1
|
Rios FJ, de Ciuceis C, Georgiopoulos G, Lazaridis A, Nosalski R, Pavlidis G, Tual-Chalot S, Agabiti-Rosei C, Camargo LL, Dąbrowska E, Quarti-Trevano F, Hellmann M, Masi S, Lopreiato M, Mavraganis G, Mengozzi A, Montezano AC, Stavropoulos K, Winklewski PJ, Wolf J, Costantino S, Doumas M, Gkaliagkousi E, Grassi G, Guzik TJ, Ikonomidis I, Narkiewicz K, Paneni F, Rizzoni D, Stamatelopoulos K, Stellos K, Taddei S, Touyz RM, Virdis A. Mechanisms of Vascular Inflammation and Potential Therapeutic Targets: A Position Paper From the ESH Working Group on Small Arteries. Hypertension 2024; 81:1218-1232. [PMID: 38511317 DOI: 10.1161/hypertensionaha.123.22483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Inflammatory responses in small vessels play an important role in the development of cardiovascular diseases, including hypertension, stroke, and small vessel disease. This involves various complex molecular processes including oxidative stress, inflammasome activation, immune-mediated responses, and protein misfolding, which together contribute to microvascular damage. In addition, epigenetic factors, including DNA methylation, histone modifications, and microRNAs influence vascular inflammation and injury. These phenomena may be acquired during the aging process or due to environmental factors. Activation of proinflammatory signaling pathways and molecular events induce low-grade and chronic inflammation with consequent cardiovascular damage. Identifying mechanism-specific targets might provide opportunities in the development of novel therapeutic approaches. Monoclonal antibodies targeting inflammatory cytokines and epigenetic drugs, show promise in reducing microvascular inflammation and associated cardiovascular diseases. In this article, we provide a comprehensive discussion of the complex mechanisms underlying microvascular inflammation and offer insights into innovative therapeutic strategies that may ameliorate vascular injury in cardiovascular disease.
Collapse
Affiliation(s)
- Francisco J Rios
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Carolina de Ciuceis
- Department of Clinical and Experimental Sciences, University of Brescia (C.d.C., C.A.-R., D.R.)
| | - Georgios Georgiopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School (G.G., G.M., K. Stamatelopoulos), National and Kapodistrian University of Athens
| | - Antonios Lazaridis
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Greece (A.L., E.G.)
| | - Ryszard Nosalski
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute, University of Edinburgh, United Kingdom (R.N., T.J.G.)
- Department of Internal Medicine, Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland (R.N., T.J.G.)
| | - George Pavlidis
- Medical School (G.P., I.I.), National and Kapodistrian University of Athens
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2-Cardiology Department, Attikon Hospital, Athens, Greece (G.P., I.I.)
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, United Kingdom (S.T.-C., K. Stellos)
| | - Claudia Agabiti-Rosei
- Department of Clinical and Experimental Sciences, University of Brescia (C.d.C., C.A.-R., D.R.)
| | - Livia L Camargo
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Edyta Dąbrowska
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Fosca Quarti-Trevano
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (F.Q.-T., G.G.)
| | - Marcin Hellmann
- Cardiac Diagnostics (M.H.), Medical University of Gdansk, Poland
| | - Stefano Masi
- Institute of Cardiovascular Science, University College London, United Kingdom (S.M.)
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| | - Mariarosaria Lopreiato
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| | - Georgios Mavraganis
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School (G.G., G.M., K. Stamatelopoulos), National and Kapodistrian University of Athens
| | - Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland (A.M., F.P.)
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa (A.M.)
| | - Augusto C Montezano
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Konstantinos Stavropoulos
- Second Medical Department, Hippokration Hospital, Aristotle University of Thessaloniki, Greece (K. Stavropoulos)
| | - Pawel J Winklewski
- Departments of Human Physiology (P.J.W.), Medical University of Gdansk, Poland
| | - Jacek Wolf
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Sarah Costantino
- University Heart Center (S.C., F.P.), University Hospital Zurich, Switzerland
| | - Michael Doumas
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Eugenia Gkaliagkousi
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Greece (A.L., E.G.)
| | - Guido Grassi
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (F.Q.-T., G.G.)
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute, University of Edinburgh, United Kingdom (R.N., T.J.G.)
- Department of Internal Medicine, Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland (R.N., T.J.G.)
| | - Ignatios Ikonomidis
- Medical School (G.P., I.I.), National and Kapodistrian University of Athens
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2-Cardiology Department, Attikon Hospital, Athens, Greece (G.P., I.I.)
| | - Krzysztof Narkiewicz
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland (A.M., F.P.)
- University Heart Center (S.C., F.P.), University Hospital Zurich, Switzerland
- Department of Research and Education (F.P.), University Hospital Zurich, Switzerland
| | - Damiano Rizzoni
- Department of Clinical and Experimental Sciences, University of Brescia (C.d.C., C.A.-R., D.R.)
- Division of Medicine, Spedali Civili di Brescia, Italy (D.R.)
| | - Kimon Stamatelopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School (G.G., G.M., K. Stamatelopoulos), National and Kapodistrian University of Athens
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, United Kingdom (S.T.-C., K. Stellos)
- Department of Cardiovascular Research, European Center for Angioscience, Medical Faculty Mannheim (K. Stellos), Heidelberg University, Germany
- Department of Cardiology, University Hospital Mannheim (K. Stellos), Heidelberg University, Germany
- German Centre for Cardiovascular Research, Heidelberg/Mannheim Partner Site (K. Stellos)
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| | - Rhian M Touyz
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| |
Collapse
|
2
|
Wackers P, Dollé MET, van Oostrom CT, van Kerkhof LW. Exploration of genome-wide DNA methylation profiles in night shift workers. Epigenetics 2023; 18:2152637. [PMID: 36457290 PMCID: PMC9980630 DOI: 10.1080/15592294.2022.2152637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
The past decades, studies indicated that night shift work is associated with adverse health effects, however, molecular mechanisms underlying these effects are poorly understood. A few previous studies have hypothesized a role for DNA-methylation (DNAm) in this relationship. We performed a cross-sectional epigenome-wide association study, to investigate if night shift work is associated with genome-wide DNAm changes and DNAm-based biological age acceleration, based on previously developed so-called 'epigenetic clocks.' Short term (2-6 years) and intermediate term (10-16 years) night shift workers, along with age and sex matched dayworkers (non-shift workers) were selected from the Lifelines Cohort Study. For genome-wide methylation analysis the Infinium Methylation EPIC array (Ilumina) was used. Linear regression analyses were used to detect differences in methylation at individual CpG-sites associated with night shift work. Pathway analysis was performed based on KEGG pathways and predictions of age acceleration in night shift workers were performed based on four previously developed epigenetic age calculators. Only in women, differences in methylation at individual CpG-sites were observed between night shift workers and non-shift workers. Most of these differentially methylated positions (DMPs) were observed in intermediate term night shift workers. Pathway analysis shows involvement of pathways related to circadian rhythm and cellular senescence. Increased age acceleration was observed only in short-term night shift workers (men and women). This might be indicative of adaptation to night shift work or a so-called healthy worker effect. In conclusion, these results show that DNA methylation changes are associated with night shift work, specifically in women.
Collapse
Affiliation(s)
- Paul Wackers
- Center for Health Protection, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Martijn E. T. Dollé
- Center for Health Protection, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Conny T.M. van Oostrom
- Center for Health Protection, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Linda W.M. van Kerkhof
- Center for Health Protection, National Institute for Public Health and the Environment, Bilthoven, The Netherlands,CONTACT Linda W.M. van Kerkhof Centre for Health Protection; National Institute for Public Health and the Environment, P.O. Box 1, 3720 BA; Bilthoven, The Netherlands
| |
Collapse
|
3
|
Abrahams Y, Willmer T, Patel O, Samodien E, Muller CJF, Windvogel S, Johnson R, Pheiffer C. A high fat, high sugar diet induces hepatic Peroxisome proliferator-activated receptor gamma coactivator 1-alpha promoter hypermethylation in male Wistar rats. Biochem Biophys Res Commun 2023; 680:25-33. [PMID: 37713959 DOI: 10.1016/j.bbrc.2023.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/31/2023] [Accepted: 09/02/2023] [Indexed: 09/17/2023]
Abstract
Previously we reported that a high fat, high sugar (HFHS) diet induced adiposity, hyperinsulinaemia, hyperleptinaemia, hypertriglyceridaemia and increased liver mass in male Wistar rats. In the present study, the mechanisms underlying the increased liver mass were further elucidated by assessing hepatic lipid accumulation and the expression and methylation status of key metabolic genes using histology, quantitative real-time PCR and pyrosequencing, respectively. The HFHS diet induced hepatic steatosis, increased hepatic triglycerides (1.8-fold, p < 0.001), and increased the expression of sterol regulatory element-binding transcription factor 1 (Srebf1) (2.0-fold, p < 0.001) and peroxisome proliferator-activated receptor gamma (Pparg) (1.7-fold, p = 0.017) in the liver. The expression of peroxisome proliferator-activated receptor gamma coactivator 1 alpha (Pgc1a) was decreased (2.6-fold, p < 0.010), which was accompanied by hypermethylation (p = 0.018) of a conserved CpG site in the promoter of Pgc1a in HFHS fed rats compared to controls. In silico analysis identified putative binding sites for CCAAT/enhancer-binding protein beta (C/EBPß) and hepatocyte nuclear factor 1 (HNF1) within proximity to the hypermethylated CpG. As Pgc1a is a co-activator of several transcription factors regulating multiple metabolic pathways, hypermethylation of this conserved CpG site in the promoter of Pgc1a may be one possible mechanism contributing to the development of hepatic steatosis in response to a HFHS diet. However, further work is required to confirm the role of Pgc1a in steatosis.
Collapse
Affiliation(s)
- Yoonus Abrahams
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa; Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Tarryn Willmer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa; Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa; Division of Cell Biology, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Oelfah Patel
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa; Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
| | - Ebrahim Samodien
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
| | - Christo J F Muller
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa; Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa; Department of Biochemistry and Microbiology, University of Zululand, Kwadlangezwa, South Africa
| | - Shantal Windvogel
- Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Rabia Johnson
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa; Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Carmen Pheiffer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa; Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa; Department of Obstetrics and Gynaecology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
4
|
Glisic M, Raguindin PF, Gemperli A, Taneri PE, Salvador DJ, Voortman T, Marques Vidal P, Papatheodorou SI, Kunutsor SK, Bano A, Ioannidis JPA, Muka T. A 7-Step Guideline for Qualitative Synthesis and Meta-Analysis of Observational Studies in Health Sciences. Public Health Rev 2023; 44:1605454. [PMID: 37260612 PMCID: PMC10227668 DOI: 10.3389/phrs.2023.1605454] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/14/2023] [Indexed: 06/02/2023] Open
Abstract
Objectives: To provide a step-by-step, easy-to-understand, practical guide for systematic review and meta-analysis of observational studies. Methods: A multidisciplinary team of researchers with extensive experience in observational studies and systematic review and meta-analysis was established. Previous guidelines in evidence synthesis were considered. Results: There is inherent variability in observational study design, population, and analysis, making evidence synthesis challenging. We provided a framework and discussed basic meta-analysis concepts to assist reviewers in making informed decisions. We also explained several statistical tools for dealing with heterogeneity, probing for bias, and interpreting findings. Finally, we briefly discussed issues and caveats for translating results into clinical and public health recommendations. Our guideline complements "A 24-step guide on how to design, conduct, and successfully publish a systematic review and meta-analysis in medical research" and addresses peculiarities for observational studies previously unexplored. Conclusion: We provided 7 steps to synthesize evidence from observational studies. We encourage medical and public health practitioners who answer important questions to systematically integrate evidence from observational studies and contribute evidence-based decision-making in health sciences.
Collapse
Affiliation(s)
- Marija Glisic
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Swiss Paraplegic Research, Nottwil, Switzerland
| | - Peter Francis Raguindin
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Swiss Paraplegic Research, Nottwil, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
- Faculty of Health Science and Medicine, University of Lucerne, Lucerne, Switzerland
| | - Armin Gemperli
- Swiss Paraplegic Research, Nottwil, Switzerland
- Institute of Primary and Community Care, University of Lucerne, Lucerne, Switzerland
| | - Petek Eylul Taneri
- HRB-Trials Methodology Research Network, National University of Ireland, Galway, Ireland
| | - Dante Jr. Salvador
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
| | - Trudy Voortman
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, Netherlands
| | - Pedro Marques Vidal
- Department of Medicine, Internal Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | | | - Setor K. Kunutsor
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester, United Kingdom
- Translational Health Sciences, Bristol Medical School, University of Bristol, Southmead Hospital, Bristol, United Kingdom
| | - Arjola Bano
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - John P. A. Ioannidis
- Stanford Prevention Research Center, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, United States
- Department of Statistics, Stanford University, Stanford, CA, United States
- Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA, United States
| | - Taulant Muka
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA, United States
- Epistudia, Bern, Switzerland
| |
Collapse
|
5
|
Svoboda LK, Wang K, Goodrich JM, Jones TR, Colacino JA, Peterson KE, Tellez-Rojo MM, Sartor MA, Dolinoy DC. Perinatal Lead Exposure Promotes Sex-Specific Epigenetic Programming of Disease-Relevant Pathways in Mouse Heart. TOXICS 2023; 11:85. [PMID: 36668811 PMCID: PMC9860846 DOI: 10.3390/toxics11010085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/21/2022] [Accepted: 12/25/2022] [Indexed: 06/17/2023]
Abstract
Environmental contaminants such as the metal lead (Pb) are associated with cardiovascular disease, but the underlying molecular mechanisms are poorly understood. In particular, little is known about how exposure to Pb during early development impacts the cardiac epigenome at any point across the life course and potential differences between sexes. In a mouse model of human-relevant perinatal exposures, we utilized RNA-seq and Enhanced Reduced Representation Bisulfite Sequencing (ERRBS) to investigate the effects of Pb exposure during gestation and lactation on gene expression and DNA methylation, respectively, in the hearts of male and female mice at weaning. For ERRBS, we identified differentially methylated CpGs (DMCs) or differentially methylated 1000 bp regions (DMRs) based on a minimum absolute change in methylation of 10% and an FDR < 0.05. For gene expression data, an FDR < 0.05 was considered significant. No individual genes met the FDR cutoff for gene expression; however, we found that Pb exposure leads to significant changes in the expression of gene pathways relevant to cardiovascular development and disease. We further found that Pb promotes sex-specific changes in DNA methylation at hundreds of gene loci (280 DMCs and 99 DMRs in males, 189 DMCs and 121 DMRs in females), and pathway analysis revealed that these CpGs and regions collectively function in embryonic development. In males, differential methylation also occurred at genes related to immune function and metabolism. We then investigated whether genes exhibiting differential methylation at weaning were also differentially methylated in hearts from a cohort of Pb-exposed mice at adulthood. We found that a single gene, Galnt2, showed differential methylation in both sexes and time points. In a human cohort investigating the influence of prenatal Pb exposure on the epigenome, we also observed an inverse association between first trimester Pb concentrations and adolescent blood leukocyte DNA methylation at a locus in GALNT2, suggesting that this gene may represent a biomarker of Pb exposure across species. Together, these data, across two time points in mice and in a human birth cohort study, collectively demonstrate that Pb exposure promotes sex-specific programming of the cardiac epigenome, and provide potential mechanistic insight into how Pb causes cardiovascular disease.
Collapse
Affiliation(s)
- Laurie K. Svoboda
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Kai Wang
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jaclyn M. Goodrich
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Tamara R. Jones
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Justin A. Colacino
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Karen E. Peterson
- Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Martha M. Tellez-Rojo
- Center for Research on Nutrition and Health, National Institute of Public Health, Cuernavaca 62100, Mexico
| | - Maureen A. Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Dana C. Dolinoy
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| |
Collapse
|
6
|
Zhou S, Zhang J, Zhou C, Gong F, Zhu X, Pan X, Sun J, Gao X, Huang Y. DNA Methylation of Patatin-Like Phospholipase Domain-Containing Protein 6 Gene Contributes to the Risk of Intracranial Aneurysm in Males. Front Aging Neurosci 2022; 14:885680. [PMID: 35898327 PMCID: PMC9309567 DOI: 10.3389/fnagi.2022.885680] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/17/2022] [Indexed: 11/16/2022] Open
Abstract
Objective: This study is aimed to investigate the contribution of patatin-like phospholipase domain-containing protein 6 (PNPLA6) DNA methylation to the risk of intracranial aneurysm (IA) in the Han Chinese population. Methods: A total of 96 age- and sex-matched participants were recruited to evaluate PNPLA6 methylation via bisulfite pyrosequencing. The PNPLA6 mRNA expression in the plasma was determined using real-time quantitative reverse transcription-polymerase chain reaction. Human primary artery smooth muscle cells (HPCASMC) were used for the in vitro function study. Results: PNPLA6 methylation was significantly higher in patients with IA than in healthy controls (p < 0.01). Sex group analysis showed that this correlation appeared in the male group (p < 0.01) but not in the female group (p > 0.05). PNPLA6 methylation was significantly associated with age in all participants (r = 0.306, p = 0.003) and in the control group (r = 0.377, p = 0.008) but not in the IA group (r = 0.127, p = 0.402). Furthermore, the PNPLA6 mRNA expression significantly decreased in patients with IA than that in the controls (p = 0.016). PNPLA6 expression was significantly inversely correlated with elevated DNA methylation in participants (r = −0.825, p < 0.0001). In addition, PNPLA6 transcription was significantly enhanced following treatment with 5-aza-2’-deoxycytidine methylation inhibitor in HPCASMC.The receiver operating characteristic analyses of curves showed that the PNPLA6 mean methylation [area under the curve (AUC) = 0.74, p < 0.001] and mRNA expression (AUC = 0.86, p < 0.001) could have a diagnostic value for patients with IA. Conclusion: Although future functional experiments are required to test our hypothesis, our study demonstrated that PNPLA6 methylation and mRNA expression were significantly associated with the risk of IA; thus, they show potential for use in the early diagnosis of IA.
Collapse
Affiliation(s)
- Shengjun Zhou
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, China
| | - Junjun Zhang
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, China
| | - Chenhui Zhou
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, China
| | - Fanyong Gong
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, China
| | - Xueli Zhu
- Department of Ultrasound, Ningbo First Hospital, Ningbo, China
| | - Xingqiang Pan
- Ningbo Center for Disease Control and Prevention, Ningbo, China
| | - Jie Sun
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, China
- *Correspondence: Jie Sun Xiang Gao Yi Huang
| | - Xiang Gao
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, China
- *Correspondence: Jie Sun Xiang Gao Yi Huang
| | - Yi Huang
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, China
- Medical Research Center, Ningbo First Hospital, Ningbo, China
- *Correspondence: Jie Sun Xiang Gao Yi Huang
| |
Collapse
|
7
|
CircLDLR Modulates the Proliferation and Apoptosis of Vascular Smooth Muscle Cells in Coronary Artery Disease Through miR-26-5p/KDM6A Axis. J Cardiovasc Pharmacol 2022; 80:132-139. [PMID: 35384910 DOI: 10.1097/fjc.0000000000001275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 03/17/2022] [Indexed: 11/25/2022]
Abstract
ABSTRACT The purpose of this study was to investigate the effect of circLDLR on the proliferation and apoptosis of vascular smooth muscle cells (VSMCs) in coronary artery disease and its regulatory mechanism. The expression of KDM6A was detected by qRT-PCR or Western blot. VSMCs were transfected with miR-26-5p mimic/inhibitor or OE KDM6A. Cell proliferation and apoptosis were assessed. Luciferase reporter gene assays were used to examine interactions between miR-26-5p and KDM6A in VSMCs. Downregulation of circLDLR was associated with increased miR-26-5p in coronary artery disease tissues. In addition, circLDLR could inhibit cell proliferation and promote cell apoptosis by regulating miR-26-5p. Moreover, the overexpression of KDM6A reduced VSMCs proliferation and increased apoptosis in an miR-26-5p/circLDLR axis-dependent manner. CircLDLR modulates the proliferation and apoptosis of VSMCs through miR-26-5p/KDM6A axis.
Collapse
|
8
|
Abstract
Sex is a key risk factor for many types of cardiovascular disease. It is imperative to understand the mechanisms underlying sex differences to devise optimal preventive and therapeutic approaches for all individuals. Both biological sex (determined by sex chromosomes and gonadal hormones) and gender (social and cultural behaviors associated with femininity or masculinity) influence differences between men and women in disease susceptibility and pathology. Here, we focus on the application of experimental mouse models that elucidate the influence of 2 components of biological sex-sex chromosome complement (XX or XY) and gonad type (ovaries or testes). These models have revealed that in addition to well-known effects of gonadal hormones, sex chromosome complement influences cardiovascular risk factors, such as plasma cholesterol levels and adiposity, as well as the development of atherosclerosis and pulmonary hypertension. One mechanism by which sex chromosome dosage influences cardiometabolic traits is through sex-biased expression of X chromosome genes that escape X inactivation. These include chromatin-modifying enzymes that regulate gene expression throughout the genome. The identification of factors that determine sex-biased gene expression and cardiometabolic traits will expand our mechanistic understanding of cardiovascular disease processes and provide insight into sex differences that remain throughout the lifespan as gonadal hormone levels alter with age.
Collapse
Affiliation(s)
- Karen Reue
- Department of Human Genetics, David Geffen School of Medicine at UCLA
- Department of Medicine, David Geffen School of Medicine at UCLA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095
| | - Carrie B. Wiese
- Department of Human Genetics, David Geffen School of Medicine at UCLA
| |
Collapse
|
9
|
Chen J, Liu Z, Ma L, Gao S, Fu H, Wang C, Lu A, Wang B, Gu X. Targeting Epigenetics and Non-coding RNAs in Myocardial Infarction: From Mechanisms to Therapeutics. Front Genet 2022; 12:780649. [PMID: 34987550 PMCID: PMC8721121 DOI: 10.3389/fgene.2021.780649] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
Myocardial infarction (MI) is a complicated pathology triggered by numerous environmental and genetic factors. Understanding the effect of epigenetic regulation mechanisms on the cardiovascular disease would advance the field and promote prophylactic methods targeting epigenetic mechanisms. Genetic screening guides individualised MI therapies and surveillance. The present review reported the latest development on the epigenetic regulation of MI in terms of DNA methylation, histone modifications, and microRNA-dependent MI mechanisms and the novel therapies based on epigenetics.
Collapse
Affiliation(s)
- Jinhong Chen
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Zhichao Liu
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Li Ma
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Shengwei Gao
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Huanjie Fu
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Can Wang
- Acupuncture Department, The First Affiliated Hospital of Tianjin University of TCM, Tianjin, China
| | - Anmin Lu
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Baohe Wang
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, Tianjin, China
| | - Xufang Gu
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, Tianjin, China
| |
Collapse
|
10
|
Age at Natural Menopause and Blood Pressure Traits: Mendelian Randomization Study. J Clin Med 2021; 10:jcm10194299. [PMID: 34640315 PMCID: PMC8509463 DOI: 10.3390/jcm10194299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/12/2021] [Accepted: 09/17/2021] [Indexed: 01/10/2023] Open
Abstract
Observational studies suggest that early onset of menopause is associated with increased risk of hypertension. Whether this association is causal or due to residual confounding and/or reverse causation remains undetermined. We aimed to evaluate the observational and causal association between age at natural menopause (ANM) and blood pressure traits in Caucasian women. A cross-sectional and one-sample Mendelian randomization (MR) study was conducted in 4451 postmenopausal women from the CoLaus and Rotterdam studies. Regression models were built with observational data to study the associations of ANM with systolic and diastolic blood pressure (SBP/DBP) and hypertension. One-sample MR analysis was performed by calculating a genetic risk score of 54 ANM-related variants, previously identified in a genome-wide association study (GWAS) on ANM. In the two-sample MR analysis we used the estimates from the ANM-GWAS and association estimates from 168,575 women of the UK Biobank to evaluate ANM-related variants and their causal association with SBP and DBP. Pooled analysis from both cohorts showed that a one-year delay in menopause onset was associated with 2% (95% CI 0; 4) increased odds of having hypertension, and that early menopause was associated with lower DBP (β = −1.31, 95% CI −2.43; −0.18). While one-sample MR did not show a causal association between ANM and blood pressure traits, the two-sample MR showed a positive causal association of ANM with SBP; the last was driven by genes related to DNA damage repair. The present study does not support the hypothesis that early onset of menopause is associated with higher blood pressure. Our results suggest different ANM-related genetic pathways could differently impact blood pressure.
Collapse
|
11
|
Masi S, Ambrosini S, Mohammed SA, Sciarretta S, Lüscher TF, Paneni F, Costantino S. Epigenetic Remodeling in Obesity-Related Vascular Disease. Antioxid Redox Signal 2021; 34:1165-1199. [PMID: 32808539 DOI: 10.1089/ars.2020.8040] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: The prevalence of obesity and cardiometabolic phenotypes is alarmingly increasing across the globe and is associated with atherosclerotic vascular complications and high mortality. In spite of multifactorial interventions, vascular residual risk remains high in this patient population, suggesting the need for breakthrough therapies. The mechanisms underpinning obesity-related vascular disease remain elusive and represent an intense area of investigation. Recent Advances: Epigenetic modifications-defined as environmentally induced chemical changes of DNA and histones that do not affect DNA sequence-are emerging as a potent modulator of gene transcription in the vasculature and might significantly contribute to the development of obesity-induced endothelial dysfunction. DNA methylation and histone post-translational modifications cooperate to build complex epigenetic signals, altering transcriptional networks that are implicated in redox homeostasis, mitochondrial function, vascular inflammation, and perivascular fat homeostasis in patients with cardiometabolic disturbances. Critical Issues: Deciphering the epigenetic landscape in the vasculature is extremely challenging due to the complexity of epigenetic signals and their function in regulating transcription. An overview of the most important epigenetic pathways is required to identify potential molecular targets to treat or prevent obesity-related endothelial dysfunction and atherosclerotic disease. This would enable the employment of precision medicine approaches in this setting. Future Directions: Current and future research efforts in this field entail a better definition of the vascular epigenome in obese patients as well as the unveiling of novel, cell-specific chromatin-modifying drugs that are able to erase specific epigenetic signals that are responsible for maladaptive transcriptional alterations and vascular dysfunction in obese patients. Antioxid. Redox Signal. 34, 1165-1199.
Collapse
Affiliation(s)
- Stefano Masi
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy
| | - Samuele Ambrosini
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| | - Shafeeq A Mohammed
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| | - Sebastiano Sciarretta
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland.,Heart Division, Royal Brompton and Harefield Hospital Trust, National Heart & Lung Institute, 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, Switzerland.,Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Sarah Costantino
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| |
Collapse
|
12
|
Mukherjee N, Arathimos R, Chen S, Kheirkhah Rahimabad P, Han L, Zhang H, Holloway JW, Relton C, Henderson AJ, Arshad SH, Ewart S, Karmaus W. DNA methylation at birth is associated with lung function development until age 26 years. Eur Respir J 2021; 57:2003505. [PMID: 33214203 DOI: 10.1183/13993003.03505-2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022]
Abstract
Little is known about whether DNA methylation (DNAm) of cytosine-phosphate-guanine (CpG) sites at birth predicts patterns of lung function development. We used heel prick DNAm from the F1-generation of Isle of Wight birth cohort (IOWBC-F1) for discovery of CpGs associated with lung function trajectories (forced expiratory volume in 1 s, forced vital capacity, their ratio, and forced expiratory flow at 25-75% of forced vital capacity) over the first 26 years, stratified by sex. We replicated the findings in the Avon Longitudinal Study of Parents and Children (ALSPAC) using cord blood DNAm.Epigenome-wide screening was applied to identify CpGs associated with lung function trajectories in 396 boys and 390 girls of IOWBC-F1. Replication in ALSPAC focussed on lung function at ages 8, 15 and 24 years. Statistically significantly replicated CpGs were investigated for consistency in direction of association between cohorts, stability of DNAm over time in IOWBC-F1, relevant biological processes and for association with gene expression (n=161) in IOWBC F2-generation (IOWBC-F2).Differential DNAm of eight CpGs on genes GLUL, MYCN, HLX, LHX1, COBL, COL18A1, STRA6, and WNT11 involved in developmental processes, were significantly associated with lung function in the same direction in IOWBC-F1 and ALSPAC, and showed stable patterns at birth, aged 10 and 18 years between high and low lung function trajectories in IOWBC-F1. CpGs on LHX1 and COL18A1 were linked to gene expression in IOWBC-F2.In two large cohorts, novel DNAm at birth were associated with patterns of lung function in adolescence and early adulthood providing possible targets for preventative interventions against adverse pulmonary function development.
Collapse
Affiliation(s)
- Nandini Mukherjee
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| | - Ryan Arathimos
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Social Genetic & Developmental Psychiatry Centre, Kings College London, London, UK
- NIHR Biomedical Research Centre for Mental Health, South London and Maudsley NHS Trust, London, UK
| | - Su Chen
- Dept of Mathematical Sciences, The University of Memphis, Memphis, TN, USA
| | - Parnian Kheirkhah Rahimabad
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| | - Luhang Han
- Dept of Mathematical Sciences, The University of Memphis, Memphis, TN, USA
| | - Hongmei Zhang
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| | - John W Holloway
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Caroline Relton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - A John Henderson
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Syed Hasan Arshad
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- The David Hide Asthma and Allergy Research Centre, Isle of Wight, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Susan Ewart
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Wilfried Karmaus
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| |
Collapse
|
13
|
Sex-Specific Alterations in Cardiac DNA Methylation in Adult Mice by Perinatal Lead Exposure. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18020577. [PMID: 33445541 PMCID: PMC7826866 DOI: 10.3390/ijerph18020577] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/27/2020] [Accepted: 01/04/2021] [Indexed: 12/27/2022]
Abstract
Environmental factors play an important role in the etiology of cardiovascular diseases. Cardiovascular diseases exhibit marked sexual dimorphism; however, the sex-specific effects of environmental exposures on cardiac health are incompletely understood. Perinatal and adult exposures to the metal lead (Pb) are linked to several adverse cardiovascular outcomes, but the sex-specific effects of this toxicant on the heart have received little attention. Perinatal environmental exposures can lead to disease through disruption of the normal epigenetic programming that occurs during early development. Using a mouse model of human-relevant perinatal environmental exposure, we investigated the effects of exposure to Pb during gestation and lactation on DNA methylation in the hearts of adult offspring mice (n = 6 per sex). Two weeks prior to mating, dams were assigned to control or Pb acetate (32 ppm) water, and exposure continued until offspring were weaned at three weeks of age. Enhanced reduced-representation bisulfite sequencing was used to measure DNA methylation in the hearts of offspring at five months of age. Although Pb exposure stopped at three weeks of age, we discovered hundreds of differentially methylated cytosines (DMCs) and regions (DMRs) in males and females at five months of age. DMCs/DMRs and their associated genes were sex-specific, with a small, but statistically significant subset overlapping between sexes. Pathway analysis revealed altered methylation of genes important for cardiac and other tissue development in males, and histone demethylation in females. Together, these data demonstrate that perinatal exposure to Pb induces sex-specific changes in cardiac DNA methylation that are present long after cessation of exposure, and highlight the importance of considering sex in environmental epigenetics and mechanistic toxicology studies.
Collapse
|
14
|
AlSiraj Y, Thatcher SE, Blalock E, Saintilnord WN, Daugherty A, Lu HS, Luo W, Shen YH, LeMaire SA, Arnold AP, Cassis LA. Monosomy X in Female Mice Influences the Regional Formation and Augments the Severity of Angiotensin II-Induced Aortopathies. Arterioscler Thromb Vasc Biol 2021; 41:269-283. [PMID: 33054396 PMCID: PMC8259710 DOI: 10.1161/atvbaha.120.314407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Turner syndrome women (monosomy X) have high risk of aortopathies consistent with a role for sex chromosomes in disease development. We demonstrated that sex chromosomes influence regional development of Ang II (angiotensin II)-induced aortopathies in mice. In this study, we determined if the number of X chromosomes regulates regional development of Ang II-induced aortopathies. Approach and Results: We used females with varying numbers of X chromosomes (XX female mice [XXF] or XO female mice [XOF]) on an C57BL/6J (ascending aortopathies) or low-density lipoprotein receptor deficient (Ldlr-/-) background (descending and abdominal aortopathies) compared with XY males (XYM). To induce aortopathies, mice were infused with Ang II. XOF (C57BL/6J) exhibited larger percent increases in ascending aortic lumen diameters than Ang II-infused XXF or XYM. Ang II-infused XOF (Ldlr-/-) exhibited similar incidences of thoracic (XOF, 50%; XYM, 71%) and abdominal aortopathies (XOF, 83%; XYM, 71%) as XYM, which were greater than XXF (XXF, 0%). Abdominal aortic lumen diameters and maximal external diameters were similar between XOF and XYM but greater than XXF, and these effects persisted with extended Ang II infusions. Larger aortic lumen diameters, abdominal aortopathy incidence (XXF, 20%; XOF, 75%), and maximal aneurysm diameters (XXF, 1.02±0.17; XOF, 1.96±0.32 mm; P=0.027) persisted in ovariectomized Ang II-infused XOF mice. Data from RNA-seq demonstrated that X chromosome genes that escape X-inactivation (histone lysine demethylases Kdm5c and Kdm6a) exhibited lower mRNA abundance in aortas of XOF than XXF (P=0.033 and 0.024, respectively). Conversely, DNA methylation was higher in aortas of XOF than XXF (P=0.038). CONCLUSIONS The absence of a second X chromosome promotes diffuse Ang II-induced aortopathies in females.
Collapse
MESH Headings
- Angiotensin II
- Animals
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Thoracic/chemically induced
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/pathology
- DNA Methylation
- Disease Models, Animal
- Female
- Histone Demethylases/genetics
- Histone Demethylases/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Ovariectomy
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Severity of Illness Index
- Turner Syndrome/complications
- Turner Syndrome/genetics
- Mice
Collapse
Affiliation(s)
- Yasir AlSiraj
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington KY
| | - Sean E. Thatcher
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington KY
| | - Eric Blalock
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington KY
| | - Wesley N. Saintilnord
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
| | - Alan Daugherty
- Department of Physiology, University of Kentucky, Lexington KY
- Saha Cardiovascular Research Center, University of Kentucky, Lexington KY
| | - Hong S. Lu
- Department of Physiology, University of Kentucky, Lexington KY
- Saha Cardiovascular Research Center, University of Kentucky, Lexington KY
| | - Wei Luo
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, and Department of Cardiovascular Surgery, Texas Heart Institute, Houston TX
| | - Ying H. Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, and Department of Cardiovascular Surgery, Texas Heart Institute, Houston TX
| | - Scott A. LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, and Department of Cardiovascular Surgery, Texas Heart Institute, Houston TX
| | - Arthur P. Arnold
- Integrative Biology and Physiology, University of California, Los Angeles CA
| | - Lisa A. Cassis
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington KY
| |
Collapse
|