stress may improve rice tolerance to subsequent stress, trough biochemical, physiological and epigenetic processes, with more significant changes visible in the tolerant genotype.
Collapse
Affiliation(s)
| | - Priscila Ariane Auler
- Department of Botany, Biology Institute, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Tatiana Rossatto
- Department of Botany, Biology Institute, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Pedro M Barros
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Genomics of Plant Stress - Plant Functional Genomics Lab, Av. da República, Oeiras, 2780-157, Portugal
| | - Maria Margarida Oliveira
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Genomics of Plant Stress - Plant Functional Genomics Lab, Av. da República, Oeiras, 2780-157, Portugal
| | | |
Collapse
|
7
|
Jiang Z, Lai Y, Beaver JM, Tsegay PS, Zhao ML, Horton JK, Zamora M, Rein HL, Miralles F, Shaver M, Hutcheson JD, Agoulnik I, Wilson SH, Liu Y. Oxidative DNA Damage Modulates DNA Methylation Pattern in Human Breast Cancer 1 (BRCA1) Gene via the Crosstalk between DNA Polymerase β and a de novo DNA Methyltransferase. Cells 2020; 9:E225. [PMID: 31963223 PMCID: PMC7016758 DOI: 10.3390/cells9010225] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/15/2019] [Accepted: 01/13/2020] [Indexed: 12/13/2022] Open
Abstract
DNA damage and base excision repair (BER) are actively involved in the modulation of DNA methylation and demethylation. However, the underlying molecular mechanisms remain unclear. In this study, we seek to understand the mechanisms by exploring the effects of oxidative DNA damage on the DNA methylation pattern of the tumor suppressor breast cancer 1 (BRCA1) gene in the human embryonic kidney (HEK) HEK293H cells. We found that oxidative DNA damage simultaneously induced DNA demethylation and generation of new methylation sites at the CpGs located at the promoter and transcribed regions of the gene ranging from -189 to +27 in human cells. We demonstrated that DNA damage-induced demethylation was mediated by nucleotide misincorporation by DNA polymerase β (pol β). Surprisingly, we found that the generation of new DNA methylation sites was mediated by coordination between pol β and the de novo DNA methyltransferase, DNA methyltransferase 3b (DNMT3b), through the interaction between the two enzymes in the promoter and encoding regions of the BRCA1 gene. Our study provides the first evidence that oxidative DNA damage can cause dynamic changes in DNA methylation in the BRCA1 gene through the crosstalk between BER and de novo DNA methylation.
Collapse
Affiliation(s)
- Zhongliang Jiang
- Biochemistry Ph.D. Program, Florida International University, Miami, FL 33199, USA; (Z.J.); (J.M.B.); (P.S.T.)
| | - Yanhao Lai
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL 33199, USA; (Y.L.); (M.Z.); (H.L.R.); (F.M.)
| | - Jill M. Beaver
- Biochemistry Ph.D. Program, Florida International University, Miami, FL 33199, USA; (Z.J.); (J.M.B.); (P.S.T.)
| | - Pawlos S. Tsegay
- Biochemistry Ph.D. Program, Florida International University, Miami, FL 33199, USA; (Z.J.); (J.M.B.); (P.S.T.)
| | - Ming-Lang Zhao
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (M.-L.Z.); (J.K.H.); (S.H.W.)
| | - Julie K. Horton
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (M.-L.Z.); (J.K.H.); (S.H.W.)
| | - Marco Zamora
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL 33199, USA; (Y.L.); (M.Z.); (H.L.R.); (F.M.)
| | - Hayley L. Rein
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL 33199, USA; (Y.L.); (M.Z.); (H.L.R.); (F.M.)
| | - Frank Miralles
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL 33199, USA; (Y.L.); (M.Z.); (H.L.R.); (F.M.)
| | - Mohammad Shaver
- Department of Biomedical Engineering, Florida International University, Miami, FL 33199, USA; (M.S.); (J.D.H.)
| | - Joshua D. Hutcheson
- Department of Biomedical Engineering, Florida International University, Miami, FL 33199, USA; (M.S.); (J.D.H.)
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, USA;
| | - Irina Agoulnik
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, USA;
- Department of Human and Molecular Genetics, Florida International University, Miami, FL 33199, USA
| | - Samuel H. Wilson
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (M.-L.Z.); (J.K.H.); (S.H.W.)
| | - Yuan Liu
- Biochemistry Ph.D. Program, Florida International University, Miami, FL 33199, USA; (Z.J.); (J.M.B.); (P.S.T.)
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL 33199, USA; (Y.L.); (M.Z.); (H.L.R.); (F.M.)
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, USA;
| |
Collapse
|
8
|
Li LT, Wang X, Zhu WT, Qian GW, Pei DS, Zheng JN. Reciprocal Role Of DNA Methylation And Sp1 Binding In Ki-67 Gene Transcription. Cancer Manag Res 2019; 11:9749-9759. [PMID: 31819613 PMCID: PMC6874502 DOI: 10.2147/cmar.s213769] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 10/16/2019] [Indexed: 01/26/2023] Open
Abstract
Purpose DNA methylation plays major regulatory roles in gene transcription. Our previous studies confirmed that Ki-67 promoter is hypomethylated and Sp1 is a transcriptional activator of Ki-67 gene in cancer cells. However, whether Sp1-mediated transcriptional activation of Ki-67 is related to its methylation has not been studied yet. Materials and methods In this study, we confirmed that methylated CpG binding protein 2 (MBD2) binding to methylated DNA hindered the binding of Sp1 to Ki-67 promoter and then repressed Ki-67 transcription through chromatin immunoprecipitation (ChIP) and quantitative real-time PCR (qRT-PCR). Co-immunoprecipitation (Co-IP), ChIP, methylation-specific PCR (MS-PCR) and Western blot were utilized to analyze the effects of Sp1 binding to Ki-67 promoter on its methylation status. Results Less DNA methyltransferase 1 (DNMT1) bound to the Ki-67 promoter in MKN45 cells than in HK-2 cells. Histone acetyltransferase p300 that was recruited by Sp1 to Ki-67 promoter could attenuate the methylation level of Ki-67 promoter. Furthermore, higher expression of Sp1 and Ki-67 was related to the overall survival (OS), first progression (FP) and post-progression survival (PPS) in gastric cancer by scrutinizing bioinformatics datasets. Conclusion Taken together, our findings suggested that hypomethylation of Ki-67 promoter enhanced the binding of Sp1, which in turn maintained hypomethylation of promoter, leading to increase Ki-67 expression in cancer cells. Sp1 and Ki-67 could act promising prognostic biomarkers for clinical diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Lian-Tao Li
- Cancer Institute, Xuzhou Medical University, Xuzhou 221000, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, People's Republic of China.,Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, People's Republic of China
| | - Xun Wang
- Department of Interventional Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, People's Republic of China
| | - Wen-Tao Zhu
- Department of Pathology, Xuzhou Medical University, Xuzhou 221000, People's Republic of China
| | - Guo-Wei Qian
- Department of Medical Oncology, Shanghai Sixth People's Hospital, Shanghai 200000, People's Republic of China
| | - Dong-Sheng Pei
- Cancer Institute, Xuzhou Medical University, Xuzhou 221000, People's Republic of China.,Department of Pathology, Xuzhou Medical University, Xuzhou 221000, People's Republic of China
| | - Jun-Nian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou 221000, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, People's Republic of China
| |
Collapse
|
9
|
He ZH, Chen Y, Chen P, He SD, Zeng HH, Ye JR, Liu D, Cao J. 5-Aza-2'-deoxycytidine protects against emphysema in mice via suppressing p16 Ink4a expression in lung tissue. Int J Chron Obstruct Pulmon Dis 2017; 12:3149-3158. [PMID: 29133977 PMCID: PMC5669795 DOI: 10.2147/copd.s131090] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background There is a growing realization that COPD, or at least emphysema, involves several processes presenting in aging and cellular senescence. Endothelial progenitor cells (EPCs) contribute to neovascularization and play an important role in the development of COPD. The gene for p16Ink4a is a major dominant senescence one. The aim of the present study was to observe changes in lung function, histomorphology of lung tissue, and expression of p16Ink4a in lung tissue and bone marrow-derived EPCs in emphysematous mice induced by cigarette-smoke extract (CSE), and further to search for a potential candidate agent protecting against emphysema induced by CSE. Materials and methods An animal emphysema model was induced by intraperitoneal injection of CSE. 5-Aza-2′-deoxycytidine (5-Aza-CdR) was administered to the emphysematous mice. Lung function and histomorphology of lung tissue were measured. The p16Ink4a protein and mRNA in EPCs and lung tissues were detected using Western blotting and quantitative reverse-transcription polymerase chain reaction, respectively. Results CSE induced emphysema with increased p16Ink4a expression in lung tissue and bone marrow-derived EPCs. 5-Aza-CdR partly protected against emphysema, especially in the lung-morphology profile, and partly protest against the overexpression of p16Ink4a in EPCs and lung tissue induced by CSE. Conclusion 5-Aza-CdR partly protected against emphysema in mice via suppressing p16Ink4a expression in EPCs and lung tissue.
Collapse
Affiliation(s)
| | - Yan Chen
- Department of Respiratory Medicine, Second Xiangya Hospital, Central South University, Changsha
| | - Ping Chen
- Department of Respiratory Medicine, Second Xiangya Hospital, Central South University, Changsha
| | - Sheng-Dong He
- Department of Respiratory Medicine, Second Xiangya Hospital, Central South University, Changsha
| | - Hui-Hui Zeng
- Department of Respiratory Medicine, Second Xiangya Hospital, Central South University, Changsha
| | - Ji-Ru Ye
- Department of Respiratory Medicine, Second Xiangya Hospital, Central South University, Changsha
| | - Da Liu
- Department of Respiratory Medicine, Second Xiangya Hospital, Central South University, Changsha
| | - Jun Cao
- Department of Respiratory Medicine, Hunan Provincial People's Hospital, Changsha, China
| |
Collapse
|
10
|
Gianoglio S, Moglia A, Acquadro A, Comino C, Portis E. The genome-wide identification and transcriptional levels of DNA methyltransferases and demethylases in globe artichoke. PLoS One 2017; 12:e0181669. [PMID: 28746368 PMCID: PMC5529103 DOI: 10.1371/journal.pone.0181669] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/05/2017] [Indexed: 11/19/2022] Open
Abstract
Changes to the cytosine methylation status of DNA, driven by the activity of C5 methyltransferases (C5-MTases) and demethylases, exert an important influence over development, transposon movement, gene expression and imprinting. Three groups of C5-MTase enzymes have been identified in plants, namely MET (methyltransferase 1), CMT (chromomethyltransferases) and DRM (domains rearranged methyltransferases). Here the repertoire of genes encoding C5-MTase and demethylase by the globe artichoke (Cynara cardunculus var. scolymus) is described, based on sequence homology, a phylogenetic analysis and a characterization of their functional domains. A total of ten genes encoding C5-MTase (one MET, five CMTs and four DRMs) and five demethylases was identified. An analysis of their predicted product's protein structure suggested an extensive level of conservation has been retained by the C5-MTases. Transcriptional profiling based on quantitative real time PCR revealed a number of differences between the genes encoding maintenance and de novo methyltransferases, sometimes in a tissue- or development-dependent manner, which implied a degree of functional specialization.
Collapse
Affiliation(s)
- Silvia Gianoglio
- Department of Agricultural, Forest and Food Sciences, University of Torino, Grugliasco, Italy
| | - Andrea Moglia
- Department of Agricultural, Forest and Food Sciences, University of Torino, Grugliasco, Italy
| | - Alberto Acquadro
- Department of Agricultural, Forest and Food Sciences, University of Torino, Grugliasco, Italy
| | - Cinzia Comino
- Department of Agricultural, Forest and Food Sciences, University of Torino, Grugliasco, Italy
| | - Ezio Portis
- Department of Agricultural, Forest and Food Sciences, University of Torino, Grugliasco, Italy
| |
Collapse
|
11
|
He Z, Chen Y, Hou C, He W, Chen P. Cigarette Smoke Extract Changes Expression of Endothelial Nitric Oxide Synthase (eNOS) and p16(INK4a) and is Related to Endothelial Progenitor Cell Dysfunction. Med Sci Monit 2017; 23:3224-3231. [PMID: 28668968 PMCID: PMC5507800 DOI: 10.12659/msm.902746] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Endothelial dysfunction is an important pathophysiologic feature in many smoke-related diseases. Endothelial progenitor cells (EPCs) are the precursors of endothelial cells and play a fundamental role in the maintenance of endothelial integrity and function. Endothelial nitric oxide synthase (eNOS) is the dominant NOS isoform in the vasculature and plays a central role in the maintenance of endothelial homeostasis. p16(INK4a) is a cyclin-dependent kinase inhibitor and could be regarded as a major dominant senescence gene. The present study aimed to determine whether the expression of eNOS and p16(INK4a) in EPCs is related to EPCs function and the possible epigenetic mechanism, if any. MATERIAL AND METHODS We investigated EPCs capacity for proliferation, adhesion, and secretion, and the expression of eNOS and p16(INK4a) in EPCs which were altered by cigarette smoke extract (CSE) in vitro. Furthermore, Decitabine (Dec), an agent of demethylation, was used to examine whether it could alter the changes induced by CSE. RESULTS The present study demonstrated that EPCs altered by CSE in vitro displayed decreased capacities of proliferation, adhesion, and secretion, which was accompanied by decreased eNOS expression and increased p16(INK4a) expression in EPCs. Furthermore, Dec could alleviate the changes in the expression of eNOS and p16(INK4a), and protect against the EPCs dysfunction caused by CSE. CONCLUSIONS The decreased eNOS expression and increased p16(INK4a) expression was associated with dysfunction of EPCs caused by CSE. The mechanism of methylation, one of the most common epigenetic mechanism, may be involved in the EPCs dysfunction caused by CSE.
Collapse
Affiliation(s)
- Zhihui He
- Department of Intensive Care Unit, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Yan Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Can Hou
- Department of Intensive Care Unit, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Wenfang He
- Department of Intensive Care Unit, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Ping Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
12
|
Furlan D, Trapani D, Berrino E, Debernardi C, Panero M, Libera L, Sahnane N, Riva C, Tibiletti MG, Sessa F, Sapino A, Venesio T. Oxidative DNA damage induces hypomethylation in a compromised base excision repair colorectal tumourigenesis. Br J Cancer 2017; 116:793-801. [PMID: 28141798 PMCID: PMC5355935 DOI: 10.1038/bjc.2017.9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/23/2016] [Accepted: 01/05/2017] [Indexed: 12/12/2022] Open
Abstract
Background: A compromised base excision repair (BER) promotes carcinogenesis by accumulating oxidative DNA-damaged products as observed in MUTYH-associated polyposis, a hereditary colorectal cancer syndrome marked by adenomas and cancers with an accumulation of 8-oxoguanine. Remarkably, DNA global demethylation has been shown to be mediated by BER, suggesting a relevant interplay with early colorectal tumourigenesis. To check this hypothesis, we investigated a cohort of 49 adenomas and 10 carcinomas, derived from 17 MUTYH-associated polyposis patients; as adenoma controls, we used a set of 36 familial adenomatous polyposis and 24 sporadic polyps. Methods: Samples were analysed for their mutational and epigenetic status, measured as global LINE-1 (long interspersed nuclear element) and gene-specific LINE-1 MET methylation by mass spectrometry and pyrosequencing. Results: MUTYH-associated polyposis adenomas were strikingly more hypomethylated than familial adenomatous and sporadic polyps for both DNA demethylation markers (P=0.032 and P=0.007 for LINE-1; P=0.004 and P<0.0001 for LINE-1 MET, respectively) with levels comparable to those of the carcinomas derived from the same patients. They also had mutations due mainly to KRAS/NRAS p.G12C, which was absent in the controls (P<0.0001 for both sets). Conclusions: Our results show that DNA demethylation, together with specific KRAS/NRAS mutations, drives the early steps of oxidative damage colorectal tumourigenesis.
Collapse
Affiliation(s)
- Daniela Furlan
- Anatomic Pathology Unit, Department of Surgical and Morphological Sciences, University of Insubria, Varese 21100, Italy.,Research Center for the Study of Hereditary and Familial Tumors, University of Insubria, Varese 21100, Italy
| | - Davide Trapani
- Anatomic Pathology Unit, Department of Surgical and Morphological Sciences, University of Insubria, Varese 21100, Italy
| | - Enrico Berrino
- Molecular Pathology Laboratory, Unit of Pathology, Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, Candiolo (Torino) 10060, Italy
| | - Carla Debernardi
- Molecular Pathology Laboratory, Unit of Pathology, Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, Candiolo (Torino) 10060, Italy
| | - Mara Panero
- Molecular Pathology Laboratory, Unit of Pathology, Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, Candiolo (Torino) 10060, Italy
| | - Laura Libera
- Anatomic Pathology Unit, Department of Surgical and Morphological Sciences, University of Insubria, Varese 21100, Italy
| | - Nora Sahnane
- Anatomic Pathology Unit, Department of Surgical and Morphological Sciences, University of Insubria, Varese 21100, Italy
| | - Cristina Riva
- Anatomic Pathology Unit, Department of Surgical and Morphological Sciences, University of Insubria, Varese 21100, Italy.,Research Center for the Study of Hereditary and Familial Tumors, University of Insubria, Varese 21100, Italy
| | - Maria Grazia Tibiletti
- Anatomic Pathology Unit, Department of Surgical and Morphological Sciences, University of Insubria, Varese 21100, Italy
| | - Fausto Sessa
- Anatomic Pathology Unit, Department of Surgical and Morphological Sciences, University of Insubria, Varese 21100, Italy.,Research Center for the Study of Hereditary and Familial Tumors, University of Insubria, Varese 21100, Italy
| | - Anna Sapino
- Molecular Pathology Laboratory, Unit of Pathology, Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, Candiolo (Torino) 10060, Italy
| | - Tiziana Venesio
- Molecular Pathology Laboratory, Unit of Pathology, Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, Candiolo (Torino) 10060, Italy
| |
Collapse
|
13
|
Wang ZL, Gao S, Li L, Li XY, Huan SL, Wang K. Demethylation of tumor necrosis factor-α converting enzyme predicts poor prognosis in acute-on-chronic hepatitis B liver failure. Clin Res Hepatol Gastroenterol 2016; 40:457-64. [PMID: 26850359 DOI: 10.1016/j.clinre.2015.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 12/03/2015] [Accepted: 12/11/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND OBJECTIVE Tumor necrosis factor-α converting enzyme (TACE) has been demonstrated to be involved in liver inflammation. However, the significance of TACE methylation in acute-on-chronic hepatitis B liver failure (ACHBLF) has not been demonstrated. This study aims to evaluate TACE methylation status in ACHBLF and determine its predictive value for prognosis. METHODS Forty-five patients with ACHBLF, 80 with chronic hepatitis B (CHB) and 54 healthy controls (HCs) were enrolled. The methylation status of TACE promoter was determined by methylation-specific polymerase chain reaction. The TACE mRNA expression was determined by quantitative real-time polymerase chain reaction. The plasma levels of TACE, TNF-α, sTNFRI, sTNFRII were measured by enzyme-linked immunosorbent assay. RESULTS TACE methylation was significantly lower in patients with ACHBLF than those with CHB (χ(2)=24.69, P<0.01) and HCs (χ(2)=35.93, P<0.01). Meanwhile, TACE methylation was significantly lower in CHB patients than HCs (χ(2)=4.03, P<0.05). TACE methylation was significantly inversely associated with its mRNA expression (r=-0.68; P<0.01). The plasma levels of TACE, TNF-α, sTNFRI, sTNFRII were significantly higher in patients with ACHBLF than those with CHB (P<0.05, respectively) and HCs (P<0.05, respectively). In patients with ACHBLF, significantly higher prothrombin activity, lower total bilirubin and MELD score were found in TACE methylated group than unmethylated group (P<0.05). ACHBLF patients with methylated TACE showed significantly better survival than those without (P<0.01). CONCLUSION This study showed that demethylation of TACE promoter occurred in ACHBLF and might serve as a potential prognostic marker.
Collapse
Affiliation(s)
- Zhen-Li Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Institute of Shandong University, Wenhuaxi Road 107#, Shandong, Jinan 250012, China
| | - Shuai Gao
- Department of Hepatology, Qilu Hospital of Shandong University, Institute of Shandong University, Wenhuaxi Road 107#, Shandong, Jinan 250012, China
| | - Lei Li
- Department of Infectious Diseases, Qingdao Women and Children Hospital, Qingdao 266034 Shandong, China
| | - Xin-You Li
- Department of Hepatology, Qilu Hospital of Shandong University, Institute of Shandong University, Wenhuaxi Road 107#, Shandong, Jinan 250012, China
| | - Shu-Ling Huan
- Department of Hepatology, Qilu Hospital of Shandong University, Institute of Shandong University, Wenhuaxi Road 107#, Shandong, Jinan 250012, China
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Institute of Shandong University, Wenhuaxi Road 107#, Shandong, Jinan 250012, China; Institute of Hepatology, Shandong University, 250012 Shandong, Jinan, China.
| |
Collapse
|
14
|
Rajput P, Pandey V, Kumar V. Stimulation of ribosomal RNA gene promoter by transcription factor Sp1 involves active DNA demethylation by Gadd45-NER pathway. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1859:953-63. [PMID: 27156884 DOI: 10.1016/j.bbagrm.2016.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 04/23/2016] [Accepted: 05/04/2016] [Indexed: 01/29/2023]
Abstract
The well-studied Pol II transcription factor Sp1 has not been investigated for its regulatory role in rDNA transcription. Here, we show that Sp1 bound to specific sites on rDNA and localized into the nucleoli during the G1 phase of cell cycle to activate rDNA transcription. It facilitated the recruitment of Pol I pre-initiation complex and impeded the binding of nucleolar remodeling complex (NoRC) to rDNA resulting in the formation of euchromatin active state. More importantly, Sp1 also orchestrated the site-specific binding of Gadd45a-nucleotide excision repair (NER) complex resulting in active demethylation and transcriptional activation of rDNA. Interestingly, knockdown of Sp1 impaired rDNA transcription due to reduced engagement of the Gadd45a-NER complex and hypermethylation of rDNA. Thus, the present study unveils a novel role of Sp1 in rDNA transcription involving promoter demethylation.
Collapse
MESH Headings
- A549 Cells
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cell Nucleolus/metabolism
- DNA Methylation
- DNA Repair
- DNA, Ribosomal/genetics
- DNA, Ribosomal/metabolism
- Epigenesis, Genetic
- Euchromatin/chemistry
- Euchromatin/metabolism
- G1 Phase
- Genes, Reporter
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- HCT116 Cells
- HEK293 Cells
- Humans
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Promoter Regions, Genetic
- RNA, Ribosomal/genetics
- RNA, Ribosomal/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction
- Sp1 Transcription Factor/antagonists & inhibitors
- Sp1 Transcription Factor/genetics
- Sp1 Transcription Factor/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- Pallavi Rajput
- Virology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi-110067, India
| | - Vijaya Pandey
- Virology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi-110067, India
| | - Vijay Kumar
- Virology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi-110067, India.
| |
Collapse
|
15
|
Sweatt JD. Dynamic DNA methylation controls glutamate receptor trafficking and synaptic scaling. J Neurochem 2016; 137:312-30. [PMID: 26849493 DOI: 10.1111/jnc.13564] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/27/2016] [Accepted: 01/30/2016] [Indexed: 12/27/2022]
Abstract
Hebbian plasticity, including long-term potentiation and long-term depression, has long been regarded as important for local circuit refinement in the context of memory formation and stabilization. However, circuit development and stabilization additionally relies on non-Hebbian, homeostatic, forms of plasticity such as synaptic scaling. Synaptic scaling is induced by chronic increases or decreases in neuronal activity. Synaptic scaling is associated with cell-wide adjustments in postsynaptic receptor density, and can occur in a multiplicative manner resulting in preservation of relative synaptic strengths across the entire neuron's population of synapses. Both active DNA methylation and demethylation have been validated as crucial regulators of gene transcription during learning, and synaptic scaling is known to be transcriptionally dependent. However, it has been unclear whether homeostatic forms of plasticity such as synaptic scaling are regulated via epigenetic mechanisms. This review describes exciting recent work that has demonstrated a role for active changes in neuronal DNA methylation and demethylation as a controller of synaptic scaling and glutamate receptor trafficking. These findings bring together three major categories of memory-associated mechanisms that were previously largely considered separately: DNA methylation, homeostatic plasticity, and glutamate receptor trafficking. This review describes exciting recent work that has demonstrated a role for active changes in neuronal DNA methylation and demethylation as a controller of synaptic scaling and glutamate receptor trafficking. These findings bring together three major categories of memory-associated mechanisms that were previously considered separately: glutamate receptor trafficking, DNA methylation, and homeostatic plasticity.
Collapse
Affiliation(s)
- J David Sweatt
- Department of Neurobiology, Evelyn F. McKnight Brain Institute, Civitan International Research Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
16
|
Kienhöfer S, Musheev MU, Stapf U, Helm M, Schomacher L, Niehrs C, Schäfer A. GADD45a physically and functionally interacts with TET1. Differentiation 2015; 90:59-68. [PMID: 26546041 PMCID: PMC4673086 DOI: 10.1016/j.diff.2015.10.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 10/22/2015] [Accepted: 10/22/2015] [Indexed: 12/30/2022]
Abstract
DNA demethylation plays a central role during development and in adult physiology. Different mechanisms of active DNA demethylation have been established. For example, Growth Arrest and DNA Damage 45-(GADD45) and Ten-Eleven-Translocation (TET) proteins act in active DNA demethylation but their functional relationship is unresolved. Here we show that GADD45a physically interacts – and functionally cooperates with TET1 in methylcytosine (mC) processing. In reporter demethylation GADD45a requires endogenous TET1 and conversely TET1 requires GADD45a. On GADD45a target genes TET1 hyperinduces 5-hydroxymethylcytosine (hmC) in the presence of GADD45a, while 5-formyl-(fC) and 5-carboxylcytosine (caC) are reduced. Likewise, in global analysis GADD45a positively regulates TET1 mediated mC oxidation and enhances fC/caC removal. Our data suggest a dual function of GADD45a in oxidative DNA demethylation, to promote directly or indirectly TET1 activity and to enhance subsequent fC/caC removal.
Collapse
Affiliation(s)
| | | | - Ulrike Stapf
- Institute of Molecular Biology, 55128 Mainz, Germany
| | - Mark Helm
- Johannes Gutenberg Universität Mainz, Institut für Pharmazie und Biochemie, 55128 Mainz, Germany
| | | | - Christof Niehrs
- Institute of Molecular Biology, 55128 Mainz, Germany; DKFZ-ZMBH Alliance, Division of Molecular Embryology, 69120 Heidelberg, Germany
| | | |
Collapse
|
17
|
He ZH, Chen Y, Chen P, He SD, Ye JR, Liu D. Decitabine enhances stem cell antigen-1 expression in cigarette smoke extract-induced emphysema in animal model. Exp Biol Med (Maywood) 2015; 241:131-9. [PMID: 26264445 DOI: 10.1177/1535370215598402] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 07/08/2015] [Indexed: 12/24/2022] Open
Abstract
Stem cell antigen-1 (Sca-1) is a mouse glycosyl phosphatidylinositol-anchored protein and a cell surface marker found on hematopoietic stem cells (HSCs). Despite decades of study, its biological functions remain little known. Sca-1 is a typical marker of bone marrow-derived HSCs, it is also expressed by a mixture of tissue-resident stem, progenitor cells in nonhematopoietic organs. Endothelial progenitor cell (EPC) is a subtype of HSC and contributes to endothelial repair by homing in on locations of injury. Abnormal genetic methylation has been detected in smoking-related diseases. The present study aimed to investigate the lung function and histomorphology, the expression of Sca-1 gene in lung tissues, and bone marrow-derived EPCs in cigarette smoke extract (CSE)-induced emphysema mice, and to further determine whether Decitabine (Dec), the most widely used inhibitor of DNA methylation, could protect against the damages caused by CSE. The results of the present study demonstrated that Dec could partly protect against CSE-induced emphysema in mice, enhance Sca-1 expression in lung tissue, and bone marrow-derived EPCs. The results suggested that the depletion of the progenitor cell pool and DNA methylation of Sca-1 gene may be involved in the progression of emphysema in mice.
Collapse
Affiliation(s)
- Zhi-Hui He
- Intensive Care Unit, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011, China
| | - Yan Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011, China Division of Respiratory Disease, Department of Internal Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011, China
| | - Ping Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011, China
| | - Sheng-Dong He
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011, China
| | - Ji-Ru Ye
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011, China
| | - Da Liu
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011, China
| |
Collapse
|
18
|
Vlková V, Štěpánek I, Hrušková V, Šenigl F, Mayerová V, Šrámek M, Šímová J, Bieblová J, Indrová M, Hejhal T, Dérian N, Klatzmann D, Six A, Reiniš M. Epigenetic regulations in the IFNγ signalling pathway: IFNγ-mediated MHC class I upregulation on tumour cells is associated with DNA demethylation of antigen-presenting machinery genes. Oncotarget 2015; 5:6923-35. [PMID: 25071011 PMCID: PMC4196173 DOI: 10.18632/oncotarget.2222] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Downregulation of MHC class I expression on tumour cells, a common mechanism by which tumour cells can escape from specific immune responses, can be associated with coordinated silencing of antigen-presenting machinery genes. The expression of these genes can be restored by IFNγ. In this study we documented association of DNA demethylation of selected antigen-presenting machinery genes located in the MHC genomic locus (TAP-1, TAP-2, LMP-2, LMP-7) upon IFNγ treatment with MHC class I upregulation on tumour cells in several MHC class I-deficient murine tumour cell lines (TC-1/A9, TRAMP-C2, MK16 and MC15). Our data also documented higher methylation levels in these genes in TC-1/A9 cells, as compared to their parental MHC class I-positive TC-1 cells. IFNγ-mediated DNA demethylation was relatively fast in comparison with demethylation induced by DNA methyltransferase inhibitor 5-azacytidine, and associated with increased histone H3 acetylation in the promoter regions of APM genes. Comparative transcriptome analysis in distinct MHC class I-deficient cell lines upon their treatment with either IFNγ or epigenetic agents revealed that a set of genes, significantly enriched for the antigen presentation pathway, was regulated in the same manner. Our data demonstrate that IFNγ acts as an epigenetic modifier when upregulating the expression of antigen-presenting machinery genes.
Collapse
Affiliation(s)
- Veronika Vlková
- Department of Tumour Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v. v. i., Prague
| | - Ivan Štěpánek
- Department of Tumour Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v. v. i., Prague
| | - Veronika Hrušková
- Department of Tumour Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v. v. i., Prague
| | - Filip Šenigl
- Department of Viral and Cellular Genetics, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v. v. i., Prague
| | - Veronika Mayerová
- Department of Tumour Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v. v. i., Prague
| | - Martin Šrámek
- Department of Tumour Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v. v. i., Prague
| | - Jana Šímová
- Department of Tumour Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v. v. i., Prague
| | - Jana Bieblová
- Department of Tumour Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v. v. i., Prague
| | - Marie Indrová
- Department of Tumour Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v. v. i., Prague
| | - Tomáš Hejhal
- Department of Tumour Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v. v. i., Prague
| | - Nicolas Dérian
- UPMC Univ Paris 06, UMR 7211, Immunology-Immunopathology-Immunotherapy (I3), Paris, France; CNRS, UMR 7211, Immunology-Immunopathology-Immunotherapy (I3), Paris, France; INSERM, UMR_S 959, Immunology-Immunopathology-Immunotherapy (I3), Paris, France; AP-HP, Hôpital Pitié-Salpêtrière, CIC-BTi Biotherapy & Département Hospitalo-Universitaire (DHU) Inflammation-Immunopathology-Biotherapy (i2B), Paris, France
| | - David Klatzmann
- UPMC Univ Paris 06, UMR 7211, Immunology-Immunopathology-Immunotherapy (I3), Paris, France; CNRS, UMR 7211, Immunology-Immunopathology-Immunotherapy (I3), Paris, France; INSERM, UMR_S 959, Immunology-Immunopathology-Immunotherapy (I3), Paris, France; AP-HP, Hôpital Pitié-Salpêtrière, CIC-BTi Biotherapy & Département Hospitalo-Universitaire (DHU) Inflammation-Immunopathology-Biotherapy (i2B), Paris, France
| | - Adrien Six
- UPMC Univ Paris 06, UMR 7211, Immunology-Immunopathology-Immunotherapy (I3), Paris, France; CNRS, UMR 7211, Immunology-Immunopathology-Immunotherapy (I3), Paris, France; INSERM, UMR_S 959, Immunology-Immunopathology-Immunotherapy (I3), Paris, France; AP-HP, Hôpital Pitié-Salpêtrière, CIC-BTi Biotherapy & Département Hospitalo-Universitaire (DHU) Inflammation-Immunopathology-Biotherapy (i2B), Paris, France
| | - Milan Reiniš
- Department of Tumour Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v. v. i., Prague
| |
Collapse
|
19
|
Abstract
The study of epigenetics in plants has a long and rich history, from initial descriptions of non-Mendelian gene behaviors to seminal discoveries of chromatin-modifying proteins and RNAs that mediate gene silencing in most eukaryotes, including humans. Genetic screens in the model plant Arabidopsis have been particularly rewarding, identifying more than 130 epigenetic regulators thus far. The diversity of epigenetic pathways in plants is remarkable, presumably contributing to the phenotypic plasticity of plant postembryonic development and the ability to survive and reproduce in unpredictable environments.
Collapse
Affiliation(s)
- Craig S Pikaard
- Department of Biology, Department of Molecular and Cellular Biochemistry, and Howard Hughes Medical Institute, Indiana University, Bloomington, Indiana 47405
| | - Ortrun Mittelsten Scheid
- Gregor Mendel-Institute of Molecular Plant Biology, Austrian Academy of Sciences, 1030 Vienna, Austria
| |
Collapse
|
20
|
Romagnolo DF, Zempleni J, Selmin OI. Nuclear receptors and epigenetic regulation: opportunities for nutritional targeting and disease prevention. Adv Nutr 2014; 5:373-85. [PMID: 25022987 PMCID: PMC4085186 DOI: 10.3945/an.114.005868] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Posttranslational modifications of histones, alterations in the recruitment and functions of non-histone proteins, DNA methylation, and changes in expression of noncoding RNAs contribute to current models of epigenetic regulation. Nuclear receptors (NRs) are a group of transcription factors that, through ligand-binding, act as sensors to changes in nutritional, environmental, developmental, pathophysiologic, and endocrine conditions and drive adaptive responses via gene regulation. One mechanism through which NRs direct gene expression is the assembly of transcription complexes with cofactors and coregulators that possess chromatin-modifying properties. Chromatin modifications can be transient or become part of the cellular "memory" and contribute to genomic imprinting. Because many food components bind to NRs, they can ultimately influence transcription of genes associated with biologic processes, such as inflammation, proliferation, apoptosis, and hormonal response, and alter the susceptibility to chronic diseases (e.g., cancer, diabetes, obesity). The objective of this review is to highlight how NRs influence epigenetic regulation and the relevance of dietary compound-NR interactions in human nutrition and for disease prevention and treatment. Identifying gene targets of unliganded and bound NRs may assist in the development of epigenetic maps for food components and dietary patterns. Progress in these areas may lead to the formulation of disease-prevention models based on epigenetic control by individual or associations of food ligands of NRs.
Collapse
Affiliation(s)
- Donato F Romagnolo
- Department of Nutritional Sciences and University of Arizona Cancer Center, University of Arizona, Tucson, AZ; and
| | - Janos Zempleni
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE
| | - Ornella I Selmin
- Department of Nutritional Sciences and University of Arizona Cancer Center, University of Arizona, Tucson, AZ; and
| |
Collapse
|
21
|
Duncan EJ, Gluckman PD, Dearden PK. Epigenetics, plasticity, and evolution: How do we link epigenetic change to phenotype? JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2014; 322:208-20. [PMID: 24719220 DOI: 10.1002/jez.b.22571] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 03/13/2014] [Accepted: 03/15/2014] [Indexed: 12/12/2022]
Abstract
Epigenetic mechanisms are proposed as an important way in which the genome responds to the environment. Epigenetic marks, including DNA methylation and Histone modifications, can be triggered by environmental effects, and lead to permanent changes in gene expression, affecting the phenotype of an organism. Epigenetic mechanisms have been proposed as key in plasticity, allowing environmental exposure to shape future gene expression. While we are beginning to understand how these mechanisms have roles in human biology and disease, we have little understanding of their roles and impacts on ecology and evolution. In this review, we discuss different types of epigenetic marks, their roles in gene expression and plasticity, methods for assaying epigenetic changes, and point out the future advances we require to understand fully the impact of this field.
Collapse
Affiliation(s)
- Elizabeth J Duncan
- Genetics Otago and Gravida, The National Centre for Growth and Development, Biochemistry Department, University of Otago, Dunedin, New Zealand
| | | | | |
Collapse
|
22
|
von Stechow L, Ruiz-Aracama A, van de Water B, Peijnenburg A, Danen E, Lommen A. Identification of cisplatin-regulated metabolic pathways in pluripotent stem cells. PLoS One 2013; 8:e76476. [PMID: 24146875 PMCID: PMC3797786 DOI: 10.1371/journal.pone.0076476] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 08/29/2013] [Indexed: 11/19/2022] Open
Abstract
The chemotherapeutic compound, cisplatin causes various kinds of DNA lesions but also triggers other pertubations, such as ER and oxidative stress. We and others have shown that treatment of pluripotent stem cells with cisplatin causes a plethora of transcriptional and post-translational alterations that, to a major extent, point to DNA damage response (DDR) signaling. The orchestrated DDR signaling network is important to arrest the cell cycle and repair the lesions or, in case of damage beyond repair, eliminate affected cells. Failure to properly balance the various aspects of the DDR in stem cells contributes to ageing and cancer. Here, we performed metabolic profiling by mass spectrometry of embryonic stem (ES) cells treated for different time periods with cisplatin. We then integrated metabolomics with transcriptomics analyses and connected cisplatin-regulated metabolites with regulated metabolic enzymes to identify enriched metabolic pathways. These included nucleotide metabolism, urea cycle and arginine and proline metabolism. Silencing of identified proline metabolic and catabolic enzymes indicated that altered proline metabolism serves as an adaptive, rather than a toxic response. A group of enriched metabolic pathways clustered around the metabolite S-adenosylmethionine, which is a hub for methylation and transsulfuration reactions and polyamine metabolism. Enzymes and metabolites with pro- or anti-oxidant functions were also enriched but enhanced levels of reactive oxygen species were not measured in cisplatin-treated ES cells. Lastly, a number of the differentially regulated metabolic enzymes were identified as target genes of the transcription factor p53, pointing to p53-mediated alterations in metabolism in response to genotoxic stress. Altogether, our findings reveal interconnecting metabolic pathways that are responsive to cisplatin and may serve as signaling modules in the DDR in pluripotent stem cells.
Collapse
Affiliation(s)
- Louise von Stechow
- Department of Toxicology, LACDR, Leiden University, Leiden, The Netherlands
- Netherlands Toxicogenomics Center, Maastricht, The Netherlands
| | - Ainhoa Ruiz-Aracama
- RIKILT - Institute of Food Safety, Wageningen, The Netherlands
- Netherlands Toxicogenomics Center, Maastricht, The Netherlands
| | - Bob van de Water
- Department of Toxicology, LACDR, Leiden University, Leiden, The Netherlands
| | - Ad Peijnenburg
- RIKILT - Institute of Food Safety, Wageningen, The Netherlands
| | - Erik Danen
- Department of Toxicology, LACDR, Leiden University, Leiden, The Netherlands
- * E-mail: (AL); (ED)
| | - Arjen Lommen
- RIKILT - Institute of Food Safety, Wageningen, The Netherlands
- * E-mail: (AL); (ED)
| |
Collapse
|
23
|
Schomacher L. Mammalian DNA demethylation: multiple faces and upstream regulation. Epigenetics 2013; 8:679-84. [PMID: 23803967 PMCID: PMC3781186 DOI: 10.4161/epi.24977] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 05/07/2013] [Accepted: 05/08/2013] [Indexed: 01/01/2023] Open
Abstract
DNA cytosine methylation is a reversible epigenetic mark regulating gene expression. Aberrant methylation profiles are concomitant with developmental defects and cancer. Numerous studies in the past decade have identified enzymes and pathways responsible for active DNA demethylation both on a genome-wide as well as gene-specific scale. Recent findings have strengthened the idea that 5-methylcytosine oxidation catalyzed by members of the ten-eleven translocation (Tet1-3) oxygenases in conjunction with replication-coupled dilution of the conversion products causes the majority of genome-wide erasure of methylation marks during early development. In contrast, short and long patch DNA excision repair seems to be implicated mainly in gene-specific demethylation. Growth arrest and DNA damage-inducible protein 45 a (Gadd45a) regulates gene-specific demethylation within regulatory sequences of limited lengths raising the question of how such site specificity is achieved. A new study identified the protein inhibitor of growth 1 (Ing1) as a reader of the active chromatin mark histone H3 lysine 4 trimethylation (H3K4me3). Ing1 binds and directs Gadd45a to target sites, thus linking the histone code with DNA demethylation.
Collapse
|
24
|
Li J, Braganza A, Sobol RW. Base excision repair facilitates a functional relationship between Guanine oxidation and histone demethylation. Antioxid Redox Signal 2013; 18:2429-43. [PMID: 23311711 PMCID: PMC3671628 DOI: 10.1089/ars.2012.5107] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
SIGNIFICANCE Appropriately controlled epigenetic regulation is critical for the normal development and health of an organism. Misregulation of epigenetic control via deoxyribonucleic acid (DNA) methylation or histone methylation has been associated with cancer and chromosomal instability syndromes. RECENT ADVANCES The main function of the proteins in the base excision repair (BER) pathway is to repair DNA single-strand breaks and deamination, oxidation, and alkylation-induced DNA base damage that may result from chemotherapy, environmental exposure, or byproducts of cellular metabolism. Recent studies have suggested that one or more BER proteins may also participate in epigenetic regulation to facilitate gene expression modulation via alteration of the state of DNA methylation or via a reaction coupled to histone modification. BER proteins have also been reported to play an essential role in pluripotent stem cell reprogramming. CRITICAL ISSUES One emerging function for BER in epigenetic regulation is the repair of base lesions induced by hydrogen peroxide as a byproduct of lysine-specific demethylase 1 (LSD1) enzymatic activity (LSD1/LSD2-coupled BER) for transcriptional regulation. FUTURE DIRECTIONS To shed light on this novel role of BER, this review focuses on the repair of oxidative lesions in nuclear DNA that are induced during LSD1-mediated histone demethylation. Further, we highlight current studies suggesting a role for BER proteins in transcriptional regulation of gene expression via BER-coupled active DNA demethylation in mammalian cells. Such efforts to address the role of BER proteins in epigenetic regulation could broaden cancer therapeutic strategies to include epigenetic modifiers combined with BER inhibitors.
Collapse
Affiliation(s)
- Jianfeng Li
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | |
Collapse
|
25
|
Huang S, Zhu Z, Wang Y, Wang Y, Xu L, Chen X, Xu Q, Zhang Q, Zhao X, Yu Y, Wu D. Tet1 is required for Rb phosphorylation during G1/S phase transition. Biochem Biophys Res Commun 2013; 434:241-4. [PMID: 23542465 DOI: 10.1016/j.bbrc.2013.02.110] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 02/16/2013] [Indexed: 01/08/2023]
Abstract
DNA methylation plays an important role in many biological processes, including regulation of gene expression, maintenance of chromatin conformation and genomic stability. TET-family proteins convert 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), which indicates that these enzymes may participate in DNA demethylation. The function of TET1 has not yet been well characterized in somatic cells. Here, we show that depletion of Tet1 in NIH3T3 cells inhibits cell growth. Furthermore, Tet1 knockdown blocks cyclin D1 accumulation in G1 phase, inhibits Rb phosphorylation and consequently delays entrance to G1/S phase. Taken together, this study demonstrates that Tet1 is required for cell proliferation and that this process is mediated through the Rb pathway.
Collapse
Affiliation(s)
- Shengsong Huang
- Department of Urology, Tongji Hospital of Tongji University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Schäfer A, Karaulanov E, Stapf U, Döderlein G, Niehrs C. Ing1 functions in DNA demethylation by directing Gadd45a to H3K4me3. Genes Dev 2013; 27:261-73. [PMID: 23388825 DOI: 10.1101/gad.186916.112] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Active DNA demethylation regulates epigenetic gene activation in numerous processes, but how the target site specificity of DNA demethylation is determined and what factors are involved are still poorly understood. Here we show that the tumor suppressor inhibitor of growth protein 1 (Ing1) is required for targeting active DNA demethylation. Ing1 functions by recruiting the regulator of DNA demethylation growth arrest and DNA damage protein 45a (Gadd45a) to histone H3 trimethylated at Lys 4 (H3K4me3). We show that reduced H3K4 methylation impairs recruitment of Gadd45a/Ing1 and gene-specific DNA demethylation. Our results indicate that histone methylation directs DNA demethylation.
Collapse
|
27
|
Abstract
Embryonic stem cells (ESCs) can self renew and retain the potential to differentiate into each of the cell types within the body. During experimental reprogramming, many of the features of ESCs can be acquired by differentiated target cells. One of these is the unusual cell division cycle that characterizes ESCs in which the Gap (G) phases are short and DNA Synthesis (S) phase predominates. Growing evidence has suggested that this atypical cell-cycle structure may be important for maintaining pluripotency and for enhancing pluripotent conversion. Here, we review current knowledge of cell-cycle regulation in ESCs and outline how this unique cell-cycle structure might contribute to successful reprogramming.
Collapse
Affiliation(s)
- Tomomi Tsubouchi
- MRC Genome Damage and Stability Centre, University of Sussex, Falmer, United Kingdom
| | | |
Collapse
|
28
|
Schäfer A. Gadd45 proteins: key players of repair-mediated DNA demethylation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 793:35-50. [PMID: 24104472 DOI: 10.1007/978-1-4614-8289-5_3] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The three growth arrest and DNA damage 45 (Gadd45) family genes encode for stress-response proteins that are rapidly induced upon cellular stress or differentiation cues. They are well-characterized regulators of cell cycle, senescence, survival, and apoptosis. More recently, it has become clear that Gadd45 proteins promote active DNA demethylation thereby mediating gene activation. This epigenetic function of Gadd45 is important for differentiation and transcriptional regulation during development. Mechanistically, Gadd45 acts as an adapter for DNA repair factors at gene-specific loci to promote removal of 5-methylcytosine from DNA. Hence, Gadd45 is a nexus between DNA repair and epigenetic gene regulation.
Collapse
|
29
|
Abstract
DNA methylation has long been considered a very stable DNA modification in mammals that could only be removed by replication in the absence of remethylation - that is, by mere dilution of this epigenetic mark (so-called passive DNA demethylation). However, in recent years, a significant number of studies have revealed the existence of active processes of DNA demethylation in mammals, with important roles in development and transcriptional regulation, allowing the molecular mechanisms of active DNA demethylation to be unraveled. In this article, we review the recent literature highlighting the prominent role played in active DNA demethylation by base excision repair and especially by TDG.
Collapse
Affiliation(s)
- Shannon R Dalton
- Cancer Biology Program, Epigenetics & Progenitor Cells Program, Fox Chase Cancer Center, PA 19111, USA
| | | |
Collapse
|
30
|
Rea M, Zheng W, Chen M, Braud C, Bhangu D, Rognan TN, Xiao W. Histone H1 affects gene imprinting and DNA methylation in Arabidopsis. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2012; 71:776-86. [PMID: 22519754 PMCID: PMC3429642 DOI: 10.1111/j.1365-313x.2012.05028.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Imprinting, i.e. parent-of-origin expression of alleles, plays an important role in regulating development in mammals and plants. DNA methylation catalyzed by DNA methyltransferases plays a pivotal role in regulating imprinting by silencing parental alleles. DEMETER (DME), a DNA glycosylase functioning in the base-excision DNA repair pathway, can excise 5-methylcytosine from DNA and regulate genomic imprinting in Arabidopsis. DME demethylates the maternal MEDEA (MEA) promoter in endosperm, resulting in expression of the maternal MEA allele. However, it is not known whether DME interacts with other proteins in regulating gene imprinting. Here we report the identification of histone H1.2 as a DME-interacting protein in a yeast two-hybrid screen, and confirmation of their interaction by the in vitro pull-down assay. Genetic analysis of the loss-of-function histone h1 mutant showed that the maternal histone H1 allele is required for DME regulation of MEA, FWA and FIS2 imprinting in Arabidopsis endosperm but the paternal allele is dispensable. Furthermore, we show that mutations in histone H1 result in an increase of DNA methylation in the maternal MEA and FWA promoter in endosperm. Our results suggest that histone H1 is involved in DME-mediated DNA methylation and gene regulation at imprinted loci.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wenyan Xiao
- For correspondence: Fax, 314-977-3658; Tel, 314-977-2547;
| |
Collapse
|
31
|
Suarez-Alvarez B, Rodriguez RM, Fraga MF, López-Larrea C. DNA methylation: a promising landscape for immune system-related diseases. Trends Genet 2012; 28:506-14. [PMID: 22824525 DOI: 10.1016/j.tig.2012.06.005] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Revised: 06/13/2012] [Accepted: 06/20/2012] [Indexed: 01/31/2023]
Abstract
During hematopoiesis, a unique hematopoietic stem cell (HSC) from the bone marrow gives rise to a subset of mature blood cells that directs all the immune responses. Recent studies have shown that this well-defined, hierarchical process is regulated in part by epigenetic mechanisms. Changes in the DNA methylation profile have a critical role in the division of these stem cells into the myeloid and lymphoid lineages and in the establishment of a specific phenotype and functionality in each terminally differentiated cell type. In this review, we describe how the DNA methylation patterns are modified during hematopoietic differentiation and what their role is in cell plasticity and immune function. An in-depth knowledge of these epigenetic mechanisms will help clarify how cell type-specific gene programs are established, and how they can be leveraged in the development of novel strategies for treating immune system-related pathologies.
Collapse
|
32
|
Epigenetic control on cell fate choice in neural stem cells. Protein Cell 2012; 3:278-90. [PMID: 22549586 DOI: 10.1007/s13238-012-2916-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 03/31/2012] [Indexed: 10/28/2022] Open
Abstract
Derived from neural stem cells (NSCs) and progenitor cells originated from the neuroectoderm, the nervous system presents an unprecedented degree of cellular diversity, interwoven to ensure correct connections for propagating information and responding to environmental cues. NSCs and progenitor cells must integrate cell-intrinsic programs and environmental cues to achieve production of appropriate types of neurons and glia at appropriate times and places during development. These developmental dynamics are reflected in changes in gene expression, which is regulated by transcription factors and at the epigenetic level. From early commitment of neural lineage to functional plasticity in terminal differentiated neurons, epigenetic regulation is involved in every step of neural development. Here we focus on the recent advance in our understanding of epigenetic regulation on orderly generation of diverse neural cell types in the mammalian nervous system, an important aspect of neural development and regenerative medicine.
Collapse
|
33
|
Ross JP, Rand KN, Molloy PL. Hypomethylation of repeated DNA sequences in cancer. Epigenomics 2012; 2:245-69. [PMID: 22121873 DOI: 10.2217/epi.10.2] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
An important feature of cancer development and progression is the change in DNA methylation patterns, characterized by the hypermethylation of specific genes concurrently with an overall decrease in the level of 5-methylcytosine. Hypomethylation of the genome can affect both single-copy genes, repeat DNA sequences and transposable elements, and is highly variable among and within cancer types. Here, we review our current understanding of genome hypomethylation in cancer, with a particular focus on hypomethylation of the different classes and families of repeat sequences. The emerging data provide insights into the importance of methylation of different repeat families in the maintenance of chromosome structural integrity and the fidelity of normal transcriptional regulation. We also consider the events underlying cancer-associated hypomethylation and the potential for the clinical use of characteristic DNA methylation changes in diagnosis, prognosis or classification of tumors.
Collapse
Affiliation(s)
- Jason P Ross
- Commonwealth Scientific & Industrial Research Organisation, Food & Nutritional Science, Preventative Health National Research Flagship, North Ryde, NSW 1670, Australia
| | | | | |
Collapse
|
34
|
Abstract
DNA hypomethylation was the initial epigenetic abnormality recognized in human tumors. However, for several decades after its independent discovery by two laboratories in 1983, it was often ignored as an unwelcome complication, with almost all of the attention on the hypermethylation of promoters of genes that are silenced in cancers (e.g., tumor-suppressor genes). Because it was subsequently shown that global hypomethylation of DNA in cancer was most closely associated with repeated DNA elements, cancer linked-DNA hypomethylation continued to receive rather little attention. DNA hypomethylation in cancer can no longer be considered an oddity, because recent high-resolution genome-wide studies confirm that DNA hypomethylation is the almost constant companion to hypermethylation of the genome in cancer, just usually (but not always) in different sequences. Methylation changes at individual CpG dyads in cancer can have a high degree of dependence not only on the regional context, but also on neighboring sites. DNA demethylation during carcinogenesis may involve hemimethylated dyads as intermediates, followed by spreading of the loss of methylation on both strands. In this review, active demethylation of DNA and the relationship of cancer-associated DNA hypomethylation to cancer stem cells are discussed. Evidence is accumulating for the biological significance and clinical relevance of DNA hypomethylation in cancer, and for cancer-linked demethylation and de novo methylation being highly dynamic processes.
Collapse
Affiliation(s)
- Melanie Ehrlich
- Hayward Genetics Program, Department of Biochemistry, Tulane Cancer Center, Tulane Medical School, 1430 TulaneAvenue, New Orleans, LA 70112, USA.
| |
Collapse
|
35
|
Niehrs C, Schäfer A. Active DNA demethylation by Gadd45 and DNA repair. Trends Cell Biol 2012; 22:220-7. [PMID: 22341196 DOI: 10.1016/j.tcb.2012.01.002] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 12/23/2011] [Accepted: 01/05/2012] [Indexed: 11/13/2022]
Abstract
How DNA methylation patterns are established, maintained and remodeled is incompletely understood, however, it has become clear that DNA methylation is reversible and dynamic as a result of enzymatic DNA demethylation. Several different mechanisms that may account for demethylation have recently been put forward and all seem to involve DNA repair. Here, we review DNA demethylation mediated by multifunctional growth arrest and DNA damage 45 (Gadd45) protein family members which mediate DNA demethylation during cell differentiation and stress response. Gadd45 recruits nucleotide and/or base excision repair factors to gene-specific loci and acts as an adapter between repair factors and chromatin, thereby creating a nexus between epigenetics and DNA repair.
Collapse
|
36
|
Li Y, Ohms SJ, Shannon FM, Sun C, Fan JY. IL-2 and GM-CSF are regulated by DNA demethylation during activation of T cells, B cells and macrophages. Biochem Biophys Res Commun 2012; 419:748-53. [PMID: 22387543 DOI: 10.1016/j.bbrc.2012.02.094] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Accepted: 02/16/2012] [Indexed: 11/19/2022]
Abstract
DNA demethylation has been found to occur at the promoters of a number of actively expressed cytokines and is believed to play a critical role in transcriptional regulation. While many DNA demethylation studies have focused on T cell activation, proliferation and differentiation, changes in DNA methylation in other types of immune cells are less well studied. We found that the expression of two cytokines (IL-2 and GM-CSF) responded differently to activation in three types of immune cells: EL4, A20 and RAW264.7 cells. Using the McrBC and MeDIP approaches, we observed decreases in DNA methylation at a genome-wide level and at the promoters of the genes of these cytokines. The expression of several potential enzymes/co-enzymes involved in the DNA demethylation pathways seemed to be associated with immune cell activation.
Collapse
Affiliation(s)
- Yan Li
- College of Animal Science & Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | | | | | | | | |
Collapse
|
37
|
Abstract
Understanding the molecular mechanisms involved in the control of cell differentiation during embryonic development is currently one of the main objectives of developmental biology. This knowledge will provide a basis for the development of new strategies in the field of regenerative medicine, one of the most promising weapons to fight many human diseases. Cell differentiation during embryonic development is controlled primarily by epigenetic factors, that is, mechanisms involved in the regulation of chromatin structure and gene expression. Here we describe the best known epigenetic modifications, and pathways, mainly focused on DNA methylation and histone modifications, and try to depict the state of art in our knowledge about epigenetic regulation of embryonic stem cell maintenance and differentiation.
Collapse
|
38
|
Teperek-Tkacz M, Pasque V, Gentsch G, Ferguson-Smith AC. Epigenetic reprogramming: is deamination key to active DNA demethylation? Reproduction 2011; 142:621-32. [PMID: 21911441 DOI: 10.1530/rep-11-0148] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
DNA demethylation processes are important for reproduction, being central in epigenetic reprogramming during embryonic and germ cell development. While the enzymes methylating DNA have been known for many years, identification of factors capable of mediating active DNA demethylation has been challenging. Recent findings suggest that cytidine deaminases may be key players in active DNA demethylation. One of the most investigated candidates is activation-induced cytidine deaminase (AID), best known for its role in generating secondary antibody diversity in B cells. We evaluate evidence for cytidine deaminases in DNA demethylation pathways in vertebrates and discuss possible models for their targeting and activity regulation. These findings are also considered along with alternative demethylation pathways involving hydroxymethylation.
Collapse
|
39
|
Khobta A, Epe B. Interactions between DNA damage, repair, and transcription. Mutat Res 2011; 736:5-14. [PMID: 21907218 DOI: 10.1016/j.mrfmmm.2011.07.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 06/22/2011] [Accepted: 07/25/2011] [Indexed: 01/16/2023]
Abstract
This review addresses a variety of mechanisms by which DNA repair interacts with transcription and vice versa. Blocking of transcriptional elongation is the best studied of these mechanisms. Transcription recovery after damage therefore has often been used as a surrogate marker of DNA repair in cells. However, it has become evident that relationships between DNA damage, repair, and transcription are more complex due to various indirect effects of DNA damage on gene transcription. These include inhibition of transcription by DNA repair intermediates as well as regulation of transcription and of the epigenetic status of the genes by DNA repair-related mechanisms. In addition, since transcription is emerging as an important endogenous source of DNA damage in cells, we briefly summarise recent advances in understanding the nature of co-transcriptionally induced DNA damage and the DNA repair pathways involved.
Collapse
Affiliation(s)
- Andriy Khobta
- Institute of Pharmacy and Biochemistry, University of Mainz, Mainz, Germany
| | | |
Collapse
|
40
|
Cortellino S, Xu J, Sannai M, Moore R, Caretti E, Cigliano A, Le Coz M, Devarajan K, Wessels A, Soprano D, Abramowitz LK, Bartolomei MS, Rambow F, Bassi MR, Bruno T, Fanciulli M, Renner C, Klein-Szanto AJ, Matsumoto Y, Kobi D, Davidson I, Alberti C, Larue L, Bellacosa A. Thymine DNA glycosylase is essential for active DNA demethylation by linked deamination-base excision repair. Cell 2011; 146:67-79. [PMID: 21722948 DOI: 10.1016/j.cell.2011.06.020] [Citation(s) in RCA: 595] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 05/17/2011] [Accepted: 06/12/2011] [Indexed: 01/19/2023]
Abstract
DNA methylation is a major epigenetic mechanism for gene silencing. Whereas methyltransferases mediate cytosine methylation, it is less clear how unmethylated regions in mammalian genomes are protected from de novo methylation and whether an active demethylating activity is involved. Here, we show that either knockout or catalytic inactivation of the DNA repair enzyme thymine DNA glycosylase (TDG) leads to embryonic lethality in mice. TDG is necessary for recruiting p300 to retinoic acid (RA)-regulated promoters, protection of CpG islands from hypermethylation, and active demethylation of tissue-specific developmentally and hormonally regulated promoters and enhancers. TDG interacts with the deaminase AID and the damage response protein GADD45a. These findings highlight a dual role for TDG in promoting proper epigenetic states during development and suggest a two-step mechanism for DNA demethylation in mammals, whereby 5-methylcytosine and 5-hydroxymethylcytosine are first deaminated by AID to thymine and 5-hydroxymethyluracil, respectively, followed by TDG-mediated thymine and 5-hydroxymethyluracil excision repair.
Collapse
Affiliation(s)
- Salvatore Cortellino
- Cancer Biology Program and Epigenetics and Progenitor Cells Keystone Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
We review experiments in which somatic cell nuclei are transplanted singly to enucleated eggs (metaphase II) in amphibia and mammals and as multiple nuclei to the germinal vesicle of amphibian oocytes (prophase I). These experiments have shown the totipotency of some somatic cell nuclei, as well as switches in cell type and changes in gene expression. Abnormalities of nuclear transplant embryo development increase greatly as nuclei are taken from progressively more differentiated donor cells. The molecular changes that accompany the reprogramming of transplanted nuclei help to indicate the mechanisms used by eggs and oocytes to reprogram gene expression. We discuss the importance of chromosomal protein exchange, of transcription factor supply, and of chromatin access in reprogramming.
Collapse
Affiliation(s)
- J B Gurdon
- Wellcome Trust Cancer Research UK Gurdon Institute, Cambridge, United Kingdom.
| | | |
Collapse
|
42
|
Xiao Y, Zhang HL, Bai LY, Wang XM, Li WG, Yang LG. [Active DNA demethylation in mammals]. YI CHUAN = HEREDITAS 2011; 33:298-306. [PMID: 21482518 DOI: 10.3724/sp.j.1005.2011.00298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
DNA methylation is a stable and heritable epigenetic mark, and it is one of the best characterized epigenetic modifications. Active DNA demethylation has been reported both in plant and animal cells, and the mechanism behind it is becoming clear in plant. Whereas a bona fide enzyme, which is responsible for active DNA demethylation, have not been identified in mammals, and active demethylation pathway is controversial. In the present review, we described that active DNA demethylation take place in a spatial- and temporal-specific way on the basis of recent literatures. Moreover, several candidate pathways such as oxygenation and deamination of 5-methyl cytosine and DNA repair pathways, which may be responsible for the active process were introduced on a cell- and tissue-specific view. The aim of this paper is to help re-searchers reveal the mechanism underlying this important event during epigenetic reprogramming in mammals.
Collapse
Affiliation(s)
- Yao Xiao
- Key Lab of Education Ministry of China in Agricultural Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
The generation of complex organisms requires that an initial population of cells with identical gene expression profiles can adopt different cell fates during development by progressively diverging transcriptional programs. These programs depend on the binding of transcritional regulators to specific genomic sites, which in turn is controlled by modifications of the chromatin. Chromatin modifications may occur directly upon DNA by methylation of specific nucleotides, or may involve post-translational modification of histones. Local regulation of histone post-translational modifications regionalizes the genome into euchromatic regions, which are more accessible to DNA-binding factors, and condensed heterochromatic regions, inhibiting the binding of such factors. In addition, these modifications may be required in a genome-wide fashion for processes such as DNA replication or chromosome condensation. From an embryologist's point of view chromatin modifications are intensively studied in the context of imprinting and have more recently received increasing attention in understanding the basis of pluripotency and cellular differentiation. Here, we describe recently uncovered roles of chromatin modifications in zebrafish development and regeneration, as well as available resources and commonly used techniques. We provide a general introduction into chromatin modifications and their respective functions with a focus on gene transcription, as well as key aspects of their roles in the early zebrafish embryo, neural development, formation of the digestive system and tissue regeneration.
Collapse
Affiliation(s)
- Jordi Cayuso Mas
- MRC National Institute for Medical Research, The Ridgeway, London, NW7 1AA, UK
| | | | | |
Collapse
|
44
|
Abstract
Memory formation and storage require long-lasting changes in memory-related neuronal circuits. Recent evidence indicates that DNA methylation may serve as a contributing mechanism in memory formation and storage. These emerging findings suggest a role for an epigenetic mechanism in learning and long-term memory maintenance and raise apparent conundrums and questions. For example, it is unclear how DNA methylation might be reversed during the formation of a memory, how changes in DNA methylation alter neuronal function to promote memory formation, and how DNA methylation patterns differ between neuronal structures to enable both consolidation and storage of memories. Here we evaluate the existing evidence supporting a role for DNA methylation in memory, discuss how DNA methylation may affect genetic and neuronal function to contribute to behavior, propose several future directions for the emerging subfield of neuroepigenetics, and begin to address some of the broader implications of this work.
Collapse
Affiliation(s)
- Jeremy J Day
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | |
Collapse
|
45
|
Pasque V, Miyamoto K, Gurdon JB. Efficiencies and mechanisms of nuclear reprogramming. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2010; 75:189-200. [PMID: 21047900 PMCID: PMC3833051 DOI: 10.1101/sqb.2010.75.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The differentiated state of somatic cells is highly stable, but it can be experimentally reversed. The resulting cells can then be redirected into many different pathways. Nuclear reprogramming has been achieved by nuclear transfer to eggs, cell fusion, and overexpression of transcription factors. The mechanisms of nuclear reprogramming are not understood, but some insight into them is provided by comparing the efficiencies of different reprogramming strategies. Here, we compare these efficiencies by describing the frequency and rapidity with which reprogramming is induced and by the proportion of cells and level of expression in which reprogramming is achieved. We comment on the mechanisms that lead to successful somatic-cell reprogramming and on those that resist in helping to maintain the differentiated state of somatic cells.
Collapse
Affiliation(s)
- V Pasque
- The Wellcome Trust/Cancer Research UK Gurdon Institute and Department of Zoology, University of Cambridge, Cambridge CB2 1QN, United Kingdom
| | | | | |
Collapse
|
46
|
Liu J, Zhang Z, Bando M, Itoh T, Deardorff MA, Li JR, Clark D, Kaur M, Tatsuro K, Kline AD, Chang C, Vega H, Jackson LG, Spinner NB, Shirahige K, Krantz ID. Genome-wide DNA methylation analysis in cohesin mutant human cell lines. Nucleic Acids Res 2010; 38:5657-71. [PMID: 20448023 PMCID: PMC2943628 DOI: 10.1093/nar/gkq346] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2010] [Revised: 04/19/2010] [Accepted: 04/21/2010] [Indexed: 12/17/2022] Open
Abstract
The cohesin complex has recently been shown to be a key regulator of eukaryotic gene expression, although the mechanisms by which it exerts its effects are poorly understood. We have undertaken a genome-wide analysis of DNA methylation in cohesin-deficient cell lines from probands with Cornelia de Lange syndrome (CdLS). Heterozygous mutations in NIPBL, SMC1A and SMC3 genes account for ∼65% of individuals with CdLS. SMC1A and SMC3 are subunits of the cohesin complex that controls sister chromatid cohesion, whereas NIPBL facilitates cohesin loading and unloading. We have examined the methylation status of 27 578 CpG dinucleotides in 72 CdLS and control samples. We have documented the DNA methylation pattern in human lymphoblastoid cell lines (LCLs) as well as identified specific differential DNA methylation in CdLS. Subgroups of CdLS probands and controls can be classified using selected CpG loci. The X chromosome was also found to have a unique DNA methylation pattern in CdLS. Cohesin preferentially binds to hypo-methylated DNA in control LCLs, whereas the differential DNA methylation alters cohesin binding in CdLS. Our results suggest that in addition to DNA methylation multiple mechanisms may be involved in transcriptional regulation in human cells and in the resultant gene misexpression in CdLS.
Collapse
Affiliation(s)
- Jinglan Liu
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Zhe Zhang
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Masashige Bando
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Takehiko Itoh
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Matthew A. Deardorff
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Jennifer R. Li
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Dinah Clark
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Maninder Kaur
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Kondo Tatsuro
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Antonie D. Kline
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Celia Chang
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Hugo Vega
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Laird G. Jackson
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Nancy B. Spinner
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Katsuhiko Shirahige
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| | - Ian D. Krantz
- Division of Human Genetics, Abramson Research Institute, Center for Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA, Laboratory of Chromosome Structure and Function, Department of Biological Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, B2C 4259, Nagatsuta, Midori-ku, Yokohama City, Kanagawa 226-8501, Japan, The University of Pennsylvania School of Medicine, PA 19104, USA, Division of Developmental Disability, Misakaenosono Mutsumi Developmental, Medical, and Welfare Center, Konagai-cho Maki 570-1, Isahaya, 859-0169, Japan, Harvey Institute for Human Genetics, Department of Pediatrics, Greater Baltimore Medical Center, Baltimore, MD 21204, Genomic and Microarray Facility, the Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, Department of Genetics and Genomics Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA and Department of Obstetrics and Gynecology, Drexel University School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
47
|
Liang CY, Wang LJ, Chen CP, Chen LF, Chen YH, Chen H. GCM1 Regulation of the Expression of Syncytin 2 and Its Cognate Receptor MFSD2A in Human Placenta1. Biol Reprod 2010; 83:387-95. [DOI: 10.1095/biolreprod.110.083915] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
48
|
Chmurzynska A. Fetal programming: link between early nutrition, DNA methylation, and complex diseases. Nutr Rev 2010; 68:87-98. [PMID: 20137054 DOI: 10.1111/j.1753-4887.2009.00265.x] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Complex traits, including those involved in diet-related diseases, are determined by multiple genes and environmental influences. Factors influencing the development of complex traits should be expanded to include epigenetic factors, such as DNA methylation, which occurs in utero. Epigenetic factors regulate gene expression and thereby cell differentiation and organogenesis. The process of epigenotype establishment is sensitive to environmental conditions, with nutrition being one of the most important related factors. For example, DNA methylation depends on the availability of several nutrients including methionine and vitamins B(6), B(12), and folate. Epidemiological studies show that undernutrition during fetal life is associated with increased susceptibility to complex diseases. Numerous studies have been conducted on prenatal caloric and protein undernutrition. A reduction in the number of cells and changes in the structure and functioning of organs, as well as permanent changes in DNA methylation and gene expression, have been considered the molecular mechanisms responsible for metabolism programming.
Collapse
Affiliation(s)
- Agata Chmurzynska
- Department of Human Nutrition and Hygiene, Poznan University of Life Sciences, Wojska Polskiego 31, Poznan, Poland.
| |
Collapse
|
49
|
Keverne EB. Epigenetically regulated imprinted genes and foetal programming. Neurotox Res 2010; 18:386-92. [PMID: 20309665 DOI: 10.1007/s12640-010-9169-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Revised: 02/25/2010] [Accepted: 03/01/2010] [Indexed: 12/18/2022]
Abstract
Genomic imprinting is a widespread epigenetic phenomenon in mammals and many imprinted genes are expressed in the developing hypothalamus and placenta. The placenta and brain are very different structures with very different roles, but in the pregnant mother they functionally interact coordinating and ensuring the provision of nutrients, timing of parturition and priming of hypothalamus for maternal care and nurturing. This interaction has been evolutionarily fine-tuned to optimise infant survival such that when resources are poor, the mother 'informs' this condition to the foetus producing a thrifty phenotype that is adapted to survive scarce resources after birth.
Collapse
Affiliation(s)
- Eric B Keverne
- Sub-Department of Animal Behaviour, University of Cambridge, Madingley, Cambridge, CB23 8AA, UK.
| |
Collapse
|
50
|
|