1
|
Liang J, Yang F, Li Z, Li Q. Epigenetic regulation of the inflammatory response in stroke. Neural Regen Res 2025; 20:3045-3062. [PMID: 39589183 PMCID: PMC11881735 DOI: 10.4103/nrr.nrr-d-24-00672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/15/2024] [Accepted: 09/20/2024] [Indexed: 11/27/2024] Open
Abstract
Stroke is classified as ischemic or hemorrhagic, and there are few effective treatments for either type. Immunologic mechanisms play a critical role in secondary brain injury following a stroke, which manifests as cytokine release, blood-brain barrier disruption, neuronal cell death, and ultimately behavioral impairment. Suppressing the inflammatory response has been shown to mitigate this cascade of events in experimental stroke models. However, in clinical trials of anti-inflammatory agents, long-term immunosuppression has not demonstrated significant clinical benefits for patients. This may be attributable to the dichotomous roles of inflammation in both tissue injury and repair, as well as the complex pathophysiologic inflammatory processes in stroke. Inhibiting acute harmful inflammatory responses or inducing a phenotypic shift from a pro-inflammatory to an anti-inflammatory state at specific time points after a stroke are alternative and promising therapeutic strategies. Identifying agents that can modulate inflammation requires a detailed understanding of the inflammatory processes of stroke. Furthermore, epigenetic reprogramming plays a crucial role in modulating post-stroke inflammation and can potentially be exploited for stroke management. In this review, we summarize current findings on the epigenetic regulation of the inflammatory response in stroke, focusing on key signaling pathways including nuclear factor-kappa B, Janus kinase/signal transducer and activator of transcription, and mitogen-activated protein kinase as well as inflammasome activation. We also discuss promising molecular targets for stroke treatment. The evidence to date indicates that therapeutic targeting of the epigenetic regulation of inflammation can shift the balance from inflammation-induced tissue injury to repair following stroke, leading to improved post-stroke outcomes.
Collapse
Affiliation(s)
- Jingyi Liang
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Fei Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Zixiao Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Center for Healthcare Quality Management in Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Engineering Research Center of Digital Healthcare for Neurological Diseases, Beijing, China
| | - Qian Li
- Laboratory for Clinical Medicine, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Mariner BL, McCoy BM, Greenier A, Brassington L, Slikas E, Adjangba C, Marye A, Harrison BR, Bamberger T, Algavi Y, Muller E, Harris A, Rout E, Avery A, Borenstein E, Promislow D, Snyder-Mackler N. DNA methylation of transposons pattern aging differences across a diverse cohort of dogs from the Dog Aging Project. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.08.617286. [PMID: 39416178 PMCID: PMC11482827 DOI: 10.1101/2024.10.08.617286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Within a species, larger individuals often have shorter lives and higher rates of age-related disease. Despite this well-known link, we still know little about underlying age-related epigenetic differences, which could help us better understand inter-individual variation in aging and the etiology, onset, and progression of age-associated disease. Dogs exhibit this negative correlation between size, health, and longevity and thus represent an excellent system in which to test the underlying mechanisms. Here, we quantified genome-wide DNA methylation in a cohort of 864 dogs in the Dog Aging Project. Age strongly patterned the dog epigenome, with the majority (66% of age-associated loci) of regions associating age-related loss of methylation. These age effects were non-randomly distributed in the genome and differed depending on genomic context. We found the LINE1 (long interspersed elements) class of TEs (transposable elements) were the most frequently hypomethylated with age (FDR < 0.05, 40% of all LINE1 regions). This LINE1 pattern differed in magnitude across breeds of different sizes- the largest dogs lost 0.26% more LINE1 methylation per year than the smallest dogs. This suggests that epigenetic regulation of TEs, particularly LINE1s, may contribute to accelerated age and disease phenotypes within a species. Since our study focused on the methylome of immune cells, we looked at LINE1 methylation changes in golden retrievers, a breed highly susceptible to hematopoietic cancers, and found they have accelerated age-related LINE1 hypomethylation compared to other breeds. We also found many of the LINE1s hypomethylated with age are located on the X chromosome and are, when considering X chromosome inactivation, counter-intuitively more methylated in males. These results have revealed the demethylation of LINE1 transposons as a potential driver of intra-species, demographic-dependent aging variation.
Collapse
|
3
|
Rroji O, Mucignat C. Factors influencing brain recovery from stroke via possible epigenetic changes. Future Sci OA 2024; 10:2409609. [PMID: 39429231 PMCID: PMC11497982 DOI: 10.1080/20565623.2024.2409609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 09/24/2024] [Indexed: 10/22/2024] Open
Abstract
Aim: To examine epigenetic changes leading to functional repair after damage to the central motor system.Data sources: A literature search was conducted using medical and health science electronic databases (PubMed, MEDLINE, Scopus) up to July 2023.Study selection: Data were summarized for type of intervention, study design, findings including human and animal studies.Data extraction: Data were extracted and double-checked independently for methodological quality. By means of the influence of environmental (calorie restriction or physical exercise) and other factors, epigenetic instructions were found to increase levels of BDNF and enhance synaptic neurotransmission, possibly leading to larger scale changes in structural and functional assets, which may end up to cognitive and motor repair after stroke.
Collapse
Affiliation(s)
- Orjon Rroji
- Department of Radiology & Imaging techniques, European University of Tirana, Albania
| | - Carla Mucignat
- Department of Molecular Medicine, University of Padova, Italy
- National Institute for Biostructures & Biosystems, Rome, Italy
| |
Collapse
|
4
|
Mehta SL, Arruri V, Vemuganti R. Role of transcription factors, noncoding RNAs, epitranscriptomics, and epigenetics in post-ischemic neuroinflammation. J Neurochem 2024; 168:3430-3448. [PMID: 38279529 PMCID: PMC11272908 DOI: 10.1111/jnc.16055] [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: 08/26/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/28/2024]
Abstract
Post-stroke neuroinflammation is pivotal in brain repair, yet persistent inflammation can aggravate ischemic brain damage and hamper recovery. Following stroke, specific molecules released from brain cells attract and activate central and peripheral immune cells. These immune cells subsequently release diverse inflammatory molecules within the ischemic brain, initiating a sequence of events, including activation of transcription factors in different brain cell types that modulate gene expression and influence outcomes; the interactive action of various noncoding RNAs (ncRNAs) to regulate multiple biological processes including inflammation, epitranscriptomic RNA modification that controls RNA processing, stability, and translation; and epigenetic changes including DNA methylation, hydroxymethylation, and histone modifications crucial in managing the genic response to stroke. Interactions among these events further affect post-stroke inflammation and shape the depth of ischemic brain damage and functional outcomes. We highlighted these aspects of neuroinflammation in this review and postulate that deciphering these mechanisms is pivotal for identifying therapeutic targets to alleviate post-stroke dysfunction and enhance recovery.
Collapse
Affiliation(s)
- Suresh L. Mehta
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Vijay Arruri
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- William S. Middleton Veterans Hospital, Madison, WI, USA
| |
Collapse
|
5
|
Kennedy K, Jusue-Torres I, Buller ID, Rossi E, Mallisetty A, Rodgers K, Lee B, Menchaca M, Pasquinelli M, Nguyen RH, Weinberg F, Rubinstein I, Herman JG, Brock M, Feldman L, Aldrich MC, Hulbert A. Neighborhood-level deprivation and survival in lung cancer. BMC Cancer 2024; 24:959. [PMID: 39107707 PMCID: PMC11301857 DOI: 10.1186/s12885-024-12720-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Despite recent advances in lung cancer therapeutics and improving overall survival, disparities persist among socially disadvantaged populations. This study aims to determine the effects of neighborhood deprivation indices (NDI) on lung cancer mortality. This is a multicenter retrospective cohort study assessing the relationship between NDI and overall survival adjusted for age, disease stage, and DNA methylation among biopsy-proven lung cancer patients. State-specific NDI for each year of sample collection were computed at the U.S. census tract level and dichotomized into low- and high-deprivation. RESULTS A total of 173 non small lung cancer patients were included, with n = 85 (49%) and n = 88 (51%) in the low and high-deprivation groups, respectively. NDI was significantly higher among Black patients when compared with White patients (p = 0.003). There was a significant correlation between DNA methylation and stage for HOXA7, SOX17, ZFP42, HOXA9, CDO1 and TAC1. Only HOXA7 DNA methylation was positively correlated with NDI. The high-deprivation group had a statistically significant shorter survival than the low-deprivation group (p = 0.02). After adjusting for age, race, stage, and DNA methylation status, belonging to the high-deprivation group was associated with higher mortality with a hazard ratio of 1.81 (95%CI: 1.03-3.19). CONCLUSIONS Increased neighborhood-level deprivation may be associated with liquid biopsy DNA methylation, shorter survival, and increased mortality. Changes in health care policies that consider neighborhood-level indices of socioeconomic deprivation may enable a more equitable increase in lung cancer survival.
Collapse
Affiliation(s)
- Kathleen Kennedy
- Department of Hematology Oncology, University of Illinois College of Medicine in Chicago, Chicago, IL, USA
| | - Ignacio Jusue-Torres
- Department of Neurologic Surgery, Mayo Clinic Health System, Eau Claire, WI, USA
| | - Ian D Buller
- Cancer Prevention Fellowship Program, Division of Cancer Prevention, National Cancer Institute, Rockville, MD, USA
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Emily Rossi
- Cancer Prevention Fellowship Program, Division of Cancer Prevention, National Cancer Institute, Rockville, MD, USA
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Apurva Mallisetty
- Department of Surgery, Cancer Center, University of Illinois College of Medicine in Chicago, 909 South Wolcott Ave COMRB Suite 5140, Chicago, IL, 60612, USA
| | - Kristen Rodgers
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Beverly Lee
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Martha Menchaca
- Department of Radiology, University of Illinois College of Medicine in Chicago, Chicago, IL, USA
| | - Mary Pasquinelli
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois College of Medicine in Chicago, Chicago, IL, USA
| | - Ryan H Nguyen
- Department of Hematology Oncology, University of Illinois College of Medicine in Chicago, Chicago, IL, USA
| | - Frank Weinberg
- Department of Hematology Oncology, University of Illinois College of Medicine in Chicago, Chicago, IL, USA
| | - Israel Rubinstein
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois College of Medicine in Chicago, Chicago, IL, USA
- Division of Research Services, Jesse Brown VA Medical Center, Chicago, IL, USA
| | - James G Herman
- Lung Cancer Program, University of Pittsburgh Cancer Institute, The Hillman Cancer Center, Pittsburgh, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Malcolm Brock
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Lawrence Feldman
- Department of Hematology Oncology, University of Illinois College of Medicine in Chicago, Chicago, IL, USA
- Division of Research Services, Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Melinda C Aldrich
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alicia Hulbert
- Department of Surgery, Cancer Center, University of Illinois College of Medicine in Chicago, 909 South Wolcott Ave COMRB Suite 5140, Chicago, IL, 60612, USA.
- Division of Research Services, Jesse Brown VA Medical Center, Chicago, IL, USA.
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.
| |
Collapse
|
6
|
Zhang L, Guo J, Liu Y, Sun S, Liu B, Yang Q, Tao J, Tian XL, Pu J, Hong H, Wang M, Chen HZ, Ren J, Wang X, Liang Z, Wang Y, Huang K, Zhang W, Qu J, Ju Z, Liu GH, Pei G, Li J, Zhang C. A framework of biomarkers for vascular aging: a consensus statement by the Aging Biomarker Consortium. LIFE MEDICINE 2023; 2:lnad033. [PMID: 40040784 PMCID: PMC11879419 DOI: 10.1093/lifemedi/lnad033] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 08/25/2023] [Indexed: 03/06/2025]
Abstract
Aging of the vasculature, which is integral to the functioning of literally all human organs, serves as a fundamental physiological basis for age-related alterations as well as a shared etiological mechanism for various chronic diseases prevalent in the elderly population. China, home to the world's largest aging population, faces an escalating challenge in addressing the prevention and management of these age-related conditions. To meet this challenge, the Aging Biomarker Consortium of China has developed an expert consensus on biomarkers of vascular aging (VA) by synthesizing literature and insights from scientists and clinicians. This consensus provides a comprehensive assessment of biomarkers associated with VA and presents a systemic framework to classify them into three dimensions: functional, structural, and humoral. Within each dimension, the expert panel recommends the most clinically relevant VA biomarkers. For the functional domain, biomarkers reflecting vascular stiffness and endothelial function are highlighted. The structural dimension encompasses metrics for vascular structure, microvascular structure, and distribution. Additionally, proinflammatory factors are emphasized as biomarkers with the humoral dimension. The aim of this expert consensus is to establish a foundation for assessing the extent of VA and conducting research related to VA, with the ultimate goal of improving the vascular health of the elderly in China and globally.
Collapse
Affiliation(s)
| | - Le Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jun Guo
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Yuehong Liu
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Shimin Sun
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine, Jena University Hospital, Jena 07743, Germany
| | - Baohua Liu
- School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen 518055, China
| | - Qi Yang
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jun Tao
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun-Yat-sen University, Guangzhou 510080, China
| | - Xiao-Li Tian
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Jun Pu
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai 200127, China
| | - Huashan Hong
- Department of Geriatrics, Fujian Key Laboratory of Vascular Aging, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Miao Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Clinical Pharmacology Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Hou-Zao Chen
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
- Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Jie Ren
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Xiaoming Wang
- Department of Geriatrics, Xijing Hospital, Air Force Medical University, Xi’an 710032, China
| | - Zhen Liang
- Shenzhen People’s Hospital, Shenzhen 518020, China
| | - Yuan Wang
- Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Kai Huang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou 510632, China
| | - Guang-Hui Liu
- University of Chinese Academy of Sciences, Beijing 100049, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Gang Pei
- Collaborative Innovation Center for Brain Science, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Cuntai Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
7
|
Phillips CM, Stamatovic SM, Keep RF, Andjelkovic AV. Epigenetics and stroke: role of DNA methylation and effect of aging on blood-brain barrier recovery. Fluids Barriers CNS 2023; 20:14. [PMID: 36855111 PMCID: PMC9972738 DOI: 10.1186/s12987-023-00414-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/10/2023] [Indexed: 03/02/2023] Open
Abstract
Incomplete recovery of blood-brain barrier (BBB) function contributes to stroke outcomes. How the BBB recovers after stroke remains largely unknown. Emerging evidence suggests that epigenetic factors play a significant role in regulating post-stroke BBB recovery. This study aimed to evaluate the epigenetic and transcriptional profile of cerebral microvessels after thromboembolic (TE) stroke to define potential causes of limited BBB recovery. RNA-sequencing and reduced representation bisulfite sequencing (RRBS) analyses were performed using microvessels isolated from young (6 months) and old (18 months) mice seven days poststroke compared to age-matched sham controls. DNA methylation profiling of poststroke brain microvessels revealed 11,287 differentially methylated regions (DMR) in old and 9818 DMR in young mice, corresponding to annotated genes. These DMR were enriched in genes encoding cell structural proteins (e.g., cell junction, and cell polarity, actin cytoskeleton, extracellular matrix), transporters and channels (e.g., potassium transmembrane transporter, organic anion and inorganic cation transporters, calcium ion transport), and proteins involved in endothelial cell processes (e.g., angiogenesis/vasculogenesis, cell signaling and transcription regulation). Integrated analysis of methylation and RNA sequencing identified changes in cell junctions (occludin), actin remodeling (ezrin) as well as signaling pathways like Rho GTPase (RhoA and Cdc42ep4). Aging as a hub of aberrant methylation affected BBB recovery processes by profound alterations (hypermethylation and repression) in structural protein expression (e.g., claudin-5) as well as activation of a set of genes involved in endothelial to mesenchymal transformation (e.g., Sox9, Snai1), repression of angiogenesis and epigenetic regulation. These findings revealed that DNA methylation plays an important role in regulating BBB repair after stroke, through regulating processes associated with BBB restoration and prevalently with processes enhancing BBB injury.
Collapse
Affiliation(s)
- Chelsea M Phillips
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Svetlana M Stamatovic
- Department of Pathology, Medical School, University of Michigan, 7520A MSRB I, 1150 W Medical Center Dr, Ann Arbor, MI, 48109-5602, USA
| | - Richard F Keep
- Department of Neurosurgery, Medical School, University of Michigan, 7520A MSRB I, 1150 W Medical Center Dr, Ann Arbor, MI, 48109-5602, USA.,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Anuska V Andjelkovic
- Department of Pathology, Medical School, University of Michigan, 7520A MSRB I, 1150 W Medical Center Dr, Ann Arbor, MI, 48109-5602, USA. .,Department of Neurosurgery, Medical School, University of Michigan, 7520A MSRB I, 1150 W Medical Center Dr, Ann Arbor, MI, 48109-5602, USA.
| |
Collapse
|
8
|
Phillips C, Stamatovic S, Keep R, Andjelkovic A. Epigenetics and stroke: role of DNA methylation and effect of aging on blood-brain barrier recovery. RESEARCH SQUARE 2023:rs.3.rs-2444060. [PMID: 36711725 PMCID: PMC9882686 DOI: 10.21203/rs.3.rs-2444060/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Incomplete recovery of blood-brain barrier (BBB) function contributes to stroke outcomes. How the BBB recovers after stroke remains largely unknown. Emerging evidence suggests that epigenetic factors play a significant role in regulating post-stroke BBB recovery. This study aimed to evaluate the epigenetic and transcriptional profile of cerebral microvessels after thromboembolic (TE) stroke to define potential causes of limited BBB recovery. RNA-sequencing and reduced representation bisulfite sequencing (RRBS) analyses were performed using microvessels isolated from young (6 months) and old (18 months) mice seven days poststroke compared to age-matched sham controls. DNA methylation profiling of poststroke brain microvessels revealed 11287 differentially methylated regions (DMR) in old and 9818 DMR in young mice, corresponding to annotated genes. These DMR were enriched in genes encoding cell structural proteins (e.g., cell junction, and cell polarity, actin cytoskeleton, extracellular matrix), transporters and channels (e.g., potassium transmembrane transporter, organic anion and inorganic cation transporters, calcium ion transport), and proteins involved in endothelial cell processes (e.g., angiogenesis/vasculogenesis, cell signaling and transcription regulation). Integrated analysis of methylation and RNA sequencing identified changes in cell junctions (occludin), actin remodeling (ezrin) as well as signaling pathways like Rho GTPase (RhoA and Cdc42ep4). Aging as a hub of aberrant methylation affected BBB recovery processes by profound alterations (hypermethylation and repression) in structural protein expression (e.g., claudin-5) as well as activation of a set of genes involved in endothelial to mesenchymal transformation (e.g., Sox17 , Snail1 ), repression of angiogenesis and epigenetic regulation. These findings revealed that DNA methylation plays an important role in regulating BBB repair after stroke, through regulating processes associated with BBB restoration and prevalently with processes enhancing BBB injury.
Collapse
|
9
|
Aitbaev KA, Murkamilov IT, Fomin VV, Rayimzhanov ZR, Yusupova TF, Yusupov FA. [New data on the pathophysiology of ischemic stroke: epigenetic mechanisms in focus]. Zh Nevrol Psikhiatr Im S S Korsakova 2023; 123:24-29. [PMID: 38148694 DOI: 10.17116/jnevro202312312224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Epigenetics is a branch of molecular biology that studies modifications able to change gene expression without changing the DNA sequence. Epigenetic modulations include DNA methylation, histone modifications, and noncoding RNAs. These heritable and modifiable gene changes can be caused by lifestyle and dietary factors. In recent years, epigenetic changes have been associated with the pathogenesis of a number of diseases, such as diabetes mellitus, obesity, renal pathology and various types of cancer. They were also associated with the pathogenesis of cardiovascular diseases, including ischemic stroke. In this regard, it is important to note that since epigenetic modifications are reversible processes, they can help in the development of new therapeutic approaches to treat human diseases. This mini-review presents the latest data on the influence of epigenetic modifications on the pathogenesis of ischemic stroke obtained both in animal models and in patients.
Collapse
Affiliation(s)
- K A Aitbaev
- Research Institute of Molecular Biology and Medicine, Bishkek, Kyrgyzstan
| | - I T Murkamilov
- Akhunbaev Kyrgyz State Medical Academy, Bishkek, Kyrgyzstan
- Kyrgyz- Russian Slavic University named after the First President of Russia B.N. Yeltsin, Bishkek, Kyrgyzstan
| | - V V Fomin
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - Z R Rayimzhanov
- Burdenko Military Clinical Hospital Ministry of Defense of Russia, Moscow, Russia
| | | | | |
Collapse
|
10
|
Epigenetic Mechanisms Involved in Inflammaging-Associated Hypertension. Curr Hypertens Rep 2022; 24:547-562. [PMID: 35796869 DOI: 10.1007/s11906-022-01214-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW This review summarizes the involvement of inflammaging in vascular damage with focus on the epigenetic mechanisms by which inflammaging-induced hypertension is triggered. RECENT FINDINGS Inflammaging in hypertension is a complex condition associated with the production of inflammatory mediators by the immune cells, enhancement of oxidative stress, and tissue remodeling in vascular smooth muscle cells and endothelial cells. Cellular processes are numerous, including inflammasome assembly and cell senescence which may involve mitochondrial dysfunction, autophagy, DNA damage response, dysbiosis, and many others. More recently, a series of noncoding RNAs, mainly microRNAs, have been described as possessing epigenetic actions on the regulation of inflammasome-related hypertension, emerging as a promising therapeutic strategy. Although there are a variety of pharmacological agents that effectively regulate inflammaging-related hypertension, a deeper understanding of the epigenetic events behind the control of vessel deterioration is needed for the treatment or even to prevent the disease onset.
Collapse
|
11
|
Monsour M, Gordon J, Lockard G, Alayli A, Elsayed B, Connolly J, Borlongan CV. Minor Changes for a Major Impact: A Review of Epigenetic Modifications in Cell-Based Therapies for Stroke. Int J Mol Sci 2022; 23:13106. [PMID: 36361891 PMCID: PMC9656972 DOI: 10.3390/ijms232113106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 08/16/2024] Open
Abstract
Epigenetic changes in stroke may revolutionize cell-based therapies aimed at reducing ischemic stroke risk and damage. Epigenetic changes are a novel therapeutic target due to their specificity and potential for reversal. Possible targets for epigenetic modification include DNA methylation and demethylation, post-translational histone modification, and the actions of non-coding RNAs such as microRNAs. Many of these epigenetic modifications have been reported to modulate atherosclerosis development and progression, ultimately contributing to stroke pathogenesis. Furthermore, epigenetics may play a major role in inflammatory responses following stroke. Stem cells for stroke have demonstrated safety in clinical trials for stroke and show therapeutic benefit in pre-clinical studies. The efficacy of these cell-based interventions may be amplified with adjunctive epigenetic modifications. This review advances the role of epigenetics in atherosclerosis and inflammation in the context of stroke, followed by a discussion on current stem cell studies modulating epigenetics to ameliorate stroke damage.
Collapse
Affiliation(s)
- Molly Monsour
- University of South Florida Morsani College of Medicine, Tampa, FL 33602, USA
| | - Jonah Gordon
- University of South Florida Morsani College of Medicine, Tampa, FL 33602, USA
| | - Gavin Lockard
- University of South Florida Morsani College of Medicine, Tampa, FL 33602, USA
| | - Adam Alayli
- University of South Florida Morsani College of Medicine, Tampa, FL 33602, USA
| | - Bassel Elsayed
- University of South Florida Morsani College of Medicine, Tampa, FL 33602, USA
| | - Jacob Connolly
- University of South Florida Morsani College of Medicine, Tampa, FL 33602, USA
| | - Cesar V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| |
Collapse
|
12
|
Peng J, Ghosh D, Zhang F, Yang L, Wu J, Pang J, Zhang L, Yin S, Jiang Y. Advancement of epigenetics in stroke. Front Neurosci 2022; 16:981726. [PMID: 36312038 PMCID: PMC9610114 DOI: 10.3389/fnins.2022.981726] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/27/2022] [Indexed: 10/14/2023] Open
Abstract
A wide plethora of intervention procedures, tissue plasminogen activators, mechanical thrombectomy, and several neuroprotective drugs were reported in stroke research over the last decennium. However, against this vivid background of newly emerging pieces of evidence, there is little to no advancement in the overall functional outcomes. With the advancement of epigenetic tools and technologies associated with intervention medicine, stroke research has entered a new fertile. The stroke involves an overabundance of inflammatory responses arising in part due to the body's immune response to brain injury. Neuroinflammation contributes to significant neuronal cell death and the development of functional impairment and even death in stroke patients. Recent studies have demonstrated that epigenetics plays a key role in post-stroke conditions, leading to inflammatory responses and alteration of the microenvironment within the injured tissue. In this review, we summarize the progress of epigenetics which provides an overview of recent advancements on the emerging key role of secondary brain injury in stroke. We also discuss potential epigenetic therapies related to clinical practice.
Collapse
Affiliation(s)
- Jianhua Peng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Dipritu Ghosh
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Fan Zhang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lei Yang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jinpeng Wu
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jinwei Pang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lifang Zhang
- Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shigang Yin
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, China
| | - Yong Jiang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
13
|
Liu Y, Shang G, Zhang X, Liu F, Zhang C, Li Z, Jia J, Xu Y, Zhang Z, Yang S, Zhou B, Luan Y, Huang Y, Peng Y, Han T, He Y, Zheng H. CAMTA1 gene affects the ischemia-reperfusion injury by regulating CCND1. Front Cell Neurosci 2022; 16:868291. [PMID: 36159397 PMCID: PMC9500443 DOI: 10.3389/fncel.2022.868291] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Epigenetic modulations lead to changes in gene expression, including DNA methylation, histone modifications, and noncoding RNAs. In recent years, epigenetic modifications have been related to the pathogenesis of different types of cancer, cardiovascular disease, and other diseases. Emerging evidence indicates that DNA methylation could be associated with ischemic stroke (IS) and plays a role in pathological progression, but the underlying mechanism has not yet been fully understood. In this study, we used human methylation 850K BeadChip to analyze the differences in gene methylation status in the peripheral blood samples from two groups (3 IS patients vs. 3 healthy controls). According to their bioinformatics profiling, we found 278 genes with significantly different methylation levels. Seven genes with the most significant methylation modifications were validated in two expanded groups (100 IS patients vs. 100 healthy controls). The CAMTA1 gene had significantly different methylation changes in patients compared to the controls. To understand the CAMTA1 function in stroke, we generated CAMTA1 knockout in SH-SY5Y cells. RNA seq results in CAMTA1 knockout cells revealed the pathways and gene set enrichments involved in cellular proliferation and cell cycle. Furthermore, a series of experiments demonstrated that in the oxygen-glucose deprivation/re-oxygenation (OGD/R) model system, the expression of cyclin D1, an essential regulator of cell cycle progression, was increased in SH-SY5Y CAMTA1 KO cells. Increasing evidence demonstrated that ischemic stress could inappropriately raise cyclin D1 levels in mature neurons. However, the molecular signals leading to an increased cyclin D1 level are unclear. Our findings demonstrate for the first time that the CAMTA1 gene could regulate cyclin D1 expression and implicate their role in strokes.
Collapse
Affiliation(s)
- Yang Liu
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Guohui Shang
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xuran Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Henan University of CM, Henan University of CM, Zhengzhou, China
| | - Fuyong Liu
- Department of Pathogenic Biology and Immunology, School of Life Sciences, Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Chi Zhang
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhihao Li
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jing Jia
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yan Xu
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhaojing Zhang
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Shangdong Yang
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Baixue Zhou
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yingying Luan
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yanyang Huang
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yue Peng
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Tianyi Han
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ying He
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Ying He
| | - Hong Zheng
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- *Correspondence: Hong Zheng
| |
Collapse
|
14
|
The Role of DNA Methylation in Stroke Recovery. Int J Mol Sci 2022; 23:ijms231810373. [PMID: 36142283 PMCID: PMC9499691 DOI: 10.3390/ijms231810373] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Epigenetic alterations affect the onset of ischemic stroke, brain injury after stroke, and mechanisms of poststroke recovery. In particular, DNA methylation can be dynamically altered by maintaining normal brain function or inducing abnormal brain damage. DNA methylation is regulated by DNA methyltransferase (DNMT), which promotes methylation, DNA demethylase, which removes methyl groups, and methyl-cytosine–phosphate–guanine-binding domain (MBD) protein, which binds methylated DNA and inhibits gene expression. Investigating the effects of modulating DNMT, TET, and MBD protein expression on neuronal cell death and neurorepair in ischemic stroke and elucidating the underlying mechanisms can facilitate the formulation of therapeutic strategies for neuroprotection and promotion of neuronal recovery after stroke. In this review, we summarize the role of DNA methylation in neuroprotection and neuronal recovery after stroke according to the current knowledge regarding the effects of DNA methylation on excitotoxicity, oxidative stress, apoptosis, neuroinflammation, and recovery after ischemic stroke. This review of the literature regarding the role of DNA methylation in neuroprotection and functional recovery after stroke may contribute to the development and application of novel therapeutic strategies for stroke.
Collapse
|
15
|
Abstract
Over the course of a human lifespan, genome integrity erodes, leading to an increased abundance of several types of chromatin changes. The abundance of DNA lesions (chemical perturbations to nucleotides) increases with age, as does the number of genomic mutations and transcriptional disruptions caused by replication or transcription of those lesions, respectively. At the epigenetic level, precise DNA methylation patterns degrade, likely causing increasingly stochastic variations in gene expression. Similarly, the tight regulation of histone modifications begins to unravel. The genomic instability caused by these mechanisms allows transposon element reactivation and remobilization, further mutations, gene dysregulation, and cytoplasmic chromatin fragments. This cumulative genomic instability promotes cell signaling events that drive cell fate decisions and extracellular communications known to disrupt tissue homeostasis and regeneration. In this Review, we focus on age-related epigenetic changes and their interactions with age-related genomic changes that instigate these events.
Collapse
Affiliation(s)
- Carolina Soto-Palma
- Institute on the Biology of Aging and Metabolism
- Department of Biochemistry, Molecular Biology, and Biophysics
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism
- Department of Biochemistry, Molecular Biology, and Biophysics
| | - Christopher D. Faulk
- Institute on the Biology of Aging and Metabolism
- Department of Animal Science, and
| | - Xiao Dong
- Institute on the Biology of Aging and Metabolism
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
16
|
Stroke and Etiopathogenesis: What Is Known? Genes (Basel) 2022; 13:genes13060978. [PMID: 35741740 PMCID: PMC9222702 DOI: 10.3390/genes13060978] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 02/05/2023] Open
Abstract
Background: A substantial portion of stroke risk remains unexplained, and a contribution from genetic factors is supported by recent findings. In most cases, genetic risk factors contribute to stroke risk as part of a multifactorial predisposition. A major challenge in identifying the genetic determinants of stroke is fully understanding the complexity of the phenotype. Aims: Our narrative review is needed to improve our understanding of the biological pathways underlying the disease and, through this understanding, to accelerate the identification of new drug targets. Methods: We report, the research in the literature until February 2022 in this narrative review. The keywords are stroke, causes, etiopathogenesis, genetic, epigenetic, ischemic stroke. Results: While better risk prediction also remains a long-term goal, its implementation is still complex given the small effect-size of genetic risk variants. Some authors encourage the use of stroke genetic panels for stroke risk assessment and further stroke research. In addition, new biomarkers for the genetic causes of stroke and new targets for gene therapy are on the horizon. Conclusion: We summarize the latest evidence and perspectives of ischemic stroke genetics that may be of interest to the physician and useful for day-to-day clinical work in terms of both prevention and treatment of ischemic stroke.
Collapse
|
17
|
Das D, Karthik N, Taneja R. Crosstalk Between Inflammatory Signaling and Methylation in Cancer. Front Cell Dev Biol 2021; 9:756458. [PMID: 34901003 PMCID: PMC8652226 DOI: 10.3389/fcell.2021.756458] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/11/2021] [Indexed: 01/08/2023] Open
Abstract
Inflammation is an intricate immune response against infection and tissue damage. While the initial immune response is important for preventing tumorigenesis, chronic inflammation is implicated in cancer pathogenesis. It has been linked to various stages of tumor development including transformation, proliferation, angiogenesis, and metastasis. Immune cells, through the production of inflammatory mediators such as cytokines, chemokines, transforming growth factors, and adhesion molecules contribute to the survival, growth, and progression of the tumor in its microenvironment. The aberrant expression and secretion of pro-inflammatory and growth factors by the tumor cells result in the recruitment of immune cells, thus creating a mutual crosstalk. The reciprocal signaling between the tumor cells and the immune cells creates and maintains a successful tumor niche. Many inflammatory factors are regulated by epigenetic mechanisms including DNA methylation and histone modifications. In particular, DNA and histone methylation are crucial forms of transcriptional regulation and aberrant methylation has been associated with deregulated gene expression in oncogenesis. Such deregulations have been reported in both solid tumors and hematological malignancies. With technological advancements to study genome-wide epigenetic landscapes, it is now possible to identify molecular mechanisms underlying altered inflammatory profiles in cancer. In this review, we discuss the role of DNA and histone methylation in regulation of inflammatory pathways in human cancers and review the merits and challenges of targeting inflammatory mediators as well as epigenetic regulators in cancer.
Collapse
Affiliation(s)
- Dipanwita Das
- Department of Physiology, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nandini Karthik
- Department of Physiology, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Reshma Taneja
- Department of Physiology, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
18
|
Qiu M, Xu E, Zhan L. Epigenetic Regulations of Microglia/Macrophage Polarization in Ischemic Stroke. Front Mol Neurosci 2021; 14:697416. [PMID: 34707480 PMCID: PMC8542724 DOI: 10.3389/fnmol.2021.697416] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/26/2021] [Indexed: 01/04/2023] Open
Abstract
Ischemic stroke is one of the leading causes of death and disability worldwide. Microglia/macrophages (MMs)-mediated neuroinflammation contributes significantly to the pathological process of ischemic brain injury. Microglia, serving as resident innate immune cells in the central nervous system, undergo pro-inflammatory phenotype or anti-inflammatory phenotype in response to the microenvironmental changes after cerebral ischemia. Emerging evidence suggests that epigenetics modifications, reversible modifications of the phenotype without changing the DNA sequence, could play a pivotal role in regulation of MM polarization. However, the knowledge of the mechanism of epigenetic regulations of MM polarization after cerebral ischemia is still limited. In this review, we present the recent advances in the mechanisms of epigenetics involved in regulating MM polarization, including histone modification, non-coding RNA, and DNA methylation. In addition, we discuss the potential of epigenetic-mediated MM polarization as diagnostic and therapeutic targets for ischemic stroke. It is valuable to identify the underlying mechanisms between epigenetics and MM polarization, which may provide a promising treatment strategy for neuronal damage after cerebral ischemia.
Collapse
Affiliation(s)
- Meiqian Qiu
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - En Xu
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Lixuan Zhan
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| |
Collapse
|
19
|
Xu H, Li S, Liu YS. Roles and Mechanisms of DNA Methylation in Vascular Aging and Related Diseases. Front Cell Dev Biol 2021; 9:699374. [PMID: 34262910 PMCID: PMC8273304 DOI: 10.3389/fcell.2021.699374] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/07/2021] [Indexed: 12/20/2022] Open
Abstract
Vascular aging is a pivotal risk factor promoting vascular dysfunction, the development and progression of vascular aging-related diseases. The structure and function of endothelial cells (ECs), vascular smooth muscle cells (VSMCs), fibroblasts, and macrophages are disrupted during the aging process, causing vascular cell senescence as well as vascular dysfunction. DNA methylation, an epigenetic mechanism, involves the alteration of gene transcription without changing the DNA sequence. It is a dynamically reversible process modulated by methyltransferases and demethyltransferases. Emerging evidence reveals that DNA methylation is implicated in the vascular aging process and plays a central role in regulating vascular aging-related diseases. In this review, we seek to clarify the mechanisms of DNA methylation in modulating ECs, VSMCs, fibroblasts, and macrophages functions and primarily focus on the connection between DNA methylation and vascular aging-related diseases. Therefore, we represent many vascular aging-related genes which are modulated by DNA methylation. Besides, we concentrate on the potential clinical application of DNA methylation to serve as a reliable diagnostic tool and DNA methylation-based therapeutic drugs for vascular aging-related diseases.
Collapse
Affiliation(s)
- Hui Xu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Aging and Age-Related Disease Research, Central South University, Changsha, China
| | - Shuang Li
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Aging and Age-Related Disease Research, Central South University, Changsha, China
| | - You-Shuo Liu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Aging and Age-Related Disease Research, Central South University, Changsha, China
| |
Collapse
|
20
|
Kumar A, Misra S, Nair P, Algahtany M. Epigenetics Mechanisms in Ischemic Stroke: A Promising Avenue? J Stroke Cerebrovasc Dis 2021; 30:105690. [PMID: 33684709 DOI: 10.1016/j.jstrokecerebrovasdis.2021.105690] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/15/2021] [Accepted: 02/15/2021] [Indexed: 12/27/2022] Open
Abstract
Stroke has emerged as the second most common cause of mortality worldwide and is a major public health problem. It is a multi-factorial disease and genetics plays an important role in its pathophysiology, however, mechanisms of genome involvement in the disease remain unclear. Both genetic and epigenetic mechanisms could play a role in the development of stroke disease. Although epigenetic characteristics may also be heritable, they can be modified during the lifetime under different environmental exposure in response to lifestyle. Recent studies provide clear evidence that epigenetic factors play an important role in the pathological mechanisms leading to an elevated risk of cardiovascular diseases and stroke. Epigenetic changes are reversible therefore; studying epigenetic factors may serve as a marker for disease progression, biomarker for disease diagnosis, and development of novel targets for therapeutic intervention. Identifying the factors which predispose the risk of stroke provides information for the mechanism of stroke and the design of new drug targets where epigenetic modifications play a significant role. Epigenetic modifications play an essential role in a large variety of multifactorial diseases. This review will focus on the evidence that epigenetic mechanisms play a crucial role in the pathophysiology of ischemic stroke.
Collapse
Affiliation(s)
- Amit Kumar
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India.
| | - Shubham Misra
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India.
| | - Pallavi Nair
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India.
| | - Mubarak Algahtany
- Division of Neurosurgery, Department of Surgery, College of Medicine, King Khalid University, Abha, Saudi Arabia.
| |
Collapse
|
21
|
Dolcini J, Kioumourtzoglou MA, Cayir A, Sanchez-Guerra M, Brennan KJ, Dereix AE, Coull BA, Spiro A, Vokonas P, Schwartz J, Baccarelli AA. Age and mitochondrial DNA copy number influence the association between outdoor temperature and cognitive function: Insights from the VA Normative Aging Study. Environ Epidemiol 2020; 4:e0108. [PMID: 32832843 PMCID: PMC7423527 DOI: 10.1097/ee9.0000000000000108] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 06/29/2020] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND General cognitive function deteriorates with aging, a change that has been linked to outdoor temperature. Older individuals have reduced ability to adapt to changes in outdoor temperature than younger people. However, to what extent short-term changes in outdoor temperature interact with mitochondria to affect cognition in older people has not yet been determined. METHODS Our study included 591 participants of the Normative Aging Study who underwent multiple examinations between 2000 and 2013. Cognitive function was evaluated via the Mini-Mental State Examination. Outdoor temperature was estimated at residential addresses 1 day before the examination using on a validated spatiotemporal temperature model. Mitochondrial DNA copy number (mtDNAcn) was determined using buffy coat samples. RESULTS We found an interaction between temperature, age, mtDNAcn, and cognition. In individuals 84 years of age or older, cooler temperature was associated with low cognition (odds ratio = 1.2; 95% confidence interval = 1.05, 1.35 for a 1°C decrease in temperature; P = 0.007). We found higher odds ratio per 1°C decrease in temperature among individuals with lower mtDNAcn (β3 = 0.12; 95% confidence interval = 0.01, 0.22; P interaction = 0.02). CONCLUSIONS Our findings, albeit potentially underpowered, suggest that older individuals may be more susceptible to the influence of short-term temperature exposure on cognition. Moreover, the level of mtDNAcn may also modify the association between temperature and cognitive function, indicating a possible role of these cellular elements in this relationship.
Collapse
Affiliation(s)
- Jacopo Dolcini
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York
- Department of Biomedical Sciences and Public Health, Section of Hygiene and Preventive Medicine, Medical School, Polytechnic University of Marche, Ancona, Italy
| | | | - Akin Cayir
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Marco Sanchez-Guerra
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Kasey J. Brennan
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York
| | - Alexandra E. Dereix
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York
| | - Brent A. Coull
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Avron Spiro
- Veterans Affairs Normative Aging Study, Veterans Affairs Boston Healthcare System, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Pantel Vokonas
- Veterans Affairs Normative Aging Study, Veterans Affairs Boston Healthcare System, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Joel Schwartz
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Andrea A. Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York
| |
Collapse
|
22
|
Micheu MM, Birsan MV, Szép R, Keresztesi Á, Nita IA. From air pollution to cardiovascular diseases: the emerging role of epigenetics. Mol Biol Rep 2020; 47:5559-5567. [PMID: 32506306 DOI: 10.1007/s11033-020-05570-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 06/05/2020] [Indexed: 12/11/2022]
Abstract
The association between air pollution and a wide-ranging spectrum of acute and chronic disorders-including cardiovascular diseases-is widely acknowledged. Exposure to airborne pollutants triggers harmful mechanisms such as oxidative stress and systemic inflammation, which lead to increased incidence of myocardial infarction, arterial hypertension, stroke, and heart failure. Sustained efforts have been made in recent years to discover how environmental exposures affect human health through epigenetic phenomena, such as DNA methylation, histone modifications and non-coding RNA-mediated gene regulation. This review summarizes the current evidences on the relationship between air pollution exposure, epigenetic alterations and cardiovascular impact, in view of present implications and future perspectives.
Collapse
Affiliation(s)
- Miruna-Mihaela Micheu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Bucharest, Romania
| | - Marius-Victor Birsan
- Department of Research and Meteo Infrastructure Projects, Meteo Romania (National Meteorological Administration), Sos. Bucuresti-Ploiesti 97, 013686, Bucharest, Romania.
| | - Róbert Szép
- Doctoral School of Chemistry, Faculty of Natural Sciences, University of Pécs, Pecs, Hungary
- Institute for R&D in Hunting and Mountain Resources, Miercurea Ciuc, Romania
- Department of Bioengineering, Faculty of Economics, Socio-Human Science and Engineering, Sapientia Hungarian University of Transylvania, Miercurea Ciuc, Romania
| | - Ágnes Keresztesi
- Doctoral School of Chemistry, Faculty of Natural Sciences, University of Pécs, Pecs, Hungary
- Institute for R&D in Hunting and Mountain Resources, Miercurea Ciuc, Romania
- Department of Bioengineering, Faculty of Economics, Socio-Human Science and Engineering, Sapientia Hungarian University of Transylvania, Miercurea Ciuc, Romania
| | - Ion-Andrei Nita
- Department of Research and Meteo Infrastructure Projects, Meteo Romania (National Meteorological Administration), Sos. Bucuresti-Ploiesti 97, 013686, Bucharest, Romania
- Doctoral School of Geosciences, Faculty of Geography and Geology, Alexandru Ioan Cuza University, Iaşi, Romania
| |
Collapse
|
23
|
|
24
|
Cai S, Rao X, Ye J, Ling Y, Mi S, Chen H, Fan C, Li Y. Relationship between urinary bisphenol a levels and cardiovascular diseases in the U.S. adult population, 2003-2014. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 192:110300. [PMID: 32058166 DOI: 10.1016/j.ecoenv.2020.110300] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/18/2020] [Accepted: 02/04/2020] [Indexed: 05/23/2023]
Abstract
BACKGROUND Emerging evidence has identified cardiovascular system as a potential target of Bisphenol A (BPA). Although a few studies have revealed the relationship between BPA and the risk of several cardiovascular diseases (CVD) outcomes and CVD risk factors, no published studies have investigated the link between urinary BPA and the risk of stroke. METHODS Data were derived from the United States National Health and Nutrition Examination Surveys (NHANES), with a representative sample aged ≥20 years (n = 9139) from 2003 to 2014. We performed multivariable logistic regression model to estimate associations between quartiles and natural logarithm transformed urinary BPA concentrations and five specific CVD outcomes and total CVD. RESULTS In quartile analysis, highest level of urinary BPA was associated with increased prevalence of myocardial infarction (MI) (OR = 1.73, 95% CI = 1.11-2.69) and stroke (OR = 1.61, 95% CI = 1.09-2.36), when compared with those at the lowest quartile. Per unit (μg/g creatinine) increment in ln-transformed BPA concentration was shown to be significantly associated with 19%, 19%, 25%, 29%, 20%, and 16% increased odds ratios of prevalence of congestive heart failure, coronary heart disease (CHD), angina pectoris, MI, stroke and total CVD among total participants, respectively. Similar associations were found in males rather than in females. CONCLUSION We provided the premier evidence of positive relationship between urinary BPA concentration and stroke in U.S. POPULATION Urinary BPA levels were also positively correlated with congestive heart failure, CHD, angina pectoris, MI, as well as total CVD. These associations were more evident in males. Well-coordinated and prospective studies are warranted to gain the human effects of BPA on CVD.
Collapse
Affiliation(s)
- Shaofang Cai
- Department of Science and Education, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, China
| | - Xianming Rao
- The Second Affiliated Hospital of Xiamen Medical College, Xiamen, China
| | - Jianhong Ye
- The Second Affiliated Hospital of Xiamen Medical College, Xiamen, China
| | - Yuxiao Ling
- Department of Epidemiology and Health Statistics, Hangzhou Medical College School of Public Health, Hangzhou, China
| | - Shuai Mi
- Department of Epidemiology and Health Statistics, Hangzhou Medical College School of Public Health, Hangzhou, China
| | - Hanzhu Chen
- Department of Epidemiology and Health Statistics, Hangzhou Medical College School of Public Health, Hangzhou, China
| | - Chunhong Fan
- Department of Epidemiology and Health Statistics, Hangzhou Medical College School of Public Health, Hangzhou, China
| | - Yingjun Li
- Department of Epidemiology and Health Statistics, Hangzhou Medical College School of Public Health, Hangzhou, China.
| |
Collapse
|
25
|
Stanzione R, Cotugno M, Bianchi F, Marchitti S, Forte M, Volpe M, Rubattu S. Pathogenesis of Ischemic Stroke: Role of Epigenetic Mechanisms. Genes (Basel) 2020; 11:genes11010089. [PMID: 31941075 PMCID: PMC7017187 DOI: 10.3390/genes11010089] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/29/2019] [Accepted: 01/09/2020] [Indexed: 12/17/2022] Open
Abstract
Epigenetics is the branch of molecular biology that studies modifications able to change gene expression without altering the DNA sequence. Epigenetic modulations include DNA methylation, histone modifications, and noncoding RNAs. These gene modifications are heritable and modifiable and can be triggered by lifestyle and nutritional factors. In recent years, epigenetic changes have been associated with the pathogenesis of several diseases such as diabetes, obesity, renal pathology, and different types of cancer. They have also been related with the pathogenesis of cardiovascular diseases including ischemic stroke. Importantly, since epigenetic modifications are reversible processes they could assist with the development of new therapeutic approaches for the treatment of human diseases. In the present review article, we aim to collect the most recent evidence concerning the impact of epigenetic modifications on the pathogenesis of ischemic stroke in both animal models and humans.
Collapse
Affiliation(s)
- Rosita Stanzione
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy; (M.C.); (F.B.); (S.M.); (M.F.); (M.V.); (S.R.)
- Correspondence: ; Tel.: +86-5915224
| | - Maria Cotugno
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy; (M.C.); (F.B.); (S.M.); (M.F.); (M.V.); (S.R.)
| | - Franca Bianchi
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy; (M.C.); (F.B.); (S.M.); (M.F.); (M.V.); (S.R.)
| | - Simona Marchitti
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy; (M.C.); (F.B.); (S.M.); (M.F.); (M.V.); (S.R.)
| | - Maurizio Forte
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy; (M.C.); (F.B.); (S.M.); (M.F.); (M.V.); (S.R.)
| | - Massimo Volpe
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy; (M.C.); (F.B.); (S.M.); (M.F.); (M.V.); (S.R.)
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Speranza Rubattu
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy; (M.C.); (F.B.); (S.M.); (M.F.); (M.V.); (S.R.)
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| |
Collapse
|
26
|
Perera BP, Faulk C, Svoboda LK, Goodrich JM, Dolinoy DC. The role of environmental exposures and the epigenome in health and disease. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2020; 61:176-192. [PMID: 31177562 PMCID: PMC7252203 DOI: 10.1002/em.22311] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 05/02/2023]
Abstract
The genetic material of every organism exists within the context of regulatory networks that govern gene expression, collectively called the epigenome. Epigenetics has taken center stage in the study of diseases such as cancer and diabetes, but its integration into the field of environmental health is still emerging. As the Environmental Mutagenesis and Genomics Society (EMGS) celebrates its 50th Anniversary this year, we have come together to review and summarize the seminal advances in the field of environmental epigenomics. Specifically, we focus on the role epigenetics may play in multigenerational and transgenerational transmission of environmentally induced health effects. We also summarize state of the art techniques for evaluating the epigenome, environmental epigenetic analysis, and the emerging field of epigenome editing. Finally, we evaluate transposon epigenetics as they relate to environmental exposures and explore the role of noncoding RNA as biomarkers of environmental exposures. Although the field has advanced over the past several decades, including being recognized by EMGS with its own Special Interest Group, recently renamed Epigenomics, we are excited about the opportunities for environmental epigenetic science in the next 50 years. Environ. Mol. Mutagen. 61:176-192, 2020. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bambarendage P.U. Perera
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Christopher Faulk
- Department of Animal Sciences, University of Minnesota, St. Paul, Minnesota
| | - Laurie K. Svoboda
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Jaclyn M. Goodrich
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Dana C. Dolinoy
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
- Correspondence to: Dana C. Dolinoy, Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan.
| |
Collapse
|
27
|
Wang C, O'Brien KM, Xu Z, Sandler DP, Taylor JA, Weinberg CR. Long-term ambient fine particulate matter and DNA methylation in inflammation pathways: results from the Sister Study. Epigenetics 2019; 15:524-535. [PMID: 31822152 DOI: 10.1080/15592294.2019.1699894] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Although underlying mechanisms of long-term exposure to air pollution and cardiovascular disease remain obscure, effects might partially act through changes in DNA methylation. We examined the associations between long-term ambient fine particulate matter (PM2.5) and methylation, considering both a global measure and methylation at several specific inflammation-related loci, in two random sub-cohorts selected from a nationwide prospective study of US women. In one sub-cohort we measured long interspersed nucleotide element (LINE-1); in the other, we measured methylation at three candidates CpG loci related to inflammatory pathways [tumour necrosis factor-alpha (TNF-α) and toll-like receptor-2 (TLR-2)]. Annual average contemporaneous ambient PM2.5 concentrations were estimated for the current residence. We used both classical least-squares and quantile regression models to estimate the long-term effects. The women in sub-cohorts 1 (n = 491) and 2 (n = 882) had mean ages of 55.8 and 56.7, respectively. Neither modelling approach showed an association between long-term PM2.5 and LINE-1 methylation or between PM2.5 and either of the two CpG sites in TLR-2. Using linear regression, there was an estimated change of -6.5% (95% confidence interval CI: -13.34%, 0.35%) in mean methylation of TNF-α per 5 µg/m3 increase in PM2.5. Quantile regression showed that the downward shift was mainly in the lower half of the distribution of DNA methylation. Long-term residence in regions with higher ambient PM2.5 may be associated with increased TNF-α through a reduction in methylation, particularly in the lower tail. Epigenetic markers and quantile regression might provide insight into mechanisms underlying the relationship between air pollution and cardiovascular disease.
Collapse
Affiliation(s)
- Cuicui Wang
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA.,Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Katie M O'Brien
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA.,Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Zongli Xu
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Jack A Taylor
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Clarice R Weinberg
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| |
Collapse
|
28
|
Li Z, Li N, Guo C, Li X, Qian Y, Wu J, Yang Y, Wei Y. Genomic DNA methylation signatures in different tissues after ambient air particulate matter exposure. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 179:175-181. [PMID: 31039460 DOI: 10.1016/j.ecoenv.2019.04.049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/13/2019] [Accepted: 04/16/2019] [Indexed: 06/09/2023]
Abstract
DNA methylation (5-mc) is one of the several epigenetic markers, and is generally associated with the inhibition of gene expression. Both hyper and hypo DNA methylation are associated with the diseases. Exposure to fine particles with a diameter of 2.5 μm or less (PM2.5) is a pervasive risk factor for cardiopulmonary mortality, metabolic disorders, cognition damage, and etc.. Recent reports pointed toward that these diseases were associated with the altered DNA methylation level of some specific-gene, potentially suggesting that the DNA methylation alteration was involved in the health hazard derived from the PM2.5 exposure. In this study, we systematically investigated the global DNA methylation level of most tissues, including lung, heart, testis, thymus, spleen, epididymal fat, hippocampus, kidney, live, after short and long term PM2.5 exposure. After acute PM2.5 exposure, the global hypo-methylation in DNA was observed in lung and heart. Notably, after chronic PM2.5 exposure, level of global DNA methylation decreased in most organs which included lung, testis, thymus, spleen, epididymal fat, hippocampus and blood. The present study systematically demonstrated the global DNA methylation changes by PM2.5 exposure, and put forward a possible orientation for further exploring the effects of ambient air particles exposure on the specific organs.
Collapse
Affiliation(s)
- Zhigang Li
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Nannan Li
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Chen Guo
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Xiaoqian Li
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Yan Qian
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Jing Wu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | - Yining Yang
- Class 5 of Grade 2 in Senior High School, Beijing No.171 High School, 100013, China
| | - Yongjie Wei
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China.
| |
Collapse
|
29
|
Placek K, Schultze JL, Aschenbrenner AC. Epigenetic reprogramming of immune cells in injury, repair, and resolution. J Clin Invest 2019; 129:2994-3005. [PMID: 31329166 DOI: 10.1172/jci124619] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Immune cells are pivotal in the reaction to injury, whereupon, under ideal conditions, repair and resolution phases restore homeostasis following initial acute inflammation. Immune cell activation and reprogramming require transcriptional changes that can only be initiated if epigenetic alterations occur. Recently, accelerated deciphering of epigenetic mechanisms has extended knowledge of epigenetic regulation, including long-distance chromatin remodeling, DNA methylation, posttranslational histone modifications, and involvement of small and long noncoding RNAs. Epigenetic changes have been linked to aspects of immune cell development, activation, and differentiation. Furthermore, genome-wide epigenetic landscapes have been established for some immune cells, including tissue-resident macrophages, and blood-derived cells including T cells. The epigenetic mechanisms underlying developmental steps from hematopoietic stem cells to fully differentiated immune cells led to development of epigenetic technologies and insights into general rules of epigenetic regulation. Compared with more advanced research areas, epigenetic reprogramming of immune cells in injury remains in its infancy. While the early epigenetic mechanisms supporting activation of the immune response to injury have been studied, less is known about resolution and repair phases and cell type-specific changes. We review prominent recent findings concerning injury-mediated epigenetic reprogramming, particularly in stroke and myocardial infarction. Lastly, we illustrate how single-cell technologies will be crucial to understanding epigenetic reprogramming in the complex sequential processes following injury.
Collapse
Affiliation(s)
- Katarzyna Placek
- Immunology and Metabolism, LIMES Institute, University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Platform for Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases and the University of Bonn, Bonn, Germany.,Genomics and Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| | - Anna C Aschenbrenner
- Genomics and Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
30
|
Di Mauro V, Crasto S, Colombo FS, Di Pasquale E, Catalucci D. Wnt signalling mediates miR-133a nuclear re-localization for the transcriptional control of Dnmt3b in cardiac cells. Sci Rep 2019; 9:9320. [PMID: 31249372 PMCID: PMC6597717 DOI: 10.1038/s41598-019-45818-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 06/14/2019] [Indexed: 12/14/2022] Open
Abstract
MiR-133a is a muscle-enriched miRNA, which plays a key role for proper skeletal and cardiac muscle function via regulation of transduction cascades, including the Wnt signalling. MiR-133a modulates its targets via canonical mRNA repression, a process that has been largely demonstrated to occur within the cytoplasm. However, recent evidence has shown that miRNAs play additional roles in other sub-cellular compartments, such as nuclei. Here, we show that miR-133a translocates to the nucleus of cardiac cells following inactivation of the canonical Wnt pathway. The nuclear miR-133a/AGO2 complex binds to a complementary miR-133a target site within the promoter of the de novo DNA methyltransferase 3B (Dnmt3b) gene, leading to its transcriptional repression, which is mediated by DNMT3B itself. Altogether, these data show an unconventional role of miR-133a that upon its relocalization to the nucleus is responsible for epigenetic repression of its target gene Dnmt3b via a DNMT3B self-regulatory negative feedback loop.
Collapse
Affiliation(s)
- Vittoria Di Mauro
- University of Milan Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milan, Italy
- CNR-IRGB UOS Milan, Via Fantoli 15/16, 20138, Milan, Italy
- Humanitas Clinical and Research Center, via Alessandro Manzoni 113, 20089, Rozzano, Milan, Italy
| | - Silvia Crasto
- CNR-IRGB UOS Milan, Via Fantoli 15/16, 20138, Milan, Italy
- Humanitas Clinical and Research Center, via Alessandro Manzoni 113, 20089, Rozzano, Milan, Italy
| | - Federico Simone Colombo
- Humanitas Clinical and Research Center, via Alessandro Manzoni 113, 20089, Rozzano, Milan, Italy
| | - Elisa Di Pasquale
- CNR-IRGB UOS Milan, Via Fantoli 15/16, 20138, Milan, Italy
- Humanitas Clinical and Research Center, via Alessandro Manzoni 113, 20089, Rozzano, Milan, Italy
| | - Daniele Catalucci
- CNR-IRGB UOS Milan, Via Fantoli 15/16, 20138, Milan, Italy.
- Humanitas Clinical and Research Center, via Alessandro Manzoni 113, 20089, Rozzano, Milan, Italy.
| |
Collapse
|
31
|
Leso V, Vetrani I, Della Volpe I, Nocera C, Iavicoli I. Welding Fume Exposure and Epigenetic Alterations: A Systematic Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16101745. [PMID: 31108839 PMCID: PMC6571852 DOI: 10.3390/ijerph16101745] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/11/2019] [Accepted: 05/13/2019] [Indexed: 11/16/2022]
Abstract
Epigenetics are heritable changes in gene expression not coded in the DNA sequence, which stand at the interface between the genome, environmental exposure and development. From an occupational health perspective, epigenetic variants may link workplace exposures and health effects. Therefore, this review aimed to overview possible epigenetic effects induced by welding fumes on exposed workers and health implications. A systematic search was performed on Pubmed, Scopus, and ISI Web of Knowledge databases. DNA methylation changes have been reported in genes responsible for the cardiac autonomic function and coagulation, i.e., LINE-1, GPR133 and F2RL3, in mitochondrial-DNA-sequences involved in the regulation of energy-generation/redox-signaling, as well as in inflammatory activated genes, i.e., iNOS. However, the limited number of retrieved articles, their cross-sectional nature, the lack of a suitable qualitative-quantitative exposure assessment, and the heterogeneity of biological-outcomes investigated, prevent the extrapolation of a definite causal relationship between welding fumes and epigenetic phenomena. Future studies should clarify the function of such epigenetic alterations as possible markers of occupational exposure and early effect, dose-response relationships, and underlying molecular mechanisms. Overall, this may be helpful to guide suitable risk assessment and management strategies to protect the health of workers exposed to welding fumes.
Collapse
Affiliation(s)
- Veruscka Leso
- Section of Occupational Medicine, Department of Public Health, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy.
| | - Ilaria Vetrani
- Section of Occupational Medicine, Department of Public Health, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy.
| | - Ilaria Della Volpe
- Section of Occupational Medicine, Department of Public Health, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy.
| | - Caterina Nocera
- Section of Occupational Medicine, Department of Public Health, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy.
| | - Ivo Iavicoli
- Section of Occupational Medicine, Department of Public Health, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
32
|
Epigenetics and Inflammatory Markers: A Systematic Review of the Current Evidence. Int J Inflam 2019; 2019:6273680. [PMID: 31205673 PMCID: PMC6530203 DOI: 10.1155/2019/6273680] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/07/2019] [Indexed: 02/07/2023] Open
Abstract
Epigenetic mechanisms have been suggested to play a role in the genetic regulation of pathways related to inflammation. Therefore, we aimed to systematically review studies investigating the association between DNA methylation and histone modifications with circulatory inflammation markers in blood. Five bibliographic databases were screened until 21 November of 2017. We included studies conducted on humans that examined the association between epigenetic marks (DNA methylation and/or histone modifications) and a comprehensive list of inflammatory markers. Of the 3,759 identified references, 24 articles were included, involving, 17,399 individuals. There was suggestive evidence for global hypomethylation but better-quality studies in the future have to confirm this. Epigenome-wide association studies (EWAS) (n=7) reported most of the identified differentially methylated genes to be hypomethylated in inflammatory processes. Candidate genes studies reported 18 differentially methylated genes related to several circulatory inflammation markers. There was no overlap in the methylated sites investigated in candidate gene studies and EWAS, except for TMEM49, which was found to be hypomethylated with higher inflammatory markers in both types of studies. The relation between histone modifications and inflammatory markers was assessed by one study only. This review supports an association between epigenetic marks and inflammation, suggesting hypomethylation of the genome. Important gaps in the quality of studies were reported such as inadequate sample size, lack of adjustment for relevant confounders, and failure to replicate the findings. While most of the studies have been focused on C-reactive protein, further efforts should investigate other inflammatory markers.
Collapse
|
33
|
The role of DNA methylation and hydroxymethylation in immunosenescence. Ageing Res Rev 2019; 51:11-23. [PMID: 30769150 DOI: 10.1016/j.arr.2019.01.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 12/12/2022]
Abstract
A healthy functioning immune system is critical to stave off infectious diseases, but as humans and other organisms age, their immune systems decline. As a result, diseases that were readily thwarted in early life pose nontrivial harm and can even be deadly in late life. Immunosenescence is defined as the general deterioration of the immune system with age, and it is characterized by functional changes in hematopoietic stem cells (HSCs) and specific blood cell types as well as changes in levels of numerous factors, particularly those involved in inflammation. Potential mechanisms underlying immunosenescence include epigenetic changes such as changes in DNA methylation (DNAm) and DNA hydroxymethylation (DNAhm) that occur with age. The purpose of this review is to describe what is currently known about the relationship between immunosenescence and the age-related changes to DNAm and DNAhm, and to discuss experimental approaches best suited to fill gaps in our understanding.
Collapse
|
34
|
Lopes LL, Bressan J, Peluzio MDCG, Hermsdorff HHM. LINE-1 in Obesity and Cardiometabolic Diseases: A Systematic Review. J Am Coll Nutr 2019; 38:478-484. [PMID: 30862304 DOI: 10.1080/07315724.2018.1553116] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Epigenetic mechanisms may play an important role in the etiology of obesity and cardiometabolic diseases, by activating or silencing the related-genes. Scientific evidence has suggested that LINE-1 methylation is associated with body composition and obesity-related diseases, including insulin resistance, type 2 diabetes mellitus, and cardiovascular disease (CVD). It also has been evaluated as predictor of weight loss. The studies' results are still conflicting, and positive and negative associations have been found to LINE-1 methylation regarding adiposity and cardiometabolic markers. Overall, this review presents observational (cross-sectional and longitudinal) studies and interventions (diet, exercises, and bariatric surgery) that evaluated the relationship of the LINE-1 methylation with obesity, weight loss, dyslipidemias, hypertension, insulin resistance, CVD, and metabolic syndrome. TEACHING POINTS Epigenetic mechanisms may play an important role in the etiology of obesity and cardiometabolic diseases. Many studies have related methylation of LINE-1 with cardiometabolic diseases; however, the results are still controversial. The relationship between the etiology of chronic diseases and the methylation of LINE-1 is not fully elucidated. With advances in epigenetic studies, related mechanisms may be early biomarkers in weight change and cardiometabolic risk.
Collapse
Affiliation(s)
- Lílian L Lopes
- a Department of Nutrition and Health , Universidade Federal de Viçosa (UFV) , Viçosa , Minas Gerais , Brazil
| | - Josefina Bressan
- a Department of Nutrition and Health , Universidade Federal de Viçosa (UFV) , Viçosa , Minas Gerais , Brazil
| | - Maria do Carmo G Peluzio
- a Department of Nutrition and Health , Universidade Federal de Viçosa (UFV) , Viçosa , Minas Gerais , Brazil
| | - Helen Hermana M Hermsdorff
- a Department of Nutrition and Health , Universidade Federal de Viçosa (UFV) , Viçosa , Minas Gerais , Brazil
| |
Collapse
|
35
|
Nardini C, Moreau JF, Gensous N, Ravaioli F, Garagnani P, Bacalini MG. The epigenetics of inflammaging: The contribution of age-related heterochromatin loss and locus-specific remodelling and the modulation by environmental stimuli. Semin Immunol 2018; 40:49-60. [PMID: 30396810 DOI: 10.1016/j.smim.2018.10.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/12/2018] [Accepted: 10/15/2018] [Indexed: 12/12/2022]
Abstract
A growing amount of evidences indicates that inflammaging - the chronic, low grade inflammation state characteristic of the elderly - is the result of genetic as well as environmental or stochastic factors. Some of these, such as the accumulation of senescent cells that are persistent during aging or accompany its progression, seem to be sufficient to initiate the aging process and to fuel it. Others, like exposure to environmental compounds or infections, are temporary and resolve within a (relatively) short time. In both cases, however, a cellular memory of the event can be established by means of epigenetic modulation of the genome. In this review we will specifically discuss the relationship between epigenetics and inflammaging. In particular, we will show how age-associated epigenetic modifications concerned with heterochromatin loss and gene-specific remodelling, can promote inflammaging. Furthermore, we will recall how the exposure to specific nutritional, environmental and microbial stimuli can affect the rate of inflammaging through epigenetic mechanisms, touching also on the recent insight given by the concept of trained immunity.
Collapse
Affiliation(s)
- Christine Nardini
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; CNR IAC "Mauro Picone", Roma, Italy; Personal Genomics S.r.l., Verona, Italy
| | - Jean-Francois Moreau
- University of Bordeaux, CNRS-UMR5164, 146 rue Léo Saignat, 33076 Bordeaux, France; CHU Bordeaux, Place Amélie Raba-Léon, Bordeaux, France
| | - Noémie Gensous
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Francesco Ravaioli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Paolo Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy; Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, Stockholm, Sweden; Laboratory of Cell Biology, Rizzoli Orthopaedic Institute, Bologna, Italy; CNR Institute of Molecular Genetics, Unit of Bologna, Bologna, Italy; Center for Applied Biomedical Research (CRBA), St. Orsola-Malpighi University Hospital, Bologna, Italy.
| | | |
Collapse
|
36
|
Endothelial cell senescence in aging-related vascular dysfunction. Biochim Biophys Acta Mol Basis Dis 2018; 1865:1802-1809. [PMID: 31109450 DOI: 10.1016/j.bbadis.2018.08.008] [Citation(s) in RCA: 266] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/31/2018] [Accepted: 08/02/2018] [Indexed: 12/20/2022]
Abstract
Increased cardiovascular disease in aging is partly a consequence of the vascular endothelial cell (EC) senescence and associated vascular dysfunction. In this contest, EC senescence is a pathophysiological process of structural and functional changes including dysregulation of vascular tone, increased endothelium permeability, arterial stiffness, impairment of angiogenesis and vascular repair, and a reduction of EC mitochondrial biogenesis. Dysregulation of cell cycle, oxidative stress, altered calcium signaling, hyperuricemia, and vascular inflammation have been implicated in the development and progression of EC senescence and vascular disease in aging. A number of abnormal molecular pathways are associated with these underlying pathophysiological changes including Sirtuin 1, Klotho, fibroblast growth factor 21, and activation of the renin angiotensin-aldosterone system. However, the molecular mechanisms of EC senescence and associated vascular impairment in aging are not completely understood. This review provides a contemporary update on molecular mechanisms, pathophysiological events, as well functional changes in EC senescence and age-associated cardiovascular disease. This article is part of a Special Issue entitled: Genetic and epigenetic regulation of aging and longevity edited by Jun Ren & Megan Yingmei Zhang.
Collapse
|
37
|
Ng GYQ, Yun-An L, Sobey CG, Dheen T, Fann DYW, Arumugam TV. Epigenetic regulation of inflammation in stroke. Ther Adv Neurol Disord 2018; 11:1756286418771815. [PMID: 29774056 PMCID: PMC5949939 DOI: 10.1177/1756286418771815] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 03/29/2018] [Indexed: 12/30/2022] Open
Abstract
Despite extensive research, treatments for clinical stroke are still limited only to the administration of tissue plasminogen activator and the recent introduction of mechanical thrombectomy, which can be used in only a limited proportion of patients due to time constraints. A plethora of inflammatory events occur during stroke, arising in part due to the body's immune response to brain injury. Neuroinflammation contributes significantly to neuronal cell death and the development of functional impairment and death in stroke patients. Therefore, elucidating the molecular and cellular mechanisms underlying inflammatory damage following stroke injury will be essential for the development of useful therapies. Research findings increasingly point to the likelihood that epigenetic mechanisms play a role in the pathophysiology of stroke. Epigenetics involves the differential regulation of gene expression, including those involved in brain inflammation and remodelling after stroke. Hence, it is conceivable that epigenetic mechanisms may contribute to differential interindividual vulnerability and injury responses to cerebral ischaemia. In this review, we summarize recent findings on the emerging role of epigenetics in the regulation of neuroinflammation in stroke. We also discuss potential epigenetic targets that may be assessed for the development of stroke therapies.
Collapse
Affiliation(s)
- Gavin Yong-Quan Ng
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Singapore
| | - Lim Yun-An
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Singapore
| | - Christopher G. Sobey
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, Australia
| | - Thameem Dheen
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - David Yang-Wei Fann
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Singapore
| | - Thiruma V. Arumugam
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Medical Drive, MD9, Singapore School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea Neurobiology/Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore
| |
Collapse
|
38
|
Fiorito G, Vlaanderen J, Polidoro S, Gulliver J, Galassi C, Ranzi A, Krogh V, Grioni S, Agnoli C, Sacerdote C, Panico S, Tsai MY, Probst-Hensch N, Hoek G, Herceg Z, Vermeulen R, Ghantous A, Vineis P, Naccarati A. Oxidative stress and inflammation mediate the effect of air pollution on cardio- and cerebrovascular disease: A prospective study in nonsmokers. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2018; 59:234-246. [PMID: 29114965 DOI: 10.1002/em.22153] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/29/2017] [Accepted: 10/08/2017] [Indexed: 05/17/2023]
Abstract
Air pollution is associated with a broad range of adverse health effects, including mortality and morbidity due to cardio- and cerebrovascular diseases (CCVD), but the molecular mechanisms involved are not entirely understood. This study aims to investigate the involvement of oxidative stress and inflammation in the causal chain, and to identify intermediate biomarkers that are associated retrospectively with the exposure and prospectively with the disease. We designed a case-control study on CCVD nested in a cohort of 18,982 individuals from the EPIC-Italy study. We measured air pollution, inflammatory biomarkers, and whole-genome DNA methylation in blood collected up to 17 years before the diagnosis. The study sample includes all the incident CCVD cases among former- and never-smokers, with available stored blood sample, that arose in the cohort during the follow-up. We identified enrichment of altered DNA methylation in "ROS/Glutathione/Cytotoxic granules" and "Cytokine signaling" pathways related genes, associated with both air pollution (multiple comparisons adjusted p for enrichment ranging from 0.01 to 0.03 depending on pollutant) and with CCVD risk (P = 0.04 and P = 0.03, respectively). Also, Interleukin-17 was associated with higher exposure to NO2 (P = 0.0004), NOx (P = 0.0005), and CCVD risk (OR = 1.79; CI 1.04-3.11; P = 0.04 comparing extreme tertiles). Our findings indicate that chronic exposure to air pollution can lead to oxidative stress, which in turn activates a cascade of inflammatory responses mainly involving the "Cytokine signaling" pathway, leading to increased risk of CCVD. Inflammatory proteins and DNA methylation alterations can be detected several years before CCVD diagnosis in blood samples, being promising preclinical biomarkers. Environ. Mol. Mutagen. 59:234-246, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Giovanni Fiorito
- Italian Institute for Genomic Medicine (IIGM), Turin, Italy
- Department of Medical Sciences, University of Turin, Italy
| | - Jelle Vlaanderen
- Utrecht University, Institute for Risk Assessment Sciences, Environmental Epidemiology Division, Utrecht, Netherlands
| | | | - John Gulliver
- MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, UK
| | - Claudia Galassi
- Unit of Cancer Epidemiology, Città Della Salute e della Scienza University-Hospital and Center for Cancer Prevention (CPO), Turin, Italy
| | - Andrea Ranzi
- Environmental Health Reference Centre, Regional Agency for Prevention, Environment and Energy of Emilia-Romagna, Modena, Italy
| | - Vittorio Krogh
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Grioni
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Claudia Agnoli
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Città Della Salute e della Scienza University-Hospital and Center for Cancer Prevention (CPO), Turin, Italy
| | - Salvatore Panico
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | - Ming-Yi Tsai
- University of Basel, Basel, Switzerland
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Nicole Probst-Hensch
- University of Basel, Basel, Switzerland
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Gerard Hoek
- Utrecht University, Institute for Risk Assessment Sciences, Environmental Epidemiology Division, Utrecht, Netherlands
| | - Zdenko Herceg
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Roel Vermeulen
- Utrecht University, Institute for Risk Assessment Sciences, Environmental Epidemiology Division, Utrecht, Netherlands
| | - Akram Ghantous
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Paolo Vineis
- Italian Institute for Genomic Medicine (IIGM), Turin, Italy
- MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, UK
| | | |
Collapse
|
39
|
Duan L, Hu J, Xiong X, Liu Y, Wang J. The role of DNA methylation in coronary artery disease. Gene 2018; 646:91-97. [DOI: 10.1016/j.gene.2017.12.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 11/07/2017] [Accepted: 12/18/2017] [Indexed: 01/09/2023]
|
40
|
Kwa FA, Jackson DE. Manipulating the epigenome for the treatment of disorders with thrombotic complications. Drug Discov Today 2018; 23:719-726. [DOI: 10.1016/j.drudis.2018.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/11/2017] [Accepted: 01/04/2018] [Indexed: 11/25/2022]
|
41
|
Cardelli M. The epigenetic alterations of endogenous retroelements in aging. Mech Ageing Dev 2018; 174:30-46. [PMID: 29458070 DOI: 10.1016/j.mad.2018.02.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/05/2018] [Accepted: 02/08/2018] [Indexed: 02/06/2023]
Abstract
Endogenous retroelements, transposons that mobilize through RNA intermediates, include some of the most abundant repetitive sequences of the human genome, such as Alu and LINE-1 sequences, and human endogenous retroviruses. Recent discoveries demonstrate that these mobile genetic elements not only act as intragenomic parasites, but also exert regulatory roles in living cells. The risk of genomic instability represented by endogenous retroelements is normally counteracted by a series of epigenetic control mechanisms which include, among the most important, CpG DNA methylation. Indeed, most of the genomic CpG sites subjected to DNA methylation in the nuclear DNA are carried by these repetitive elements. As other parts of the genome, endogenous retroelements and other transposable elements are subjected to deep epigenetic alterations during aging, repeatedly observed in the context of organismal and cellular senescence, in human and other species. This review summarizes the current status of knowledge about the epigenetic alterations occurring in this large, non-genic portion of the genome in aging and age-related conditions, with a focus on the causes and the possible functional consequences of these alterations.
Collapse
Affiliation(s)
- Maurizio Cardelli
- Advanced Technology Center for Aging Research, Scientific Technological Area, Italian National Research Center on Aging (INRCA), via Birarelli 8, 60121 Ancona, Italy.
| |
Collapse
|
42
|
Pathophysiology of cardiovascular disease in diabetes mellitus. Cardiovasc Endocrinol Metab 2018; 7:4-9. [PMID: 31646271 DOI: 10.1097/xce.0000000000000141] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 11/22/2017] [Indexed: 01/12/2023]
Abstract
Diabetes mellitus elicits cellular, epigenetic, and post-translational changes that directly or indirectly affect the biology of the vasculature and other metabolic systems resulting in the apparition of cardiovascular disease. In this review, we provide a current perspective on the most recent discoveries in this field, with particular focus on hyperglycemia- induced pathology in the cardiovascular system. We also provide perspective on the clinical importance of molecular targeting of cardiovascular and diabetes mellitus therapies to treat hyperglycemia, inflammation, thrombosis, dyslipidemia, atherosclerosis, and hypertension.
Collapse
|
43
|
Correlation between global methylation level of peripheral blood leukocytes and serum C reactive protein level modified by MTHFR polymorphism: a cross-sectional study. BMC Cancer 2018; 18:184. [PMID: 29439678 PMCID: PMC5812223 DOI: 10.1186/s12885-018-4089-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 02/05/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Chronic inflammatory conditions are associated with higher tumor incidence through epigenetic and genetic alterations. Here, we focused on an association between an inflammation marker, C-reactive-protein (CRP), and global DNA methylation levels of peripheral blood leukocytes. METHODS The subjects were 384 healthy Japanese women enrolled as the control group of a case-control study for breast cancer conducted from 2001 to 2005. Global DNA methylation was quantified by Luminometric Methylation Assay (LUMA). RESULTS With adjustment for lifestyle-related factors, including folate intake, the global DNA methylation level of peripheral blood leukocytes was significantly but weakly increased by 0.43% per quartile category for CRP (P for trend = 0.010). Estimated methylation levels stratified by CRP quartile were 70.0%, 70.8%, 71.4%, and 71.3%, respectively. In addition, interaction between polymorphism of MTHFR (rs1801133, known as C677T) and CRP was significant (P for interaction = 0.046); the global methylation level was significantly increased by 0.61% per quartile category for CRP in the CT/TT group (those with the minor allele T, P for trend = 0.001), whereas no association was observed in the CC group (wild type). CONCLUSIONS Our study suggests that CRP concentration is weakly associated with global DNA methylation level. However, this association was observed more clearly in individuals with the minor allele of the MTHFR missense SNP rs1801133. By elucidating the complex mechanism of the regulation of DNA methylation by both acquired and genetic factors, our results may be important for cancer prevention.
Collapse
|
44
|
Jones MJ, Moore SR, Kobor MS. Principles and Challenges of Applying Epigenetic Epidemiology to Psychology. Annu Rev Psychol 2018; 69:459-485. [DOI: 10.1146/annurev-psych-122414-033653] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Meaghan J. Jones
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital, Vancouver, British Columbia V6H 3N1, Canada;, ,
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V6H 3N1, Canada
| | - Sarah R. Moore
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital, Vancouver, British Columbia V6H 3N1, Canada;, ,
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V6H 3N1, Canada
| | - Michael S. Kobor
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital, Vancouver, British Columbia V6H 3N1, Canada;, ,
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V6H 3N1, Canada
- Human Early Learning Partnership, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| |
Collapse
|
45
|
Hao G, Youssef NA, Davis CL, Su S. The role of DNA methylation in the association between childhood adversity and cardiometabolic disease. Int J Cardiol 2017; 255:168-174. [PMID: 29288057 DOI: 10.1016/j.ijcard.2017.12.063] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 11/17/2017] [Accepted: 12/16/2017] [Indexed: 02/07/2023]
Abstract
Growing evidence suggests that adverse environmental stimuli, especially during sensitive periods in early life, may lead to cardiometabolic disease in later life. However, the underlying biological mechanisms remain a mystery. Recent studies inferred that epigenetic modifications are likely involved. We review recent studies, primarily focused on the findings from human studies, to indicate the role of DNA methylation in the associations between childhood adversity and cardiometabolic disease in adulthood. In particular, we focused on DNA methylation modifications in genes regulating the hypothalamus-pituitary-adrenal axis as well as the immune system.
Collapse
Affiliation(s)
- Guang Hao
- Department of Population Health Sciences, Medical College of Georgia, Augusta University, Augusta, GA, United States.
| | - Nagy A Youssef
- Department of Psychiatry & Health Behavior, Medical College of Georgia, Augusta University, Augusta, GA, United States.
| | - Catherine L Davis
- Department of Population Health Sciences, Georgia Prevention Institute, Medical College of Georgia, Augusta University, Augusta, GA, United States.
| | - Shaoyong Su
- Department of Population Health Sciences, Medical College of Georgia, Augusta University, Augusta, GA, United States.
| |
Collapse
|
46
|
Improta G, Ritter C, Pettinato A, Vasta V, Schrama D, Fraggetta F, Becker JC. MGMT promoter methylation status in Merkel cell carcinoma: in vitro versus invivo. J Cancer Res Clin Oncol 2017; 143:1489-1497. [PMID: 28405827 DOI: 10.1007/s00432-017-2413-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/30/2017] [Indexed: 12/17/2022]
Abstract
PURPOSE Expression of O6-methylguanine-DNA methyltransferase (MGMT) in Merkel cell carcinoma (MCC) is very variable; thus, we tested whether this may be due to differential methylation of the MGMT gene promoter. METHODS Quantitative analysis of MGMT mRNA and protein expression, as well as MGMT promoter methylation status, was performed in a series of tissue samples of MCC tumors, representing both primary and metastatic lesions, as well as in six MCC cell lines. RESULTS These analyses revealed a very heterogeneous MGMT mRNA and protein expression in MCC both in vivo and in vitro. However, neither the MGMT mRNA nor protein expression correlated with the sensitivity of MCC cell lines toward the alkylating agent dacarbazine in vitro. Notably, increased methylation at the promoter of the MGMT gene was observed in 2/6 (33%) of the MCC cell lines; however, MGMT promoter methylation was absent in all MCC tissue samples. According to our results, albeit aberrant methylation of MGMT gene promoter can be observed in in vitro propagated MCC cell lines, it seems to be absent or very rare in MCC lesions in situ. CONCLUSION Thus, the evaluation of this marker has no or only little significance for predicting response to therapy or for improving efficacy of demethylating agents in the treatment of MCC. Microenvironmental factors may play a role in explaining the different results between MCC cell lines and MCC samples.
Collapse
Affiliation(s)
- Giuseppina Improta
- Laboratory of Clinical Research and Advanced Diagnostics, IRCCS-CROB, Rionero in Vulture, Italy
| | - Cathrin Ritter
- Department of Translational Skin Cancer Research (tscr), University Hospital Essen, Universitätstraße 1, 45141, Essen, Germany
- German Cancer Consortium (DKTK), Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Angela Pettinato
- Department of Pathology, Azienda Ospedaliera per l' Emergenza Cannizzaro, Catania, Italy
| | - Valeria Vasta
- Department of Pathology, Azienda Ospedaliera per l' Emergenza Cannizzaro, Catania, Italy
| | - David Schrama
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Filippo Fraggetta
- Department of Pathology, Azienda Ospedaliera per l' Emergenza Cannizzaro, Catania, Italy
| | - Jürgen C Becker
- Department of Translational Skin Cancer Research (tscr), University Hospital Essen, Universitätstraße 1, 45141, Essen, Germany.
- German Cancer Consortium (DKTK), Essen, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
47
|
Li J, Zhu X, Yu K, Jiang H, Zhang Y, Deng S, Cheng L, Liu X, Zhong J, Zhang X, He M, Chen W, Yuan J, Gao M, Bai Y, Han X, Liu B, Luo X, Mei W, He X, Sun S, Zhang L, Zeng H, Sun H, Liu C, Guo Y, Zhang B, Zhang Z, Huang J, Pan A, Yuan Y, Angileri F, Ming B, Zheng F, Zeng Q, Mao X, Peng Y, Mao Y, He P, Wang QK, Qi L, Hu FB, Liang L, Wu T. Genome-Wide Analysis of DNA Methylation and Acute Coronary Syndrome. Circ Res 2017; 120:1754-1767. [DOI: 10.1161/circresaha.116.310324] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 03/22/2017] [Accepted: 03/27/2017] [Indexed: 12/17/2022]
Abstract
Rationale:
Acute coronary syndrome (ACS) is a leading cause of death worldwide. Immune functions play a vital role in ACS development; however, whether epigenetic modulation contributes to the regulation of blood immune cells in this disease has not been investigated.
Objective:
We conducted an epigenome-wide analysis with circulating immune cells to identify differentially methylated genes in ACS.
Methods and Results:
We examined genome-wide methylation of whole blood in 102 ACS patients and 101 controls using HumanMethylation450 array, and externally replicated significant discoveries in 100 patients and 102 controls. For the replicated loci, we further analyzed their association with ACS in 6 purified leukocyte subsets, their correlation with the expressions of annotated genes, and their association with cardiovascular traits/risk factors. We found novel and reproducible association of ACS with blood methylation at 47 cytosine-phosphoguanine sites (discovery: false discovery rate <0.005; replication: Bonferroni corrected
P
<0.05). The association of methylation levels at these cytosine-phosphoguanine sites with ACS was further validated in at least 1 of the 6 leukocyte subsets, with predominant contributions from CD8
+
T cells, CD4
+
T cells, and B cells. Blood methylation of 26 replicated cytosine-phosphoguanine sites showed significant correlation with expressions of annotated genes (including
IL6R
,
FASLG
, and
CCL18
;
P
<5.9×10
−4
), and differential gene expression in case versus controls corroborated the observed differential methylation. The replicated loci suggested a role in ACS-relevant functions including chemotaxis, coronary thrombosis, and T-cell–mediated cytotoxicity. Functional analysis using the top ACS-associated methylation loci in purified T and B cells revealed vital pathways related to atherogenic signaling and adaptive immune response. Furthermore, we observed a significant enrichment of the replicated cytosine-phosphoguanine sites associated with smoking and low-density lipoprotein cholesterol (
P
enrichment
≤1×10
−5
).
Conclusions:
Our study identified novel blood methylation alterations associated with ACS and provided potential clinical biomarkers and therapeutic targets. Our results may suggest that immune signaling and cellular functions might be regulated at an epigenetic level in ACS.
Collapse
Affiliation(s)
- Jun Li
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Xiaoyan Zhu
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Kuai Yu
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Haijing Jiang
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Yizhi Zhang
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Siyun Deng
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Longxian Cheng
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Xuezhen Liu
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Jia Zhong
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Xiaomin Zhang
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Meian He
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Weihong Chen
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Jing Yuan
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Ming Gao
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Yansen Bai
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Xu Han
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Bing Liu
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Xiaoting Luo
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Wenhua Mei
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Xiaosheng He
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Shunchang Sun
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Liyun Zhang
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Hesong Zeng
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Huizhen Sun
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Chuanyao Liu
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Yanjun Guo
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Bing Zhang
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Zhihong Zhang
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Jinyan Huang
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - An Pan
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Yu Yuan
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Francesca Angileri
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Bingxia Ming
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Fang Zheng
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Qiutang Zeng
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Xiaobo Mao
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Yudong Peng
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Yi Mao
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Ping He
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Qing K. Wang
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Lu Qi
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Frank B. Hu
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Liming Liang
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| | - Tangchun Wu
- From the Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (J.L., X. Zhu, K.Y., H.J., Y.Z., S.D., X. Liu, X. Zhang, M.H., W.C., J.Y., Y.B., X. Han, B.L., X. He, H.S., C.L., Y.G., B.Z., Z.Z., A.P., Y.Y., F.A., T.W.); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (J.L., L.Q., F.B
| |
Collapse
|
48
|
Epigenetic changes in blood leukocytes following an omega-3 fatty acid supplementation. Clin Epigenetics 2017; 9:43. [PMID: 28450971 PMCID: PMC5405524 DOI: 10.1186/s13148-017-0345-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/14/2017] [Indexed: 12/24/2022] Open
Abstract
Background Omega-3 polyunsaturated fatty acids (n-3 FAs) have several beneficial effects on cardiovascular (CV) disease risk factors. These effects on CV risk profile may be mediated by several factors, including epigenetic modifications. Our objective is to investigate, using genome-wide DNA methylation analyses, methylation changes following an n-3 FA supplementation in overweight and obese subjects and to identify specific biological pathways potentially altered by the supplementation. Results Blood leukocytes genome-wide DNA methylation profiles of 36 overweight and obese subjects before and after a 6-week supplementation with 3 g of n-3 FAs were compared using GenomeStudio software. After supplementation, 308 CpG sites, assigned to 231 genes, were differentially methylated (FDR-corrected Diffscore ≥│13│~ P ≤ 0.05). Using Ingenuity Pathway Analysis system, a total of 55 pathways were significantly overrepresented following supplementation. Among these pathways, 16 were related to inflammatory and immune response, lipid metabolism, type 2 diabetes, and cardiovascular signaling. Changes in methylation levels of CpG sites within AKT3, ATF1, HDAC4, and IGFBP5 were correlated with changes in plasma triglyceride and glucose levels as well as with changes in the ratio of total cholesterol/HDL-cholesterol following the supplementation. Conclusions These data provide key differences in blood leukocytes DNA methylation profiles of subjects following an n-3 FA supplementation, which brings new, potential insights on metabolic pathways underlying the effects of n-3 FAs on CV health. Electronic supplementary material The online version of this article (doi:10.1186/s13148-017-0345-3) contains supplementary material, which is available to authorized users.
Collapse
|
49
|
Colicino E, Wilson A, Frisardi MC, Prada D, Power MC, Hoxha M, Dioni L, Spiro A, Vokonas PS, Weisskopf MG, Schwartz JD, Baccarelli AA. Telomere Length, Long-Term Black Carbon Exposure, and Cognitive Function in a Cohort of Older Men: The VA Normative Aging Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2017; 125:76-81. [PMID: 27259001 PMCID: PMC5226701 DOI: 10.1289/ehp241] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 09/21/2015] [Accepted: 05/11/2016] [Indexed: 05/03/2023]
Abstract
BACKGROUND Long-term air pollution exposure has been associated with age-related cognitive impairment, possibly because of enhanced inflammation. Leukocytes with longer telomere length (TL) are more responsive to inflammatory stimuli, yet TL has not been evaluated in relation to air pollution and cognition. OBJECTIVES We assessed whether TL modifies the association of 1-year exposure to black carbon (BC), a marker of traffic-related air pollution, with cognitive function in older men, and we examined whether this modification is independent of age and of C-reactive protein (CRP), a marker of inflammation. METHODS Between 1999 and 2007, we conducted 1-3 cognitive examinations of 428 older men in the Veterans Affairs (VA) Normative Aging Study. We used covariate-adjusted repeated-measure logistic regression to estimate associations of 1-year BC exposure with relative odds of being a low scorer (≤ 25) on the Mini-Mental State Examination (MMSE), which is a proxy of poor cognition. Confounders included age, CRP, and lifestyle and sociodemographic factors. RESULTS Each doubling in BC level was associated with 1.57 (95% CI: 1.20, 2.05) times higher odds of low MMSE scores. The BC-MMSE association was greater only among individuals with longer blood TL (5th quintile) (OR = 3.23; 95% CI: 1.37, 7.59; p = 0.04 for BC-by-TL-interaction). TL and CRP were associated neither with each other nor with MMSE. However, CRP modified the BC-MMSE relationship, with stronger associations only at higher CRP (5th quintile) and reference TL level (1st quintile) (OR = 2.68; 95% CI: 1.06, 6.79; p = 0.04 for BC-by-CRP-interaction). CONCLUSIONS TL and CRP levels may help predict the impact of BC exposure on cognitive function in older men. Citation: Colicino E, Wilson A, Frisardi MC, Prada D, Power MC, Hoxha M, Dioni L, Spiro A III, Vokonas PS, Weisskopf MG, Schwartz JD, Baccarelli AA. 2017. Telomere length, long-term black carbon exposure, and cognitive function in a cohort of older men: the VA Normative Aging Study. Environ Health Perspect 125:76-81; http://dx.doi.org/10.1289/EHP241.
Collapse
Affiliation(s)
- Elena Colicino
- Department of Environmental Health, and
- Address correspondence to E. Colicino, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave., Building 1, Room G03, Boston, MA 02115 USA. Telephone: (617) 432-1979. E-mail:
| | - Ander Wilson
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | | | - Diddier Prada
- Department of Environmental Health, and
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología–Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Melinda C. Power
- Department of Environmental Health, and
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Mirjam Hoxha
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Epidemiology Unit, Department of Preventive Medicine, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Laura Dioni
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Epidemiology Unit, Department of Preventive Medicine, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Avron Spiro
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Pantel S. Vokonas
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, USA
| | | | | | | |
Collapse
|
50
|
Dai L, Mehta A, Mordukhovich I, Just AC, Shen J, Hou L, Koutrakis P, Sparrow D, Vokonas PS, Baccarelli AA, Schwartz JD. Differential DNA methylation and PM 2.5 species in a 450K epigenome-wide association study. Epigenetics 2016; 12:139-148. [PMID: 27982729 DOI: 10.1080/15592294.2016.1271853] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Although there is growing evidence that exposure to ambient particulate matter is associated with global DNA methylation and gene-specific methylation, little is known regarding epigenome-wide changes in DNA methylation in relation to particles and, especially, particle components. Using the Illumina Infinium HumanMethylation450 BeadChip, we examined the relationship between one-year moving averages of PM2.5 species (Al, Ca, Cu, Fe, K, Na, Ni, S, Si, V, and Zn) and DNA methylation at 484,613 CpG probes in a longitudinal cohort that included 646 subjects. Bonferroni correction was applied to adjust for multiple comparisons. Bioinformatics analysis of the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment was also performed. We observed 20 Bonferroni significant (P-value < 9.4× 10-9) CpGs for Fe, 8 for Ni, and 1 for V. Particularly, methylation at Schlafen Family Member 11 (SLFN11) cg10911913 was positively associated with measured levels of all 3 species. The SLFN11 gene codes for an interferon-induced protein that inhibits retroviruses and sensitizes cancer cells to DNA-damaging agents. Bioinformatics analysis suggests that gene targets may be relevant to pathways including cancers, signal transduction, and cell growth and death. Ours is the first study to examine the epigenome-wide association between ambient particles species and DNA methylation. We found that long-term exposures to specific components of ambient particle pollution, especially particles emitted during oil combustion, were associated with methylation changes in genes relevant to immune responses. Our findings provide insight into potential biologic mechanisms on an epigenetic level.
Collapse
Affiliation(s)
- Lingzhen Dai
- a Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Amar Mehta
- a Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Irina Mordukhovich
- a Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Allan C Just
- b Department of Preventive Medicine , Icahn School of Medicine at Mount Sinai , New York , NY , USA
| | - Jincheng Shen
- c Department of Biostatistics , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Lifang Hou
- d Department of Preventive Medicine , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Petros Koutrakis
- a Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - David Sparrow
- e Veterans Affairs Normative Aging Study, Veterans Affairs Boston Healthcare System , Department of Medicine, Boston University School of Medicine , Boston , MA , USA
| | - Pantel S Vokonas
- e Veterans Affairs Normative Aging Study, Veterans Affairs Boston Healthcare System , Department of Medicine, Boston University School of Medicine , Boston , MA , USA
| | - Andrea A Baccarelli
- a Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Joel D Schwartz
- a Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| |
Collapse
|