1
|
Maimaiti A, Ma J, Hao C, Han D, Wang Y, Wang Z, Abudusalamu R. DNA methylation-estimated phenotypes, telomere length and risk of ischemic stroke: epigenetic age acceleration of screening and a Mendelian randomization study. Aging (Albany NY) 2024; 16:11970-11993. [PMID: 39159130 PMCID: PMC11386914 DOI: 10.18632/aging.206072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 07/16/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND Aging is a complex biological process that may be accelerated in certain pathological conditions. DNA methylation age (DNAmAge) has emerged as a biomarker for biological age, which can differ from chronological age. This research peels back the layers of the relationship between fast-forward aging and ischemic stroke, poking and prodding the potential two-way causal influences between stroke and biological aging indicators. METHODS We analyzed a cohort of ischemic stroke patients, comparing DNAmAge with chronological age to measure age acceleration. We assessed variations in age acceleration among stroke subtypes and between sexes. Using Mendelian randomization, we examined the causal links between stroke, aging biomarkers like telomere length, and age acceleration's effect on stroke risk. RESULTS Our investigation reveals a pronounced association between ischemic stroke and age acceleration, most notably in patients with cardioembolic strokes, who exhibited a striking median difference of 9 years between DNAmAge and chronological age. Furthermore, age acceleration differed significantly across stroke subtypes and was higher in women than in men. In terms of causality, MR analysis indicated a modest negative effect of stroke on telomere length, but no causal effect of age phenotypes on stroke or its subtypes. However, some indication of a potential causal effect of ischemic stroke on PhenoAge acceleration was observed. CONCLUSION The study provides insight into the relationship between DNAmAge and ischemic stroke, particularly cardioembolic stroke, and suggests possible gender differences. These insights carry profound clinical significance and set stage for future investigations into the entwined pathways of stroke and accelerated aging.
Collapse
Affiliation(s)
- Aierpati Maimaiti
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Jianhua Ma
- Department of Neurology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Chenguang Hao
- Department of Neurology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Dengfeng Han
- Department of Neurology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Yongxin Wang
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Zengliang Wang
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Rena Abudusalamu
- Department of Neurology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| |
Collapse
|
2
|
Singh A, Bhatt KS, Nguyen HC, Frisbee JC, Singh KK. Endothelial-to-Mesenchymal Transition in Cardiovascular Pathophysiology. Int J Mol Sci 2024; 25:6180. [PMID: 38892367 PMCID: PMC11173124 DOI: 10.3390/ijms25116180] [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: 05/10/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Under different pathophysiological conditions, endothelial cells lose endothelial phenotype and gain mesenchymal cell-like phenotype via a process known as endothelial-to-mesenchymal transition (EndMT). At the molecular level, endothelial cells lose the expression of endothelial cell-specific markers such as CD31/platelet-endothelial cell adhesion molecule, von Willebrand factor, and vascular-endothelial cadherin and gain the expression of mesenchymal cell markers such as α-smooth muscle actin, N-cadherin, vimentin, fibroblast specific protein-1, and collagens. EndMT is induced by numerous different pathways triggered and modulated by multiple different and often redundant mechanisms in a context-dependent manner depending on the pathophysiological status of the cell. EndMT plays an essential role in embryonic development, particularly in atrioventricular valve development; however, EndMT is also implicated in the pathogenesis of several genetically determined and acquired diseases, including malignant, cardiovascular, inflammatory, and fibrotic disorders. Among cardiovascular diseases, aberrant EndMT is reported in atherosclerosis, pulmonary hypertension, valvular disease, fibroelastosis, and cardiac fibrosis. Accordingly, understanding the mechanisms behind the cause and/or effect of EndMT to eventually target EndMT appears to be a promising strategy for treating aberrant EndMT-associated diseases. However, this approach is limited by a lack of precise functional and molecular pathways, causes and/or effects, and a lack of robust animal models and human data about EndMT in different diseases. Here, we review different mechanisms in EndMT and the role of EndMT in various cardiovascular diseases.
Collapse
Affiliation(s)
- Aman Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (A.S.); (K.S.B.); (H.C.N.); (J.C.F.)
| | - Kriti S. Bhatt
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (A.S.); (K.S.B.); (H.C.N.); (J.C.F.)
| | - Hien C. Nguyen
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (A.S.); (K.S.B.); (H.C.N.); (J.C.F.)
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Jefferson C. Frisbee
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (A.S.); (K.S.B.); (H.C.N.); (J.C.F.)
| | - Krishna K. Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (A.S.); (K.S.B.); (H.C.N.); (J.C.F.)
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
3
|
Porter GA. Environmental Signals. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:397-416. [PMID: 38884722 DOI: 10.1007/978-3-031-44087-8_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Environmental factors have long been known to play a role in the pathogenesis of congenital heart disease (CHD), but this has not been a major focus of research in the modern era. Studies of human exposures and animal models demonstrate that demographics (age, race, socioeconomic status), diseases (e.g., diabetes, hypertension, obesity, stress, infection, high altitude), recreational and therapeutic drug use, and chemical exposures are associated with an increased risk for CHD. Unfortunately, although studies suggest that exposures to these factors may cause CHD, in most cases, the data are not strong, are inconclusive, or are contradictory. Although most studies concentrate on the effects of maternal exposure, paternal exposure to some agents can also modify this risk. From a mechanistic standpoint, recent delineation of signaling and genetic controls of cardiac development has revealed molecular pathways that may explain the effects of environmental signals on cardiac morphogenesis and may provide further tools to study the effects of environmental stimuli on cardiac development. For example, environmental factors likely regulate cellular signaling pathways, transcriptional and epigenetic regulation, proliferation, and physiologic processes that can control the development of the heart and other organs. However, understanding of the epidemiology and risk of these exposures and the mechanistic basis for any effects on cardiac development remains incomplete. Further studies defining the relationship between environmental exposures and human CHD and the mechanisms involved should reveal strategies to prevent, diagnose, and treat CHD induced by environmental signals.
Collapse
Affiliation(s)
- George A Porter
- Departments of Pediatrics (Cardiology), Pharmacology and Physiology, and Medicine (Aab Cardiovascular Research Institute), University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
4
|
Geiger M, Gorica E, Mohammed SA, Mongelli A, Mengozi A, Delfine V, Ruschitzka F, Costantino S, Paneni F. Epigenetic Network in Immunometabolic Disease. Adv Biol (Weinh) 2024; 8:e2300211. [PMID: 37794610 DOI: 10.1002/adbi.202300211] [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: 06/08/2023] [Revised: 09/08/2023] [Indexed: 10/06/2023]
Abstract
Although a large amount of data consistently shows that genes affect immunometabolic characteristics and outcomes, epigenetic mechanisms are also heavily implicated. Epigenetic changes, including DNA methylation, histone modification, and noncoding RNA, determine gene activity by altering the accessibility of chromatin to transcription factors. Various factors influence these alterations, including genetics, lifestyle, and environmental cues. Moreover, acquired epigenetic signals can be transmitted across generations, thus contributing to early disease traits in the offspring. A closer investigation is critical in this aspect as it can help to understand the underlying molecular mechanisms further and gain insights into potential therapeutic targets for preventing and treating diseases arising from immuno-metabolic dysregulation. In this review, the role of chromatin alterations in the transcriptional modulation of genes involved in insulin resistance, systemic inflammation, macrophage polarization, endothelial dysfunction, metabolic cardiomyopathy, and nonalcoholic fatty liver disease (NAFLD), is discussed. An overview of emerging chromatin-modifying drugs and the importance of the individual epigenetic profile for personalized therapeutic approaches in patients with immuno-metabolic disorders is also presented.
Collapse
Affiliation(s)
- Martin Geiger
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Era Gorica
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Shafeeq Ahmed Mohammed
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Alessia Mongelli
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Alessandro Mengozi
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Valentina Delfine
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- University Heart Center, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- University Heart Center, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- Department of Research and Education, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| |
Collapse
|
5
|
Gladwell LR, Ahiarah C, Rasheed S, Rahman SM, Choudhury M. Traditional Therapeutics and Potential Epidrugs for CVD: Why Not Both? Life (Basel) 2023; 14:23. [PMID: 38255639 PMCID: PMC10820772 DOI: 10.3390/life14010023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide. In addition to the high mortality rate, people suffering from CVD often endure difficulties with physical activities and productivity that significantly affect their quality of life. The high prevalence of debilitating risk factors such as obesity, type 2 diabetes mellitus, smoking, hypertension, and hyperlipidemia only predicts a bleak future. Current traditional CVD interventions offer temporary respite; however, they compound the severe economic strain of health-related expenditures. Furthermore, these therapeutics can be prescribed indefinitely. Recent advances in the field of epigenetics have generated new treatment options by confronting CVD at an epigenetic level. This involves modulating gene expression by altering the organization of our genome rather than altering the DNA sequence itself. Epigenetic changes are heritable, reversible, and influenced by environmental factors such as medications. As CVD is physiologically and pathologically diverse in nature, epigenetic interventions can offer a ray of hope to replace or be combined with traditional therapeutics to provide the prospect of addressing more than just the symptoms of CVD. This review discusses various risk factors contributing to CVD, perspectives of current traditional medications in practice, and a focus on potential epigenetic therapeutics to be used as alternatives.
Collapse
Affiliation(s)
- Lauren Rae Gladwell
- Department of Pharmaceutical Sciences, Texas A&M Irma Lerma Rangel College of Pharmacy, 1114 TAMU, College Station, TX 77843, USA
| | - Chidinma Ahiarah
- Department of Pharmaceutical Sciences, Texas A&M Irma Lerma Rangel College of Pharmacy, 1114 TAMU, College Station, TX 77843, USA
| | - Shireen Rasheed
- Department of Pharmaceutical Sciences, Texas A&M Irma Lerma Rangel College of Pharmacy, 1114 TAMU, College Station, TX 77843, USA
| | - Shaikh Mizanoor Rahman
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al-Mouz, Nizwa 616, Oman
| | - Mahua Choudhury
- Department of Pharmaceutical Sciences, Texas A&M Irma Lerma Rangel College of Pharmacy, 1114 TAMU, College Station, TX 77843, USA
| |
Collapse
|
6
|
Dowaidar M. Gene-environment interactions that influence CVD, lipid traits, obesity, diabetes, and hypertension appear to be able to influence gene therapy. Mol Aspects Med 2023; 94:101213. [PMID: 37703607 DOI: 10.1016/j.mam.2023.101213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023]
Abstract
Most mind boggling diseases are accepted to be impacted by both genetic and environmental elements. As of late, there has been a flood in the improvement of different methodologies, concentrate on plans, and measurable and logical techniques to examine gene-environment cooperations (G × Es) in enormous scope studies including human populaces. The many-sided exchange between genetic elements and environmental openings has long charmed the consideration of clinicians and researchers looking to grasp the complicated starting points of diseases. While single variables can add to disease, the blend of genetic variations and environmental openings frequently decides disease risk. The fundamental point of this paper is to talk about the Gene-Environment Associations That Impact CVD, Lipid Characteristics, Obesity, Diabetes, and Hypertension Have all the earmarks of being Ready to Impact Gene Therapy. This survey paper investigates the meaning of gene-environment collaborations (G × E) in disease advancement. The intricacy of genetic and environmental communications in disease causation is explained, underlining the multifactorial idea of many circumstances. The job of gene-environment cooperations in cardiovascular disease, lipid digestion, diabetes, obesity, and hypertension is investigated. This audit fixates on Gene by Environment (G × E) collaborations, investigating their importance in disease etiology.
Collapse
Affiliation(s)
- Moataz Dowaidar
- Department of Bioengineering, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261, Saudi Arabia; Interdisciplinary Research Center for Hydrogen and Energy Storage (IRC-HES), King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261, Saudi Arabia; Interdisciplinary Research Center for Health & Biosciences, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261, Saudi Arabia.
| |
Collapse
|
7
|
Bergonzini M, Loreni F, Lio A, Russo M, Saitto G, Cammardella A, Irace F, Tramontin C, Chello M, Lusini M, Nenna A, Ferrisi C, Ranocchi F, Musumeci F. Panoramic on Epigenetics in Coronary Artery Disease and the Approach of Personalized Medicine. Biomedicines 2023; 11:2864. [PMID: 37893238 PMCID: PMC10604795 DOI: 10.3390/biomedicines11102864] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/02/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Epigenetic modifications play a fundamental role in the progression of coronary artery disease (CAD). This panoramic review aims to provide an overview of the current understanding of the epigenetic mechanisms involved in CAD pathogenesis and highlights the potential implications for personalized medicine approaches. Epigenetics is the study of heritable changes that do not influence alterations in the DNA sequence of the genome. It has been shown that epigenetic processes, including DNA/histone methylation, acetylation, and phosphorylation, play an important role. Additionally, miRNAs, lncRNAs, and circRNAs are also involved in epigenetics, regulating gene expression patterns in response to various environmental factors and lifestyle choices. In the context of CAD, epigenetic alterations contribute to the dysregulation of genes involved in inflammation, oxidative stress, lipid metabolism, and vascular function. These epigenetic changes can occur during early developmental stages and persist throughout life, predisposing individuals to an increased risk of CAD. Furthermore, in recent years, the concept of personalized medicine has gained significant attention. Personalized medicine aims to tailor medical interventions based on an individual's unique genetic, epigenetic, environmental, and lifestyle factors. In the context of CAD, understanding the interplay between genetic variants and epigenetic modifications holds promise for the development of more precise diagnostic tools, risk stratification models, and targeted therapies. This review summarizes the current knowledge of epigenetic mechanisms in CAD and discusses the fundamental principles of personalized medicine.
Collapse
Affiliation(s)
- Marcello Bergonzini
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Francesco Loreni
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Antonio Lio
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Marco Russo
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Guglielmo Saitto
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Antonio Cammardella
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Francesco Irace
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Corrado Tramontin
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Massimo Chello
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Mario Lusini
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Antonio Nenna
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Chiara Ferrisi
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Federico Ranocchi
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Francesco Musumeci
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, 00152 Rome, Italy
| |
Collapse
|
8
|
Shetty SS, D D, S H, Sonkusare S, Naik PB, Kumari N S, Madhyastha H. Environmental pollutants and their effects on human health. Heliyon 2023; 9:e19496. [PMID: 37662771 PMCID: PMC10472068 DOI: 10.1016/j.heliyon.2023.e19496] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/05/2023] Open
Abstract
Numerous environmental contaminants significantly contribute to human disease, affecting climate change and public and individual health, resulting in increased mortality and morbidity. Because of the scarcity of information regarding pollution exposure from less developed nations with inadequate waste management, higher levels of poverty, and limited adoption of new technology, the relationship between pollutants and health effects needs to be investigated more. A similar situation is present in many developed countries, where solutions are only discovered after the harm has already been done and the necessity for safeguards has subsided. The connection between environmental toxins and health needs to be better understood due to difficulties in quantifying exposure levels and a lack of systematic monitoring. Different pollutants are to blame for both chronic and acute disorders. Additionally, research becomes challenging when disease problems are seen after prolonged exposure. This review aims to discuss the present understanding of the association between environmental toxins and human health in bridging this knowledge gap. The genesis of cancer and the impact of various environmental pollutants on the human body's cardiovascular, respiratory, reproductive, prenatal, and neural health are discussed in this overview.
Collapse
Affiliation(s)
- Shilpa S. Shetty
- CentralResearch laboratory, Cellomics Laboratory, K. S. Hegde Medical Academy, Nitte (Deemed to be University), Mangaluru., Karnataka, India
| | - Deepthi D
- CentralResearch laboratory, Cellomics Laboratory, K. S. Hegde Medical Academy, Nitte (Deemed to be University), Mangaluru., Karnataka, India
| | - Harshitha S
- CentralResearch laboratory, Cellomics Laboratory, K. S. Hegde Medical Academy, Nitte (Deemed to be University), Mangaluru., Karnataka, India
| | - Shipra Sonkusare
- Department of Obstetrics and Gynecology, K. S. Hegde Medical Academy, Mangaluru, 576018, Karnataka, India
| | - Prashanth B. Naik
- Department of Pediatrics, K. S. Hegde Medical Academy, Mangaluru, 576018, Karnataka, India
| | - Suchetha Kumari N
- CentralResearch laboratory, Cellomics Laboratory, K. S. Hegde Medical Academy, Nitte (Deemed to be University), Mangaluru., Karnataka, India
- Department of Biochemistry, K. S. Hegde Medical Academy, Mangaluru, 576018, Karnataka, India
| | - Harishkumar Madhyastha
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| |
Collapse
|
9
|
Martino F, Bassareo PP, Martino E, Romeo F, Calcaterra G, Perrone Filardi P, Indolfi C, Nodari S, Montemurro V, Guccione P, Salvo GD, Chessa M, Pedrinelli R, Mercuro G, Barillà F. Cardiovascular prevention in childhood: a consensus document of the Italian Society of Cardiology Working Group on Congenital Heart Disease and Cardiovascular Prevention in Paediatric Age. J Cardiovasc Med (Hagerstown) 2023; 24:492-505. [PMID: 37409595 DOI: 10.2459/jcm.0000000000001488] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
Cardiovascular diseases (CVD) may be manifested from a very early age. Genetic and environmental (epigenetic) factors interact to affect development and give rise to an abnormal phenotypical expression of genetic information, although not eliciting changes in the nucleotide sequence of DNA. It has been scientifically proven that increased oxidative stress (OS) caused by disease (overweight, obesity, diabetes), nutritional imbalances, unhealthy lifestyles (smoking, alcohol, substance abuse) in the mother during pregnancy may induce placental dysfunction, intrauterine growth restriction, prematurity, low birth weight, postnatal adiposity rebound, metabolic alterations and consequent onset of traditional cardiovascular risk factors. OS represents the cornerstone in the onset of atherosclerosis and manifestation of CVD following an extended asymptomatic period. OS activates platelets and monocytes eliciting the release of pro-inflammatory, pro-atherogenic and pro-oxidising substances resulting in endothelial dysfunction, decrease in flow-mediated arterial dilatation and increase in carotid intima-media thickness. The prevention of CVD is defined as primordial (aimed at preventing risk factors development), primary (aimed at early identification and treatment of risk factors), secondary (aimed at reducing risk of future events in patients who have already manifested a cardiovascular event), and tertiary (aimed at limiting the complex outcome of disease). Atherosclerosis prevention should be implemented as early as possible. Appropriate screening should be carried out to identify children at high risk who are apparently healthy and implement measures including dietary and lifestyle changes, addition of nutritional supplements and, lastly, pharmacological treatment if risk profiles fail to normalise. Reinstating endothelial function during the reversible stage of atherosclerosis is crucial.
Collapse
Affiliation(s)
- Francesco Martino
- Department of Internal Clinical, Anesthesiological and Cardiovascular Sciences, La Sapienza University, Rome, Italy
| | - Pier Paolo Bassareo
- University College of Dublin, School of Medicine, Mater Misericordiae University Hospital and Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Eliana Martino
- Department of Internal Clinical, Anesthesiological and Cardiovascular Sciences, La Sapienza University, Rome, Italy
| | | | | | | | - Ciro Indolfi
- Division of Cardiology, Research Centre for Cardiovascular Diseases, Magna Graecia University, Catanzaro
| | - Savina Nodari
- Department of Medical Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, ASST Spedali Civili, Brescia
| | | | - Paolo Guccione
- Department of Cardiology, Cardiac Surgery, Cardio-pulmonary Transplantation, IRCCS Bambino Gesu'Paediatric Hospital, Rome
| | - Giovanni Di Salvo
- Division of Paediatric Cardiology, Department of Women's and Children's Health, University of Padua, Padua
| | - Massimo Chessa
- ACHD UNIT, Pediatric and Adult Congenital Heart Centre, IRCCS-Policlinico San Donato, San Donato Milanese, Vita Salute San Raffaele University, Milan
| | - Roberto Pedrinelli
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa
| | | | | |
Collapse
|
10
|
Skouras AZ, Antonakis-Karamintzas D, Tsantes AG, Triantafyllou A, Papagiannis G, Tsolakis C, Koulouvaris P. The Acute and Chronic Effects of Resistance and Aerobic Exercise in Hemostatic Balance: A Brief Review. Sports (Basel) 2023; 11:sports11040074. [PMID: 37104148 PMCID: PMC10143125 DOI: 10.3390/sports11040074] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Hemostatic balance refers to the dynamic balance between blood clot formation (coagulation), blood clot dissolution (fibrinolysis), anticoagulation, and innate immunity. Although regular habitual exercise may lower the incidence of cardiovascular diseases (CVD) by improving an individual’s hemostatic profile at rest and during exertion, vigorous exercise may increase the risk of sudden cardiac death and venous thromboembolism (VTE). This literature review aims to investigate the hemostatic system’s acute and chronic adaptive responses to different types of exercise in healthy and patient populations. Compared to athletes, sedentary healthy individuals demonstrate similar post-exercise responses in platelet function and coagulatory and fibrinolytic potential. However, hemostatic adaptations of patients with chronic diseases in regular training is a promising field. Despite the increased risk of thrombotic events during an acute bout of vigorous exercise, regular exposure to high-intensity exercise might desensitize exercise-induced platelet aggregation, moderate coagulatory parameters, and up-regulate fibrinolytic potential via increasing tissue plasminogen activator (tPA) and decreasing plasminogen activator inhibitor (PAI-1) response. Future research might focus on combining different types of exercise, manipulating each training characteristic (frequency, intensity, time, and volume), or investigating the minimal exercise dosage required to maintain hemostatic balance, especially in patients with various health conditions.
Collapse
|
11
|
Holliday KM, Gondalia R, Baldassari A, Justice AE, Stewart JD, Liao D, Yanosky JD, Jordahl KM, Bhatti P, Assimes TL, Pankow JS, Guan W, Fornage M, Bressler J, North KE, Conneely KN, Li Y, Hou L, Vokonas PS, Ward-Caviness CK, Wilson R, Wolf K, Waldenberger M, Cyrys J, Peters A, Boezen HM, Vonk JM, Sayols-Baixeras S, Lee M, Baccarelli AA, Whitsel EA. Gaseous air pollutants and DNA methylation in a methylome-wide association study of an ethnically and environmentally diverse population of U.S. adults. ENVIRONMENTAL RESEARCH 2022; 212:113360. [PMID: 35500859 PMCID: PMC9354583 DOI: 10.1016/j.envres.2022.113360] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 06/03/2023]
Abstract
Epigenetic mechanisms may underlie air pollution-health outcome associations. We estimated gaseous air pollutant-DNA methylation (DNAm) associations using twelve subpopulations within Women's Health Initiative (WHI) and Atherosclerosis Risk in Communities (ARIC) cohorts (n = 8397; mean age 61.3 years; 83% female; 46% African-American, 46% European-American, 8% Hispanic/Latino). We used geocoded participant address-specific mean ambient carbon monoxide (CO), nitrogen oxides (NO2; NOx), ozone (O3), and sulfur dioxide (SO2) concentrations estimated over the 2-, 7-, 28-, and 365-day periods before collection of blood samples used to generate Illumina 450 k array leukocyte DNAm measurements. We estimated methylome-wide, subpopulation- and race/ethnicity-stratified pollutant-DNAm associations in multi-level, linear mixed-effects models adjusted for sociodemographic, behavioral, meteorological, and technical covariates. We combined stratum-specific estimates in inverse variance-weighted meta-analyses and characterized significant associations (false discovery rate; FDR<0.05) at Cytosine-phosphate-Guanine (CpG) sites without among-strata heterogeneity (PCochran's Q > 0.05). We attempted replication in the Cooperative Health Research in Region of Augsburg (KORA) study and Normative Aging Study (NAS). We observed a -0.3 (95% CI: -0.4, -0.2) unit decrease in percent DNAm per interquartile range (IQR, 7.3 ppb) increase in 28-day mean NO2 concentration at cg01885635 (chromosome 3; regulatory region 290 bp upstream from ZNF621; FDR = 0.03). At intragenic sites cg21849932 (chromosome 20; LIME1; intron 3) and cg05353869 (chromosome 11; KLHL35; exon 2), we observed a -0.3 (95% CI: -0.4, -0.2) unit decrease (FDR = 0.04) and a 1.2 (95% CI: 0.7, 1.7) unit increase (FDR = 0.04), respectively, in percent DNAm per IQR (17.6 ppb) increase in 7-day mean ozone concentration. Results were not fully replicated in KORA and NAS. We identified three CpG sites potentially susceptible to gaseous air pollution-induced DNAm changes near genes relevant for cardiovascular and lung disease. Further harmonized investigations with a range of gaseous pollutants and averaging durations are needed to determine the effect of gaseous air pollutants on DNA methylation and ultimately gene expression.
Collapse
Affiliation(s)
- Katelyn M Holliday
- Department of Family Medicine and Community Health, School of Medicine, Duke University, Durham, NC, USA; Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
| | - Rahul Gondalia
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Antoine Baldassari
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | | | - James D Stewart
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Duanping Liao
- Division of Epidemiology, Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Jeff D Yanosky
- Division of Epidemiology, Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Kristina M Jordahl
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | - Parveen Bhatti
- Cancer Control Research, BC Cancer, Vancouver, BC, Canada
| | | | - James S Pankow
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA
| | - Weihua Guan
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA; Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jan Bressler
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Karen N Conneely
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Yun Li
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA; Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA; Department of Computer Science, University of North Carolina, Chapel Hill, NC, USA
| | - Lifang Hou
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University Chicago, Evanston, IL, USA; Center for Population Epigenetics, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University Chicago, Evanston, IL, USA
| | - Pantel S Vokonas
- VA Normative Aging Study, VA Boston Healthcare System, Schools of Medicine and Public Health, Boston University, Boston, MA, USA
| | - Cavin K Ward-Caviness
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, 104 Mason Farm Rd, Chapel Hill, NC, 27514, USA
| | - Rory Wilson
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany; Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Kathrin Wolf
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany; Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Josef Cyrys
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany; Institute for Medical Information Processing, Biometry and Epidemiology, Medical Faculty, Ludwig Maximilians University, Munich, Germany
| | - H Marike Boezen
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, the Netherlands; University of Groningen, University Medical Center Groningen, GRIAC Research Institute, the Netherlands
| | - Judith M Vonk
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, the Netherlands; University of Groningen, University Medical Center Groningen, GRIAC Research Institute, the Netherlands
| | - Sergi Sayols-Baixeras
- Cardiovascular Epidemiology and Genetics Research Group, Hospital Del Mar Medical Research Institute (IMIM), Campus Del Mar, Universitat Pompeu Fabra, Barcelona, Spain; Consorcio CIBER, M.P. Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; Molecular Epidemiology and Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Mikyeong Lee
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Andrea A Baccarelli
- Laboratory of Environmental Epigenetics, Departments of Environmental Health Sciences and Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Eric A Whitsel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA; Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
12
|
Studying Epigenetics of Cardiovascular Diseases on Chip Guide. CARDIOGENETICS 2022. [DOI: 10.3390/cardiogenetics12030021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Epigenetics is defined as the study of inheritable changes in the gene expressions and phenotypes that occurs without altering the normal DNA sequence. These changes are mainly due to an alteration in chromatin or its packaging, which changes the DNA accessibility. DNA methylation, histone modification, and noncoding or microRNAs can best explain the mechanism of epigenetics. There are various DNA methylated enzymes, histone-modifying enzymes, and microRNAs involved in the cause of various CVDs (cardiovascular diseases) such as cardiac hypertrophy, heart failure, and hypertension. Moreover, various CVD risk factors such as diabetes mellitus, hypoxia, aging, dyslipidemia, and their epigenetics are also discussed together with CVDs such as CHD (coronary heart disease) and PAH (pulmonary arterial hypertension). Furthermore, different techniques involved in epigenetic chromatin mapping are explained. Among these techniques, the ChIP-on-chip guide is explained with regard to its role in cardiac hypertrophy, a final form of heart failure. This review focuses on different epigenetic factors that are involved in causing cardiovascular diseases.
Collapse
|
13
|
Zhang Q, Meng XH, Qiu C, Shen H, Zhao Q, Zhao LJ, Tian Q, Sun CQ, Deng HW. Integrative analysis of multi-omics data to detect the underlying molecular mechanisms for obesity in vivo in humans. Hum Genomics 2022; 16:15. [PMID: 35568907 PMCID: PMC9107154 DOI: 10.1186/s40246-022-00388-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 05/04/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Obesity is a complex, multifactorial condition in which genetic play an important role. Most of the systematic studies currently focuses on individual omics aspect and provide insightful yet limited knowledge about the comprehensive and complex crosstalk between various omics levels. SUBJECTS AND METHODS Therefore, we performed a most comprehensive trans-omics study with various omics data from 104 subjects, to identify interactions/networks and particularly causal regulatory relationships within and especially those between omic molecules with the purpose to discover molecular genetic mechanisms underlying obesity etiology in vivo in humans. RESULTS By applying differentially analysis, we identified 8 differentially expressed hub genes (DEHGs), 14 differentially methylated regions (DMRs) and 12 differentially accumulated metabolites (DAMs) for obesity individually. By integrating those multi-omics biomarkers using Mendelian Randomization (MR) and network MR analyses, we identified 18 causal pathways with mediation effect. For the 20 biomarkers involved in those 18 pairs, 17 biomarkers were implicated in the pathophysiology of obesity or related diseases. CONCLUSIONS The integration of trans-omics and MR analyses may provide us a holistic understanding of the underlying functional mechanisms, molecular regulatory information flow and the interactive molecular systems among different omic molecules for obesity risk and other complex diseases/traits.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Community Nursing, School of Nursing and Health, Zhengzhou University, High-Tech Development Zone of States, Zhengzhou, 450001, Henan, People's Republic of China
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Xiang-He Meng
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
- Center for System Biology, Data Sciences, and Reproductive Health, School of Basic Medical Science, Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Chuan Qiu
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Hui Shen
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Qi Zhao
- Department of Preventive Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Lan-Juan Zhao
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Qing Tian
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Chang-Qing Sun
- Department of Community Nursing, School of Nursing and Health, Zhengzhou University, High-Tech Development Zone of States, Zhengzhou, 450001, Henan, People's Republic of China
- Department of Social Medicine and Health Management, College of Public Health, Zhengzhou University, High-Tech Development Zone of States, Zhengzhou, 450001, Henan, People's Republic of China
| | - Hong-Wen Deng
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|
14
|
Gentry AE, Robins J, Makowski M, Kliewer W. Differential DNA Methylation and Cardiometabolic Risk in African American Mother-Adolescent Dyads. Biol Res Nurs 2022; 24:75-84. [PMID: 34719281 PMCID: PMC9248288 DOI: 10.1177/10998004211039017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Cardiovascular disease disproportionately affects African Americans as the leading cause of morbidity and mortality. Among African Americans, compared to other racial groups, cardiovascular disease onset occurs at an earlier age due to a higher prevalence of cardiometabolic risk factors, particularly obesity, hypertension and type 2 diabetes. Emerging evidence suggests that heritable epigenetic processes are related to increased cardiovascular disease risk, but this is largely unexplored in adolescents or across generations. MATERIALS AND METHODS In a cross-sectional descriptive pilot study in low-income African American mother-adolescent dyads, we examined associations between DNA methylation and the cardiometabolic indicators of body mass index, waist circumference, and insulin resistance. RESULTS Four adjacent cytosine and guanine nucleotides (CpG) sites were significantly differentially methylated and associated with C-reactive protein (CRP), 62 with waist circumference, and none to insulin resistance in models for both mothers and adolescents. CONCLUSION Further study of the relations among psychological and environmental stressors, indicators of cardiovascular disease, risk, and epigenetic factors will improve understanding of cardiovascular disease risk so that preventive measures can be instituted earlier and more effectively. To our knowledge this work is the first to examine DNA methylation and cardiometabolic risk outcomes in mother-adolescent dyads.
Collapse
Affiliation(s)
- Amanda Elswick Gentry
- Department of Psychiatry, Virginia
Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University,
Richmond, VA, USA,Amanda Elswick Gentry, PhD, Department of
Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia
Commonwealth University, 800 East Leigh Street, Suite 100, Room 130-B, Richmond,
VA 23219, USA.
| | - Jo Robins
- School of Nursing, Virginia
Commonwealth University, Richmond, VA, USA
| | | | - Wendy Kliewer
- Department of Psychology, College of
Humanities and Sciences, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
15
|
Ghosh AK. Acetyltransferase p300 Is a Putative Epidrug Target for Amelioration of Cellular Aging-Related Cardiovascular Disease. Cells 2021; 10:cells10112839. [PMID: 34831061 PMCID: PMC8616404 DOI: 10.3390/cells10112839] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 12/31/2022] Open
Abstract
Cardiovascular disease is the leading cause of accelerated as well as chronological aging-related human morbidity and mortality worldwide. Genetic, immunologic, unhealthy lifestyles including daily consumption of high-carb/high-fat fast food, lack of exercise, drug addiction, cigarette smoke, alcoholism, and exposure to environmental pollutants like particulate matter (PM)-induced stresses contribute profoundly to accelerated and chronological cardiovascular aging and associated life threatening diseases. All these stressors alter gene expression epigenetically either through activation or repression of gene transcription via alteration of chromatin remodeling enzymes and chromatin landscape by DNA methylation or histone methylation or histone acetylation. Acetyltransferase p300, a major epigenetic writer of acetylation on histones and transcription factors, contributes significantly to modifications of chromatin landscape of genes involved in cellular aging and cardiovascular diseases. In this review, the key findings those implicate acetyltransferase p300 as a major contributor to cellular senescence or aging related cardiovascular pathologies including vascular dysfunction, cardiac hypertrophy, myocardial infarction, cardiac fibrosis, systolic/diastolic dysfunction, and aortic valve calcification are discussed. The efficacy of natural or synthetic small molecule inhibitor targeting acetyltransferase p300 in amelioration of stress-induced dysregulated gene expression, cellular aging, and cardiovascular disease in preclinical study is also discussed.
Collapse
Affiliation(s)
- Asish K Ghosh
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
16
|
Sumi MP, Mahajan B, Sattar RSA, Nimisha, Apurva, Kumar A, Sharma AK, Ahmad E, Ali A, Saluja SS. Elucidation of Epigenetic Landscape in Coronary Artery Disease: A Review on Basic Concept to Personalized Medicine. Epigenet Insights 2021; 14:2516865720988567. [PMID: 33598635 PMCID: PMC7863167 DOI: 10.1177/2516865720988567] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/26/2020] [Indexed: 12/11/2022] Open
Abstract
Despite extensive clinical research and management protocols applied in the field of coronary artery diseases (CAD), it still holds the number 1 position in mortality worldwide. This indicates that we need to work on precision medicine to discover the diagnostic, therapeutic, and prognostic targets to improve the outcome of CAD. In precision medicine, epigenetic changes play a vital role in disease onset and progression. Epigenetics is the study of heritable changes that do not affect the alterations of DNA sequence in the genome. It comprises various covalent modifications that occur in DNA or histone proteins affecting the spatial arrangement of the DNA and histones. These multiple modifications include DNA/histone methylation, acetylation, phosphorylation, and SUMOylation. Besides these covalent modifications, non-coding RNAs-viz. miRNA, lncRNA, and circRNA are also involved in epigenetics. Smoking, alcohol, diet, environmental pollutants, obesity, and lifestyle are some of the prime factors affecting epigenetic alterations. Novel molecular techniques such as next-generation sequencing, chromatin immunoprecipitation, and mass spectrometry have been developed to identify important cross points in the epigenetic web in relation to various diseases. The studies regarding exploration of epigenetics, have led researchers to identify multiple diagnostic markers and therapeutic targets that are being used in different disease diagnosis and management. Here in this review, we will discuss various ground-breaking contributions of past and recent studies in the epigenetic field in concert with coronary artery diseases. Future prospects of epigenetics and its implication in CAD personalized medicine will also be discussed in brief.
Collapse
Affiliation(s)
- Mamta P Sumi
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research (GIPMER), University of Delhi, New Delhi, India
| | - Bhawna Mahajan
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research (GIPMER), University of Delhi, New Delhi, India
- Department of Biochemistry, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research (GIPMER), University of Delhi, New Delhi, India
| | - Real Sumayya Abdul Sattar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research (GIPMER), University of Delhi, New Delhi, India
| | - Nimisha
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research (GIPMER), University of Delhi, New Delhi, India
| | - Apurva
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research (GIPMER), University of Delhi, New Delhi, India
| | - Arun Kumar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research (GIPMER), University of Delhi, New Delhi, India
| | - Abhay Kumar Sharma
- Department of Biochemistry, All India Institute of Medical Science, Patna, Bihar, India
| | - Ejaz Ahmad
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research (GIPMER), University of Delhi, New Delhi, India
| | - Asgar Ali
- Department of Biochemistry, All India Institute of Medical Science, Patna, Bihar, India
| | - Sundeep Singh Saluja
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research (GIPMER), University of Delhi, New Delhi, India
| |
Collapse
|
17
|
Abstract
Emerging evidence suggests the growing importance of "nongenetic factors" in the pathogenesis of atherosclerotic vascular disease. Indeed, the inherited genome determines only part of the risk profile as genomic approaches do not take into account additional layers of biological regulation by "epi"-genetic changes. Epigenetic modifications are defined as plastic chemical changes of DNA/histone complexes which critically affect gene activity without altering the DNA sequence. These modifications include DNA methylation, histone posttranslational modifications, and non-coding RNAs and have the ability to modulate gene expression at both transcriptional and posttranscriptional level. Notably, epigenetic signals are mainly induced by environmental factors (i.e., pollution, smoking, noise) and, once acquired, may be transmitted to the offspring. The inheritance of adverse epigenetic changes may lead to premature deregulation of pathways involved in vascular damage and endothelial dysfunction. Here, we describe the emerging role of epigenetic modifications as fine-tuners of gene transcription in atherosclerosis. Specifically, the following aspects are described in detail: (1) discovery and impact of the epigenome in cardiovascular disease, (2) the epigenetic landscape in atherosclerosis; (3) inheritance of epigenetic signals and premature vascular disease; (4) epigenetic control of lipid metabolism, vascular oxidative stress, inflammation, autophagy, and apoptosis; (5) epigenetic biomarkers in patients with atherosclerosis; (6) novel therapeutic strategies to modulate epigenetic marks. Understanding the individual epigenetic profile may pave the way for new approaches to determine cardiovascular risk and to develop personalized therapies to treat atherosclerosis and its complications.
Collapse
|
18
|
Datta A, Alam MJ, Khaleda L, Al-Forkan M. Protective effects of Corchorus olitorius and Butea monosperma against Arsenic induced aberrant methylation and mitochondrial DNA damage in wistar rat model. Toxicol Rep 2021; 8:30-37. [PMID: 33391994 PMCID: PMC7772721 DOI: 10.1016/j.toxrep.2020.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/07/2020] [Accepted: 12/16/2020] [Indexed: 11/26/2022] Open
Abstract
Arsenic exposure through food causes genotoxic effects in vivo. Sub chronic exposure to Arsenic causes pre-cancerous changes. C. olitorious & B. monosperma have significant nephro & hepato protective potential. Protective effect of both plants species is evident in molecular level. Potent use as readily available food supplement to minimize Arsenic toxicity.
Millions of people around the world are chronically exposed to Arsenic (As) through food and drinking water. Studies revealed that Arsenic is genotoxic and causes damage to DNA. In this study, we evaluated Corchorus olitorius and Butea monosperma for their alleviative properties against Arsenic induced genotoxicity in vivo using Wistar Rat model. Arsenic exposed rats were given C. olitorius leaf powder and B. monosperma flower powder as supplementation with normal food. Methylation status of p53 promoter was measured using Methylation Sensitive Restriction Endonuclease PCR (MSRE-PCR) assay and mitochondrial DNA (mtDNA) copy number as well as occurrence of a common deletion in mtDNA in liver and kidney tissue was determined through quantitative realtime PCR (qPCR). Arsenic exposed rats after supplementation showed relatively less severe effects of toxicity evident by significantly higher amount of (p<0.05) mtDNA copy number and reduced occurrence of deletion containing mtDNA as well as lower levels of methylation in p53 gene promoter. Histopathological analysis revealed less severe histopathological changes of liver and kidney and normal liver and kidney function parameters in supplemented rats. So, the protective properties of B. monosperma and C. olitorius against Arsenic toxicity is evident in molecular level.
Collapse
Affiliation(s)
- Amit Datta
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Md Jibran Alam
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Laila Khaleda
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Mohammad Al-Forkan
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chittagong, 4331, Bangladesh
| |
Collapse
|
19
|
Tesfai B, Kibreab F, Dawit A, Mekonen Z, Ghebrezghi S, Kefele S. Cardiovascular Risk Prediction, Glycemic Control, and Determinants in Diabetic and Hypertensive Patients in Massawa Hospital, Eritrea: Cross-Sectional Study on 600 Subjects. Diabetes Metab Syndr Obes 2021; 14:3035-3046. [PMID: 34262310 PMCID: PMC8275095 DOI: 10.2147/dmso.s312448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/04/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Hypertension and diabetes are key determinants of cardiovascular risks. The objective of this study was to calculate 10-year incidence of cardiovascular risk, determine cardiovascular risk factors, and evaluate how diabetes and hypertension are controlled in patients in Massawa Hospital, Eritrea. METHODS This was a hospital-based cross-sectional study using census sampling. A checklist and interview were used as data-collection tool from October 10 to November 20, 2020. Written consent was obtained from each study participant before starting the study. Descriptive statistics were wasused, and results are presented in percentages in tables, p<0.05 was considered significant. RESULTS A total of 600 patients were enrolled in the study, dominated by the Tigrigna (58.7%) and Tigre (26.7%) ethnic groups. About half the patients (58.8%) had a body-mass index of 18-25 kg/m2, with abdominal circumference of <95 cm (74%). Most (93.5%) patients had <10% risk of cardiovascular complications in the coming 10 years. Age showed significant association with hypertension, diabetes mellitus, cardiovascular risk, and poor glycemic and blood-pressure control (p<0.001). Body-mass index, abdominal obesity, and history of stroke were associated with hypertension and diabetes mellitus (p<0.001). Moreover, smoking, hypertension, and monthly income were associated with higher cardiovascular risk (p<0.001). In addition, hypertension and abdominal obesity were associated with glycemic control (p<0.001), and blood-pressure control was significantly associated with diabetes and hypertension (p<0.001). CONCLUSION Age and hypertension were associated with diabetes, cardiovascular risk and poor glycemic control, and smoking, abdominal obesity, and monthly income also significant associations with higher cardiovascular risk and glycemic control. Cessation and adjustment of modifiable factors, such as smoking, hypertension, and regular exercise are highly recommended.
Collapse
Affiliation(s)
- Berhe Tesfai
- Medical Department, Massawa Hospital, Northern Red Sea Zone, Ministry of Health, Massawa, Eritrea
- Correspondence: Berhe Tesfai Medical Department, Massawa Hospital, Northern Red Sea Zone, Ministry of Health, Massawa, Eritrea Email
| | - Fitsum Kibreab
- Health Research and Resources Center Divisiony, Ministry of Health, Asmara, Eritrea
| | - Abraham Dawit
- Medical Department, Massawa Hospital, Northern Red Sea Zone, Ministry of Health, Massawa, Eritrea
| | - Zemui Mekonen
- Medical Department, Massawa Hospital, Northern Red Sea Zone, Ministry of Health, Massawa, Eritrea
| | - Solomon Ghebrezghi
- Medical Department, Massawa Hospital, Northern Red Sea Zone, Ministry of Health, Massawa, Eritrea
| | - Senait Kefele
- Medical Department, Massawa Hospital, Northern Red Sea Zone, Ministry of Health, Massawa, Eritrea
| |
Collapse
|
20
|
Praveena SM, Munisvaradass R, Masiran R, Rajendran RK, Lin CC, Kumar S. Phthalates exposure and attention-deficit/hyperactivity disorder in children: a systematic review of epidemiological literature. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:44757-44770. [PMID: 32895790 DOI: 10.1007/s11356-020-10652-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 08/26/2020] [Indexed: 06/11/2023]
Abstract
Epidemiological studies have proven that children mental health can be affected by environmental pollutants which are believed to be visible in the form of psychological disorder later in their childhood. Moreover, the effects of children mental health are evidently clear in the case of phthalates which have been observed to increase psychological disorder, specifically attention-deficit hyperactivity disorder (ADHD). Hence, the present study aims to conduct a systematic review and provide an overview of the existing literature on the association between urinary phthalate metabolite concentrations and ADHD symptoms among children by emphasizing the confounding factors and limitations. Additionally, this review addressed the possible phthalate mechanism insights in human body including its impact on ADHD symptoms. In this case, 16 epidemiological studies (five cross-sectional, nine cohort and two case control studies) that met all the inclusion criteria were selected out of the total of 427 papers screened to show varying quantitative associations between phthalate exposure and ADHD symptoms among children with confounding factors and limitations in the existing studies in regard to the exposure and outcomes. This review also attempted to present possible explanation on phthalate mechanism in children body and its connection on neurodevelopment and ADHD symptom development which remains unclear in most of the studies. Finally, it is highly recommended for further research to carefully design cohort studies from prenatal to later childhood development with a complete sample size in order to understand phthalate impacts on children health.
Collapse
Affiliation(s)
- Sarva Mangala Praveena
- Department of Environmental and Occupational Health, Faculty of Medicine and Health Science, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor, Malaysia.
- Food Safety and Food Integrity, Institute of Tropical, Agriculture and Food Security, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor, Malaysia.
| | - Rusheni Munisvaradass
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Science, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor, Malaysia
| | - Ruziana Masiran
- Department of Psychiatry, Faculty of Medicine and Health Science, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor, Malaysia
| | - Ranjith Kumar Rajendran
- Graduate Institute of Environmental Engineering, National Central University, No.300, Zhongda Rd., Zhongli District, Taoyuan City, 32001, Taiwan
| | - Chu-Ching Lin
- Graduate Institute of Environmental Engineering, National Central University, No.300, Zhongda Rd., Zhongli District, Taoyuan City, 32001, Taiwan
| | - Suresh Kumar
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Science, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor, Malaysia
- Department of Biotechnology, BIHER, Bharath University, Chennai, 600 073, Tamil Nadu, India
| |
Collapse
|
21
|
Lin CY, Lee HL, Hwang YT, Huang PC, Wang C, Sung FC, Wu C, Su TC. Urinary heavy metals, DNA methylation, and subclinical atherosclerosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 204:111039. [PMID: 32738627 DOI: 10.1016/j.ecoenv.2020.111039] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 07/05/2020] [Accepted: 07/13/2020] [Indexed: 06/11/2023]
Abstract
PURPOSE Lead (Pb) or cadmium (Cd) exposure has been linked to atherosclerosis. Co-exposure of these two heavy metals often occurs in humans. Recent evidence has indicated a crucial role of DNA methylation in atherosclerosis, while Pb or Cd exposure has also been shown to alter DNA methylation. However, it is still unknown whether DNA methylation plays a role in the pathological mechanism of these two heavy metals in atherosclerosis. APPROACH AND RESULTS We enrolled 738 participants (12-30 years) to investigate the association among concentrations of urine Pb or Cd, the 5mdC/dG value (a global DNA methylation marker) and the carotid intima-media thickness (CIMT). When each heavy metal was modeled separately, the results showed urine Pb and Cd concentrations were positively associated with the 5mdC/dG value and CIMT, respectively. When the two heavy metals were analyzed in the same model, urinary Pb concentrations were positively associated with the 5mdC/dG value and CIMT, while urinary Cd concentrations were only positively associated with the CIMT. When Pb and Cd are simultaneously considered in the same logistic regression model, the odds ratios (OR) of thicker CIMT (greater than 75th percentile) with one unit increase in ln-Pb level was 1.67 (95% C.I. = 1.17-2.46, P = 0.005) when levels of 5mdC/dG were above 50th percentile, which is higher than 5mdC/dG bellow the 50th percentile (OR = 1.50 (95% C.I. = 0.96-2.35), P = 0.076). In structural equation model (SEM), Pb or Cd levels are directly associated with CIMT. Moreover, Pb or Cd had an indirect association with CIMT through the 5mdC/dG. When we considered Pb and Cd together, Pb levels had a direct association with CIMT and an indirect association with CIMT through the 5mdC/dG value, while Cd only had a direct association with CIMT. CONCLUSIONS Our findings imply that Pb and Cd exposure might be associated with subclinical atherosclerosis, and global DNA methylation might mediate Pb-associated subclinical atherosclerosis in this young population. Future effort is necessary to elucidate the causal relationship.
Collapse
Affiliation(s)
- Chien-Yu Lin
- Department of Internal Medicine, En Chu Kong Hospital, New Taipei City, 237, Taiwan; School of Medicine, Fu Jen Catholic University, New Taipei City, 242, Taiwan; Department of Environmental Engineering and Health, Yuanpei University of Medical Technology, Hsinchu, 300, Taiwan.
| | - Hui-Ling Lee
- Department of Chemistry, Fu Jen Catholic University, New Taipei City, 242, Taiwan
| | - Yi-Ting Hwang
- Department of Statistics, National Taipei University, New Taipei City, 237, Taiwan
| | - Po-Chin Huang
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, 350, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, 404, Taiwan
| | - Chikang Wang
- Department of Environmental Engineering and Health, Yuanpei University of Medical Technology, Hsinchu, 300, Taiwan
| | - Fung-Chang Sung
- Department of Health Services Administration, College of Public Health, China Medical University, Taichung, 404, Taiwan
| | - Charlene Wu
- Global Health Program, National Taiwan University, College of Public Health, 10055, Taiwan
| | - Ta-Chen Su
- Department of Environmental and Occupational Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan; Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan; Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, 100, Taiwan.
| |
Collapse
|
22
|
Ambrosini S, Mohammed SA, Costantino S, Paneni F. Disentangling the epigenetic landscape in cardiovascular patients: a path toward personalized medicine. Minerva Cardiol Angiol 2020; 69:331-345. [PMID: 32996305 DOI: 10.23736/s2724-5683.20.05326-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Despite significant advances in our understanding of cardiovascular disease (CVD) we are still far from having developed breakthrough strategies to combat coronary atherosclerosis and heart failure, which account for most of CV deaths worldwide. Available cardiovascular therapies have failed to show to be equally effective in all patients, suggesting that inter-individual diversity is an important factor when it comes to conceive and deliver effective personalized treatments. Genome mapping has proved useful in identifying patients who could benefit more from specific drugs depending on genetic variances; however, our genetic make-up determines only a limited part of an individual's risk profile. Recent studies have demonstrated that epigenetic changes - defined as dynamic changes of DNA and histones which do not affect DNA sequence - are key players in the pathophysiology of cardiovascular disease and may participate to delineate cardiovascular risk trajectories over the lifetime. Epigenetic modifications include changes in DNA methylation, histone modifications and non-coding RNAs and these epigenetic signals have shown to cooperate in modulating chromatin accessibility to transcription factors and gene expression. Environmental factors such as air pollution, smoking, psychosocial context, and unhealthy diet regimens have shown to significantly modify the epigenome thus leading to altered transcriptional programs and CVD phenotypes. Therefore, the integration of genetic and epigenetic information might be invaluable to build individual maps of cardiovascular risk and hence, could be employed for the design of customized diagnostic and therapeutic strategies. In the present review, we discuss the growing importance of epigenetic information and its putative implications in cardiovascular precision medicine.
Collapse
Affiliation(s)
- Samuele Ambrosini
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| | - Shafeeq A Mohammed
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| | - Sarah Costantino
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland - .,Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
23
|
New Mechanisms of Vascular Dysfunction in Cardiometabolic Patients: Focus on Epigenetics. High Blood Press Cardiovasc Prev 2020; 27:363-371. [PMID: 32740853 DOI: 10.1007/s40292-020-00400-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 07/21/2020] [Indexed: 12/28/2022] Open
Abstract
Epigenetic processing takes centre stage in cardiometabolic diseases (obesity, metabolic syndrome, type 2 diabetes, hypertension), where it participates in adiposity, inflammation, endothelial dysfunction, vascular insulin resistance and atherosclerosis. Epigenetic modifications, defined as heritable changes in gene expression that do not entail mutation in the DNA sequence, are mainly induced by environmental stimuli (stress, pollution, cigarette smoking) and are gaining considerable interest due to their causal role in cardiovascular disease, and their amenability to pharmacological intervention. Importantly, epigenetic modifications acquired during life can be transmitted to the offspring and exert their biological effects across multiple generations. Indeed, such transgenerational transmission of epigenetic signals may contribute to anticipating cardiovascular and metabolic disease phenotypes already in children and young adults. A deeper understanding of environmental factors and their effects on the epigenetic machinery and transcriptional programs is warranted to develop effective mechanism-based therapeutic strategies. The clinical application of epigenetic drugs-also known as "epi-drugs"-is currently exploding in the field of cardiovascular disease. The present review describes the main epigenetic networks underlying cardiometabolic alterations and sheds light on specific points of intervention for pharmacological reprogramming in this setting.
Collapse
|
24
|
Wang Z, Zhao YT, Zhao TC. Histone deacetylases in modulating cardiac disease and their clinical translational and therapeutic implications. Exp Biol Med (Maywood) 2020; 246:213-225. [PMID: 32727215 DOI: 10.1177/1535370220944128] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Cardiovascular diseases are the leading cause of mortality and morbidity worldwide. Histone deacetylases (HDACs) play an important role in the epigenetic regulation of genetic transcription in response to stress or pathological conditions. HDACs interact with a complex co-regulatory network of transcriptional regulators, deacetylate histones or non-histone proteins, and modulate gene expression in the heart. The selective HDAC inhibitors have been considered to be a critical target for the treatment of cardiac disease, especially for ameliorating cardiac dysfunction. In this review, we discuss our current knowledge of the cellular and molecular basis of HDACs in mediating cardiac development and hypertrophy and related pharmacologic interventions in heart disease.
Collapse
Affiliation(s)
- Zhengke Wang
- Department of Surgery, Boston University Medical School, Roger Williams Medical Center, Providence, RI 02908, USA
| | - Yu Tina Zhao
- University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Ting C Zhao
- Departments of Surgery and Plastic Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| |
Collapse
|
25
|
Koseler A, Ma F, Kilic ID, Morselli M, Kilic O, Pellegrini M. Genome-wide DNA Methylation Profiling of Blood from Monozygotic Twins Discordant for Myocardial Infarction. In Vivo 2020; 34:361-367. [PMID: 31882500 DOI: 10.21873/invivo.11782] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 01/24/2023]
Abstract
BACKGROUND/AIM This study aimed to measure the DNA methylation state of thousands of CpG islands in the blood of two monozygotic twins that were discordant for cardiovascular disease (CVD). Twin 1 had suffered myocardial infarction, while the other was healthy. PATIENTS AND METHODS Since the aim of this study was to identify differentially methylated regions which might act as potential markers, reduced-representation bisulfite libraries were used for whole-genome methylation analysis. RESULTS According to the analysis, 11 genes lipid droplet associated hydrolase (LDAH), apolipoprotein B (APOB), acyl-CoA synthetase medium chain family member 2A (ACSM2A), acyl-CoA synthetase medium chain family member 5(ACSM5), acyl-CoA synthetase family member 3 (ACSF3), carboxylesterase 1 (CES1), carboxylesterase 1 pseudogene 1 (CES1P1), AFG3 like matrix AAA peptidase subunit 2 (AFG3L2), iron-sulfur cluster assembly enzyme (ISCU), SEC14 like lipid binding 2 (SEC14L2) and microsomal triglyceride transfer protein (MTTP) were all hypomethylated in DNA from twin 2, the unaffected twin. Methylation changes were observed at different multiple loci between the twins, suggesting loci that are affected by disease status in identical genetic backgrounds. CONCLUSION This twin study may contribute significantly to the understanding of the genetic basis of CVD and resulting myocardial infarction. This approach may allow identification of possible target loci associated with aberrant epigenetic regulation in CVD.
Collapse
Affiliation(s)
- Aylin Koseler
- Department of Biophysics, Pamukkale University School of Medicine, Denizli, Turkey
| | - Feiyang Ma
- Department of Molecular, Cell and Developmental Biology, David Geffen School of Medicine, University of California, Los Angeles, CA, U.S.A
| | - Ismail Dogu Kilic
- Department of Cardiology, Pamukkale University School of Medicine, Denizli, Turkey
| | - Marco Morselli
- Department of Molecular, Cell and Developmental Biology, David Geffen School of Medicine, University of California, Los Angeles, CA, U.S.A
| | - Oguz Kilic
- Department of Cardiology, Pamukkale University School of Medicine, Denizli, Turkey
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, David Geffen School of Medicine, University of California, Los Angeles, CA, U.S.A.
| |
Collapse
|
26
|
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. [DOI: 10.1007/s11033-020-05570-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 06/05/2020] [Indexed: 12/11/2022]
|
27
|
Abstract
Retrotransposon activity and genomic template stability (GTS) are one of the most significant rearranging mechanisms in environmental stress. Therefore, in this study, it is aimed to elucidate effecting of Cobalt (Co) on the instability of genomes and Long Terminal Repeat retrotransposon polymorphism in Zea mays and whether humic acid (HA) has any role on these parameters. For this purpose, Retrotransposon-microsatellite amplified polymorphism (REMAP) and Inter-Retrotransposon Amplified Polymorphism (IRAP) markers were applied to evaluate retrotransposon polymorphism and the GTS levels. It was found that IRAP and REMAP primers generate unique polymorphic band structures on maize plants treated with various doses of Co. Retrotransposon polymorphism increased and GTS decreased while increasing Co concentration. On the other hand, there was a reduction in negative effects of Co on retrotransposon GTS and polymorphism after treatment with HA. The results indicate that HA may be used effectively for the protection of maize seedlings from the destructive effects of Co toxicity.
Collapse
|
28
|
Dutta S, Haggerty DK, Rappolee DA, Ruden DM. Phthalate Exposure and Long-Term Epigenomic Consequences: A Review. Front Genet 2020; 11:405. [PMID: 32435260 PMCID: PMC7218126 DOI: 10.3389/fgene.2020.00405] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 03/30/2020] [Indexed: 12/27/2022] Open
Abstract
Phthalates are esters of phthalic acid which are used in cosmetics and other daily personal care products. They are also used in polyvinyl chloride (PVC) plastics to increase durability and plasticity. Phthalates are not present in plastics by covalent bonds and thus can easily leach into the environment and enter the human body by dermal absorption, ingestion, or inhalation. Several in vitro and in vivo studies suggest that phthalates can act as endocrine disruptors and cause moderate reproductive and developmental toxicities. Furthermore, phthalates can pass through the placental barrier and affect the developing fetus. Thus, phthalates have ubiquitous presence in food and environment with potential adverse health effects in humans. This review focusses on studies conducted in the field of toxicogenomics of phthalates and discusses possible transgenerational and multigenerational effects caused by phthalate exposure during any point of the life-cycle.
Collapse
Affiliation(s)
- Sudipta Dutta
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Diana K Haggerty
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, United States
| | - Daniel A Rappolee
- Department of Obstetrics and Gynecology, Reproductive Endocrinology and Infertility, CS Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, United States.,Reproductive Stress, Inc., Grosse Pointe Farms, MI, United States
| | - Douglas M Ruden
- Department of Obstetrics and Gynecology, Reproductive Endocrinology and Infertility, CS Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, United States.,Institutes for Environmental Health Science, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
29
|
Al-Hasani K, Khurana I, Farhat T, Eid A, El-Osta A. Epigenetics of Diabetic Nephropathy: From Biology to Therapeutics. EUROPEAN MEDICAL JOURNAL 2020. [DOI: 10.33590/emj/19-00137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Diabetic nephropathy (DN) is a lethal microvascular complication associated with Type 1 and Type 2 diabetes mellitus, and is the leading single cause of end-stage renal disease. Although genetic influences are important, epigenetic mechanisms have been implicated in several aspects of the disease. The current therapeutic methods to treat DN are limited to slowing disease progression without repair and regeneration of the damaged nephrons. Replacing dying or diseased kidney cells with new nephrons is an attractive strategy. This review considers the genetic and epigenetic control of nephrogenesis, together with the epigenetic mechanisms that accompany kidney development and recent advances in induced reprogramming and kidney cell regeneration in the context of DN.
Collapse
Affiliation(s)
- Keith Al-Hasani
- Department of Diabetes, Epigenetics in Human Health and Disease Laboratory, Monash University, Melbourne, Australia
| | - Ishant Khurana
- Department of Diabetes, Epigenetics in Human Health and Disease Laboratory, Monash University, Melbourne, Australia
| | - Theresa Farhat
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Assaad Eid
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Assam El-Osta
- Department of Diabetes, Epigenetics in Human Health and Disease Laboratory, Monash University, Melbourne, Australia; Department of Clinical Pathology, The University of Melbourne, Victoria, Australia; Faculty of Health, Department of Technology, Biomedical Laboratory Science, University College Copenhagen, Copenhagen, Denmark; Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong
| |
Collapse
|
30
|
Mohammed SA, Ambrosini S, Lüscher T, Paneni F, Costantino S. Epigenetic Control of Mitochondrial Function in the Vasculature. Front Cardiovasc Med 2020; 7:28. [PMID: 32195271 PMCID: PMC7064473 DOI: 10.3389/fcvm.2020.00028] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/19/2020] [Indexed: 12/24/2022] Open
Abstract
The molecular signatures of epigenetic regulation and chromatin architecture are emerging as pivotal regulators of mitochondrial function. Recent studies unveiled a complex intersection among environmental factors, epigenetic signals, and mitochondrial metabolism, ultimately leading to alterations of vascular phenotype and increased cardiovascular risk. Changing environmental conditions over the lifetime induce covalent and post-translational chemical modification of the chromatin template which sensitize the genome to establish new transcriptional programs and, hence, diverse functional states. On the other hand, metabolic alterations occurring in mitochondria affect the availability of substrates for chromatin-modifying enzymes, thus leading to maladaptive epigenetic signatures altering chromatin accessibility and gene transcription. Indeed, several components of the epigenetic machinery require intermediates of cellular metabolism (ATP, AcCoA, NADH, α-ketoglutarate) for enzymatic function. In the present review, we describe the emerging role of epigenetic modifications as fine tuners of gene transcription in mitochondrial dysfunction and vascular disease. Specifically, the following aspects are described in detail: (i) mitochondria and vascular function, (ii) mitochondrial ROS, (iii) epigenetic regulation of mitochondrial function; (iv) the role of mitochondrial metabolites as key effectors for chromatin-modifying enzymes; (v) epigenetic therapies. Understanding epigenetic routes may pave the way for new approaches to develop personalized therapies to prevent mitochondrial insufficiency and its complications.
Collapse
Affiliation(s)
- Shafeeq A Mohammed
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| | - Samuele Ambrosini
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| | - Thomas Lüscher
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland.,Research, Education and Development, Royal Brompton and Harefield Hospital Trust and Imperial College, London, United Kingdom
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland.,Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Sarah Costantino
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| |
Collapse
|
31
|
Davis FM, Gallagher KA. Epigenetic Mechanisms in Monocytes/Macrophages Regulate Inflammation in Cardiometabolic and Vascular Disease. Arterioscler Thromb Vasc Biol 2020; 39:623-634. [PMID: 30760015 DOI: 10.1161/atvbaha.118.312135] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cardiometabolic and vascular disease, with their associated secondary complications, are the leading cause of morbidity and mortality in Western society. Chronic inflammation is a common theme that underlies initiation and progression of cardiovascular disease. In this regard, monocytes/macrophages are key players in the development of a chronic inflammatory state. Over the past decade, epigenetic modifications, such as DNA methylation and posttranslational histone processing, have emerged as important regulators of immune cell phenotypes. Accumulating studies reveal the importance of epigenetic enzymes in the dynamic regulation of key signaling pathways that alter monocyte/macrophage phenotypes in response to environmental stimuli. In this review, we highlight the current paradigms of monocyte/macrophage polarization and the emerging role of epigenetic modification in the regulation of monocyte/macrophage phenotype in obesity, diabetes mellitus, atherosclerosis, and abdominal aortic aneurysms.
Collapse
Affiliation(s)
- Frank M Davis
- From the Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor
| | - Katherine A Gallagher
- From the Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor
| |
Collapse
|
32
|
Wang N, Li Q, Liu H, Lin L, Han W, Hao W. Role of C/EBPα hypermethylation in diesel engine exhaust exposure-induced lung inflammation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 183:109500. [PMID: 31450033 DOI: 10.1016/j.ecoenv.2019.109500] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/27/2019] [Accepted: 07/30/2019] [Indexed: 06/10/2023]
Abstract
Exposure to diesel engine exhaust (DEE) impairs lung function. But the underlying mechanisms are still not fully understood. The aim of this study was to investigate the effects of long-term DEE exposure on lung inflammation and the underlying mechanisms. Sprague-Dawley male rats were exposed to DEE with 3 mg/m3 of diesel exhaust particles (DEP) for 12 weeks. Then urine, blood, bronchoalveolar lavage fluid (BALF), and lung tissue were collected for the determination of biochemistry indexes, DNA methylation status, and histological changes in the lung. The results showed that the metabolites of polycyclic aromatic hydrocarbons (PAHs) 2-hydroxyphenanthrene (2-OHPh) and 9-OHPh, and 8-hydroxy-2'-deoxyguanosine (8-OHdG), and malondialdehyde (MDA) level were higher in urine of DEE-exposed rats than control group. The level of proinflammatory cytokines IL-8, IL-6, and TNF-α was significantly higher in serum (1.8, 3.5, and nearly 1.0-fold increase, respectively), BALF (2.2, 3.8, and 2.0-fold increase, respectively) and lung tissues (3.5, 4.3, and 2.4-fold increase, respectively) of DEE-exposed rats than control group. While the level of clara cell secretory protein (CC16) and pulmonary surfactant protein D (SP-D) with anti-inflammatory property was obviously lower in serum (reduction of 29% and 38%, respectively), BALF (reduction of 50% and 46%, respectively) and lung tissues (reduction of 50% and 55%, respectively) of DEE-exposed rats than control group. Exposure to DEE also resulted in significant increases in total white blood cell (WBC), neutrophil, eosinophil, and lymphocyte number in BALF. Airway inflammation and remolding were apparent in DEE group. The methylation level of CCAAT/enhancer-binding protein alpha (C/EBPα) promoter was markedly increased (about 3.2-fold increase), and its mRNA and protein expression were significantly decreased (about 62% and 68% decrease, respectively) in the lungs of DEE-exposed rats compared with the group. Further, cell experiments were performed to investigate the relationship between C/EBPα and CC16, and CC16 function under DEP conditions. The results showed that DEP inhibited CC16 expression via methylation of C/EBPα promoter, and the increase of CC16 level significantly relieved the proinflammatory effects caused by DEP exposure. In conclusion, our data indicated that long-term exposure to DEE can cause lung inflammation, at least in part via methylation of C/EBPα promoter, and inhibition of CC16 expression.
Collapse
Affiliation(s)
- Ning Wang
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, 266011, China
| | - Qinghai Li
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, 266011, China
| | - Hong Liu
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, 266011, China
| | - Li Lin
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, 266011, China
| | - Wei Han
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, 266011, China.
| | - Wanming Hao
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, 266011, China.
| |
Collapse
|
33
|
CaM kinase II regulates cardiac hemoglobin expression through histone phosphorylation upon sympathetic activation. Proc Natl Acad Sci U S A 2019; 116:22282-22287. [PMID: 31619570 DOI: 10.1073/pnas.1816521116] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Sympathetic activation of β-adrenoreceptors (β-AR) represents a hallmark in the development of heart failure (HF). However, little is known about the underlying mechanisms of gene regulation. In human ventricular myocardium from patients with end-stage HF, we found high levels of phosphorylated histone 3 at serine-28 (H3S28p). H3S28p was increased by inhibition of the catecholamine-sensitive protein phosphatase 1 and decreased by β-blocker pretreatment. By a series of in vitro and in vivo experiments, we show that the β-AR downstream protein kinase CaM kinase II (CaMKII) directly binds and phosphorylates H3S28. Whereas, in CaMKII-deficient myocytes, acute catecholaminergic stimulation resulted in some degree of H3S28p, sustained catecholaminergic stimulation almost entirely failed to induce H3S28p. Genome-wide analysis of CaMKII-mediated H3S28p in response to chronic β-AR stress by chromatin immunoprecipitation followed by massive genomic sequencing led to the identification of CaMKII-dependent H3S28p target genes. Forty percent of differentially H3S28p-enriched genomic regions were associated with differential, mostly increased expression of the nearest genes, pointing to CaMKII-dependent H3S28p as an activating histone mark. Remarkably, the adult hemoglobin genes showed an H3S28p enrichment close to their transcriptional start or end sites, which was associated with increased messenger RNA and protein expression. In summary, we demonstrate that chronic β-AR activation leads to CaMKII-mediated H3S28p in cardiomyocytes. Thus, H3S28p-dependent changes may play an unexpected role for cardiac hemoglobin regulation in the context of sympathetic activation. These data also imply that CaMKII may be a yet unrecognized stress-responsive regulator of hematopoesis.
Collapse
|
34
|
Patsouras MD, Vlachoyiannopoulos PG. Evidence of epigenetic alterations in thrombosis and coagulation: A systematic review. J Autoimmun 2019; 104:102347. [PMID: 31607428 DOI: 10.1016/j.jaut.2019.102347] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023]
Abstract
Thrombosis in the context of Cardiovascular disease (CVD) affects mainly the blood vessels supplying the heart, brain and peripheries and it is the leading cause of death worldwide. The pathophysiological thrombotic mechanisms are largely unknown. Heritability contributes to a 30% of the incidence of CVD. The remaining variation can be explained by life style factors such as smoking, dietary and exercise habits, environmental exposure to toxins, and drug usage and other comorbidities. Epigenetic variation can be acquired or inherited and constitutes an interaction between genes and the environment. Epigenetics have been implicated in atherosclerosis, ischemia/reperfusion damage and the cardiovascular response to hypoxia. Epigenetic regulators of gene expression are mainly the methylation of CpG islands, histone post translational modifications (PTMs) and microRNAs (miRNAs). These epigenetic regulators control gene expression either through activation or silencing. Epigenetic control is mostly dynamic and can potentially be manipulated to prevent or reverse the uncontrolled expression of genes, a trait that renders them putative therapeutic targets. In the current review, we systematically studied and present available data on epigenetic alterations implicated in thrombosis derived from human studies. Evidence of epigenetic alterations is observed in several thrombotic diseases such as Coronary Artery Disease and Cerebrovascular Disease, Preeclampsia and Antiphospholipid Syndrome. Differential CpG methylation and specific histone PTMs that control transcription of prothrombotic and proinflammatory genes have also been associated with predisposing factors of thrombosis and CVD, such us smoking, air pollution, hypertriglyceridemia, occupational exposure to particulate matter and comorbidities including cancer, Chronic Obstructive Pulmonary Disease and Chronic Kidney Disease. These clinical observations are further supported by in vitro experiments and indicate that epigenetic regulation affects the pathophysiology of thrombotic disorders with potential diagnostic or therapeutic utility.
Collapse
Affiliation(s)
- M D Patsouras
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - P G Vlachoyiannopoulos
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
35
|
Prenatal high-salt diet impaired vasodilatation with reprogrammed renin-angiotensin system in offspring rats. J Hypertens 2019; 36:2369-2379. [PMID: 30382958 DOI: 10.1097/hjh.0000000000001865] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIMS High-salt diet is linked to hypertension, and prenatal high-salt diet increases the risk of cardiovascular diseases in the offspring. The present study investigated whether and how prenatal high-salt diet influenced nitric oxide-mediated vasodilatation in the offspring. METHODS AND RESULTS Pregnant rats were fed either normal-salt (1% sodium chloride) or high-salt (8% sodium chloride) diet during gestation. Experiments were conducted in 5-month-old male offspring. Sodium nitroprusside (SNP, nitric oxide donor)-induced hypotensive responses (in vivo) and vascular dilatation (in vitro) was significantly attenuated (Emax: 84 ± 2 vs. 51 ± 2, high-salt vs. control, P < 0.001) in the high-salt offspring, indicating reduced vascular relaxations. Pretreatment with Tempol (reactive oxygen species scavenger) alleviated this attenuation. The high-salt offspring showed an increased level of oxidative stress markers in both mesenteric arteries and plasma samples. The antioxidant activity, serum superoxide dismutase and catalase were significantly reduced, whereas malondialdehyde was increased in the high-salt offspring. O2 production, and protein expression of Nox2 and Nox4 in mesenteric arteries was significantly increased in the high-salt offspring whereas Nox1 showed no changes. The local renin-angiotensin system in mesenteric arteries was activated, associated with an increased NADPH oxidase. DNA methylation at the proximal promoter of angiotensin-converting enzyme gene in the lung was significantly increased in the high-salt offspring (P = 0.004). CONCLUSION The results suggest that prenatal high-salt diet impairs nitric oxide-mediated vasodilatation because of the increased oxidative stress-affected renin-angiotensin system in the high-salt offspring, providing new information for understanding, and early prevention of cardiovascular diseases in fetal origins.
Collapse
|
36
|
Lei X, Chen R, Wang C, Shi J, Zhao Z, Li W, Yan B, Chillrud S, Cai J, Kan H. Personal Fine Particulate Matter Constituents, Increased Systemic Inflammation, and the Role of DNA Hypomethylation. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2019; 53:9837-9844. [PMID: 31328512 PMCID: PMC7092684 DOI: 10.1021/acs.est.9b02305] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Limited evidence is available on the effects of various fine particulate matter (PM2.5) components on inflammatory cytokines and DNA methylation. We examined whether 16 PM2.5 components are associated with changes in four blood biomarkers, that is, tumor necrosis factor-α (TNF-α), soluble cluster of differentiation 40 ligand (sCD40L), soluble intercellular adhesion molecule-1 (sICAM-1), and fibrinogen, as well as their corresponding DNA methylation levels in a panel of 36 healthy college students in Shanghai, China. We used linear mixed-effect models to evaluate the associations, with controls of potential confounders. We further conducted mediation analysis to evaluate the potential mediation effects of components on inflammatory markers through change in DNA methylation. We observed that several components were consistently associated with TNF-α and fibrinogen as well as their DNA hypomethylation. For example, an interquartile range increase in personal exposure to PM2.5-lead (Pb) was associated with 65.20% (95% CI: 37.07, 99.10) increase in TNF-α and 2.66 (95% CI: 37.07, 99.10) decrease in TNF-α methylation, 30.51% (95% CI: 0.72, 69.11) increase in fibrinogen and 1.25 (95% CI: 0.67, 1.83) decrease in F3 methylation. PM2.5 components were significantly associated with sICAM-1 methylation but not with sICAM-1 protein. DNA methylation mediated 19.89%-41.75% of the elevation in TNF-α expression by various PM2.5 constituents. Our findings provide clues that personal PM2.5 constituents exposure may contribute to increased systemic inflammation through DNA hypomethylation.
Collapse
Affiliation(s)
- Xiaoning Lei
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, 200433, China
| | - Renjie Chen
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, 200433, China
| | - Cuicui Wang
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, 200433, China
| | - Jingjin Shi
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, 200433, China
| | - Zhuohui Zhao
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, 200433, China
| | - Weihua Li
- Key Laboratory of Reproduction Regulation of National Population and Family Planning Commission, Shanghai Institute of Planned Research, Institute of Reproduction and Development, Fudan University, Shanghai, 200433, China
| | - Beizhan Yan
- Division of Geochemistry, Lamont-Doherty Earth Observatory of Columbia University, Palisades, New York 10964, United States
| | - Steve Chillrud
- Division of Geochemistry, Lamont-Doherty Earth Observatory of Columbia University, Palisades, New York 10964, United States
| | - Jing Cai
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, 200433, China
- Shanghai Key Laboratory of Meteorology and Health, Shanghai, 200030, China
- Corresponding Authors: Phone/fax: +86 (21) 54237908; . Phone/fax: +86 (21) 54237908;
| | - Haidong Kan
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, 200433, China
- Key Laboratory of Reproduction Regulation of National Population and Family Planning Commission, Shanghai Institute of Planned Research, Institute of Reproduction and Development, Fudan University, Shanghai, 200433, China
- Corresponding Authors: Phone/fax: +86 (21) 54237908; . Phone/fax: +86 (21) 54237908;
| |
Collapse
|
37
|
Cellular Effects of Butyrate on Vascular Smooth Muscle Cells are Mediated through Disparate Actions on Dual Targets, Histone Deacetylase (HDAC) Activity and PI3K/Akt Signaling Network. Int J Mol Sci 2019; 20:ijms20122902. [PMID: 31197106 PMCID: PMC6628026 DOI: 10.3390/ijms20122902] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/08/2019] [Accepted: 06/11/2019] [Indexed: 12/19/2022] Open
Abstract
Vascular remodeling is a characteristic feature of cardiovascular diseases. Altered cellular processes of vascular smooth muscle cells (VSMCs) is a crucial component in vascular remodeling. Histone deacetylase inhibitor (HDACI), butyrate, arrests VSMC proliferation and promotes cell growth. The objective of the study is to determine the mechanism of butyrate-induced VSMC growth. Using proliferating VSMCs exposed to 5 mM butyrate, immunoblotting studies are performed to determine whether PI3K/Akt pathway that regulates different cellular effects is a target of butyrate-induced VSMC growth. Butyrate inhibits phosphorylation-dependent activation of PI3K, PDK1, and Akt, eliciting differential effects on downstream targets of Akt. Along with previously reported Ser9 phosphorylation-mediated GSK3 inactivation leading to stability, increased expression and accumulation of cyclin D1, and epigenetic histone modifications, inactivation of Akt by butyrate results in: transcriptional activation of FOXO1 and FOXO3 promoting G1 arrest through p21Cip1/Waf1 and p15INK4B upregulation; inactivation of mTOR inhibiting activation of its targets p70S6K and 4E-BP1 impeding protein synthesis; inhibition of caspase 3 cleavage and downregulation of PARP preventing apoptosis. Our findings imply butyrate abrogates Akt activation, causing differential effects on Akt targets promoting convergence of cross-talk between their complimentary actions leading to VSMC growth by arresting proliferation and inhibiting apoptosis through its effect on dual targets, HDAC activity and PI3K/Akt pathway network.
Collapse
|
38
|
Ganguly BB. Exposure index of methyl isocyanate (MIC) gas disaster and a comprehensive spectrum of cytogenetic analysis after 30 years. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:18208-18229. [PMID: 31041706 DOI: 10.1007/s11356-019-04439-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 01/30/2019] [Indexed: 06/09/2023]
Abstract
Severity of clinical expression and high mortality could not facilitate establishing exposure index/association following MIC disaster in Bhopal. Mortality-based exposure stratification was critiqued by the International Medical Commission on Bhopal (IMCB). IMCB stratified exposure considering distance as surrogate at 2 km intervals after 10 years. The first follow-up cytogenetic screening of the pre-screened survivors after 30 years has demonstrated chromosome abnormalities (CA). Exposure stratification was attempted considering cytogenetic screening conducted during 1986-1988. Elevation of CA appeared proportional to exposure status and authenticated the initial mortality-based stratification. The one-on-one comparison of the previous and present cytogenetics has described the individual response to MIC exposure over 30 years. Chi-square test has been carried out for checking the cytogenetic changes at the individual level statistically, which revealed that differences of chromosomal aberrations collected immediately post-disaster and 30 years later are nonsignificant. The prominence of interindividual variation was noticed in general. The impact of overall exposure was higher in males. Constitutional abnormalities in 8.5% of the study population, including translocation, inversion, deletion, fragile sites, etc., necessitate screening of blood-linked members. The incidence of acrocentric association was prominent in the study population. Normal karyotype in children born to severely exposed parents with congenital anomalies indicates necessity of molecular karyotyping and/or screening of mutations. The study highlights follow-up of the health of the index cases at shorter (3-6 months) intervals. This comprehensive spectrum of cytogenetic report highlights immediate post-disaster chromosomal aberrations, the changes that occurred over 30 years in conjunction with other environmental factors at the individual level, constitutive genomic aberrations, polymorphic variations, and chromosomal patterns in congenitally malformed children of the survivors, which collectively indicate the possibility of acquisition/persistence of stable aberrations in MIC-exposed lymphocytes through interaction with environmental/biological confounders.
Collapse
Affiliation(s)
- Bani Bandana Ganguly
- MGM Center for Genetic Research & Diagnosis, MGM New Bombay Hospital, Vashi Sector 3, Navi Mumbai, 400703, India.
| |
Collapse
|
39
|
Grau-Perez M, Agha G, Pang Y, Bermudez JD, Tellez-Plaza M. Mendelian Randomization and the Environmental Epigenetics of Health: a Systematic Review. Curr Environ Health Rep 2019; 6:38-51. [DOI: 10.1007/s40572-019-0226-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
40
|
Li D, Tolleson WH, Yu D, Chen S, Guo L, Xiao W, Tong W, Ning B. Regulation of cytochrome P450 expression by microRNAs and long noncoding RNAs: Epigenetic mechanisms in environmental toxicology and carcinogenesis. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2019; 37:180-214. [PMID: 31305208 PMCID: PMC6737535 DOI: 10.1080/10590501.2019.1639481] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Environmental exposures to hazardous chemicals are associated with a variety of human diseases and disorders, including cancers. Phase I metabolic activation and detoxification reactions catalyzed by cytochrome P450 enzymes (CYPs) affect the toxicities of many xenobiotic compounds. Proper regulation of CYP expression influences their biological effects. Noncoding RNAs (ncRNAs) are involved in regulating CYP expression, and ncRNA expression is regulated in response to environmental chemicals. The mechanistic interactions between ncRNAs and CYPs associated with the toxicity and carcinogenicity of environmental chemicals are described in this review, focusing on microRNA-dependent CYP regulation. The role of long noncoding RNAs in regulating CYP expression is also presented and new avenues of research concerning this regulatory mechanism are described.
Collapse
Affiliation(s)
- Dongying Li
- a National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA) , Jefferson , AR , USA
| | - William H Tolleson
- a National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA) , Jefferson , AR , USA
| | - Dianke Yu
- a National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA) , Jefferson , AR , USA
| | - Si Chen
- a National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA) , Jefferson , AR , USA
| | - Lei Guo
- a National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA) , Jefferson , AR , USA
| | - Wenming Xiao
- a National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA) , Jefferson , AR , USA
| | - Weida Tong
- a National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA) , Jefferson , AR , USA
| | - Baitang Ning
- a National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA) , Jefferson , AR , USA
| |
Collapse
|
41
|
Wang T, Pehrsson EC, Purushotham D, Li D, Zhuo X, Zhang B, Lawson HA, Province MA, Krapp C, Lan Y, Coarfa C, Katz TA, Tang WY, Wang Z, Biswal S, Rajagopalan S, Colacino JA, Tsai ZTY, Sartor MA, Neier K, Dolinoy DC, Pinto J, Hamanaka RB, Mutlu GM, Patisaul HB, Aylor DL, Crawford GE, Wiltshire T, Chadwick LH, Duncan CG, Garton AE, McAllister KA, Bartolomei MS, Walker CL, Tyson FL. The NIEHS TaRGET II Consortium and environmental epigenomics. Nat Biotechnol 2018; 36:225-227. [PMID: 29509741 DOI: 10.1038/nbt.4099] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Ting Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA.,The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Erica C Pehrsson
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA.,The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Deepak Purushotham
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA.,The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Daofeng Li
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA.,The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Xiaoyu Zhuo
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA.,The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Bo Zhang
- Center for Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Heather A Lawson
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael A Province
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Christopher Krapp
- Epigenetics Institute, Center for Excellence in Environmental Toxicology, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yemin Lan
- Epigenetics Institute, Center for Excellence in Environmental Toxicology, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Cristian Coarfa
- Center for Precision Environmental Health, Departments of Molecular & Cellular Biology and Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Tiffany A Katz
- Center for Precision Environmental Health, Departments of Molecular & Cellular Biology and Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Wan Yee Tang
- Department of Environmental Health Sciences, Johns Hopkins School of Public Health, Baltimore, Maryland, USA
| | - Zhibin Wang
- Department of Environmental Health Sciences, Johns Hopkins School of Public Health, Baltimore, Maryland, USA
| | - Shyam Biswal
- Department of Environmental Health Sciences, Johns Hopkins School of Public Health, Baltimore, Maryland, USA
| | - Sanjay Rajagopalan
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Justin A Colacino
- Department of Environmental Health Sciences and Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Zing Tsung-Yeh Tsai
- Department of Environmental Health Sciences and Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Maureen A Sartor
- Department of Environmental Health Sciences and Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Kari Neier
- Department of Environmental Health Sciences and Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Dana C Dolinoy
- Department of Environmental Health Sciences and Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Jayant Pinto
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Robert B Hamanaka
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Gokhan M Mutlu
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Heather B Patisaul
- Department of Biological Sciences, Center for Human Health and the Environment, Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, USA
| | - David L Aylor
- Department of Biological Sciences, Center for Human Health and the Environment, Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, USA
| | - Gregory E Crawford
- Center for Genomic & Computational Biology, Division of Medical Genetics, Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Tim Wiltshire
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Lisa H Chadwick
- Genes Environment and Health Branch, Division of Extramural Research and Training, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Christopher G Duncan
- Genes Environment and Health Branch, Division of Extramural Research and Training, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Amanda E Garton
- Genes Environment and Health Branch, Division of Extramural Research and Training, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Kimberly A McAllister
- Genes Environment and Health Branch, Division of Extramural Research and Training, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | | | - Marisa S Bartolomei
- Epigenetics Institute, Center for Excellence in Environmental Toxicology, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Cheryl L Walker
- Center for Precision Environmental Health, Departments of Molecular & Cellular Biology and Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Frederick L Tyson
- Genes Environment and Health Branch, Division of Extramural Research and Training, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| |
Collapse
|
42
|
Costantino S, Libby P, Kishore R, Tardif JC, El-Osta A, Paneni F. Epigenetics and precision medicine in cardiovascular patients: from basic concepts to the clinical arena. Eur Heart J 2018; 39:4150-4158. [PMID: 29069341 PMCID: PMC6293269 DOI: 10.1093/eurheartj/ehx568] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/04/2017] [Accepted: 09/22/2017] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of mortality worldwide and also inflict major burdens on morbidity, quality of life, and societal costs. Considering that CVD preventive medications improve vascular outcomes in less than half of patients (often relative risk reductions range from 12% to 20% compared with placebo), precision medicine offers an attractive approach to refine the targeting of CVD medications to responsive individuals in a population and thus allocate resources more wisely and effectively. New tools furnished by advances in basic science and translational medicine could help achieve this goal. This approach could reach beyond the practitioners 'eyeball' assessment or venerable markers derived from the physical examination and standard laboratory evaluation. Advances in genetics have identified novel pathways and targets that operate in numerous diseases, paving the way for 'precision medicine'. Yet the inherited genome determines only part of an individual's risk profile. Indeed, standard genomic approaches do not take into account the world of regulation of gene expression by modifications of the 'epi'genome. Epigenetic modifications defined as 'heritable changes to the genome that do not involve changes in DNA sequence' have emerged as a new layer of biological regulation in CVD and could advance individualized risk assessment as well as devising and deploying tailored therapies. This review, therefore, aims to acquaint the cardiovascular community with the rapidly advancing and evolving field of epigenetics and its implications in cardiovascular precision medicine.
Collapse
Affiliation(s)
- Sarah Costantino
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Zurich, Switzerland
| | - Peter Libby
- Brigham and Women’s Hospital, Division of Cardiovascular Medicine, Boston, MA, USA
| | - Raj Kishore
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, MERB-953, 3500 N Broad Street, Philadelphia, PA, USA
- Department of Pharmacology, Temple University, Philadelphia, PA, USA
| | - Jean-Claude Tardif
- Montreal Health Innovations Coordinating Center (MHICC), Montreal, Canada
- Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - Assam El-Osta
- Central Clinical School, Faculty of Medicine, Monash University, Victoria, Australia
- Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
- Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Zurich, Switzerland
- University Heart Center, Cardiology, University Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
43
|
Tibaut M, Caprnda M, Kubatka P, Sinkovič A, Valentova V, Filipova S, Gazdikova K, Gaspar L, Mozos I, Egom EE, Rodrigo L, Kruzliak P, Petrovic D. Markers of Atherosclerosis: Part 2 - Genetic and Imaging Markers. Heart Lung Circ 2018; 28:678-689. [PMID: 30318392 DOI: 10.1016/j.hlc.2018.09.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/30/2018] [Accepted: 06/25/2018] [Indexed: 12/18/2022]
Abstract
This is Part 2 of a two-part review summarising current knowledge on biomarkers of atherosclerosis. Part 1 addressed serological biomarkers. Here, in part 2 we address genetic and imaging markers, and other developments in predicting risk. Further improvements in risk stratification are expected with the addition of genetic risk scores. In addition to single nucleotide polymorphisms (SNPs), recent advances in epigenetics offer DNA methylation profiles, histone chemical modifications, and micro-RNAs as other promising indicators of atherosclerosis. Imaging biomarkers are better studied and already have a higher degree of clinical applicability in cardiovascular (CV) event prediction and detection of preclinical atherosclerosis. With new methodologies, such as proteomics and metabolomics, discoveries of new clinically applicable biomarkers are expected.
Collapse
Affiliation(s)
- Miha Tibaut
- General Hospital Murska Sobota, Murska Sobota, Slovenia
| | - Martin Caprnda
- 1st Department of Internal Medicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia; Department of Experimental Carcinogenesis, Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Andreja Sinkovič
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | | | - Slavomira Filipova
- Department of Cardiology, National Institute of Cardiovascular Diseases and Slovak Medical University, Bratislava, Slovakia
| | - Katarina Gazdikova
- Department of Nutrition, Faculty of Nursing and Professional Health Studies, Slovak Medical University, Bratislava, Slovakia; Department of General Medicine, Faculty of Medicine, Slovak Medical University, Bratislava, Slovakia.
| | - Ludovit Gaspar
- 1st Department of Internal Medicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia.
| | - Ioana Mozos
- Department of Functional Sciences, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania; Center for Translational Research and Systems Medicine, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Emmanuel E Egom
- Jewish General Hospital and Lady Davis Institute for Medical Research, Montreal, Canada; Department of Cardiology, The Adelaide and Meath Hospital Dublin, Incorporating the National Children Hospital, Dublin, Ireland
| | - Luis Rodrigo
- Faculty of Medicine, University of Oviedo, Central University Hospital of Asturias (HUCA), Oviedo, Spain
| | - Peter Kruzliak
- 2nd Department of Surgery, Center for Vascular Disease, Faculty of Medicine, Masaryk University and St. Anne's University Hospital, Brno, Czech Republic; Department of Internal Medicine, Brothers of Mercy Hospital, Brno, Czech Republic.
| | - Daniel Petrovic
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
44
|
Epigenetic processing in cardiometabolic disease. Atherosclerosis 2018; 281:150-158. [PMID: 30290963 DOI: 10.1016/j.atherosclerosis.2018.09.029] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/24/2018] [Accepted: 09/20/2018] [Indexed: 02/07/2023]
Abstract
Albeit a consistent body of evidence supports the notion that genes influence cardiometabolic features and outcomes, the "non-genetic regulation" of this process is gaining increasing attention. Plastic chemical changes of DNA/histone complexes - known as epigenetic changes - critically determine gene activity by rapidly modifying chromatin accessibility to transcription factors. In this review, we describe the emerging role of chromatin modifications as fine tuners of gene transcription in adipogenesis, insulin resistance, macrophage polarization, immuno-metabolism, endothelial dysfunction and metabolic cardiomyopathy. Epigenetic processing participates in the dynamic interplay among different organs in the cardiometabolic patient. DNA methylation and post-translational histone modifications in both visceral and subcutaneous adipose tissue enable the transcription of genes implicated in lipo- and adipogenesis, inflammation and insulin resistance. Along the same line, complex networks of chromatin modifying enzymes are responsible for impaired nitric oxide bioavailability and defective insulin signalling in the vasculature, thus leading to reduced capillary recruitment and insulin delivery in the liver, skeletal muscle and adipose tissue. Furthermore, changes in methylation status of IL-4, IFNγ and Forkhead box P3 (Foxp3) gene loci are crucial for the polarization of immune cells, thus leading to adipose tissue inflammation and atherosclerosis. Cell-specific epigenetic information could advance our understanding of cardiometabolic processes, thus leading to individualized risk assessment and personalized therapeutic approaches in patients with cardiometabolic disturbances. The development of new chromatin modifying drugs indicates that targeting epigenetic changes is a promising approach to reduce the burden of cardiovascular disease in this setting.
Collapse
|
45
|
Meta-Prediction of MTHFR Gene Polymorphisms and Air Pollution on the Risk of Hypertensive Disorders in Pregnancy Worldwide. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:ijerph15020326. [PMID: 29438331 PMCID: PMC5858395 DOI: 10.3390/ijerph15020326] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/24/2018] [Accepted: 02/05/2018] [Indexed: 11/16/2022]
Abstract
Hypertensive disorders in pregnancy (HDP) are devastating health hazards for both women and children. Both methylenetetrahydrofolate reductase (MTHFR) gene polymorphisms and air pollution can affect health status and result in increased risk of HDP for women. The major objective of this study was to investigate the effect of MTHFR polymorphisms, air pollution, and their interaction on the risk of HDP by using meta-predictive analytics. We searched various databases comprehensively to access all available studies conducted for various ethnic populations from countries worldwide, from 1997 to 2017. Seventy-one studies with 8064 cases and 13,232 controls for MTHFR C677T and 11 studies with 1425 cases and 1859 controls for MTHFR A1298C were included. MTHFR C677T homozygous TT (risk ratio (RR) = 1.28, p < 0.0001) and CT plus TT (RR = 1.07, p = 0.0002) were the risk genotypes, while wild-type CC played a protective role (RR = 0.94, p = 0.0017) for HDP. The meta-predictive analysis found that the percentage of MTHFR C677T TT plus CT (p = 0.044) and CT (p = 0.043) genotypes in the HDP case group were significantly increased with elevated levels of air pollution worldwide. Additionally, in countries with higher air pollution levels, the pregnant women with wild-type CC MTHFR 677 had a protection effect against HDP (p = 0.014), whereas, the homozygous TT of MTHFR C677T polymorphism was a risk genotype for developing HDP. Air pollution level is an environmental factor interacting with increased MTHFR C677T polymorphisms, impacting the susceptibility of HDP for women.
Collapse
|
46
|
Smolkova B, Dusinska M, Gabelova A. Nanomedicine and epigenome. Possible health risks. Food Chem Toxicol 2017; 109:780-796. [PMID: 28705729 DOI: 10.1016/j.fct.2017.07.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 07/08/2017] [Indexed: 02/07/2023]
Abstract
Nanomedicine is an emerging field that combines knowledge of nanotechnology and material science with pharmaceutical and biomedical sciences, aiming to develop nanodrugs with increased efficacy and safety. Compared to conventional therapeutics, nanodrugs manifest higher stability and circulation time, reduced toxicity and improved targeted delivery. Despite the obvious benefit, the accumulation of imaging agents and nanocarriers in the body following their therapeutic or diagnostic application generates concerns about their safety for human health. Numerous toxicology studies have demonstrated that exposure to nanomaterials (NMs) might pose serious risks to humans. Epigenetic modifications, representing a non-genotoxic mechanism of toxicant-induced health effects, are becoming recognized as playing a potential causative role in the aetiology of many diseases including cancer. This review i) provides an overview of recent advances in medical applications of NMs and ii) summarizes current evidence on their possible epigenetic toxicity. To discern potential health risks of NMs, since current data are mostly based upon in vitro and animal models, a better understanding of functional relationships between NM exposure, epigenetic deregulation and phenotype is required.
Collapse
Affiliation(s)
- Bozena Smolkova
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia.
| | - Maria Dusinska
- Health Effects Laboratory MILK, NILU- Norwegian Institute for Air Research, 2007 Kjeller, Norway
| | - Alena Gabelova
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| |
Collapse
|
47
|
The Impact of Environmental Factors in Influencing Epigenetics Related to Oxidative States in the Cardiovascular System. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:2712751. [PMID: 28607629 PMCID: PMC5457758 DOI: 10.1155/2017/2712751] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 04/03/2017] [Accepted: 04/12/2017] [Indexed: 12/14/2022]
Abstract
Oxidative states exert a significant influence on a wide range of biological and molecular processes and functions. When their balance is shifted towards enhanced amounts of free radicals, pathological phenomena can occur, as the generation of reactive oxygen species (ROS) in tissue microenvironment or in the systemic circulation can be detrimental. Epidemic chronic diseases of western societies, such as cardiovascular disease, obesity, and diabetes correlate with the imbalance of redox homeostasis. Current advances in our understanding of epigenetics have revealed a parallel scenario showing the influence of oxidative stress as a major regulator of epigenetic gene regulation via modification of DNA methylation, histones, and microRNAs. This has provided both the biological link and a potential molecular explanation between oxidative stress and cardiovascular/metabolic phenomena. Accordingly, in this review, we will provide current insights on the physiological and pathological impact of changes in oxidative states on cardiovascular disorders, by specifically focusing on the influence of epigenetic regulation. A special emphasis will highlight the effect on epigenetic regulation of human's current life habits, external and environmental factors, including food intake, tobacco, air pollution, and antioxidant-based approaches. Additionally, the strategy to quantify oxidative states in humans in order to determine which biological marker could best match a subject's profile will be discussed.
Collapse
|
48
|
Martino F, Magenta A, Pannarale G, Martino E, Zanoni C, Perla FM, Puddu PE, Barillà F. Epigenetics and cardiovascular risk in childhood. J Cardiovasc Med (Hagerstown) 2017; 17:539-46. [PMID: 27367935 DOI: 10.2459/jcm.0000000000000334] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiovascular disease (CVD) can arise at the early stages of development and growth. Genetic and environmental factors may interact resulting in epigenetic modifications with abnormal phenotypic expression of genetic information without any change in the nucleotide sequence of DNA. Maternal dietary imbalance, inadequate to meet the nutritional needs of the fetus can lead to intrauterine growth retardation, decreased gestational age, low birth weight, excessive post-natal growth and metabolic alterations, with subsequent appearance of CVD risk factors. Fetal exposure to high cholesterol, diabetes and maternal obesity is associated with increased risk and progression of atherosclerosis. Maternal smoking during pregnancy and exposure to various environmental pollutants induce epigenetic alterations of gene expression relevant to the onset or progression of CVD. In children with hypercholesterolemia and/or obesity, oxidative stress activates platelets and monocytes, which release proinflammatory and proatherogenic substances, inducing endothelial dysfunction, decreased Doppler flow-mediated dilation and increased carotid intima-media thickness. Primary prevention of atherosclerosis should be implemented early. It is necessary to identify, through screening, high-risk apparently healthy children and take care of them enforcing healthy lifestyle (mainly consisting of Mediterranean diet and physical activity), prescribing nutraceuticals and eventual medications, if required by a high-risk profile. The key issue is the restoration of endothelial function in the reversible stage of atherosclerosis. Epigenetics may provide new markers for an early identification of children at risk and thereby develop innovative therapies and specific nutritional interventions in critical times.
Collapse
Affiliation(s)
- Francesco Martino
- aDepartment of Pediatrics and Child Neuropsychiatry, Sapienza University of RomebVascular Pathology Laboratory, Fondazione Luigi Monti, Istituto Dermopatico dell'Immacolata-IRCCScDepartment of Cardiovascular, Respiratory, Nephrological, Anesthesiological and Geriatric Sciences, 'Sapienza' University of Rome, Rome, Italy*The authors contributed equally to this work
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Lippi G, Cervellin G. The interplay between genetics, epigenetics and environment in modulating the risk of coronary heart disease. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:460. [PMID: 28090516 DOI: 10.21037/atm.2016.12.29] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Giuseppe Lippi
- Section of Clinical Biochemistry, University of Verona, Verona, Italy
| | | |
Collapse
|
50
|
Visani G, Manti A, Valentini L, Canonico B, Loscocco F, Isidori A, Gabucci E, Gobbi P, Montanari S, Rocchi M, Papa S, Gatti AM. Environmental nanoparticles are significantly over-expressed in acute myeloid leukemia. Leuk Res 2016; 50:50-56. [PMID: 27669365 DOI: 10.1016/j.leukres.2016.09.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 08/05/2016] [Accepted: 09/01/2016] [Indexed: 12/29/2022]
Abstract
The increase in the incidence of acute myeloid leukemia (AML) may suggest a possible environmental etiology. PM2.5 was declared by IARC a Class I carcinogen. No report has focused on particulate environmental pollution together with AML. The study investigated the presence and composition of particulate matter in blood with a Scanning Electron Microscope coupled with an Energy Dispersive Spectroscope, a sensor capable of identifying the composition of foreign bodies. 38 peripheral blood samples, 19 AML cases and 19 healthy controls, were analyzed. A significant overload of particulate matter-derived nanoparticles linked or aggregated to blood components was found in AML patients, while almost absent in matched healthy controls. Two-tailed Student's t-test, MANOVA and Principal Component Analysis indicated that the total numbers of aggregates and particles were statistically different between cases and controls (MANOVA, P<0.001 and P=0.009 respectively). The particles detected showed to contain highly-reactive, non-biocompatible and non-biodegradable metals; in particular, micro- and nano-sized particles grouped in organic/inorganic clusters, with statistically higher frequency of a subgroup of elements in AML samples. The demonstration, for the first time, of an overload of nanoparticles linked to blood components in AML patients could be the basis for a possible, novel pathogenetic mechanism for AML development.
Collapse
Affiliation(s)
- G Visani
- Hematology and Stem Cell Transplant Center, AORMN, Pesaro, Italy.
| | - A Manti
- Dept. of Earth, Life and Environment Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - L Valentini
- Dept. of Earth, Life and Environment Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - B Canonico
- Dept. of Earth, Life and Environment Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - F Loscocco
- Hematology and Stem Cell Transplant Center, AORMN, Pesaro, Italy
| | - A Isidori
- Hematology and Stem Cell Transplant Center, AORMN, Pesaro, Italy
| | - E Gabucci
- Hematology and Stem Cell Transplant Center, AORMN, Pesaro, Italy
| | - P Gobbi
- Dept. of Earth, Life and Environment Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - S Montanari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - M Rocchi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - S Papa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - A M Gatti
- National Council of Research of Italy, Institute for the Science and Technology of Ceramic Materials, Faenza, Italy
| |
Collapse
|