1
|
Ozuynuk-Ertugrul AS, Kirsan CB, Erkan AF, Ekici B, Komurcu-Bayrak E, Coban N. Genetic variants of ANRIL and coronary artery disease: Insights from a Turkish study population. Gene 2024; 917:148475. [PMID: 38631653 DOI: 10.1016/j.gene.2024.148475] [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: 01/24/2024] [Revised: 03/27/2024] [Accepted: 04/15/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND AND AIM Coronary artery disease (CAD) remains a leading cause of morbidity and mortality globally despite advancements in treatment. Long non-coding RNAs (lncRNAs) play crucial roles in the atherosclerotic process, with ANRIL being one such lncRNA. This study explored the association between ANRIL polymorphisms (rs1333049:C > G, rs564398:T > C, and rs10757274:A > G) and CAD along with CAD risk factors in a Turkish patient group. METHODS The study included 1285 participants, consisting of 736 patients diagnosed with CAD (mean age = 63.3 ± 10.5 years) and 549 non-CAD controls (mean age = 57.52 ± 11.01 years). Genotypes for rs1333049, rs564398, and rs10757274 were determined using qRT-PCR. RESULTS G allele carriage of both rs1333049 and rs10757274 polymorphisms were associated with higher Gensini score, SYNTAX score, total cholesterol, and triglyceride levels in female CAD patients and non-CAD males. Females with rs564398 CC genotype were more susceptible to CAD (p = 0.02) and severe CAD (p = 0.05). Moreover, the G and T alleles of rs10757274 and rs564398 were more prevalent among hypertensive males. Also, carrying the C allele for rs564398 was associated with a decreased risk for type 2 diabetes mellitus (T2DM) (p = 0.02). Besides, carriers of the rs1333049 C allele for decreased risk for T2DM (p = 0.03) and CAD complexed with T2DM (p = 0.04) in logistic regression analyses. CONCLUSIONS In conclusion, selected ANRIL polymorphisms were associated with CAD presence/severity and CAD risk factors, T2DM, and hypertension. Notably, this study, the largest sample-sized study examining the effects of selected polymorphisms on CAD and its risk factors among Turkish individuals, supported the findings of previous studies conducted on different ethnicities.
Collapse
Affiliation(s)
- Aybike Sena Ozuynuk-Ertugrul
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Genetics, Istanbul, Turkey; Istanbul University Institute of Graduate Studies in Health Sciences, Istanbul, Turkey
| | - Cemre Buse Kirsan
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Genetics, Istanbul, Turkey; Istanbul University Institute of Graduate Studies in Health Sciences, Istanbul, Turkey
| | - Aycan Fahri Erkan
- Ufuk University, Faculty of Medicine, Department of Cardiology, Ankara, Turkey
| | - Berkay Ekici
- Ufuk University, Faculty of Medicine, Department of Cardiology, Ankara, Turkey
| | - Evrim Komurcu-Bayrak
- Istanbul University, Istanbul Faculty of Medicine, Department of Medical Genetics, Istanbul, Turkey
| | - Neslihan Coban
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Genetics, Istanbul, Turkey.
| |
Collapse
|
2
|
Le A, Peng H, Golinsky D, Di Scipio M, Lali R, Paré G. What Causes Premature Coronary Artery Disease? Curr Atheroscler Rep 2024; 26:189-203. [PMID: 38573470 DOI: 10.1007/s11883-024-01200-y] [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] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
PURPOSE OF REVIEW This review provides an overview of genetic and non-genetic causes of premature coronary artery disease (pCAD). RECENT FINDINGS pCAD refers to coronary artery disease (CAD) occurring before the age of 65 years in women and 55 years in men. Both genetic and non-genetic risk factors may contribute to the onset of pCAD. Recent advances in the genetic epidemiology of pCAD have revealed the importance of both monogenic and polygenic contributions to pCAD. Familial hypercholesterolemia (FH) is the most common monogenic disorder associated with atherosclerotic pCAD. However, clinical overreliance on monogenic genes can result in overlooked genetic causes of pCAD, especially polygenic contributions. Non-genetic factors, notably smoking and drug use, are also important contributors to pCAD. Cigarette smoking has been observed in 25.5% of pCAD patients relative to 12.2% of non-pCAD patients. Finally, myocardial infarction (MI) associated with spontaneous coronary artery dissection (SCAD) may result in similar clinical presentations as atherosclerotic pCAD. Recognizing the genetic and non-genetic causes underlying pCAD is important for appropriate prevention and treatment. Despite recent progress, pCAD remains incompletely understood, highlighting the need for both awareness and research.
Collapse
Affiliation(s)
- Ann Le
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Department of Medical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Helen Peng
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Danielle Golinsky
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- School of Nursing, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Matteo Di Scipio
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Department of Medical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Ricky Lali
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Guillaume Paré
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada.
- Department of Medical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
- Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada.
- Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada.
| |
Collapse
|
3
|
Xie F, Wang D, Cheng M. CDKN2B-AS1 may act as miR-92a-3p sponge in coronary artery disease. Minerva Cardiol Angiol 2024; 72:125-133. [PMID: 38231078 DOI: 10.23736/s2724-5683.23.06441-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
BACKGROUND LncRNAs, miRNAs, and the sponge effect between them exert diverse biological influences on the pathogenesis and progression of coronary artery disease (CAD), thus necessitating an exploration of the lncRNA-miRNA-gene regulatory network in CAD. METHODS Expression profile GSE98583 was obtained from NCBI, containing the data of 12 CAD patients and 6 controls. Limma package was utilized to determine the differentially expressed genes (DEGs). Functional enrichment analysis was performed by DAVID. The CAD-related miRNA-DEG associations were retrieved via HMDD and miRTarBase, and the CAD-related lncRNA-miRNA associations were retrieved via LncRNADisease and starBase. The CAD-related lncRNA-miRNA-DEG regulatory network was constructed by combining these associations. The dual luciferase test was carried out to validate the connections among lncRNA, miRNA, and gene. RESULTS Overall, 534 DEGs were identified between CAD samples and controls, including 243 up-regulated and 291 down-regulated, and were enriched in various gene ontology biological processes and KEGG pathways. The CAD-related miRNAs targeting DEGs included hsa-miR-206, has-miR-320b, has-miR-4513, has-miR-765, and has-miR-92a-3p, and hsa-miR-92a-3p regulated the most DEGs. In the lncRNA-miRNA associations, only CDKN2B-AS1 regulated the CAD-related miRNA, hsa-miR-92a-3p, which was validated using the dual luciferase test. CONCLUSIONS CDKN2B-AS1 may act as an hsa-miR-92a-3p sponge to regulate the downstream DEGs in CAD. CDKN2B-AS1/ hsa-miR-92a-3p/GATA2 might be a novel mechanism for CAD.
Collapse
Affiliation(s)
- Fei Xie
- Department of Cardiac Surgery, The Second Hospital Affiliated to Harbin Medical University, Harbin, Heilongjiang, China
| | - Dan Wang
- Department of Cardiac Surgery, The Second Hospital Affiliated to Harbin Medical University, Harbin, Heilongjiang, China
| | - Ming Cheng
- Department of Cardiac Surgery, The Second Hospital Affiliated to Harbin Medical University, Harbin, Heilongjiang, China -
| |
Collapse
|
4
|
Jiao Y, Meng F, Ma G, Lei H, Liu J. An increase in a long noncoding RNA ANRIL in peripheral plasma is an indicator of stable angina. Clinics (Sao Paulo) 2023; 78:100289. [PMID: 37852142 PMCID: PMC10585623 DOI: 10.1016/j.clinsp.2023.100289] [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: 03/10/2023] [Revised: 09/15/2023] [Accepted: 09/26/2023] [Indexed: 10/20/2023] Open
Abstract
INTRODUCTION Stable angina develops during physical activity or stress, and it is typically an aspect of Coronary Heart Disease (CHD) that can lead to arrhythmia, heart failure and even sudden death. ANRIL, an Antisense Noncoding RNA gene in the INK4 Locus, is associated with multiple disorders including CHD; however, expressional levels of ANRIL in between patients with stable angina and myocardial infarction, one of the acute coronary syndrome, have not been clarified yet. METHODS The authors enrolled 62 patients with myocardial infarction and 59 with stable angina before primary percutaneous coronary intervention, as well as 48 healthy volunteers. Their peripheral blood was collected for analysis of ANRIL and cardiac troponin I, a traditional diagnostic index of CHD by real-time PCR. RESULTS The data showed that ANRIL is a better diagnostic indicator than cardiac troponin I in patients with stable angina and that the levels of ANRIL are higher in patients with stable angina than those with the myocardial infarction. DISCUSSION The levels of ANRIL in peripheral plasma could be used as a good biomarker for stable angina.
Collapse
Affiliation(s)
- Yunjuan Jiao
- Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, China; Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, China
| | - Fanming Meng
- School of Forensic, School of Basic Medical Sciences, Central South University, China
| | - Gaoen Ma
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, China.
| | - Hetian Lei
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, China.
| | - Junwen Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, China; China-Africa Research Center of Infectious Diseases, School of Basic Medical Sciences, Central South University, Hunan Province, China.
| |
Collapse
|
5
|
Zhu Y, Tazearslan C, Rosenfeld MG, Fiser A, Suh Y. Identification and functional validation of an enhancer variant in the 9p21.3 locus associated with glaucoma risk and elevated expression of p16 INK4a. Aging Cell 2023; 22:e13908. [PMID: 37345431 PMCID: PMC10497822 DOI: 10.1111/acel.13908] [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: 12/11/2017] [Revised: 05/05/2023] [Accepted: 05/30/2023] [Indexed: 06/23/2023] Open
Abstract
Glaucoma is a leading cause of irreversible blindness, with advanced age being the single most significant risk factor. However, the mechanisms underlying the relationship between aging and glaucoma remain unclear. Genome-wide association studies (GWAS) have successfully identified genetic variants strongly associated with increased glaucoma risk. Understanding how these variants function in pathogenesis is crucial for translating genetic associations into molecular mechanisms and, ultimately, clinical applications. The chromosome 9p21.3 locus is among the most replicated glaucoma risk loci discovered by GWAS. Nonetheless, the absence of protein-coding genes in the locus makes interpreting the disease association challenging, leaving the causal variant and molecular mechanism elusive. In this study, we report the identification of a functional glaucoma risk variant, rs6475604. By employing computational and experimental methods, we demonstrated that rs6475604 resides in a repressive regulatory element. Risk allele of rs6475604 disrupts the binding of YY1, a transcription factor known to repress the expression of a neighboring gene in 9p21.3, p16INK4A, which plays a crucial role in cellular senescence and aging. These findings suggest that the glaucoma disease variant contributes to accelerated senescence, providing a molecular link between glaucoma risk and an essential cellular mechanism for human aging.
Collapse
Affiliation(s)
- Yizhou Zhu
- Department of Obstetrics and GynecologyColumbia UniversityNew YorkNew YorkUSA
| | - Cagdas Tazearslan
- Department of GeneticsAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Michael G. Rosenfeld
- Department of MedicineSchool of MedicineUniversity of CaliforniaLa JollaCaliforniaUSA
- Howard Hughes Medical InstituteUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Andras Fiser
- Department of Systems & Computational BiologyAlbert Einstein College of MedicineNew YorkNew YorkUSA
- Department of BiochemistryAlbert Einstein College of MedicineNew YorkNew YorkUSA
| | - Yousin Suh
- Department of Obstetrics and GynecologyColumbia UniversityNew YorkNew YorkUSA
- Department of Genetics and DevelopmentColumbia UniversityNew YorkNew YorkUSA
| |
Collapse
|
6
|
Zhu Y, Tazearslan C, Rosenfeld MG, Fiser A, Suh Y. Identification and functional validation of an enhancer variant in the 9p21.3 locus associated with glaucoma risk and elevated expression of p16 INK4a. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.18.541339. [PMID: 37292862 PMCID: PMC10245730 DOI: 10.1101/2023.05.18.541339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Glaucoma is a leading cause of irreversible blindness, with advanced age being the single most significant risk factor. However, the mechanisms underlying the relationship between aging and glaucoma remain unclear. Genome-wide association studies (GWAS) have successfully identified genetic variants strongly associated with increased glaucoma risk. Understanding how these variants function in pathogenesis is crucial for translating genetic associations into molecular mechanisms and, ultimately, clinical applications. The chromosome 9p21.3 locus is among the most replicated glaucoma risk loci discovered by GWAS. Nonetheless, the absence of protein-coding genes in the locus makes interpreting the disease association challenging, leaving the causal variant and molecular mechanism elusive. In this study, we report the identification of a functional glaucoma risk variant, rs6475604. By employing computational and experimental methods, we demonstrated that rs6475604 resides in a repressive regulatory element. Risk allele of rs6475604 disrupts the binding of YY1, a transcription factor known to repress the expression of a neighboring gene in 9p21.3, p16INK4A, which plays a crucial role in cellular senescence and aging. These findings suggest that the glaucoma disease variant contributes to accelerated senescence, providing a molecular link between glaucoma risk and an essential cellular mechanism for human aging.
Collapse
Affiliation(s)
- Yizhou Zhu
- Department of Obstetrics and Gynecology, Columbia University, New York, NY10032, USA
| | - Cagdas Tazearslan
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY10461, USA
| | - Michael G. Rosenfeld
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093
| | - Andras Fiser
- Department of Systems & Computational Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Yousin Suh
- Department of Obstetrics and Gynecology, Columbia University, New York, NY10032, USA
- Department of Genetics and Development, Columbia University, New York, NY10032, USA
| |
Collapse
|
7
|
Jiang Y, Zhao Y, Li ZY, Chen S, Fang F, Cai JH. Potential roles of microRNAs and long noncoding RNAs as diagnostic, prognostic and therapeutic biomarkers in coronary artery disease. Int J Cardiol 2023:S0167-5273(23)00478-3. [PMID: 37019219 DOI: 10.1016/j.ijcard.2023.03.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/27/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023]
Abstract
Coronary artery disease (CAD), which is mainly caused by atherosclerotic processes in coronary arteries, became a significant health issue. MicroRNAs (miRNAs), and long noncoding RNAs (lncRNAs), have been shown to be stable in plasma and could thereby be adopted as biomarkers for CAD diagnosis and treatment. MiRNAs can regulate CAD development through different pathways and mechanisms, including modulation of vascular smooth muscle cell (VSMC) activity, inflammatory responses, myocardial injury, angiogenesis, and leukocyte adhesion. Similarly, previously studies have indicated that the causal effects of lncRNAs in CAD pathogenesis and their utility in CAD diagnosis and treatment, has been found to lead to cell cycle transition, proliferation dysregulation, and migration in favour of CAD development. Differential expression of miRNAs and lncRNAs in CAD patients has been identified and served as diagnostic, prognostic and therapeutic biomarkers for the assessment of CAD patients. Thus, in the current review, we summarize the functions of miRNAs and lncRNAs, which aimed to identify novel targets for the CAD diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Yong Jiang
- Department of Laboratory Medicine, Jilin Medical University, No. 5 Jilin Street, Jilin 132013, China.
| | - Ying Zhao
- Department of Cardiology, Jilin Central Hospital, Jilin 132011, China
| | - Zheng-Yi Li
- Department of Laboratory Medicine, Jilin Medical University, No. 5 Jilin Street, Jilin 132013, China
| | - Shuang Chen
- Department of Laboratory Medicine, Jilin Medical University, No. 5 Jilin Street, Jilin 132013, China
| | - Fang Fang
- Department of Laboratory Medicine, Jilin Medical University, No. 5 Jilin Street, Jilin 132013, China.
| | - Jian-Hui Cai
- Department of Clinical Medicine, Jilin Medical University, Jilin 132013, China; Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin 132013, China.
| |
Collapse
|
8
|
Epigenetics and Gut Microbiota Crosstalk: A potential Factor in Pathogenesis of Cardiovascular Disorders. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120798. [PMID: 36551003 PMCID: PMC9774431 DOI: 10.3390/bioengineering9120798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
Cardiovascular diseases (CVD) are the leading cause of mortality, morbidity, and "sudden death" globally. Environmental and lifestyle factors play important roles in CVD susceptibility, but the link between environmental factors and genetics is not fully established. Epigenetic influence during CVDs is becoming more evident as its direct involvement has been reported. The discovery of epigenetic mechanisms, such as DNA methylation and histone modification, suggested that external factors could alter gene expression to modulate human health. These external factors also influence our gut microbiota (GM), which participates in multiple metabolic processes in our body. Evidence suggests a high association of GM with CVDs. Although the exact mechanism remains unclear, the influence of GM over the epigenetic mechanisms could be one potential pathway in CVD etiology. Both epigenetics and GM are dynamic processes and vary with age and environment. Changes in the composition of GM have been found to underlie the pathogenesis of metabolic diseases via modulating epigenetic changes in the form of DNA methylation, histone modifications, and regulation of non-coding RNAs. Several metabolites produced by the GM, including short-chain fatty acids, folates, biotin, and trimethylamine-N-oxide, have the potential to regulate epigenetics, apart from playing a vital role in normal physiological processes. The role of GM and epigenetics in CVDs are promising areas of research, and important insights in the field of early diagnosis and therapeutic approaches might appear soon.
Collapse
|
9
|
Wei B, Liu Y, Li H, Peng Y, Luo Z. Effect of 9p21.3 (lncRNA and CDKN2A/2B) variant on lipid profile. Front Cardiovasc Med 2022; 9:946289. [PMID: 36158791 PMCID: PMC9489913 DOI: 10.3389/fcvm.2022.946289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
Background Several 9p21.3 variants, such as rs1333049, rs4977574, rs10757274, rs10757278, and rs10811661, identified from recent genome-wide association studies (GWASs) are reported to be associated with coronary artery disease (CAD) susceptibility but independent of dyslipidemia. This study investigated whether these 9p21.3 variants influenced lipid profiles. Methods and results By searching the PubMed and Cochrane databases, 101,099 individuals were included in the analysis. The consistent finding for the rs1333049 C allele on lipid profiles increased the triglyceride (TG) levels. Moreover, the rs4977574 G allele and the rs10757274 G allele, respectively, increased low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol (HDL-C) levels. However, the rs10811661 C allele largely reduced LDL-C levels. Subgroup analyses indicated that the effects of the rs1333049 C allele, rs4977574 G allele, and rs10757274 G allele on lipid profiles were stronger in Whites compared with Asians. In contrast, the effect of the rs10811661 C allele on lipid profiles was stronger in Asians compared with Whites. Conclusion The rs1333049 C allele, rs4977574 G allele, and rs10757274 G allele of lncRNA, and the rs10811661 G allele of CDKN2A/2B had a significant influence on lipid levels, which may help the understanding of the underlying mechanisms between 9p21.3 variants and CAD.
Collapse
Affiliation(s)
- Baozhu Wei
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, China
- *Correspondence: Baozhu Wei,
| | - Yang Liu
- Department of Endocrinology, China Resources and WISCO General Hospital, Wuhan, China
| | - Hang Li
- Department of Gerontology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yuanyuan Peng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Zhi Luo
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Zhi Luo,
| |
Collapse
|
10
|
Association of lipid metabolism-related gene promoter methylation with risk of coronary artery disease. Mol Biol Rep 2022; 49:9373-9378. [PMID: 35941416 DOI: 10.1007/s11033-022-07789-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 10/15/2022]
Abstract
BACKGROUND Coronary artery disease (CAD) is a complex disease that is influenced by environmental and genetic factors. Lipid levels are regarded as a major risk factor for CAD, and epigenetic mechanisms might be involved in the regulation of CAD development. This study was designed to investigate the association between the DNA methylation status of 8 lipid metabolism-related genes and the risk of CAD in the Chinese Han population. METHODS A total of 260 individuals were sampled in this study, including 120 CAD cases and 140 normal healthy controls. DNA methylation status was tested via targeted bisulfite sequencing. RESULTS The results indicated a significant association between hypomethylation of the APOC3, CETP and APOC1 gene promoters and the risk of CAD. Individuals with higher methylation levels of the APOA5 and LIPC gene promoters had increased risks for CAD. In addition, ANGPTL4 methylation level was significantly associated with CAD in males but not females. There were no significant differences in the methylation levels of the APOB and PCSK9 gene promoters between CAD patients and controls. CONCLUSIONS The methylation status of the APOC3, APOA5, LIPC, CETP and APOC1 gene promoters may be associated with the development of CAD.
Collapse
|
11
|
Xiong T, Xiao B, Wu Y, Liu Y, Li Q. Upregulation of the Long Non-coding RNA LINC01480 Is Associated With Immune Infiltration in Coronary Artery Disease Based on an Immune-Related lncRNA-mRNA Co-expression Network. Front Cardiovasc Med 2022; 9:724262. [PMID: 35557532 PMCID: PMC9086407 DOI: 10.3389/fcvm.2022.724262] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 03/18/2022] [Indexed: 12/03/2022] Open
Abstract
Coronary artery disease (CAD) is considered one of the leading causes of death worldwide. Although dysregulation of long non-coding RNAs (lncRNAs) has been reported to be associated with the initiation and progression of CAD, the knowledge regarding their specific functions as well their physiological/pathological significance in CAD is very limited. In this study, we aimed to systematically analyze immune-related lncRNAs in CAD and explore the relationship between key immune-related lncRNAs and the immune cell infiltration process. Based on differential expression analysis of mRNAs and lncRNAs, an immune-related lncRNA-mRNA weighted gene co-expression network containing 377 lncRNAs and 119 mRNAs was constructed. LINC01480 and AL359237.1 were identified as the hub immune-related lncRNAs in CAD using the random forest-recursive feature elimination and least absolute shrinkage and selection operator logistic regression. Furthermore, 93 CAD samples were divided into two subgroups according to the expression values of LINC01480 and AL359237.1 by consensus clustering analysis. By performing gene set enrichment analysis, we found that cluster 2 enriched more cardiovascular risk pathways than cluster 1. The immune cell infiltration analysis of ischemic cardiomyopathy (ICM; an advanced stage of CAD) samples revealed that the proportion of macrophage M2 was upregulated in the LINC01480 highly expressed samples, thus suggesting that LINC01480 plays a protective role in the progression of ICM. Based on the findings of this study, lncRNA LINC01480 may be used as a novel biomarker and therapeutic target for CAD.
Collapse
Affiliation(s)
- Ting Xiong
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
- Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, South China University of Technology, Guangzhou, China
| | - Botao Xiao
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Yueheng Wu
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong General Hospital, Guangzhou, China
| | - Yunfeng Liu
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Quhuan Li
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
- Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, South China University of Technology, Guangzhou, China
- *Correspondence: Quhuan Li,
| |
Collapse
|
12
|
Bian W, Jiang XX, Wang Z, Zhu YR, Zhang H, Li X, Liu Z, Xiong J, Zhang DM. Comprehensive analysis of the ceRNA network in coronary artery disease. Sci Rep 2021; 11:24279. [PMID: 34930980 PMCID: PMC8688464 DOI: 10.1038/s41598-021-03688-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 12/08/2021] [Indexed: 12/14/2022] Open
Abstract
With the rapid aging of the population, coronary artery disease (CAD) has become one of the most fatal chronic diseases. However, the genetic mechanism of CAD is still unclear. The purpose of this study is to construct the lncRNA-miRNA-mRNA regulatory network for CAD diseases and systematically identify differentially expressed genes in patients with coronary heart disease. In this study, two lncRNA datasets (GSE69587 and GSE113079) and a microRNA dataset (GSE105449) which contained 393 and 38 CAD samples were selected. In addition, two mRNA datasets which named GSE113079 (98 CAD samples) and GSE9820 (8 CAD samples) were selected to search the differentially expressed genes (DEGs). By comparing the expression data between CAD and control samples, a total of 1111 lncRNAs, 2595 mRNAs and 22 miRNAs were identified. Based on the DEGs, a lncRNA-miRNA-mRNA ceRNA network was constructed to explore the hub nodes in CAD. In the ceRNA network, the lncRNAs KCNQ1OT1 and H19 showed high connectivity with the nine miRNAs. GO and KEGG results showed that genes in ceRNA networks were mainly involved in nitrogen compound metabolic process, PI3K-Akt signaling pathway and retrograde endocannabinoid signaling. These findings will improve the understanding of the occurrence and development mechanism of CAD.
Collapse
Affiliation(s)
- Weikang Bian
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Xiao-Xin Jiang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Zhicheng Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Yan-Rong Zhu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Hongsong Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Xiaobo Li
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Zhizhong Liu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Jing Xiong
- Department of Pharmacology, China Pharmaceutical University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Dai-Min Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China.
| |
Collapse
|
13
|
Xu B, Xu Z, Chen Y, Lu N, Shu Z, Tan X. Genetic and epigenetic associations of ANRIL with coronary artery disease and risk factors. BMC Med Genomics 2021; 14:240. [PMID: 34615528 PMCID: PMC8496081 DOI: 10.1186/s12920-021-01094-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 09/30/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Both DNA genotype and methylation of antisense non-coding RNA in the INK4 locus (ANRIL) have been robustly associated with coronary artery disease (CAD), but the interdependent mechanisms of genotype and methylation remain unclear. METHODS Eighteen tag single nucleotide polymorphisms (SNPs) of ANRIL were genotyped in a matched case-control study (cases 503 and controls 503). DNA methylation of ANRIL and the INK4/ARF locus (p14ARF, p15INK4b and p16INK4a) was measured using pyrosequencing in the same set of samples (cases 100 and controls 100). RESULTS Polymorphisms of ANRIL (rs1004638, rs1333048 and rs1333050) were significantly associated with CAD (p < 0.05). The incidence of CAD, multi-vessel disease, and modified Gensini scores demonstrated a strong, direct association with ANRIL gene dosage (p < 0.05). There was no significant association between ANRIL polymorphisms and myocardial infarction/acute coronary syndrome (MI/ACS) (p > 0.05). Methylation levels of ANRIL were similar between the two studied groups (p > 0.05), but were different in the rs1004638 genotype, with AA and AT genotype having a higher level of ANRIL methylation (pos4, p = 0.006; pos8, p = 0.019). Further Spearman analyses indicated that methylation levels of ANRIL were positively associated with systolic blood pressure (pos6, r = 0.248, p = 0.013), diastolic blood pressure (pos3, r = 0.213, p = 0.034; pos6, r = 0.220, p = 0.028), and triglyceride (pos4, r = 0.253, p = 0.013), and negatively associated with high-density lipoprotein cholesterol (pos2, r = - 0.243, p = 0.017). Additionally, we identified 12 transcription factor binding sites (TFBS) within the methylated ANRIL region, and functional annotation indicated these TFBS were associated with basal transcription. Methylation at the INK4/ARF locus was not associated with ANRIL genotype. CONCLUSIONS These results indicate that ANRIL genotype (tag SNPs rs1004638, rs1333048 and rs1333050) mainly affects coronary atherosclerosis, but not MI/ACS. There may be allele-related DNA methylation and allele-related binding of transcription factors within the ANRIL promoter.
Collapse
Affiliation(s)
- Bayi Xu
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Zhixia Xu
- Department of Medical Service, Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Yequn Chen
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Nan Lu
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Zhouwu Shu
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xuerui Tan
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China.
| |
Collapse
|
14
|
Copur S, Rossing P, Afsar B, Sag AA, Siriopol D, Kuwabara M, Ortiz A, Kanbay M. A primer on metabolic memory: why existing diabesity treatments fail. Clin Kidney J 2021; 14:756-767. [PMID: 34512957 PMCID: PMC8422888 DOI: 10.1093/ckj/sfaa143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Indexed: 11/28/2022] Open
Abstract
Despite massive government and private sector investments into prevention of cardiovascular disease, diabetes mellitus and obesity, efforts have largely failed, and the burden of cost remains in the treatment of downstream morbidity and mortality, with overall stagnating outcomes. A new paradigm shift in the approach to these patients may explain why existing treatment strategies fail, and offer new treatment targets. This review aims to provide a clinician-centred primer on metabolic memory, defined as the sum of irreversible genetic, epigenetic, cellular and tissue-level alterations that occur with long-time exposure to metabolic derangements.
Collapse
Affiliation(s)
- Sidar Copur
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - Baris Afsar
- Department of Internal Medicine, Division of Nephrology, Suleyman Demirel University School of Medicine, Isparta, Turkey
| | - Alan A Sag
- Department of Radiology, Division of Vascular and Interventional Radiology, Duke University Medical Center, Durham, NC, USA
| | - Dimitrie Siriopol
- Nephrology Clinic, Dialysis and Renal Transplant Center, 'C.I. PARHON' University Hospital, 'Grigore T. Popa' University of Medicine, Iasi, Romania
| | | | - Alberto Ortiz
- School of Medicine, Dialysis Unit, IIS-Fundacion Jimenez Diaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Mehmet Kanbay
- Department of Medicine, Division of Nephrology, Koc University School of Medicine, Istanbul, Turkey
| |
Collapse
|
15
|
Sharma AR, Shashikiran U, Uk AR, Shetty R, Satyamoorthy K, Rai PS. Aberrant DNA methylation and miRNAs in coronary artery diseases and stroke: a systematic review. Brief Funct Genomics 2021; 19:259-285. [PMID: 31950130 DOI: 10.1093/bfgp/elz043] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/31/2019] [Accepted: 12/12/2019] [Indexed: 01/01/2023] Open
Abstract
Coronary artery disease (CAD) and ischemic stroke are the two most predominant forms of cardiovascular diseases (CVDs) caused by genetic, epigenetic and environmental risk factors. Although studies on the impact of 'epigenetics' in CVDs is not new, its effects are increasingly being realized as a key regulatory determinant that may drive predisposition, pathophysiology and therapeutic outcome. The most widely studied epigenetic risk factors are regulated by DNA methylation and miRNA expression. To keep pace with growing developments and discoveries, a comprehensive review was performed using Pubmed, Science Direct and Scopus databases to highlight the role of DNA methylation and miRNAs in CAD and stroke subjects. Network analysis was performed using ClueGO software and miRTargetLink database. We identified 32 studies of DNA methylation on CAD and stroke, of which, 6 studies showed differences in global DNA methylation, 10 studies reported the genome-wide difference in DNA methylation and 16 studies demonstrated altered DNA methylation at 14 candidate loci. The network analysis showed positive regulation of nitric oxide biosynthetic process, homocysteine metabolic process and negative regulation of lipid storage. About, 155 miRNAs were associated with CAD, stroke and related phenotypes in 83 studies. Interestingly, mir-223 hypomethylation and altered expression were associated with cerebral infarction and stroke. The target prediction for 18 common miRNAs between CAD and stroke showed strong interaction with SP3 and SP1 genes. This systematic review addresses the present knowledge on DNA methylation and miRNAs in CAD and stroke, whose abnormal regulation has been implicated in etiology or progression of the diseases.
Collapse
|
16
|
Ghafouri-Fard S, Gholipour M, Taheri M. The Emerging Role of Long Non-coding RNAs and Circular RNAs in Coronary Artery Disease. Front Cardiovasc Med 2021; 8:632393. [PMID: 33708807 PMCID: PMC7940190 DOI: 10.3389/fcvm.2021.632393] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/15/2021] [Indexed: 12/21/2022] Open
Abstract
Coronary artery disease (CAD) is a common disorder caused by atherosclerotic processes in the coronary arteries. This condition results from abnormal interactions between numerous cell types in the artery walls. The main participating factors in this process are accumulation of lipid deposits, endothelial cell dysfunction, macrophage induction, and changes in smooth muscle cells. Several lines of evidence underscore participation of long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) in the pathogenesis of CAD. Several lncRNAs such as H19, ANRIL, MIAT, lnc-DC, IFNG-AS1, and LEF1-AS1 have been shown to be up-regulated in the biological materials obtained from CAD patients. On the other hand, Gas5, Chast, HULC, DICER1-AS1, and MEG3 have been down-regulated in CAD patients. Meanwhile, a number of circRNAs have been demonstrated to influence function of endothelial cells or vascular smooth muscle cells, thus contributing to the pathogenesis of CAD. In the current review, we summarize the function of lncRNAs and circRNAs in the development and progression of CAD.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholipour
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Association between genetic variants at chromosome 9p21 and risk of coronary artery disease in Emirati Type 2 Diabetes patients. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
18
|
Wang Z, Li C, Sun X, Li Z, Li J, Wang L, Sun Y. Hypermethylation of miR-181b in monocytes is associated with coronary artery disease and promotes M1 polarized phenotype via PIAS1-KLF4 axis. Cardiovasc Diagn Ther 2020; 10:738-751. [PMID: 32968630 DOI: 10.21037/cdt-20-407] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Dysregulated microRNAs are involved in the macrophage polarization and atherosclerotic development. Apart from microRNAs, alteration in DNA methylation is considered as one of the most frequent epigenetic changes. The purpose of the research is to investigate the altered methylation status of miR-181b in the circulating monocytes from patients with coronary artery disease (CAD) and explore the underlying mechanisms. Methods We examined the methylation status of miR-181b in purified circulating monocytes from patients with CAD and healthy controls. We then transfected monocytes with miR-181b mimics and determined the role of miR-181b on the phenotypic switch of macrophages and inflammatory response. DNA methylation levels determined by MethyLight PCR and pyrosequencing at the promoter of miR-181b significantly increased in CAD patients. Based on TargetScan database, we identified PIAS1 as the target gene of miR-181b and explored the interaction of miR-181b and PIAS1 by Dual-Luciferase assay, quantitative PCR and immunoblots. We also investigated the role of miR-181b and PIAS1 on macrophage polarization and inflammation. Results Hypermethylation at the promoter of miR-181b directly contributed to the decrease of miR-181b activity and expression. Overexpression of miR-181b reduced M1 polarization and facilitated M2 polarization determined by quantitative PCR. While knockdown of PIAS1 induced KLF4 degradation and SUMOylation in monocytes, miR-181b mimics reverse the KLF4 SUMOylation via suppression of PIAS1. Moreover, KLF4 SUMOylation by PIAS1 reversed M1 polarization induced by depletion of miR-181b in monocytes. Conclusions Hypermethylation of miR-181b induces M1 polarization and promotes atherosclerosis through activation of PIAS1 and KLF4 SUMOylation in macrophages.
Collapse
Affiliation(s)
- Zhonghua Wang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chunlei Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinyong Sun
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhuqin Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jia Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lanfeng Wang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanming Sun
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
19
|
Probing the epigenetic signatures in subjects with coronary artery disease. Mol Biol Rep 2020; 47:6693-6703. [PMID: 32803503 DOI: 10.1007/s11033-020-05723-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/02/2020] [Indexed: 12/16/2022]
Abstract
Depletion of S-adenosyl methionine and 5-methyltetrahydrofolate; and elevation of total plasma homocysteine were documented in CAD patients, which might modulate the gene-specific methylation status and alter their expression. In this study, we have aimed to delineate CAD-specific epigenetic signatures by investigating the methylation and expression of 11 candidate genes i.e. ABCG1, LIPC, PLTP, IL-6, TNF-α, CDKN2A, CDKN2B, F2RL3, FGF2, P66 and TGFBR3. The methylation-specific PCR and qRT-PCR were used to assess the methylation status and the expression of candidate genes, respectively. CAD patients showed the upregulation of IL-6, TNF-α, CDKN2A, CDKN2B, F2RL3, FGF2, P66, and TGFBR3. Hypomethylation of CDKN2A loci was shown to increase risk for CAD by 1.79-folds (95% CI 1.22-2.63). Classification and regression tree (CART) model of gene expression showed increased risk for CAD with F2RL3 > 3.4-fold, while demonstrating risk reduction with F2RL3 < 3.4-fold and IL-6 < 7.7-folds. This CAD prediction model showed the excellent sensitivity (0.98, 95% CI 0.88-1.00), specificity (0.91, 95% CI 0.86-0.92), positive predictive value (0.82, 95% CI 0.75-0.84), and negative predictive value (0.99, 95% CI 0.94-1.00) with an overall accuracy of 92.8% (95% CI 87.0-94.1%). Folate and B12 deficiencies were observed in CAD cases, which were shown to contribute to hypomethylation and upregulation of the prime candidate genes i.e. CDKN2A and F2RL3. Early onset diabetes was associated with IL-6 and TNF-α hypomethylation and upregulation of CDKN2A. The expression of F2RL3 and IL-6 (or) hypomethylation status at CDKN2A locus are potential biomarkers in CAD risk prediction. Early epigenetic imprints of CAD were observed in early onset diabetes. Folate and B12 deficiencies are the contributing factors to these changes in CAD-specific epigenetic signatures.
Collapse
|
20
|
Yu Y, Chen Q, Zhang X, Yang J, Lin K, Ji C, Xu A, Yang L, Miao L. Long noncoding RNA ANRIL promotes the malignant progression of cholangiocarcinoma by epigenetically repressing ERRFI1 expression. Cancer Sci 2020; 111:2297-2309. [PMID: 32378752 PMCID: PMC7385372 DOI: 10.1111/cas.14447] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 04/28/2020] [Accepted: 04/30/2020] [Indexed: 12/11/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) have recently been verified to have significant regulatory functions in many types of human cancers. The lncRNA ANRIL is transcribed from the INK4b-ARF-INK4a gene cluster in the opposite direction. Whether ANRIL can act as an oncogenic molecule in cholangiocarcinoma (CCA) remains unknown. Our data show that ANRIL knockdown greatly inhibited CCA cell proliferation and migration in vitro and in vivo. According to the results of RNA sequencing analysis, ANRIL knockdown dramatically altered target genes associated with the cell cycle, cell proliferation, and apoptosis. By binding to a component of the epigenetic modification complex enhancer of zeste homolog 2 (EZH2), ANRIL could maintain lysine residue 27 of histone 3 (H3K27me3) levels in the promoter of ERBB receptor feedback inhibitor 1 (ERRFI1), which is a tumor suppressor gene in CCA. In this way, ERRFI1 expression was suppressed in CCA cells. These data verified the key role of the epigenetic regulation of ANRIL in CCA oncogenesis and indicate its potential as a target for CCA intervention.
Collapse
Affiliation(s)
- Yang Yu
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Qiaoyu Chen
- Department of Pathology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, China
| | - Xunlei Zhang
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Jian Yang
- Department of Urology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kaibo Lin
- Department of Assisted Reproduction, Shanghai Ninth Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Congfei Ji
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Aibing Xu
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Lei Yang
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Lin Miao
- Medical Centre for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
21
|
Xu LB, Zhang YQ, Zhang NN, Li B, Weng JY, Li XY, Lu WC, Yu PR, Wang X, Li Y, Han Z, Chen L, He HT, Zhou YF, Ma XX, Xu GD. Rs10757274 gene polymorphisms in coronary artery disease: A systematic review and a meta-analysis. Medicine (Baltimore) 2020; 99:e18841. [PMID: 32011499 PMCID: PMC7220330 DOI: 10.1097/md.0000000000018841] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND It has been reported the rs10757274 SNP (present on locus 9p21 in the gene for CDKN2BAS1) might be associated with susceptibility to coronary artery disease (CAD). Owing to mixed and inconclusive results, we conducted a meta-analysis to investigate the association between rs10757274 polymorphism and the risk of CAD. OBJECTIVES The present study aimed to investigate the relationship between rs10757274 polymorphism and the risk of CAD. METHODS All studies of the rs10757274 SNP with CAD that were published between 2007 and 2018 were retrieved from the PubMed database. Meta-analysis was performed with Stata 14.0 software. The effect size of the rs10757274 SNP with CAD risk was assessed based on the odds ratios (ORs) with calculation of 95% confidence interval (CI). RESULTS Eleven studies including 52,209 subjects (cases: 7990, controls: 44,219) were included in the final data combination. Pooled overall analyses showed that rs10757274 (allele model: P < .001; dominant model: P < .001; recessive model: P < .001; Heterozygote codominant: P = .002; Homozygote codominant: P < .001) polymorphisms were significantly associated with the likelihood of CAD. Significant heterogeneity between individual studies appears in all 5 models. Further subgroup analyses revealed that rs10757274 polymorphisms were all significantly correlated with the likelihood of CAD and no heterogeneity were observed in West Asians. CONCLUSIONS Our findings indicated that rs10757274 polymorphisms may serve as genetic biomarkers of CAD, especially in West Asians.
Collapse
Affiliation(s)
- Lang-Biao Xu
- Department of Cardiology, Suzhou Municipal Hospital, Nanjing Medical University
| | - Yi-Qing Zhang
- Department of Cardiology, Suzhou Municipal Hospital, Nanjing Medical University
| | - Nan-Nan Zhang
- Department of Cardiology, Suzhou Municipal Hospital, Nanjing Medical University
| | - Biao Li
- Department of Cardiology, Suzhou Municipal Hospital, Nanjing Medical University
| | - Jia-Yi Weng
- Department of Cardiology, Suzhou Municipal Hospital, Nanjing Medical University
| | - Xiao-Yang Li
- Department of Cardiology, Suzhou Municipal Hospital, Nanjing Medical University
| | - Wen-Chao Lu
- Department of Cardiology, Suzhou Municipal Hospital, Nanjing Medical University
| | - Pei-Ran Yu
- Department of Cardiology, Suzhou Municipal Hospital, Nanjing Medical University
| | - Xi Wang
- Department of Cardiology, Suzhou Municipal Hospital, Nanjing Medical University
| | - Yuan Li
- Department of Cardiology, Suzhou Municipal Hospital, Nanjing Medical University
| | - Zhen Han
- Department of Cardiology, Suzhou Municipal Hospital, Nanjing Medical University
| | - Lu Chen
- Department of Cardiology, Suzhou Municipal Hospital, Nanjing Medical University
| | - Hong-Tao He
- Department of Cardiology, Suzhou Municipal Hospital, Nanjing Medical University
| | - Ya-Feng Zhou
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou City 215006, Jiang su Province, PR China
| | - Xue-Xing Ma
- Department of Cardiology, Suzhou Municipal Hospital, Nanjing Medical University
| | - Gui-Dong Xu
- Department of Cardiology, Suzhou Municipal Hospital, Nanjing Medical University
| |
Collapse
|
22
|
Chen L, Qu H, Guo M, Zhang Y, Cui Y, Yang Q, Bai R, Shi D. ANRIL and atherosclerosis. J Clin Pharm Ther 2019; 45:240-248. [PMID: 31703157 DOI: 10.1111/jcpt.13060] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/26/2019] [Accepted: 09/09/2019] [Indexed: 12/15/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE The 3.8-kb-long antisense non-coding RNA at the INK4 locus (ANRIL) is transcribed from the short arm of human chromosome 9 on P21 and is associated with malfunction of the vascular endothelium, vascular smooth muscle cell (VSMC) proliferation/migration/senescence/apoptosis, mononuclear cell adhesion and proliferation, glycolipid metabolism disorder and DNA damage. Hence, ANRIL plays an important role in atherogenesis. Moreover, genome-wide association studies (GWAS) have identified ANRIL as a biomarker that is closely related to coronary heart disease (CHD). The objective of this review was to discuss the pathological mechanism of ANRIL in atherosclerotic development and its significance as a predictor of cardiovascular disease. METHODS Review of the PubMed, EMBASE and Cochrane databases for articles demonstrating the roles of ANRIL in the development of atherosclerotic diseases. RESULTS AND DISCUSSION The abnormal expression of ANRIL is linked to vascular endothelium injury; the proliferation, migration, senescence and apoptosis of VSMCs; mononuclear cell adhesion and proliferation; glycolipid metabolism disorder; DNA damage; and competing endogenous RNAs. Moreover, ANRIL accelerates the progression of CHD by regulating its single nucleotide polymorphisms (SNPs). WHAT IS NEW AND CONCLUSION Considering that ANRIL accelerates atherosclerosis (AS) development and is a risk factor for CHD, it is reasonable for us to explore an efficacious ANRIL-based therapy for AS in CHD.
Collapse
Affiliation(s)
- Li Chen
- Peking University Traditional Chinese Medicine Clinical Medical School (Xi yuan), Beijing, China
| | - Hua Qu
- Graduate School of China Academy of Chinese Medical Sciences, Beijing, China
| | - Ming Guo
- Cardiovascular Diseases Center, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Zhang
- Cardiovascular Diseases Center, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuanyuan Cui
- Cardiovascular Diseases Center, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiaoning Yang
- Cardiovascular Diseases Center, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Ruina Bai
- Cardiovascular Diseases Center, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Dazhuo Shi
- Cardiovascular Diseases Center, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
23
|
Zhao CH, Cao HT, Zhang J, Jia QW, An FH, Chen ZH, Li LH, Wang LS, Ma WZ, Yang ZJ, Jia EZ. DNA methylation of antisense noncoding RNA in the INK locus (ANRIL) is associated with coronary artery disease in a Chinese population. Sci Rep 2019; 9:15340. [PMID: 31653960 PMCID: PMC6814794 DOI: 10.1038/s41598-019-51921-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 10/10/2019] [Indexed: 12/11/2022] Open
Abstract
To explore the association between methylation of antisense non-coding RNA in the INK4 locus (ANRIL) and coronary artery disease (CAD) development. Methylation levels of ANRIL in 100 subjects with CAD and 100 controls were quantitatively analyzed using Sequenom MassARRAY. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis was used to identify novel pathways. Our analyses indicated that 7 to 8 CpG sites within the 2nd CpG island located upstream of ANRIL, also known as cyclin-dependent kinase inhibitor 2B - antisense 1 (CDKN2B-AS1), are hyper-methylated in CAD subjects compared to controls (p = 0.034). The 40th CpG site within the 2nd CpG island located upstream of CDKN2B-AS1 was methylated to a lesser extent in CAD subjects compared to controls (p = 0.045). Both Pearson and Spearman analyses indicated that methylation levels were significantly associated with total cholesterol (r = 0.204, p = 0.004), fasting high-density lipoprotein cholesterol (r = 0.165, p = 0.020), and fasting low-density lipoprotein cholesterol (r = 0.265, p = 0.000). KEGG pathway analysis revealed a significant enrichment of genes associated with the tumor necrosis factor (TNF) signaling pathway. Among them, CCAAT/enhancer binding protein (C/EBPβ) was identified as a key transcription factor that promotes expression of CDKN2B-AS1 through promotor interaction. DNA methylation of the ANRIL promoter was significantly associated with CAD development in our study. Our analyses suggest that C/EBPβ is a key transcription factor that promotes CDKN2B-AS1 expression by directly interacting with the gene promotor mediated by TNF signaling.
Collapse
Affiliation(s)
- Chen-Hui Zhao
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Hai-Tao Cao
- Department of Cardiovascular Medicine, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu Province, China
| | - Jing Zhang
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Qiao-Wei Jia
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Feng-Hui An
- Department of Cardiovascular Medicine, the Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yining, 835000, Xinjiang, China
| | - Zhao-Hong Chen
- Department of Cardiovascular Medicine, the Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yining, 835000, Xinjiang, China
| | - Li-Hua Li
- Department of Cardiovascular Medicine, the Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yining, 835000, Xinjiang, China
| | - Lian-Sheng Wang
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Wen-Zhu Ma
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Zhi-Jian Yang
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - En-Zhi Jia
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
24
|
Li C, Zong W, Zhang M, Tu Y, Zhou Q, Ni M, Li Z, Liu H, Zhang J. Increased Ratio of Circulating T-Helper 1 to T-Helper 2 Cells and Severity of Coronary Artery Disease in Patients with Acute Myocardial Infarction: A Prospective Observational Study. Med Sci Monit 2019; 25:6034-6042. [PMID: 31407674 PMCID: PMC6703085 DOI: 10.12659/msm.913891] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background This study aimed to determine the association between CD4-positive T-helper (Th) cell subsets, T-helper 1 (Th1) and T-helper 2 (Th2) in patients with acute myocardial infarction (AMI) and the severity of coronary artery disease (CAD) determined by coronary artery angiography. Material/Methods Three groups of patients with AMI who underwent coronary angiography and percutaneous coronary intervention (PCI) included patients with stable CAD (n=35), ST-segment elevation myocardial infarction (STEMI) (n=30), and non-STEMI (NSTEMI) (n=35), and controls (n=33). Measurement of high-sensitivity cardiac troponin T (hs-cTnT) was performed. The numbers of circulating CD4-positive Th1 and Th2 cells were measured using flow cytometry. Plasma levels of interferon-γ (IFN-γ) and interleukin-4 (IL-4) were measured using enzyme-linked immunosorbent assay (ELISA). Results An increase in the Th1 lymphocyte population was associated with more CAD, and an increased Th1/Th2 ratio was found in patients with NSTEMI and STEMI (controls 7.27±2.98; stable CAD 7.58±2.52; NSTEMI 16.62±2.74; and STEMI 22.32±7.35) (P<0.001). The proportion of Th1 cells and the Th1/Th2 ratio increased as the number of affected arteries, the degree of stenosis, and the lesion length increased. At a median follow-up of 18.2 months, patients with CAD and an increased Th1/Th2 ratio had a significant increase in adverse cardiac events compared with patients with a reduced Th1/Th2 ratio (log-rank, P=0.042). Conclusions An increased ratio of circulating Th1 to Th2 cells in patients with AMI was associated with the severity of CAD determined by angiography.
Collapse
Affiliation(s)
- Chang Li
- Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei, China (mainland)
| | - Wenxia Zong
- Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei, China (mainland)
| | - Ming Zhang
- Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei, China (mainland)
| | - Yanming Tu
- Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei, China (mainland)
| | - Qiyu Zhou
- Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei, China (mainland)
| | - Mingke Ni
- Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei, China (mainland)
| | - Zhiyong Li
- Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei, China (mainland)
| | - Haizhen Liu
- Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei, China (mainland)
| | - Jingyi Zhang
- Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei, China (mainland)
| |
Collapse
|
25
|
Hu Y, Hu J. Diagnostic value of circulating lncRNA ANRIL and its correlation with coronary artery disease parameters. ACTA ACUST UNITED AC 2019; 52:e8309. [PMID: 31411246 PMCID: PMC6694403 DOI: 10.1590/1414-431x20198309] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/24/2019] [Indexed: 02/06/2023]
Abstract
This study aimed to detect the expression of the long non-coding RNA (lncRNA) antisense non-coding RNA in the INK4 locus (ANRIL) and evaluate its correlation with disease risk, stenosis degree, inflammation, as well as overall survival (OS) in coronary artery disease (CAD) patients. A total of 230 patients who underwent diagnostic coronary angiography were consecutively recruited and assigned to CAD group (n=125) or control group (n=105) according to presence or absence of CAD. Gensini score was calculated to assess the severity of coronary artery damage. Plasma samples were collected and the expression ANRIL was detected in all participants. High-sensitivity C-reactive protein (hs-CRP), erythrocyte sedimentation rate (ESR), and cytokines including tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6, IL-8, IL-10, and IL-17 in CAD patients were measured and OS was calculated. The relative expression of ANRIL was higher in CAD patients compared to controls (P<0.001). Receiver operating characteristic disclosed that ANRIL could distinguish CAD patients from controls with an area under the curve of 0.789 (95%CI: 0.731–0.847). Spearman's rank correlation test revealed that expression of ANRIL was positively correlated with Gensini score (P=0.001), levels of hs-CRP (P=0.001), ESR (P=0.038), TNF-α (P=0.004), and IL-6 (P<0.001), while negatively correlated with IL-10 level (P=0.008) in CAD patients. Kaplan-Meier curve revealed that high expression of ANRIL was associated with shorter OS (P=0.013). In conclusion, circulating ANRIL presented a good diagnostic value for CAD, and its high expression was associated with increased stenosis degree, raised inflammation, and poor OS in CAD patients.
Collapse
Affiliation(s)
- Yao Hu
- Department of Cardiovascular Medicine, Jiangxi Provincial People's Hospital, Affiliated to Nanchang University, Nanchang, China
| | - Jing Hu
- Department of Cardiovascular Medicine, The First Hospital of Nanchang, Affiliated to Nanchang University, Nanchang, China
| |
Collapse
|
26
|
Chen X, Yu X, Shen E. Overexpression of CDKN2B is involved in poor gastric cancer prognosis. J Cell Biochem 2019; 120:19825-19831. [PMID: 31297846 DOI: 10.1002/jcb.29287] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 02/28/2019] [Indexed: 12/13/2022]
Abstract
The objective of this investigation is to elucidate the clinical significance of cyclin-dependent kinase inhibitor 2B (CDKN2B) expression regarding gastric cancer (GC), as well as to detect the involvement of CDKN2B expression in the clinicopathological indexes and prognosis of GC. Immunohistochemical analysis was used for identification of CDKN2B expression in GC specimens. Chi-square (χ2 ) test was applied to detect the association of CDKN2B expression and clinicopathological parameters of GC. The involvement of CDKN2B expression in the prognosis was analyzed via univariate and multivariate analysis. It was indicated that relative to the corresponding para-carcinoma tissues, CDKN2B expression was notably upregulated in GC specimens. Moreover, the expression of CDKN2B was strongly correlated with the differentiation (r = -0.182; P = .015), invasion (r = -0.157; P = .038), distant metastases (r = -0.196; P = .004), and TNM stage (r = -0.204; P = .005). Nevertheless, no remarkable variance was related to age, tumor loci, or sex. Kaplan-Meier survival curve and univariate analysis showed that CDKN2B overexpression predicted poorer disease-free survival (P = .007) and overall survival (P = .005) in those with GC. In addition, Cox proportional hazards regression model revealed that CDKN2B was an isolated biomarker of disease-free survival and overall survival in patients with GC. Taken together, our data demonstrated that the overexpression of CDKN2B could be an isolated factor for GC prognostic in patients. CDKN2B gene may be a useful target and new treatment for improving the prognosis of GC.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pathology, Taizhou Municipal Hospital, Taizhou, Zhejiang, China
| | - Xingtong Yu
- Department of Pathology, Taizhou Municipal Hospital, Taizhou, Zhejiang, China
| | - Enjian Shen
- Department of Pathology, Taizhou Municipal Hospital, Taizhou, Zhejiang, China
| |
Collapse
|
27
|
Guo Z, Li L, Gao Y, Zhang X, Cheng M. RETRACTED ARTICLE: Overexpression of lncRNA ANRIL aggravated hydrogen peroxide-disposed injury in PC-12 cells via inhibiting miR-499a/PDCD4 axis-mediated PI3K/Akt/mTOR/p70S6K pathway. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2624-2633. [DOI: 10.1080/21691401.2019.1629953] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Zhiliang Guo
- Department of Orthopedic, The 89 Hospital of Chinese PLA, Weifang, China
| | - Lanlan Li
- Clinic Medical College, Weifang Medical University, Weifang, China
| | - Yu Gao
- Clinic Medical College, Weifang Medical University, Weifang, China
| | - Xiaoyun Zhang
- Clinic Medical College, Weifang Medical University, Weifang, China
| | - Min Cheng
- Clinic Medical College, Weifang Medical University, Weifang, China
| |
Collapse
|
28
|
Sun QM, Hu B, Fu PY, Tang WG, Zhang X, Zhan H, Sun C, He YF, Song K, Xiao YS, Sun J, Xu Y, Zhou J, Fan J. Long non-coding RNA 00607 as a tumor suppressor by modulating NF-κB p65/p53 signaling axis in hepatocellular carcinoma. Carcinogenesis 2019; 39:1438-1446. [PMID: 30169594 DOI: 10.1093/carcin/bgy113] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 08/24/2018] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence suggests that long non-coding RNA (lncRNA) plays important roles in some malignant tumors. However, the mechanism underlying how lncRNA regulates hepatocellular carcinoma (HCC) process remains largely unknown. In this study, we explored the potential role of lncRNA 00607 as a novel tumor suppressor in HCC. In this study, we examined the regulation of lncRNA 00607 by the important inflammatory cytokine tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). We also determined the expression of LINC000607 in 159 HCC tumors and paired adjacent tissues. Effects of LINC000607 in HCC proliferation and apoptosis were examined in vitro in HCC cell lines and in vivo tumor xenografts. Furthermore, we also examine underlying mechanism by which lncRNA 00607 regulates NF-κB p65 and how LIN00607 exerts its tumor suppressor role in HCC. We found that lncRNA 00607 expression level is lower in HCC tumors compared with matched normal liver tissue, and its low expression predicts worse prognosis in HCC. Functionally, lncRNA 00607 overexpression leads to decreased HCC cell proliferation in vitro and in vivo, enhanced apoptosis and chemotherapeutic drug sensitivity. Mechanistically, lncRNA 00607 inhibits the p65 transcription by binding to the p65 promoter region, therefore contributing to increased p53 levels in HCC. Taken together, the findings of this study show that the TNF-α/IL-6-lncRNA 00607-NF-κB p65/p53 signaling axis represents a novel therapeutic avenue in cancer chemotherapy.
Collapse
Affiliation(s)
- Qi-Man Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Zhongshan Hospital of Fudan University, Shanghai, P.R. China
| | - Bo Hu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Zhongshan Hospital of Fudan University, Shanghai, P.R. China
| | - Pei-Yao Fu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Zhongshan Hospital of Fudan University, Shanghai, P.R. China
| | - Wei-Guo Tang
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, P. R. China
| | - Xin Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Zhongshan Hospital of Fudan University, Shanghai, P.R. China
| | - Hao Zhan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Zhongshan Hospital of Fudan University, Shanghai, P.R. China
| | - Chao Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Zhongshan Hospital of Fudan University, Shanghai, P.R. China
| | - Yi-Feng He
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Zhongshan Hospital of Fudan University, Shanghai, P.R. China
| | - Kang Song
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Zhongshan Hospital of Fudan University, Shanghai, P.R. China
| | - Yong-Sheng Xiao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Zhongshan Hospital of Fudan University, Shanghai, P.R. China
| | - Jian Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Zhongshan Hospital of Fudan University, Shanghai, P.R. China
| | - Yang Xu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Zhongshan Hospital of Fudan University, Shanghai, P.R. China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Zhongshan Hospital of Fudan University, Shanghai, P.R. China.,Institute of Biomedical Sciences, Fudan University, Shanghai, P.R. China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Zhongshan Hospital of Fudan University, Shanghai, P.R. China.,Institute of Biomedical Sciences, Fudan University, Shanghai, P.R. China
| |
Collapse
|
29
|
Li M, Duan L, Li Y, Liu B. Long noncoding RNA/circular noncoding RNA-miRNA-mRNA axes in cardiovascular diseases. Life Sci 2019; 233:116440. [PMID: 31047893 DOI: 10.1016/j.lfs.2019.04.066] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/23/2019] [Accepted: 04/29/2019] [Indexed: 02/01/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide. Non-coding RNAs including long non-coding RNAs (lncRNAs), circular RNAs (circRNAs) and microRNAs (miRNAs) have been reported to participate in pathological developments of CVDs through various mechanisms. Among them, the networks among lncRNAs/circRNAs, miRNAs, and mRNAs have recently attracted attention. Understanding the molecular mechanism could aid the discovery of therapeutic targets or strategies in CVDs including atherosclerosis, myocardial infarction (MI), hypertrophy, heart failure (HF) and cardiomyopathy. In this review, we summarize the latest research involving the lncRNA/circRNA-miRNA-mRNA axis in CVDs, with emphasis on the molecular mechanism.
Collapse
Affiliation(s)
- Ming Li
- Department of Gastroenterology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun 130041, China
| | - Liwei Duan
- Department of Gastroenterology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun 130041, China
| | - Yangxue Li
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun 130041, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun 130041, China.
| |
Collapse
|
30
|
Zhang Y, Zhang L, Wang Y, Ding H, Xue S, Qi H, Li P. MicroRNAs or Long Noncoding RNAs in Diagnosis and Prognosis of Coronary Artery Disease. Aging Dis 2019; 10:353-366. [PMID: 31011482 PMCID: PMC6457061 DOI: 10.14336/ad.2018.0617] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 06/17/2018] [Indexed: 12/14/2022] Open
Abstract
Coronary artery disease (CAD) is the result of atherosclerotic plaque development in the wall of the coronary arteries. The underlying mechanism involves atherosclerosis of the arteries of the heart which is a relatively complex process comprising several steps. In CAD, atherosclerosis induces functional and structural changes. The pathogenesis of CAD results from various changes in and interactions between multiple cell types in the artery walls; these changes mainly include endothelial cell (EC) dysfunction, vascular smooth muscle cell (SMC) alteration, lipid deposition and macrophage activation. Various blood markers associated with an increased risk for cardiovascular endpoints have been identified; however, few have yet been shown to have a diagnostic impact or important clinical implications that would affect patient management. Noncoding RNAs, especially microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), can be stable in plasma and other body fluids and could therefore serve as biomarkers for some diseases. Many studies have shown that some miRNAs and lncRNAs play key roles in heart and vascular development and in cardiac pathophysiology. Thus, we summarize here the latest research progress, focusing on the molecular mechanism of miRNAs and lncRNAs in CAD, with the intent of seeking new targets for the treatment of heart disease.
Collapse
Affiliation(s)
- Yuan Zhang
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Lei Zhang
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Yu Wang
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Han Ding
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Sheng Xue
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Hongzhao Qi
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Peifeng Li
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| |
Collapse
|
31
|
Langmia IM, Kräker K, Weiss SE, Haase N, Schütte T, Herse F, Dechend R. Cardiovascular Programming During and After Diabetic Pregnancy: Role of Placental Dysfunction and IUGR. Front Endocrinol (Lausanne) 2019; 10:215. [PMID: 31024453 PMCID: PMC6466995 DOI: 10.3389/fendo.2019.00215] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 03/18/2019] [Indexed: 12/31/2022] Open
Abstract
Intrauterine growth restriction (IUGR) is a condition whereby a fetus is unable to achieve its genetically determined potential size. IUGR is a global health challenge due to high mortality and morbidity amongst affected neonates. It is a multifactorial condition caused by maternal, fetal, placental, and genetic confounders. Babies born of diabetic pregnancies are usually large for gestational age but under certain conditions whereby prolonged uncontrolled hyperglycemia leads to placental dysfunction, the outcome of the pregnancy is an intrauterine growth restricted fetus with clinical features of malnutrition. Placental dysfunction leads to undernutrition and hypoxia, which triggers gene modification in the developing fetus due to fetal adaptation to adverse utero environmental conditions. Thus, in utero gene modification results in future cardiovascular programming in postnatal and adult life. Ongoing research aims to broaden our understanding of the molecular mechanisms and pathological pathways involved in fetal programming due to IUGR. There is a need for the development of effective preventive and therapeutic strategies for the management of growth-restricted infants. Information on the mechanisms involved with in utero epigenetic modification leading to development of cardiovascular disease in adult life will increase our understanding and allow the identification of susceptible individuals as well as the design of targeted prevention strategies. This article aims to systematically review the latest molecular mechanisms involved in the pathogenesis of IUGR in cardiovascular programming. Animal models of IUGR that used nutrient restriction and hypoxia to mimic the clinical conditions in humans of reduced flow of nutrients and oxygen to the fetus will be discussed in terms of cardiac remodeling and epigenetic programming of cardiovascular disease. Experimental evidence of long-term fetal programming due to IUGR will also be included.
Collapse
Affiliation(s)
- Immaculate M. Langmia
- Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrueck Center for Molecular Medicine and the Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Alexander von Humboldt Foundation, Bonn, Germany
| | - Kristin Kräker
- Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrueck Center for Molecular Medicine and the Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
- Max-Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Sara E. Weiss
- Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrueck Center for Molecular Medicine and the Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nadine Haase
- Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrueck Center for Molecular Medicine and the Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
- Max-Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Till Schütte
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Center for Cardiovascular Research, Institute of Pharmacology, Charité -Universitätsmedizin Berlin, Berlin, Germany
| | - Florian Herse
- Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrueck Center for Molecular Medicine and the Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Max-Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrueck Center for Molecular Medicine and the Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- HELIOS-Klinikum, Berlin, Germany
- *Correspondence: Ralf Dechend
| |
Collapse
|
32
|
Zhang L, Ji H, Huang Y, Hu H, Li B, Yang Y, Yu H, Chen X, Li W, Liu F, Wang S, Wang C, Chen K, Bao Y, Liu H, Duan S. Association of BAX hypermethylation with coronary heart disease is specific to individuals aged over 70. Medicine (Baltimore) 2019; 98:e14130. [PMID: 30681575 PMCID: PMC6358363 DOI: 10.1097/md.0000000000014130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION As a member of B-cell lymphoma-2 (BCL-2) gene family, BCL-2 associated X (BAX) is important for cell apoptosis. In this work, we investigated the association of BAX promoter DNA methylation with coronary heart disease (CHD) in Han Chinese. METHODS A SYBR green-based quantitative methylation specific PCR (qMSP) was used to test BAX methylation levels in 959 CHD cases and 514 controls. RESULTS Although BAX methylation was not associated with CHD in the total samples, further breakdown analysis by age showed that BAX hypermethylation was significantly associated with CHD for individuals aged over 70 (median percentage of methylation ratio [PMR], 10.70% in cases versus (vs) 2.25% in controls, P =.046). Moreover, BAX methylation was associated with smoking and lipoprotein A (Lp(a)) for individuals aged over 70 (CHD: smoking P = .012, Lp(a) P = .001; non-CHD: smoking P = .051, Lp(a) P = .004). Further analysis of Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) data showed BAX expression was upregulated by 5-aza-2'-deoxycytidine demethylation agent (fold = 1.66, P = .038) and inversely correlated with BAX methylation (r = -0.428, P = 8E-05). CONCLUSIONS Our study supported that BAX hypermethylation might contribute to CHD risk via downregulation of BAX expression for individuals aged over 70.
Collapse
Affiliation(s)
- Limei Zhang
- Department of Cardiology, Yinzhou People's Hospital, Ningbo University, Ningbo
| | - Huihui Ji
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo
| | - Yi Huang
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo
| | - Haochang Hu
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo
| | - Bin Li
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo
| | - Yong Yang
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo
| | - Hang Yu
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo
| | - Xiaoying Chen
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo
| | - Wenxia Li
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo
| | - Fang Liu
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo
| | - Shi Wang
- Department of Cardiology, Yinzhou People's Hospital, Ningbo University, Ningbo
| | - Chunming Wang
- Department of Cardiology, Yinzhou People's Hospital, Ningbo University, Ningbo
| | - Ke Chen
- Department of Cardiology, Yinzhou People's Hospital, Ningbo University, Ningbo
| | - Yingchun Bao
- Department of Cardiology, Yinzhou People's Hospital, Ningbo University, Ningbo
| | - Haibo Liu
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shiwei Duan
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo
| |
Collapse
|
33
|
Holdt LM, Teupser D. Long Noncoding RNA ANRIL: Lnc-ing Genetic Variation at the Chromosome 9p21 Locus to Molecular Mechanisms of Atherosclerosis. Front Cardiovasc Med 2018; 5:145. [PMID: 30460243 PMCID: PMC6232298 DOI: 10.3389/fcvm.2018.00145] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 10/01/2018] [Indexed: 12/24/2022] Open
Abstract
Ever since the first genome-wide association studies (GWAS) on coronary artery disease (CAD), the Chr9p21 risk locus has emerged as a top signal in GWAS of atherosclerotic cardiovascular disease, including stroke and peripheral artery disease. The CAD risk SNPs on Chr9p21 lie within a stretch of 58 kilobases of non-protein-coding DNA, containing the gene body of the long noncoding RNA (lncRNA) antisense non coding RNA in the INK4 locus (ANRIL). How risk is affected by the Chr9p21 locus in molecular detail is a matter of ongoing research. Here we will review recent advances in the understanding that ANRIL serves as a key risk effector molecule of atherogenesis at the locus. One focus of this review is the shift in understanding that genetic variation at Chr9p21 not only affects the abundance of ANRIL, and in some cases expression of the adjacent CDKN2A/B tumor suppressors, but also impacts ANRIL splicing, such that 3′-5′-linked circular noncoding ANRIL RNA species are produced. We describe how the balance of linear and circular ANRIL RNA, determined by the Chr9p21 genotype, regulates molecular pathways and cellular functions involved in atherogenesis. We end with an outlook on how manipulating circular ANRIL abundance may be exploited for therapeutic purposes.
Collapse
Affiliation(s)
- Lesca M Holdt
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Daniel Teupser
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
34
|
Chen X, Jiang D, Xu L, Han L, Hu H, Huang Y, Lu D, Ji H, Li B, Yang Y, Zhou C, Xu X, Wu N, Xu X, Xu Y, Shen Y, Li J, Duan S. Elevated methylation of cyclin dependent kinase inhibitor 2B contributes to the risk of coronary heart disease in women. Exp Ther Med 2018; 17:205-213. [PMID: 30651784 PMCID: PMC6307461 DOI: 10.3892/etm.2018.6920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 01/18/2018] [Indexed: 12/12/2022] Open
Abstract
Cyclin dependent kinase inhibitor 2B (CDKN2B) encodes a cyclin-dependent kinase inhibitor that may enhance the formation of atherosclerotic plaques. The aim of the present study was to investigate the contribution of CDKN2B promoter methylation on the risk of coronary heart disease (CHD). The present results indicated a significant association between increased CDKN2B methylation and the risk of CHD (adjusted P=0.043). A breakdown analysis according to sex demonstrated that CDKN2B methylation was significantly associated with the risk of CHD in women (adjusted P=0.010), but not in men. A further breakdown analysis by age indicated a significant association of CHD in the women >60 years (P=0.024). Luciferase reporter gene assay results indicated that the CDKN2B promoter fragment significantly enhanced luciferase activity (P<0.001). In addition, CDKN2B transcription was significantly enhanced following treatment with 5-aza-2′-deoxycytidine methylation inhibitor in human aortic endothelial cells (HAEC) and human primary coronary artery smooth muscle cells (HPCASMC; P<0.05 and P<0.01), but not in 293 cells. Notably, estrogen treatment reduced CDKN2B methylation of several CpGs and significantly increased CDKN2B gene expression levels in HAEC, HPCASMC and 293 cells (P<0.05 and P<0.01). Additionally, treatment of HAEC and HPCASMC with simvastatin and γ-carboxy-L-glutamic acid reduced CDKN2B promoter methylation and increased CDKN2B transcription concomitantly. The present study suggests that CDKN2B promoter methylation may be associated with sex dimorphism in the pathogenesis of CHD.
Collapse
Affiliation(s)
- Xiaomin Chen
- Key Laboratory of Ningbo First Hospital and Cardiovascular Center of Ningbo First Hospital, Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Danjie Jiang
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Limin Xu
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Liyuan Han
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Haochang Hu
- Key Laboratory of Ningbo First Hospital and Cardiovascular Center of Ningbo First Hospital, Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Yi Huang
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Deyi Lu
- Richard and Loan Hill Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Huihui Ji
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Bin Li
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Yong Yang
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Cong Zhou
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xuting Xu
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Nan Wu
- Key Laboratory of Ningbo First Hospital and Cardiovascular Center of Ningbo First Hospital, Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Xiaofeng Xu
- Key Laboratory of Ningbo First Hospital and Cardiovascular Center of Ningbo First Hospital, Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Yan Xu
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Yusheng Shen
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jiyi Li
- Key Laboratory of Ningbo First Hospital and Cardiovascular Center of Ningbo First Hospital, Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Shiwei Duan
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
35
|
Wang Y, Song X, Li Z, Liu B. Long non-coding RNAs in coronary atherosclerosis. Life Sci 2018; 211:189-197. [PMID: 30195033 DOI: 10.1016/j.lfs.2018.08.072] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 12/26/2022]
Abstract
Coronary atherosclerosis (CAS), a leading cause of cardiovascular disease, is a major cause of death worldwide. CAS is a chronic disease in the aorta that can be caused by dyslipidemia, abnormal glucose metabolism, endothelial cell dysfunction, vascular smooth muscle cell (VSMC) or fibrous connective tissue hyperplasia, immune inflammatory reactions, and many other factors. The pathogenesis of CAS is not fully understood, as it is a complex lesion complicated by multiple factors. Damage-response theories have put forward endothelial cell (EC) injury as the initiating factor for CAS; the addition of lipid metabolism disorders may enhance monocyte adhesion, increase the proliferation and migration of fibroblasts and VSMCs, and accelerate the development of CAS. Furthermore, inflammatory and immune responses can create a vicious cycle of endothelial injury, which also plays key roles in the formation of CAS. Therefore, in order to elucidate the mechanisms controlling CAS, it is important to study the etiology of vascular cell dysfunction, abnormal energy and metabolism disorders, and immune and inflammatory reactions. Non-coding RNAs play regulatory roles in the pathogenesis of CAS, especially long non-coding RNAs (lncRNAs); lncRNAs have recently become a major focus for cardiovascular disease mechanisms, as they play numerous roles in the progression of CAS. Therefore, in this review, we discuss the role of lncRNAs in the pathogenesis of coronary CAS, and their role in the prevention and treatment of coronary CAS.
Collapse
Affiliation(s)
- Yiran Wang
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xianjing Song
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Zhibo Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
36
|
Kong Y, Hsieh CH, Alonso LC. ANRIL: A lncRNA at the CDKN2A/B Locus With Roles in Cancer and Metabolic Disease. Front Endocrinol (Lausanne) 2018; 9:405. [PMID: 30087655 PMCID: PMC6066557 DOI: 10.3389/fendo.2018.00405] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 06/29/2018] [Indexed: 12/12/2022] Open
Abstract
The CDKN2A/B genomic locus is associated with risk of human cancers and metabolic disease. Although the locus contains several important protein-coding genes, studies suggest disease roles for a lesser-known antisense lncRNA encoded at this locus, called ANRIL. ANRIL is a complex gene containing at least 21 exons in simians, with many reported linear and circular isoforms. Like other genes, abundance of ANRIL is regulated by epigenetics, classic transcription regulation, splicing, and post-transcriptional influences such as RNA stability and microRNAs. Known molecular functions of ANRIL include in cis and in trans gene regulation through chromatin modification complexes, and influence over microRNA signaling networks. Polymorphisms at the ANRIL gene are linked to risk for many different cancers, as well as risk of atherosclerotic cardiovascular disease, bone mass, obesity and type 2 diabetes. A broad array of variable reported impacts of polymorphisms on ANRIL abundance, splicing and function suggests that ANRIL has cell-type and context-dependent regulation and actions. In cancer cells, ANRIL gain of function increases proliferation, metastasis, cell survival and epithelial-mesenchymal transformation, whereas ANRIL loss of function decreases tumor size and growth, invasion and metastasis, and increases apoptosis and senescence. In metabolic disease, polymorphisms at the ANRIL gene are linked to risk of type 2 diabetes, coronary artery disease, coronary artery calcium score, myocardial infarction, and stroke. Intriguingly, with the exception of one polymorphism in exon 2 of ANRIL, the single nucleotide polymorphisms (SNPs) associated with atherosclerosis and diabetes are non-overlapping. Evidence suggests that ANRIL gain of function increases atherosclerosis; in diabetes, a risk-SNP reduced the pancreatic beta cell proliferation index. Studies are limited by the uncertain relevance of rodent models to ANRIL studies, since most ANRIL exons do not exist in mouse. Diverse cell-type-dependent results suggest it is necessary to perform studies in the relevant primary human tissue for each disease. Much remains to be learned about the biology of ANRIL in human health and disease; this research area may lead to insight into disease mechanisms and therapeutic approaches.
Collapse
Affiliation(s)
| | | | - Laura C. Alonso
- Department of Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
37
|
Genetic and Epigenetic Regulations of Post-prandial Lipemia. CURRENT GENETIC MEDICINE REPORTS 2018. [DOI: 10.1007/s40142-018-0146-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
38
|
An integrated transcriptomic and epigenomic analysis identifies CD44 gene as a potential biomarker for weight loss within an energy-restricted program. Eur J Nutr 2018; 58:1971-1980. [PMID: 29948221 DOI: 10.1007/s00394-018-1750-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 06/11/2018] [Indexed: 12/15/2022]
Abstract
PURPOSE The interindividual variable response to weight-loss treatments requires the search for new predictive biomarkers for improving the success of weight-loss programs. The aim of this study is to identify novel genes that distinguish individual responses to a weight-loss dietary treatment by using the integrative analysis of mRNA expression and DNA methylation arrays. METHODS Subjects from Metabolic Syndrome Reduction in Navarra (RESMENA) project were classified as low (LR) or high (HR) responders depending on their weight loss. Transcriptomic (n = 24) and epigenomic (n = 47) patterns were determined by array-based genome-wide technologies in human white blood cells at the baseline of the treatment period. CD44 expression was validated by qRT-PCR and methylation degree of CpGs of the gene was validated by MassARRAY® EpiTYPER™ in a subsample of 47 subjects. CD44 protein levels were measured by ELISA in human plasma. RESULTS Different expression and DNA methylation profiles were identified in LR in comparison to HR. The integrative analysis of both array data identified four genes: CD44, ITPR1, MTSS1 and FBXW5 that were differently methylated and expressed between groups. CD44 showed higher expression and lower DNA methylation levels in LR than in HR. Although differences in CD44 protein levels between LR and HR were not statistically significant, a positive association was observed between CD44 mRNA expression and protein levels. CONCLUSIONS In summary, the combination of a genome-wide methylation and expression array dataset can be a useful strategy to identify novel genes that might be considered as predictors of the dietary response. CD44 gene transcription and methylation may be a possible candidate biomarker for weight-loss prediction.
Collapse
|
39
|
Dual Effect of IL-6 -174 G/C Polymorphism and Promoter Methylation in the Risk of Coronary Artery Disease Among South Indians. Indian J Clin Biochem 2018; 34:180-187. [PMID: 31092991 DOI: 10.1007/s12291-018-0740-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/08/2018] [Indexed: 02/07/2023]
Abstract
Inflammation plays an important role in the pathogenesis of atherosclerosis and coronary syndromes; moreover, various lines of evidence suggest that genetic factors do contribute to the risk of coronary artery disease (CAD). The proinflammatory cytokine IL-6 is a central mediator of inflammation associated with CAD. The present study is aimed to investigate the association of single nucleotide polymorphism in the promoter region of the IL-6 gene (-174 G > C) and methylation with the susceptibility of CAD. Genotyping of IL-6 -174 G/C polymorphism was performed by PCR-RFLP. Methylation-specific PCR method was used to study the IL-6 gene promoter methylation. Analysis of 470 subjects (265 CAD patients and 205 controls) showed association of the -174 G/C variant with the CAD risk in dominant model (OR 1.58, 95% CI, 1.024-2.23, P = 0.04). Further, the analysis of the distribution of genotypes and alleles of -174 G > C polymorphism according to clinical features of CAD, revealed significant association of genotype and allele (OR 1.86, 95% CI 1.18-2.84 P = 0.01, and OR 1.71, 95% CI 1.09-2.23 P = 0.02 respectively) with diabetes, and we found no association with hypertension (OR 0.95, 95% CI 0.57-1.59, P = 0.8). We also analyzed the methylation status of IL-6 promoter region between cases and controls showed significant hypo methylation in CAD subjects (OR 2.36, 95% CI 1.51-4.259, P = 0.006). Additionally, GC, CC genotypes and C allele carriers show hypomethylation in CAD cases compared to controls (54.58 vs. 76.85%, 29.83 vs. 40% respectively). In conclusion, the promoter polymorphism -174 G/C is associated with CAD risk and further carriers of 'C' allele at -174 locus showed significant hypo methylation which could contribute to increased risk of CAD. The present study highlights the association of allele and genotypes with differential DNA methylation of CpG islands in the IL-6 promoter region which may affect IL-6 gene regulation.
Collapse
|
40
|
Yari M, Bitarafan S, Broumand MA, Fazeli Z, Rahimi M, Ghaderian SMH, Mirfakhraie R, Omrani MD. Association between Long Noncoding RNA ANRIL Expression Variants and Susceptibility to Coronary Artery Disease. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2018; 7:1-7. [PMID: 30234067 PMCID: PMC6134424 DOI: 10.22088/ijmcm.bums.7.1.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 01/06/2018] [Indexed: 11/25/2022]
Abstract
Animal cells possess thousands of long non-coding (lnc) RNAs, such as antisense noncoding RNA in the INK4 locus (ANRIL), which have regulatory roles in the cells’ molecular mechanisms, including X-chromosome inactivation, and developmental processes. These lnc RNAs are known to influence the extensive spectrum of age-related disorders. Accordingly, there is evidence for the role of these lnc RNAs in cardiovascular diseases, particularly coronary artery diseases (CAD). The aim of this study was to assess whether the expression of the lnc RNA ANRIL was associated with a susceptibility to CAD by evaluating the expression level of the two transcripts of ANRIL. Peripheral blood was taken from fifty patients affected by CAD and relative expression of ANRIL was determined by Real-Time PCR assay. The obtained data indicated that the EU741058 transcript expression level significantly decreased in CAD patients in comparison with the healthy individuals (P= 0.001). Furthermore, there was no significant association between the NR_003529 transcript expression, and CAD risk in Iranian patients (P=0.751). Our results suggest that the expression level of the EU741058 transcript of ANRIL may be implicated in CAD development, creating a predictive biomarker for CAD patients in future.
Collapse
Affiliation(s)
- Mohsen Yari
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Bitarafan
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Broumand
- Department of Molecular Pathology, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Fazeli
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahnoosh Rahimi
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Bioinformatics and Genomics , Pharmacogenetic Research Center, Simple LIMS, San Diego, CA, USA
| | | | - Reza Mirfakhraie
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mir Davood Omrani
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Musunuru K, Ingelsson E, Fornage M, Liu P, Murphy AM, Newby LK, Newton-Cheh C, Perez MV, Voora D, Woo D. The Expressed Genome in Cardiovascular Diseases and Stroke: Refinement, Diagnosis, and Prediction: A Scientific Statement From the American Heart Association. ACTA ACUST UNITED AC 2018; 10:HCG.0000000000000037. [PMID: 28760750 DOI: 10.1161/hcg.0000000000000037] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There have been major advances in our knowledge of the contribution of DNA sequence variations to cardiovascular disease and stroke. However, the inner workings of the body reflect the complex interplay of factors beyond the DNA sequence, including epigenetic modifications, RNA transcripts, proteins, and metabolites, which together can be considered the "expressed genome." The emergence of high-throughput technologies, including epigenomics, transcriptomics, proteomics, and metabolomics, is now making it possible to address the contributions of the expressed genome to cardiovascular disorders. This statement describes how the expressed genome can currently and, in the future, potentially be used to diagnose diseases and to predict who will develop diseases such as coronary artery disease, stroke, heart failure, and arrhythmias.
Collapse
|
42
|
Duan L, Liu C, Hu J, Liu Y, Wang J, Chen G, Li Z, Chen H. Epigenetic mechanisms in coronary artery disease: The current state and prospects. Trends Cardiovasc Med 2017; 28:311-319. [PMID: 29366539 DOI: 10.1016/j.tcm.2017.12.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 12/18/2017] [Accepted: 12/23/2017] [Indexed: 12/12/2022]
Abstract
Coronary artery disease (CAD) is the leading cause of morbidity and mortality. CAD has both genetic and environmental causes. In the past two decades, the understanding of epigenetics has advanced swiftly and vigorously. It has been demonstrated that epigenetic modifications are associated with the onset and progression of CAD. This review aims to improve the understanding of the epigenetic mechanisms closely related to CAD and to provide a novel perspective on the onset and development of CAD. Epigenetic changes include DNA methylation, histone modification, microRNA and lncRNA, which are interrelated with critical genes and influence the expression of those genes. In addition, miRNA plays a diverse role in the pathological process of CAD. Numerous studies have found that some cardiac-specific miRNAs have potential as certain diagnostic biomarkers and treatment targets for CAD. In this review, the aberrant epigenetic mechanisms that contribute to CAD will be discussed. We will also provide novel insight into the epigenetic mechanisms that target CAD.
Collapse
Affiliation(s)
- Lian Duan
- Department of Cardiology, Guang' an men Hospital, No. 5 Beixiange, Xicheng District, Beijing, China; Beijing University of Traditional Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, China
| | - Chao Liu
- Department of Cardiology, Guang' an men Hospital, No. 5 Beixiange, Xicheng District, Beijing, China; Beijing University of Traditional Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, China
| | - Junyuan Hu
- Department of Cardiology, Guang' an men Hospital, No. 5 Beixiange, Xicheng District, Beijing, China; Beijing University of Traditional Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, China
| | - Yongmei Liu
- Department of Cardiology, Guang' an men Hospital, No. 5 Beixiange, Xicheng District, Beijing, China
| | - Jie Wang
- Department of Cardiology, Guang' an men Hospital, No. 5 Beixiange, Xicheng District, Beijing, China.
| | - Guang Chen
- Department of Cardiology, Guang' an men Hospital, No. 5 Beixiange, Xicheng District, Beijing, China; Beijing University of Traditional Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, China
| | - Zhaoling Li
- Department of Cardiology, Guang' an men Hospital, No. 5 Beixiange, Xicheng District, Beijing, China
| | - Hengwen Chen
- Department of Cardiology, Guang' an men Hospital, No. 5 Beixiange, Xicheng District, Beijing, China
| |
Collapse
|
43
|
Giannakopoulou E, Konstantinou F, Ragia G, Tavridou A, Karaglani M, Chatzaki E, Papapetropoulos A, Mikroulis D, Manolopoulos VG. Epigenetics-by-Sex Interaction for Coronary Artery Disease Risk Conferred by the Cystathionine γ-Lyase Gene Promoter Methylation. ACTA ACUST UNITED AC 2017; 21:741-748. [DOI: 10.1089/omi.2017.0149] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Efstathia Giannakopoulou
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Fotios Konstantinou
- Department of Cardiothoracic Surgery, Academic General Hospital of Evros, Alexandroupolis, Greece
| | - Georgia Ragia
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Anna Tavridou
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
- Clinical Pharmacology Unit, Academic General Hospital of Evros, Alexandroupolis, Greece
| | - Makrina Karaglani
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Ekaterini Chatzaki
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Mikroulis
- Department of Cardiothoracic Surgery, Academic General Hospital of Evros, Alexandroupolis, Greece
| | - Vangelis G. Manolopoulos
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
- Clinical Pharmacology Unit, Academic General Hospital of Evros, Alexandroupolis, Greece
| |
Collapse
|
44
|
Zhong J, Chen X, Ye H, Wu N, Chen X, Duan S. CDKN2A and CDKN2B methylation in coronary heart disease cases and controls. Exp Ther Med 2017; 14:6093-6098. [PMID: 29285163 DOI: 10.3892/etm.2017.5310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 03/24/2017] [Indexed: 01/13/2023] Open
Abstract
The aim of the present study was to investigate the association between cyclin-dependent kinase inhibitor 2A (CDKN2A) and cyclin-dependent kinase inhibitor 2B (CDKN2B) methylation, and coronary heart disease (CHD), and to explore the interaction between methylation status and CHD clinical characteristics in Han Chinese patients. A total of 189 CHD (96 males, 93 females) and 190 well-matched non-CHD controls (96 males, 94 females) were recruited for the study. Methylation-specific polymerase chain reaction technology was used to examine gene promoter methylation status. Comparisons of methylation frequencies between CHD and non-CHD patients were carried out using the Chi-square test. Methylation levels of CDKN2A and CDKN2B genes were not found to be associated with the risk of CHD. However, the mean age of CDKN2A-hypermethylated participants was significantly lower than CDKN2A-unmethylated participants (58.73±5.88 vs. 62.62±5.36 years, adjusted P<0.001). Conversely, the mean age of CDKN2B-hypermethylated participants was significantly higher compared with CDKN2B-unmethylated participants (62.26±5.48 vs. 58.33±7.47 years, adjusted P=0.048). In addition, CDKN2B methylation frequencies were significantly increased in female participants compared with males (99.47 vs. 11.98%, P=0.032). In conclusion, the results indicated that CDKN2A and CDKN2B promoter methylation frequencies were significantly associated with age, and there was a gender dimorphism in CDKN2B methylation.
Collapse
Affiliation(s)
- Jinyan Zhong
- Cardiology Center, Ningbo First Hospital, Ningbo University, Ningbo, Zhejiang 315010, P.R. China.,Department of Cardiology, Ningbo Second Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Xiaoying Chen
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Huadan Ye
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Nan Wu
- Cardiology Center, Ningbo First Hospital, Ningbo University, Ningbo, Zhejiang 315010, P.R. China.,Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xiaomin Chen
- Cardiology Center, Ningbo First Hospital, Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Shiwei Duan
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
45
|
Curtis EM, Murray R, Titcombe P, Cook E, Clarke-Harris R, Costello P, Garratt E, Holbrook JD, Barton S, Inskip H, Godfrey KM, Bell CG, Cooper C, Lillycrop KA, Harvey NC. Perinatal DNA Methylation at CDKN2A Is Associated With Offspring Bone Mass: Findings From the Southampton Women's Survey. J Bone Miner Res 2017; 32:2030-2040. [PMID: 28419547 PMCID: PMC5528139 DOI: 10.1002/jbmr.3153] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 03/28/2017] [Accepted: 04/07/2017] [Indexed: 12/21/2022]
Abstract
Poor intrauterine and childhood growth has been linked with the risk of osteoporosis in later life, a relationship that may in part be mediated through altered epigenetic regulation of genes. We previously identified a region within the promoter of the long non-coding RNA ANRIL encoded by the CDKN2A locus, at which differential DNA methylation at birth showed correlations with offspring adiposity. Given the common lineage of adipocytes and osteoblasts, we investigated the relationship between perinatal CDKN2A methylation and bone mass at ages 4 and 6 years. Using sodium bisulfite pyrosequencing, we measured the methylation status of the 9 CpGs within this region in umbilical cord samples from discovery (n = 332) and replication (n = 337) cohorts of children from the Southampton Women's Survey, whose bone mass was assessed by dual-energy X-ray absorptiomietry (DXA; Hologic Discovery). Inverse associations were found between perinatal CDKN2A methylation and whole-body minus head bone area (BA), bone mineral content (BMC), and areal bone mineral density (BMD). This was confirmed in replication and combined data sets (all p < 0.01), with each 10% increase in methylation being associated with a decrease in BMC of 4 to 9 g at age 4 years (p ≤ 0.001). Relationships were similar with 6-year bone mass. Functional investigation of the differentially methylated region in the SaOS-2 osteosarcoma cell line showed that transcription factors bound to the identified CpGs in a methylation-specific manner and that CpG mutagenesis modulated ANRIL expression. In conclusion, perinatal methylation at CDKN2A is associated with childhood bone development and has significance for cell function. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Elizabeth M Curtis
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Robert Murray
- Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Philip Titcombe
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Eloïse Cook
- Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | | | - Paula Costello
- Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Emma Garratt
- Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Joanna D Holbrook
- Institute of Developmental Sciences, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK.,Singapore Institute for Clinical Sciences (SICS), A*STAR, Brenner Centre for Molecular Medicine, Singapore
| | - Sheila Barton
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Hazel Inskip
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK.,Institute of Developmental Sciences, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Christopher G Bell
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK.,Institute of Developmental Sciences, University of Southampton, Southampton, UK.,Centre for Biological Sciences, University of Southampton, Southampton, UK
| | - Cyrus Cooper
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK.,NIHR Oxford Musculoskeletal Biomedical Research Unit, University of Oxford, Oxford, UK
| | - Karen A Lillycrop
- Institute of Developmental Sciences, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK.,Centre for Biological Sciences, University of Southampton, Southampton, UK
| | - Nicholas C Harvey
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
46
|
Li Q, Peng W, Li H, Zhuang J, Luo X, Xu Y. Association of the single nucleotide polymorphism in chromosome 9p21 and chromosome 9q33 with coronary artery disease in Chinese population. BMC Cardiovasc Disord 2017; 17:255. [PMID: 28962556 PMCID: PMC5622451 DOI: 10.1186/s12872-017-0685-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 09/12/2017] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Our study aims to explore the association of rs7025486 single-nucleotide polymorphisms (SNP) in DAB2IP and rs1333049 on chromosome 9p21.3 with the coronary artery disease in Chinese population. METHODS All patients came from the east China area and underwent coronary angiography. Rs7025486 and rs1333049 polymorphism were genotyped in 555 patients with CAD and in 480 healthy controls that underwent coronary angiography. RESULTS In Chinese population, the rs7025486 genotype in the case group was no significant different than the control group (P = 0.531).Meanwhile, the rs1333049 SNP has statistically significant (P = 0.006), which was the independent risk factors for CAD (OR1.252, P = 0.039), and consistent with the past studies conclusion. CONCLUSION Genotype of rs1333049 on chromosome 9p21, but not rs7025486 on chromosome 9q33, is an independent determinant of the incidence of CAD in Chinese population.
Collapse
Affiliation(s)
- Qi Li
- No. 113 Hospital of Chinese People's Liberation Army (PLA), Ningbo, China
| | - Wenhui Peng
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hailing Li
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianhui Zhuang
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xuesheng Luo
- No. 113 Hospital of Chinese People's Liberation Army (PLA), Ningbo, China
| | - Yawei Xu
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
47
|
Cheng M, An S, Li J. CDKN2B - AS may indirectly regulate coronary artery disease-associated genes via targeting miR - 92a. Gene 2017; 629:101-107. [DOI: 10.1016/j.gene.2017.07.070] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/20/2017] [Accepted: 07/27/2017] [Indexed: 12/27/2022]
|
48
|
Fernández-Sanlés A, Sayols-Baixeras S, Subirana I, Degano IR, Elosua R. Association between DNA methylation and coronary heart disease or other atherosclerotic events: A systematic review. Atherosclerosis 2017; 263:325-333. [PMID: 28577936 DOI: 10.1016/j.atherosclerosis.2017.05.022] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/05/2017] [Accepted: 05/17/2017] [Indexed: 11/24/2022]
Abstract
BACKGROUND AND AIMS The aim of this study was to perform a systematic review of the association between DNA methylation and coronary heart disease (CHD) or related atherosclerotic traits. METHODS A systematic review was designed. The condition of interest was DNA methylation, and the outcome was CHD or other atherosclerosis-related traits. Three DNA methylation approaches were considered: global methylation, candidate-gene, and epigenome-wide association studies (EWAS). A functional analysis was undertaken using the Ingenuity Pathway Analysis software. RESULTS In total, 51 articles were included in the analysis: 12 global methylation, 34 candidate-gene and 11 EWAS, with six studies using more than one approach. The results of the global methylation studies were inconsistent. The candidate-gene results were consistent for some genes, suggesting that hypermethylation in ESRα, ABCG1 and FOXP3 and hypomethylation in IL-6 were associated with CHD. The EWAS identified 84 genes showing differential methylation associated with CHD in more than one study. The probability of these findings was <1.37·10-5. One third of these genes have been related to obesity in genome-wide association studies. The functional analysis identified several diseases and functions related to these set of genes: inflammatory, metabolic and cardiovascular disease. CONCLUSIONS Global DNA methylation seems to be not associated with CHD. The evidence from candidate-gene studies was limited. The EWAS identified a set of 84 genes highlighting the relevance of obesity, inflammation, lipid and carbohydrate metabolism in CHD. This set of genes could be prioritized in future studies assessing the role of DNA methylation in CHD.
Collapse
Affiliation(s)
- Alba Fernández-Sanlés
- Cardiovascular Epidemiology and Genetics Research Group, REGICOR Study Group, IMIM (Hospital Del Mar Medical Research Institute), Barcelona, Catalonia, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain
| | - Sergi Sayols-Baixeras
- Cardiovascular Epidemiology and Genetics Research Group, REGICOR Study Group, IMIM (Hospital Del Mar Medical Research Institute), Barcelona, Catalonia, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain; CIBER Cardiovascular Diseases (CIBERCV), Barcelona, Catalonia, Spain
| | - Isaac Subirana
- Cardiovascular Epidemiology and Genetics Research Group, REGICOR Study Group, IMIM (Hospital Del Mar Medical Research Institute), Barcelona, Catalonia, Spain; CIBER Epidemiology and Public Health (CIBERESP), Barcelona, Catalonia, Spain
| | - Irene R Degano
- Cardiovascular Epidemiology and Genetics Research Group, REGICOR Study Group, IMIM (Hospital Del Mar Medical Research Institute), Barcelona, Catalonia, Spain; CIBER Cardiovascular Diseases (CIBERCV), Barcelona, Catalonia, Spain
| | - Roberto Elosua
- Cardiovascular Epidemiology and Genetics Research Group, REGICOR Study Group, IMIM (Hospital Del Mar Medical Research Institute), Barcelona, Catalonia, Spain; CIBER Cardiovascular Diseases (CIBERCV), Barcelona, Catalonia, Spain.
| |
Collapse
|
49
|
Guo T, Chai X, Liu Q, Peng W, Peng Z, Cai Y. Downregulation of P16 promotes cigarette smoke extract-induced vascular smooth muscle cell proliferation via preventing G1/S phase transition. Exp Ther Med 2017; 14:214-220. [PMID: 28672917 PMCID: PMC5488496 DOI: 10.3892/etm.2017.4468] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 12/09/2016] [Indexed: 12/24/2022] Open
Abstract
The proliferation of vascular smooth muscle cells (VSMCs) serves an important role in cigarette smoking-associated vascular diseases; however, the underlying mechanisms responsible for this remain unclear. The aim of the present study was to elucidate the role of P16 in cigarette smoke extract (CSE)-induced VSMC proliferation and the underlying mechanism responsible. Human aortic smooth muscle cells (HAOSMCs) were exposed to CSE, and an MTT assay and flow cytometry were performed to evaluate cell proliferation and cell cycle distribution. Western blotting was conducted to examine protein expression and bisulfite genomic sequencing polymerase chain reaction was used to determine the methylation status of the P16 promoter CpG island. It was demonstrated that treatment with CSE significantly promoted the proliferation of HAOSMCs in a concentration- and time-dependent manner and induced a downregulation in P16 expression (all P<0.05). A luciferase reporter gene assay data demonstrated that CSE treatment induced hypermethylation of the P16 promoter, which led to a significant decrease in its transcriptional activity and significantly reduced P16 protein expression in HAOSMCs (both P<0.01). Furthermore, P16 downregulation induced a significant increase in the expression of cyclin-dependent kinase (CDK) 4, CDK6 and phosphorylated retinoblastoma (p-Rb) protein (all P<0.001) and significantly increased the ratio of cells in S phase in CSE-treated HAOSMCs (P<0.001). Overexpression of P16 inhibited CSE-induced cell proliferation through inducing cell cycle arrest in G1 phase (P<0.001), and led to decreased levels of CDK4 (P<0.01), CDK6 (P<0.01) and p-Rb (P<0.001) in HASMCs. The results of the present study therefore demonstrate that P16 may be associated with the CSE-induced proliferation of VSMCs, suggesting that P16 serves a role in the development of cigarette smoke-associated vascular diseases.
Collapse
Affiliation(s)
- Tao Guo
- Department of Emergency Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Xiangping Chai
- Department of Emergency Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Qiming Liu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Wen Peng
- Department of Emergency Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Zhenyu Peng
- Department of Emergency Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Yuzhong Cai
- Department of Emergency Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
50
|
Lillycrop K, Murray R, Cheong C, Teh AL, Clarke-Harris R, Barton S, Costello P, Garratt E, Cook E, Titcombe P, Shunmuganathan B, Liew SJ, Chua YC, Lin X, Wu Y, Burdge GC, Cooper C, Inskip HM, Karnani N, Hopkins JC, Childs CE, Chavez CP, Calder PC, Yap F, Lee YS, Chong YS, Melton PE, Beilin L, Huang RC, Gluckman PD, Harvey N, Hanson MA, Holbrook JD, Godfrey KM. ANRIL Promoter DNA Methylation: A Perinatal Marker for Later Adiposity. EBioMedicine 2017; 19:60-72. [PMID: 28473239 PMCID: PMC5440605 DOI: 10.1016/j.ebiom.2017.03.037] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 03/13/2017] [Accepted: 03/27/2017] [Indexed: 12/22/2022] Open
Abstract
Experimental studies show a substantial contribution of early life environment to obesity risk through epigenetic processes. We examined inter-individual DNA methylation differences in human birth tissues associated with child's adiposity. We identified a novel association between the level of CpG methylation at birth within the promoter of the long non-coding RNA ANRIL (encoded at CDKN2A) and childhood adiposity at age 6-years. An association between ANRIL methylation and adiposity was also observed in three additional populations; in birth tissues from ethnically diverse neonates, in peripheral blood from adolescents, and in adipose tissue from adults. Additionally, CpG methylation was associated with ANRIL expression in vivo, and CpG mutagenesis in vitro inhibited ANRIL promoter activity. Furthermore, CpG methylation enhanced binding to an Estrogen Response Element within the ANRIL promoter. Our findings demonstrate that perinatal methylation at loci relevant to gene function may be a robust marker of later adiposity, providing substantial support for epigenetic processes in mediating long-term consequences of early life environment on human health.
Collapse
Affiliation(s)
- Karen Lillycrop
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK; NIHR Southampton Biomedical Research Centre, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Robert Murray
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK.
| | - Clara Cheong
- Singapore Institute for Clinical Sciences (SICS), Agency for Science Technology and Research (A*STAR), Singapore
| | - Ai Ling Teh
- Singapore Institute for Clinical Sciences (SICS), Agency for Science Technology and Research (A*STAR), Singapore
| | - Rebecca Clarke-Harris
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Sheila Barton
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Paula Costello
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Emma Garratt
- NIHR Southampton Biomedical Research Centre, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Eloise Cook
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Philip Titcombe
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Bhuvaneshwari Shunmuganathan
- Singapore Institute for Clinical Sciences (SICS), Agency for Science Technology and Research (A*STAR), Singapore
| | - Samantha J Liew
- Singapore Institute for Clinical Sciences (SICS), Agency for Science Technology and Research (A*STAR), Singapore
| | - Yong-Cai Chua
- Singapore Institute for Clinical Sciences (SICS), Agency for Science Technology and Research (A*STAR), Singapore
| | - Xinyi Lin
- Singapore Institute for Clinical Sciences (SICS), Agency for Science Technology and Research (A*STAR), Singapore
| | - Yonghui Wu
- Singapore Institute for Clinical Sciences (SICS), Agency for Science Technology and Research (A*STAR), Singapore
| | - Graham C Burdge
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Cyrus Cooper
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Hazel M Inskip
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Neerja Karnani
- Singapore Institute for Clinical Sciences (SICS), Agency for Science Technology and Research (A*STAR), Singapore
| | - James C Hopkins
- Academic Unit of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Caroline E Childs
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Carolina Paras Chavez
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Philip C Calder
- NIHR Southampton Biomedical Research Centre, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fabian Yap
- Department of Paediatrics, KK Women's and Children's Hospital, Singapore; Duke NUS Graduate School of Medicine, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Yung Seng Lee
- Singapore Institute for Clinical Sciences (SICS), Agency for Science Technology and Research (A*STAR), Singapore; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences (SICS), Agency for Science Technology and Research (A*STAR), Singapore; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Philip E Melton
- Centre for Genetics of Health and Disease, University of Western, Australia; Faculty of Health Science, Curtin University, Australia
| | - Lawrie Beilin
- School of Medicine and Pharmacology, University of Western Australia, Australia
| | - Rae-Chi Huang
- Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Peter D Gluckman
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Nick Harvey
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Mark A Hanson
- NIHR Southampton Biomedical Research Centre, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Joanna D Holbrook
- Singapore Institute for Clinical Sciences (SICS), Agency for Science Technology and Research (A*STAR), Singapore
| | - Keith M Godfrey
- NIHR Southampton Biomedical Research Centre, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| |
Collapse
|