1
|
Abudahab S, Kronfol MM, Dozmorov MG, Campbell T, Jahr FM, Nguyen J, AlAzzeh O, Al Saeedy DY, Victor A, Lee S, Malay S, Lapato DM, Halquist MS, McRae M, Deshpande LS, Slattum PW, Price ET, McClay JL. Genome-wide analysis of hepatic DNA methylation reveals impact of epigenetic aging on xenobiotic metabolism and transport genes in an aged mouse model. GeroScience 2024; 46:5967-5980. [PMID: 38558216 PMCID: PMC11493898 DOI: 10.1007/s11357-024-01137-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
Hepatic xenobiotic metabolism and transport decline with age, while intact xenobiotic metabolism is associated with longevity. However, few studies have examined the genome-wide impact of epigenetic aging on these processes. We used reduced representation bisulfite sequencing (RRBS) to map DNA methylation changes in liver DNA from mice ages 4 and 24 months. We identified several thousand age-associated differentially methylated sites (a-DMS), many of which overlapped genes encoding Phase I and Phase II drug metabolizing enzymes, in addition to ABC and SLC classes of transporters. Notable genes harboring a-DMS were Cyp1a2, Cyp2d9, and Abcc2 that encode orthologs of the human drug metabolizing enzymes CYP1A2 and CYP2D6, and the multidrug resistance protein 2 (MRP2) transporter. Cyp2d9 hypermethylation with age was significantly associated with reduced gene expression, while Abcc2 expression was unchanged with age. Cyp1a2 lost methylation with age while, counterintuitively, its expression also reduced with age. We hypothesized that age-related dysregulation of the hepatic transcriptional machinery caused down-regulation of genes despite age-related hypomethylation. Bioinformatic analysis of hypomethylated a-DMS in our sample found them to be highly enriched for hepatic nuclear factor 4 alpha (HNF4α) binding sites. HNF4α promotes Cyp1a2 expression and is downregulated with age, which could explain the reduction in Cyp1a2 expression. Overall, our study supports the broad impact of epigenetic aging on xenobiotic metabolism and transport. Future work should evaluate the interplay between hepatic nuclear receptor function and epigenetic aging. These results may have implications for studies of longevity and healthy aging.
Collapse
Affiliation(s)
- Sara Abudahab
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Dr. Joseph L. McClay, 6Th floor Smith Building, 410 North 12Th Street, Medical College of Virginia Campus, Richmond, VA, 23298-0533, USA
| | - Mohamad M Kronfol
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Dr. Joseph L. McClay, 6Th floor Smith Building, 410 North 12Th Street, Medical College of Virginia Campus, Richmond, VA, 23298-0533, USA
| | - Mikhail G Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, USA
| | - Thomas Campbell
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, VA, USA
| | - Fay M Jahr
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Dr. Joseph L. McClay, 6Th floor Smith Building, 410 North 12Th Street, Medical College of Virginia Campus, Richmond, VA, 23298-0533, USA
| | - Jasmine Nguyen
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Dr. Joseph L. McClay, 6Th floor Smith Building, 410 North 12Th Street, Medical College of Virginia Campus, Richmond, VA, 23298-0533, USA
| | - Ola AlAzzeh
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Dr. Joseph L. McClay, 6Th floor Smith Building, 410 North 12Th Street, Medical College of Virginia Campus, Richmond, VA, 23298-0533, USA
| | - Dalia Y Al Saeedy
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Dr. Joseph L. McClay, 6Th floor Smith Building, 410 North 12Th Street, Medical College of Virginia Campus, Richmond, VA, 23298-0533, USA
| | - Ashley Victor
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Dr. Joseph L. McClay, 6Th floor Smith Building, 410 North 12Th Street, Medical College of Virginia Campus, Richmond, VA, 23298-0533, USA
| | - Sera Lee
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Dr. Joseph L. McClay, 6Th floor Smith Building, 410 North 12Th Street, Medical College of Virginia Campus, Richmond, VA, 23298-0533, USA
| | - Shravani Malay
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Dr. Joseph L. McClay, 6Th floor Smith Building, 410 North 12Th Street, Medical College of Virginia Campus, Richmond, VA, 23298-0533, USA
| | - Dana M Lapato
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Matthew S Halquist
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, VA, USA
| | - MaryPeace McRae
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Dr. Joseph L. McClay, 6Th floor Smith Building, 410 North 12Th Street, Medical College of Virginia Campus, Richmond, VA, 23298-0533, USA
| | - Laxmikant S Deshpande
- Department of Neurology, Virginia Commonwealth University, Richmond, VA, USA
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Patricia W Slattum
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Dr. Joseph L. McClay, 6Th floor Smith Building, 410 North 12Th Street, Medical College of Virginia Campus, Richmond, VA, 23298-0533, USA
- Virginia Center On Aging, Virginia Commonwealth University, Richmond, VA, USA
| | - Elvin T Price
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Dr. Joseph L. McClay, 6Th floor Smith Building, 410 North 12Th Street, Medical College of Virginia Campus, Richmond, VA, 23298-0533, USA
| | - Joseph L McClay
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Dr. Joseph L. McClay, 6Th floor Smith Building, 410 North 12Th Street, Medical College of Virginia Campus, Richmond, VA, 23298-0533, USA.
| |
Collapse
|
2
|
Wang K, Sartor MA, Colacino JA, Dolinoy DC, Svoboda LK. Sex-Specific Deflection of Age-Related DNA Methylation and Gene Expression in Mouse Heart by Perinatal Toxicant Exposures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.25.591125. [PMID: 38712146 PMCID: PMC11071472 DOI: 10.1101/2024.04.25.591125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Background Global and site-specific changes in DNA methylation and gene expression are associated with cardiovascular aging and disease, but how toxicant exposures during early development influence the normal trajectory of these age-related molecular changes, and whether there are sex differences, has not yet been investigated. Objectives We used an established mouse model of developmental exposures to investigate the effects of perinatal exposure to either lead (Pb) or diethylhexyl phthalate (DEHP), two ubiquitous environmental contaminants strongly associated with CVD, on age-related cardiac DNA methylation and gene expression. Methods Dams were randomly assigned to receive human physiologically relevant levels of Pb (32 ppm in water), DEHP (25 mg/kg chow), or control water and chow. Exposures started two weeks prior to mating and continued until weaning at postnatal day 21 (3 weeks of age). Approximately one male and one female offspring per litter were followed to 3 weeks, 5 months, or 10 months of age, at which time whole hearts were collected (n ≥ 5 per sex per exposure). Enhanced reduced representation bisulfite sequencing (ERRBS) was used to assess the cardiac DNA methylome at 3 weeks and 10 months, and RNA-seq was conducted at all 3 time points. MethylSig and edgeR were used to identify age-related differentially methylated regions (DMRs) and differentially expressed genes (DEGs), respectively, within each sex and exposure group. Cell type deconvolution of bulk RNA-seq data was conducted using the MuSiC algorithm and publicly available single cell RNA-seq data. Results Thousands of DMRs and hundreds of DEGs were identified in control, DEHP, and Pb-exposed hearts across time between 3 weeks and 10 months of age. A closer look at the genes and pathways showing differential DNA methylation revealed that the majority were unique to each sex and exposure group. Overall, pathways governing development and differentiation were most frequently altered with age in all conditions. A small number of genes in each group showed significant changes in DNA methylation and gene expression with age, including several that were altered by both toxicants but were unchanged in control. We also observed subtle, but significant changes in the proportion of several cell types due to age, sex, and developmental exposure. Discussion Together these data show that perinatal Pb or DEHP exposures deflect normal age-related gene expression, DNA methylation programs, and cellular composition across the life course, long after cessation of exposure, and highlight potential biomarkers of developmental toxicant exposures. Further studies are needed to investigate how these epigenetic and transcriptional changes impact cardiovascular health across the life course.
Collapse
|
3
|
Li Z, Wang W, Li W, Duan H, Xu C, Tian X, Ning F, Zhang D. Co-methylation analyses identify CpGs associated with lipid traits in Chinese discordant monozygotic twins. Hum Mol Genet 2024; 33:583-593. [PMID: 38142287 DOI: 10.1093/hmg/ddad207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/29/2023] [Accepted: 12/05/2023] [Indexed: 12/25/2023] Open
Abstract
To control genetic background and early life milieu in genome-wide DNA methylation analysis for blood lipids, we recruited Chinese discordant monozygotic twins to explore the relationships between DNA methylations and total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), and triglycerides (TG). 132 monozygotic (MZ) twins were included with discordant lipid levels and completed data. A linear mixed model was conducted in Epigenome-wide association study (EWAS). Generalized estimating equation model was for gene expression analysis. We conducted Weighted correlation network analysis (WGCNA) to build co-methylated interconnected network. Additional Qingdao citizens were recruited for validation. Inference about Causation through Examination of Familial Confounding (ICE FALCON) was used to infer the possible direction of these relationships. A total of 476 top CpGs reached suggestively significant level (P < 10-4), of which, 192 CpGs were significantly associated with TG (FDR < 0.05). They were used to build interconnected network and highlight crucial genes from WGCNA. Finally, four CpGs in GATA4 were validated as risk factors for TC; six CpGs at ITFG2-AS1 were negatively associated with TG; two CpGs in PLXND1 played protective roles in HDL-C. ICE FALCON indicated abnormal TC was regarded as the consequence of DNA methylation in CpGs at GATA4, rather than vice versa. Four CpGs in ITFG2-AS1 were both causes and consequences of modified TG levels. Our results indicated that DNA methylation levels of 12 CpGs in GATA4, ITFG2-AS1, and PLXND1 were relevant to TC, TG, and HDL-C, respectively, which might provide new epigenetic insights into potential clinical treatment of dyslipidemia.
Collapse
Affiliation(s)
- Zhaoying Li
- Department of Epidemiology and Health Statistics, The College of Public Health of Qingdao University, No. 308 Ning Xia Street, Qingdao 266071, Shandong Province, People's Republic of China
| | - Weijing Wang
- Department of Epidemiology and Health Statistics, The College of Public Health of Qingdao University, No. 308 Ning Xia Street, Qingdao 266071, Shandong Province, People's Republic of China
| | - Weilong Li
- Epidemiology and Biostatistics, Department of Public Health, University of Southern Denmark, J.B. Winsløws Vej 9 B, st. tv. Odense C DK-5000, Denmark
| | - Haiping Duan
- Qingdao Municipal Center for Disease Control and Prevention, No. 175 Shandong Road, Qingdao 266000, Shandong Province, People's Republic of China
- Qingdao Institute of Preventive Medicine, No. 175 Shandong Road, Qingdao 266000, Shandong Province, People's Republic of China
| | - Chunsheng Xu
- Qingdao Municipal Center for Disease Control and Prevention, No. 175 Shandong Road, Qingdao 266000, Shandong Province, People's Republic of China
- Qingdao Institute of Preventive Medicine, No. 175 Shandong Road, Qingdao 266000, Shandong Province, People's Republic of China
| | - Xiaocao Tian
- Qingdao Municipal Center for Disease Control and Prevention, No. 175 Shandong Road, Qingdao 266000, Shandong Province, People's Republic of China
- Qingdao Institute of Preventive Medicine, No. 175 Shandong Road, Qingdao 266000, Shandong Province, People's Republic of China
| | - Feng Ning
- Qingdao Municipal Center for Disease Control and Prevention, No. 175 Shandong Road, Qingdao 266000, Shandong Province, People's Republic of China
- Qingdao Institute of Preventive Medicine, No. 175 Shandong Road, Qingdao 266000, Shandong Province, People's Republic of China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, The College of Public Health of Qingdao University, No. 308 Ning Xia Street, Qingdao 266071, Shandong Province, People's Republic of China
| |
Collapse
|
4
|
Abudahab S, Slattum PW, Price ET, McClay JL. Epigenetic regulation of drug metabolism in aging: utilizing epigenetics to optimize geriatric pharmacotherapy. Pharmacogenomics 2024; 25:41-54. [PMID: 38126340 PMCID: PMC10794944 DOI: 10.2217/pgs-2023-0199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
We explore the relationship between epigenetic aging and drug metabolism. We review current evidence for changes in drug metabolism in normal aging, followed by a description of how epigenetic modifications associated with age can regulate the expression and functionality of genes. In particular, we focus on the role of epigenome-wide studies of human and mouse liver in understanding these age-related processes with respect to xenobiotic processing. We highlight genes encoding drug metabolizing enzymes and transporters revealed to be affected by epigenetic aging in these studies. We conclude that substantial evidence exists for epigenetic aging impacting drug metabolism and transport genes, but more work is needed. We further highlight the promise of pharmacoepigenetics applied to enhancing drug safety in older adults.
Collapse
Affiliation(s)
- Sara Abudahab
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Patricia W Slattum
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
- Virginia Center on Aging, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Elvin T Price
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Joseph L McClay
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
5
|
Varshavsky M, Harari G, Glaser B, Dor Y, Shemer R, Kaplan T. Accurate age prediction from blood using a small set of DNA methylation sites and a cohort-based machine learning algorithm. CELL REPORTS METHODS 2023; 3:100567. [PMID: 37751697 PMCID: PMC10545910 DOI: 10.1016/j.crmeth.2023.100567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/18/2023] [Accepted: 08/03/2023] [Indexed: 09/28/2023]
Abstract
Chronological age prediction from DNA methylation sheds light on human aging, health, and lifespan. Current clocks are mostly based on linear models and rely upon hundreds of sites across the genome. Here, we present GP-age, an epigenetic non-linear cohort-based clock for blood, based upon 11,910 methylomes. Using 30 CpG sites alone, GP-age outperforms state-of-the-art models, with a median accuracy of ∼2 years on held-out blood samples, for both array and sequencing-based data. We show that aging-related changes occur at multiple neighboring CpGs, with implications for using fragment-level analysis of sequencing data in aging research. By training three independent clocks, we show enrichment of donors with consistent deviation between predicted and actual age, suggesting individual rates of biological aging. Overall, we provide a compact yet accurate alternative to array-based clocks for blood, with applications in longitudinal aging research, forensic profiling, and monitoring epigenetic processes in transplantation medicine and cancer.
Collapse
Affiliation(s)
- Miri Varshavsky
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel; The Center for Computational Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gil Harari
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel; Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Benjamin Glaser
- Department of Endocrinology and Metabolism, Hadassah Medical Center and Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yuval Dor
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel; The Center for Computational Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ruth Shemer
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Tommy Kaplan
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel; Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel; The Center for Computational Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
6
|
López-Gil L, Pascual-Ahuir A, Proft M. Genomic Instability and Epigenetic Changes during Aging. Int J Mol Sci 2023; 24:14279. [PMID: 37762580 PMCID: PMC10531692 DOI: 10.3390/ijms241814279] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Aging is considered the deterioration of physiological functions along with an increased mortality rate. This scientific review focuses on the central importance of genomic instability during the aging process, encompassing a range of cellular and molecular changes that occur with advancing age. In particular, this revision addresses the genetic and epigenetic alterations that contribute to genomic instability, such as telomere shortening, DNA damage accumulation, and decreased DNA repair capacity. Furthermore, the review explores the epigenetic changes that occur with aging, including modifications to histones, DNA methylation patterns, and the role of non-coding RNAs. Finally, the review discusses the organization of chromatin and its contribution to genomic instability, including heterochromatin loss, chromatin remodeling, and changes in nucleosome and histone abundance. In conclusion, this review highlights the fundamental role that genomic instability plays in the aging process and underscores the need for continued research into these complex biological mechanisms.
Collapse
Affiliation(s)
- Lucía López-Gil
- Department of Biotechnology, Instituto de Biología Molecular y Celular de Plantas, Universitat Politècnica de València, Ingeniero Fausto Elio s/n, 46022 Valencia, Spain;
- Department of Molecular and Cellular Pathology and Therapy, Instituto de Biomedicina de Valencia IBV-CSIC, Consejo Superior de Investigaciones Científicas CSIC, Jaime Roig 11, 46010 Valencia, Spain
| | - Amparo Pascual-Ahuir
- Department of Biotechnology, Instituto de Biología Molecular y Celular de Plantas, Universitat Politècnica de València, Ingeniero Fausto Elio s/n, 46022 Valencia, Spain;
| | - Markus Proft
- Department of Molecular and Cellular Pathology and Therapy, Instituto de Biomedicina de Valencia IBV-CSIC, Consejo Superior de Investigaciones Científicas CSIC, Jaime Roig 11, 46010 Valencia, Spain
| |
Collapse
|
7
|
Maji RK, Czepukojc B, Scherer M, Tierling S, Cadenas C, Gianmoena K, Gasparoni N, Nordström K, Gasparoni G, Laggai S, Yang X, Sinha A, Ebert P, Falk-Paulsen M, Kinkley S, Hoppstädter J, Chung HR, Rosenstiel P, Hengstler JG, Walter J, Schulz MH, Kessler SM, Kiemer AK. Alterations in the hepatocyte epigenetic landscape in steatosis. Epigenetics Chromatin 2023; 16:30. [PMID: 37415213 DOI: 10.1186/s13072-023-00504-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 06/21/2023] [Indexed: 07/08/2023] Open
Abstract
Fatty liver disease or the accumulation of fat in the liver, has been reported to affect the global population. This comes with an increased risk for the development of fibrosis, cirrhosis, and hepatocellular carcinoma. Yet, little is known about the effects of a diet containing high fat and alcohol towards epigenetic aging, with respect to changes in transcriptional and epigenomic profiles. In this study, we took up a multi-omics approach and integrated gene expression, methylation signals, and chromatin signals to study the epigenomic effects of a high-fat and alcohol-containing diet on mouse hepatocytes. We identified four relevant gene network clusters that were associated with relevant pathways that promote steatosis. Using a machine learning approach, we predict specific transcription factors that might be responsible to modulate the functionally relevant clusters. Finally, we discover four additional CpG loci and validate aging-related differential CpG methylation. Differential CpG methylation linked to aging showed minimal overlap with altered methylation in steatosis.
Collapse
Affiliation(s)
- Ranjan Kumar Maji
- Institute for Cardiovascular Regeneration, Goethe-University, 60590, Frankfurt, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 60590, Frankfurt, Germany
| | - Beate Czepukojc
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123, Saarbrücken, Germany
| | - Michael Scherer
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), 08003, Barcelona, Spain
| | - Sascha Tierling
- Department of Genetics, Saarland University, 66123, Saarbrücken, Germany
| | - Cristina Cadenas
- IfADo: Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Kathrin Gianmoena
- IfADo: Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Nina Gasparoni
- Department of Genetics, Saarland University, 66123, Saarbrücken, Germany
| | - Karl Nordström
- Department of Genetics, Saarland University, 66123, Saarbrücken, Germany
| | - Gilles Gasparoni
- Department of Genetics, Saarland University, 66123, Saarbrücken, Germany
| | - Stephan Laggai
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123, Saarbrücken, Germany
| | - Xinyi Yang
- Institute of Medical Bioinformatics and Biostatistics, Philipps University of Marburg, 35032, Marburg, Germany
| | - Anupam Sinha
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, 24105, Kiel, Germany
| | - Peter Ebert
- Core Unit Bioinformatics, Medical Faculty, Heinrich Heine University, 40225, Düsseldorf, Germany
- Department of Computational Biology and Applied Algorithmics, Max Planck Institute for Informatics, 66123, Saarbrücken, Germany
| | - Maren Falk-Paulsen
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, 24105, Kiel, Germany
| | - Sarah Kinkley
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, 14195, Berlin, Germany
| | - Jessica Hoppstädter
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123, Saarbrücken, Germany
| | - Ho-Ryun Chung
- Institute of Medical Bioinformatics and Biostatistics, Philipps University of Marburg, 35032, Marburg, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, 24105, Kiel, Germany
| | - Jan G Hengstler
- IfADo: Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Jörn Walter
- Department of Genetics, Saarland University, 66123, Saarbrücken, Germany
| | - Marcel H Schulz
- Institute for Cardiovascular Regeneration, Goethe-University, 60590, Frankfurt, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 60590, Frankfurt, Germany.
- Department of Computational Biology and Applied Algorithmics, Max Planck Institute for Informatics, 66123, Saarbrücken, Germany.
- Excellence Cluster on Multimodal Computing and Interaction, Saarland University, 66123, Saarbrücken, Germany.
| | - Sonja M Kessler
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123, Saarbrücken, Germany.
- Institute of Pharmacy, Experimental Pharmacology for Natural Sciences, Martin Luther University Halle-Wittenberg, Halle, Germany.
- Halle Research Centre for Drug Therapy (HRCDT), Halle, Germany.
| | - Alexandra K Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123, Saarbrücken, Germany.
| |
Collapse
|
8
|
Bazi Bushen mitigates epigenetic aging and extends healthspan in naturally aging mice. Biomed Pharmacother 2023; 160:114384. [PMID: 36764132 DOI: 10.1016/j.biopha.2023.114384] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/18/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Bazi Bushen (BZBS), a traditional Chinese medicine, has been proven effective in the treatment of age-related disease in mouse models. However, whether its therapeutic effects are due to antiaging mechanism has not yet been explored. In the present study, we investigated the antiaging effects of BZBS in naturally aging mice by using behavioral tests, liver DNA methylome sequencing, methylation age estimation, and frailty index assessment. The methylome analysis revealed a decrease of mCpG levels in the aged mouse liver. BZBS treatment tended to restore age-associated methylation decline and prune the methylation pattern toward that of young mice. More importantly, BZBS significantly rejuvenated methylation age of the aged mice, which was computed by an upgraded DNA methylation clock. These results were consistent with enhanced memory and muscular endurance, as well as decreased frailty score and liver pathological changes. KEGG analysis together with aging-related database screening identified methylation-targeted pathways upon BZBS treatment, including oxidative stress, DNA repair, MAPK signaling, and inflammation. Upregulation of key effectors and their downstream effects on elevating Sod2 expression and diminishing DNA damage were further investigated. Finally, in vitro experiments with senescent HUVECs proved a direct effect of BZBS extracts on the regulation of methylation enzymes during cellular aging. In summary, our work has revealed for the first time the antiaging effects of BZBS by slowing the methylation aging. These results suggest that BZBS might have great potential to extend healthspan and also explored the mechanism of BZBS action in the treatment of age-related diseases.
Collapse
|
9
|
Szukiewicz D, Trojanowski S, Kociszewska A, Szewczyk G. Modulation of the Inflammatory Response in Polycystic Ovary Syndrome (PCOS)-Searching for Epigenetic Factors. Int J Mol Sci 2022; 23:ijms232314663. [PMID: 36498989 PMCID: PMC9736994 DOI: 10.3390/ijms232314663] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in women of reproductive age. Despite its incidence, the syndrome is poorly understood and remains underdiagnosed, and female patients are diagnosed with a delay. The heterogenous nature of this complex disorder results from the combined occurrence of genetic, environmental, endocrine, and behavioral factors. Primary clinical manifestations of PCOS are derived from the excess of androgens (anovulation, polycystic ovary morphology, lack of or scanty, irregular menstrual periods, acne and hirsutism), whereas the secondary manifestations include multiple metabolic, cardiovascular, and psychological disorders. Dietary and lifestyle factors play important roles in the development and course of PCOS, which suggests strong epigenetic and environmental influences. Many studies have shown a strong association between PCOS and chronic, low-grade inflammation both in the ovarian tissue and throughout the body. In the vast majority of PCOS patients, elevated values of inflammatory markers or their gene markers have been reported. Development of the vicious cycle of the chronic inflammatory state in PCOS is additionally stimulated by hyperinsulinemia and obesity. Changes in DNA methylation, histone acetylation and noncoding RNA levels are presented in this review in the context of oxidative stress, reactive oxygen species, and inflammatory signaling in PCOS. Epigenetic modulation of androgenic activity in response to inflammatory signaling is also discussed.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
- Correspondence:
| | - Seweryn Trojanowski
- Chair and Department of Obstetrics, Gynecology and Gynecological Oncology, Medical University of Warsaw, 03-242 Warsaw, Poland
| | - Anna Kociszewska
- Chair and Department of Obstetrics, Gynecology and Gynecological Oncology, Medical University of Warsaw, 03-242 Warsaw, Poland
| | - Grzegorz Szewczyk
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
10
|
Liver Regeneration: Changes in Oxidative Stress, Immune System, Cytokines, and Epigenetic Modifications Associated with Aging. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9018811. [PMID: 35936214 PMCID: PMC9352489 DOI: 10.1155/2022/9018811] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/25/2022] [Accepted: 06/29/2022] [Indexed: 01/10/2023]
Abstract
The regenerative capacity of the liver decreases with increase in age. In recent years, studies in mice have found that the regenerative capacity of the liver is associated with changes in the immune system of the liver, cytokines in the body, aging-related epigenetic modifications in the cell, and intracellular signaling pathways. In the immune system of the aging liver, monocytes and macrophages play an important role in tissue repair. During tissue repair, monocytes and macrophages undergo a series of functional and phenotypic changes to initiate and maintain tissue repair. Studies have discovered that knocking out macrophages in the liver during the repair phase results in significant impairment of liver regeneration. Furthermore, as the body ages, the secretion and function of cytokines undergo a series of changes. For example, the levels of interleukin-6, transforming growth factor-alpha, hepatocyte growth factor, and vascular endothelial growth factor undergo changes that alter hepatocyte regulation, thereby affecting its proliferation. In addition, body aging is accompanied by cellular aging, which leads to changes in gene expression and epigenetic modifications. Additionally, this in turn causes alterations in cell function, morphology, and division and affects the regenerative capacity of the liver. As the body ages, the activity of associated functional proteins, such as CCAAT-enhancer-binding proteins, p53, and switch/sucrose nonfermentable complex, changes in the liver, leading to alterations in several signaling pathways, such as the Hippo, PI3K-Akt, mTOR, and STAT3 pathways. Therefore, in recent years, research on aging and liver regeneration has primarily focused on the immune system, signaling pathways, epigenetic changes of senescent cells, and cytokine secretion in the liver. Hence, this review details the roles of these influencing factors in liver regeneration and impact of aging-related factors.
Collapse
|
11
|
Jima DD, Skaar DA, Planchart A, Motsinger-Reif A, Cevik SE, Park SS, Cowley M, Wright F, House J, Liu A, Jirtle RL, Hoyo C. Genomic map of candidate human imprint control regions: the imprintome. Epigenetics 2022; 17:1920-1943. [PMID: 35786392 PMCID: PMC9665137 DOI: 10.1080/15592294.2022.2091815] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Imprinted genes – critical for growth, metabolism, and neuronal function – are expressed from one parental allele. Parent-of-origin-dependent CpG methylation regulates this expression at imprint control regions (ICRs). Since ICRs are established before tissue specification, these methylation marks are similar across cell types. Thus, they are attractive for investigating the developmental origins of adult diseases using accessible tissues, but remain unknown. We determined genome-wide candidate ICRs in humans by performing whole-genome bisulphite sequencing (WGBS) of DNA derived from the three germ layers and from gametes. We identified 1,488 hemi-methylated candidate ICRs, including 19 of 25 previously characterized ICRs (https://humanicr.org/). Gamete methylation approached 0% or 100% in 332 ICRs (178 paternally and 154 maternally methylated), supporting parent-of-origin-specific methylation, and 65% were in well-described CTCF-binding or DNaseI hypersensitive regions. This draft of the human imprintome will allow for the systematic determination of the role of early-acquired imprinting dysregulation in the pathogenesis of human diseases and developmental and behavioural disorders.
Collapse
Affiliation(s)
- Dereje D Jima
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.,Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA
| | - David A Skaar
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.,Toxicology Program, North Carolina State University, Raleigh, NC, USA
| | - Antonio Planchart
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.,Toxicology Program, North Carolina State University, Raleigh, NC, USA
| | - Alison Motsinger-Reif
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.,National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Sebnem E Cevik
- Toxicology Program, North Carolina State University, Raleigh, NC, USA
| | - Sarah S Park
- Toxicology Program, North Carolina State University, Raleigh, NC, USA.,Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Michael Cowley
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.,Toxicology Program, North Carolina State University, Raleigh, NC, USA
| | - Fred Wright
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.,Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA
| | - John House
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.,Toxicology Program, North Carolina State University, Raleigh, NC, USA.,National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Andy Liu
- Department of Neurology, Duke University, School of Medicine, Durham, NC, USA
| | - Randy L Jirtle
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.,Toxicology Program, North Carolina State University, Raleigh, NC, USA
| | - Cathrine Hoyo
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.,Toxicology Program, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
12
|
Ling C, Bacos K, Rönn T. Epigenetics of type 2 diabetes mellitus and weight change - a tool for precision medicine? Nat Rev Endocrinol 2022; 18:433-448. [PMID: 35513492 DOI: 10.1038/s41574-022-00671-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/29/2022] [Indexed: 12/12/2022]
Abstract
Pioneering studies performed over the past few decades demonstrate links between epigenetics and type 2 diabetes mellitus (T2DM), the metabolic disorder with the most rapidly increasing prevalence in the world. Importantly, these studies identified epigenetic modifications, including altered DNA methylation, in pancreatic islets, adipose tissue, skeletal muscle and the liver from individuals with T2DM. As non-genetic factors that affect the risk of T2DM, such as obesity, unhealthy diet, physical inactivity, ageing and the intrauterine environment, have been associated with epigenetic modifications in healthy individuals, epigenetics probably also contributes to T2DM development. In addition, genetic factors associated with T2DM and obesity affect the epigenome in human tissues. Notably, causal mediation analyses found DNA methylation to be a potential mediator of genetic associations with metabolic traits and disease. In the past few years, translational studies have identified blood-based epigenetic markers that might be further developed and used for precision medicine to help patients with T2DM receive optimal therapy and to identify patients at risk of complications. This Review focuses on epigenetic mechanisms in the development of T2DM and the regulation of body weight in humans, with a special focus on precision medicine.
Collapse
Affiliation(s)
- Charlotte Ling
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden.
| | - Karl Bacos
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden
| | - Tina Rönn
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden
| |
Collapse
|
13
|
Reale A, Tagliatesta S, Zardo G, Zampieri M. Counteracting aged DNA methylation states to combat ageing and age-related diseases. Mech Ageing Dev 2022; 206:111695. [PMID: 35760211 DOI: 10.1016/j.mad.2022.111695] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 06/09/2022] [Accepted: 06/22/2022] [Indexed: 12/18/2022]
Abstract
DNA methylation (DNAm) overwrites information about multiple extrinsic factors on the genome. Age is one of these factors. Age causes characteristic DNAm changes that are thought to be not only major drivers of normal ageing but also precursors to diseases, cancer being one of these. Although there is still much to learn about the relationship between ageing, age-related diseases and DNAm, we now know how to interpret some of the effects caused by age in the form of changes in methylation marks at specific loci. In fact, these changes form the basis of the so called "epigenetic clocks", which translate the genomic methylation profile into an "epigenetic age". Epigenetic age does not only estimate chronological age but can also predict the risk of chronic diseases and mortality. Epigenetic age is believed to be one of the most accurate metrics of biological age. Initial evidence has recently been gathered pointing to the possibility that the rate of epigenetic ageing can be slowed down or even reversed. In this review, we discuss some of the most relevant advances in this field. Expected outcome is that this approach can provide insights into how to preserve health and reduce the impact of ageing diseases in humans.
Collapse
Affiliation(s)
- Anna Reale
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy.
| | - Stefano Tagliatesta
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00161 Rome, Italy.
| | - Giuseppe Zardo
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy.
| | - Michele Zampieri
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy.
| |
Collapse
|
14
|
Habibe JJ, Clemente-Olivo MP, de Vries CJ. How (Epi)Genetic Regulation of the LIM-Domain Protein FHL2 Impacts Multifactorial Disease. Cells 2021; 10:2611. [PMID: 34685595 PMCID: PMC8534169 DOI: 10.3390/cells10102611] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 01/13/2023] Open
Abstract
Susceptibility to complex pathological conditions such as obesity, type 2 diabetes and cardiovascular disease is highly variable among individuals and arises from specific changes in gene expression in combination with external factors. The regulation of gene expression is determined by genetic variation (SNPs) and epigenetic marks that are influenced by environmental factors. Aging is a major risk factor for many multifactorial diseases and is increasingly associated with changes in DNA methylation, leading to differences in gene expression. Four and a half LIM domains 2 (FHL2) is a key regulator of intracellular signal transduction pathways and the FHL2 gene is consistently found as one of the top hyper-methylated genes upon aging. Remarkably, FHL2 expression increases with methylation. This was demonstrated in relevant metabolic tissues: white adipose tissue, pancreatic β-cells, and skeletal muscle. In this review, we provide an overview of the current knowledge on regulation of FHL2 by genetic variation and epigenetic DNA modification, and the potential consequences for age-related complex multifactorial diseases.
Collapse
Affiliation(s)
- Jayron J. Habibe
- Department of Medical Biochemistry, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, 1105 AZ Amsterdam, The Netherlands; (J.J.H.); (M.P.C.-O.)
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands
| | - Maria P. Clemente-Olivo
- Department of Medical Biochemistry, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, 1105 AZ Amsterdam, The Netherlands; (J.J.H.); (M.P.C.-O.)
| | - Carlie J. de Vries
- Department of Medical Biochemistry, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, 1105 AZ Amsterdam, The Netherlands; (J.J.H.); (M.P.C.-O.)
| |
Collapse
|
15
|
Clemente-Olivo MP, Habibe JJ, Vos M, Ottenhoff R, Jongejan A, Herrema H, Zelcer N, Kooijman S, Rensen PCN, van Raalte DH, Nieuwdorp M, Eringa EC, de Vries CJ. Four-and-a-half LIM domain protein 2 (FHL2) deficiency protects mice from diet-induced obesity and high FHL2 expression marks human obesity. Metabolism 2021; 121:154815. [PMID: 34119536 DOI: 10.1016/j.metabol.2021.154815] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/26/2021] [Accepted: 06/08/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Four-and-a-Half-LIM-domain-protein 2 (FHL2) modulates multiple signal transduction pathways but has not been implicated in obesity or energy metabolism. In humans, methylation and expression of the FHL2 gene increases with age, and high FHL2 expression is associated with increased body weight in humans and mice. This led us to hypothesize that FHL2 is a determinant of diet-induced obesity. METHODS FHL2-deficient (FHL2-/-) and wild type male mice were fed a high-fat diet. Metabolic phenotyping of these mice, as well as transcriptional analysis of key metabolic tissues was performed. Correlation of the expression of FHL2 and relevant genes was assessed in datasets from white adipose tissue of individuals with and without obesity. RESULTS FHL2 Deficiency protects mice from high-fat diet-induced weight gain, whereas glucose handling is normal. We observed enhanced energy expenditure, which may be explained by a combination of changes in multiple tissues; mild activation of brown adipose tissue with increased fatty acid uptake, increased cardiac glucose uptake and browning of white adipose tissue. Corroborating our findings in mice, expression of FHL2 in human white adipose tissue positively correlates with obesity and negatively with expression of browning-associated genes. CONCLUSION Our results position FHL2 as a novel regulator of obesity and energy expenditure in mice and human. Given that FHL2 expression increases during aging, we now show that low FHL2 expression associates with a healthy metabolic state.
Collapse
Affiliation(s)
- Maria P Clemente-Olivo
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Jayron J Habibe
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Department of Physiology, Amsterdam UMC, Amsterdam Cardiovascular Sciences, location VUmc, Amsterdam, the Netherlands
| | - Mariska Vos
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Roelof Ottenhoff
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Aldo Jongejan
- Department of Bioinformatics, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Hilde Herrema
- Department of Experimental Vascular Medicine, Amsterdam UMC, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Noam Zelcer
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Sander Kooijman
- Department of Medicine, Division of Endocrinology, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Daniël H van Raalte
- Department of Internal Medicine, Diabetes Center, Amsterdam UMC, Amsterdam Cardiovascular Sciences Amsterdam, the Netherlands
| | - Max Nieuwdorp
- Department of Experimental Vascular Medicine, Amsterdam UMC, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Etto C Eringa
- Department of Physiology, Amsterdam UMC, Amsterdam Cardiovascular Sciences, location VUmc, Amsterdam, the Netherlands
| | - Carlie J de Vries
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands.
| |
Collapse
|
16
|
Oblak L, van der Zaag J, Higgins-Chen AT, Levine ME, Boks MP. A systematic review of biological, social and environmental factors associated with epigenetic clock acceleration. Ageing Res Rev 2021; 69:101348. [PMID: 33930583 DOI: 10.1016/j.arr.2021.101348] [Citation(s) in RCA: 213] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 04/01/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Abstract
Aging involves a diverse set of biological changes accumulating over time that leads to increased risk of morbidity and mortality. Epigenetic clocks are now widely used to quantify biological aging, in order to investigate determinants that modify the rate of aging and to predict age-related outcomes. Numerous biological, social and environmental factors have been investigated for their relationship to epigenetic clock acceleration and deceleration. The aim of this review was to synthesize general trends concerning the associations between human epigenetic clocks and these investigated factors. We conducted a systematic review of all available literature and included 156 publications across 4 resource databases. We compiled a list of all presently existing blood-based epigenetic clocks. Subsequently, we created an extensive dataset of over 1300 study findings in which epigenetic clocks were utilized in blood tissue of human subjects to assess the relationship between these clocks and numeral environmental exposures and human traits. Statistical analysis was possible on 57 such relationships, measured across 4 different epigenetic clocks (Hannum, Horvath, Levine and GrimAge). We found that the Horvath, Hannum, Levine and GrimAge epigenetic clocks tend to agree in direction of effects, but vary in size. Body mass index, HIV infection, and male sex were significantly associated with acceleration of one or more epigenetic clocks. Acceleration of epigenetic clocks was also significantly related to mortality, cardiovascular disease, cancer and diabetes. Our findings provide a graphical and numerical synopsis of the past decade of epigenetic age estimation research and indicate areas where further attention could be focused in the coming years.
Collapse
|
17
|
Yang HJ, Koh E, Sung MK, Kang H. Changes Induced by Mind-Body Intervention Including Epigenetic Marks and Its Effects on Diabetes. Int J Mol Sci 2021; 22:ijms22031317. [PMID: 33525677 PMCID: PMC7865217 DOI: 10.3390/ijms22031317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 12/20/2022] Open
Abstract
Studies have evidenced that epigenetic marks associated with type 2 diabetes (T2D) can be inherited from parents or acquired through fetal and early-life events, as well as through lifelong environments or lifestyles, which can increase the risk of diabetes in adulthood. However, epigenetic modifications are reversible, and can be altered through proper intervention, thus mitigating the risk factors of T2D. Mind-body intervention (MBI) refers to interventions like meditation, yoga, and qigong, which deal with both physical and mental well-being. MBI not only induces psychological changes, such as alleviation of depression, anxiety, and stress, but also physiological changes like parasympathetic activation, lower cortisol secretion, reduced inflammation, and aging rate delay, which are all risk factors for T2D. Notably, MBI has been reported to reduce blood glucose in patients with T2D. Herein, based on recent findings, we review the effects of MBI on diabetes and the mechanisms involved, including epigenetic modifications.
Collapse
Affiliation(s)
- Hyun-Jeong Yang
- Korea Institute of Brain Science, Seoul 06022, Korea; (M.-K.S.); (H.K.)
- Department of Integrative Health Care, University of Brain Education, Cheonan 31228, Korea
- Correspondence:
| | - Eugene Koh
- Temasek Life Sciences Laboratories, Singapore 117604, Singapore;
| | - Min-Kyu Sung
- Korea Institute of Brain Science, Seoul 06022, Korea; (M.-K.S.); (H.K.)
| | - Hojung Kang
- Korea Institute of Brain Science, Seoul 06022, Korea; (M.-K.S.); (H.K.)
| |
Collapse
|
18
|
Characterization of the effects of age and childhood maltreatment on ELOVL2 DNA methylation. Dev Psychopathol 2021; 34:864-874. [PMID: 33461631 DOI: 10.1017/s0954579420001972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
DNA methylation of the elongation of very long chain fatty acids protein 2 (ELOVL2) was suggested as a biomarker of biological aging, while childhood maltreatment (CM) has been associated with accelerated biological aging. We investigated the association of age and CM experiences with ELOVL2 methylation in peripheral blood mononuclear cells (PBMC). Furthermore, we investigated ELOVL2 methylation in the umbilical cord blood mononuclear cells (UBMC) of newborns of mothers with and without CM. PBMC and UBMC were isolated from 113 mother-newborn dyads and genomic DNA was extracted. Mothers with and without CM experiences were recruited directly postpartum. Mass array spectrometry and pyrosequencing were used for methylation analyses of ELOVL2 intron 1, and exon 1 and 5' end, respectively. ELOVL2 5' end and intron 1 methylation increased with higher age but were not associated with CM experiences. On the contrary, overall ELOVL2 exon 1 methylation increased with higher CM, but these changes were minimal and did not increase with age. Maternal CM experiences and neonatal methylation of ELOVL2 intron 1 or exon 1 were not significantly correlated. Our study suggests region-specific effects of chronological age and experienced CM on ELOVL2 methylation and shows that the epigenetic biomarker for age within the ELOVL2 gene does not show accelerated biological aging years after CM exposure.
Collapse
|
19
|
Maude H, Sanchez-Cabanillas C, Cebola I. Epigenetics of Hepatic Insulin Resistance. Front Endocrinol (Lausanne) 2021; 12:681356. [PMID: 34046015 PMCID: PMC8147868 DOI: 10.3389/fendo.2021.681356] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 04/20/2021] [Indexed: 01/14/2023] Open
Abstract
Insulin resistance (IR) is largely recognized as a unifying feature that underlies metabolic dysfunction. Both lifestyle and genetic factors contribute to IR. Work from recent years has demonstrated that the epigenome may constitute an interface where different signals may converge to promote IR gene expression programs. Here, we review the current knowledge of the role of epigenetics in hepatic IR, focusing on the roles of DNA methylation and histone post-translational modifications. We discuss the broad epigenetic changes observed in the insulin resistant liver and its associated pathophysiological states and leverage on the wealth of 'omics' studies performed to discuss efforts in pinpointing specific loci that are disrupted by these changes. We envision that future studies, with increased genomic resolution and larger cohorts, will further the identification of biomarkers of early onset hepatic IR and assist the development of targeted interventions. Furthermore, there is growing evidence to suggest that persistent epigenetic marks may be acquired over prolonged exposure to disease or deleterious exposures, highlighting the need for preventative medicine and long-term lifestyle adjustments to avoid irreversible or long-term alterations in gene expression.
Collapse
Affiliation(s)
| | | | - Inês Cebola
- *Correspondence: Hannah Maude, ; Inês Cebola,
| |
Collapse
|
20
|
FeSTwo, a two-step feature selection algorithm based on feature engineering and sampling for the chronological age regression problem. Comput Biol Med 2020; 125:104008. [DOI: 10.1016/j.compbiomed.2020.104008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 08/26/2020] [Accepted: 08/26/2020] [Indexed: 02/02/2023]
|
21
|
Guevara EE, Lawler RR, Staes N, White CM, Sherwood CC, Ely JJ, Hopkins WD, Bradley BJ. Age-associated epigenetic change in chimpanzees and humans. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190616. [PMID: 32951551 DOI: 10.1098/rstb.2019.0616] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Methylation levels have been shown to change with age at sites across the human genome. Change at some of these sites is so consistent across individuals that it can be used as an 'epigenetic clock' to predict an individual's chronological age to within a few years. Here, we examined how the pattern of epigenetic ageing in chimpanzees compares with humans. We profiled genome-wide blood methylation levels by microarray for 113 samples from 83 chimpanzees aged 1-58 years (26 chimpanzees were sampled at multiple ages during their lifespan). Many sites (greater than 65 000) showed significant change in methylation with age and around one-third (32%) of these overlap with sites showing significant age-related change in humans. At over 80% of sites showing age-related change in both species, chimpanzees displayed a significantly faster rate of age-related change in methylation than humans. We also built a chimpanzee-specific epigenetic clock that predicted age in our test dataset with a median absolute deviation from known age of only 2.4 years. However, our chimpanzee clock showed little overlap with previously constructed human clocks. Methylation at CpGs comprising our chimpanzee clock showed moderate heritability. Although the use of a human microarray for profiling chimpanzees biases our results towards regions with shared genomic sequence between the species, nevertheless, our results indicate that there is considerable conservation in epigenetic ageing between chimpanzees and humans, but also substantial divergence in both rate and genomic distribution of ageing-associated sites. This article is part of the theme issue 'Evolution of the primate ageing process'.
Collapse
Affiliation(s)
- Elaine E Guevara
- Department of Evolutionary Anthropology, Duke University, Durham, NC 27708, USA.,Center for the Advanced Study of Human Paleobiology, Department of Anthropology, The George Washington University, Washington, DC 20052, USA
| | - Richard R Lawler
- Department of Sociology and Anthropology, James Madison University, Harrisonburg, VA 22807, USA
| | - Nicky Staes
- Center for the Advanced Study of Human Paleobiology, Department of Anthropology, The George Washington University, Washington, DC 20052, USA.,Behavioural Ecology and Ecophysiology Group, Department of Biology, University of Antwerp, Wilrijk, Belgium.,Centre for Research and Conservation, Royal Zoological Society of Antwerp, Antwerp, Belgium
| | - Cassandra M White
- Center for the Advanced Study of Human Paleobiology, Department of Anthropology, The George Washington University, Washington, DC 20052, USA
| | - Chet C Sherwood
- Center for the Advanced Study of Human Paleobiology, Department of Anthropology, The George Washington University, Washington, DC 20052, USA
| | | | - William D Hopkins
- Keeling Center for Comparative Medicine and Research, University of Texas MD Anderson Cancer Center, Bastrop, TX 78602, USA
| | - Brenda J Bradley
- Center for the Advanced Study of Human Paleobiology, Department of Anthropology, The George Washington University, Washington, DC 20052, USA
| |
Collapse
|
22
|
Ling C. Epigenetic regulation of insulin action and secretion - role in the pathogenesis of type 2 diabetes. J Intern Med 2020; 288:158-167. [PMID: 32363639 DOI: 10.1111/joim.13049] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/17/2020] [Accepted: 01/30/2020] [Indexed: 12/21/2022]
Abstract
The prevalence of type 2 diabetes (T2D) is rapidly increasing worldwide. Obesity, physical inactivity and ageing increase the risk of T2D. Epigenetic modifications can change due to environmental exposures and may thereby predispose to disease. This review aims at summarizing recent advances in epigenetics related to T2D, with a special focus on impaired insulin action and secretion in humans. There will be an emphasis on analyses in human tissues; both from T2D case-control cohorts and intervention studies. Current data support an important role for epigenetics in the pathogenesis of T2D. Numerous studies have found differential DNA methylation and gene expression in skeletal muscle, adipose tissue, the liver and pancreatic islets from subjects with T2D compared with nondiabetic controls. For example, PDX1 has increased DNA methylation and decreased expression in pancreatic islets from patients with T2D compared with nondiabetic controls. Nongenetic risk factors for T2D such as ageing, unhealthy diets and physical activity do also impact the epigenome in human tissues. Interestingly, physical activity altered DNA methylation of candidate genes for T2D such as THADA in muscle and FTO, KCNQ1 and TCF7L2 in adipose tissue. There is also a strong interaction between genetic and epigenetic factors that together seem to affect T2D. mQTL studies in human adipose tissue and pancreatic islets showed that SNPs associated with DNA methylation levels in numerous sites. Several of these SNPs are also associated with T2D. Recent data also support that DNA methylation of some sites in blood may be developed into biomarkers that predict T2D since methylation of, for example TXNIP, ABCG1 and SREBF1 associated with future T2D. Future studies should use this information for development of new therapies and biomarkers and thereby improve prediction, prevention and treatment of T2D and its complications.
Collapse
Affiliation(s)
- C Ling
- From the, Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden
| |
Collapse
|
23
|
Chen X, Lin Q, Wen J, Lin W, Liang J, Huang H, Li L, Huang J, Chen F, Liu D, Chen G. Whole genome bisulfite sequencing of human spermatozoa reveals differentially methylated patterns from type 2 diabetic patients. J Diabetes Investig 2020; 11:856-864. [PMID: 31869513 PMCID: PMC7378413 DOI: 10.1111/jdi.13201] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 12/15/2019] [Accepted: 12/19/2019] [Indexed: 12/14/2022] Open
Abstract
AIMS/INTRODUCTION The incidence of type 2 diabetes mellitus is increasing worldwide, and it might partly cause metabolic disorder and type 2 diabetes mellitus susceptibility in patients' offspring through epigenetic modification. However, the underlying mechanisms remain largely unclear. Recent studies have shown a potential link between deoxyribonucleic acid methylation in paternal sperm and susceptibility to type 2 diabetes mellitus in offspring, so this article focuses on whether the whole-genome methylation profiles of spermatozoa in type 2 diabetes mellitus patients have changed. MATERIALS AND METHODS We investigated the genome-wide deoxyribonucleic acid methylation profiles in spermatozoa by comparing eight individuals with type 2 diabetes mellitus and nine non-diabetic controls using whole-genome bisulfite sequencing method. RESULTS First, we found that the proportion of methylated cytosine in the whole genome of the type 2 diabetes mellitus group was slightly lower than that of the control group. Interestingly, the proportion of methylated cytosines in the CG context decreased, and the proportion of methylated cytosines in the CHG context (H = A, T or C) increased in the type 2 diabetes mellitus group, but the proportion of methylated cytosines in the CHH context (H = A, T or C) barely changed. The methylated cytosines in the CG context were mainly distributed at the high methylated level, whereas methylated cytosines in the CHG context and methylated cytosines in the CHH context were mainly distributed at the low and middle methylated level in both groups. Second, functional enrichment analysis showed that differentially methylated genes played a significant role in nervous system development and cell metabolism. Finally, we identified 10 top type 2 diabetes mellitus-related differentially methylated genes, including IRS1, PRKCE, FTO, PPARGC1A, KCNQ1, ATP10A, GHR, CREB1, PRKAR1A and HNF1B. CONCLUSIONS Our study provides the first evidence for deoxyribonucleic acid methylation reprogramming in spermatozoa of type 2 diabetes mellitus patients, and provides a new basis for explaining the complex mechanism of type 2 diabetes mellitus susceptibility in offspring.
Collapse
Affiliation(s)
- Xiongfeng Chen
- Department of Scientific ResearchFujian Provincial HospitalFuzhouFujianChina
| | - Qinghua Lin
- Department of Scientific ResearchFujian Academy of Medical SciencesFuzhouFujianChina
| | - Junping Wen
- Department of Endocrinology and MetabolismFujian Provincial HospitalShengli Clinical Medical College of Fujian Medical UniversityFuzhouFujianChina
| | - Wei Lin
- Department of Endocrinology and MetabolismFujian Provincial HospitalShengli Clinical Medical College of Fujian Medical UniversityFuzhouFujianChina
| | - Jixing Liang
- Department of Endocrinology and MetabolismFujian Provincial HospitalShengli Clinical Medical College of Fujian Medical UniversityFuzhouFujianChina
| | - Huibin Huang
- Department of Endocrinology and MetabolismFujian Provincial HospitalShengli Clinical Medical College of Fujian Medical UniversityFuzhouFujianChina
| | - Liantao Li
- Department of Endocrinology and MetabolismFujian Provincial HospitalShengli Clinical Medical College of Fujian Medical UniversityFuzhouFujianChina
| | - Jianxin Huang
- Department of Clinical LaboratoryFujian Provincial HospitalFuzhouFujianChina
| | - Falin Chen
- Department of Clinical LaboratoryFujian Provincial HospitalFuzhouFujianChina
| | - Deli Liu
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational BiomedicineNew York CityNew YorkUSA
| | - Gang Chen
- Department of Scientific ResearchFujian Academy of Medical SciencesFuzhouFujianChina
- Department of Endocrinology and MetabolismFujian Provincial HospitalShengli Clinical Medical College of Fujian Medical UniversityFuzhouFujianChina
| |
Collapse
|
24
|
Liu Y, Shen Y, Guo T, Parnell LD, Westerman KE, Smith CE, Ordovas JM, Lai CQ. Statin Use Associates With Risk of Type 2 Diabetes via Epigenetic Patterns at ABCG1. Front Genet 2020; 11:622. [PMID: 32612641 PMCID: PMC7308584 DOI: 10.3389/fgene.2020.00622] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/22/2020] [Indexed: 11/13/2022] Open
Abstract
Statin is the medication most widely prescribed to reduce plasma cholesterol levels. Yet, how the medication contributes to diabetes risk and impaired glucose metabolism is not clear. This study aims to examine the epigenetic mechanisms of ABCG1 through which statin use associates with risk of type 2 diabetes. We determined the association between the statin use, DNA methylation at ABCG1 and type 2 diabetes/glycemic traits in the Framingham Heart Study Offspring (FHS, n = 2741), with validation in the Women’s Health Initiative Study (WHI, n = 2020). The causal effect of statin use on the risk of type 2 diabetes was examined using a two-step Mendelian randomization approach. Next, based on transcriptome analysis, we determined the links between the medication-associated epigenetic status of ABCG1 and biological pathways on the pathogenesis of type 2 diabetes. Our results showed that DNA methylation levels at cg06500161 of ABCG1 were positively associated with the use of statin, type 2 diabetes and related traits (fasting glucose and insulin) in FHS and WHI. Two-step Mendelian randomization suggested a causal effect of statin use on type 2 diabetes and related traits through epigenetic mechanisms, specifically, DNA methylation at cg06500161. Our results highlighted that gene expression of ABCG1, ABCA1 and ACSL3, involved in both cholesterol metabolism and glycemic pathways, was inversely associated with statin use, CpG methylation, and diabetic signatures. We concluded that DNA methylation site cg06500161 at ABCG1 is a mediator of the association between statins and risk of type 2 diabetes.
Collapse
Affiliation(s)
- Yuwei Liu
- School of Public Health, Fudan University, Shanghai, China.,Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Yu Shen
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Tao Guo
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States.,Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Laurence D Parnell
- USDA Agricultural Research Service, Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Kenneth E Westerman
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Caren E Smith
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Jose M Ordovas
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States.,IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain.,Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Chao-Qiang Lai
- USDA Agricultural Research Service, Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| |
Collapse
|
25
|
Treviño LS, Dong J, Kaushal A, Katz TA, Jangid RK, Robertson MJ, Grimm SL, Ambati CSR, Putluri V, Cox AR, Kim KH, May TD, Gallo MR, Moore DD, Hartig SM, Foulds CE, Putluri N, Coarfa C, Walker CL. Epigenome environment interactions accelerate epigenomic aging and unlock metabolically restricted epigenetic reprogramming in adulthood. Nat Commun 2020; 11:2316. [PMID: 32385268 PMCID: PMC7210260 DOI: 10.1038/s41467-020-15847-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 03/19/2020] [Indexed: 12/13/2022] Open
Abstract
Our early-life environment has a profound influence on developing organs that impacts metabolic function and determines disease susceptibility across the life-course. Using a rat model for exposure to an endocrine disrupting chemical (EDC), we show that early-life chemical exposure causes metabolic dysfunction in adulthood and reprograms histone marks in the developing liver to accelerate acquisition of an adult epigenomic signature. This epigenomic reprogramming persists long after the initial exposure, but many reprogrammed genes remain transcriptionally silent with their impact on metabolism not revealed until a later life exposure to a Western-style diet. Diet-dependent metabolic disruption was largely driven by reprogramming of the Early Growth Response 1 (EGR1) transcriptome and production of metabolites in pathways linked to cholesterol, lipid and one-carbon metabolism. These findings demonstrate the importance of epigenome:environment interactions, which early in life accelerate epigenomic aging, and later in adulthood unlock metabolically restricted epigenetic reprogramming to drive metabolic dysfunction. Early life exposure to environmental stressors, including endocrine disrupting chemicals (EDCs), can impact health later in life. Here, the authors show that neonatal EDC exposure in rats causes epigenetic reprogramming in the liver, which is transcriptionally silent until animals are placed on a Western-style diet.
Collapse
Affiliation(s)
- Lindsey S Treviño
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,Division of Health Equities, Department of Population Sciences, City of Hope, Duarte, CA, 91010, USA
| | - Jianrong Dong
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ahkilesh Kaushal
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Tiffany A Katz
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Rahul Kumar Jangid
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Matthew J Robertson
- Advanced Technology Cores, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Sandra L Grimm
- Advanced Technology Cores, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chandra Shekar R Ambati
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Vasanta Putluri
- Advanced Technology Cores, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Aaron R Cox
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kang Ho Kim
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Thaddeus D May
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Morgan R Gallo
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030, USA
| | - David D Moore
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Sean M Hartig
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Charles E Foulds
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Cristian Coarfa
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Cheryl Lyn Walker
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA. .,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
26
|
Fraszczyk E, Luijten M, Spijkerman AMW, Snieder H, Wackers PFK, Bloks VW, Nicoletti CF, Nonino CB, Crujeiras AB, Buurman WA, Greve JW, Rensen SS, Wolffenbuttel BHR, van Vliet-Ostaptchouk JV. The effects of bariatric surgery on clinical profile, DNA methylation, and ageing in severely obese patients. Clin Epigenetics 2020; 12:14. [PMID: 31959221 PMCID: PMC6972025 DOI: 10.1186/s13148-019-0790-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/27/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Severe obesity is a growing, worldwide burden and conventional therapies including radical change of diet and/or increased physical activity have limited results. Bariatric surgery has been proposed as an alternative therapy showing promising results. It leads to substantial weight loss and improvement of comorbidities such as type 2 diabetes. Increased adiposity is associated with changes in epigenetic profile, including DNA methylation. We investigated the effect of bariatric surgery on clinical profile, DNA methylation, and biological age estimated using Horvath's epigenetic clock. RESULTS To determine the impact of bariatric surgery and subsequent weight loss on clinical traits, a cohort of 40 severely obese individuals (BMI = 30-73 kg/m2) was examined at the time of surgery and at three follow-up visits, i.e., 3, 6, and 12 months after surgery. The majority of the individuals were women (65%) and the mean age at surgery was 45.1 ± 8.1 years. We observed a significant decrease over time in BMI, fasting glucose, HbA1c, HOMA-IR, insulin, total cholesterol, triglycerides, LDL and free fatty acids levels, and a significant small increase in HDL levels (all p values < 0.05). Epigenome-wide association analysis revealed 4857 differentially methylated CpG sites 12 months after surgery (at Bonferroni-corrected p value < 1.09 × 10-7). Including BMI change in the model decreased the number of significantly differentially methylated CpG sites by 51%. Gene set enrichment analysis identified overrepresentation of multiple processes including regulation of transcription, RNA metabolic, and biosynthetic processes in the cell. Bariatric surgery in severely obese patients resulted in a decrease in both biological age and epigenetic age acceleration (EAA) (mean = - 0.92, p value = 0.039). CONCLUSIONS Our study shows that bariatric surgery leads to substantial BMI decrease and improvement of clinical outcomes observed 12 months after surgery. These changes explained part of the association between bariatric surgery and DNA methylation. We also observed a small, but significant improvement of biological age. These epigenetic changes may be modifiable by environmental lifestyle factors and could be used as potential biomarkers for obesity and in the future for obesity related comorbidities.
Collapse
Affiliation(s)
- Eliza Fraszczyk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Mirjam Luijten
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Annemieke M W Spijkerman
- Centre for Nutrition, Prevention and Health services, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Paul F K Wackers
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Vincent W Bloks
- Department of Pediatrics, section of Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Carolina F Nicoletti
- Laboratory of Nutrigenomics Studies, Department of Internal Medicine, Ribeirão Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Carla B Nonino
- Laboratory of Nutrigenomics Studies, Department of Health Sciences, Ribeirão Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Ana B Crujeiras
- Epigenomics in Endocrinology and Nutrition, Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS) and Santiago de Compostela University (USC), Santiago de Compostela, Spain.,CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), Madrid, Spain
| | - Wim A Buurman
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Jan Willem Greve
- Department of Surgery, Zuyderland Medical Center Heerlen, Dutch Obesity Clinic South, Heerlen, The Netherlands.,Department of Surgery, Maastricht University Medical Center, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht, The Netherlands
| | - Sander S Rensen
- Department of Surgery, Maastricht University Medical Center, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht, The Netherlands
| | - Bruce H R Wolffenbuttel
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jana V van Vliet-Ostaptchouk
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands. .,Genomics Coordination Center, Department of Genetics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713, GZ, Groningen, The Netherlands.
| |
Collapse
|
27
|
Bacalini MG, Franceschi C, Gentilini D, Ravaioli F, Zhou X, Remondini D, Pirazzini C, Giuliani C, Marasco E, Gensous N, Di Blasio AM, Ellis E, Gramignoli R, Castellani G, Capri M, Strom S, Nardini C, Cescon M, Grazi GL, Garagnani P. Molecular Aging of Human Liver: An Epigenetic/Transcriptomic Signature. J Gerontol A Biol Sci Med Sci 2019; 74:1-8. [PMID: 29554203 DOI: 10.1093/gerona/gly048] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Indexed: 12/12/2022] Open
Abstract
The feasibility of liver transplantation from old healthy donors suggests that this organ is able to preserve its functionality during aging. To explore the biological basis of this phenomenon, we characterized the epigenetic profile of liver biopsies collected from 45 healthy liver donors ranging from 13 to 90 years old using the Infinium HumanMethylation450 BeadChip. The analysis indicates that a large remodeling in DNA methylation patterns occurs, with 8,823 age-associated differentially methylated CpG probes. Notably, these age-associated changes tended to level off after the age of 60, as confirmed by Horvath's clock. Using stringent selection criteria, we further identified a DNA methylation signature of aging liver including 75 genomic regions. We demonstrated that this signature is specific for liver compared to other tissues and that it is able to detect biological age-acceleration effects associated with obesity. Finally, we combined DNA methylation measurements with available expression data. Although the intersection between the two omic characterizations was low, both approaches suggested a previously unappreciated role of epithelial-mesenchymal transition and Wnt-signaling pathways in the aging of human liver.
Collapse
Affiliation(s)
| | - Claudio Franceschi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Italy.,DIMES-Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, Bologna, Italy.,CIG, Interdepartmental Center 'L. Galvani', Alma Mater Studiorum, Bologna, Italy
| | - Davide Gentilini
- Department of Brain and Behavioral Sciences, University of Pavia, Italy
| | - Francesco Ravaioli
- DIMES-Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, Bologna, Italy
| | - Xiaoyuan Zhou
- Group of Clinical Genomic Networks, Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, PR China.,University of Chinese Academy of Sciences, Beijing, PR China.,Department of Neurology, University of San Francisco, California
| | - Daniel Remondini
- Department of Physics and Astronomy (DIFA) and INFN Sez. Bologna, Alma Mater Studiorum, Italy
| | | | - Cristina Giuliani
- Department of Biological Geological and Environmental Sciences, Laboratory of Molecular Anthropology and Centre for Genome Biology, University of Bologna, Italy
| | - Elena Marasco
- CIG, Interdepartmental Center 'L. Galvani', Alma Mater Studiorum, Bologna, Italy
| | - Noémie Gensous
- DIMES-Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, Bologna, Italy
| | | | - Ewa Ellis
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Gastone Castellani
- CIG, Interdepartmental Center 'L. Galvani', Alma Mater Studiorum, Bologna, Italy.,Department of Biological Geological and Environmental Sciences, Laboratory of Molecular Anthropology and Centre for Genome Biology, University of Bologna, Italy
| | - Miriam Capri
- DIMES-Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, Bologna, Italy.,CIG, Interdepartmental Center 'L. Galvani', Alma Mater Studiorum, Bologna, Italy
| | - Stephen Strom
- Department of Laboratory Medicine, Karolinska Institute and Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Christine Nardini
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,CNR IAC "Mauro Picone", Roma, Italy.,Personal Genomics S.r.l., Verona, Italy
| | - Matteo Cescon
- General Surgery and Transplant Unit, Department of Medical and Surgical Sciences, Sant'Orsola-Malpighi Hospital, Bologna, Italy
| | | | - Paolo Garagnani
- DIMES-Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, Bologna, Italy.,CIG, Interdepartmental Center 'L. Galvani', Alma Mater Studiorum, Bologna, Italy.,Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Applied Biomedical Research Center, S. Orsola-Malpighi Polyclinic, Bologna, Italy.,Institute of Molecular Genetics (IGM)-CNR, Unit of Bologna, Italy.,Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopaedic Institute, Bologna, Italy
| |
Collapse
|
28
|
Liu Y, Wang M, Marcora EM, Zhang B, Goate AM. Promoter DNA hypermethylation - Implications for Alzheimer's disease. Neurosci Lett 2019; 711:134403. [PMID: 31351091 PMCID: PMC6759378 DOI: 10.1016/j.neulet.2019.134403] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 07/12/2019] [Accepted: 07/23/2019] [Indexed: 12/31/2022]
Abstract
Recent methylome-wide association studies (MWAS) in humans have solidified the concept that aberrant DNA methylation is associated with Alzheimer's disease (AD). We summarize these findings to improve the understanding of mechanisms governing DNA methylation pertinent to transcriptional regulation, with an emphasis of AD-associated promoter DNA hypermethylation, which establishes an epigenetic barrier for transcriptional activation. By considering brain cell type specific expression profiles that have been published only for non-demented individuals, we detail functional activities of selected neuron, microglia, and astrocyte-enriched genes (AGAP2, DUSP6 and GPR37L1, respectively), which are DNA hypermethylated at promoters in AD. We highlight future directions in MWAS including experimental confirmation, functional relevance to AD, cell type-specific temporal characterization, and mechanism investigation.
Collapse
Affiliation(s)
- Yiyuan Liu
- Department of Neuroscience and Department of Genetics and Genomic Sciences, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY, 10029, USA
| | - Minghui Wang
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA.
| | - Edoardo M Marcora
- Department of Neuroscience and Department of Genetics and Genomic Sciences, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY, 10029, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Alison M Goate
- Department of Neuroscience and Department of Genetics and Genomic Sciences, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY, 10029, USA
| |
Collapse
|
29
|
Gillberg L, Rönn T, Jørgensen SW, Perfilyev A, Hjort L, Nilsson E, Brøns C, Vaag A, Ling C. Fasting unmasks differential fat and muscle transcriptional regulation of metabolic gene sets in low versus normal birth weight men. EBioMedicine 2019; 47:341-351. [PMID: 31439477 PMCID: PMC6796584 DOI: 10.1016/j.ebiom.2019.08.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/19/2019] [Accepted: 08/06/2019] [Indexed: 12/17/2022] Open
Abstract
Background Individuals born with low birth weight (LBW) have an increased risk of metabolic diseases when exposed to diets rich in calories and fat but may respond to fasting in a metabolically preferential manner. We hypothesized that impaired foetal growth is associated with differential regulation of gene expression and epigenetics in metabolic tissues in response to fasting in young adulthood. Methods Genome-wide expression and DNA methylation were analysed in subcutaneous adipose tissue (SAT) and skeletal muscle from LBW and normal birth weight (NBW) men after 36 h fasting and after an isocaloric control study using microarrays. Findings Transcriptome analyses revealed that expression of genes involved in oxidative phosphorylation (OXPHOS) and other key metabolic pathways were lower in SAT from LBW vs NBW men after the control study, but paradoxically higher in LBW vs NBW men after 36 h fasting. Thus, fasting was associated with downregulated OXPHOS and metabolic gene sets in NBW men only. Likewise, in skeletal muscle only NBW men downregulated OXPHOS genes with fasting. Few epigenetic changes were observed in SAT and muscle between the groups. Interpretation Our results provide insights into the molecular mechanisms in muscle and adipose tissue governing a differential metabolic response in subjects with impaired foetal growth when exposed to fasting in adulthood. The results support the concept of developmental programming of metabolic diseases including type 2 diabetes. Fund The Swedish Research Council, the Danish Council for Strategic Research, the Novo Nordisk foundation, the Swedish Foundation for Strategic Research, The European Foundation for the Study of Diabetes, The EU 6th Framework EXGENESIS grant and Rigshospitalet.
Collapse
Affiliation(s)
- Linn Gillberg
- Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Tina Rönn
- Department of Clinical Sciences, Epigenetics and Diabetes Unit, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | | | - Alexander Perfilyev
- Department of Clinical Sciences, Epigenetics and Diabetes Unit, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Line Hjort
- Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
| | - Emma Nilsson
- Department of Clinical Sciences, Epigenetics and Diabetes Unit, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Charlotte Brøns
- Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
| | - Allan Vaag
- Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular, Renal and Metabolism (CVRM), Translational Medical Unit, Early Clinical Development, IMED Biotech Unit, AstraZeneca, Mölndal, Sweden
| | - Charlotte Ling
- Department of Clinical Sciences, Epigenetics and Diabetes Unit, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden.
| |
Collapse
|
30
|
Chen J, Zou Q, Lv D, Raza MA, Wang X, Chen Y, Xi X, Li P, Wen A, Zhu L, Tang G, Li M, Li X, Jiang Y. Comprehensive transcriptional profiling of aging porcine liver. PeerJ 2019; 7:e6949. [PMID: 31149403 PMCID: PMC6526898 DOI: 10.7717/peerj.6949] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 04/09/2019] [Indexed: 01/04/2023] Open
Abstract
Background Aging is a major risk factor for the development of many diseases, and the liver, as the most important metabolic organ, is significantly affected by aging. It has been shown that the liver weight tends to increase in rodents and decrease in humans with age. Pigs have a genomic structure, with physiological as well as biochemical features that are similar to those of humans, and have therefore been used as a valuable model for studying human diseases. The molecular mechanisms of the liver aging of large mammals on a comprehensive transcriptional level remain poorly understood. The pig is an ideal model animal to clearly and fully understand the molecular mechanism underlying human liver aging. Methods In this study, four healthy female Yana pigs (an indigenous Chinese breed) were investigated: two young sows (180-days-old) and two old sows (8-years-old). High throughput RNA sequencing was performed to evaluate the expression profiles of messenger RNA, long non-coding RNAs, micro RNAs, and circular RNAs during the porcine liver aging process. Gene Ontology (GO) analysis was performed to investigate the biological functions of age-related genes. Results A number of age-related genes were identified in the porcine liver. GO annotation showed that up-regulated genes were mainly related to immune response, while the down-regulated genes were mainly related to metabolism. Moreover, several lncRNAs and their target genes were also found to be differentially expressed during liver aging. In addition, the multi-group cooperative control relationships and constructed circRNA-miRNA co-expression networks were assessed during liver aging. Conclusions Numerous age-related genes were identified and circRNA-miRNA co-expression networks that are active during porcine liver aging were constructed. These findings contribute to the understanding of the transcriptional foundations of liver aging and also provide further references that clarify human liver aging at the molecular level.
Collapse
Affiliation(s)
- Jianning Chen
- Department of Zoology, College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan, China
| | - Qin Zou
- Department of Zoology, College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan, China
| | - Daojun Lv
- Sichuan Weimu Modern Agricultural Science and Technology Co., Ltd, Chengdu, Sichuan, China
| | - Muhammad Ali Raza
- Department of Crop Cultivation and Farming System, College of Agronomy, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xue Wang
- Department of Zoology, College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan, China
| | - Yan Chen
- Department of Zoology, College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan, China
| | - Xiaoyu Xi
- Department of Zoology, College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan, China
| | - Peilin Li
- Sichuan Weimu Modern Agricultural Science and Technology Co., Ltd, Chengdu, Sichuan, China
| | - Anxiang Wen
- Department of Zoology, College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan, China
| | - Li Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Guoqing Tang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Mingzhou Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xuewei Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yanzhi Jiang
- Department of Zoology, College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan, China
| |
Collapse
|
31
|
Ling C, Rönn T. Epigenetics in Human Obesity and Type 2 Diabetes. Cell Metab 2019; 29:1028-1044. [PMID: 30982733 PMCID: PMC6509280 DOI: 10.1016/j.cmet.2019.03.009] [Citation(s) in RCA: 494] [Impact Index Per Article: 98.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/05/2019] [Accepted: 03/18/2019] [Indexed: 12/16/2022]
Abstract
Epigenetic mechanisms control gene activity and the development of an organism. The epigenome includes DNA methylation, histone modifications, and RNA-mediated processes, and disruption of this balance may cause several pathologies and contribute to obesity and type 2 diabetes (T2D). This Review summarizes epigenetic signatures obtained from human tissues of relevance for metabolism-i.e., adipose tissue, skeletal muscle, pancreatic islets, liver, and blood-in relation to obesity and T2D. Although this research field is still young, these comprehensive data support not only a role for epigenetics in disease development, but also epigenetic alterations as a response to disease. Genetic predisposition, as well as aging, contribute to epigenetic variability, and several environmental factors, including exercise and diet, further interact with the human epigenome. The reversible nature of epigenetic modifications holds promise for future therapeutic strategies in obesity and T2D.
Collapse
Affiliation(s)
- Charlotte Ling
- Epigenetics and Diabetes Unit, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden.
| | - Tina Rönn
- Epigenetics and Diabetes Unit, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| |
Collapse
|
32
|
Steiger H, Booij L, Kahan `E, McGregor K, Thaler L, Fletcher E, Labbe A, Joober R, Israël M, Szyf M, Agellon LB, Gauvin L, St-Hilaire A, Rossi E. A longitudinal, epigenome-wide study of DNA methylation in anorexia nervosa: results in actively ill, partially weight-restored, long-term remitted and non-eating-disordered women. J Psychiatry Neurosci 2019; 44:205-213. [PMID: 30693739 PMCID: PMC6488489 DOI: 10.1503/jpn.170242] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 05/30/2018] [Accepted: 09/04/2018] [Indexed: 12/12/2022] Open
Abstract
Background This study explored state-related tendencies in DNA methylation in people with anorexia nervosa. Methods We measured genome-wide DNA methylation in 75 women with active anorexia nervosa (active), 31 women showing stable remission of anorexia nervosa (remitted) and 41 women with no eating disorder (NED). We also obtained post-intervention methylation data from 52 of the women from the active group. Results Comparisons between members of the active and NED groups showed 58 differentially methylated sites (Q < 0.01) that corresponded to genes relevant to metabolic and nutritional status (lipid and glucose metabolism), psychiatric status (serotonin receptor activity) and immune function. Methylation levels in members of the remitted group differed from those in the active group on 265 probes that also involved sites associated with genes for serotonin and insulin activity, glucose metabolism and immunity. Intriguingly, the direction of methylation effects in remitted participants tended to be opposite to those seen in active participants. The chronicity of Illness correlated (usually inversely, at Q < 0.01) with methylation levels at 64 sites that mapped onto genes regulating glutamate and serotonin activity, insulin function and epigenetic age. In contrast, body mass index increases coincided (at Q < 0.05) with generally increased methylation-level changes at 73 probes associated with lipid and glucose metabolism, immune and inflammatory processes, and olfaction. Limitations Sample sizes were modest for this type of inquiry, and findings may have been subject to uncontrolled effects of medication and substance use. Conclusion Findings point to the possibility of reversible epigenetic alterations in anorexia nervosa, and suggest that an adequate pathophysiological model would likely need to include psychiatric, metabolic and immune components.
Collapse
Affiliation(s)
- Howard Steiger
- From the Eating Disorders Program, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Israël, St-Hilaire, Rossi); the Research Centre, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Joober, Israël, St-Hilaire, Rossi); the Department of Psychiatry, McGill University (Steiger, Booij, Thaler, Joober, Israël, St-Hilaire); the Department of Psychology, Concordia University (Booij); the Sainte-Justine Hospital Research Centre, University of Montreal (Booij); the Department of Epidemiology, Biostatistics, and Occupational Health, McGill University (McGregor); the Department of Decision Sciences, HEC Montreal (Labbe); the Department of Pharmacology and Therapeutics, McGill University (Szyf); the School of Human Nutrition, McGill University (Agellon); and the Centre de recherche du Centre Hospitalier, de l’Université de Montréal (CRCHUM) (Gauvin), Montreal, Que., Canada
| | - Linda Booij
- From the Eating Disorders Program, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Israël, St-Hilaire, Rossi); the Research Centre, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Joober, Israël, St-Hilaire, Rossi); the Department of Psychiatry, McGill University (Steiger, Booij, Thaler, Joober, Israël, St-Hilaire); the Department of Psychology, Concordia University (Booij); the Sainte-Justine Hospital Research Centre, University of Montreal (Booij); the Department of Epidemiology, Biostatistics, and Occupational Health, McGill University (McGregor); the Department of Decision Sciences, HEC Montreal (Labbe); the Department of Pharmacology and Therapeutics, McGill University (Szyf); the School of Human Nutrition, McGill University (Agellon); and the Centre de recherche du Centre Hospitalier, de l’Université de Montréal (CRCHUM) (Gauvin), Montreal, Que., Canada
| | - `Esther Kahan
- From the Eating Disorders Program, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Israël, St-Hilaire, Rossi); the Research Centre, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Joober, Israël, St-Hilaire, Rossi); the Department of Psychiatry, McGill University (Steiger, Booij, Thaler, Joober, Israël, St-Hilaire); the Department of Psychology, Concordia University (Booij); the Sainte-Justine Hospital Research Centre, University of Montreal (Booij); the Department of Epidemiology, Biostatistics, and Occupational Health, McGill University (McGregor); the Department of Decision Sciences, HEC Montreal (Labbe); the Department of Pharmacology and Therapeutics, McGill University (Szyf); the School of Human Nutrition, McGill University (Agellon); and the Centre de recherche du Centre Hospitalier, de l’Université de Montréal (CRCHUM) (Gauvin), Montreal, Que., Canada
| | - Kevin McGregor
- From the Eating Disorders Program, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Israël, St-Hilaire, Rossi); the Research Centre, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Joober, Israël, St-Hilaire, Rossi); the Department of Psychiatry, McGill University (Steiger, Booij, Thaler, Joober, Israël, St-Hilaire); the Department of Psychology, Concordia University (Booij); the Sainte-Justine Hospital Research Centre, University of Montreal (Booij); the Department of Epidemiology, Biostatistics, and Occupational Health, McGill University (McGregor); the Department of Decision Sciences, HEC Montreal (Labbe); the Department of Pharmacology and Therapeutics, McGill University (Szyf); the School of Human Nutrition, McGill University (Agellon); and the Centre de recherche du Centre Hospitalier, de l’Université de Montréal (CRCHUM) (Gauvin), Montreal, Que., Canada
| | - Lea Thaler
- From the Eating Disorders Program, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Israël, St-Hilaire, Rossi); the Research Centre, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Joober, Israël, St-Hilaire, Rossi); the Department of Psychiatry, McGill University (Steiger, Booij, Thaler, Joober, Israël, St-Hilaire); the Department of Psychology, Concordia University (Booij); the Sainte-Justine Hospital Research Centre, University of Montreal (Booij); the Department of Epidemiology, Biostatistics, and Occupational Health, McGill University (McGregor); the Department of Decision Sciences, HEC Montreal (Labbe); the Department of Pharmacology and Therapeutics, McGill University (Szyf); the School of Human Nutrition, McGill University (Agellon); and the Centre de recherche du Centre Hospitalier, de l’Université de Montréal (CRCHUM) (Gauvin), Montreal, Que., Canada
| | - Emilie Fletcher
- From the Eating Disorders Program, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Israël, St-Hilaire, Rossi); the Research Centre, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Joober, Israël, St-Hilaire, Rossi); the Department of Psychiatry, McGill University (Steiger, Booij, Thaler, Joober, Israël, St-Hilaire); the Department of Psychology, Concordia University (Booij); the Sainte-Justine Hospital Research Centre, University of Montreal (Booij); the Department of Epidemiology, Biostatistics, and Occupational Health, McGill University (McGregor); the Department of Decision Sciences, HEC Montreal (Labbe); the Department of Pharmacology and Therapeutics, McGill University (Szyf); the School of Human Nutrition, McGill University (Agellon); and the Centre de recherche du Centre Hospitalier, de l’Université de Montréal (CRCHUM) (Gauvin), Montreal, Que., Canada
| | - Aurelie Labbe
- From the Eating Disorders Program, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Israël, St-Hilaire, Rossi); the Research Centre, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Joober, Israël, St-Hilaire, Rossi); the Department of Psychiatry, McGill University (Steiger, Booij, Thaler, Joober, Israël, St-Hilaire); the Department of Psychology, Concordia University (Booij); the Sainte-Justine Hospital Research Centre, University of Montreal (Booij); the Department of Epidemiology, Biostatistics, and Occupational Health, McGill University (McGregor); the Department of Decision Sciences, HEC Montreal (Labbe); the Department of Pharmacology and Therapeutics, McGill University (Szyf); the School of Human Nutrition, McGill University (Agellon); and the Centre de recherche du Centre Hospitalier, de l’Université de Montréal (CRCHUM) (Gauvin), Montreal, Que., Canada
| | - Ridha Joober
- From the Eating Disorders Program, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Israël, St-Hilaire, Rossi); the Research Centre, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Joober, Israël, St-Hilaire, Rossi); the Department of Psychiatry, McGill University (Steiger, Booij, Thaler, Joober, Israël, St-Hilaire); the Department of Psychology, Concordia University (Booij); the Sainte-Justine Hospital Research Centre, University of Montreal (Booij); the Department of Epidemiology, Biostatistics, and Occupational Health, McGill University (McGregor); the Department of Decision Sciences, HEC Montreal (Labbe); the Department of Pharmacology and Therapeutics, McGill University (Szyf); the School of Human Nutrition, McGill University (Agellon); and the Centre de recherche du Centre Hospitalier, de l’Université de Montréal (CRCHUM) (Gauvin), Montreal, Que., Canada
| | - Mimi Israël
- From the Eating Disorders Program, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Israël, St-Hilaire, Rossi); the Research Centre, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Joober, Israël, St-Hilaire, Rossi); the Department of Psychiatry, McGill University (Steiger, Booij, Thaler, Joober, Israël, St-Hilaire); the Department of Psychology, Concordia University (Booij); the Sainte-Justine Hospital Research Centre, University of Montreal (Booij); the Department of Epidemiology, Biostatistics, and Occupational Health, McGill University (McGregor); the Department of Decision Sciences, HEC Montreal (Labbe); the Department of Pharmacology and Therapeutics, McGill University (Szyf); the School of Human Nutrition, McGill University (Agellon); and the Centre de recherche du Centre Hospitalier, de l’Université de Montréal (CRCHUM) (Gauvin), Montreal, Que., Canada
| | - Moshe Szyf
- From the Eating Disorders Program, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Israël, St-Hilaire, Rossi); the Research Centre, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Joober, Israël, St-Hilaire, Rossi); the Department of Psychiatry, McGill University (Steiger, Booij, Thaler, Joober, Israël, St-Hilaire); the Department of Psychology, Concordia University (Booij); the Sainte-Justine Hospital Research Centre, University of Montreal (Booij); the Department of Epidemiology, Biostatistics, and Occupational Health, McGill University (McGregor); the Department of Decision Sciences, HEC Montreal (Labbe); the Department of Pharmacology and Therapeutics, McGill University (Szyf); the School of Human Nutrition, McGill University (Agellon); and the Centre de recherche du Centre Hospitalier, de l’Université de Montréal (CRCHUM) (Gauvin), Montreal, Que., Canada
| | - Luis B. Agellon
- From the Eating Disorders Program, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Israël, St-Hilaire, Rossi); the Research Centre, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Joober, Israël, St-Hilaire, Rossi); the Department of Psychiatry, McGill University (Steiger, Booij, Thaler, Joober, Israël, St-Hilaire); the Department of Psychology, Concordia University (Booij); the Sainte-Justine Hospital Research Centre, University of Montreal (Booij); the Department of Epidemiology, Biostatistics, and Occupational Health, McGill University (McGregor); the Department of Decision Sciences, HEC Montreal (Labbe); the Department of Pharmacology and Therapeutics, McGill University (Szyf); the School of Human Nutrition, McGill University (Agellon); and the Centre de recherche du Centre Hospitalier, de l’Université de Montréal (CRCHUM) (Gauvin), Montreal, Que., Canada
| | - Lise Gauvin
- From the Eating Disorders Program, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Israël, St-Hilaire, Rossi); the Research Centre, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Joober, Israël, St-Hilaire, Rossi); the Department of Psychiatry, McGill University (Steiger, Booij, Thaler, Joober, Israël, St-Hilaire); the Department of Psychology, Concordia University (Booij); the Sainte-Justine Hospital Research Centre, University of Montreal (Booij); the Department of Epidemiology, Biostatistics, and Occupational Health, McGill University (McGregor); the Department of Decision Sciences, HEC Montreal (Labbe); the Department of Pharmacology and Therapeutics, McGill University (Szyf); the School of Human Nutrition, McGill University (Agellon); and the Centre de recherche du Centre Hospitalier, de l’Université de Montréal (CRCHUM) (Gauvin), Montreal, Que., Canada
| | - Annie St-Hilaire
- From the Eating Disorders Program, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Israël, St-Hilaire, Rossi); the Research Centre, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Joober, Israël, St-Hilaire, Rossi); the Department of Psychiatry, McGill University (Steiger, Booij, Thaler, Joober, Israël, St-Hilaire); the Department of Psychology, Concordia University (Booij); the Sainte-Justine Hospital Research Centre, University of Montreal (Booij); the Department of Epidemiology, Biostatistics, and Occupational Health, McGill University (McGregor); the Department of Decision Sciences, HEC Montreal (Labbe); the Department of Pharmacology and Therapeutics, McGill University (Szyf); the School of Human Nutrition, McGill University (Agellon); and the Centre de recherche du Centre Hospitalier, de l’Université de Montréal (CRCHUM) (Gauvin), Montreal, Que., Canada
| | - Erika Rossi
- From the Eating Disorders Program, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Israël, St-Hilaire, Rossi); the Research Centre, Douglas University Institute (Steiger, Kahan, Thaler, Fletcher, Joober, Israël, St-Hilaire, Rossi); the Department of Psychiatry, McGill University (Steiger, Booij, Thaler, Joober, Israël, St-Hilaire); the Department of Psychology, Concordia University (Booij); the Sainte-Justine Hospital Research Centre, University of Montreal (Booij); the Department of Epidemiology, Biostatistics, and Occupational Health, McGill University (McGregor); the Department of Decision Sciences, HEC Montreal (Labbe); the Department of Pharmacology and Therapeutics, McGill University (Szyf); the School of Human Nutrition, McGill University (Agellon); and the Centre de recherche du Centre Hospitalier, de l’Université de Montréal (CRCHUM) (Gauvin), Montreal, Que., Canada
| |
Collapse
|
33
|
Hack LM, Fries GR, Eyre HA, Bousman CA, Singh AB, Quevedo J, John VP, Baune BT, Dunlop BW. Moving pharmacoepigenetics tools for depression toward clinical use. J Affect Disord 2019; 249:336-346. [PMID: 30802699 PMCID: PMC6763314 DOI: 10.1016/j.jad.2019.02.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/01/2019] [Accepted: 02/05/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Major depressive disorder (MDD) is a leading cause of disability worldwide, and over half of patients do not achieve symptom remission following an initial antidepressant course. Despite evidence implicating a strong genetic basis for the pathophysiology of MDD, there are no adequately validated biomarkers of treatment response routinely used in clinical practice. Pharmacoepigenetics is an emerging field that has the potential to combine both genetic and environmental information into treatment selection and further the goal of precision psychiatry. However, this field is in its infancy compared to the more established pharmacogenetics approaches. METHODS We prepared a narrative review using literature searches of studies in English pertaining to pharmacoepigenetics and treatment of depressive disorders conducted in PubMed, Google Scholar, PsychINFO, and Ovid Medicine from inception through January 2019. We reviewed studies of DNA methylation and histone modifications in both humans and animal models of depression. RESULTS Emerging evidence from human and animal work suggests a key role for epigenetic marks, including DNA methylation and histone modifications, in the prediction of antidepressant response. The challenges of heterogeneity of patient characteristics and loci studied as well as lack of replication that have impacted the field of pharmacogenetics also pose challenges to the development of pharmacoepigenetic tools. Additionally, given the tissue specific nature of epigenetic marks as well as their susceptibility to change in response to environmental factors and aging, pharmacoepigenetic tools face additional challenges to their development. LIMITATIONS This is a narrative and not systematic review of the literature on the pharmacoepigenetics of antidepressant response. We highlight key studies pertaining to pharmacoepigenetics and treatment of depressive disorders in humans and depressive-like behaviors in animal models, regardless of sample size or methodology. While we discuss DNA methylation and histone modifications, we do not cover microRNAs, which have been reviewed elsewhere recently. CONCLUSIONS Utilization of genome-wide approaches and reproducible epigenetic assays, careful selection of the tissue assessed, and integration of genetic and clinical information into pharmacoepigenetic tools will improve the likelihood of developing clinically useful tests.
Collapse
Affiliation(s)
- Laura M Hack
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Emory University, Atlanta, GA, USA; Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 401 Quarry Road, Palo Alto, CA 94305, USA; Sierra Pacific Mental Illness Research Education and Clinical Centers, VA Palo Alto Health Care System, Palo Alto, CA, USA.
| | - Gabriel R Fries
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Harris A Eyre
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 401 Quarry Road, Palo Alto, CA 94305, USA; Innovation Institute, Texas Medical Center, Houston, TX, USA; IMPACT SRC, School of Medicine, Deakin University, Geelong, Victoria, Australia; Department of Psychiatry, University of Melbourne, Melbourne, Victoria, Australia
| | - Chad A Bousman
- Departments of Medical Genetics, Psychiatry, Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Ajeet B Singh
- IMPACT SRC, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Joao Quevedo
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Vineeth P John
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Bernhard T Baune
- Department of Psychiatry, University of Melbourne, Melbourne, Victoria, Australia
| | - Boadie W Dunlop
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Emory University, Atlanta, GA, USA
| |
Collapse
|
34
|
Perera BPU, Svoboda L, Dolinoy DC. Genomic Tools for Environmental Epigenetics and Implications for Public Health. CURRENT OPINION IN TOXICOLOGY 2019; 18:27-33. [PMID: 31763499 DOI: 10.1016/j.cotox.2019.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Epigenetics refers to the study of mitotically heritable and potentially reversible changes in gene expression unrelated to the DNA sequence itself, influenced by epigenetic marks including chromatin modifications, non-coding RNA and alterations to DNA itself via methylation and hydroxymethylation. Epigenetics has taken center stage in the study of diseases such as cancer, diabetes, and neurodegeneration; however, its integration into the field of environmental health sciences and toxicology (e.g. Toxicoepigenetics) is in its infancy. This review highlights the need to evaluate surrogate and target tissues in the field of toxicoepigenetics as the National Institute of Environmental Health Sciences (NIEHS) multi-phased Toxicant Exposure and Response by Genomic and Epigenomic Regulators of Transcription (TaRGET) consortia make headway, and the emergence of non-coding RNA biomarkers. The review also discusses lead (Pb) as a potential toxicoepigenetic exposure, where pre- and post-natal Pb exposure is associated with reprogramming of DNA methylation, histone modifications, and microRNA expression, representing potential biomarkers or predictors for Pb-induced health outcomes. Finally, new advances in epigenome editing, highlighting the potential of small ncRNA, will be explored for environmental health sciences research.
Collapse
Affiliation(s)
- Bambarendage P U Perera
- University of Michigan School of Public Health, Department of Environmental Health Sciences, Ann Arbor, MI
| | - Laurie Svoboda
- University of Michigan School of Public Health, Department of Environmental Health Sciences, Ann Arbor, MI
| | - Dana C Dolinoy
- University of Michigan School of Public Health, Department of Environmental Health Sciences, Ann Arbor, MI
- University of Michigan School of Public Health, Department of Nutritional Sciences, Ann Arbor, MI
| |
Collapse
|
35
|
Ramos-Lopez O, Riezu-Boj JI, Milagro FI, Alfredo Martinez J. Association of Methylation Signatures at Hepatocellular Carcinoma Pathway Genes with Adiposity and Insulin Resistance Phenotypes. Nutr Cancer 2018; 71:840-851. [PMID: 30457363 DOI: 10.1080/01635581.2018.1531136] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Omar Ramos-Lopez
- Department of Nutrition, Food Science and Physiology, and Center for Nutrition Research, University of Navarra, Pamplona, Spain
| | - Jose I. Riezu-Boj
- Department of Nutrition, Food Science and Physiology, and Center for Nutrition Research, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Fermin I. Milagro
- Department of Nutrition, Food Science and Physiology, and Center for Nutrition Research, University of Navarra, Pamplona, Spain
- CIBERobn, Fisiopatología de la Obesidad y la Nutrición, Carlos III Health Institute, Madrid, Spain
| | - J. Alfredo Martinez
- Department of Nutrition, Food Science and Physiology, and Center for Nutrition Research, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- CIBERobn, Fisiopatología de la Obesidad y la Nutrición, Carlos III Health Institute, Madrid, Spain
- Madrid Institute of Advanced Studies (IMDEA Food), Madrid, Spain
| | | |
Collapse
|
36
|
Abstract
Recent research suggests that epigenetics, especially DNA methylation, plays a mechanistic role in aging. Epigenetic clocks, which measure changes in a few hundred specific CpG sites, can accurately predict chronological age in a variety of species, including humans. These clocks are currently the bestbiomarkers for predicting mortality in humans. Additionally, several studies have characterized the effects of aging across the methylome in a wide variety of tissues from humans and mice. A small fraction (~2%) of the CpG sites show age-related changes, either hypermethylation or hypomethylation with aging. Evaluation of non-CpG site methylation has only been examined in a few studies, with about ~0.5% of these sites showing achange with age. Therefore, while only a small fraction of cytosines in the genome show changes in DNA methylation with age, this represents 2 to 3 million cytosines in the genome. Importantly, the only study to compare the effect of aging on DNA methylation in male and female mice and humans found that N95% of the age-related changes in DNA methylation in the hippocampus were sexually divergent, i.e., the methylation did not differ between males and females atyoung age but age-related changes occurred in one sex but not the other. The age-related changes in DNA methylation tend to be enriched and under-represented in specific genomic contexts, with some commonalities between tissues and species that require further investigation. The strongest evidence that the age-related changes in DNA methylation play a role in aging comes from studies of anti-aging interventions (e.g., caloric restriction, dwarfism, and rapamycin treatment) in mice. These anti-aging interventions deaccelerate the epigenetic clocks and reverse/prevent 20 to 40% of the age-related changes in DNA methylation. It will be important in the future to demonstrate that at least some of the age-related changes in DNA methylation directly lead to alterations in the transcriptome of cells/tissues that could potentially contribute to aging.
Collapse
|
37
|
DNA methylation dynamics in aging: how far are we from understanding the mechanisms? Mech Ageing Dev 2018; 174:3-17. [DOI: 10.1016/j.mad.2017.12.002] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/14/2017] [Accepted: 12/16/2017] [Indexed: 02/07/2023]
|
38
|
Najar RA, Wani NA, Bhat JA, Dar NJ, Rahat B, Gupta AP, Kaur J, Kaur J, Hamid A. Modulation of dietary folate with age confers selective hepatocellular epigenetic imprints through DNA methylation. J Nutr Biochem 2018; 53:121-132. [PMID: 29220669 DOI: 10.1016/j.jnutbio.2017.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/15/2017] [Accepted: 10/16/2017] [Indexed: 02/03/2023]
Abstract
The present study has been designed to determine the effect of folate modulation (deficiency/supplementation) with aging on the promoter methylation of tumor suppressor and proto-oncogenes to understand the underlying mechanism of epigenetic alterations. Folate deficiency was induced for 3 and 5 months in weanling, young and adult groups, and after 3 months of folate deficiency, they were repleted with physiological folate (2 mg/kg diet) and folate oversupplementation (8 mg/kg diet) for another 2 months. The methylation facet in the present study revealed that the combined effect of folate deficiency and aging decreased the methylation index. Folate deficiency with age resulted in the up-regulation of proto-oncogenes (C-MYC and C-JUN) and cell cycle regulator gene Cyclin E as a result of promoter hypomethylation. However, in case of tumor suppressor genes (p53, p15ink4b and p16ink4a), the expression levels were found to be decreased at transcriptional level due to promoter hypermethylation. Upon repletion with physiological folate and folate oversupplementation, we found down-regulation of proto-oncogenes and up-regulation of tumor suppressor genes as a result of promoter hypermethylation and hypomethylation, respectively. Deregulation of these important genes due to folate deficiency may contribute toward the pathogenesis at cellular level.
Collapse
Affiliation(s)
- Rauf Ahmad Najar
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Nissar Ahmad Wani
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Javeed Ahmad Bhat
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Nawab John Dar
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Beenish Rahat
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Ajai Prakash Gupta
- Quality Control and Quality Assurance Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Jaspreet Kaur
- University Institute of Engineering and Technology, Panjab University, Chandigarh 160016, India
| | - Jyotdeep Kaur
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Abid Hamid
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India.
| |
Collapse
|
39
|
Davegårdh C, García-Calzón S, Bacos K, Ling C. DNA methylation in the pathogenesis of type 2 diabetes in humans. Mol Metab 2018; 14:12-25. [PMID: 29496428 PMCID: PMC6034041 DOI: 10.1016/j.molmet.2018.01.022] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 01/29/2018] [Accepted: 01/29/2018] [Indexed: 02/08/2023] Open
Abstract
Background Type 2 diabetes (T2D) is a multifactorial, polygenic disease caused by impaired insulin secretion and insulin resistance. Genome-wide association studies (GWAS) were expected to resolve a large part of the genetic component of diabetes; yet, the single nucleotide polymorphisms identified by GWAS explain less than 20% of the estimated heritability for T2D. There was subsequently a need to look elsewhere to find disease-causing factors. Mechanisms mediating the interaction between environmental factors and the genome, such as epigenetics, may be of particular importance in the pathogenesis of T2D. Scope of Review This review summarizes knowledge of the impact of epigenetics on the pathogenesis of T2D in humans. In particular, the review will focus on alterations in DNA methylation in four human tissues of importance for the disease; pancreatic islets, skeletal muscle, adipose tissue, and the liver. Case–control studies and studies examining the impact of non-genetic and genetic risk factors on DNA methylation in humans will be considered. These studies identified epigenetic changes in tissues from subjects with T2D versus non-diabetic controls. They also demonstrate that non-genetic factors associated with T2D such as age, obesity, energy rich diets, physical activity and the intrauterine environment impact the epigenome in humans. Additionally, interactions between genetics and epigenetics seem to influence the pathogenesis of T2D. Conclusions Overall, previous studies by our group and others support a key role for epigenetics in the growing incidence of T2D.
Collapse
Affiliation(s)
- Cajsa Davegårdh
- Epigenetics and Diabetes, Lund University Diabetes Centre (LUDC), Box 50332, 20213 Malmö, Sweden.
| | - Sonia García-Calzón
- Epigenetics and Diabetes, Lund University Diabetes Centre (LUDC), Box 50332, 20213 Malmö, Sweden
| | - Karl Bacos
- Epigenetics and Diabetes, Lund University Diabetes Centre (LUDC), Box 50332, 20213 Malmö, Sweden
| | - Charlotte Ling
- Epigenetics and Diabetes, Lund University Diabetes Centre (LUDC), Box 50332, 20213 Malmö, Sweden
| |
Collapse
|
40
|
Cheng Z, Zheng L, Almeida FA. Epigenetic reprogramming in metabolic disorders: nutritional factors and beyond. J Nutr Biochem 2017; 54:1-10. [PMID: 29154162 DOI: 10.1016/j.jnutbio.2017.10.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 09/26/2017] [Accepted: 10/10/2017] [Indexed: 12/13/2022]
Abstract
Environmental factors (e.g., malnutrition and physical inactivity) contribute largely to metabolic disorders including obesity, type 2 diabetes, cardiometabolic disease and nonalcoholic fatty liver diseases. The abnormalities in metabolic activity and pathways have been increasingly associated with altered DNA methylation, histone modification and noncoding RNAs, whereas lifestyle interventions targeting diet and physical activity can reverse the epigenetic and metabolic changes. Here we review recent evidence primarily from human studies that links DNA methylation reprogramming to metabolic derangements or improvements, with a focus on cross-tissue (e.g., the liver, skeletal muscle, pancreas, adipose tissue and blood samples) epigenetic markers, mechanistic mediators of the epigenetic reprogramming, and the potential of using epigenetic traits to predict disease risk and intervention response. The challenges in epigenetic studies addressing the mechanisms of metabolic diseases and future directions are also discussed and prospected.
Collapse
Affiliation(s)
- Zhiyong Cheng
- Department of Human Nutrition, Foods, and Exercise, Fralin Translational Obesity Research Center, College of Agriculture and Life Science, Virginia Tech, Blacksburg, VA 24061, USA.
| | - Louise Zheng
- Department of Human Nutrition, Foods, and Exercise, Fralin Translational Obesity Research Center, College of Agriculture and Life Science, Virginia Tech, Blacksburg, VA 24061, USA
| | - Fabio A Almeida
- Department of Health Promotion, Social & Behavioral Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
41
|
Anzai Á, Marcondes RR, Gonçalves TH, Carvalho KC, Simões MJ, Garcia N, Soares JM, Padmanabhan V, Baracat EC, da Silva IDCG, Maciel GAR. Impaired branched-chain amino acid metabolism may underlie the nonalcoholic fatty liver disease-like pathology of neonatal testosterone-treated female rats. Sci Rep 2017; 7:13167. [PMID: 29030588 PMCID: PMC5640623 DOI: 10.1038/s41598-017-13451-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/20/2017] [Indexed: 12/25/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is frequently associated with non-alcoholic fatty liver disease (NAFLD), but the mechanisms involved in the development of NAFLD in PCOS are not well known. We investigated histological changes and metabolomic profile in the liver of rat models of PCOS phenotype induced by testosterone or estradiol. Two-day old female rats received sc injections of 1.25 mg testosterone propionate (Testos; n = 10), 0.5 mg estradiol benzoate (E2; n = 10), or vehicle (control group, CNT; n = 10). Animals were euthanized at 90-94 d of age and the liver was harvested for histological and metabolomic analyses. Findings showed only Testos group exhibited fatty liver morphology and higher levels of ketogenic and branched-chain amino acids (BCAA). Enrichment analysis showed effects of testosterone on BCAA degradation pathway and mitochondrial enzymes related to BCAA metabolism. Testos group also had a decreased liver fatty acid elongase 2 (ELOVL2) activity. E2 group had reduced lipid and acylcarnitine metabolites in the liver. Both groups had increased organic cation transporters (SLC22A4 and SLC16A9) activity. These findings indicate that neonatal testosterone treatment, but not estradiol, produces histological changes in female rat liver that mimic NAFLD with testosterone-treated rats showing impaired BCAA metabolism and dysfunctions in ELOVL2, SLC22A4 and SLC16A9 activity.
Collapse
Affiliation(s)
- Álvaro Anzai
- Laboratorio de Ginecologia Estrutural e Molecular (LIM 58), Disciplina de Ginecologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246903, Brazil
| | - Rodrigo R Marcondes
- Laboratorio de Ginecologia Estrutural e Molecular (LIM 58), Disciplina de Ginecologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246903, Brazil.
| | - Thiago H Gonçalves
- Laboratorio de Ginecologia Estrutural e Molecular (LIM 58), Disciplina de Ginecologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246903, Brazil
| | - Kátia C Carvalho
- Laboratorio de Ginecologia Estrutural e Molecular (LIM 58), Disciplina de Ginecologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246903, Brazil
| | - Manuel J Simões
- Departamento de Morfologia e Genetica, Disciplina de Histologia e Biologia Estrutural, Universidade Federal de Sao Paulo, Sao Paulo, SP, 04023900, Brazil
| | - Natália Garcia
- Laboratorio de Ginecologia Estrutural e Molecular (LIM 58), Disciplina de Ginecologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246903, Brazil
| | - José M Soares
- Laboratorio de Ginecologia Estrutural e Molecular (LIM 58), Disciplina de Ginecologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246903, Brazil
| | - Vasantha Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Edmund C Baracat
- Laboratorio de Ginecologia Estrutural e Molecular (LIM 58), Disciplina de Ginecologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246903, Brazil
| | - Ismael D C G da Silva
- Laboratorio de Ginecologia Molecular e Proteomica, Departamento de Ginecologia, Universidade Federal de Sao Paulo, Sao Paulo, SP, 04024002, Brazil
| | - Gustavo A R Maciel
- Laboratorio de Ginecologia Estrutural e Molecular (LIM 58), Disciplina de Ginecologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, 01246903, Brazil.
| |
Collapse
|
42
|
Nilsson E, Ling C. DNA methylation links genetics, fetal environment, and an unhealthy lifestyle to the development of type 2 diabetes. Clin Epigenetics 2017; 9:105. [PMID: 29026446 PMCID: PMC5627472 DOI: 10.1186/s13148-017-0399-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/29/2017] [Indexed: 01/09/2023] Open
Abstract
Type 2 diabetes is a complex trait with both environmental and hereditary factors contributing to the overall pathogenesis. One link between genes, environment, and disease is epigenetics influencing gene transcription and, consequently, organ function. Genome-wide studies have shown altered DNA methylation in tissues important for glucose homeostasis including pancreas, liver, skeletal muscle, and adipose tissue from subjects with type 2 diabetes compared with nondiabetic controls. Factors predisposing for type 2 diabetes including an adverse intrauterine environment, increasing age, overweight, physical inactivity, a family history of the disease, and an unhealthy diet have all shown to affect the DNA methylation pattern in target tissues for insulin resistance in humans. Epigenetics including DNA methylation may therefore improve our understanding of the type 2 diabetes pathogenesis, contribute to development of novel treatments, and be a useful tool to identify individuals at risk for developing the disease.
Collapse
Affiliation(s)
- Emma Nilsson
- Department of Clinical Sciences, Epigenetics and Diabetes Unit, Lund University Diabetes Centre, Scania University Hospital, Jan Waldenströms gata 35, 205 02 Malmö, Sweden
| | - Charlotte Ling
- Department of Clinical Sciences, Epigenetics and Diabetes Unit, Lund University Diabetes Centre, Scania University Hospital, Jan Waldenströms gata 35, 205 02 Malmö, Sweden
| |
Collapse
|
43
|
Gilbert KM, Blossom SJ, Reisfeld B, Erickson SW, Vyas K, Maher M, Broadfoot B, West K, Bai S, Cooney CA, Bhattacharyya S. Trichloroethylene-induced alterations in DNA methylation were enriched in polycomb protein binding sites in effector/memory CD4 + T cells. ENVIRONMENTAL EPIGENETICS 2017; 3:dvx013. [PMID: 29129997 PMCID: PMC5676456 DOI: 10.1093/eep/dvx013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 06/30/2017] [Accepted: 07/03/2017] [Indexed: 06/07/2023]
Abstract
Exposure to industrial solvent and water pollutant trichloroethylene (TCE) can promote autoimmunity, and expand effector/memory (CD62L) CD4+ T cells. In order to better understand etiology reduced representation bisulfite sequencing was used to study how a 40-week exposure to TCE in drinking water altered methylation of ∼337 770 CpG sites across the entire genome of effector/memory CD4+ T cells from MRL+/+ mice. Regardless of TCE exposure, 62% of CpG sites in autosomal chromosomes were hypomethylated (0-15% methylation), and 25% were hypermethylated (85-100% methylation). In contrast, only 6% of the CpGs on the X chromosome were hypomethylated, and 51% had mid-range methylation levels. In terms of TCE impact, TCE altered (≥ 10%) the methylation of 233 CpG sites in effector/memory CD4+ T cells. Approximately 31.7% of these differentially methylated sites occurred in regions known to bind one or more Polycomb group (PcG) proteins, namely Ezh2, Suz12, Mtf2 or Jarid2. In comparison, only 23.3% of CpG sites not differentially methylated by TCE were found in PcG protein binding regions. Transcriptomics revealed that TCE altered the expression of ∼560 genes in the same effector/memory CD4+ T cells. At least 80% of the immune genes altered by TCE had binding sites for PcG proteins flanking their transcription start site, or were regulated by other transcription factors that were in turn ordered by PcG proteins at their own transcription start site. Thus, PcG proteins, and the differential methylation of their binding sites, may represent a new mechanism by which TCE could alter the function of effector/memory CD4+ T cells.
Collapse
Affiliation(s)
- Kathleen M. Gilbert
- Arkansas Children’s Research Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Sarah J. Blossom
- Arkansas Children’s Research Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Brad Reisfeld
- Colorado State University, Fort Collins, CO 80523, USA
| | - Stephen W. Erickson
- Arkansas Children’s Research Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Kanan Vyas
- Arkansas Children’s Research Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Mary Maher
- Arkansas Children’s Research Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Brannon Broadfoot
- Arkansas Children’s Research Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Kirk West
- Arkansas Children’s Research Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Shasha Bai
- Arkansas Children’s Research Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Craig A. Cooney
- Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Sudeepa Bhattacharyya
- Arkansas Children’s Research Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| |
Collapse
|