1001
|
Cazaly E, Charlesworth J, Dickinson JL, Holloway AF. Genetic Determinants of Epigenetic Patterns: Providing Insight into Disease. Mol Med 2015; 21:400-9. [PMID: 25822796 DOI: 10.2119/molmed.2015.00001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 03/26/2015] [Indexed: 02/06/2023] Open
Abstract
The field of epigenetics and our understanding of the mechanisms that regulate the establishment, maintenance and heritability of epigenetic patterns continue to grow at a remarkable rate. This information is providing increased understanding of the role of epigenetic changes in disease, insight into the underlying causes of these epigenetic changes and revealing new avenues for therapeutic intervention. Epigenetic modifiers are increasingly being pursued as therapeutic targets in a range of diseases, with a number of agents targeting epigenetic modifications already proving effective in diseases such as cancer. Although it is well established that DNA mutations and aberrant expression of epigenetic modifiers play a key role in disease, attention is now turning to the interplay between genetic and epigenetic factors in complex disease etiology. The role of genetic variability in determining epigenetic profiles, which can then be modified by environmental and stochastic factors, is becoming more apparent. Understanding the interplay between genetic and epigenetic factors is likely to aid in identifying individuals most likely to benefit from epigenetic therapies. This goal is coming closer to realization because of continual advances in laboratory and statistical tools enabling improvements in the integration of genomic, epigenomic and phenotypic data.
Collapse
Affiliation(s)
- Emma Cazaly
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Jac Charlesworth
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Joanne L Dickinson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Adele F Holloway
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
1002
|
Chen Y, Palczewski K. Systems Pharmacology Links GPCRs with Retinal Degenerative Disorders. Annu Rev Pharmacol Toxicol 2015; 56:273-98. [PMID: 25839098 DOI: 10.1146/annurev-pharmtox-010715-103033] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In most biological systems, second messengers and their key regulatory and effector proteins form links between multiple cellular signaling pathways. Such signaling nodes can integrate the deleterious effects of genetic aberrations, environmental stressors, or both in complex diseases, leading to cell death by various mechanisms. Here we present a systems (network) pharmacology approach that, together with transcriptomics analyses, was used to identify different G protein-coupled receptors that experimentally protected against cellular stress and death caused by linked signaling mechanisms. We describe the application of this concept to degenerative and diabetic retinopathies in appropriate mouse models as an example. Systems pharmacology also provides an attractive framework for devising strategies to combat complex diseases by using (repurposing) US Food and Drug Administration-approved pharmacological agents.
Collapse
Affiliation(s)
- Yu Chen
- Yueyang Hospital and.,Clinical Research Institute of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Krzysztof Palczewski
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106;
| |
Collapse
|
1003
|
Loharch S, Bhutani I, Jain K, Gupta P, Sahoo DK, Parkesh R. EpiDBase: a manually curated database for small molecule modulators of epigenetic landscape. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2015; 2015:bav013. [PMID: 25776023 PMCID: PMC4360624 DOI: 10.1093/database/bav013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We have developed EpiDBase (www.epidbase.org), an interactive database of small molecule ligands of epigenetic protein families by bringing together experimental, structural and chemoinformatic data in one place. Currently, EpiDBase encompasses 5784 unique ligands (11 422 entries) of various epigenetic markers such as writers, erasers and readers. The EpiDBase includes experimental IC50 values, ligand molecular weight, hydrogen bond donor and acceptor count, XlogP, number of rotatable bonds, number of aromatic rings, InChIKey, two-dimensional and three-dimensional (3D) chemical structures. A catalog of all epidbase ligands based on the molecular weight is also provided. A structure editor is provided for 3D visualization of ligands. EpiDBase is integrated with tools like text search, disease-specific search, advanced search, substructure, and similarity analysis. Advanced analysis can be performed using substructure and OpenBabel-based chemical similarity fingerprints. The EpiDBase is curated to identify unique molecular scaffolds. Initially, molecules were selected by removing peptides, macrocycles and other complex structures and then processed for conformational sampling by generating 3D conformers. Subsequent filtering through Zinc Is Not Commercial (ZINC: a free database of commercially available compounds for virtual screening) and Lilly MedChem regular rules retained many distinctive drug-like molecules. These molecules were then analyzed for physicochemical properties using OpenBabel descriptors and clustered using various methods such as hierarchical clustering, binning partition and multidimensional scaling. EpiDBase provides comprehensive resources for further design, development and refinement of small molecule modulators of epigenetic markers. Database URL:www.epidbase.org
Collapse
Affiliation(s)
- Saurabh Loharch
- Department of Advanced Protein Science, Institute of Microbial Technology, Chandigarh 160036, India
| | - Isha Bhutani
- Department of Advanced Protein Science, Institute of Microbial Technology, Chandigarh 160036, India
| | - Kamal Jain
- Department of Advanced Protein Science, Institute of Microbial Technology, Chandigarh 160036, India
| | - Pawan Gupta
- Department of Advanced Protein Science, Institute of Microbial Technology, Chandigarh 160036, India
| | - Debendra K Sahoo
- Department of Advanced Protein Science, Institute of Microbial Technology, Chandigarh 160036, India
| | - Raman Parkesh
- Department of Advanced Protein Science, Institute of Microbial Technology, Chandigarh 160036, India
| |
Collapse
|
1004
|
Schiano C, Vietri MT, Grimaldi V, Picascia A, De Pascale MR, Napoli C. Epigenetic-related therapeutic challenges in cardiovascular disease. Trends Pharmacol Sci 2015; 36:226-35. [PMID: 25758254 DOI: 10.1016/j.tips.2015.02.005] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 02/06/2015] [Accepted: 02/09/2015] [Indexed: 02/08/2023]
Abstract
Progress in human genetic and genomic research has led to the identification of genetic variants associated with specific cardiovascular diseases (CVDs), but the pathogenic mechanisms remain unclear. Recent studies have analyzed the involvement of epigenetic mechanisms such as DNA methylation and histone modifications in the development and progression of CVD. Preliminary work has investigated the correlations between DNA methylation, histone modifications, and RNA-based mechanisms with CVDs including atherosclerosis, heart failure (HF), myocardial infarction (MI), and cardiac hypertrophy. Remarkably, both in utero programming and postnatal hypercholesterolemia may affect the epigenetic signature in the human cardiovascular system, thereby providing novel early epigenetic-related pharmacological insights. Interestingly, some dietary compounds, including polyphenols, cocoa, and folic acid, can modulate DNA methylation status, whereas statins may promote epigenetic-based control in CVD prevention through histone modifications. We review recent findings on the epigenetic control of cardiovascular system and new challenges for therapeutic strategies in CVDs.
Collapse
Affiliation(s)
- Concetta Schiano
- Institute of Diagnostic and Nuclear Development (SDN), Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS), Via Emanuele Gianturco 113, 80143 Naples, Italy
| | - Maria Teresa Vietri
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Via Luigi De Crecchio 7, 80138 Naples, Italy
| | - Vincenzo Grimaldi
- Unità Operativa Complessa Division of Immunohematology, Transfusion Medicine and Transplant Immunology (SIMT), Regional Reference Laboratory of Transplant Immunology (LIT), Azienda Ospedaliera Universitaria (AOU), Second University of Naples, Piazza Luigi Miraglia 2, 80138, Naples, Italy.
| | - Antonietta Picascia
- Unità Operativa Complessa Division of Immunohematology, Transfusion Medicine and Transplant Immunology (SIMT), Regional Reference Laboratory of Transplant Immunology (LIT), Azienda Ospedaliera Universitaria (AOU), Second University of Naples, Piazza Luigi Miraglia 2, 80138, Naples, Italy
| | - Maria Rosaria De Pascale
- Unità Operativa Complessa Division of Immunohematology, Transfusion Medicine and Transplant Immunology (SIMT), Regional Reference Laboratory of Transplant Immunology (LIT), Azienda Ospedaliera Universitaria (AOU), Second University of Naples, Piazza Luigi Miraglia 2, 80138, Naples, Italy
| | - Claudio Napoli
- Institute of Diagnostic and Nuclear Development (SDN), Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS), Via Emanuele Gianturco 113, 80143 Naples, Italy; Unità Operativa Complessa Division of Immunohematology, Transfusion Medicine and Transplant Immunology (SIMT), Regional Reference Laboratory of Transplant Immunology (LIT), Azienda Ospedaliera Universitaria (AOU), Second University of Naples, Piazza Luigi Miraglia 2, 80138, Naples, Italy
| |
Collapse
|
1005
|
Pulido Fontes L, Quesada Jimenez P, Mendioroz Iriarte M. Epigenetics and epilepsy. NEUROLOGÍA (ENGLISH EDITION) 2015. [DOI: 10.1016/j.nrleng.2014.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
1006
|
Kantanen J, Løvendahl P, Strandberg E, Eythorsdottir E, Li MH, Kettunen-Præbel A, Berg P, Meuwissen T. Utilization of farm animal genetic resources in a changing agro-ecological environment in the Nordic countries. Front Genet 2015; 6:52. [PMID: 25767477 PMCID: PMC4341116 DOI: 10.3389/fgene.2015.00052] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 02/05/2015] [Indexed: 12/16/2022] Open
Abstract
Livestock production is the most important component of northern European agriculture and contributes to and will be affected by climate change. Nevertheless, the role of farm animal genetic resources in the adaptation to new agro-ecological conditions and mitigation of animal production’s effects on climate change has been inadequately discussed despite there being several important associations between animal genetic resources and climate change issues. The sustainability of animal production systems and future food security require access to a wide diversity of animal genetic resources. There are several genetic questions that should be considered in strategies promoting adaptation to climate change and mitigation of environmental effects of livestock production. For example, it may become important to choose among breeds and even among farm animal species according to their suitability to a future with altered production systems. Some animals with useful phenotypes and genotypes may be more useful than others in the changing environment. Robust animal breeds with the potential to adapt to new agro-ecological conditions and tolerate new diseases will be needed. The key issue in mitigation of harmful greenhouse gas effects induced by livestock production is the reduction of methane (CH4) emissions from ruminants. There are differences in CH4 emissions among breeds and among individual animals within breeds that suggest a potential for improvement in the trait through genetic selection. Characterization of breeds and individuals with modern genomic tools should be applied to identify breeds that have genetically adapted to marginal conditions and to get critical information for breeding and conservation programs for farm animal genetic resources. We conclude that phenotyping and genomic technologies and adoption of new breeding approaches, such as genomic selection introgression, will promote breeding for useful characters in livestock species.
Collapse
Affiliation(s)
- Juha Kantanen
- Green Technology, Natural Resources Institute Finland , Jokioinen, Finland ; Department of Biology, University of Eastern Finland , Kuopio, Finland
| | - Peter Løvendahl
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University , Tjele, Denmark
| | - Erling Strandberg
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences , Uppsala, Sweden
| | - Emma Eythorsdottir
- Faculty of Land and Animal Resources, Agricultural University of Iceland , Reykjavik, Iceland
| | - Meng-Hua Li
- Green Technology, Natural Resources Institute Finland , Jokioinen, Finland ; Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing, China
| | | | - Peer Berg
- NordGen - Nordic Genetic Resource Center , Aas, Norway
| | - Theo Meuwissen
- Department of Animal and Aquacultural Sciences, Norwegian University of Life Sciences , Aas, Norway
| |
Collapse
|
1007
|
Mitochondria in health, aging and diseases: the epigenetic perspective. Biogerontology 2015; 16:569-85. [DOI: 10.1007/s10522-015-9562-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 02/19/2015] [Indexed: 01/15/2023]
|
1008
|
Zampieri M, Ciccarone F, Calabrese R, Franceschi C, Bürkle A, Caiafa P. Reconfiguration of DNA methylation in aging. Mech Ageing Dev 2015; 151:60-70. [PMID: 25708826 DOI: 10.1016/j.mad.2015.02.002] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 01/20/2015] [Accepted: 02/19/2015] [Indexed: 12/12/2022]
Abstract
A complex interplay between multiple biological effects shapes the aging process. The advent of genome-wide quantitative approaches in the epigenetic field has highlighted the effective impact of epigenetic deregulation, particularly of DNA methylation, on aging. Age-associated alterations in DNA methylation are commonly grouped in the phenomenon known as "epigenetic drift" which is characterized by gradual extensive demethylation of genome and hypermethylation of a number of promoter-associated CpG islands. Surprisingly, specific DNA regions show directional epigenetic changes in aged individuals suggesting the importance of these events for the aging process. However, the epigenetic information obtained until now in aging needs a re-consideration due to the recent discovery of 5-hydroxymethylcytosine, a new DNA epigenetic mark present on genome. A recapitulation of the factors involved in the regulation of DNA methylation and the changes occurring in aging will be described in this review also considering the data available on 5 hmC.
Collapse
Affiliation(s)
- Michele Zampieri
- Department of Cellular Biotechnologies and Hematology, "Sapienza" University of Rome, Rome 00161, Italy; Pasteur Institute-Fondazione Cenci Bolognetti, Rome 00161, Italy
| | - Fabio Ciccarone
- Department of Cellular Biotechnologies and Hematology, "Sapienza" University of Rome, Rome 00161, Italy; Pasteur Institute-Fondazione Cenci Bolognetti, Rome 00161, Italy
| | - Roberta Calabrese
- Department of Cellular Biotechnologies and Hematology, "Sapienza" University of Rome, Rome 00161, Italy; Pasteur Institute-Fondazione Cenci Bolognetti, Rome 00161, Italy
| | - Claudio Franceschi
- Department of Experimental Pathology, Alma Mater Studiorum, University of Bologna, Bologna 40126, Italy
| | - Alexander Bürkle
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Konstanz D-78457, Germany
| | - Paola Caiafa
- Department of Cellular Biotechnologies and Hematology, "Sapienza" University of Rome, Rome 00161, Italy; Pasteur Institute-Fondazione Cenci Bolognetti, Rome 00161, Italy.
| |
Collapse
|
1009
|
Jiang R, Jones MJ, Chen E, Neumann SM, Fraser HB, Miller GE, Kobor MS. Discordance of DNA methylation variance between two accessible human tissues. Sci Rep 2015; 5:8257. [PMID: 25660083 PMCID: PMC4321176 DOI: 10.1038/srep08257] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 01/13/2015] [Indexed: 01/22/2023] Open
Abstract
Population epigenetic studies have been seeking to identify differences in DNA methylation between specific exposures, demographic factors, or diseases in accessible tissues, but relatively little is known about how inter-individual variability differs between these tissues. This study presents an analysis of DNA methylation differences between matched peripheral blood mononuclear cells (PMBCs) and buccal epithelial cells (BECs), the two most accessible tissues for population studies, in 998 promoter-located CpG sites. Specifically we compared probe-wise DNA methylation variance, and how this variance related to demographic factors across the two tissues. PBMCs had overall higher DNA methylation than BECs, and the two tissues tended to differ most at genomic regions of low CpG density. Furthermore, although both tissues showed appreciable probe-wise variability, the specific regions and magnitude of variability differed strongly between tissues. Lastly, through exploratory association analysis, we found indication of differential association of BEC and PBMC with demographic variables. The work presented here offers insight into variability of DNA methylation between individuals and across tissues and helps guide decisions on the suitability of buccal epithelial or peripheral mononuclear cells for the biological questions explored by epigenetic studies in human populations.
Collapse
Affiliation(s)
- Ruiwei Jiang
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Meaghan J Jones
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Edith Chen
- Department of Psychology and Institute for Policy Research, Northwestern University, Evanston, IL 60208, United States
| | - Sarah M Neumann
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Hunter B Fraser
- Department of Biology, Stanford University, Stanford, CA 94305, United States
| | - Gregory E Miller
- Department of Psychology and Institute for Policy Research, Northwestern University, Evanston, IL 60208, United States
| | - Michael S Kobor
- 1] Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada [2] Human Early Learning Partnership, School of Population and Public Health, University of British Columbia, Vancouver, BC, V6T 1Z8, Canada
| |
Collapse
|
1010
|
Abstract
Obesity rates continue to rise throughout the world. Recent evidence has suggested that environmental factors contribute to altered energy balance regulation. However, the role of epigenetic modifications to the central control of energy homeostasis remains unknown. To investigate the role of DNA methylation in the regulation of energy balance, we investigated the role of the de novo DNA methyltransferase, Dnmt3a, in Single-minded 1 (Sim1) cells, including neurons in the paraventricular nucleus of the hypothalamus (PVH). Dnmt3a expression levels were decreased in the PVH of high-fat-fed mice. Mice lacking Dnmt3a specifically in the Sim1 neurons, which are expressed in the forebrain, including PVH, became obese with increased amounts of abdominal and subcutaneous fat. The mice were also found to have hyperphagia, decreased energy expenditure, and glucose intolerance with increased serum insulin and leptin. Furthermore, these mice developed hyper-LDL cholesterolemia when fed a high-fat diet. Gene expression profiling and DNA methylation analysis revealed that the expression of tyrosine hydroxylase and galanin were highly upregulated in the PVH of Sim1-specific Dnmt3a deletion mice. DNA methylation levels of the tyrosine hydroxylase promoter were decreased in the PVH of the deletion mice. These results suggest that Dnmt3a in the PVH is necessary for the normal control of body weight and energy homeostasis and that tyrosine hydroxylase is a putative target of Dnmt3a in the PVH. These results provide evidence for a role for Dnmt3a in the PVH to link environmental conditions to altered energy homeostasis.
Collapse
|
1011
|
Ishikawa K, Tsunekawa S, Ikeniwa M, Izumoto T, Iida A, Ogata H, Uenishi E, Seino Y, Ozaki N, Sugimura Y, Hamada Y, Kuroda A, Shinjo K, Kondo Y, Oiso Y. Long-term pancreatic beta cell exposure to high levels of glucose but not palmitate induces DNA methylation within the insulin gene promoter and represses transcriptional activity. PLoS One 2015; 10:e0115350. [PMID: 25658116 PMCID: PMC4319953 DOI: 10.1371/journal.pone.0115350] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 11/22/2014] [Indexed: 01/06/2023] Open
Abstract
Recent studies have implicated epigenetics in the pathophysiology of diabetes. Furthermore, DNA methylation, which irreversibly deactivates gene transcription, of the insulin promoter, particularly the cAMP response element, is increased in diabetes patients. However, the underlying mechanism remains unclear. We aimed to investigate insulin promoter DNA methylation in an over-nutrition state. INS-1 cells, the rat pancreatic beta cell line, were cultured under normal-culture-glucose (11.2 mmol/l) or experimental-high-glucose (22.4 mmol/l) conditions for 14 days, with or without 0.4 mmol/l palmitate. DNA methylation of the rat insulin 1 gene (Ins1) promoter was investigated using bisulfite sequencing and pyrosequencing analysis. Experimental-high-glucose conditions significantly suppressed insulin mRNA and increased DNA methylation at all five CpG sites within the Ins1 promoter, including the cAMP response element, in a time-dependent and glucose concentration-dependent manner. DNA methylation under experimental-high-glucose conditions was unique to the Ins1 promoter; however, palmitate did not affect DNA methylation. Artificial methylation of Ins1 promoter significantly suppressed promoter-driven luciferase activity, and a DNA methylation inhibitor significantly improved insulin mRNA suppression by experimental-high-glucose conditions. Experimental-high-glucose conditions significantly increased DNA methyltransferase activity and decreased ten-eleven-translocation methylcytosine dioxygenase activity. Oxidative stress and endoplasmic reticulum stress did not affect DNA methylation of the Ins1 promoter. High glucose but not palmitate increased ectopic triacylglycerol accumulation parallel to DNA methylation. Metformin upregulated insulin gene expression and suppressed DNA methylation and ectopic triacylglycerol accumulation. Finally, DNA methylation of the Ins1 promoter increased in isolated islets from Zucker diabetic fatty rats. This study helps to clarify the effect of an over-nutrition state on DNA methylation of the Ins1 promoter in pancreatic beta cells. It provides new insights into the irreversible pathophysiology of diabetes.
Collapse
Affiliation(s)
- Kota Ishikawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466–8550, Japan
| | - Shin Tsunekawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466–8550, Japan
- * E-mail:
| | - Makoto Ikeniwa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466–8550, Japan
| | - Takako Izumoto
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsushi Iida
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466–8550, Japan
| | - Hidetada Ogata
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466–8550, Japan
| | - Eita Uenishi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466–8550, Japan
| | - Yusuke Seino
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466–8550, Japan
| | - Nobuaki Ozaki
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466–8550, Japan
| | - Yoshihisa Sugimura
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466–8550, Japan
| | - Yoji Hamada
- Department of Metabolic Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akio Kuroda
- Diabetes Therapeutics and Research Center, The University of Tokushima, Tokushima, Japan
| | - Keiko Shinjo
- Department of Epigenomics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yutaka Kondo
- Department of Epigenomics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yutaka Oiso
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466–8550, Japan
| |
Collapse
|
1012
|
Chen B, Li S, Ren Q, Tong X, Zhang X, Kang L. Paternal epigenetic effects of population density on locust phase-related characteristics associated with heat-shock protein expression. Mol Ecol 2015; 24:851-62. [DOI: 10.1111/mec.13072] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 01/02/2015] [Accepted: 01/07/2015] [Indexed: 12/28/2022]
Affiliation(s)
- Bing Chen
- State Key Laboratory of Integrated Management of Pest Insects and Rodents; Institute of Zoology; Chinese Academy of Sciences; Beijing 100101 China
| | - Shaoqin Li
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory; College of Plant Science and Technology; Huazhong Agricultural University; Wuhan 430070 China
| | - Qiang Ren
- State Key Laboratory of Integrated Management of Pest Insects and Rodents; Institute of Zoology; Chinese Academy of Sciences; Beijing 100101 China
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory; College of Plant Science and Technology; Huazhong Agricultural University; Wuhan 430070 China
| | - Xiwen Tong
- State Key Laboratory of Integrated Management of Pest Insects and Rodents; Institute of Zoology; Chinese Academy of Sciences; Beijing 100101 China
| | - Xia Zhang
- Beijing Institute of Life Sciences; Chinese Academy of Sciences; Beijing 100101 China
| | - Le Kang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents; Institute of Zoology; Chinese Academy of Sciences; Beijing 100101 China
- Beijing Institute of Life Sciences; Chinese Academy of Sciences; Beijing 100101 China
| |
Collapse
|
1013
|
Brain-derived neurotrophic factor (BDNF) Val66Met polymorphism influences the association of the methylome with maternal anxiety and neonatal brain volumes. Dev Psychopathol 2015; 27:137-50. [DOI: 10.1017/s0954579414001357] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
AbstractEarly life environments interact with genotype to determine stable phenotypic outcomes. Here we examined the influence of a variant in the brain-derived neurotropic factor (BDNF) gene (Val66Met), which underlies synaptic plasticity throughout the central nervous system, on the degree to which antenatal maternal anxiety associated with neonatal DNA methylation. We also examined the association between neonatal DNA methylation and brain substructure volume, as a function of BDNF genotype. Infant, but not maternal, BDNF genotype dramatically influences the association of antenatal anxiety on the epigenome at birth as well as that between the epigenome and neonatal brain structure. There was a greater impact of antenatal maternal anxiety on the DNA methylation of infants with the methionine (Met)/Met compared to both Met/valine (Val) and Val/Val genotypes. There were significantly more cytosine–phosphate–guanine sites where methylation levels covaried with right amygdala volume among Met/Met compared with both Met/Val and Val/Val carriers. In contrast, more cytosine–phosphate–guanine sites covaried with left hippocampus volume in Val/Val infants compared with infants of the Met/Val or Met/Met genotype. Thus, antenatal Maternal Anxiety × BDNF Val66Met Polymorphism interactions at the level of the epigenome are reflected differently in the structure of the amygdala and the hippocampus. These findings suggest that BDNF genotype regulates the sensitivity of the methylome to early environment and that differential susceptibility to specific environmental conditions may be both tissue and function specific.
Collapse
|
1014
|
Oh G, Wang SC, Pal M, Chen ZF, Khare T, Tochigi M, Ng C, Yang YA, Kwan A, Kaminsky ZA, Mill J, Gunasinghe C, Tackett JL, Gottesman II, Willemsen G, de Geus EJ, Vink JM, Slagboom PE, Wray NR, Heath AC, Montgomery GW, Turecki G, Martin NG, Boomsma DI, McGuffin P, Kustra R, Petronis A. DNA modification study of major depressive disorder: beyond locus-by-locus comparisons. Biol Psychiatry 2015; 77:246-255. [PMID: 25108803 PMCID: PMC4277915 DOI: 10.1016/j.biopsych.2014.06.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 06/06/2014] [Accepted: 06/08/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Major depressive disorder (MDD) exhibits numerous clinical and molecular features that are consistent with putative epigenetic misregulation. Despite growing interest in epigenetic studies of psychiatric diseases, the methodologies guiding such studies have not been well defined. METHODS We performed DNA modification analysis in white blood cells from monozygotic twins discordant for MDD, in brain prefrontal cortex, and germline (sperm) samples from affected individuals and control subjects (total N = 304) using 8.1K CpG island microarrays and fine mapping. In addition to the traditional locus-by-locus comparisons, we explored the potential of new analytical approaches in epigenomic studies. RESULTS In the microarray experiment, we detected a number of nominally significant DNA modification differences in MDD and validated selected targets using bisulfite pyrosequencing. Some MDD epigenetic changes, however, overlapped across brain, blood, and sperm more often than expected by chance. We also demonstrated that stratification for disease severity and age may increase the statistical power of epimutation detection. Finally, a series of new analytical approaches, such as DNA modification networks and machine-learning algorithms using binary and quantitative depression phenotypes, provided additional insights on the epigenetic contributions to MDD. CONCLUSIONS Mapping epigenetic differences in MDD (and other psychiatric diseases) is a complex task. However, combining traditional and innovative analytical strategies may lead to identification of disease-specific etiopathogenic epimutations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Art Petronis
- Institute of Systems Biology and Bioinformatics, National Central University, Chungli, Taiwan.
| |
Collapse
|
1015
|
Abstract
Clinical observations and epidemiological studies have highlighted some important differences in disease course and phenotypes between pediatric inflammatory bowel disease (IBD) and adult-onset IBD. Also from a therapeutic angle, the approach to young-onset IBD is different with a more rapid introduction of azathioprine and a high threshold for long and systemic steroid use, which may affect bone mineral density and growth. The observed clinical differences have been an area of scientific research and genetic studies have been the focus of attention. Specific candidate gene studies as well as genome-wide association studies have been performed in pediatric IBD. With the exception of very early-onset IBD occurring before the age of 2 years; no overt differences in genetic susceptibility have been identified. In contrast, very early-onset IBD seems in particular to be a genetic disease with defects in the IL10 signaling pathway being the principal example. This review aims to answer some straightforward questions arising in this topic by giving concise information.
Collapse
Affiliation(s)
- Thomas Billiet
- Department of Gastroenterology, University Hospitals Leuven, Herestraat 49 - B-3000 Leuven Belgium
| | | |
Collapse
|
1016
|
Fan J, Krautkramer KA, Feldman JL, Denu JM. Metabolic regulation of histone post-translational modifications. ACS Chem Biol 2015; 10:95-108. [PMID: 25562692 DOI: 10.1021/cb500846u] [Citation(s) in RCA: 242] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Histone post-translational modifications regulate transcription and other DNA-templated functions. This process is dynamically regulated by specific modifying enzymes whose activities require metabolites that either serve as cosubstrates or act as activators/inhibitors. Therefore, metabolism can influence histone modification by changing local concentrations of key metabolites. Physiologically, the epigenetic response to metabolism is important for nutrient sensing and environment adaption. In pathologic states, the connection between metabolism and histone modification mediates epigenetic abnormality in complex disease. In this review, we summarize recent studies of the molecular mechanisms involved in metabolic regulation of histone modifications and discuss their biological significance.
Collapse
Affiliation(s)
- Jing Fan
- Department of Biomolecular Chemistry and the Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin 53715, United States
| | - Kimberly A. Krautkramer
- Department of Biomolecular Chemistry and the Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin 53715, United States
| | - Jessica L. Feldman
- Department of Biomolecular Chemistry and the Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin 53715, United States
| | - John M. Denu
- Department of Biomolecular Chemistry and the Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin 53715, United States
| |
Collapse
|
1017
|
Fazeli A, Moein Vaziri N, Holt WV. Proteomics of the periconception milieu. Proteomics 2015; 15:649-55. [PMID: 25404351 DOI: 10.1002/pmic.201400362] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 11/09/2014] [Accepted: 11/13/2014] [Indexed: 01/11/2023]
Abstract
There is increasing realisation that human health status in adulthood depends critically upon environmental conditions pertaining around the time of conception and during pregnancy. Poor maternal diet or adverse environmental conditions around the periconception period somehow induces the resultant embryo to adapt predictively in order to survive this level of stress for the whole of its life. However, if there is a mismatch between expectation and reality, where the conditions during later life are better than expected, things go wrong and the adult suffers a range of illnesses, including diabetes, heart disease, hypertension and stroke. Understanding the molecular signals that direct the early embryo to adopt appropriate adaptations to suit its future life would be extremely valuable. However, although it appears to be an ideal task for proteomic applications, there are technical, ethical and practical limitations to what can be achieved with the current framework of proteomic technology. Here, we review what has been achieved to date, explain some of the experimental problems and suggest some strategies for taking this field forward.
Collapse
Affiliation(s)
- Alireza Fazeli
- Academic Unit of Reproductive and Developmental Medicine, University of Sheffield, Sheffield, UK
| | | | | |
Collapse
|
1018
|
Na HK, Woo JH. Helicobacter pylori Induces Hypermethylation of CpG Islands Through Upregulation of DNA Methyltransferase: Possible Involvement of Reactive Oxygen/Nitrogen Species. J Cancer Prev 2015; 19:259-64. [PMID: 25574460 PMCID: PMC4285956 DOI: 10.15430/jcp.2014.19.4.259] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 12/23/2014] [Accepted: 12/24/2014] [Indexed: 12/27/2022] Open
Abstract
Helicobacter pylori infection has been considered to be one of the major factors implicated in etiology of gastric cancer. Aberrant DNA methylation accounts for epigenetic modifications induced by H. pylori. H. pylori-induced hypermethylation has been linked to enhancement of the rates of metastasis and recurrence in gastric cancer patients. H. pylori-induced gene hypermethylation has been known to be associated with inflammation. However, the molecular mechanisms underlying H. pylori-induced hypermethylation remain largely unknown. This review highlights possible involvement of reactive oxygen/nitrogen species in H. pylori-induced hypermethylation and gastric carcinogenesis.
Collapse
Affiliation(s)
- Hye-Kyung Na
- Department of Food and Nutrition, College of Human Ecology, Sungshin Women's University, Seoul, Korea
| | - Jeong-Hwa Woo
- Department of Food and Nutrition, College of Human Ecology, Sungshin Women's University, Seoul, Korea
| |
Collapse
|
1019
|
Hagmann J, Becker C, Müller J, Stegle O, Meyer RC, Wang G, Schneeberger K, Fitz J, Altmann T, Bergelson J, Borgwardt K, Weigel D. Century-scale methylome stability in a recently diverged Arabidopsis thaliana lineage. PLoS Genet 2015; 11:e1004920. [PMID: 25569172 PMCID: PMC4287485 DOI: 10.1371/journal.pgen.1004920] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 11/24/2014] [Indexed: 01/17/2023] Open
Abstract
There has been much excitement about the possibility that exposure to specific environments can induce an ecological memory in the form of whole-sale, genome-wide epigenetic changes that are maintained over many generations. In the model plant Arabidopsis thaliana, numerous heritable DNA methylation differences have been identified in greenhouse-grown isogenic lines, but it remains unknown how natural, highly variable environments affect the rate and spectrum of such changes. Here we present detailed methylome analyses in a geographically dispersed A. thaliana population that constitutes a collection of near-isogenic lines, diverged for at least a century from a common ancestor. Methylome variation largely reflected genetic distance, and was in many aspects similar to that of lines raised in uniform conditions. Thus, even when plants are grown in varying and diverse natural sites, genome-wide epigenetic variation accumulates mostly in a clock-like manner, and epigenetic divergence thus parallels the pattern of genome-wide DNA sequence divergence. It continues to be hotly debated to what extent environmentally induced epigenetic change is stably inherited and thereby contributes to short-term adaptation. It has been shown before that natural Arabidopsis thaliana lines differ substantially in their methylation profiles. How much of this is independent of genetic changes remains, however, unclear, especially given that there is very little conservation of methylation between species, simply because the methylated sequences themselves, mostly repeats, are not conserved over millions of years. On the other hand, there is no doubt that artificially induced epialleles can contribute to phenotypic variation. To investigate whether epigenetic differentiation, at least in the short term, proceeds very differently from genetic variation, and whether genome-wide epigenetic fingerprints can be used to uncover local adaptation, we have taken advantage of a near-clonal North American A. thaliana population that has diverged under natural conditions for at least a century. We found that both patterns and rates of methylome variation were in many aspects similar to those of lines grown in stable environments, which suggests that environment-induced changes are only minor contributors to durable genome-wide heritable epigenetic variation.
Collapse
Affiliation(s)
- Jörg Hagmann
- Department of Molecular Biology, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Claude Becker
- Department of Molecular Biology, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Jonas Müller
- Department of Molecular Biology, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Oliver Stegle
- Machine Learning and Computational Biology Research Group, Max Planck Institute for Developmental Biology and Max Planck Institute for Intelligent Systems, Tübingen, Germany
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Rhonda C. Meyer
- The Leibniz Institute of Plant Genetics and Crop Plant Research, Gatersleben, Germany
| | - George Wang
- Department of Molecular Biology, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Korbinian Schneeberger
- Department of Plant Developmental Biology, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Joffrey Fitz
- Department of Molecular Biology, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Thomas Altmann
- The Leibniz Institute of Plant Genetics and Crop Plant Research, Gatersleben, Germany
| | - Joy Bergelson
- Department of Ecology and Evolution, University of Chicago, Chicago, Illinois, United States of America
| | - Karsten Borgwardt
- Machine Learning and Computational Biology Research Group, Max Planck Institute for Developmental Biology and Max Planck Institute for Intelligent Systems, Tübingen, Germany
- Center for Bioinformatics (ZBIT), Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Detlef Weigel
- Department of Molecular Biology, Max Planck Institute for Developmental Biology, Tübingen, Germany
- * E-mail:
| |
Collapse
|
1020
|
Schamberger AC, Mise N, Meiners S, Eickelberg O. Epigenetic mechanisms in COPD: implications for pathogenesis and drug discovery. Expert Opin Drug Discov 2015; 9:609-28. [PMID: 24850530 DOI: 10.1517/17460441.2014.913020] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Chronic obstructive pulmonary disease (COPD) is the fourth leading cause of death worldwide. The growing burden of COPD is due to continuous tobacco use, which is the most important risk factor of the disease, indoor fumes, occupational exposures and also aging of the world's population. Epigenetic mechanisms significantly contribute to COPD pathophysiology. AREAS COVERED This review focuses on disease-relevant changes in DNA modification, histone modification and non-coding RNA expression in COPD, and provides insight into novel therapeutic approaches modulating epigenetic mechanisms. Recent findings revealed, among others, globally changed DNA methylation patterns, decreased levels of histone deacetylases and reduced microRNAs levels in COPD. The authors also discuss a potential role of the chromatin silencing Polycomb group of proteins in COPD. EXPERT OPINION COPD is a highly complex disease and therapy development is complicated by the fact that many smokers develop both COPD and lung cancer. Of interest, combination therapies involving DNA methyltransferase inhibitors and anti-inflammatory drugs provide a promising approach, as they might be therapeutic for both COPD and cancer. Although the field of epigenetic research has virtually exploded over the last 10 years, particular efforts are required to enhance our knowledge of the COPD epigenome in order to successfully establish epigenetic-based therapies for this widespread disease.
Collapse
Affiliation(s)
- Andrea C Schamberger
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, University Hospital and Ludwig-Maximilians-University, Member of the German Center for Lung Research (DZL) , Max-Lebsche-Platz 31, 81377 Munich , Germany
| | | | | | | |
Collapse
|
1021
|
O'Doherty AM, McGettigan PA. Epigenetic processes in the male germline. Reprod Fertil Dev 2015; 27:725-38. [DOI: 10.1071/rd14167] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 08/01/2014] [Indexed: 12/12/2022] Open
Abstract
Sperm undergo some of the most extensive chromatin modifications seen in mammalian biology. During male germline development, paternal DNA methylation marks are erased and established on a global scale through waves of demethylation and de novo methylation. As spermatogenesis progresses, the majority of the histones are removed and replaced by protamines, enabling a tighter packaging of the DNA and transcriptional shutdown. Following fertilisation, the paternal genome is rapidly reactivated, actively demethylated, the protamines are replaced with histones and the embryonic genome is activated. The development of new assays, made possible by high-throughput sequencing technology, has resulted in the revisiting of what was considered settled science regarding the state of DNA packaging in mammalian spermatozoa. Researchers have discovered that not all histones are replaced by protamines and, in certain experiments, various species of RNA have been detected in what was previously considered transcriptionally quiescent spermatozoa. Most controversially, several groups have suggested that environmental modifications of the epigenetic state of spermatozoa may operate as a non-DNA-based form of inheritance, a process known as ‘transgenerational epigenetic inheritance’. Other developments in the field include the increased focus on the involvement of short RNAs, such as microRNAs, long non-coding RNAs and piwi-interacting RNAs. There has also been an accumulation of evidence illustrating associations between defects in sperm DNA packaging and disease and fertility. In this paper we review the literature, recent findings and areas of controversy associated with epigenetic processes in the male germline, focusing on DNA methylation dynamics, non-coding RNAs, the biology of sperm chromatin packaging and transgenerational inheritance.
Collapse
|
1022
|
Fernández AF, Bayón GF, Urdinguio RG, Toraño EG, García MG, Carella A, Petrus-Reurer S, Ferrero C, Martinez-Camblor P, Cubillo I, García-Castro J, Delgado-Calle J, Pérez-Campo FM, Riancho JA, Bueno C, Menéndez P, Mentink A, Mareschi K, Claire F, Fagnani C, Medda E, Toccaceli V, Brescianini S, Moran S, Esteller M, Stolzing A, de Boer J, Nisticò L, Stazi MA, Fraga MF. H3K4me1 marks DNA regions hypomethylated during aging in human stem and differentiated cells. Genome Res 2015; 25:27-40. [PMID: 25271306 PMCID: PMC4317171 DOI: 10.1101/gr.169011.113] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 09/23/2014] [Indexed: 12/17/2022]
Abstract
In differentiated cells, aging is associated with hypermethylation of DNA regions enriched in repressive histone post-translational modifications. However, the chromatin marks associated with changes in DNA methylation in adult stem cells during lifetime are still largely unknown. Here, DNA methylation profiling of mesenchymal stem cells (MSCs) obtained from individuals aged 2 to 92 yr identified 18,735 hypermethylated and 45,407 hypomethylated CpG sites associated with aging. As in differentiated cells, hypermethylated sequences were enriched in chromatin repressive marks. Most importantly, hypomethylated CpG sites were strongly enriched in the active chromatin mark H3K4me1 in stem and differentiated cells, suggesting this is a cell type-independent chromatin signature of DNA hypomethylation during aging. Analysis of scedasticity showed that interindividual variability of DNA methylation increased during aging in MSCs and differentiated cells, providing a new avenue for the identification of DNA methylation changes over time. DNA methylation profiling of genetically identical individuals showed that both the tendency of DNA methylation changes and scedasticity depended on nongenetic as well as genetic factors. Our results indicate that the dynamics of DNA methylation during aging depend on a complex mixture of factors that include the DNA sequence, cell type, and chromatin context involved and that, depending on the locus, the changes can be modulated by genetic and/or external factors.
Collapse
Affiliation(s)
- Agustín F Fernández
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Gustavo F Bayón
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Rocío G Urdinguio
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Estela G Toraño
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, 33006 Oviedo, Spain
| | - María G García
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Antonella Carella
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Sandra Petrus-Reurer
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Cecilia Ferrero
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Pablo Martinez-Camblor
- Oficina de Investigación Biosanitaria (OIB-FICYT) de Asturias, 33005 Oviedo, Spain and Universidad Autónoma de Chile, Chile
| | - Isabel Cubillo
- Unidad de Biotecnología Celular, Área de Genética Humana, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier García-Castro
- Unidad de Biotecnología Celular, Área de Genética Humana, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jesús Delgado-Calle
- Department of Internal Medicine, Hospital U.M. Valdecilla, University of Cantabria, IDIVAL, 39011 Santander, Spain
| | - Flor M Pérez-Campo
- Department of Internal Medicine, Hospital U.M. Valdecilla, University of Cantabria, IDIVAL, 39011 Santander, Spain
| | - José A Riancho
- Department of Internal Medicine, Hospital U.M. Valdecilla, University of Cantabria, IDIVAL, 39011 Santander, Spain
| | - Clara Bueno
- Josep Carreras Leukemia Research Institute, School of Medicine, University of Barcelona, 08036 Barcelona, Spain
| | - Pablo Menéndez
- Josep Carreras Leukemia Research Institute, School of Medicine, University of Barcelona, 08036 Barcelona, Spain; Institut Català de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Anouk Mentink
- MIRA Institute of Biomedical Technology and Technical Medicine, University of Twente, 7500 AE Enschede, The Netherlands
| | - Katia Mareschi
- Pediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, City of Science and Health of Turin, Regina Margherita Children's Hospital, 10126 Turin, Italy; Department of Public Health and Pediatrics, University of Turin, 10126 Turin, Italy
| | - Fabian Claire
- Translational Centre for Regenerative Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Corrado Fagnani
- Genetic Epidemiology Unit, National Centre of Epidemiology, Surveillance and Health Promotion; Istituto Superiore di Sanità; Viale Regina Elena 299, 00161, Rome, Italy
| | - Emanuela Medda
- Genetic Epidemiology Unit, National Centre of Epidemiology, Surveillance and Health Promotion; Istituto Superiore di Sanità; Viale Regina Elena 299, 00161, Rome, Italy
| | - Virgilia Toccaceli
- Genetic Epidemiology Unit, National Centre of Epidemiology, Surveillance and Health Promotion; Istituto Superiore di Sanità; Viale Regina Elena 299, 00161, Rome, Italy
| | - Sonia Brescianini
- Genetic Epidemiology Unit, National Centre of Epidemiology, Surveillance and Health Promotion; Istituto Superiore di Sanità; Viale Regina Elena 299, 00161, Rome, Italy
| | - Sebastián Moran
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), 08908 Barcelona, Catalonia, Spain
| | - Manel Esteller
- Institut Català de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain; Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), 08908 Barcelona, Catalonia, Spain; Department of Physiological Sciences II, School of Medicine, University of Barcelona, 08036 Barcelona, Catalonia, Spain
| | - Alexandra Stolzing
- Translational Centre for Regenerative Medicine, University of Leipzig, 04103 Leipzig, Germany; Loughborough University, Wolfson School of Mechanical and Manufacturing Engineering, LE11 3TU Loughborough, United Kingdom
| | - Jan de Boer
- MIRA Institute of Biomedical Technology and Technical Medicine, University of Twente, 7500 AE Enschede, The Netherlands; cBITE laboratory, Merln Institute of Technology-inspired Regenerative Medicine, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Lorenza Nisticò
- Genetic Epidemiology Unit, National Centre of Epidemiology, Surveillance and Health Promotion; Istituto Superiore di Sanità; Viale Regina Elena 299, 00161, Rome, Italy
| | - Maria A Stazi
- Genetic Epidemiology Unit, National Centre of Epidemiology, Surveillance and Health Promotion; Istituto Superiore di Sanità; Viale Regina Elena 299, 00161, Rome, Italy
| | - Mario F Fraga
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, 33006 Oviedo, Spain; Department of Immunology and Oncology, National Center for Biotechnology, CNB-CSIC, Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
1023
|
Mansoori AA, Jain SK. Molecular Links between Alcohol and Tobacco Induced DNA Damage, Gene Polymorphisms and Patho-physiological Consequences: A Systematic Review of Hepatic Carcinogenesis. Asian Pac J Cancer Prev 2015; 16:4803-12. [PMID: 26163595 DOI: 10.7314/apjcp.2015.16.12.4803] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Chronic alcohol and tobacco abuse plays a crucial role in the development of different liver associated disorders. Intake promotes the generation of reactive oxygen species within hepatic cells exposing their DNA to continuous oxidative stress which finally leads to DNA damage. However in response to such damage an entangled protective repair machinery comprising different repair proteins like ATM, ATR, H2AX, MRN complex becomes activated. Under abnormal conditions the excessive reactive oxygen species generation results in genetic predisposition of various genes (as ADH, ALDH, CYP2E1, GSTT1, GSTP1 and GSTM1) involved in xenobiotic metabolic pathways, associated with susceptibility to different liver related diseases such as fibrosis, cirrhosis and hepatocellular carcinoma. There is increasing evidence that the inflammatory process is inherently associated with many different cancer types, including hepatocellular carcinomas. The generated reactive oxygen species can also activate or repress epigenetic elements such as chromatin remodeling, non-coding RNAs (micro-RNAs), DNA (de) methylation and histone modification that affect gene expression, hence leading to various disorders. The present review provides comprehensive knowledge of different molecular mechanisms involved in gene polymorphism and their possible association with alcohol and tobacco consumption. The article also showcases the necessity of identifying novel diagnostic biomarkers for early cancer risk assessment among alcohol and tobacco users.
Collapse
Affiliation(s)
- Abdul Anvesh Mansoori
- Molecular Biology Laboratory, Department of Biotechnology, Dr. Hari Singh Gour Central University, Sagar, M.P. India E-mail :
| | | |
Collapse
|
1024
|
Abstract
The mammalian gastrointestinal tract is home to trillions of commensal microorganisms that collectively make up the intestinal microbiota. These microbes are important environmental factors that regulate homeostasis, and alterations in the composition of the microbiota have been associated with several diseases, including inflammatory bowel disease, diabetes, and cancer. New research is beginning to uncover epigenomic pathways that may regulate this relationship with the microbiota. Epigenomic modifications alter the structure of the chromatin and therefore regulate the transcriptional program of a cell. These modifications are maintained by the dynamic activity of various modifying and demodifying enzymes, the activities of which can be influenced by metabolites and other environmental cues. Histone deacetylases (HDACs) are a class of epigenomic-modifying enzymes that are regulated by both endogenous and exogenous factors, and recent studies have suggested that host HDAC expression is important for regulating communication between the intestinal microbiota and mammalian host cells.
Collapse
Affiliation(s)
- Theresa Alenghat
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
1025
|
|
1026
|
Beaujean N. Epigenetics, embryo quality and developmental potential. Reprod Fertil Dev 2015; 27:53-62. [DOI: 10.1071/rd14309] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
It is very important for embryologists to understand how parental inherited genomes are reprogrammed after fertilisation in order to obtain good-quality embryos that will sustain further development. In mammals, it is now well established that important epigenetic modifications occur after fertilisation. Although gametes carry special epigenetic signatures, they should attain embryo-specific signatures, some of which are crucial for the production of healthy embryos. Indeed, it appears that proper establishment of different epigenetic modifications and subsequent scaffolding of the chromatin are crucial steps during the first cleavages. This ‘reprogramming’ is promoted by the intimate contact between the parental inherited genomes and the oocyte cytoplasm after fusion of the gametes. This review introduces two main epigenetic players, namely histone post-translational modifications and DNA methylation, and highlights their importance during early embryonic development.
Collapse
|
1027
|
PGC1α -1 Nucleosome Position and Splice Variant Expression and Cardiovascular Disease Risk in Overweight and Obese Individuals. PPAR Res 2014; 2014:895734. [PMID: 25614734 PMCID: PMC4295622 DOI: 10.1155/2014/895734] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 11/26/2014] [Indexed: 11/17/2022] Open
Abstract
PGC1α, a transcriptional coactivator, interacts with PPARs and others to regulate skeletal muscle metabolism. PGC1α undergoes splicing to produce several mRNA variants, with the NTPGC1α variant having a similar biological function to the full length PGC1α (FLPGC1α). CVD is associated with obesity and T2D and a lower percentage of type 1 oxidative fibers and impaired mitochondrial function in skeletal muscle, characteristics determined by PGC1α expression. PGC1α expression is epigenetically regulated in skeletal muscle to determine mitochondrial adaptations, and epigenetic modifications may regulate mRNA splicing. We report in this paper that skeletal muscle PGC1α -1 nucleosome (-1N) position is associated with splice variant NTPGC1α but not FLPGC1α expression. Division of participants based on the -1N position revealed that those individuals with a -1N phased further upstream from the transcriptional start site (UP) expressed lower levels of NTPGC1α than those with the -1N more proximal to TSS (DN). UP showed an increase in body fat percentage and serum total and LDL cholesterol. These findings suggest that the -1N may be a potential epigenetic regulator of NTPGC1α splice variant expression, and -1N position and NTPGC1α variant expression in skeletal muscle are linked to CVD risk. This trial is registered with clinicaltrials.gov, identifier NCT00458133.
Collapse
|
1028
|
Dicer and Hsp104 function in a negative feedback loop to confer robustness to environmental stress. Cell Rep 2014; 10:47-61. [PMID: 25543137 DOI: 10.1016/j.celrep.2014.12.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 11/06/2014] [Accepted: 12/02/2014] [Indexed: 11/22/2022] Open
Abstract
Epigenetic mechanisms can be influenced by environmental cues and thus evoke phenotypic variation. This plasticity can be advantageous for adaptation but also detrimental if not tightly controlled. Although having attracted considerable interest, it remains largely unknown if and how environmental cues such as temperature trigger epigenetic alterations. Using fission yeast, we demonstrate that environmentally induced discontinuous phenotypic variation is buffered by a negative feedback loop that involves the RNase Dicer and the protein disaggregase Hsp104. In the absence of Hsp104, Dicer accumulates in cytoplasmic inclusions and heterochromatin becomes unstable at elevated temperatures, an epigenetic state inherited for many cell divisions after the heat stress. Loss of Dicer leads to toxic aggregation of an exogenous prionogenic protein. Our results highlight the importance of feedback regulation in building epigenetic memory and uncover Hsp104 and Dicer as homeostatic controllers that buffer environmentally induced stochastic epigenetic variation and toxic aggregation of prionogenic proteins.
Collapse
|
1029
|
Jiang R, Jones MJ, Sava F, Kobor MS, Carlsten C. Short-term diesel exhaust inhalation in a controlled human crossover study is associated with changes in DNA methylation of circulating mononuclear cells in asthmatics. Part Fibre Toxicol 2014; 11:71. [PMID: 25487561 PMCID: PMC4268899 DOI: 10.1186/s12989-014-0071-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 11/24/2014] [Indexed: 11/10/2022] Open
Abstract
Background Changes in DNA methylation have been associated with traffic-related air pollution in observational studies, but the specific mechanisms and temporal dynamics therein have not been explored in a controlled study of asthmatics. In this study, we investigate short-term effects of diesel exhaust inhalation on DNA methylation levels at CpG sites across the genome in circulating blood in asthmatics. Methods A double-blind crossover study of filtered air and diesel exhaust exposures was performed on sixteen non-smoking asthmatic subjects. Blood samples were collected pre-exposure, and then 6 and 30 hours post-exposure. Peripheral blood mononuclear cell DNA methylation was interrogated using the Illumina Infinium HumanMethylation450 Array. Exposure-related changes in DNA methylation were identified. In addition, CpG sites overlapping with Alu or LINE1 repetitive elements and candidate microRNA loci were also analyzed. Results DNA methylation at 2827 CpG sites were affected by exposure to diesel exhaust but not filtered air; these sites enriched for genes involved in protein kinase and NFkB pathways. CpG sites with significant changes in response to diesel exhaust exposure primarily became less methylated, with a site residing within GSTP1 being among the significant hits. Diesel exhaust-associated change was also found for CpG sites overlapping with Alu and LINE1 elements as well as for a site within miR-21. Conclusion Short-term exposure to diesel exhaust resulted in DNA methylation changes at CpG sites residing in genes involved in inflammation and oxidative stress response, repetitive elements, and microRNA. This provides plausibility for the role of DNA methylation in pathways by which airborne particulate matter impacts gene expression and offers support for including DNA methylation analysis in future efforts to understand the interactions between environmental exposures and biological systems. Electronic supplementary material The online version of this article (doi:10.1186/s12989-014-0071-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ruiwei Jiang
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, 950 west 28th Avenue, Vancouver, V5Z4H4, Canada.
| | - Meaghan J Jones
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, 950 west 28th Avenue, Vancouver, V5Z4H4, Canada.
| | - Francesco Sava
- Air Pollution Exposure Laboratory, Chan-Yeung Centre for Occupational and Environmental Lung Disease, Department of Medicine, Division of Respiratory Medicine, University of British Columbia, 2775 Laurel Street, Vancouver, British Columbia, V5Z1L9, Canada.
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, 950 west 28th Avenue, Vancouver, V5Z4H4, Canada. .,Human Early Learning Partnership, School of Population and Public Health, University of British Columbia, 2206 East Mall, Vancouver, British Columbia, V6T1Z3, Canada.
| | - Christopher Carlsten
- Air Pollution Exposure Laboratory, Chan-Yeung Centre for Occupational and Environmental Lung Disease, Department of Medicine, Division of Respiratory Medicine, University of British Columbia, 2775 Laurel Street, Vancouver, British Columbia, V5Z1L9, Canada.
| |
Collapse
|
1030
|
Choi Y, Mango SE. Hunting for Darwin's gemmules and Lamarck's fluid: Transgenerational signaling and histone methylation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1839:1440-53. [DOI: 10.1016/j.bbagrm.2014.05.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Revised: 05/07/2014] [Accepted: 05/13/2014] [Indexed: 01/22/2023]
|
1031
|
Martins-Júnior HA, Pinaffi FLV, Simas RC, Tarouco AK, Ferreira CR, Silva LA, Nogueira GP, Meirelles FV, Eberlin MN, Perecin F. Plasma steroid dynamics in late- and near-term naturally and artificially conceived bovine pregnancies as elucidated by multihormone high-resolution LC-MS/MS. Endocrinology 2014; 155:5011-23. [PMID: 25299569 DOI: 10.1210/en.2013-2166] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The plasma levels of corticosteroids and sex steroids during pregnancy are key indicators of mammalian placental function and the onset of parturition. Steroid hormones are believed to be disturbed in pregnancies produced using assisted reproductive technologies (ARTs) due to placental dysfunction and the frequently observed lack of parturition signals. To elucidate the plasma steroid dynamics, a liquid chromatography-tandem mass spectrometry method was developed and used to determine the levels of corticosteroids (corticosterone, 11-deoxycortisol, and cortisol) and their direct precursors (progesterone and 17α-OH-progesterone) as well as sex steroids (androstenedione, estrone, estrone sulfate, testosterone, and 17β-estradiol) in bovine plasma. The levels of these 10 steroids in recipient cows carrying naturally conceived (control), in vitro fertilized (IVF), or cloned (somatic cell nuclear transfer) conceptuses were compared during late-term pregnancy (30 days before parturition), during near-term pregnancy (1 day before parturition), and on the day of parturition (day 0). Significant differences were observed among the corticosteroid levels: higher levels of corticosterone, 11-deoxycortisol, and cortisol were detected in cloned pregnancies at day 30; lower levels of corticosterone were observed in ART-derived pregnancies at days 1 and 0; and estrone and estradiol levels were higher in IVF pregnancies throughout the final development. These results suggested an upregulation of the P450C11 and P450C21 enzymes 30 days before parturition in somatic cell nuclear transfer pregnancies and an overactivation of the aromatase enzyme in IVF pregnancies. Taken together, the monitoring of multiple steroid hormones revealed that the pregnancies obtained using ART exhibited plasma steroid concentration dynamics compatible with the dysregulation of steroidogenic tissues.
Collapse
Affiliation(s)
- Helio A Martins-Júnior
- Thomson Mass Spectrometry Laboratory (H.A.M.-J., R.C.S., C.R.F., M.N.E.), Institute of Chemistry, University of Campinas, Campinas, São Paulo, Brazil 13083-970; AB SCIEX of Brazil (H.A.M.-J.), São Paulo, São Paulo, Brazil 04719-002; Laboratory of Theriogenology Dr O. J. Ginther (F.L.V.P., L.A.S.), Department of Veterinary Medicine, School of Animal Sciences and Food Engineering (FZEA), University of São Paulo, Pirassununga, São Paulo, Brazil 13635-900; Laboratory of Molecular Morphophysiology and Development (A.K.T., F.V.M., F.P.), Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil 13635-900; Department of Production and Animal Health (R.C.S., G.P.N.), School of Veterinary Medicine, São Paulo State University, Araçatuba, São Paulo, Brazil 16050-680; and State Foundation of Agricultural Research (A.K.T.), Research Center Iwar Beckman, Hulha Negra, Rio Grande do Sul, Brazil 96400-970
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1032
|
Ramakrishna N, Meeker HC, Brown WT. Novel Epigenetic Regulation of Alpha-Synuclein Expression in Down Syndrome. Mol Neurobiol 2014; 53:155-162. [DOI: 10.1007/s12035-014-8979-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 10/31/2014] [Indexed: 10/24/2022]
|
1033
|
Daskalakis NP, Yehuda R. Site-specific methylation changes in the glucocorticoid receptor exon 1F promoter in relation to life adversity: systematic review of contributing factors. Front Neurosci 2014; 8:369. [PMID: 25484853 PMCID: PMC4240065 DOI: 10.3389/fnins.2014.00369] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 10/28/2014] [Indexed: 12/20/2022] Open
Abstract
There has been recent interest in epigenetics in psychiatry since it offers a means of understanding how stressful life experiences, in interaction with the genotype, result in epigenetic changes that result in altered gene expression, ultimately affecting the risk for mental disorders. Many studies focused on methylation of the glucocorticoid receptor exon 1F promoter following an initial observation that changes in this region could be modulated by the environment. This review examines all published studies that have attempted to measure methylation in this region using different techniques, several tissue types, populations at different behavioral state and stages of development. Methodological issues have been raised with the aim of attempting to understand methylation quantification and site of action. We propose that it is useful to examine whether methylation at specific sites within the promoter region may be particularly relevant to psychiatric vulnerability to stress-related outcomes.
Collapse
Affiliation(s)
- Nikolaos P Daskalakis
- Traumatic Stress Studies Division, Department of Psychiatry, Icahn School of Medicine at Mount Sinai New York, NY, USA ; Mental Health Patient Care Center, James J. Peters Veterans Affairs Medical Center Bronx, New York, NY, USA ; Laboratory of Molecular Neuropsychiatry, Department of Psychiatry, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Rachel Yehuda
- Traumatic Stress Studies Division, Department of Psychiatry, Icahn School of Medicine at Mount Sinai New York, NY, USA ; Mental Health Patient Care Center, James J. Peters Veterans Affairs Medical Center Bronx, New York, NY, USA ; Department of Neuroscience, Icahn School of Medicine at Mount Sinai New York, NY, USA
| |
Collapse
|
1034
|
Harrisson KA, Pavlova A, Telonis-Scott M, Sunnucks P. Using genomics to characterize evolutionary potential for conservation of wild populations. Evol Appl 2014; 7:1008-25. [PMID: 25553064 PMCID: PMC4231592 DOI: 10.1111/eva.12149] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 02/10/2014] [Indexed: 12/16/2022] Open
Abstract
Genomics promises exciting advances towards the important conservation goal of maximizing evolutionary potential, notwithstanding associated challenges. Here, we explore some of the complexity of adaptation genetics and discuss the strengths and limitations of genomics as a tool for characterizing evolutionary potential in the context of conservation management. Many traits are polygenic and can be strongly influenced by minor differences in regulatory networks and by epigenetic variation not visible in DNA sequence. Much of this critical complexity is difficult to detect using methods commonly used to identify adaptive variation, and this needs appropriate consideration when planning genomic screens, and when basing management decisions on genomic data. When the genomic basis of adaptation and future threats are well understood, it may be appropriate to focus management on particular adaptive traits. For more typical conservations scenarios, we argue that screening genome-wide variation should be a sensible approach that may provide a generalized measure of evolutionary potential that accounts for the contributions of small-effect loci and cryptic variation and is robust to uncertainty about future change and required adaptive response(s). The best conservation outcomes should be achieved when genomic estimates of evolutionary potential are used within an adaptive management framework.
Collapse
Affiliation(s)
| | - Alexandra Pavlova
- School of Biological Sciences, Monash UniversityMelbourne, Vic., Australia
| | | | - Paul Sunnucks
- School of Biological Sciences, Monash UniversityMelbourne, Vic., Australia
| |
Collapse
|
1035
|
Shahabi P, Siest G, Meyer UA, Visvikis-Siest S. Human cytochrome P450 epoxygenases: Variability in expression and role in inflammation-related disorders. Pharmacol Ther 2014; 144:134-61. [DOI: 10.1016/j.pharmthera.2014.05.011] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 05/15/2014] [Indexed: 12/19/2022]
|
1036
|
Davinelli S, Calabrese V, Zella D, Scapagnini G. Epigenetic nutraceutical diets in Alzheimer's disease. J Nutr Health Aging 2014; 18:800-5. [PMID: 25389957 DOI: 10.1007/s12603-014-0552-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
There is growing support that environmental influences and individual genetic susceptibility may increase the incidence and accelerate the onset of Alzheimer's disease (AD). Epigenetic mechanisms encompass a complex regulatory network of modifications with considerable impact on health and disease risk. Abnormal epigenetic regulation is a hallmark in many pathological conditions including AD. It is well recognized that numerous bioactive dietary components mediate epigenetic modifications associated with the pathophysiology of several diseases. Although the influences of dietary factors on epigenetic regulation have been extensively investigated, only few studies have explored the effects of specific food components in regulating epigenetic patterns during neurodegeneration and AD. Epigenetic nutritional research has substantial potential for AD and may represent a window of opportunity to complement other interventions. Here, we provide a brief overview of the main mechanisms involved in AD, some of which may be epigenetically modulated by bioactive food.
Collapse
Affiliation(s)
- S Davinelli
- Giovanni Scapagnini, Department of Medicine and Health Sciences, University of Molise, Campobasso 86100, Italy,
| | | | | | | |
Collapse
|
1037
|
Epigenomic regulation of host-microbiota interactions. Trends Immunol 2014; 35:518-25. [PMID: 25443494 DOI: 10.1016/j.it.2014.09.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 09/21/2014] [Accepted: 09/22/2014] [Indexed: 02/07/2023]
Abstract
The trillions of beneficial commensal microorganisms that normally reside in the gastrointestinal tract have emerged as a critical source of environmentally-derived stimuli that can impact health and disease. However, the underlying cellular and molecular mechanisms that recognize commensal bacteria-derived signals and regulate mammalian homeostasis are just beginning to be defined. Highly coordinated epigenomic modifications allow mammals to alter the transcriptional program of a cell in response to environmental cues. These modifications may play a key role in regulating the dynamic relationship between mammals and their microbiota. We review recent advances in understanding the interplay between the microbiota and mammalian epigenomic pathways, and highlight emerging findings that implicate a central role for histone deacetylases (HDACs) in orchestrating host-microbiota interactions.
Collapse
|
1038
|
Campanella G, Polidoro S, Di Gaetano C, Fiorito G, Guarrera S, Krogh V, Palli D, Panico S, Sacerdote C, Tumino R, Elliott P, Matullo G, Chadeau-Hyam M, Vineis P. Epigenetic signatures of internal migration in Italy. Int J Epidemiol 2014; 44:1442-1449. [PMID: 25324151 PMCID: PMC4588856 DOI: 10.1093/ije/dyu198] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Observational studies have suggested that the risks of non-communicable diseases in voluntary migrants become similar to those in the host population after one or more generations, supporting the hypothesis that these diseases have a predominantly environmental (rather than inherited) origin. However, no study has been conducted thus far to identify alterations at the molecular level that might mediate these changes in disease risk after migration. METHODS Using genome-wide DNA methylation profiles from more than 1000 Italian participants, we conducted an epigenome-wide association study (EWAS) to identify differences between south-to-north migrants and their origin (southern natives) and host (north-western natives) populations. RESULTS We identified several differentially methylated CpG loci, in particular when comparing south-to-north migrants with north-western natives. We hypothesise that these alterations may underlie an adaptive response to exposure differentials that exist between origin and host populations. CONCLUSIONS Our study is the first large agnostic investigation of DNA methylation changes linked to migratory processes, and shows the potential of EWAS to investigate their biological effects.
Collapse
Affiliation(s)
- Gianluca Campanella
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Silvia Polidoro
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Cornelia Di Gaetano
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Giovanni Fiorito
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Simonetta Guarrera
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Vittorio Krogh
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Domenico Palli
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Salvatore Panico
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Carlotta Sacerdote
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Rosario Tumino
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Giuseppe Matullo
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Marc Chadeau-Hyam
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Paolo Vineis
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK Department of Epidemiology and Biostatistics, Imperial College London, London, UK,Human Genetics Foundation (HuGeF), Turin, Italy, Department of Medical Sciences, University of Turin, Turin, Italy, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy, Istituto per lo Studio e la Prevenzione Oncologica (ISPO Toscana), Florence, Italy, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy, Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), Turin, Italy Cancer Registry and Histopathology Unit, Azienda Ospedaliera 'Civile - M. P. Arezzo', Ragusa, Italy and MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| |
Collapse
|
1039
|
Pierron F, Bureau du Colombier S, Moffett A, Caron A, Peluhet L, Daffe G, Lambert P, Elie P, Labadie P, Budzinski H, Dufour S, Couture P, Baudrimont M. Abnormal ovarian DNA methylation programming during gonad maturation in wild contaminated fish. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2014; 48:11688-11695. [PMID: 25203663 DOI: 10.1021/es503712c] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
There is increasing evidence that pollutants may cause diseases via epigenetic modifications. Epigenetic mechanisms such as DNA methylation participate in the regulation of gene transcription. Surprisingly, epigenetics research is still limited in ecotoxicology. In this study, we investigated whether chronic exposure to contaminants experienced by wild female fish (Anguilla anguilla) throughout their juvenile phase can affect the DNA methylation status of their oocytes during gonad maturation. Thus, fish were sampled in two locations presenting a low or a high contamination level. Then, fish were transferred to the laboratory and artificially matured. Before hormonal treatment, the DNA methylation levels of the genes encoding for the aromatase and the receptor of the follicle stimulating hormone were higher in contaminated fish than in fish from the clean site. For the hormone receptor, this hypermethylation was positively correlated with the contamination level of fish and was associated with a decrease in its transcription level. In addition, whereas gonad growth was associated with an increase in DNA methylation in fish from the clean site, no changes were observed in contaminated fish in response to hormonal treatment. Finally, a higher gonad growth was observed in fish from the reference site in comparison to contaminated fish.
Collapse
Affiliation(s)
- Fabien Pierron
- University of Bordeaux, EPOC, UMR 5805 , F-33400 Talence, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1040
|
Osella M, Riba A, Testori A, Corà D, Caselle M. Interplay of microRNA and epigenetic regulation in the human regulatory network. Front Genet 2014; 5:345. [PMID: 25339974 PMCID: PMC4186481 DOI: 10.3389/fgene.2014.00345] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 09/16/2014] [Indexed: 11/13/2022] Open
Abstract
The expression of protein-coding genes is controlled by a complex network of regulatory interactions. It is becoming increasingly appreciated that post-transcriptional repression by microRNAs, a class of small non-coding RNAs, is a key layer of regulation in several biological processes. In this contribution, we discuss the interplay between microRNAs and epigenetic regulators. Among the mixed genetic circuits composed by these two different kinds of regulation, it seems that a central role is played by double-negative feedback loops in which a microRNA inhibits an epigenetic regulator and in turn is controlled at the epigenetic level by the same regulator. We discuss a few relevant properties of this class of network motifs and their potential role in cell differentiation. In particular, using mathematical modeling we show how this particular circuit can exhibit a switch-like behavior between two alternative steady states, while being robust to stochastic transitions between these two states, a feature presumably required for circuits involved in cell fate decision. Finally, we present a list of putative double-negative feedback loops from a literature survey combined with bioinformatic analysis, and discuss in detail a few examples.
Collapse
Affiliation(s)
- Matteo Osella
- Dipartimento di Fisica, Istituto Nazionale di Fisica Nucleare, Università di Torino Torino, Italy
| | - Andrea Riba
- Dipartimento di Fisica, Istituto Nazionale di Fisica Nucleare, Università di Torino Torino, Italy
| | - Alessandro Testori
- Dipartimento di Fisica, Istituto Nazionale di Fisica Nucleare, Università di Torino Torino, Italy
| | - Davide Corà
- Dipartimento di Oncologia, Istituto per la Ricerca sul Cancro di Candiolo-Istituto di Ricovero e Cura a Carattere Scientifico, Università di Torino Torino, Italy
| | - Michele Caselle
- Dipartimento di Fisica, Istituto Nazionale di Fisica Nucleare, Università di Torino Torino, Italy
| |
Collapse
|
1041
|
Wilde JJ, Petersen JR, Niswander L. Genetic, epigenetic, and environmental contributions to neural tube closure. Annu Rev Genet 2014; 48:583-611. [PMID: 25292356 DOI: 10.1146/annurev-genet-120213-092208] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The formation of the embryonic brain and spinal cord begins as the neural plate bends to form the neural folds, which meet and adhere to close the neural tube. The neural ectoderm and surrounding tissues also coordinate proliferation, differentiation, and patterning. This highly orchestrated process is susceptible to disruption, leading to neural tube defects (NTDs), a common birth defect. Here, we highlight genetic and epigenetic contributions to neural tube closure. We describe an online database we created as a resource for researchers, geneticists, and clinicians. Neural tube closure is sensitive to environmental influences, and we discuss disruptive causes, preventative measures, and possible mechanisms. New technologies will move beyond candidate genes in small cohort studies toward unbiased discoveries in sporadic NTD cases. This will uncover the genetic complexity of NTDs and critical gene-gene interactions. Animal models can reveal the causative nature of genetic variants, the genetic interrelationships, and the mechanisms underlying environmental influences.
Collapse
Affiliation(s)
- Jonathan J Wilde
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, Colorado 80045;
| | | | | |
Collapse
|
1042
|
Gu L, Liu H, Gu X, Boots C, Moley KH, Wang Q. Metabolic control of oocyte development: linking maternal nutrition and reproductive outcomes. Cell Mol Life Sci 2014; 72:251-71. [PMID: 25280482 DOI: 10.1007/s00018-014-1739-4] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Revised: 09/12/2014] [Accepted: 09/22/2014] [Indexed: 02/01/2023]
Abstract
Obesity, diabetes, and related metabolic disorders are major health issues worldwide. As the epidemic of metabolic disorders continues, the associated medical co-morbidities, including the detrimental impact on reproduction, increase as well. Emerging evidence suggests that the effects of maternal nutrition on reproductive outcomes are likely to be mediated, at least in part, by oocyte metabolism. Well-balanced and timed energy metabolism is critical for optimal development of oocytes. To date, much of our understanding of oocyte metabolism comes from the effects of extrinsic nutrients on oocyte maturation. In contrast, intrinsic regulation of oocyte development by metabolic enzymes, intracellular mediators, and transport systems is less characterized. Specifically, decreased acid transport proteins levels, increased glucose/lipid content and elevated reactive oxygen species in oocytes have been implicated in meiotic defects, organelle dysfunction and epigenetic alteration. Therefore, metabolic disturbances in oocytes may contribute to the diminished reproductive potential experienced by women with metabolic disorders. In-depth research is needed to further explore the underlying mechanisms. This review also discusses several approaches for metabolic analysis. Metabolomic profiling of oocytes, the surrounding granulosa cells, and follicular fluid will uncover the metabolic networks regulating oocyte development, potentially leading to the identification of oocyte quality markers and prevention of reproductive disease and poor outcomes in offspring.
Collapse
Affiliation(s)
- Ling Gu
- College of Animal Science and Technology, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, Jiangsu, China,
| | | | | | | | | | | |
Collapse
|
1043
|
Brucato N, DeLisi LE, Fisher SE, Francks C. Hypomethylation of the paternally inherited LRRTM1 promoter linked to schizophrenia. Am J Med Genet B Neuropsychiatr Genet 2014; 165B:555-63. [PMID: 25111784 DOI: 10.1002/ajmg.b.32258] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 06/26/2014] [Indexed: 12/25/2022]
Abstract
Epigenetic effects on psychiatric traits remain relatively under-studied, and it remains unclear what the sizes of individual epigenetic effects may be, or how they vary between different clinical populations. The gene LRRTM1 (chromosome 2p12) has previously been linked and associated with schizophrenia in a parent-of-origin manner in a set of affected siblings (LOD = 4.72), indirectly suggesting a disruption of paternal imprinting at this locus in these families. From the same set of siblings that originally showed strong linkage at this locus, we analyzed 99 individuals using 454-bisulfite sequencing, from whole blood DNA, to measure the level of DNA methylation in the promoter region of LRRTM1. We also assessed seven additional loci that would be informative to compare. Paternal identity-by-descent sharing at LRRTM1, within sibling pairs, was linked to their similarity of methylation at the gene's promoter. Reduced methylation at the promoter showed a significant association with schizophrenia. Sibling pairs concordant for schizophrenia showed more similar methylation levels at the LRRTM1 promoter than diagnostically discordant pairs. The alleles of common SNPs spanning the locus did not explain this epigenetic linkage, which can therefore be considered as largely independent of DNA sequence variation and would not be detected in standard genetic association analysis. Our data suggest that hypomethylation at the LRRTM1 promoter, particularly of the paternally inherited allele, was a risk factor for the development of schizophrenia in this set of siblings affected with familial schizophrenia, and that had previously showed linkage at this locus in an affected-sib-pair context.
Collapse
Affiliation(s)
- Nicolas Brucato
- Language & Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
| | | | | | | |
Collapse
|
1044
|
|
1045
|
DNA methylation biomarkers: cancer and beyond. Genes (Basel) 2014; 5:821-64. [PMID: 25229548 PMCID: PMC4198933 DOI: 10.3390/genes5030821] [Citation(s) in RCA: 193] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Revised: 08/17/2014] [Accepted: 09/01/2014] [Indexed: 12/23/2022] Open
Abstract
Biomarkers are naturally-occurring characteristics by which a particular pathological process or disease can be identified or monitored. They can reflect past environmental exposures, predict disease onset or course, or determine a patient's response to therapy. Epigenetic changes are such characteristics, with most epigenetic biomarkers discovered to date based on the epigenetic mark of DNA methylation. Many tissue types are suitable for the discovery of DNA methylation biomarkers including cell-based samples such as blood and tumor material and cell-free DNA samples such as plasma. DNA methylation biomarkers with diagnostic, prognostic and predictive power are already in clinical trials or in a clinical setting for cancer. Outside cancer, strong evidence that complex disease originates in early life is opening up exciting new avenues for the detection of DNA methylation biomarkers for adverse early life environment and for estimation of future disease risk. However, there are a number of limitations to overcome before such biomarkers reach the clinic. Nevertheless, DNA methylation biomarkers have great potential to contribute to personalized medicine throughout life. We review the current state of play for DNA methylation biomarkers, discuss the barriers that must be crossed on the way to implementation in a clinical setting, and predict their future use for human disease.
Collapse
|
1046
|
El Hajj N, Schneider E, Lehnen H, Haaf T. Epigenetics and life-long consequences of an adverse nutritional and diabetic intrauterine environment. Reproduction 2014; 148:R111-20. [PMID: 25187623 PMCID: PMC4241689 DOI: 10.1530/rep-14-0334] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The phenomenon that adverse environmental exposures in early life are associated with increased susceptibilities for many adult, particularly metabolic diseases, is now referred to as ‘developmental origins of health and disease (DOHAD)’ or ‘Barker’ hypothesis. Fetal overnutrition and undernutrition have similar long-lasting effects on the setting of the neuroendocrine control systems, energy homeostasis, and metabolism, leading to life-long increased morbidity. There are sensitive time windows during early development, where environmental cues can program persistent epigenetic modifications which are generally assumed to mediate these gene–environment interactions. Most of our current knowledge on fetal programing comes from animal models and epidemiological studies in humans, in particular the Dutch famine birth cohort. In industrialized countries, there is more concern about adverse long-term consequences of fetal overnutrition, i.e. by exposure to gestational diabetes mellitus and/or maternal obesity which affect 10–20% of pregnancies. Epigenetic changes due to maternal diabetes/obesity may predispose the offspring to develop metabolic disease later in life and, thus, transmit the adverse environmental exposure to the next generation. This vicious cycle could contribute significantly to the worldwide metabolic disease epidemics. In this review article, we focus on the epigenetics of an adverse intrauterine environment, in particular gestational diabetes, and its implications for the prevention of complex disease.
Collapse
Affiliation(s)
- Nady El Hajj
- Institute of Human GeneticsJulius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, GermanyDepartment of Gynecology and ObstetricsStädtische Kliniken, 41239 Mönchengladbach, Germany
| | - Eberhard Schneider
- Institute of Human GeneticsJulius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, GermanyDepartment of Gynecology and ObstetricsStädtische Kliniken, 41239 Mönchengladbach, Germany
| | - Harald Lehnen
- Institute of Human GeneticsJulius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, GermanyDepartment of Gynecology and ObstetricsStädtische Kliniken, 41239 Mönchengladbach, Germany
| | - Thomas Haaf
- Institute of Human GeneticsJulius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, GermanyDepartment of Gynecology and ObstetricsStädtische Kliniken, 41239 Mönchengladbach, Germany
| |
Collapse
|
1047
|
Affiliation(s)
- Swapan Kumar Das
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| |
Collapse
|
1048
|
Abstract
Most of what is known about the pathogenesis of inflammatory bowel disease (IBD) pertains to complex interplay between host genetics, immunity, and environmental factors. Epigenetic modifications play pivotal roles in intestinal immunity and mucosal homeostasis as well as mediating gene-environment interactions. In this article, we provide a historical account of epigenetic research either directly related or pertinent to the pathogenesis and management of IBD. We further collate emerging evidence supporting roles for epigenetic mechanisms in relevant aspects of IBD biology, including deregulated immunity, host-pathogen recognition and mucosal integrity. Finally, we highlight key epigenetic mechanisms that link chronic inflammation to specific IBD comorbidities, including colitis-associated cancer and discuss their potential utility as novel biomarkers or pharmacologic targets in IBD therapy.
Collapse
|
1049
|
Resendiz M, Mason S, Lo CL, Zhou FC. Epigenetic regulation of the neural transcriptome and alcohol interference during development. Front Genet 2014; 5:285. [PMID: 25206361 PMCID: PMC4144008 DOI: 10.3389/fgene.2014.00285] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 08/02/2014] [Indexed: 01/07/2023] Open
Abstract
Alcohol intoxicated cells broadly alter their metabolites – among them methyl and acetic acid can alter the DNA and histone epigenetic codes. Together with the promiscuous effect of alcohol on enzyme activities (including DNA methyltransferases) and the downstream effect on microRNA and transposable elements, alcohol is well placed to affect intrinsic transcriptional programs of developing cells. Considering that the developmental consequences of early alcohol exposure so profoundly affect neural systems, it is not unfounded to reason that alcohol exploits transcriptional regulators to challenge canonical gene expression and in effect, intrinsic developmental pathways to achieve widespread damage in the developing nervous system. To fully evaluate the role of epigenetic regulation in alcohol-related developmental disease, it is important to first gather the targets of epigenetic players in neurodevelopmental models. Here, we attempt to review the cellular and genomic windows of opportunity for alcohol to act on intrinsic neurodevelopmental programs. We also discuss some established targets of fetal alcohol exposure and propose pathways for future study. Overall, this review hopes to illustrate the known epigenetic program and its alterations in normal neural stem cell development and further, aims to depict how alcohol, through neuroepigenetics, may lead to neurodevelopmental deficits observed in fetal alcohol spectrum disorders.
Collapse
Affiliation(s)
- Marisol Resendiz
- Stark Neuroscience Research Institute Indianapolis, IN, USA ; Indiana Alcohol Research Center, Indiana University School of Medicine Indianapolis, IN, USA
| | - Stephen Mason
- Department of Anatomy and Cell Biology, Indiana University School of Medicine Indianapolis, IN, USA
| | - Chiao-Ling Lo
- Indiana Alcohol Research Center, Indiana University School of Medicine Indianapolis, IN, USA ; Department of Anatomy and Cell Biology, Indiana University School of Medicine Indianapolis, IN, USA
| | - Feng C Zhou
- Stark Neuroscience Research Institute Indianapolis, IN, USA ; Indiana Alcohol Research Center, Indiana University School of Medicine Indianapolis, IN, USA ; Department of Anatomy and Cell Biology, Indiana University School of Medicine Indianapolis, IN, USA
| |
Collapse
|
1050
|
Genetic Dissection of the Physiological Role of Skeletal Muscle in Metabolic Syndrome. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/635146] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The primary deficiency underlying metabolic syndrome is insulin resistance, in which insulin-responsive peripheral tissues fail to maintain glucose homeostasis. Because skeletal muscle is the major site for insulin-induced glucose uptake, impairments in skeletal muscle’s insulin responsiveness play a major role in the development of insulin resistance and type 2 diabetes. For example, skeletal muscle of type 2 diabetes patients and their offspring exhibit reduced ratios of slow oxidative muscle. These observations suggest the possibility of applying muscle remodeling to recover insulin sensitivity in metabolic syndrome. Skeletal muscle is highly adaptive to external stimulations such as exercise; however, in practice it is often not practical or possible to enforce the necessary intensity to obtain measurable benefits to the metabolic syndrome patient population. Therefore, identifying molecular targets for inducing muscle remodeling would provide new approaches to treat metabolic syndrome. In this review, the physiological properties of skeletal muscle, genetic analysis of metabolic syndrome in human populations and model organisms, and genetically engineered mouse models will be discussed in regard to the prospect of applying skeletal muscle remodeling as possible therapy for metabolic syndrome.
Collapse
|