1
|
Li F, Wang Y, Chen S, Liu J, Wu X, Maimati Y, Ding F, Wang X, Shen Y, Chen Q, Li Y, Shen W, Zhang R, Dai Y, Lu L. Nuclear receptor Dax1 promotes atherosclerosis by lipid transport inhibition and autophagy suppression in macrophages. Eur Heart J 2025:ehaf241. [PMID: 40259807 DOI: 10.1093/eurheartj/ehaf241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/27/2024] [Accepted: 03/25/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND AND AIMS Nuclear receptors (NRs) are involved in cardiovascular physiology and pathology. Dosage-sensitive sex reversal, adrenal hypoplasia congenita critical region on the X chromosome, gene 1 (Dax1) is a co-repressor of several protective NRs. However, whether Dax1 influences atherosclerosis remains unclear. This study aims to explore the role of Dax1 in atherogenesis and find a pharmacological approach targeting Dax1 to prevent atherosclerosis. METHODS Dax1 levels were examined in human atherosclerotic arteries. Atherosclerosis animal models were established in mice with macrophage-specific Dax1 knockdown following AAV8-PCSK9 administration and double knockout of macrophage Dax1 and ApoE to evaluate the role of Dax1. Transcriptomic and proteomic analyses were employed to decipher the underlying mechanisms. 2'-Deoxycytidine, an inhibitor of Dax1, was used to verify the effects of Dax1 in macrophages and in mice with atherosclerosis. RESULTS Dax1 mRNA level was up-regulated among NRs in atherosclerotic arteries compared to non-atherosclerotic arteries. The elevation of Dax1 was prominent in the macrophages of atherosclerotic arteries. Macrophage-specific Dax1 knockout mice had less atherosclerosis than controls. Mechanistically, Dax1 inhibited liver X receptor alpha (LXRα), and interacted directly with transcription factor EB (TFEB) to suppress autophagy, resulting in lipid accumulation and inflammation in macrophages. Additionally, 2'-deoxycytidine concentration dependently decreased Dax1 levels, enhanced autophagy, reduced lipid accumulation, and inhibited atherosclerosis in mice. CONCLUSIONS This study demonstrates that Dax1 levels are increased in atherosclerotic plaques. Dax1 promotes atherosclerosis by interacting with TFEB to suppress autophagy and inhibiting LXRα for lipid transport in macrophage, indicating that Dax1 is a potential target for atherosclerosis.
Collapse
Affiliation(s)
- Feifei Li
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Yixuan Wang
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Shuai Chen
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Jingmeng Liu
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Xinrui Wu
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Yipaerguli Maimati
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Fenghua Ding
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Xiaoqun Wang
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Ying Shen
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Qiujing Chen
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Yunqi Li
- National Research Center for Translational Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Weifeng Shen
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Yang Dai
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| | - Lin Lu
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
- National Research Center for Translational Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, PR China
| |
Collapse
|
2
|
Kayzuka C, Rondon-Pereira VC, Nogueira Tavares C, Pacheco Pachado M, Monica FZ, Tanus-Santos JE, Lacchini R. Epigenetics is involved in the pleiotropic effects of statins. Expert Opin Drug Metab Toxicol 2025:1-13. [PMID: 40208655 DOI: 10.1080/17425255.2025.2491732] [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: 08/31/2024] [Revised: 02/18/2025] [Accepted: 04/07/2025] [Indexed: 04/11/2025]
Abstract
INTRODUCTION Statins have significantly reduced mortality from cardiovascular diseases by lowering serum cholesterol levels. Beyond their lipid-lowering effects, statins improve vascular function, reduce inflammation, decrease reactive oxygen species (ROS) formation, and stabilize atherosclerotic plaques. However, the mechanisms underlying these pleiotropic effects remain unclear. AREA COVERED This narrative review summarizes and discusses epigenetic mechanisms that may explain part of the pleiotropic effects of statins. This approach allows for a reevaluation of statin use beyond its cholesterol-lowering benefits. A structured search was conducted in the PubMed and Scopus databases using specific search terms, including articles published up to August 2024. EXPERT OPINION The pleiotropic effects of statins, including those mediated by the isoprenoid pathway, partially explain their clinical benefits. By inhibiting histone deacetylases (HDACs, the 'erasers') and DNA methyltransferases (DNMTs, the 'writers'), statins promote increased histone acetylation and reduced DNA methylation at gene promoter regions. These epigenetic modifications enhance chromatin accessibility, facilitating gene transcription and protecting the cardiovascular system. Further investigation into these epigenetic mechanisms could support the repositioning of statins for broader therapeutic applications. Statins may have benefits extending beyond their role in managing hypercholesterolemia, as their pleiotropic effects contribute to the prevention of cardiovascular disease-related mortality through mechanisms independent of LDL cholesterol reduction.
Collapse
Affiliation(s)
- Cezar Kayzuka
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto, Brazil
| | | | - Cecilia Nogueira Tavares
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Mayra Pacheco Pachado
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Fabiola Zakia Monica
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Jose Eduardo Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Riccardo Lacchini
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
3
|
Zaina S. The functional significance of vascular DNA hypermethylation in atherosclerosis: a historical perspective. Front Pharmacol 2025; 16:1562674. [PMID: 40303930 PMCID: PMC12037548 DOI: 10.3389/fphar.2025.1562674] [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] [Received: 01/17/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
A decade ago, independent mechanistic and descriptive epigenomics data demonstrated for the first time that vascular DNA hypermethylation is a landmark of and causal factor in human and murine atherosclerosis. Since then, a flurry of converging evidence has assigned a prominent role to vascular DNA hypermethylation across the natural history of cardiovascular disease (CVD), from the exposure to risk factors, to the onset and progression of the atheroma. DNA hypermethylation is induced by and mediates the metabolic outcomes of high-fat diets and CVD risk-enhancing lipids in several models. Early-stage atheroma DNA is hypermethylated compared to normal adjacent tissue, and that trend is amplified as the atheroma progresses. That evidence has resulted in a strong interest for epigenetic drugs in CVD. Crucially, the DNA methylation inhibitor azacytidine has been singled out as a potent guardian of the contractile, anti-atherogenic phenotype of smooth muscle cells (SMC). Those findings are gaining relevance, as the antiatherogenic effects of the anticancer drugs azacytidine and decitabine fit into the recently revived hypothesis that the atheroma is a SMC-driven cancer-like mass. Finally, this 10-year anniversary has been marked by the first report that nanoparticles loaded with a DNA methyltransferase inhibitor drug are anti-inflammatory and inhibit murine atherosclerosis. Exciting work lies ahead to assess whether DNA hypermethylation is a practical and effective target to prevent or cure human atherosclerosis.
Collapse
Affiliation(s)
- Silvio Zaina
- Department of Medical Sciences, Division of Health Sciences, Leon Campus, University of Guanajuato, Leon, Mexico
| |
Collapse
|
4
|
Hou X, Li Z, Lin J, Lin W, Li L, Zheng X, Lai X, Zhu L, Guo P, Cai F, Zhang J, Li W, Yang C, Dai Y. Single-cell transcriptome integrated with genome-wide association study reveals heterogeneity of carotid and femoral plaques and its association with plaque stability. Sci Rep 2025; 15:11812. [PMID: 40189611 PMCID: PMC11973204 DOI: 10.1038/s41598-025-96434-4] [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: 01/28/2025] [Accepted: 03/28/2025] [Indexed: 04/09/2025] Open
Abstract
Carotid and femoral plaques exhibit varying degrees of stability; however, the relationships of different genes/cell types with plaque embolism are poorly understood. We evaluated differential gene/cell expression and investigated the cells/genes associated with carotid and femoral artery plaque embolism. sc-RNA-seq and bulk RNA data were obtained to identify differentially expressed genes (DEGs). Seven machine learning models were trained, and the top 10 DEGs across all models were selected. The most disturbed cells in carotid and femoral artery plaques were identified using Augur, while the genes and cells in the carotid plaque associated with embolism were analyzed through scPagwas. The differences in most disturbed cells and embolism-related cells were further analyzed. Compared with femoral plaques, carotid plaques had 80 downregulated and 90 upregulated genes. Machine learning identified the key DEGs between carotid and femoral plaques were predominantly from the HOX gene family. Natural Killer (NK) cells were the most significantly disturbed cells between carotid and femoral plaques, and they may be most strongly associated with plaque embolism. Among the differential genes in NK cells, CD2 was most associated with embolism. Our research may offer new insights into atherosclerosis at different locations.
Collapse
Affiliation(s)
- Xinhuang Hou
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China.
- Department of Vascular Surgery, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| | - Zhipeng Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Jun Lin
- Trauma Center and Emergency Surgery Department, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Wei Lin
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
| | - Luyao Li
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
| | - Xiaoqi Zheng
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
| | - Xiaoling Lai
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
| | - Lin Zhu
- Department of Nutrition and Food Safety, School of Public Health, Fujian Medical University, Fuzhou, 350122, China
| | - Pingfan Guo
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
- Department of Vascular Surgery, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Fanggang Cai
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
- Department of Vascular Surgery, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Jinchi Zhang
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
- Department of Vascular Surgery, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Wanglong Li
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
- Department of Vascular Surgery, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Changwei Yang
- Department of Nutrition and Food Safety, School of Public Health, Fujian Medical University, Fuzhou, 350122, China.
| | - Yiquan Dai
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China.
- Department of Vascular Surgery, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
5
|
Ni Y, Cao J, Li Y, Qi X. SOX11 silence inhibits atherosclerosis progression in ApoE-deficient mice by alleviating endothelial dysfunction. Exp Cell Res 2025; 445:114422. [PMID: 39805338 DOI: 10.1016/j.yexcr.2025.114422] [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: 09/25/2024] [Revised: 01/02/2025] [Accepted: 01/11/2025] [Indexed: 01/16/2025]
Abstract
SRY-Box Transcription Factor-11 (SOX11) is a transcriptional regulatory factor that plays a crucial role in inflammatory responses. However, its involvement in atherosclerosis (AS), a cardiovascular disease driven by endothelial cell inflammation, remains unknown. This study aims to elucidate the role of SOX11 in AS. The expression of SOX11 was found to be elevated in the aortic tissue of AS mice induced by feeding ApoE-deficient mice a high-fat diet. Knockdown of SOX11 using lentiviral-mediated SOX11-specific shRNA via tail vein injection resulted in a reduction in plaque area and lipid deposition within plaques at the aortic root. Furthermore, silencing SOX11 led to decreased expression of cell adhesion factors Intercellular Cell Adhesion Molecule-1 and Vascular Cell Adhesion Molecule-1, as well as reduced levels of inflammatory factors Interleukin (IL)-6, IL-1β, and chemokine Monocyte Chemotactic Protein-1. In the human umbilical vein endothelial cells (HUVECs) induced by Tumor Necrosis Factor (TNF)-α, increased inflammation was observed at the cellular level, along with enhanced monocyte adhesion. Infection of HUVECs with lentivirus carrying specific shRNA targeting SOX11 inhibited inflammatory response. Mechanistically, chromatin immunoprecipitation (ChIP)-PCR results revealed that SOX11 bound to the promoters of downstream target genes Tumor Necrosis Factor Receptor-Associated Factor-1 (TRAF1), Cluster of Differentiation (CD)40, and CD36, positively regulating their transcription. In conclusion, SOX11 plays a pivotal role in promoting endothelial cell inflammation. Suppression of SOX11 reduces endothelial cell inflammation by inhibiting the transcription of TRAF1, CD40, and CD36, thereby impeding the progression of atherosclerosis.
Collapse
Affiliation(s)
- Yanhui Ni
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, Hebei, China; Department of Cardiology, Hebei General Hospital, Shijiazhuang, 050051, Hebei, China
| | - Jingjing Cao
- Department of Rheumatology and Immunology, Hebei General Hospital, Shijiazhuang, 050051, Hebei, China
| | - Yuxuan Li
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, 050051, Hebei, China
| | - Xiaoyong Qi
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, Hebei, China; Department of Cardiology, Hebei General Hospital, Shijiazhuang, 050051, Hebei, China.
| |
Collapse
|
6
|
Mishra S, Puzhankara L. Periodontal Tissue Homoeostasis, Immunity, the Red Complex Pathogens, and Dysbiosis: Unraveling the microRNA Effect. Microrna 2025; 14:9-18. [PMID: 39069708 DOI: 10.2174/0122115366305491240708060422] [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: 02/12/2024] [Revised: 05/20/2024] [Accepted: 06/10/2024] [Indexed: 07/30/2024]
Abstract
microRNAs are a family of small, non-coding RNA molecules that can regulate the translation of messenger RNAs (mRNAs). Numerous miRNAs have been proposed as potential indicators for periodontal disease, and their regulation might serve as a potent means of restricting the disease process. MiRNAs act as important immune system regulators that promote the production of many cytokines, including interferon (IFN), tumour necrosis factor (TNF), and IL-1as well as RANK. Investigations pertaining to the use of specific miRNAs as therapeutic agents are underway. They can influence a variety of regulatory organs and target several genes. Additionally, distinct components of the same expression pathway can be controlled by combining miRNAs and their antagonists. In recent years, many miRNA delivery methods have been created for therapeutic applications. Studies pertaining to the role of miRNAs in periodontal disease pathogenesis may pave the way for the use of miRNAs as biomarkers of periodontal disease. A complete understanding of the role of miRNA in periodontal disease and its mechanism of action can pave the way towards therapeutic strategies that can reduce or even prevent the progression of periodontal diseases.
Collapse
Affiliation(s)
- Swastik Mishra
- Department of Periodontology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher education, Manipal, Karnataka, 576104, India
| | - Lakshmi Puzhankara
- Department of Periodontology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher education, Manipal, Karnataka, 576104, India
| |
Collapse
|
7
|
Qiu Y, Xu Q, Xie P, He C, Li Q, Yao X, Mao Y, Wu X, Zhang T. Epigenetic modifications and emerging therapeutic targets in cardiovascular aging and diseases. Pharmacol Res 2025; 211:107546. [PMID: 39674563 DOI: 10.1016/j.phrs.2024.107546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
The complex mechanisms underlying the development of cardiovascular diseases remain not fully elucidated. Epigenetics, which modulates gene expression without DNA sequence changes, is shedding light on these mechanisms and their heritable effects. This review focus on epigenetic regulation in cardiovascular aging and diseases, detailing specific epigenetic enzymes such as DNA methyltransferases (DNMTs), histone acetyltransferases (HATs), and histone deacetylases (HDACs), which serve as writers or erasers that modify the epigenetic landscape. We also discuss the readers of these modifications, such as the 5-methylcytosine binding domain proteins, and the erasers ten-eleven translocation (TET) proteins. The emerging role of RNA methylation, particularly N6-methyladenosine (m6A), in cardiovascular pathogenesis is also discussed. We summarize potential therapeutic targets, such as key enzymes and their inhibitors, including DNMT inhibitors like 5-azacytidine and decitabine, HDAC inhibitors like belinostat and givinotide, some of which have been approved by the FDA for various malignancies, suggesting their potential in treating cardiovascular diseases. Furthermore, we highlight the role of novel histone modifications and their associated enzymes, which are emerging as potential therapeutic targets in cardiovascular diseases. Thus, by incorporating the recent studies involving patients with cardiovascular aging and diseases, we aim to provide a more detailed and updated review that reflects the advancements in the field of epigenetic modification in cardiovascular diseases.
Collapse
Affiliation(s)
- Yurou Qiu
- GMU-GIBH Joint School of Life Sciences, Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, The Sixth School of Clinical Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Qing Xu
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Peichen Xie
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Chenshuang He
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Qiuchan Li
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Xin Yao
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Yang Mao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Xiaoqian Wu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, PR China.
| | - Tiejun Zhang
- GMU-GIBH Joint School of Life Sciences, Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, The Sixth School of Clinical Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
8
|
Andersen CJ, Fernandez ML. Emerging Biomarkers and Determinants of Lipoprotein Profiles to Predict CVD Risk: Implications for Precision Nutrition. Nutrients 2024; 17:42. [PMID: 39796476 PMCID: PMC11722654 DOI: 10.3390/nu17010042] [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: 12/12/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Biomarkers constitute a valuable tool to diagnose both the incidence and the prevalence of chronic diseases and may help to inform the design and effectiveness of precision nutrition interventions. Cardiovascular disease (CVD) continues to be the foremost cause of death all over the world. While the reasons that lead to increased risk for CVD are multifactorial, dyslipidemias, plasma concentrations of specific lipoproteins, and dynamic measures of lipoprotein function are strong biomarkers to predict and document coronary heart disease incidence. The aim of this review is to provide a comprehensive evaluation of the biomarkers and emerging approaches that can be utilized to characterize lipoprotein profiles as predictive tools for assessing CVD risk, including the assessment of traditional clinical lipid panels, measures of lipoprotein efflux capacity and inflammatory and antioxidant activity, and omics-based characterization of lipoprotein composition and regulators of lipoprotein metabolism. In addition, we discuss demographic, genetic, metagenomic, and lifestyle determinants of lipoprotein profiles-such as age, sex, gene variants and single-nucleotide polymorphisms, gut microbiome profiles, dietary patterns, physical inactivity, obesity status, smoking and alcohol intake, and stress-which are likely to be essential factors to explain interindividual responses to precision nutrition recommendations to mitigate CVD risk.
Collapse
Affiliation(s)
- Catherine J. Andersen
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA;
| | - Maria Luz Fernandez
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA;
- School of Nutrition and Wellness, University of Arizona, Tucson, AZ 85712, USA
| |
Collapse
|
9
|
Tanashyan MM, Shabalina AA, Annushkin VA, Mazur AS, Kuznetsova PI, Raskurazhev AA. Circulating microRNAs in Carotid Atherosclerosis: Complex Interplay and Possible Associations with Atherothrombotic Stroke. Int J Mol Sci 2024; 25:10026. [PMID: 39337512 PMCID: PMC11432131 DOI: 10.3390/ijms251810026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disorder which remains the main cause of cardiovascular morbidity and mortality, with carotid atherosclerosis (CA) being a major cause of ischemic stroke. Epigenetic regulation plays a significant role in CA progression and stroke, yet the impact of circulating microRNA expression, associated with atherogenesis, has not been clearly defined. We included 81 patients with moderate-severe CA (mean age 67 ± 7 years, 53% male), 42% of whom had prior ipsilateral ischemic stroke (i.e., were symptomatic). A total of 24 miRs were identified and their plasma expression levels were measured. We observed that several microRNAs were up-regulated in stroke survivors, namely miR-200c-3p (30.6 vs. 29.7, p = 0.047), miR-106b-3p (31.01 vs. 30.25, p = 0.004), and miR-494-5p (39 vs. 33, p < 0.001), while others (miR183-3p [25.5 vs. 28.6, p < 0.001], miR-126-5p [35.6 vs. 37.1, p = 0.03], and miR-216-3p [12.34 vs. 16.2, p < 0.001]) had lower plasma levels in symptomatic patients. In a multivariable logistic regression model for symptomatic CA, the only miRs showing statistical significance were miR-106b-5p, miR-183-3p, miR-216-3p, and miR-494-5p. Cluster analysis demonstrated differential miR expression in CA patients depending on their stroke status. Epigenetic modulation, represented as complex interplay between circulating miRs of different atherogenic potential, may play a significant role in CA development and progression. In our study, we show possible candidates for future research regarding CA and stroke.
Collapse
Affiliation(s)
- Marine M Tanashyan
- Research Center of Neurology, 80, Volokolamskoe Shosse, 125367 Moscow, Russia
| | - Alla A Shabalina
- Laboratory of Hemorheology, Hemostasis and Pharmacokinetics with Clinical Laboratory Diagnostics, Research Center of Neurology, 80 Volokolamskoye Shosse, 125367 Moscow, Russia
| | | | - Andrey S Mazur
- Research Center of Neurology, 80, Volokolamskoe Shosse, 125367 Moscow, Russia
| | - Polina I Kuznetsova
- Research Center of Neurology, 80, Volokolamskoe Shosse, 125367 Moscow, Russia
| | - Anton A Raskurazhev
- Research Center of Neurology, 80, Volokolamskoe Shosse, 125367 Moscow, Russia
| |
Collapse
|
10
|
Liu Y, Wu Y, Wang C, Hu W, Zou S, Ren H, Zuo Y, Qu L. MiR-127-3p enhances macrophagic proliferation via disturbing fatty acid profiles and oxidative phosphorylation in atherosclerosis. J Mol Cell Cardiol 2024; 193:36-52. [PMID: 38795767 DOI: 10.1016/j.yjmcc.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 05/05/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND Atherosclerosis is a chronic pathology, leading to acute coronary heart disease or stroke. MiR-127 has been found significantly upregulated in advanced atherosclerosis. But its function in atherosclerosis remains unexplored. We explored the role of miR-127-3p in regulating atherosclerosis development and its downstream mechanisms. METHODS The expression profile of miR-127 in carotid atherosclerotic plaques of 23 patients with severe carotid stenosis was detected by RT-qPCR and in situ hybridization. Primary bone marrow-derived macrophages (BMDM) stimulated with oxidized low-density lipoprotein were used as an in vitro model. CCK-8, EdU, RT-qPCR, and flow cytometry were used to detect the proliferative capacity and polarization of BMDM, which were infected by lentivirus-carrying plasmid to upregulate or downregulate miR-127-3p expression, respectively. RNA sequencing combined with bioinformatic analysis and targeted fatty acid metabolomics approach were used to detect the transcriptome and lipid metabolites. The association between miR-127-3p and its target was verified by dual-luciferase activity reporting and Western blotting. Oxygen consumption rate of BMDM were detected using seahorse analysis. High-cholesterol-diet-fed low density lipoprotein deficient (LDLR-/-) mice, with-or-without carotid tandem-stenosis surgery, were treated with miR-127-3p agomir or antagomir to examine its effect on plaque development and stability. RESULTS miR-127-3p, not -5p, is elevated in human advanced carotid atheroma and its expression is positively associated with macrophage accummulation in plaques. In vitro, miR-127-3p-overexpressed macrophage exhibites increased proliferation capacity and facilitates M1 polariztion whereas the contrary trend is present in miR-127-3p-inhibited macrophage. Stearoyl-CoA desaturase-1 (SCD1) is one potential target of miR-127-3p. miR-127-3p mimics decreases the activity of 3' untranslated regions of SCD-1. Furthermore, miR-127-3p downregulates SCD1 expression, and reversing the expression of SCD1 attenuates the increased proliferation induced by miR-127-3p overexpression in macrophage. miR-127-3p overexpression could also lead to decreased content of unsaturated fatty acids (UFAs), increased content of acetyl CoA and increased level of oxidative phosphorylation. In vivo, miR-127-3p agomir significantly increases atherosclerosis progression, macrophage proliferation and decreases SCD1 expression and the content of UFAs in aortic plaques of LDLR-/- mice. Conversely, miR-127-3p antagomir attenuated atherosclerosis, macrophage proliferation in LDLR-/- mice, and enhanced carotid plaque stability in mice with vulnerable plaque induced. CONCLUSION MiR-127-3p enhances proliferation in macrophages through downregulating SCD-1 expression and decreasing the content of unsaturated fatty acid, thereby promoting atherosclerosis development and decreasing plaque stability. miR-127-3p/SCD1/UFAs might provide potential therapeutic target for anti-inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Yandong Liu
- Department of Geriatrics, 905th Hospital of PLA NAVY, Shanghai, China; Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Yicheng Wu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Chao Wang
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Weilin Hu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Sili Zou
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Huiqiong Ren
- Department of Geriatrics, 905th Hospital of PLA NAVY, Shanghai, China.
| | - Yong Zuo
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lefeng Qu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
11
|
Kolaszyńska O, Lorkowski J. Artificial Intelligence in Cardiology and Atherosclerosis in the Context of Precision Medicine: A Scoping Review. Appl Bionics Biomech 2024; 2024:2991243. [PMID: 38715681 PMCID: PMC11074834 DOI: 10.1155/2024/2991243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/01/2024] [Accepted: 03/07/2024] [Indexed: 01/31/2025] Open
Abstract
Cardiovascular diseases remain the main cause of death worldwide which makes it essential to better understand, diagnose, and treat atherosclerosis. Artificial intelligence (AI) and novel technological solutions offer us new possibilities and enable the practice of individually tailored medicine. The study was performed using the PRISMA protocol. As of January 10, 2023, the analysis has been based on a review of 457 identified articles in PubMed and MEDLINE databases. The search covered reviews, original articles, meta-analyses, comments, and editorials published in the years 2009-2023. In total, 123 articles met inclusion criteria. The results were divided into the subsections presented in the review (genome-wide association studies, radiomics, and other studies). This paper presents actual knowledge concerning atherosclerosis, in silico, and big data analyses in cardiology that affect the way medicine is practiced in order to create an individual approach and adjust the therapy of atherosclerosis.
Collapse
Affiliation(s)
- Oliwia Kolaszyńska
- Department of Internal Medicine, Asklepios Clinic Uckermark, Am Klinikum 1, 16303, Schwedt/Oder, Germany
| | - Jacek Lorkowski
- Department of Orthopedics, Traumatology and Sports Medicine, Central Clinical Hospital of the Ministry of Internal Affairs and Administration, 137 Woloska Street, Warsaw 02-507, Poland
- Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
12
|
Mandić K, Milutin Gašperov N, Božinović K, Dediol E, Krasić J, Sinčić N, Grce M, Sabol I, Barešić A. Integrative analysis in head and neck cancer reveals distinct role of miRNome and methylome as tumour epigenetic drivers. Sci Rep 2024; 14:9062. [PMID: 38643268 PMCID: PMC11032388 DOI: 10.1038/s41598-024-59312-z] [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: 01/11/2024] [Accepted: 04/09/2024] [Indexed: 04/22/2024] Open
Abstract
Head and neck cancer is the sixth most common malignancy worldwide, with the relatively low 5-year survival rate, mainly because it is diagnosed at a late stage. Infection with HPV is a well known aetiology, which affects the nature of these cancers and patients' survival. Besides, it is considered that the main driving force for this type of cancer could be epigenetics. In this study we aimed to find potential epigenetic biomarkers, by integrating miRNome, methylome, and transcriptome analyses. From the fresh head and neck cancer tissue samples, we chose a group for miRNome, methylome and transcriptome profiling, in comparison to adequate control samples. Bioinformatics analyses are performed in R v4.2.2. Count normalisation and group differential expression for mRNA and the previously obtained miRNA count data was performed with DESeq2 v1.36. Gene set enrichment analysis was performed and visualised using gProfiler2 v0.2.1 Identification of miRNA targets was performed by querying in miRTarBase using multiMiR v1.18.0. Annotation of CpG sites merging into islands was obtained from RnBeads.hg19 v1.28.0. package. For the integrative analysis we performed kmeans clustering using stats v4.2.2 package, using 8-12 clusters and nstart 100. We found that transcriptome analysis divides samples into cancers and controls clusters, with no relation to HPV status or cancer anatomical location. Differentially expressed genes (n = 2781) were predominantly associated with signalling pathways of tumour progression. We identified a cluster of genes under the control of the transcription factor E2F that are significantly underexpressed in cancer tissue, as well as T cell immunity genes and genes related to regulation of transcription. Among overexpressed genes in tumours we found those that belong to cell cycle regulation and vasculature. A small number of genes were found significantly differentially expressed in HPV-positive versus HPV-negative tumours (for example NEFH, ZFR2, TAF7L, ZNF541, and TYMS). In this comprehensive study on an overlapping set of samples where the integration of miRNome, methylome and transcriptome analysis were performed for head and neck cancer, we demonstrated that the majority of genes were associated exclusively with miRNome or methylome and, to a lesser extent, under the control of both epigenetic mechanisms.
Collapse
Affiliation(s)
- Katarina Mandić
- Division of Electronics, Ruđer Bošković Institute, Zagreb, Croatia
| | | | - Ksenija Božinović
- Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia
| | - Emil Dediol
- Department of Maxillofacial Surgery, Clinical Hospital Dubrava, Zagreb, Croatia
| | - Jure Krasić
- Department of Medical Biology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Nino Sinčić
- Department of Medical Biology, University of Zagreb School of Medicine, Zagreb, Croatia
- Centre of Excellence in Reproductive and Regenerative Medicine, University of Zagreb School of Medicine, Zagreb, Croatia
- Biomedical Research Centre Šalata, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Magdalena Grce
- Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia
| | - Ivan Sabol
- Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia.
| | - Anja Barešić
- Division of Electronics, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
13
|
Wu X, Zhang H. Omics Approaches Unveiling the Biology of Human Atherosclerotic Plaques. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:482-498. [PMID: 38280419 PMCID: PMC10988765 DOI: 10.1016/j.ajpath.2023.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 12/16/2023] [Accepted: 12/20/2023] [Indexed: 01/29/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall, characterized by the buildup of plaques with the accumulation and transformation of lipids, immune cells, vascular smooth muscle cells, and necrotic cell debris. Plaques with collagen-poor thin fibrous caps infiltrated by macrophages and lymphocytes are considered unstable because they are at the greatest risk of rupture and clinical events. However, the current histologic definition of plaque types may not fully capture the complex molecular nature of atherosclerotic plaque biology and the underlying mechanisms contributing to plaque progression, rupture, and erosion. The advances in omics technologies have changed the understanding of atherosclerosis plaque biology, offering new possibilities to improve risk prediction and discover novel therapeutic targets. Genomic studies have shed light on the genetic predisposition to atherosclerosis, and integrative genomic analyses expedite the translation of genomic discoveries. Transcriptomic, proteomic, metabolomic, and lipidomic studies have refined the understanding of the molecular signature of atherosclerotic plaques, aiding in data-driven hypothesis generation for mechanistic studies and offering new prospects for biomarker discovery. Furthermore, advancements in single-cell technologies and emerging spatial analysis techniques have unveiled the heterogeneity and plasticity of plaque cells. This review discusses key omics-based discoveries that have advanced the understanding of human atherosclerotic plaque biology, focusing on insights derived from omics profiling of human atherosclerotic vascular specimens.
Collapse
Affiliation(s)
- Xun Wu
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Hanrui Zhang
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York.
| |
Collapse
|
14
|
Jiang QL, Li T, Xu Q, Zeng Y, Wang W, Zhang BT, Yao QP, Jiang R, Jiang J. Methyl donor diet attenuates intimal hyperplasia after vascular injury in rats. J Nutr Biochem 2024; 123:109486. [PMID: 37844765 DOI: 10.1016/j.jnutbio.2023.109486] [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: 03/05/2023] [Revised: 09/16/2023] [Accepted: 10/12/2023] [Indexed: 10/18/2023]
Abstract
Environmental factors, particularly dietary habits, play an important role in cardiovascular disease susceptibility and progression through epigenetic modification. Previous studies have shown that hyperplastic vascular intima after endarterectomy is characterized by genome-wide hypomethylation. The purpose of this study was to investigate whether methyl donor diet affects intimal hyperplasia and the possible mechanisms involved. Intimal hyperplasia was induced in SD rats by carotid artery balloon injury. From 8 d before surgery to 28 d after surgery, the animals were fed a normal diet (ND) or a methyl donor diet (MD) supplemented with folic acid, vitamin B12, choline, betaine, and zinc. Carotid artery intimal hyperplasia was observed by histology, the effect of MD on carotid protein expression was analyzed by proteomics, functional clustering, signaling pathway, and upstream-downstream relationship of differentially expressed proteins were analyzed by bioinformatics. Results showed that MD attenuated balloon injury-induced intimal hyperplasia in rat carotid arteries. Proteomic analysis showed that there were many differentially expressed proteins in the common carotid arteries of rats fed with two different diets. The differentially expressed proteins are mainly related to the composition and function of the extracellular matrix (EMC), and changes in the EMC can lead to vascular remodeling by affecting fibrosis and stiffness of the blood vessel wall. Changes in the levels of vasculotropic proteins such as S100A9, ILF3, Serpinh1, Fbln5, LOX, HSPG2, and Fmod may be the reason why MD attenuates intimal hyperplasia. Supplementation with methyl donor nutrients may be a beneficial measure to prevent pathological vascular remodeling after injury.
Collapse
Affiliation(s)
- Qi-Lan Jiang
- Department of Clinical Nutrition, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease/Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Qin Xu
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yang Zeng
- Department of Orthodontics, the Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wei Wang
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Bo-Tao Zhang
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qing-Ping Yao
- Institute of Mechanobiology & Medical Engineering, School of Life Science & Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| | - Rui Jiang
- Department of Urology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
15
|
Zhang L, Xia C, Yang Y, Sun F, Zhang Y, Wang H, Liu R, Yuan M. DNA methylation and histone post-translational modifications in atherosclerosis and a novel perspective for epigenetic therapy. Cell Commun Signal 2023; 21:344. [PMID: 38031118 PMCID: PMC10688481 DOI: 10.1186/s12964-023-01298-8] [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: 08/08/2022] [Accepted: 08/27/2023] [Indexed: 12/01/2023] Open
Abstract
Atherosclerosis, which is a vascular pathology characterized by inflammation and plaque build-up within arterial vessel walls, acts as the important cause of most cardiovascular diseases. Except for a lipid-depository and chronic inflammatory, increasing evidences propose that epigenetic modifications are increasingly associated with atherosclerosis and are of interest from both therapeutic and biomarker perspectives. The chronic progressive nature of atherosclerosis has highlighted atherosclerosis heterogeneity and the fact that specific cell types in the complex milieu of the plaque are, by far, not the only initiators and drivers of atherosclerosis. Instead, the ubiquitous effects of cell type are tightly controlled and directed by the epigenetic signature, which, in turn, is affected by many proatherogenic stimuli, including low-density lipoprotein, proinflammatory, and physical forces of blood circulation. In this review, we summarize the role of DNA methylation and histone post-translational modifications in atherosclerosis. The future research directions and potential therapy for the management of atherosclerosis are also discussed. Video Abstract.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Chenhai Xia
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Yongjun Yang
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Fangfang Sun
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Yu Zhang
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Huan Wang
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Rui Liu
- Department of Rehabilitation, Tangdu Hospital, Air Force Military Medical University, No. 1 Xinsi Road, Xi'an 710000, China.
| | - Ming Yuan
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
16
|
Aravindraja C, Jeepipalli S, Duncan W, Vekariya KM, Bahadekar S, Chan EKL, Kesavalu L. Unique miRomics Expression Profiles in Tannerella forsythia-Infected Mandibles during Periodontitis Using Machine Learning. Int J Mol Sci 2023; 24:16393. [PMID: 38003583 PMCID: PMC10671577 DOI: 10.3390/ijms242216393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/01/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
T. forsythia is a subgingival periodontal bacterium constituting the subgingival pathogenic polymicrobial milieu during periodontitis (PD). miRNAs play a pivotal role in maintaining periodontal tissue homeostasis at the transcriptional, post-transcriptional, and epigenetic levels. The aim of this study was to characterize the global microRNAs (miRNA, miR) expression kinetics in 8- and 16-week-old T. forsythia-infected C57BL/6J mouse mandibles and to identify the miRNA bacterial biomarkers of disease process at specific time points. We examined the differential expression (DE) of miRNAs in mouse mandibles (n = 10) using high-throughput NanoString nCounter® miRNA expression panels, which provided significant advantages over specific candidate miRNA or pathway analyses. All the T. forsythia-infected mice at two specific time points showed bacterial colonization (100%) in the gingival surface, along with a significant increase in alveolar bone resorption (ABR) (p < 0.0001). We performed a NanoString analysis of specific miRNA signatures, miRNA target pathways, and gene network analysis. A total of 115 miRNAs were DE in the mandible tissue during 8 and 16 weeks The T. forsythia infection, compared with sham infection, and the majority (99) of DE miRNAs were downregulated. nCounter miRNA expression kinetics identified 67 downregulated miRNAs (e.g., miR-375, miR-200c, miR-200b, miR-34b-5p, miR-141) during an 8-week infection, whereas 16 upregulated miRNAs (e.g., miR-1902, miR-let-7c, miR-146a) and 32 downregulated miRNAs (e.g., miR-2135, miR-720, miR-376c) were identified during a 16-week infection. Two miRNAs, miR-375 and miR-200c, were highly downregulated with >twofold change during an 8-week infection. Six miRNAs in the 8-week infection (miR-200b, miR-141, miR-205, miR-423-3p, miR-141-3p, miR-34a-5p) and two miRNAs in the 16-week infection (miR-27a-3p, miR-15a-5p) that were downregulated have also been reported in the gingival tissue and saliva of periodontitis patients. This preclinical in vivo study identified T. forsythia-specific miRNAs (miR-let-7c, miR-210, miR-146a, miR-423-5p, miR-24, miR-218, miR-26b, miR-23a-3p) and these miRs have also been reported in the gingival tissues and saliva of periodontitis patients. Further, several DE miRNAs that are significantly upregulated (e.g., miR-101b, miR-218, miR-127, miR-24) are also associated with many systemic diseases such as atherosclerosis, Alzheimer's disease, rheumatoid arthritis, osteoarthritis, diabetes, obesity, and several cancers. In addition to DE analysis, we utilized the XGBoost (eXtreme Gradient boost) and Random Forest machine learning (ML) algorithms to assess the impact that the number of miRNA copies has on predicting whether a mouse is infected. XGBoost found that miR-339-5p was most predictive for mice infection at 16 weeks. miR-592-5p was most predictive for mice infection at 8 weeks and also when the 8-week and 16-week results were grouped together. Random Forest predicted miR-592 as most predictive at 8 weeks as well as the combined 8-week and 16-week results, but miR-423-5p was most predictive at 16 weeks. In conclusion, the expression levels of miR-375 and miR-200c family differed significantly during disease process, and these miRNAs establishes a link between T. forsythia and development of periodontitis genesis, offering new insights regarding the pathobiology of this bacterium.
Collapse
Affiliation(s)
- Chairmandurai Aravindraja
- Department of Periodontology, College of Dentistry, University of Florida, Gainesville, FL 32610, USA; (C.A.); (S.J.); (K.M.V.)
| | - Syam Jeepipalli
- Department of Periodontology, College of Dentistry, University of Florida, Gainesville, FL 32610, USA; (C.A.); (S.J.); (K.M.V.)
| | - William Duncan
- Department of Community Dentistry, College of Dentistry, University of Florida, Gainesville, FL 32610, USA;
| | - Krishna Mukesh Vekariya
- Department of Periodontology, College of Dentistry, University of Florida, Gainesville, FL 32610, USA; (C.A.); (S.J.); (K.M.V.)
| | - Sakshee Bahadekar
- Department of Computer and Information Science and Engineering, University of Florida, Gainesville, FL 32610, USA;
| | - Edward K. L. Chan
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL 32610, USA;
| | - Lakshmyya Kesavalu
- Department of Periodontology, College of Dentistry, University of Florida, Gainesville, FL 32610, USA; (C.A.); (S.J.); (K.M.V.)
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL 32610, USA;
| |
Collapse
|
17
|
Malhi NK, Southerland KW, Lai L, Chen ZB. Epigenetic Regulation of Angiogenesis in Peripheral Artery Disease. Methodist Debakey Cardiovasc J 2023; 19:47-57. [PMID: 38028966 PMCID: PMC10655766 DOI: 10.14797/mdcvj.1294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 12/01/2023] Open
Abstract
Peripheral arterial disease (PAD) represents a global health concern with a rising prevalence attributed to factors such as obesity, diabetes, aging, and smoking. Among patients with PAD, chronic limb-threatening ischemia (CLTI) is the most severe manifestation, associated with substantial morbidity and mortality. While revascularization remains the primary therapy for CLTI, not all patients are candidates for such interventions, highlighting the need for alternative approaches. Impaired angiogenesis, the growth of new blood vessels, is a central feature of PAD, and despite decades of research, effective clinical treatments remain elusive. Epigenetics, the study of heritable changes in gene expression, has gained prominence in understanding PAD pathogenesis. Here, we explore the role of epigenetic regulation in angiogenesis within the context of PAD, with a focus on long non-coding RNAs and fibroblast-endothelial cell transdifferentiation. Additionally, we discuss the interplay between metabolic control and epigenetic regulation, providing insights into potential novel therapeutic avenues for improving PAD treatments. This review aims to offer a concise update on the application of epigenetics in angiogenesis and PAD research, inspiring further investigations in this promising field.
Collapse
Affiliation(s)
| | | | - Li Lai
- Houston Methodist Research Institute, Houston, Texas, US
| | | |
Collapse
|
18
|
Downes N, Niskanen H, Tomas Bosch V, Taipale M, Godiwala M, Väänänen MA, Turunen TA, Aavik E, Laham-Karam N, Ylä-Herttuala S, Kaikkonen MU. Hypoxic regulation of hypoxia inducible factor 1 alpha via antisense transcription. J Biol Chem 2023; 299:105291. [PMID: 37748649 PMCID: PMC10630634 DOI: 10.1016/j.jbc.2023.105291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 09/11/2023] [Accepted: 09/20/2023] [Indexed: 09/27/2023] Open
Abstract
Impaired oxygen homeostasis is a frequently encountered pathophysiological factor in multiple complex diseases, including cardiovascular disease and cancer. While the canonical hypoxia response pathway is well characterized, less is known about the role of noncoding RNAs in this process. Here, we investigated the nascent and steady-state noncoding transcriptional responses in endothelial cells and their potential roles in regulating the hypoxic response. Notably, we identify a novel antisense long noncoding RNA that convergently overlaps the majority of the hypoxia inducible factor 1 alpha (HIF1A) locus, which is expressed across several cell types and elevated in atherosclerotic lesions. The antisense (HIF1A-AS) is produced as a stable, unspliced, and polyadenylated nuclear retained transcript. HIF1A-AS is highly induced in hypoxia by both HIF1A and HIF2A and exhibits anticorrelation with the coding HIF1A transcript and protein expression. We further characterized this functional relationship by CRISPR-mediated bimodal perturbation of the HIF1A-AS promoter. We provide evidence that HIF1A-AS represses the expression of HIF1a in cis by repressing transcriptional elongation and deposition of H3K4me3, and that this mechanism is dependent on the act of antisense transcription itself. Overall, our results indicate a critical regulatory role of antisense mediated transcription in regulation of HIF1A expression and cellular response to hypoxia.
Collapse
Affiliation(s)
- Nicholas Downes
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, North-Savo, Finland
| | - Henri Niskanen
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, North-Savo, Finland
| | - Vanesa Tomas Bosch
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, North-Savo, Finland
| | - Mari Taipale
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, North-Savo, Finland
| | - Mehvash Godiwala
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, North-Savo, Finland
| | - Mari-Anna Väänänen
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, North-Savo, Finland
| | - Tiia A Turunen
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, North-Savo, Finland
| | - Einari Aavik
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, North-Savo, Finland
| | - Nihay Laham-Karam
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, North-Savo, Finland
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, North-Savo, Finland; School of Medicine, University of Eastern Finland, Kuopio, North-Savo, Finland; Heart Center, Kuopio University Hospital, Kuopio, Finland.
| | - Minna U Kaikkonen
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, North-Savo, Finland.
| |
Collapse
|
19
|
Omar M, Alexiou M, Rekhi UR, Lehmann K, Bhardwaj A, Delyea C, Elahi S, Febbraio M. DNA methylation changes underlie the long-term association between periodontitis and atherosclerotic cardiovascular disease. Front Cardiovasc Med 2023; 10:1164499. [PMID: 37153468 PMCID: PMC10160482 DOI: 10.3389/fcvm.2023.1164499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 03/22/2023] [Indexed: 05/09/2023] Open
Abstract
Periodontitis, the leading cause of adult tooth loss, has been identified as an independent risk factor for cardiovascular disease (CVD). Studies suggest that periodontitis, like other CVD risk factors, shows the persistence of increased CVD risk even after mitigation. We hypothesized that periodontitis induces epigenetic changes in hematopoietic stem cells in the bone marrow (BM), and such changes persist after the clinical elimination of the disease and underlie the increased CVD risk. We used a BM transplant approach to simulate the clinical elimination of periodontitis and the persistence of the hypothesized epigenetic reprogramming. Using the low-density lipoprotein receptor knockout (LDLRo ) atherosclerosis mouse model, BM donor mice were fed a high-fat diet to induce atherosclerosis and orally inoculated with Porphyromonas gingivalis (Pg), a keystone periodontal pathogen; the second group was sham-inoculated. Naïve LDLR o mice were irradiated and transplanted with BM from one of the two donor groups. Recipients of BM from Pg-inoculated donors developed significantly more atherosclerosis, accompanied by cytokine/chemokines that suggested BM progenitor cell mobilization and were associated with atherosclerosis and/or PD. Using whole-genome bisulfite sequencing, 375 differentially methylated regions (DMRs) and global hypomethylation in recipients of BM from Pg-inoculated donors were observed. Some DMRs pointed to the involvement of enzymes with major roles in DNA methylation and demethylation. In validation assays, we found a significant increase in the activity of ten-eleven translocase-2 and a decrease in the activity of DNA methyltransferases. Plasma S-adenosylhomocysteine levels were significantly higher, and the S-adenosylmethionine to S-adenosylhomocysteine ratio was decreased, both of which have been associated with CVD. These changes may be related to increased oxidative stress as a result of Pg infection. These data suggest a novel and paradigm-shifting mechanism in the long-term association between periodontitis and atherosclerotic CVD.
Collapse
|
20
|
Jia Z, Mei J, Zhang Y, Wang Y, Wang H, Wang A, Xu F, Zhou Q. Whole genome methylation combined with RNA-seq reveals the protective effects of Gualou-Xiebai herb pair in foam cells through DNA methylation mediated PI3K-AKT signaling pathway. Front Immunol 2023; 14:1054014. [PMID: 36911738 PMCID: PMC9992180 DOI: 10.3389/fimmu.2023.1054014] [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: 09/26/2022] [Accepted: 02/10/2023] [Indexed: 02/25/2023] Open
Abstract
DNA methylation, including aberrant hypomethylation and hypermethylation, plays a significant role in atherosclerosis (AS); therefore, targeting the unbalanced methylation in AS is a potential treatment strategy. Gualou-xiebai herb pair (GXHP), a classic herb combination, have been used for the treatment of atherosclerotic-associated diseases in traditional Chinese medicine. However, the effects and underlying mechanism of GXHP on AS remain nebulous. In this study, the CCK-8 method was applied to determine the non-toxic treatment concentrations for GXHP. The formation of foam cells played a critical role in AS, so the foam cells model was established after RAW264.7 cells were treated with ox-LDL. The contents of total cholesterol (TC) and free cholesterol (FC) were determined by Gas chromatography-mass spectrometry (GC-MS). Enzyme-linked immunosorbent assay (ELISA) was used to check the expressions of inflammatory factors including IL-1β, TNF-α, and VCAM-1. Methyl-capture sequencing (MC-seq) and RNA-seq were applied to observe the changes in genome-wide DNA methylation and gene expression, respectively. Kyoto Encyclopedia of Genes and Genomes (KEGG) were performed to analyze differentially methylated genes (DMGs) and differentially expressed genes (DEGs). The targeted signaling pathway was selected and verified using western blotting (WB). The results showed that the lipids and inflammatory factors in foam cells significantly increased. GXHP significantly reduced the expression of TC, FC, and inflammatory factors. MC-seq and RNA-seq showed that GXHP not only corrected the aberrant DNA hypermethylation, but also DNA hypomethylation, thus restored the aberrant DEGs in foam cells induced by ox-LDL. GXHP treatment may target the PI3K-Akt signaling pathway. GXHP reduced the protein levels of phosphorylated(p)-PI3K and p-AKT in foam cells. Our data suggest that treatment with GXHP showed protective effects against AS through the inhibition of DNA methylation mediated PI3K-AKT signaling pathway, suggesting GXHP as a novel methylation-based agent.
Collapse
Affiliation(s)
- Zijun Jia
- Xiyuan Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China.,Institute of Geriatric Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jun Mei
- Institute of Geriatric Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yan Zhang
- Institute of Geriatric Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ya Wang
- Institute of Geriatric Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongqin Wang
- Institute of Geriatric Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Anlu Wang
- Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengqin Xu
- Institute of Geriatric Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingbing Zhou
- Institute of Geriatric Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
21
|
Gkaliagkousi E, Lazaridis A, Dogan S, Fraenkel E, Tuna BG, Mozos I, Vukicevic M, Yalcin O, Gopcevic K. Theories and Molecular Basis of Vascular Aging: A Review of the Literature from VascAgeNet Group on Pathophysiological Mechanisms of Vascular Aging. Int J Mol Sci 2022; 23:ijms23158672. [PMID: 35955804 PMCID: PMC9368987 DOI: 10.3390/ijms23158672] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
Vascular aging, characterized by structural and functional alterations of the vascular wall, is a hallmark of aging and is tightly related to the development of cardiovascular mortality and age-associated vascular pathologies. Over the last years, extensive and ongoing research has highlighted several sophisticated molecular mechanisms that are involved in the pathophysiology of vascular aging. A more thorough understanding of these mechanisms could help to provide a new insight into the complex biology of this non-reversible vascular process and direct future interventions to improve longevity. In this review, we discuss the role of the most important molecular pathways involved in vascular ageing including oxidative stress, vascular inflammation, extracellular matrix metalloproteinases activity, epigenetic regulation, telomere shortening, senescence and autophagy.
Collapse
Affiliation(s)
- Eugenia Gkaliagkousi
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Faculty of Medicine, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece
- Correspondence: (E.G.); (K.G.)
| | - Antonios Lazaridis
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Faculty of Medicine, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece
| | - Soner Dogan
- Department of Medical Biology, School of Medicine, Yeditepe University, 34755 Istanbul, Turkey
| | - Emil Fraenkel
- 1st Department of Internal Medicine, University Hospital, Pavol Jozef Šafárik University of Košice, Trieda SNP 1, 04066 Košice, Slovakia
| | - Bilge Guvenc Tuna
- Department of Biophysics, School of Medicine, Yeditepe University, 34755 Istanbul, Turkey
| | - Ioana Mozos
- Department of Functional Sciences-Pathophysiology, Center for Translational Research and Systems Medicine, “Victor Babes” University of Medicine and Pharmacy, 300173 Timisoara, Romania
| | - Milica Vukicevic
- Cardiac Surgery Clinic, Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Ozlem Yalcin
- Department of Physiology, School of Medicine, Koc University, 34450 Istanbul, Turkey
| | - Kristina Gopcevic
- Laboratory for Analytics of Biomolecules, Department of Chemistry in Medicine, Faculty of Medicine, 11000 Belgrade, Serbia
- Correspondence: (E.G.); (K.G.)
| |
Collapse
|
22
|
Chen Y, Liang L, Wu C, Cao Z, Xia L, Meng J, Wang Z. Epigenetic Control of Vascular Smooth Muscle Cell Function in Atherosclerosis: A Role for DNA Methylation. DNA Cell Biol 2022; 41:824-837. [PMID: 35900288 DOI: 10.1089/dna.2022.0278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Atherosclerosis is a complex vascular inflammatory disease in which multiple cell types are involved, including vascular smooth muscle cells (VSMCs). In response to vascular injury and inflammatory stimuli, VSMCs undergo a "phenotypic switching" characterized by extracellular matrix secretion, loss of contractility, and abnormal proliferation and migration, which play a key role in the progression of atherosclerosis. DNA methylation modification is an important epigenetic mechanism that plays an important role in atherosclerosis. Studies investigating abnormal DNA methylation in patients with atherosclerosis have determined a specific DNA methylation profile, and proposed multiple pathways and genes involved in the etiopathogenesis of atherosclerosis. Recent studies have also revealed that DNA methylation modification controls VSMC function by regulating gene expression involved in atherosclerosis. In this review, we summarize the recent advances regarding the epigenetic control of VSMC function by DNA methylation in atherosclerosis and provide insights into the development of VSMC-centered therapeutic strategies.
Collapse
Affiliation(s)
- Yanjun Chen
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| | - Lingli Liang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| | - Chunyan Wu
- The Third Affiliated Hospital of University of South China, Hengyang, China
| | - Zitong Cao
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| | - Linzhen Xia
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| | - Jun Meng
- Functional Department, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Zuo Wang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
23
|
Hilser JR, Hartiala JA, Sriprasert I, Kono N, Cai Z, Karim R, DeYoung J, Mack WJ, Hodis HN, Allayee H. Effect of menopausal hormone therapy on methylation levels in early and late postmenopausal women. Clin Epigenetics 2022; 14:90. [PMID: 35850911 PMCID: PMC9295504 DOI: 10.1186/s13148-022-01311-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 06/28/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) remains the leading cause of death among postmenopausal women but standard primary prevention strategies in women are not as effective as in men. By comparison, the Early versus Late Intervention Trial with Estradiol (ELITE) study demonstrated that hormone therapy (HT) was associated with significant reduction in atherosclerosis progression in women who were within six years of menopause compared to those who were 10 or more years from menopause. These findings are consistent with other studies showing significant reductions in all-cause mortality and CVD with HT, particularly when initiated in women younger than 60 years of age or within 10 years since menopause. To explore the biological mechanisms underlying the age-related atheroprotective effects of HT, we investigated changes in methylation of blood cells of postmenopausal women who participated in ELITE. RESULTS We first validated the epigenetic data generated from blood leukocytes of ELITE participants by replicating previously known associations between smoking and methylation levels at previously identified CpG sites, such as cg05575921 at the AHRR locus. An epigenome-wide association study (EWAS) evaluating changes in methylation through interactions with time-since-menopause and HT revealed two significantly associated CpG sites on chromosomes 12 (cg19552895; p = 1.1 × 10-9) and 19 (cg18515510; p = 2.4 × 10-8). Specifically, HT resulted in modest, but significant, increases in methylation levels at both CpGs but only in women who were 10 or more years since menopause and randomized to HT. Changes in carotid artery intima-media thickness (CIMT) from baseline to 36 months after HT were not significantly correlated with changes in methylation levels at either cg19552895 or cg18515510. Evaluation of other previously identified CpG sites at which methylation levels in either blood or vascular tissue were associated with atherosclerosis also did not reveal any differences in methylation as a function of HT and time-since-menopause or with changes in CIMT. CONCLUSIONS We identified specific methylation differences in blood in response to HT among women who were 10 or more years since menopause. The functional consequence of these change with respect to atherosclerosis progression and protective effects of HT remains to be determined and will require additional studies.
Collapse
Affiliation(s)
- James R. Hilser
- grid.42505.360000 0001 2156 6853Departments of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC202, Los Angeles, CA 90033 USA ,grid.42505.360000 0001 2156 6853Departments of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| | - Jaana A. Hartiala
- grid.42505.360000 0001 2156 6853Departments of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC202, Los Angeles, CA 90033 USA
| | - Intira Sriprasert
- grid.42505.360000 0001 2156 6853Departments of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| | - Naoko Kono
- grid.42505.360000 0001 2156 6853Departments of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC202, Los Angeles, CA 90033 USA ,grid.42505.360000 0001 2156 6853Atherosclerosis Research Unit, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| | - Zhiheng Cai
- grid.42505.360000 0001 2156 6853Departments of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC202, Los Angeles, CA 90033 USA ,grid.42505.360000 0001 2156 6853Departments of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| | - Roksana Karim
- grid.42505.360000 0001 2156 6853Departments of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC202, Los Angeles, CA 90033 USA ,grid.42505.360000 0001 2156 6853Atherosclerosis Research Unit, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| | - Joseph DeYoung
- grid.19006.3e0000 0000 9632 6718Department of Psychiatry and Biobehavioral Sciences, Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095 USA
| | - Wendy J. Mack
- grid.42505.360000 0001 2156 6853Departments of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC202, Los Angeles, CA 90033 USA ,grid.42505.360000 0001 2156 6853Atherosclerosis Research Unit, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| | - Howard N. Hodis
- grid.42505.360000 0001 2156 6853Departments of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC202, Los Angeles, CA 90033 USA ,grid.42505.360000 0001 2156 6853Departments of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA ,grid.42505.360000 0001 2156 6853Atherosclerosis Research Unit, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| | - Hooman Allayee
- grid.42505.360000 0001 2156 6853Departments of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC202, Los Angeles, CA 90033 USA ,grid.42505.360000 0001 2156 6853Departments of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| |
Collapse
|
24
|
Nazarenko MS, Koroleva IA, Zarubin AA, Sleptcov AA. miRNA Regulome in Different Atherosclerosis Phenotypes. Mol Biol 2022. [DOI: 10.1134/s0026893322020108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
25
|
GENG H, CHEN L, SU Y, XU Q, FAN M, HUANG R, LI X, LU X, PAN M. miR-431-5p Regulates Apoptosis of Cardiomyocytes After Acute Myocardial Infarction via Targeting Selenoprotein T. Physiol Res 2022; 71:55-62. [PMID: 35043644 DOI: 10.33549/physiolres.934683] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Acute myocardial infarction (AMI) represents the acute manifestation of coronary artery disease. In recent years, microRNAs (miRNAs) have been extensively studied in AMI. This study focused on the role of miR-431-5p in AMI and its effect on cardiomyocyte apoptosis after AMI. The expression of miR-431-5p was analyzed by quantitative real-time PCR (qRT-PCR). By interfering with miR-431-5p in hypoxia-reoxygenation (H/R)-induced HL-1 cardiomyocytes, the effect of miR-431-5p on cardiomyocyte apoptosis after AMI was examined. The interaction between miR-431-5p and selenoprotein T (SELT) mRNA was verified by dual-luciferase reporter assay. Cell apoptosis was determined by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay and flow cytometry. Cell viability was examined by 3-(4,5)-dimethylthiahiazo(-z-y1)-3,5-di-phenytetrazoliumromide (MTT) assay. The results of qRT-PCR showed that the expression of miR-431-5p in AMI myocardial tissues and H/R-induced HL-1 cardiomyocytes was significantly increased. After interfering with miR-431-5p, the expression of SELT in HL-1 cells was up-regulated, cell apoptosis was decreased, cell viability was increased, and lactate dehydrogenase (LDH) activity was decreased. The dual-luciferase reporter assay confirmed the targeting relationship between miR-431-5p and SELT1 3’ untranslated region (UTR). In H/R-induced HL-1 cells, the simultaneous silencing of SELT and miR-431-5p resulted in a decrease of Bcl-2 expression, an increase of Bax expression, and an increase of cleaved-caspase 3 expression compared with silencing miR-431-5p alone. Also, cell viability was decreased, while LDH activity was increased by the simultaneous silencing of SELT and miR-431-5p. Interfering miR-431-5p protected cardiomyocytes from AMI injury via restoring the expression of SELT, providing new ideas for the treatment of AMI.
Collapse
Affiliation(s)
- H GENG
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - L CHEN
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Y SU
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Q XU
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - M FAN
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - R HUANG
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - X LI
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - X LU
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - M PAN
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
26
|
Dai Y, Chen D, Xu T. DNA Methylation Aberrant in Atherosclerosis. Front Pharmacol 2022; 13:815977. [PMID: 35308237 PMCID: PMC8927809 DOI: 10.3389/fphar.2022.815977] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/24/2022] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis (AS) is a pathological process involving lipid oxidation, immune system activation, and endothelial dysfunction. The activated immune system could lead to inflammation and oxidative stress. Risk factors like aging and hyperhomocysteinemia also promote the progression of AS. Epigenetic modifications, including DNA methylation, histone modification, and non-coding RNA, are involved in the modulation of genes between the environment and AS formation. DNA methylation is one of the most important epigenetic mechanisms in the pathogenesis of AS. However, the relationship between the progression of AS and DNA methylation is not completely understood. This review will discuss the abnormal changes of DNA methylation in AS, including genome-wide hypermethylation dominating in AS with an increase of age, hypermethylation links with methyl supply and generating hyperhomocysteinemia, and the influence of oxidative stress with the demethylation process by interfering with the hydroxyl-methylation of TET proteins. The review will also summarize the current status of epigenetic treatment, which may provide new direction and potential therapeutic targets for AS.
Collapse
|
27
|
Sallam M, Benotmane MA, Baatout S, Guns PJ, Aerts A. Radiation-induced cardiovascular disease: an overlooked role for DNA methylation? Epigenetics 2022; 17:59-80. [PMID: 33522387 PMCID: PMC8812767 DOI: 10.1080/15592294.2021.1873628] [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: 07/03/2020] [Revised: 11/27/2020] [Accepted: 01/04/2021] [Indexed: 11/25/2022] Open
Abstract
Radiotherapy in cancer treatment involves the use of ionizing radiation for cancer cell killing. Although radiotherapy has shown significant improvements on cancer recurrence and mortality, several radiation-induced adverse effects have been documented. Of these adverse effects, radiation-induced cardiovascular disease (CVD) is particularly prominent among patients receiving mediastinal radiotherapy, such as breast cancer and Hodgkin's lymphoma patients. A number of mechanisms of radiation-induced CVD pathogenesis have been proposed such as endothelial inflammatory activation, premature endothelial senescence, increased ROS and mitochondrial dysfunction. However, current research seems to point to a so-far unexamined and potentially novel involvement of epigenetics in radiation-induced CVD pathogenesis. Firstly, epigenetic mechanisms have been implicated in CVD pathophysiology. In addition, several studies have shown that ionizing radiation can cause epigenetic modifications, especially DNA methylation alterations. As a result, this review aims to provide a summary of the current literature linking DNA methylation to radiation-induced CVD and thereby explore DNA methylation as a possible contributor to radiation-induced CVD pathogenesis.
Collapse
Affiliation(s)
- Magy Sallam
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- Laboratory of Physiopharmacology, University of Antwerp, Wilrijk, Belgium
| | - Mohammed Abderrafi Benotmane
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, University of Antwerp, Wilrijk, Belgium
| | - An Aerts
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| |
Collapse
|
28
|
Miroshnikova VV, Panteleeva AA, Pobozheva IA, Razgildina ND, Polyakova EA, Markov AV, Belyaeva OD, Berkovich OA, Baranova EI, Nazarenko MS, Puzyrev VP, Pchelina SN. ABCA1 and ABCG1 DNA methylation in epicardial adipose tissue of patients with coronary artery disease. BMC Cardiovasc Disord 2021; 21:566. [PMID: 34837967 PMCID: PMC8627066 DOI: 10.1186/s12872-021-02379-7] [Citation(s) in RCA: 10] [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: 07/27/2020] [Accepted: 11/10/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Recent studies have focused on the potential role of epicardial adipose tissue (EAT) in the development of coronary artery disease (CAD). ABCA1 and ABCG1 transporters regulate cell cholesterol content and reverse cholesterol transport. We aimed to determine whether DNA methylation and mRNA levels of the ABCA1 and ABCG1 genes in EAT and subcutaneous adipose tissue (SAT) were associated with CAD. METHODS Paired EAT and SAT samples were collected from 82 patients undergoing elective cardiac surgery either for coronary artery bypass grafting (CAD group, N = 66) or valve surgery (NCAD group, N = 16). ABCA1 and ABCG1 mRNA levels in EAT and SAT samples were analyzed using real time polymerase chain reaction, ABCA1 protein levels in EAT samples were assessed by western blotting. ABCA1 and ABCG1 DNA methylation analysis was performed in 24 samples from the CAD group and 9 samples from the NCAD group via pyrosequencing. RESULTS DNA methylation levels in the ABCA1 promoter and ABCG1 cg27243685 and cg06500161 CpG sites were higher in EAT samples from patients with CAD compared with NCAD (21.92% vs 10.81%, p = 0.003; 71.51% vs 68.42%, p = 0.024; 46.11% vs 37.79%, p = 0.016, respectively). In patients with CAD, ABCA1 and ABCG1 DNA methylation levels were higher in EAT than in SAT samples (p < 0.05). ABCA1 mRNA levels in EAT samples were reduced in the subgroup of patients with CAD and concomitant carotid artery disease or peripheral artery disease compared with the NCAD group (p = 0.024). ABCA1 protein levels in EAT samples tended to be lower in CAD patients than in the NCAD group (p = 0.053). DNA methylation levels at the ABCG1 cg27243685 site positively correlated with plasma triglyceride concentration (r = 0.510, p = 0.008), body mass index (r = 0.556, p = 0.013) and waist-to-hip ratio (r = 0.504, p = 0.012) in SAT samples. CONCLUSION CAD is associated with ABCA1 and ABCG1 DNA hypermethylation in EAT. CAD with concomitant carotid artery disease or peripheral artery disease is accompanied by decreased ABCA1 gene expression in EAT. DNA methylation levels at the ABCG1 cg27243685 locus in SAT are associated with hypertriglyceridemia and obesity.
Collapse
Affiliation(s)
- Valentina V Miroshnikova
- Petersburg Nuclear Physics Institute Named By B.P. Konstantinov of National Research Center "Kurchatov Institute", Gatchina, Russian Federation.
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation.
| | - Alexandra A Panteleeva
- Petersburg Nuclear Physics Institute Named By B.P. Konstantinov of National Research Center "Kurchatov Institute", Gatchina, Russian Federation
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
- National Research Centre "Kurchatov Institute", Moscow, Russia
| | - Irina A Pobozheva
- Petersburg Nuclear Physics Institute Named By B.P. Konstantinov of National Research Center "Kurchatov Institute", Gatchina, Russian Federation
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
- National Research Centre "Kurchatov Institute", Moscow, Russia
| | - Natalia D Razgildina
- Petersburg Nuclear Physics Institute Named By B.P. Konstantinov of National Research Center "Kurchatov Institute", Gatchina, Russian Federation
| | - Ekaterina A Polyakova
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
| | - Anton V Markov
- Laboratory of Population Genetics, Research Institute of Medical Genetics, Tomsk, Russian Federation
| | - Olga D Belyaeva
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
| | - Olga A Berkovich
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
| | - Elena I Baranova
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
| | - Maria S Nazarenko
- Laboratory of Population Genetics, Research Institute of Medical Genetics, Tomsk, Russian Federation
| | - Valery P Puzyrev
- Laboratory of Population Genetics, Research Institute of Medical Genetics, Tomsk, Russian Federation
| | - Sofya N Pchelina
- Petersburg Nuclear Physics Institute Named By B.P. Konstantinov of National Research Center "Kurchatov Institute", Gatchina, Russian Federation
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
- National Research Centre "Kurchatov Institute", Moscow, Russia
| |
Collapse
|
29
|
Su Y, Guan P, Li D, Hang Y, Ye X, Han L, Lu Y, Bai X, Zhang P, Hu W. Intermedin attenuates macrophage phagocytosis via regulation of the long noncoding RNA Dnm3os/miR-27b-3p/SLAMF7 axis in a mouse model of atherosclerosis in diabetes. Biochem Biophys Res Commun 2021; 583:35-42. [PMID: 34717123 DOI: 10.1016/j.bbrc.2021.10.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/08/2021] [Accepted: 10/16/2021] [Indexed: 12/25/2022]
Abstract
Atherosclerosis in diabetes is a leading cause of cardiovascular complications. Intermedin (IMD) is a calcitonin peptide that is known to inhibit macrophage phagocytosis in atherosclerosis, but the exact mechanism is unclear. We investigate genes that are differentially expressed in response to IMD in hyperglycemic conditions and determine whether they delay the progression of atherosclerosis. An atherosclerotic and diabetic-murine model was generated in 8-week-old male ApoE-/- mice receiving streptozotocin and a high-fat diet. The mouse model was treated with IMD and the expression levels of NF-κB, Dnm3os, miR-27b-3p, and SLAMF7 were detected in plaque tissue and macrophages cultured with high glucose concentrations. Phagocytosis was determined by oxidized-low-density lipoprotein (Ox-LDL) uptake and the interactions among Dnm3os, SLAMF7 and miR-27b-3p were assessed by dual-luciferase reporter assays. The expression of NF-κB, Dnm3os, and SLAMF7 was enhanced in atherosclerotic plaques but decreased by IMD. The suppression of Dnm3os reduced plaque formation in IMD-treated mice even further whereas increased by miR-27b-3p. Dnm3os and SLAMF7 were competitively bind to miR-27b-3p in vivo. In vitro, ox-LDL uptake is elevated in macrophages cultured in hyperglycemic conditions but reduced by IMD. Dual-luciferase assays indicate that Dnm3os positively regulates SLAMF7 through miR-27b-3p expression. In conclusion, Dnm3os is involved in macrophage phagocytosis through the competitive binding of SLAMF7 with miR-27b-3p. IMD induces the suppression of Dnm3os to inhibit macrophage phagocytosis and alleviate atherosclerosis in diabetes.
Collapse
Affiliation(s)
- Yanling Su
- Department of Cardiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Ping Guan
- Department of Cardiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Dandan Li
- Department of Cardiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Yanwen Hang
- Department of Cardiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Xiaomiao Ye
- Department of Cardiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Lu Han
- Department of Cardiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Yi Lu
- Department of Cardiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Xiaolu Bai
- Department of Cardiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Peng Zhang
- Department of Cardiology, Minhang Hospital, Fudan University, Shanghai, China.
| | - Wei Hu
- Department of Cardiology, Minhang Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
30
|
Li J, Zhang X, Yang M, Yang H, Xu N, Fan X, Liu G, Jiang X, Fan J, Zhang L, Zhang H, Zhou Y, Li R, Gao S, Jin J, Jin Z, Zheng J, Tu Q, Ren J. DNA methylome profiling reveals epigenetic regulation of lipoprotein-associated phospholipase A 2 in human vulnerable atherosclerotic plaque. Clin Epigenetics 2021; 13:161. [PMID: 34419168 PMCID: PMC8379831 DOI: 10.1186/s13148-021-01152-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 08/12/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Atherosclerotic plaque vulnerability is a key feature of atheroprogression and precipitating acute cardiovascular events. Although the pivotal role of epigenetic regulation in atherosclerotic plaque destabilization is being recognized, the DNA methylation profile and its potential role in driving the progression and destabilization of atherosclerotic cardiovascular disease remains largely unknown. We conducted a genome-wide analysis to identify differentially methylated genes in vulnerable and non-vulnerable atherosclerotic lesions to understand more about pathogenesis. RESULTS We compared genome-wide DNA methylation profiling between carotid artery plaques of patients with clinically symptomatic (recent stroke or transient ischemic attack) and asymptomatic disease (no recent stroke) using Infinium Methylation BeadChip arrays, which revealed 90,368 differentially methylated sites (FDR < 0.05, |delta beta|> 0.03) corresponding to 14,657 annotated genes. Among these genomic sites, 30% were located at the promoter regions and 14% in the CpG islands, according to genomic loci and genomic proximity to the CpG islands, respectively. Moreover, 67% displayed hypomethylation in symptomatic plaques, and the differentially hypomethylated genes were found to be involved in various aspects of inflammation. Subsequently, we focus on CpG islands and revealed 14,596 differentially methylated sites (|delta beta|> 0.1) located at the promoter regions of 7048 genes. Integrated analysis of methylation and gene expression profiles identified that 107 genes were hypomethylated in symptomatic plaques and showed elevated expression levels in both advanced plaques and ruptured plaques. The imprinted gene PLA2G7, which encodes lipoprotein-associated phospholipase A2 (Lp-PLA2), was one of the top hypomethylated genes with an increased expression upon inflammation. Further, the hypomethylated CpG site at the promoter region of PLA2G7 was identified as cg11874627, demethylation of which led to increased binding of Sp3 and expression of Lp-PLA2 through bisulfate sequencing, chromatin immunoprecipitation assay and enzyme-linked immunosorbent assay. These effects were further enhanced by deacetylase. CONCLUSION Extensive DNA methylation modifications serve as a new and critical layer of biological regulation that contributes to atheroprogression and destabilization via inflammatory processes. Revelation of this hitherto unknown epigenetic regulatory mechanism could rejuvenate the prospects of Lp-PLA2 as a therapeutic target to stabilize the atherosclerotic plaque and reduce clinical sequelae.
Collapse
Affiliation(s)
- Jingjin Li
- Department of Cardiology, Beijing Tiantan Hospital of Capital Medical University, Beijing, China
| | - Xiaoping Zhang
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Mengxi Yang
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Hang Yang
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Ning Xu
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Xueqiang Fan
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Gang Liu
- Department of Cardiovascular Surgery, Peking University People's Hospital, Beijing, China
| | - Xintong Jiang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Jiasai Fan
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Lifang Zhang
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Hu Zhang
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Ying Zhou
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Rui Li
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Si Gao
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Jiangli Jin
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Zening Jin
- Department of Cardiology, Beijing Tiantan Hospital of Capital Medical University, Beijing, China
| | - Jingang Zheng
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Qiang Tu
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jingyi Ren
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China. .,Vascular Health Research Center of Peking University Health Science Center, Beijing, China.
| |
Collapse
|
31
|
Zurek M, Aavik E, Mallick R, Ylä-Herttuala S. Epigenetic Regulation of Vascular Smooth Muscle Cell Phenotype Switching in Atherosclerotic Artery Remodeling: A Mini-Review. Front Genet 2021; 12:719456. [PMID: 34422021 PMCID: PMC8375552 DOI: 10.3389/fgene.2021.719456] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/19/2021] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by extensive remodeling of medium and large-sized arteries. Inward remodeling (=lumen shrinkage) of the vascular walls is the underlying cause for ischemia in target organs. Therefore, inward remodeling can be considered the predominant feature of atherosclerotic pathology. Outward remodeling (=lumen enlargement) is a physiological response compensating for lumen shrinkage caused by neointimal hyperplasia, but as a pathological response to changes in blood flow, outward remodeling leads to substantial arterial wall thinning. Thinned vascular walls are prone to rupture, and subsequent thrombus formation accounts for the majority of acute cardiovascular events. Pathological remodeling is driven by inflammatory cells which induce vascular smooth muscle cells to switch from quiescent to a proliferative and migratory phenotype. After decades of intensive research, the molecular mechanisms of arterial remodeling are starting to unfold. In this mini-review, we summarize the current knowledge of the epigenetic and transcriptional regulation of vascular smooth muscle cell phenotype switching from the contractile to the synthetic phenotype involved in arterial remodeling and discuss potential therapeutic options.
Collapse
Affiliation(s)
- Michelle Zurek
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | | | | |
Collapse
|
32
|
DNA Methylation in Atherosclerosis: A New Perspective. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6623657. [PMID: 34257689 PMCID: PMC8249120 DOI: 10.1155/2021/6623657] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 06/08/2021] [Indexed: 01/14/2023]
Abstract
Atherosclerotic cardiovascular diseases, in which atherosclerosis (AS) is the main pathologic basis, are currently the primary diseases leading to human deaths. Emerging evidence showed that DNA methylation, which could affect the transcription and expression of critical regulatory genes, has key roles in AS. Aberrant DNA methylation including aberrant hypomethylation and hypermethylation plays key roles in endothelial-cell dysfunction, macrophage inflammation, abnormal proliferation of vascular smooth muscle cells, plaque rupture, and thrombosis in AS. Chinese herbal medicines, including single compounds and formulations, showed light on the treatment of AS through regulating the aberrant DNA methylation in AS. Targeting the aberrant DNA methylation may be one of the most important treatment strategies in the cure and prevention of AS. In this review, we focus on the relationship between DNA methylation and AS, as well as the beneficial effects of Chinese herbal medicines on DNA methylation in AS.
Collapse
|
33
|
Essential Role of the 14q32 Encoded miRNAs in Endocrine Tumors. Genes (Basel) 2021; 12:genes12050698. [PMID: 34066712 PMCID: PMC8151414 DOI: 10.3390/genes12050698] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The 14q32 cluster is among the largest polycistronic miRNA clusters. miRNAs encoded here have been implicated in tumorigenesis of multiple organs including endocrine glands. METHODS Critical review of miRNA studies performed in endocrine tumors have been performed. The potential relevance of 14q32 miRNAs through investigating their targets, and integrating the knowledge provided by literature data and bioinformatics predictions have been indicated. RESULTS Pituitary adenoma, papillary thyroid cancer and a particular subset of pheochromocytoma and adrenocortical cancer are characterized by the downregulation of miRNAs encoded by the 14q32 cluster. Pancreas neuroendocrine tumors, most of the adrenocortical cancer and medullary thyroid cancer are particularly distinct, as 14q32 miRNAs were overexpressed. In pheochromocytoma and growth-hormone producing pituitary adenoma, however, both increased and decreased expression of 14q32 miRNAs cluster members were observed. In the background of this phenomenon methodological, technical and biological factors are hypothesized and discussed. The functions of 14q32 miRNAs were also revealed by bioinformatics and literature data mining. CONCLUSIONS 14q32 miRNAs have a significant role in the tumorigenesis of endocrine organs. Regarding their stable expression in the circulation of healthy individuals, further investigation of 14q32 miRNAs could provide a potential for use as biomarkers (diagnostic or prognostic) in endocrine neoplasms.
Collapse
|
34
|
Fischer MA, Vondriska TM. Clinical epigenomics for cardiovascular disease: Diagnostics and therapies. J Mol Cell Cardiol 2021; 154:97-105. [PMID: 33561434 PMCID: PMC8330446 DOI: 10.1016/j.yjmcc.2021.01.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 01/05/2021] [Accepted: 01/10/2021] [Indexed: 12/28/2022]
Abstract
The study of epigenomics has advanced in recent years to span the regulation of a single genetic locus to the structure and orientation of entire chromosomes within the nucleus. In this review, we focus on the challenges and opportunities of clinical epigenomics in cardiovascular disease. As an integrator of genetic and environmental inputs, and because of advances in measurement techniques that are highly reproducible and provide sequence information, the epigenome is a rich source of potential biosignatures of cardiovascular health and disease. Most of the studies to date have focused on the latter, and herein we discuss observations on epigenomic changes in human cardiovascular disease, examining the role of protein modifiers of chromatin, noncoding RNAs and DNA modification. We provide an overview of cardiovascular epigenomics, discussing the challenges of data sovereignty, data analysis, doctor-patient ethics and innovations necessary to implement precision health.
Collapse
Affiliation(s)
- Matthew A Fischer
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine at UCLA, USA.
| | - Thomas M Vondriska
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine at UCLA, USA
| |
Collapse
|
35
|
Moretti R, Giuffré M, Caruso P, Gazzin S, Tiribelli C. Homocysteine in Neurology: A Possible Contributing Factor to Small Vessel Disease. Int J Mol Sci 2021; 22:2051. [PMID: 33669577 PMCID: PMC7922986 DOI: 10.3390/ijms22042051] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 12/19/2022] Open
Abstract
Homocysteine (Hcy) is a sulfur-containing amino acid generated during methionine metabolism, accumulation of which may be caused by genetic defects or the deficit of vitamin B12 and folate. A serum level greater than 15 micro-mols/L is defined as hyperhomocysteinemia (HHcy). Hcy has many roles, the most important being the active participation in the transmethylation reactions, fundamental for the brain. Many studies focused on the role of homocysteine accumulation in vascular or degenerative neurological diseases, but the results are still undefined. More is known in cardiovascular disease. HHcy is a determinant for the development and progression of inflammation, atherosclerotic plaque formation, endothelium, arteriolar damage, smooth muscle cell proliferation, and altered-oxidative stress response. Conversely, few studies focused on the relationship between HHcy and small vessel disease (SVD), despite the evidence that mice with HHcy showed a significant end-feet disruption of astrocytes with a diffuse SVD. A severe reduction of vascular aquaporin-4-water channels, lower levels of high-functioning potassium channels, and higher metalloproteinases are also observed. HHcy modulates the N-homocysteinylation process, promoting a pro-coagulative state and damage of the cellular protein integrity. This altered process could be directly involved in the altered endothelium activation, typical of SVD and protein quality, inhibiting the ubiquitin-proteasome system control. HHcy also promotes a constant enhancement of microglia activation, inducing the sustained pro-inflammatory status observed in SVD. This review article addresses the possible role of HHcy in small-vessel disease and understands its pathogenic impact.
Collapse
Affiliation(s)
- Rita Moretti
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (M.G.); (P.C.)
| | - Mauro Giuffré
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (M.G.); (P.C.)
| | - Paola Caruso
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (M.G.); (P.C.)
| | - Silvia Gazzin
- Italian Liver Foundation, AREA SCIENCE PARK, 34149 Trieste, Italy; (S.G.); (C.T.)
| | - Claudio Tiribelli
- Italian Liver Foundation, AREA SCIENCE PARK, 34149 Trieste, Italy; (S.G.); (C.T.)
| |
Collapse
|
36
|
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
|
37
|
Colpaert RMW, Calore M. Epigenetics and microRNAs in cardiovascular diseases. Genomics 2021; 113:540-551. [PMID: 33482325 DOI: 10.1016/j.ygeno.2020.12.042] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/12/2020] [Accepted: 12/05/2020] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases are among the leading causes of mortality worldwide. Besides environmental and genetic changes, these disorders can be influenced by processes which do not affect DNA sequence yet still play an important role in gene expression and which can be inherited. These so-called 'epigenetic' changes include DNA methylation, histone modifications, and ATP-dependent chromatin remodeling enzymes, which influence chromatin remodeling and gene expression. Next to these, microRNAs are non-coding RNA molecules that silence genes post-transcriptionally. Both epigenetic factors and microRNAs are known to influence cardiac development and homeostasis, in an individual fashion but also in a complex regulatory network. In this review, we will discuss how epigenetic factors and microRNAs interact with each other and how together they can influence cardiovascular diseases.
Collapse
Affiliation(s)
- Robin M W Colpaert
- Department of Molecular Genetics, Faculty of Health, Medicine and Life Sciences, Faculty of Science and Engineering, Maastricht University, the Netherlands
| | - Martina Calore
- Department of Molecular Genetics, Faculty of Health, Medicine and Life Sciences, Faculty of Science and Engineering, Maastricht University, the Netherlands.
| |
Collapse
|
38
|
Rui T, Xu S, Zhang X, Huang H, Feng S, Zhan S, Xie H, Zhou L, Ling Q, Zheng S. The chromosome 19 microRNA cluster, regulated by promoter hypomethylation, is associated with tumour burden and poor prognosis in patients with hepatocellular carcinoma. J Cell Physiol 2020; 235:6103-6112. [PMID: 31975381 DOI: 10.1002/jcp.29538] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 01/07/2020] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) is still one of the major malignant tumours with poor prognosis. The chromosome 19 microRNA cluster (C19MC) is the largest miRNA cluster, and its functions and regulatory mechanisms remain unclear in HCC. We extracted data from 373 HCC samples and 50 non-tumour samples from The Cancer Genome Atlas database. The differential expression levels and methylation levels of C19MC as well as the correlation between them were analysed. We evaluated the correlation between the expression levels of C19MC and the clinical features. We further performed prognostic analysis for C19MC and analysed the bioinformatic function. C19MC had upregulated expression levels and promoter hypomethylation in HCC. A significant negative correlation between the high expression and low methylation level of C19MC was obtained. In addition, the positive correlation between the expression levels of C19MC and the tumour grade, tumour stage and T-stage is shown. Three miRNAs (mir-512-1, mir-516a-1, mir-519a-2) were negatively associated with overall survival on the basis of the Kaplan-Meier analysis and the 3-miRNA signature was significant for the prognostic assessment of HCC. A bioinformatic enrichment analysis suggested that the target genes of the 3 miRNAs may be associated with mitogen-activated protein kinase pathways related to cancer invasion. In summary, our novel study demonstrated that the hypomethylation of promoters upregulates the expression levels of C19MC and that C19MC may represent a potential new candidate for the diagnosis and therapy of HCC.
Collapse
Affiliation(s)
- Tao Rui
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, Zhejiang, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, Zhejiang, China
- Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Siyi Xu
- Department of Electrocardiographic and Cardiac Examination, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Xueyou Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, Zhejiang, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, Zhejiang, China
- Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haitao Huang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, Zhejiang, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, Zhejiang, China
- Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shi Feng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, Zhejiang, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, Zhejiang, China
- Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shaowei Zhan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, Zhejiang, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, Zhejiang, China
- Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haiyang Xie
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, Zhejiang, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, Zhejiang, China
- Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, Zhejiang, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, Zhejiang, China
- Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qi Ling
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, Zhejiang, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, Zhejiang, China
- Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, Zhejiang, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, Zhejiang, China
- Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
39
|
de la Rocha C, Zaina S, Lund G. Is Any Cardiovascular Disease-Specific DNA Methylation Biomarker Within Reach? Curr Atheroscler Rep 2020; 22:62. [DOI: 10.1007/s11883-020-00875-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
40
|
Carbone F, Montecucco F, Xu S, Banach M, Jamialahmadi T, Sahebkar A. Epigenetics in atherosclerosis: key features and therapeutic implications. Expert Opin Ther Targets 2020; 24:719-721. [PMID: 32354276 DOI: 10.1080/14728222.2020.1764535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 04/30/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Federico Carbone
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network , Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa , Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa , Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa , Genoa, Italy
| | - Suowen Xu
- Division of Life Sciences and Medicine, University of Science and Technology of China , Hefei, Anhui, China
| | - Maciej Banach
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz , Poland
- Polish Mother's Memorial Hospital Research Institute (PMMHRI) , Lodz, Poland
| | - Tannaz Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences , Mashhad, Iran
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University , Quchan, Iran
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences , Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences , Mashhad, Iran
- Halal Research Center of IRI, FDA , Tehran, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences , Mashhad, Iran
| |
Collapse
|
41
|
Salameh Y, Bejaoui Y, El Hajj N. DNA Methylation Biomarkers in Aging and Age-Related Diseases. Front Genet 2020; 11:171. [PMID: 32211026 PMCID: PMC7076122 DOI: 10.3389/fgene.2020.00171] [Citation(s) in RCA: 168] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 02/13/2020] [Indexed: 12/11/2022] Open
Abstract
Recent research efforts provided compelling evidence of genome-wide DNA methylation alterations in aging and age-related disease. It is currently well established that DNA methylation biomarkers can determine biological age of any tissue across the entire human lifespan, even during development. There is growing evidence suggesting epigenetic age acceleration to be strongly linked to common diseases or occurring in response to various environmental factors. DNA methylation based clocks are proposed as biomarkers of early disease risk as well as predictors of life expectancy and mortality. In this review, we will summarize key advances in epigenetic clocks and their potential application in precision health. We will also provide an overview of progresses in epigenetic biomarker discovery in Alzheimer's, type 2 diabetes, and cardiovascular disease. Furthermore, we will highlight the importance of prospective study designs to identify and confirm epigenetic biomarkers of disease.
Collapse
Affiliation(s)
| | | | - Nady El Hajj
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
42
|
Schiano C, Benincasa G, Franzese M, Della Mura N, Pane K, Salvatore M, Napoli C. Epigenetic-sensitive pathways in personalized therapy of major cardiovascular diseases. Pharmacol Ther 2020; 210:107514. [PMID: 32105674 DOI: 10.1016/j.pharmthera.2020.107514] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The complex pathobiology underlying cardiovascular diseases (CVDs) has yet to be explained. Aberrant epigenetic changes may result from alterations in enzymatic activities, which are responsible for putting in and/or out the covalent groups, altering the epigenome and then modulating gene expression. The identification of novel individual epigenetic-sensitive trajectories at single cell level might provide additional opportunities to establish predictive, diagnostic and prognostic biomarkers as well as drug targets in CVDs. To date, most of studies investigated DNA methylation mechanism and miRNA regulation as epigenetics marks. During atherogenesis, big epigenetic changes in DNA methylation and different ncRNAs, such as miR-93, miR-340, miR-433, miR-765, CHROME, were identified into endothelial cells, smooth muscle cells, and macrophages. During man development, lipid metabolism, inflammation and homocysteine homeostasis, alter vascular transcriptional mechanism of fundamental genes such as ABCA1, SREBP2, NOS, HIF1. At histone level, increased HDAC9 was associated with matrix metalloproteinase 1 (MMP1) and MMP2 expression in pro-inflammatory macrophages of human carotid plaque other than to have a positive effect on toll like receptor signaling and innate immunity. HDAC9 deficiency promoted inflammation resolution and reverse cholesterol transport, which might block atherosclerosis progression and promote lesion regression. Here, we describe main human epigenetic mechanisms involved in atherosclerosis, coronary heart disease, ischemic stroke, peripheral artery disease; cardiomyopathy and heart failure. Different epigenetics mechanisms are activated, such as regulation by circular RNAs, as MICRA, and epitranscriptomics at RNA level. Moreover, in order to open new frontiers for precision medicine and personalized therapy, we offer a panoramic view on the most innovative bioinformatic tools designed to identify putative genes and molecular networks underlying CVDs in man.
Collapse
Affiliation(s)
- Concetta Schiano
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Giuditta Benincasa
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | | | | | | | - Claudio Napoli
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; IRCCS SDN, Naples, Italy
| |
Collapse
|
43
|
Stone G, Choi A, Meritxell O, Gorham J, Heydarpour M, Seidman CE, Seidman JG, Aranki SF, Body SC, Carey VJ, Raby BA, Stranger BE, Muehlschlegel JD. Sex differences in gene expression in response to ischemia in the human left ventricular myocardium. Hum Mol Genet 2020; 28:1682-1693. [PMID: 30649309 DOI: 10.1093/hmg/ddz014] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/20/2018] [Accepted: 01/09/2019] [Indexed: 01/28/2023] Open
Abstract
Sex differences exist in the prevalence, presentation and outcomes of ischemic heart disease (IHD). Females have higher risk of heart failure post-myocardial infarction relative to males and are two to three times more likely to die after coronary artery bypass grafting surgery. We examined sex differences in human myocardial gene expression in response to ischemia. Left ventricular biopsies from 68 male/46 female patients undergoing aortic valve replacement surgery were obtained at baseline and after a median 74 min of cold cardioplegic arrest/ischemia. Transcriptomes were quantified by RNA-sequencing. Cell-type enrichment analysis was used to estimate the identity and relative proportions of different cell types in each sample. A sex-specific response to ischemia was observed for 271 genes. Notably, the expression FAM5C, PLA2G4E and CYP1A1 showed an increased expression in females compared to males due to ischemia and DIO3, MT1G and CMA1 showed a decreased expression in females compared to males due to ischemia. Functional annotation analysis revealed sex-specific modulation of the oxytocin signaling pathway and common pathway of fibrin clot formation. Expression quantitative trait locus (eQTL) analysis identified variant-by-sex interaction eQTLs, indicative of sex differences in the genotypic effects on gene expression. Cell-type enrichment analysis showed sex-bias in proportion of specific cell types. Common lymphoid progenitor cells and M2 macrophages were found to increase in female samples from pre- to post-ischemia, but no change was observed in male samples. These differences in response to myocardial ischemia provide insight into the sexual dimorphism of IHD and may aid in the development of sex-specific therapies that reduce myocardial injury.
Collapse
Affiliation(s)
- Gregory Stone
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ashley Choi
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Oliva Meritxell
- Institute for Genomics and Systems Biology, Section of Genetic Medicine, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Joshua Gorham
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Mahyar Heydarpour
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Jon G Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Sary F Aranki
- Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Simon C Body
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vincent J Carey
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Benjamin A Raby
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Barbara E Stranger
- Institute for Genomics and Systems Biology, Section of Genetic Medicine, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Jochen D Muehlschlegel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
44
|
Zhang Q, Nettleship I, Schmelzer E, Gerlach J, Zhang X, Wang J, Liu C. Tissue Engineering and Regenerative Medicine Therapies for Cell Senescence in Bone and Cartilage. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:64-78. [DOI: 10.1089/ten.teb.2019.0215] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Qinghao Zhang
- Department of Materials Science and Engineering, East China University of Science and Technology, Shanghai, P.R. China
- Department of Mechanical Engineering and Materials Science, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ian Nettleship
- Department of Mechanical Engineering and Materials Science, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Eva Schmelzer
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jorg Gerlach
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xuewei Zhang
- Department of Materials Science and Engineering, East China University of Science and Technology, Shanghai, P.R. China
| | - Jing Wang
- Department of Materials Science and Engineering, East China University of Science and Technology, Shanghai, P.R. China
| | - Changsheng Liu
- Department of Materials Science and Engineering, East China University of Science and Technology, Shanghai, P.R. China
| |
Collapse
|
45
|
SULT2B1b inhibits reverse cholesterol transport and promotes cholesterol accumulation and inflammation in lymphocytes from AMI patients with low LDL-C levels. Clin Sci (Lond) 2020; 134:273-287. [PMID: 31957803 DOI: 10.1042/cs20190459] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 01/19/2023]
Abstract
The current main treatment for coronary artery disease (CAD) is to reduce low-density lipoprotein cholesterol (LDL-C) by statins, which could decrease the incidence of major adverse cardiovascular events (MACEs) by 30%. However, many residual risks still remain. To clarify the mechanism involved, we studied patients with acute myocardial infarction (AMI) with low LDL-C levels. Lymphocytes were isolated, and it was found that despite no difference in plasma LDL-C level, the lymphocyte cholesterol content was higher in AMI patient than those in non-CAD patients; thus, the decrease in intracellular cholesterol content was inconsistent with that in the plasma. Additionally, [3H]-cholesterol efflux rates were lower and mRNA levels of the inflammatory factors tumour necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) higher in AMI lymphocytes. It was found that sulphotransferase 2B1b (SULT2B1b) expression was higher in AMI lymphocytes. Further research using Jurkat T lymphocytes confirmed that SULT2B1b knockdown increased cholesterol efflux capacity and decreased mRNA levels of TNF-α and IFN-γ by increasing liver X receptor (LXR)-β levels. Furthermore, the degree of CpG island methylation in the SULT2B1b promoter was reduced in cells from AMI patients. In conclusion, SULT2B1b up-regulation due to hypomethylation of its promoter promotes cholesterol accumulation and inflammation by inhibiting LXR-β in lymphocytes of AMI patients with low LDL-C levels. Therefore, reducing intracellular cholesterol is also important as plasma cholesterol levels. Therapeutic approaches to decrease SULT2B1b expression might be potentially beneficial for CAD prevention by decreasing intracellular cholesterol.
Collapse
|
46
|
Shen W, Gao C, Cueto R, Liu L, Fu H, Shao Y, Yang WY, Fang P, Choi ET, Wu Q, Yang X, Wang H. Homocysteine-methionine cycle is a metabolic sensor system controlling methylation-regulated pathological signaling. Redox Biol 2020; 28:101322. [PMID: 31605963 PMCID: PMC6812029 DOI: 10.1016/j.redox.2019.101322] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/03/2019] [Accepted: 09/06/2019] [Indexed: 12/14/2022] Open
Abstract
Homocysteine-Methionine (HM) cycle produces universal methyl group donor S-adenosylmethione (SAM), methyltransferase inhibitor S-adenosylhomocysteine (SAH) and homocysteine (Hcy). Hyperhomocysteinemia (HHcy) is established as an independent risk factor for cardiovascular disease (CVD) and other degenerative disease. We selected 115 genes in the extended HM cycle (31 metabolic enzymes and 84 methyltransferases), examined their protein subcellular location/partner protein, investigated their mRNA levels and mapped their corresponding histone methylation status in 35 disease conditions via mining a set of public databases and intensive literature research. We have 6 major findings. 1) All HM metabolic enzymes are located only in the cytosol except for cystathionine-β-synthase (CBS), which was identified in both cytosol and nucleus. 2) Eight disease conditions encountered only histone hypomethylation on 8 histone residues (H3R2/K4/R8/K9/K27/K36/K79 and H4R3). Nine disease conditions had only histone hypermethylation on 8 histone residues (H3R2/K4/K9/K27/K36/K79 and H4R3/K20). 3) We classified 9 disease types with differential HM cycle expression pattern. Eleven disease conditions presented most 4 HM cycle pathway suppression. 4) Three disease conditions had all 4 HM cycle pathway suppression and only histone hypomethylation on H3R2/K4/R8/K9/K36 and H4R3. 5) Eleven HM cycle metabolic enzymes interact with 955 proteins. 6) Five paired HM cycle proteins interact with each other. We conclude that HM cycle is a key metabolic sensor system which mediates receptor-independent metabolism-associated danger signal recognition and modulates SAM/SAH-dependent methylation in disease conditions and that hypomethylation on frequently modified histone residues is a key mechanism for metabolic disorders, autoimmune disease and CVD. We propose that HM metabolism takes place in the cytosol, that nuclear methylation equilibration requires a nuclear-cytosol transfer of SAM/SAH/Hcy, and that Hcy clearance is essential for genetic protection.
Collapse
Affiliation(s)
- Wen Shen
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China; Centers for Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Chao Gao
- Centers for Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Ramon Cueto
- Centers for Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Lu Liu
- Centers for Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Hangfei Fu
- Centers for Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Ying Shao
- Centers for Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - William Y Yang
- Centers for Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Pu Fang
- Centers for Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Eric T Choi
- Centers for Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA; Division of Vascular & Endovascular Surgery, Department of Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Qinghua Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| | - Xiaofeng Yang
- Centers for Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Hong Wang
- Centers for Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.
| |
Collapse
|
47
|
Ikeda KT, Hale PT, Pauciulo MW, Dasgupta N, Pastura PA, Le Cras TD, Pandey MK, Nichols WC. Hypoxia-induced Pulmonary Hypertension in Different Mouse Strains: Relation to Transcriptome. Am J Respir Cell Mol Biol 2019; 60:106-116. [PMID: 30134121 DOI: 10.1165/rcmb.2017-0435oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Patients with pulmonary arterial hypertension (PAH) can harbor mutations in several genes, most commonly in BMPR2. However, disease penetrance in patients with BMPR2 mutations is low. In addition, most patients do not carry known PAH gene mutations, suggesting that other factors determine susceptibility to PAH. To begin to identify additional genomic factors contributing to PAH pathogenesis, we exposed 32 mouse strains to chronic hypoxia. We found that the PL/J strain has extremely high right ventricular systolic pressure (RVSP; 86.58 mm Hg) but minimal lung remodeling. To identify potential genomic factors contributing to the high RVSP, RNAseq analysis of PL/J lung mRNAs and microRNAs (miRNAs) after hypoxia was performed, and it demonstrated that 4 of 43 upregulated miRNAs in the Dlk1-Dio3 imprinting region are predicted to target T cell marker mRNAs. These target mRNAs, as well as the numbers of T cells were downregulated. In addition, C5a and its receptor, C5AR1, were increased. Analysis of Rho-associated protein kinase (Rock) 2 mRNA expression, in the RhoA/Rock pathway, demonstrated a significant increase in PL/J. Inhibition of Rock2 ameliorated a portion of the elevated RVSP. In addition, we identified miR-150-5p as a potential regulator of Rock2 expression. In conclusion, we identified two possible pathways contributing to the hypoxia pulmonary hypertension phenotype of extreme RVSP elevation: aberrant T cell expression driven by hypoxia-induced miRNAs and increased expression of C5a and C5AR1. We suggest that the PL/J mouse will be a good model for seeking mechanism(s) of RVSP elevation in hypoxia-induced PAH.
Collapse
Affiliation(s)
| | | | - Michael W Pauciulo
- 1 Division of Human Genetics and.,2 Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | - Patricia A Pastura
- 3 Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; and
| | - Timothy D Le Cras
- 3 Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; and.,2 Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | - William C Nichols
- 1 Division of Human Genetics and.,2 Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
48
|
Huo KG, Richer C, Berillo O, Mahjoub N, Fraulob-Aquino JC, Barhoumi T, Ouerd S, Coelho SC, Sinnett D, Paradis P, Schiffrin EL. miR-431-5p Knockdown Protects Against Angiotensin II-Induced Hypertension and Vascular Injury. Hypertension 2019; 73:1007-1017. [PMID: 30929512 DOI: 10.1161/hypertensionaha.119.12619] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Vascular injury is an early manifestation in hypertension and a cause of end-organ damage. MicroRNAs play an important role in cardiovascular disease, but their implication in vascular injury in hypertension remains unclear. This study revealed using an unbiased approach, microRNA and mRNA sequencing with molecular interaction analysis, a microRNA-transcription factor coregulatory network involved in vascular injury in mice made hypertensive by 14-day Ang II (angiotensin II) infusion. A candidate gene approach identified upregulated miR-431-5p encoded in the conserved 12qF1 (14q32 in humans) microRNA cluster, whose expression correlated with blood pressure, and which has been shown to be upregulated in human atherosclerosis, as a potential key regulator in Ang II-induced vascular injury. Gain- and loss-of-function in human vascular smooth muscle cells demonstrated that miR-431-5p regulates in part gene expression by targeting ETS homologous factor. In vivo miR-431-5p knockdown delayed Ang II-induced blood pressure elevation and reduced vascular injury in mice, which demonstrated its potential as a target for treatment of hypertension and vascular injury.
Collapse
Affiliation(s)
- Ku-Geng Huo
- From the Vascular and Hypertension Research Unit, Lady Davis Institute for Medical Research (K.-G.H., O.B., N.M., J.C.F.-A., T.B., S.O., S.C.C., P.P., E.L.S.), McGill University, Montreal, Quebec, Canada
| | - Chantal Richer
- Division of Hematology-Oncology, Research Center, Centre Hospitalier Universitaire Sainte-Justine (C.R., D.S.), Université de Montréal, Montreal, Quebec, Canada
| | - Olga Berillo
- From the Vascular and Hypertension Research Unit, Lady Davis Institute for Medical Research (K.-G.H., O.B., N.M., J.C.F.-A., T.B., S.O., S.C.C., P.P., E.L.S.), McGill University, Montreal, Quebec, Canada
| | - Nada Mahjoub
- From the Vascular and Hypertension Research Unit, Lady Davis Institute for Medical Research (K.-G.H., O.B., N.M., J.C.F.-A., T.B., S.O., S.C.C., P.P., E.L.S.), McGill University, Montreal, Quebec, Canada
| | - Julio C Fraulob-Aquino
- From the Vascular and Hypertension Research Unit, Lady Davis Institute for Medical Research (K.-G.H., O.B., N.M., J.C.F.-A., T.B., S.O., S.C.C., P.P., E.L.S.), McGill University, Montreal, Quebec, Canada
| | - Tlili Barhoumi
- From the Vascular and Hypertension Research Unit, Lady Davis Institute for Medical Research (K.-G.H., O.B., N.M., J.C.F.-A., T.B., S.O., S.C.C., P.P., E.L.S.), McGill University, Montreal, Quebec, Canada
| | - Sofiane Ouerd
- From the Vascular and Hypertension Research Unit, Lady Davis Institute for Medical Research (K.-G.H., O.B., N.M., J.C.F.-A., T.B., S.O., S.C.C., P.P., E.L.S.), McGill University, Montreal, Quebec, Canada
| | - Suellen C Coelho
- From the Vascular and Hypertension Research Unit, Lady Davis Institute for Medical Research (K.-G.H., O.B., N.M., J.C.F.-A., T.B., S.O., S.C.C., P.P., E.L.S.), McGill University, Montreal, Quebec, Canada
| | - Daniel Sinnett
- Division of Hematology-Oncology, Research Center, Centre Hospitalier Universitaire Sainte-Justine (C.R., D.S.), Université de Montréal, Montreal, Quebec, Canada.,Department of Pediatrics, Faculty of Medicine (D.S.), Université de Montréal, Montreal, Quebec, Canada
| | - Pierre Paradis
- From the Vascular and Hypertension Research Unit, Lady Davis Institute for Medical Research (K.-G.H., O.B., N.M., J.C.F.-A., T.B., S.O., S.C.C., P.P., E.L.S.), McGill University, Montreal, Quebec, Canada
| | - Ernesto L Schiffrin
- From the Vascular and Hypertension Research Unit, Lady Davis Institute for Medical Research (K.-G.H., O.B., N.M., J.C.F.-A., T.B., S.O., S.C.C., P.P., E.L.S.), McGill University, Montreal, Quebec, Canada.,Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital (E.L.S.), McGill University, Montreal, Quebec, Canada
| |
Collapse
|
49
|
Suades R, Cosentino F. The environment, epigenetic landscape and cardiovascular risk. Cardiovasc Res 2019; 115:e147-e150. [PMID: 31230064 DOI: 10.1093/cvr/cvz150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Rosa Suades
- Cardiology Unit, Department of Medicine Solna, Karolinska Institute and Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Francesco Cosentino
- Cardiology Unit, Department of Medicine Solna, Karolinska Institute and Karolinska University Hospital, Solna, Stockholm, Sweden
| |
Collapse
|
50
|
Xia Z, Gu M, Jia X, Wang X, Wu C, Guo J, Zhang L, Du Y, Wang J. Integrated DNA methylation and gene expression analysis identifies SLAMF7 as a key regulator of atherosclerosis. Aging (Albany NY) 2019; 10:1324-1337. [PMID: 29905534 PMCID: PMC6046250 DOI: 10.18632/aging.101470] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 06/04/2018] [Indexed: 12/27/2022]
Abstract
Atherosclerosis (AS) is a multifactorial disease. Exploration of DNA methylation in regulating gene transcription in a cell type- and stage-specific manner will shed light on understanding the biological processes associated with plaque stability. We identified 174 up-regulated genes with hypo-methylation in the promoter, and 86 down-regulated genes with hyper-methylation in the promoter, in AS vs. healthy controls. Among them, high expression of signaling lymphocytic activation molecule 7 (SLAM7) was examined in carotid plaque vs. intact tissue, in advanced plaque vs. early atherosclerotic tissue, and SLAMF7 protein expressed significantly higher in the unstable plaques than that in the stable plaques, especially in the CD68-positive macrophages. Depletion of SLAMF7 in plaque-derived macrophages induced a suppressed secretion of proinflammatory cytokines, and inhibited proliferation of vascular smooth muscle cells. These data provide emerging evidence that SLAMF7 could be a target of potential therapeutic intervention in carotid AS.
Collapse
Affiliation(s)
- Zhangyong Xia
- Department of Neurology Liaocheng People's Hospital and Liaocheng Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Mingliang Gu
- Joint Laboratory for Translational Medicine Research, Beijing Institute of Genomics, Chinese Academy of Sciences & Liaocheng People's Hospital, CAS Key Laboratory of Genomic Science and Information Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Xiaodong Jia
- Joint Laboratory for Translational Medicine Research, Beijing Institute of Genomics, Chinese Academy of Sciences and Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Xiaoting Wang
- Taishan Medical University, Taian, Shandong 271016, P.R. China
| | - Chunxia Wu
- Department of Ultrasonic Liaocheng People's Hospital and Liaocheng Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Jiangwen Guo
- Deparment of Neurology Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Liyong Zhang
- Department of Neurosurgery Liaocheng People's Hospital and Liaocheng Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, PR China
| | - Yifeng Du
- Department of Neurology Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Jiyue Wang
- Department of Neurosurgery Liaocheng People's Hospital and Liaocheng Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, PR China
| |
Collapse
|