1
|
Morgan M, Yellapu V, Short D, Ruggeri C. Trends in In-Hospital Mortality in Patients Admitted With Cardiovascular Diseases in the United States With Demographics and Risk Factors of All Cardiovascular In-Hospital Mortality: Analysis of the 2021 National Inpatient Sample Database. Cureus 2024; 16:e70620. [PMID: 39483569 PMCID: PMC11526619 DOI: 10.7759/cureus.70620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
Introduction and background Cardiovascular diseases (CVDs) encompass a range of disorders involving coronary artery diseases, valvular heart diseases, myocardial diseases, pericardial diseases, hypertensive heart diseases, heart failure (HF), and pulmonary artery diseases. Given the high prevalence of CVDs, understanding both overall and in-hospital mortality rates from these diseases is crucial. Unsurprisingly, most research, procedures, and new pharmacological interventions aim to reduce these rates. No recent studies have comprehensively detailed in-hospital mortality rates, demographics, and risk factors for all CVDs combined. Yet, in-hospital mortality rates due to CVD significantly impact patients' families and healthcare teams and serve as a critical measure of healthcare system development and effectiveness. Therefore, analyzing in-hospital mortality rates is essential for filling the gap in the recent comprehensive analysis of in-hospital mortality rates, demographics, and risk factors of all CVDs. Method The study used data from the National Inpatient Sample and the Nationwide Inpatient Sample (NIS) Databases of 2021 and HCUP tools. The NIS database extrapolates national estimates based on a stratified sample of 20% of US hospital discharges. Results were expressed as probability and relative risk using the t-test, with a P-value <0.05 being statistically significant. Statistical analyses were done using Stata statistical software version 18 (StataCorp LLC, College Station, TX, US). Results This study included 6,666,752 hospital admissions in the United States. Of these, 2,337,589 patients were admitted with CVDs and related symptoms, with 70,552 deaths occurring during hospitalization, resulting in an in-hospital mortality rate of 3.01% due to CVDs. Our study showed all CVD-induced in-hospital mortality combined was found to have a higher association with diabetes but a lower association with hypertension, hyperlipidemia, alcohol, and smoking. Conclusion The highest rates of cardiovascular disease in-hospital mortality are cardiac arrest, rupture of the cardiac wall as a complication of acute myocardial infarction, cardiogenic shock, rupture of papillary muscle as a complication of acute myocardial infarction, and rupture of chorda tendinea as a complication of acute myocardial infarction. The most common causes of CVD in-hospital mortality are non-ST-elevation myocardial infarction (NSTEMI) (19.20%), ST-elevation myocardial infarction (STEMI) (17.80%), cardiac arrest (15.10%), hypertensive heart disease with heart failure (12.50%), ventricular fibrillation (4.70%), ventricular tachycardia (3.30%), and aortic stenosis (2.10%). The most common risk factors for CVD in-hospital mortality are age, male gender, and diabetes. Proper diabetes control and management might be the highest preventive measure for all CVD-induced in-hospital mortality.
Collapse
Affiliation(s)
| | - Vikas Yellapu
- Cardiology, St. Luke's University Health Network, Bethlehem, USA
| | - Daryn Short
- Medicine, Temple University, Philadelphia, USA
| | - Cara Ruggeri
- Internal Medicine, St. Luke's University Health Network, Bethlehem, USA
| |
Collapse
|
2
|
Lu X. Structure and Function of Angiopoietin-like Protein 3 (ANGPTL3) in Atherosclerosis. Curr Med Chem 2020; 27:5159-5174. [PMID: 31223079 DOI: 10.2174/0929867326666190621120523] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/24/2019] [Accepted: 04/30/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Angiopoietin-Like Proteins (ANGPTLs) are structurally related to the angiopoietins. A total of eight ANGPTLs (from ANGPTL1 to ANGPTL8) have been identified so far. Most ANGPTLs possess multibiological functions on lipid metabolism, atherosclerosis, and cancer. Among them, ANGPTL3 has been shown to regulate the levels of Very Low-Density Lipoprotein (VLDL) made by the liver and play a crucial role in human lipoprotein metabolism. METHOD A systematic appraisal of ANGPTLs was conducted, focusing on the main features of ANGPTL3 that has a significant role in atherosclerosis. RESULTS Angiopoietins including ANGPTL3 are vascular growth factors that are highly specific for endothelial cells, perform a variety of other regulatory activities to influence inflammation, and have been shown to possess both pro-atherosclerotic and atheroprotective effects. CONCLUSION ANGPTL3 has been demonstrated as a promising target in the pharmacological management of atherosclerosis. However, many questions remain about its biological functions.
Collapse
Affiliation(s)
- Xinjie Lu
- The Mary and Garry Weston Molecular Immunology Laboratory, Thrombosis Research Institute, London SW3 6LR, England, United Kingdom
| |
Collapse
|
3
|
A Genomic Approach to Characterize the Vulnerable Patient – a Clinical Update. JOURNAL OF INTERDISCIPLINARY MEDICINE 2019. [DOI: 10.2478/jim-2019-0023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Atherosclerosis is the elemental precondition for any cardiovascular disease and the predominant cause of ischemic heart disease that often leads to myocardial infarction. Systemic risk factors play an important role in the starting and progression of atherosclerosis. The complexity of the disease is caused by its multifactorial origin. Besides the traditional risk factors, genetic predisposition is also a strong risk factor. Many studies have intensively researched cardioprotective drugs, which can relieve myocardial ischemia and reperfusion injury, thereby reducing infarct size. A better understanding of abnormal epigenetic pathways in the myocardial pathology may result in new treatment options. Individualized therapy based on genome sequencing is important for an effective future medical treatment. Studies based on multiomics help to better understand the pathophysiological mechanism of several diseases at a molecular level. Epigenomic, transcriptomic, proteomic, and metabolomic research may be essential in detecting the pathological phenotype of myocardial ischemia and ischemic heart failure.
Collapse
|
4
|
LDLR rs688 TT Genotype and T Allele Are Associated with Increased Susceptibility to Coronary Artery Disease-A Case-Control Study. J Cardiovasc Dev Dis 2018; 5:jcdd5020031. [PMID: 29843469 PMCID: PMC6023456 DOI: 10.3390/jcdd5020031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/21/2018] [Accepted: 05/23/2018] [Indexed: 11/16/2022] Open
Abstract
PURPOSE The low-density lipoprotein receptor is responsible for the binding and uptake of plasma LDL particles and plays a critical role in maintaining cellular cholesterol homeostasis. LDLR gene SNP rs688 has been reported to be associated with increased plasma total and LDL cholesterol in several populations and can lead to elevated plasma LDL levels, resulting in an increased risk for atherosclerosis and coronary artery disease. This study aimed to explore genetic LDLR variant rs688 for its potential roles in coronary artery disease. METHODOLOGY This study recruited 200 coronary artery disease patients and 200 healthy individuals. Genotyping of LDLR-rs688C > T gene variations was performed using the allele specific PCR method. Correlation of LDLR-rs688C > T gene variants with different clinicopathological features of coronary artery disease patients was performed. Pooled odds ratios (ORs) and 95% confidence intervals (CIs) were applied to evaluate the correlation of this microRNA polymorphism with coronary artery disease risk. RESULTS A significant difference was observed in genotype distribution among the coronary artery disease and matched healthy controls (p = 0.003). The frequencies of all three genotypes CC, CT, TT reported in the patient samples were 14%, 65% and 21% and in the healthy controls samples were 18%, 73% and 9%, respectively. The increased risk of developing CAD in Indian patients was found to be associated with LDLR rs688 TT genotype (OR = 3.0, 95% CI, 1.43 × 6.2; p = 0.003) RR 1.87 (1.20⁻2.91) p = 0.0037) and also the increased risk of developing CAD was reported to be associated with LDLR rs688 T allele (OR = 0.74, 95% CI, 1.57⁻0.97; p = 0.03) RR 0.85 (0.73⁻0.99) p = 0.03) compared to the C allele. Therefore, it was observed that more than a 3.0- and 0.74-fold increase risk of developing CAD was associated with TT genotype and T allele in Indian coronary artery disease patients. CONCLUSION The findings indicated that LDLR rs688 TT genotype and T allele are associated with an increased susceptibility to coronary artery disease patients. LDLR-rs688C > T gene variation can be used as a predisposing genetic marker for coronary artery disease. Further studies with larger sample sizes are necessary to confirm our findings.
Collapse
|
5
|
Veljkovic N, Zaric B, Djuric I, Obradovic M, Sudar-Milovanovic E, Radak D, Isenovic ER. Genetic Markers for Coronary Artery Disease. MEDICINA (KAUNAS, LITHUANIA) 2018; 54:E36. [PMID: 30344267 PMCID: PMC6122104 DOI: 10.3390/medicina54030036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/18/2018] [Accepted: 05/22/2018] [Indexed: 12/22/2022]
Abstract
Coronary artery disease (CAD) and myocardial infarction (MI) are recognized as leading causes of mortality in developed countries. Although typically associated with behavioral risk factors, such as smoking, sedentary lifestyle, and poor dietary habits, such vascular phenotypes have also long been recognized as being related to genetic background. We review the currently available data concerning genetic markers for CAD in English and non-English articles with English abstracts published between 2003 and 2018. As genetic testing is increasingly available, it may be possible to identify adequate genetic markers representing the risk profile and to use them in a clinical setting.
Collapse
Affiliation(s)
- Nevena Veljkovic
- Centre for Multidisciplinary Research and Engineering, Institute of Nuclear Science Vinca, University of Belgrade, 11000 Belgrade, Serbia.
| | - Bozidarka Zaric
- Laboratory of Radiobiology and Molecular Genetics, Institute of Nuclear Science Vinca, University of Belgrade, 11000 Belgrade, Serbia.
| | - Ilona Djuric
- Department for Endocrinology and Immunoradiology 11080 Zemun, Institute for the Application of Nuclear Energy-INEP, University of Belgrade, 11000 Belgrade, Serbia.
| | - Milan Obradovic
- Laboratory of Radiobiology and Molecular Genetics, Institute of Nuclear Science Vinca, University of Belgrade, 11000 Belgrade, Serbia.
| | - Emina Sudar-Milovanovic
- Laboratory of Radiobiology and Molecular Genetics, Institute of Nuclear Science Vinca, University of Belgrade, 11000 Belgrade, Serbia.
| | - Djordje Radak
- School of Medicine, Dedinje Cardiovascular Institute, University of Belgrade, 11000 Belgrade, Serbia.
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia.
- Serbian Academy of Sciences and Arts, 11000 Belgrade, Serbia.
| | - Esma R Isenovic
- Laboratory of Radiobiology and Molecular Genetics, Institute of Nuclear Science Vinca, University of Belgrade, 11000 Belgrade, Serbia.
| |
Collapse
|
6
|
Abstract
PURPOSE OF REVIEW The current review is to describe the genetic risk variants that have been discovered predisposing to coronary artery disease (CAD) and how they are utilized to stratify for risk of CAD. RECENT FINDINGS Over 90 genetic risk variants have been discovered that predispose to risk for CAD. SUMMARY The total genetic risk burden for CAD is proportional to the number of risk variants inherited and can be combined into a single number referred to as the genetic risk score (GRS). GRS has been utilized in multiple studies and shown to be more effective in risk stratification for CAD than conventional risk factors. There is a major advantage to risk stratification based on the GRS since the risk can be determined at birth or anytime throughout one's lifetime since the individual's DNA does not change. Widespread application of the GRS is likely to enable a paradigm shift in the primary prevention of CAD.
Collapse
|
7
|
Genome-Wide Association Studies and Risk Scores for Coronary Artery Disease: Sex Biases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1065:627-642. [PMID: 30051411 DOI: 10.1007/978-3-319-77932-4_38] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Phenotypic sex differences in coronary artery disease (CAD) and its risk factors have been apparent for many decades in basic and clinical research; however, whether these are also present at the gene level and thus influence genome-wide association and genetic risk prediction studies has often been ignored. From fundamental and medical standpoints, this is critically important to assess in order to fully understand the underlying genetic architecture that predisposes to CAD and better predict disease outcomes based on the interaction between genes, sex effects, and environment. In this chapter we aimed to (1) integrate the history and latest research from genome-wide association studies for CAD and clinical and genetic risk scores for prediction of CAD, (2) highlight sex-specific differences in these areas of research, and (3) discuss reasons why sex differences have often not been considered and, where present, why sex differences exist at genetic and phenotypic levels and how important they are for consideration in future research. While we find interesting examples of sex differences in effects of genetic variants on CAD, genome-wide association and genetic risk studies have typically not tested for sex-specific effects despite mounting evidence from diverse fields that these are likely very important to consider at both the genetic and phenotypic levels. In-depth testing for sex effects in large-scale genome-wide association studies that include autosomal and often excluded sex chromosomes alongside parallel improvements in resolution of sex-specific differences for risk factors and disease outcomes for CAD has the potential to substantially improve clinical and genetic risk prediction studies. Developing sex-tailored genetic risk scores as has been done recently for other disorders might be also warranted for CAD. In the era of precision medicine, this level of accuracy is essential for such a common and costly disease.
Collapse
|
8
|
Xu YX, Redon V, Yu H, Querbes W, Pirruccello J, Liebow A, Deik A, Trindade K, Wang X, Musunuru K, Clish CB, Cowan C, Fizgerald K, Rader D, Kathiresan S. Role of angiopoietin-like 3 (ANGPTL3) in regulating plasma level of low-density lipoprotein cholesterol. Atherosclerosis 2017; 268:196-206. [PMID: 29183623 DOI: 10.1016/j.atherosclerosis.2017.08.031] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 08/02/2017] [Accepted: 08/30/2017] [Indexed: 10/18/2022]
Abstract
BACKGROUND AND AIMS Angiopoietin-like 3 (ANGPTL3) has emerged as a key regulator of lipoprotein metabolism in humans. Homozygous loss of ANGPTL3 function causes familial combined hypolipidemia characterized by low plasma levels of triglycerides (TG), high-density lipoprotein cholesterol (HDL-C), and low-density lipoprotein cholesterol (LDL-C). While known effects of ANGPTL3 in inhibiting lipoprotein lipase and endothelial lipase contribute to the low TG and HDL-C, respectively, the basis of low LDL-C remains unclear. Our aim was to explore the role of ANGPTL3 in modulating plasma LDL-C. METHODS We performed RNAi-mediated gene silencing of ANGPTL3 in five mouse models and in human hepatoma cells. We validated results by deleting ANGPTL3 gene using the CRISPR/Cas9 genome editing system. RESULTS RNAi-mediated Angptl3 silencing in mouse livers resulted in very low TG, HDL-C and LDL-C, a pattern similar to the human phenotype. The effect was observed in wild-type and obese mice, while in hCETP/apolipoprotein (Apo) B-100 double transgenic mice, the silencing decreased LDL-C and TG, but not HDL-C. In a humanized mouse model (Apobec1-/- carrying human ApoB-100 transgene) deficient in the LDL receptor (LDLR), Angptl3 silencing had minimum effect on LDL-C, suggesting the effect being linked to LDLR. This observation is supported by an additive effect on LDL-C between ANGPTL3 and PCSK9 siRNAs. ANGPTL3 gene deletion induced cellular long-chain TG and ApoB-100 accumulation with elevated LDLR and LDLR-related protein (LRP) 1 expression. Consistent with this, ANGPTL3 deficiency by gene deletion or silencing reduced nascent ApoB-100 secretion and increased LDL/VLDL uptake. CONCLUSIONS Reduced secretion and increased uptake of ApoB-containing lipoproteins may contribute to the low LDL-C observed in mice and humans with genetic ANGPTL3 deficiency.
Collapse
Affiliation(s)
- Yu-Xin Xu
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Valeska Redon
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, 11-125 Translational Research Center, 3400 Civic Center Blvd, Building 421, Philadelphia, PA 19104-5158, USA
| | - Haojie Yu
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - William Querbes
- Alnylam Pharmaceuticals, 300 Third Street, 3rd Floor, Cambridge, MA 02142, USA
| | - James Pirruccello
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Abigail Liebow
- Alnylam Pharmaceuticals, 300 Third Street, 3rd Floor, Cambridge, MA 02142, USA
| | - Amy Deik
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Kevin Trindade
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, 11-125 Translational Research Center, 3400 Civic Center Blvd, Building 421, Philadelphia, PA 19104-5158, USA
| | - Xiao Wang
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia 19104, USA
| | - Kiran Musunuru
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia 19104, USA
| | - Clary B Clish
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Chad Cowan
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Kevin Fizgerald
- Alnylam Pharmaceuticals, 300 Third Street, 3rd Floor, Cambridge, MA 02142, USA
| | - Daniel Rader
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, 11-125 Translational Research Center, 3400 Civic Center Blvd, Building 421, Philadelphia, PA 19104-5158, USA
| | - Sekar Kathiresan
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
9
|
Genetics: Implications for Prevention and Management of Coronary Artery Disease. J Am Coll Cardiol 2017; 68:2797-2818. [PMID: 28007143 DOI: 10.1016/j.jacc.2016.10.039] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/12/2016] [Accepted: 10/24/2016] [Indexed: 12/21/2022]
Abstract
An exciting new era has dawned for the prevention and management of coronary artery disease (CAD) utilizing genetic risk variants. The recent identification of over 60 susceptibility loci for CAD confirms not only the importance of established risk factors, but also the existence of many novel causal pathways that are expected to improve our understanding of the genetic basis of CAD and facilitate the development of new therapeutic agents over time. Concurrently, Mendelian randomization studies have provided intriguing insights on the causal relationship between CAD-related traits, and highlight the potential benefits of long-term modifications of risk factors. Last, genetic risk scores of CAD may serve not only as prognostic, but also as predictive markers, and carry the potential to considerably improve the delivery of established prevention strategies. This review will summarize the evolution and discovery of genetic risk variants for CAD and their current and future clinical applications.
Collapse
|
10
|
Wirtwein M, Melander O, Sjőgren M, Hoffmann M, Narkiewicz K, Gruchala M, Sobiczewski W. Relationship between selected DNA polymorphisms and coronary artery disease complications. Int J Cardiol 2017; 228:814-820. [DOI: 10.1016/j.ijcard.2016.11.060] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 11/05/2016] [Indexed: 11/16/2022]
|
11
|
Dai X, Wiernek S, Evans JP, Runge MS. Genetics of coronary artery disease and myocardial infarction. World J Cardiol 2016; 8:1-23. [PMID: 26839654 PMCID: PMC4728103 DOI: 10.4330/wjc.v8.i1.1] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 10/18/2015] [Accepted: 11/10/2015] [Indexed: 02/06/2023] Open
Abstract
Atherosclerotic coronary artery disease (CAD) comprises a broad spectrum of clinical entities that include asymptomatic subclinical atherosclerosis and its clinical complications, such as angina pectoris, myocardial infarction (MI) and sudden cardiac death. CAD continues to be the leading cause of death in industrialized society. The long-recognized familial clustering of CAD suggests that genetics plays a central role in its development, with the heritability of CAD and MI estimated at approximately 50% to 60%. Understanding the genetic architecture of CAD and MI has proven to be difficult and costly due to the heterogeneity of clinical CAD and the underlying multi-decade complex pathophysiological processes that involve both genetic and environmental interactions. This review describes the clinical heterogeneity of CAD and MI to clarify the disease spectrum in genetic studies, provides a brief overview of the historical understanding and estimation of the heritability of CAD and MI, recounts major gene discoveries of potential causal mutations in familial CAD and MI, summarizes CAD and MI-associated genetic variants identified using candidate gene approaches and genome-wide association studies (GWAS), and summarizes the current status of the construction and validations of genetic risk scores for lifetime risk prediction and guidance for preventive strategies. Potential protective genetic factors against the development of CAD and MI are also discussed. Finally, GWAS have identified multiple genetic factors associated with an increased risk of in-stent restenosis following stent placement for obstructive CAD. This review will also address genetic factors associated with in-stent restenosis, which may ultimately guide clinical decision-making regarding revascularization strategies for patients with CAD and MI.
Collapse
Affiliation(s)
- Xuming Dai
- Xuming Dai, Szymon Wiernek, Marschall S Runge, Division of Cardiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Szymon Wiernek
- Xuming Dai, Szymon Wiernek, Marschall S Runge, Division of Cardiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - James P Evans
- Xuming Dai, Szymon Wiernek, Marschall S Runge, Division of Cardiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Marschall S Runge
- Xuming Dai, Szymon Wiernek, Marschall S Runge, Division of Cardiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| |
Collapse
|
12
|
Justesen JM, Allin KH, Sandholt CH, Borglykke A, Krarup NT, Grarup N, Linneberg A, Jørgensen T, Hansen T, Pedersen O. Interactions of Lipid Genetic Risk Scores With Estimates of Metabolic Health in a Danish Population. ACTA ACUST UNITED AC 2015; 8:465-72. [DOI: 10.1161/circgenetics.114.000637] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 02/09/2015] [Indexed: 11/16/2022]
Abstract
Background—
There are several well-established lifestyle factors influencing dyslipidemia and currently; 157 genetic susceptibility loci have been reported to be associated with serum lipid levels at genome-wide statistical significance. However, the interplay between lifestyle risk factors and these susceptibility loci has not been fully elucidated. We tested whether genetic risk scores (GRS) of lipid-associated single nucleotide polymorphisms associate with fasting serum lipid traits and whether the effects are modulated by lifestyle factors or estimates of metabolic health.
Methods and Results—
The single nucleotide polymorphisms were genotyped in 2 Danish cohorts: inter99 (n=5961) for discovery analyses and Health2006 (n=2565) for replication. On the basis of published effect sizes of single nucleotide polymorphisms associated with circulating fasting levels of total cholesterol, low-density lipoprotein-cholesterol, high-density lipoprotein-cholesterol, or triglyceride, 4 weighted GRS were constructed. In a cross-sectional design, we investigated whether the effect of these weighted GRSs on lipid levels were modulated by diet, alcohol consumption, physical activity, and smoking or the individual metabolic health status as estimated from body mass index, waist circumference, and insulin resistance assessed using homeostasis model assessment of insulin resistance. All 4 lipid weighted GRSs associated strongly with their respective trait (from
P
=3.3×10
–69
to
P
=1.1×10
–123
). We found interactions between the triglyceride weighted GRS and body mass index and waist circumference on fasting triglyceride levels in Inter99 and replicated these findings in Health2006 (
P
interaction
=9.8×10
–5
and 2.0×10
–5
, respectively, in combined analysis).
Conclusions—
Our findings suggest that individuals who are obese may be more susceptible to the cumulative genetic burden of triglyceride single nucleotide polymorphisms. Therefore, it is suggested that especially these genetically at-risk individuals may benefit more from targeted interventions aiming at obesity prevention.
Collapse
Affiliation(s)
- Johanne M. Justesen
- From The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics (J.M.J., K.H.A., C.H.S., N.T.K., N.G., T.H., O.P.), Department of Clinical Medicine (A.L.) and Department of Public Health (T.J.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Research Centre for Prevention and Health (A.B., A.L., T.J.) and Department of Clinical Experimental Research (A.L.), Glostrup University Hospital, Glostrup, Denmark; Department of
| | - Kristine H. Allin
- From The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics (J.M.J., K.H.A., C.H.S., N.T.K., N.G., T.H., O.P.), Department of Clinical Medicine (A.L.) and Department of Public Health (T.J.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Research Centre for Prevention and Health (A.B., A.L., T.J.) and Department of Clinical Experimental Research (A.L.), Glostrup University Hospital, Glostrup, Denmark; Department of
| | - Camilla H. Sandholt
- From The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics (J.M.J., K.H.A., C.H.S., N.T.K., N.G., T.H., O.P.), Department of Clinical Medicine (A.L.) and Department of Public Health (T.J.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Research Centre for Prevention and Health (A.B., A.L., T.J.) and Department of Clinical Experimental Research (A.L.), Glostrup University Hospital, Glostrup, Denmark; Department of
| | - Anders Borglykke
- From The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics (J.M.J., K.H.A., C.H.S., N.T.K., N.G., T.H., O.P.), Department of Clinical Medicine (A.L.) and Department of Public Health (T.J.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Research Centre for Prevention and Health (A.B., A.L., T.J.) and Department of Clinical Experimental Research (A.L.), Glostrup University Hospital, Glostrup, Denmark; Department of
| | - Nikolaj T. Krarup
- From The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics (J.M.J., K.H.A., C.H.S., N.T.K., N.G., T.H., O.P.), Department of Clinical Medicine (A.L.) and Department of Public Health (T.J.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Research Centre for Prevention and Health (A.B., A.L., T.J.) and Department of Clinical Experimental Research (A.L.), Glostrup University Hospital, Glostrup, Denmark; Department of
| | - Niels Grarup
- From The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics (J.M.J., K.H.A., C.H.S., N.T.K., N.G., T.H., O.P.), Department of Clinical Medicine (A.L.) and Department of Public Health (T.J.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Research Centre for Prevention and Health (A.B., A.L., T.J.) and Department of Clinical Experimental Research (A.L.), Glostrup University Hospital, Glostrup, Denmark; Department of
| | - Allan Linneberg
- From The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics (J.M.J., K.H.A., C.H.S., N.T.K., N.G., T.H., O.P.), Department of Clinical Medicine (A.L.) and Department of Public Health (T.J.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Research Centre for Prevention and Health (A.B., A.L., T.J.) and Department of Clinical Experimental Research (A.L.), Glostrup University Hospital, Glostrup, Denmark; Department of
| | - Torben Jørgensen
- From The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics (J.M.J., K.H.A., C.H.S., N.T.K., N.G., T.H., O.P.), Department of Clinical Medicine (A.L.) and Department of Public Health (T.J.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Research Centre for Prevention and Health (A.B., A.L., T.J.) and Department of Clinical Experimental Research (A.L.), Glostrup University Hospital, Glostrup, Denmark; Department of
| | - Torben Hansen
- From The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics (J.M.J., K.H.A., C.H.S., N.T.K., N.G., T.H., O.P.), Department of Clinical Medicine (A.L.) and Department of Public Health (T.J.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Research Centre for Prevention and Health (A.B., A.L., T.J.) and Department of Clinical Experimental Research (A.L.), Glostrup University Hospital, Glostrup, Denmark; Department of
| | - Oluf Pedersen
- From The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics (J.M.J., K.H.A., C.H.S., N.T.K., N.G., T.H., O.P.), Department of Clinical Medicine (A.L.) and Department of Public Health (T.J.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Research Centre for Prevention and Health (A.B., A.L., T.J.) and Department of Clinical Experimental Research (A.L.), Glostrup University Hospital, Glostrup, Denmark; Department of
| |
Collapse
|
13
|
Sasidhar MV, Reddy S, Naik A, Naik S. Genetics of coronary artery disease - a clinician's perspective. Indian Heart J 2014; 66:663-71. [PMID: 25634402 DOI: 10.1016/j.ihj.2014.12.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 11/21/2014] [Indexed: 02/02/2023] Open
Abstract
Coronary artery disease (CAD) is the major cause of fatality and disability among all cardiovascular diseases (CVD). Intricate interactions of genes and environment dictate the outcomes of CAD. Technological advances in the different fields of genetics including linkage studies (LS), candidate gene studies (CGS) and genome-wide association studies (GWA studies) have augmented the knowledge of pathogenesis of CAD. LS were more successful in identifying genetic variants among monogenic disease. GWA studies were relatively popular in identification of variation in polygenic disease. Until now, GWA studies recognized about 50 loci determining around 6% of the heritability in CAD. Clinical utility of the above knowledge would result in better CAD management, but validation of the variants in native population is warranted for active adoption into the clinic. The major aim of this review is to provide an adequate perspective of our current understanding and advances of genetics in CAD.
Collapse
Affiliation(s)
- Manda V Sasidhar
- Senior Scientist, Apollo Hospitals Educational and Research Foundation (AHERF), Hyderabad, India
| | - Satyajit Reddy
- MD Candidate, Alpert Medical School of Brown University, Providence, USA
| | - Ashutosh Naik
- Honorary Assistant Professor, Srinidhi Institute of Science, and Technology, Hyderabad, India
| | - Sudhir Naik
- Senior Consultant, Cardiology Department, Apollo Hospitals, Hyderabad, India; Adjunct Professor, AHERF, Hyderabad, India.
| |
Collapse
|
14
|
Anderson CD, Rosand J. Genome-wide linkage approach yields novel early onset myocardial infarction locus in East Asians. ACTA ACUST UNITED AC 2014; 6:531-2. [PMID: 24347618 DOI: 10.1161/circgenetics.113.000378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Christopher D Anderson
- Center for Human Genetic Research and Division of Neurocritical Care and Emergency Neurology, Massachusetts General Hospital, Boston, MA
| | | |
Collapse
|
15
|
Abstract
Heart failure explains a large portion of heart diseases. Molecular mechanisms determining cardiac function, by inference dysfunction in heart failure, are incompletely understood, especially in the common (or congestive) systolic (SHF) and diastolic heart failure (DHF). Limited genome-wide association studies (GWASs) in humans are reported on SHF and no GWAS has been performed on DHF. Genetic analyses in a rodent model of true DHF, Dahl salt-sensitive (DSS) rats, have begun to unravel the genetic components determining diastolic function. Diastolic dysfunction of DSS rats can be ameliorated or even normalized by distinct quantitative trait loci (QTLs), designated as diastolic function/blood pressure QTLs (DF/BP QTLs), which also affect blood pressure (BP). However, an improvement in diastolic dysfunction is merely transitory from a single DF/BP QTL, despite a permanent lowering of BP. A long-term protection against diastolic dysfunction can be realized only through combining specific DF/BP QTLs. Moreover, the worsening diastolic dysfunction with age can also be reversed in a different combination of DF/BP QTLs. Thus, distinct genes in combinations must be involved in the physiological mechanisms ameliorating or reversing diastolic dysfunction. As not all the QTLs that influence BP can affect diastolic function, it is not BP reduction itself that restores diastolic function, but rather specific genes that are uniquely integrated into the pathways of blood pressure homeostasis as well as diastolic function. Thus, the elucidation of pathophysiological mechanisms causal to hypertensive diastolic dysfunction will not only provide new diagnostic tools, but also novel therapeutic targets and strategies in reducing, curing, and even reversing DHF.
Collapse
|
16
|
İzmirli M, Göktekin Ö, Bacaksız A, Uysal Ö, Kılıç Ü. The effect of the SIRT1 2827 A>G polymorphism, resveratrol, exercise, age and occupation in Turkish population with cardiovascular disease. Anatol J Cardiol 2014; 15:103-6. [PMID: 25252293 PMCID: PMC5336992 DOI: 10.5152/akd.2014.5214] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Objective: Cardiovascular disease (CVD) is the leading cause of death in Europe. One of the candidate molecule affecting epigenetic mechanisms of CVD is the SIRT1, a subclass of sirtuins, is located on the long arm of chromosome 10 (10q21.3). Particularly, the relation between 2827 A>G polymorphism of the SIRT1 positioned on exon 2, leading to conversion of histidine to arginine, and the formation of CVD is not known yet. One of the activator of SIRT1, resveratrol, is also known as a cardioprotective molecule. On the other hand, the parameters including exercise, occupation and age affect CVD. The aim of the present study was to investigate the effect of the rs144124002 (2827 A>G) single nucleotide polymorphisms (SNP) of SIRT1 and exercise-occupation-age parameters on CVD. Methods: SNP of SIRT1 were analyzed using DNA isolation, the polymerase chain reaction (PCR) and restriction fragment length polymorphism. To do so, large cohorts of CVD patients (n=293) and healthy controls (n=117) who directed Cardiology Department of Bezmialem Vakıf University, Bezmialem Vakıf University Hospital were used. Results: In this study, when we assessed CVD and control groups about 2827 A>G polymorphism, all individuals were determined as homozygous genotype. We found a positive effect between the modifications of resveratrol, exercise, age and occupation and CVD (OR=0.17; CI 95%, 0.1-0.2; p≤0.001). Conclusion: This is the first study demonstrating the correlation between the SIRT1 rs144124002 polymorphism and CVD in Turkish population.
Collapse
Affiliation(s)
- Müzeyyen İzmirli
- Department of Medical Biology, Faculty of Medicine, Bezmialem Vakıf University; İstanbul-Turkey; Department of Medical Biology, Faculty of Medicine, Mustafa Kemal University; Hatay-Turkey.
| | | | | | | | | |
Collapse
|
17
|
Kilic U, Gok O, Bacaksiz A, Izmirli M, Elibol-Can B, Uysal O. SIRT1 gene polymorphisms affect the protein expression in cardiovascular diseases. PLoS One 2014; 9:e90428. [PMID: 24587358 PMCID: PMC3938716 DOI: 10.1371/journal.pone.0090428] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 01/29/2014] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease (CVD), the leading cause of death worldwide, is related to gene-environment interactions due to epigenetic factors. SIRT1 protein and its downstream pathways are critical for both normal homeostasis and protection from CVD-induced defects. The aim of this study was to investigate the association between SIRT1 single nucleotide polymorphisms (SNPs) (rs7895833 A>G in the promoter region, rs7069102 C>G in intron 4 and rs2273773 C>T in exon 5 silent mutation) and SIRT1 and eNOS (endothelial nitric oxide synthase) protein expression as well as total antioxidant status (TAS), total oxidant status (TOS) and oxidative stress index (OSI) in CVD patients as compared to controls. The frequencies of mutant genotypes and alleles for rs7069102 and rs2273773 were significantly higher in patients with CVD compared to control group. The risk for CVD was increased by 2.4 times for rs7069102 and 1.9 times for rs2273773 in carriers of mutant allele compared with carriers of wild-type allele pointing the protective role of C allele for both SNPs against CVD. For rs7895833, there was no significant difference in genotype and allele distributions between groups. SIRT1 protein, TAS, TOS and OSI levels significantly increased in patients as compared to control group. In contrast, level of eNOS protein was considerably low in the CVD patients. An increase in the SIRT1 expression in the CVD patients carrying mutant genotype for rs7069102 and heterozygote genotype for all three SNPs was observed. This is the first study reporting an association between SIRT1 gene polymorphisms and the levels of SIRT1 and eNOS expressions as well as TAS, TOS and OSI.
Collapse
Affiliation(s)
- Ulkan Kilic
- Department of Medical Biology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
- * E-mail:
| | - Ozlem Gok
- Department of Medical Biology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Ahmet Bacaksiz
- Department of Cardiology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Muzeyyen Izmirli
- Department of Medical Biology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
- Department of Medical Biology, Faculty of Medicine, Mustafa Kemal University, Hatay, Turkey
| | - Birsen Elibol-Can
- Department of Medical Biology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Omer Uysal
- Department of Biostatistics, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| |
Collapse
|
18
|
Biopsychosocial Determinants of Health and Quality of life Among Young Women with Coronary Heart Disease. CURRENT CARDIOVASCULAR RISK REPORTS 2013. [DOI: 10.1007/s12170-013-0366-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
19
|
Ganesh SK, Arnett DK, Assimes TL, Basson CT, Chakravarti A, Ellinor PT, Engler MB, Goldmuntz E, Herrington DM, Hershberger RE, Hong Y, Johnson JA, Kittner SJ, McDermott DA, Meschia JF, Mestroni L, O’Donnell CJ, Psaty BM, Vasan RS, Ruel M, Shen WK, Terzic A, Waldman SA. Genetics and Genomics for the Prevention and Treatment of Cardiovascular Disease: Update. Circulation 2013; 128:2813-51. [DOI: 10.1161/01.cir.0000437913.98912.1d] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
20
|
Tikkanen E, Havulinna AS, Palotie A, Salomaa V, Ripatti S. Genetic risk prediction and a 2-stage risk screening strategy for coronary heart disease. Arterioscler Thromb Vasc Biol 2013; 33:2261-6. [PMID: 23599444 PMCID: PMC4210840 DOI: 10.1161/atvbaha.112.301120] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 04/01/2013] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Genome-wide association studies have identified several genetic variants associated with coronary heart disease (CHD). The aim of this study was to evaluate the genetic risk discrimination and reclassification and apply the results for a 2-stage population risk screening strategy for CHD. APPROACH AND RESULTS We genotyped 28 genetic variants in 24 124 participants in 4 Finnish population-based, prospective cohorts (recruitment years 1992-2002). We constructed a multilocus genetic risk score and evaluated its association with incident cardiovascular disease events. During the median follow-up time of 12 years (interquartile range 8.75-15.25 years), we observed 1093 CHD, 1552 cardiovascular disease, and 731 acute coronary syndrome events. Adding genetic information to conventional risk factors and family history improved risk discrimination of CHD (C-index 0.856 versus 0.851; P=0.0002) and other end points (cardiovascular disease: C-index 0.840 versus 0.837, P=0.0004; acute coronary syndrome: C-index 0.859 versus 0.855, P=0.001). In a standard population of 100 000 individuals, additional genetic screening of subjects at intermediate risk for CHD would reclassify 2144 subjects (12%) into high-risk category. Statin allocation for these subjects is estimated to prevent 135 CHD cases over 14 years. Similar results were obtained by external validation, where the effects were estimated from a training data set and applied for a test data set. CONCLUSIONS Genetic risk score improves risk prediction of CHD and helps to identify individuals at high risk for the first CHD event. Genetic screening for individuals at intermediate cardiovascular risk could help to prevent future cases through better targeting of statins.
Collapse
Affiliation(s)
- Emmi Tikkanen
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | | | | | | | | |
Collapse
|
21
|
Agirbasli M, Eren F, Agirbasli D, White MJ, Williams SM. Multi-locus candidate gene analyses of lipid levels in a pediatric Turkish cohort: lessons learned on LPL, CETP, LIPC, ABCA1, and SHBG. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2013; 17:636-45. [PMID: 23988150 DOI: 10.1089/omi.2013.0066] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cardiovascular risk factors and atherosclerosis precursors were examined in 365 Turkish children and adolescents. Study participants were recruited at five different state schools. We tested single and multi-locus effects of six polymorphisms from five candidate genes, chosen based on prior known association with lipid levels in adults, for association with low (≤10th percentile) high density lipoprotein cholesterol (HDL-C) and high (≥90th percentile) triglycerides (TG), and the related continuous outcomes. We observed an association between CETP variant rs708272 and low HDL-C (allelic p=0.020, genotypic p=0.046), which was supported by an independent analysis, PRAT (PRAT control p=0.027). Sex-stratified logistic regression analysis showed that the B2 allele of rs708272 decreased odds of being in the lower tenth percentile of HDL-C measurements (OR=0.36, p=0.02) in girls; this direction of effect was also seen in boys but was not significant (OR=0.64, p=0.21). Logistic regression analysis also revealed that the T allele of rs6257 (SHBG) decreased odds of being in the top tenth percentile of TG measurements in boys (OR=0.43, p=0.03). Analysis of lipid levels as a continuous trait revealed a significant association between rs708272 (CETP) and LDL-C levels in males (p=0.02) with the B2B2 genotype group having the lowest mean LDL-C; the same direction of effect was also seen in females (p=0.05). An effect was also seen between rs708272 and HDL-C levels in girls (p=0.01), with the B2B2 genotype having the highest mean HDL-C levels. Multi-locus analysis, using quantitative multifactor dimensionality reduction (qMDR) identified the previously mentioned CETP variant as the best single locus model, and overall model, for predicting HDL-C levels in children. This study provides evidence for association between CETP and low HDL-C phenotype in children, but the results appear to be weaker in children than previous results in adults and may also be subject to gender effects.
Collapse
Affiliation(s)
- Mehmet Agirbasli
- 1 Department of Cardiology, Marmara University School of Medicine , Istanbul, Turkey
| | | | | | | | | |
Collapse
|
22
|
Gene-smoking interactions in multiple Rho-GTPase pathway genes in an early-onset coronary artery disease cohort. Hum Genet 2013; 132:1371-82. [PMID: 23907653 DOI: 10.1007/s00439-013-1339-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 07/14/2013] [Indexed: 10/26/2022]
Abstract
We performed a gene-smoking interaction analysis using families from an early-onset coronary artery disease cohort (GENECARD). This analysis was focused on validating and expanding results from previous studies implicating single nucleotide polymorphisms (SNPs) on chromosome 3 in smoking-mediated coronary artery disease. We analyzed 430 SNPs on chromosome 3 and identified 16 SNPs that showed a gene-smoking interaction at P < 0.05 using association in the presence of linkage--ordered subset analysis, a method that uses permutations of the data to empirically estimate the strength of the association signal. Seven of the 16 SNPs were in the Rho-GTPase pathway indicating a 1.87-fold enrichment for this pathway. A meta-analysis of gene-smoking interactions in three independent studies revealed that rs9289231 in KALRN had a Fisher's combined P value of 0.0017 for the interaction with smoking. In a gene-based meta-analysis KALRN had a P value of 0.026. Finally, a pathway-based analysis of the association results using WebGestalt revealed several enriched pathways including the regulation of the actin cytoskeleton pathway as defined by the Kyoto Encyclopedia of Genes and Genomes.
Collapse
|
23
|
Ding SL, Wang JX, Jiao JQ, Tu X, Wang Q, Liu F, Li Q, Gao J, Zhou QY, Gu DF, Li PF. A pre-microRNA-149 (miR-149) genetic variation affects miR-149 maturation and its ability to regulate the Puma protein in apoptosis. J Biol Chem 2013; 288:26865-77. [PMID: 23873935 DOI: 10.1074/jbc.m112.440453] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
MicroRNAs (miRNAs) are small, single-stranded, noncoding RNAs that function as negative regulators of gene expression. They are transcribed from endogenous DNA and form hairpin structures (termed as pre-miRNAs) that are processed to form mature miRNAs. It remains largely unknown as to the molecular consequences of the natural genetic variation in pre-miRNAs. Here, we report that an A→G polymorphism (rs71428439) is located in Homo sapiens miR-149 stem-loop region. This polymorphism results in a change in the structure of the miR-149 precursor. Our results showed that the genotype distribution of this polymorphism in myocardial infarction cases was significantly different from that in the control subjects. We examined the biological significance of this polymorphism on the production of mature miR-149, and we observed that the G-allelic miR-149 precursor displayed a lower production of mature miR-149 compared with the A-allelic one. Further investigations disclosed that miR-149 could withstand mitochondrial fission and apoptosis through targeting the pro-apoptotic factor p53-up-regulated modulator of apoptosis (Puma). Enforced expression of miR-149 promoted cell survival, whereas knockdown of miR-149 rendered cells to be sensitive to apoptotic stimulation. Intriguingly, the A to G variation led pre-miR-149 to elicit an attenuated effect on the inhibition of mitochondrial fission and apoptosis. Finally, this polymorphism exerts its influence on cardiac function in the mouse model of myocardial infarction. These data suggest that this polymorphism in the miR-149 precursor may result in important phenotypic traits of myocardial infarction. Our findings warrant further investigations on the relationship between miR-149 polymorphism and myocardial infarction.
Collapse
Affiliation(s)
- Su-Ling Ding
- From the Division of Cardiovascular Research, State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Wojczynski MK, Li M, Bielak LF, Kerr KF, Reiner AP, Wong ND, Yanek LR, Qu L, White CC, Lange LA, Ferguson JF, He J, Young T, Mosley TH, Smith JA, Kral BG, Guo X, Wong Q, Ganesh SK, Heckbert SR, Griswold ME, O'Leary DH, Budoff M, Carr JJ, Taylor HA, Bluemke DA, Demissie S, Hwang SJ, Paltoo DN, Polak JF, Psaty BM, Becker DM, Province MA, Post WS, O'Donnell CJ, Wilson JG, Harris TB, Kavousi M, Cupples LA, Rotter JI, Fornage M, Becker LC, Peyser PA, Borecki IB, Reilly MP. Genetics of coronary artery calcification among African Americans, a meta-analysis. BMC MEDICAL GENETICS 2013; 14:75. [PMID: 23870195 PMCID: PMC3733595 DOI: 10.1186/1471-2350-14-75] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 07/18/2013] [Indexed: 02/02/2023]
Abstract
Background Coronary heart disease (CHD) is the major cause of death in the United States. Coronary artery calcification (CAC) scores are independent predictors of CHD. African Americans (AA) have higher rates of CHD but are less well-studied in genomic studies. We assembled the largest AA data resource currently available with measured CAC to identify associated genetic variants. Methods We analyzed log transformed CAC quantity (ln(CAC + 1)), for association with ~2.5 million single nucleotide polymorphisms (SNPs) and performed an inverse-variance weighted meta-analysis on results for 5,823 AA from 8 studies. Heritability was calculated using family studies. The most significant SNPs among AAs were evaluated in European Ancestry (EA) CAC data; conversely, the significance of published SNPs for CAC/CHD in EA was queried within our AA meta-analysis. Results Heritability of CAC was lower in AA (~30%) than previously reported for EA (~50%). No SNP reached genome wide significance (p < 5E-08). Of 67 SNPs with p < 1E-05 in AA there was no evidence of association in EA CAC data. Four SNPs in regions previously implicated in CAC/CHD (at 9p21 and PHACTR1) in EA reached nominal significance for CAC in AA, with concordant direction. Among AA, rs16905644 (p = 4.08E-05) had the strongest association in the 9p21 region. Conclusions While we observed substantial heritability for CAC in AA, we failed to identify loci for CAC at genome-wide significant levels despite having adequate power to detect alleles with moderate to large effects. Although suggestive signals in AA were apparent at 9p21 and additional CAC and CAD EA loci, overall the data suggest that even larger samples and an ethnic specific focus will be required for GWAS discoveries for CAC in AA populations.
Collapse
Affiliation(s)
- Mary K Wojczynski
- Department of Genetics, Division of Statistical Genomics, Washington University School of Medicine, St. Louis, MO, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Xie F, Song Q. Association of the G1359A Polymorphism in the CNR1Gene with the Presence of Coronary Artery Disease in Patients with Metabolic Syndrome. Lab Med 2013. [DOI: 10.1309/lmwjisddz1ol8u7f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
26
|
Farhan SM, Hegele RA. Genetics 101 for Cardiologists: Rare Genetic Variants and Monogenic Cardiovascular Disease. Can J Cardiol 2013. [DOI: 10.1016/j.cjca.2012.10.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
27
|
Stender S, Frikke-Schmidt R, Nordestgaard BG, Grande P, Tybjaerg-Hansen A. Genetically elevated bilirubin and risk of ischaemic heart disease: three Mendelian randomization studies and a meta-analysis. J Intern Med 2013; 273:59-68. [PMID: 22805420 DOI: 10.1111/j.1365-2796.2012.02576.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Elevated plasma levels of bilirubin, an endogenous antioxidant, have been associated with reduced risk of ischaemic heart disease (IHD) and myocardial infarction (MI). Whether this is a causal relationship remains unclear. OBJECTIVE We tested the hypothesis that elevated plasma bilirubin is causally related to decreased risk of IHD and MI. DESIGN We used a Mendelian randomization approach and three independent studies from Copenhagen, Denmark. We measured bilirubin in 43 708 white individuals from the general population, and genotyped rs6742078 G>T in the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene in 67 068 individuals, of whom 11 686 had IHD. RESULTS Third versus first tertile of baseline bilirubin levels was associated with 134% increased bilirubin levels, with sex- and age-adjusted hazard ratios (HRs) of 0.86 [95% confidence interval (CI), 0.76-0.98; P = 0.02] for IHD and 0.81 (95% CI, 0.66-0.99; P = 0.04) for MI, but with corresponding multifactorially adjusted HRs of 0.93 (95% CI, 0.82-1.06; P = 0.29) and 0.90 (95% CI, 0.73-1.12; P = 0.35). UGT1A1 rs6742078 TT versus GG genotype was associated with 95% increased bilirubin levels (P < 0.001); TT versus GG genotype was associated with odds ratios (ORs) of 1.03 (95% CI, 0.96-1.11; P = 0.73) for IHD and 1.01 (95% CI, 0.92-1.12; P = 0.68) for MI. Finally, in a meta-analysis of the present three studies and eight previous studies including a total of 14 711 cases and 60 324 controls, the random effects OR for ischaemic cardiovascular disease for genotypes with approximately 100% increased bilirubin levels versus reference genotypes was 1.01 (95% CI, 0.88-1.16). CONCLUSION These data suggest that plasma bilirubin is not causally associated with risk of IHD.
Collapse
Affiliation(s)
- S Stender
- Department of Clinical Biochemistry, Copenhagen University, Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
28
|
Leeper NJ, Raiesdana A, Kojima Y, Kundu RK, Cheng H, Maegdefessel L, Toh R, Ahn GO, Ali ZA, Anderson DR, Miller CL, Roberts SC, Spin JM, de Almeida PE, Wu JC, Xu B, Cheng K, Quertermous M, Kundu S, Kortekaas KE, Berzin E, Downing KP, Dalman RL, Tsao PS, Schadt EE, Owens GK, Quertermous T. Loss of CDKN2B promotes p53-dependent smooth muscle cell apoptosis and aneurysm formation. Arterioscler Thromb Vasc Biol 2012; 33:e1-e10. [PMID: 23162013 DOI: 10.1161/atvbaha.112.300399] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Genomewide association studies have implicated allelic variation at 9p21.3 in multiple forms of vascular disease, including atherosclerotic coronary heart disease and abdominal aortic aneurysm. As for other genes at 9p21.3, human expression quantitative trait locus studies have associated expression of the tumor suppressor gene CDKN2B with the risk haplotype, but its potential role in vascular pathobiology remains unclear. METHODS AND RESULTS Here we used vascular injury models and found that Cdkn2b knockout mice displayed the expected increase in proliferation after injury, but developed reduced neointimal lesions and larger aortic aneurysms. In situ and in vitro studies suggested that these effects were attributable to increased smooth muscle cell apoptosis. Adoptive bone marrow transplant studies confirmed that the observed effects of Cdkn2b were mediated through intrinsic vascular cells and were not dependent on bone marrow-derived inflammatory cells. Mechanistic studies suggested that the observed increase in apoptosis was attributable to a reduction in MDM2 and an increase in p53 signaling, possibly due in part to compensation by other genes at the 9p21.3 locus. Dual inhibition of both Cdkn2b and p53 led to a reversal of the vascular phenotype in each model. CONCLUSIONS These results suggest that reduced CDKN2B expression and increased smooth muscle cell apoptosis may be one mechanism underlying the 9p21.3 association with aneurysmal disease.
Collapse
Affiliation(s)
- Nicholas J Leeper
- Department of Surgery, Stanford University, 300 Pasteur Dr, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
In the primary prevention of cardiovascular disease, the study of biomarkers to identify at-risk individuals is an expanding field. Several developments have fueled this trend, including improved understanding of the pathophysiological processes underlying atherosclerosis, advances in imaging technology to enable the quantification of subclinical disease burden, and the identification of new genetic susceptibility variants for cardiovascular disease. Furthermore, the advent of high-throughput platforms for molecular profiling has increased the pace of biomarker discovery. The rising interest in biomarkers has been balanced by the recognition that standardized and rigorous statistical approaches are needed to evaluate the clinical utility of candidate risk markers. This article reviews the issues surrounding the evaluation of biomarkers, evidence from studies of existing biomarkers, and recent applications of biomarker discovery platforms.
Collapse
Affiliation(s)
- Yin Ge
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | |
Collapse
|
30
|
Abstract
Transcriptomics is the study of how our genes are regulated and expressed in different biological settings. Technical advances now enable quantitative assessment of all expressed genes (ie, the entire "transcriptome") in a given tissue at a given time. These approaches provide a powerful tool for understanding complex biological systems and for developing novel biomarkers. This chapter will introduce basic concepts in transcriptomics and available technologies for developing transcriptomic biomarkers. We will then review current and emerging applications in cardiovascular medicine.
Collapse
Affiliation(s)
- Dawn M Pedrotty
- Penn Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | |
Collapse
|
31
|
Cui Y, Wang H, Chen H, Pang S, Wang L, Liu D, Yan B. Genetic analysis of the SIRT1 gene promoter in myocardial infarction. Biochem Biophys Res Commun 2012; 426:232-6. [PMID: 22935421 DOI: 10.1016/j.bbrc.2012.08.071] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 08/15/2012] [Indexed: 12/13/2022]
Abstract
Myocardial infarction (MI) is a restrictive phenotype of coronary artery disease. To date, a group of genes and genetic loci have been associated to MI. However, the genetic causes and underlying molecular mechanisms for MI remain largely unknown. SIRT1, one of highly conserved NAD-dependent class III deacetylases, has been involved in several cellular processes and implicated in human diseases. Autophagy is one of major cellular degradative pathways, which plays important roles in lipid metabolism. Recent studies have shown that SIRT1 deacetylates autophagy-related genes, and the expressions of autophagic genes are altered in MI patients. Accordingly, we hypothesized that SIRT1 may be linked to the MI pathogenesis. In this study, the SIRT1 gene promoter were genetically analyzed in large cohorts of MI patients (n = 327) and controls (n = 358). The results showed that six single-nucleotide polymorphisms and 14 sequence variants were identified. Among these, five novel heterozygous variants (g.69643743Ins, g.69643840Ins, g.69643903G > C, g.69644235G > C and g.69644353G > T) and one single-nucleotide polymorphism (rs35706870) were identified in MI patients, but in none of controls. Moreover, five novel heterozygous variants (g.69643672G > A, g.69644226C > T, g.69644278A > G, g.69644408G > A and g.69644408G > T) were only found in controls. The rest variants were found in MI patients and controls with similar frequencies. Taken together, the variants identified in MI patients may alter the transcriptional activities of SIRT1 gene promoter, which may change SIRT1 levels, contributing to the MI pathogenesis as a risk factor.
Collapse
Affiliation(s)
- Yinghua Cui
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Jining Medical University Affiliated Hospital, Jining Medical University, Jining, Shandong 272029, China
| | | | | | | | | | | | | |
Collapse
|
32
|
Referral, Enrollment, and Delivery of Cardiac Rehabilitation for Women. CURRENT CARDIOVASCULAR RISK REPORTS 2012. [DOI: 10.1007/s12170-012-0255-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
33
|
Heckman MG, Soto-Ortolaza AI, Diehl NN, Rayaprolu S, Brott TG, Wszolek ZK, Meschia JF, Ross OA. Genetic variants associated with myocardial infarction in the PSMA6 gene and Chr9p21 are also associated with ischaemic stroke. Eur J Neurol 2012; 20:300-8. [PMID: 22882272 DOI: 10.1111/j.1468-1331.2012.03846.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 07/05/2012] [Indexed: 11/30/2022]
Abstract
BACKGROUND Ischaemic stroke shares common traditional risk factors with coronary artery disease (CAD) and myocardial infarction (MI). This study evaluated whether genetic risk factors for CAD and MI also affect susceptibility to ischaemic stroke in Caucasians and African Americans. METHODS Included in the study were a Caucasian series (713 ischaemic stroke patients, 708 controls) and a small African American series (166 ischaemic stroke patients, 117 controls). Twenty single-nucleotide polymorphisms (SNPs) previously shown to be associated with CAD or MI were genotyped and assessed for association with ischaemic stroke and ischaemic stroke subtypes using odds ratios (ORs) from multivariable logistic regression models. RESULTS In Caucasians, four SNPs on chromosome 9p21 were significantly associated with risk of cardioembolic stroke, the strongest of which was rs1333040 (OR 1.55, P = 0.0007); similar but weaker trends were observed for small vessel stroke, with no associations observed regarding large vessel stroke. Chromosome 9p21 SNPs were also associated with risk of ischaemic stroke in African Americans (rs1333040, OR 0.65, P = 0.023; rs1333042, OR 0.55, P = 0.070; rs2383207, OR 0.55, P = 0.070). The PSMA6 SNP rs1048990 on chromosome 14q13 was associated with overall ischaemic stroke in both Caucasians (OR 0.80, P = 0.036) and African Americans (OR 0.31, P = 0.020). CONCLUSIONS Our results provide evidence that chromosome 9p21 variants are associated with cardioembolic ischaemic stroke in Caucasians and with overall ischaemic stroke in African Americans. The PSMA6 variant rs1048990 also appears to affect susceptibility to ischaemic stroke in both populations. These findings require validation, particularly the preliminary findings regarding African Americans given the small size of that series.
Collapse
Affiliation(s)
- M G Heckman
- Biostatistics Unit, Mayo Clinic, Jacksonville, FL 32224, USA.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Cardiovascular disease encompasses a range of conditions extending from myocardial infarction to congenital heart disease, most of which are heritable. Enormous effort has been invested in understanding the genes and specific DNA sequence variants that are responsible for this heritability. Here, we review the lessons learned for monogenic and common, complex forms of cardiovascular disease. We also discuss key challenges that remain for gene discovery and for moving from genomic localization to mechanistic insights, with an emphasis on the impact of next-generation sequencing and the use of pluripotent human cells to understand the mechanism by which genetic variation contributes to disease.
Collapse
Affiliation(s)
- Sekar Kathiresan
- Center for Human Genetic Research and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA.
| | | |
Collapse
|
35
|
Brown WV, Breslow J, Ballantyne C. Clinical use of genetic typing in human lipid disorders†. J Clin Lipidol 2012; 6:199-207. [DOI: 10.1016/j.jacl.2012.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 02/09/2012] [Accepted: 02/13/2012] [Indexed: 10/28/2022]
|
36
|
Timpson NJ, Wade KH, Smith GD. Mendelian randomization: application to cardiovascular disease. Curr Hypertens Rep 2012; 14:29-37. [PMID: 22161218 DOI: 10.1007/s11906-011-0242-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In the absence of an ethical, practical, and economical randomized trial, the epidemiologist is left to explore other methods in efforts to assert causality. An approach based on genotypic variation has the potential to mitigate against some of the problems found within conventional observational studies. Genetic variations associated with risk factors of interest at the population level can be used as proxy measures for these risk factors and to generate estimates of causal effect. The potential and the possible limitations of this approach within the cardiovascular field are presented in this review.
Collapse
Affiliation(s)
- Nicholas J Timpson
- MRC CAiTE Centre, School of Social and Community Medicine, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK.
| | | | | |
Collapse
|
37
|
Lusis AJ. Genetics of atherosclerosis. Trends Genet 2012; 28:267-75. [PMID: 22480919 DOI: 10.1016/j.tig.2012.03.001] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 02/29/2012] [Accepted: 03/01/2012] [Indexed: 12/13/2022]
Abstract
Genome-wide association studies (GWAS) from the past several years have provided the first unbiased evidence of the genes contributing to common cardiovascular disease traits in European and some Asian populations. The results not only confirmed the importance of prior knowledge, such as the central role of lipoproteins, but also revealed that there is still much to learn about the underlying mechanisms of this disease, as most of the associated genes do not appear to be involved in pathways previously connected to atherosclerosis. In this review, I focus on the common forms of the disease and look at both human and animal model studies. I summarize what was known before GWAS, highlight how the field has been changed by GWAS, and discuss future considerations, such as the limitations of GWAS and strategies that may lead to a more complete, mechanistic understanding of atherosclerosis.
Collapse
Affiliation(s)
- Aldons J Lusis
- University of California, Los Angeles, Department of Medicine/Division of Cardiology, Los Angeles, CA 90095-1679, USA.
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW To discuss if and how the combined analysis of large-scale datasets from multiple independent sources benefits the mapping of novel genetic elements with relevance to lipoprotein metabolism and allows for conclusions on underlying molecular mechanisms. RECENT FINDINGS Genome-wide association studies (GWAS) have identified numerous genomic loci associated with plasma lipid levels and cardiovascular disease. Yet, despite being highly successful in mapping novel loci the GWAS approach falls short to systematically extract functional information from genomic data. With the aim to complement GWAS for a better insight into disease mechanisms and identification of the most promising targets for drug development, a number of high-throughput functional genomics strategies have now been applied. These include computational approaches, consideration of gene-gene and gene-environment interactions, as well as unbiased gene-expression analyses in relevant tissues. For a limited number of loci, mechanistic insight has been gained through in-vitro and in-vivo studies by knockdown and overexpression of candidate genes. SUMMARY The integration of GWAS data with existing functional genomics strategies has contributed to ascertain the relevance of a number of novel factors for lipoprotein biology and disease. However, technologies are warranted that provide a more systematic insight into the molecular function and pathogenic relevance of promising candidate genes.
Collapse
Affiliation(s)
- Heiko Runz
- Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
39
|
Young EH, Papamarkou T, Wainwright NWJ, Sandhu MS. Genetic determinants of lipid homeostasis. Best Pract Res Clin Endocrinol Metab 2012; 26:203-9. [PMID: 22498249 DOI: 10.1016/j.beem.2011.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Circulating levels of blood lipids are heritable risk factors for atherosclerosis and heart disease, and are the target of therapeutic intervention. Studies of monogenic disorders and - more recently - genome-wide association studies have identified several important genetic determinants of blood lipid levels. These have the potential to provide new drug targets to alter blood lipid levels and may improve prediction of cardiovascular disease. Better functional validation of lipid loci is required to clarify the biological role of proteins encoded by specific genomic regions and understand how they influence lipid metabolism and confer disease risk.
Collapse
Affiliation(s)
- Elizabeth H Young
- Genetic Epidemiology Group, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK.
| | | | | | | |
Collapse
|
40
|
Variant within CELSR2/PSRC1/SORT1, but not within CILP2/PBX4, PCSK9 and APOB genes, has a potential to influence statin treatment efficacy. J Appl Biomed 2012. [DOI: 10.2478/v10136-012-0001-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
41
|
Narula N, Rapezzi C, Tavazzi L, Arbustini E. "My parents died of myocardial infarction: is that my destiny?". Med Clin North Am 2012; 96:67-86. [PMID: 22391252 DOI: 10.1016/j.mcna.2011.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
This article presents an overview of clinical and molecular genetics of myocardial infarction (MI). Discussion includes the partial overlapping of risk factors for myocardial infarction and atherosclerosis, the impact of a positive family history on the risk of MI, the "familial" nongenetic, environmental factors, the inherited risk associated with the low-dose input of many genes, and a simple approach to stratify the individual risk in genetic counseling.
Collapse
Affiliation(s)
- Nupoor Narula
- Centre for Inherited Cardiovascular Diseases, Foundation IRCCS Policlinico San Matteo, P.le Golgi n. 19 27100 Pavia, Italy
| | | | | | | |
Collapse
|
42
|
Tikkanen E, Tuovinen T, Widén E, Lehtimäki T, Viikari J, Kähönen M, Peltonen L, Raitakari OT, Ripatti S. Association of Known Loci With Lipid Levels Among Children and Prediction of Dyslipidemia in Adults. ACTA ACUST UNITED AC 2011; 4:673-80. [DOI: 10.1161/circgenetics.111.960369] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Recent genome-wide association studies have found 95 distinct genetic loci associated with high-density (HDL-C) and low-density (LDL-C) lipoprotein cholesterol, total cholesterol (TC), and triglycerides (TG), using adult samples. It is not known if these variants are associated with lipid levels in children and adolescents and if the genetic risk score (GRS), based on these variants, could improve adulthood dyslipidemia prediction over the childhood lipid measurements.
Methods and Results—
We used 2443 participants of the Cardiovascular Risk in Young Finns study cohort with up to 5 measurements of serum lipids taken between ages 3 and 45 years to estimate the effect of individual single-nucleotide polymorphisms and the GRS on lipids. The GRSs were strongly associated with lipids in all age groups (1.5×10
−20
<
P
<8.7×10
−12
for HDL-C, 3.5×10
−27
<
P
<5.6×10
−09
for LDL-C, 2.0×10
−25
<
P
<5.2×10
−09
for TC, and 4.1×10
−20
<
P
<8.4×10
−05
for TG). Jointly, the lipid loci explained 11.8–26.7% of the total variance in lipids among 3- to 6-year-old children, and the proportion dropped over age, except for TG. The discrimination of adult hypertriglyceridemia improved when GRS was added to childhood lipid measurement (
C
statistic=0.04,
P
=0.01).
Conclusions—
Previously identified lipid loci are associated with lipid levels in children and adolescents and explain up to more than 2 times of the lipid variation in children compared with adults. The TG-GRS improves the risk discrimination over childhood lipid measurement for adult hypertriglyceridemia.
Collapse
Affiliation(s)
- Emmi Tikkanen
- From the Institute for Molecular Medicine, Finland FIMM, University of Helsinki, Helsinki, Finland (E.T., T.T., E.W., L.P., S.R.); National Institute for Health and Welfare, Helsinki, Finland (E.T., T.T., L.P., S.R.); the Department of Clinical Chemistry, Tampere University Hospital and University of Tampere, Tampere, Finland (T.L.); the Department of Medicine, University of Turku and Turku University Hospital, Turku, Finland (J.V.); the Department of Clinical Physiology, Tampere University Hospital
| | - Tarja Tuovinen
- From the Institute for Molecular Medicine, Finland FIMM, University of Helsinki, Helsinki, Finland (E.T., T.T., E.W., L.P., S.R.); National Institute for Health and Welfare, Helsinki, Finland (E.T., T.T., L.P., S.R.); the Department of Clinical Chemistry, Tampere University Hospital and University of Tampere, Tampere, Finland (T.L.); the Department of Medicine, University of Turku and Turku University Hospital, Turku, Finland (J.V.); the Department of Clinical Physiology, Tampere University Hospital
| | - Elisabeth Widén
- From the Institute for Molecular Medicine, Finland FIMM, University of Helsinki, Helsinki, Finland (E.T., T.T., E.W., L.P., S.R.); National Institute for Health and Welfare, Helsinki, Finland (E.T., T.T., L.P., S.R.); the Department of Clinical Chemistry, Tampere University Hospital and University of Tampere, Tampere, Finland (T.L.); the Department of Medicine, University of Turku and Turku University Hospital, Turku, Finland (J.V.); the Department of Clinical Physiology, Tampere University Hospital
| | - Terho Lehtimäki
- From the Institute for Molecular Medicine, Finland FIMM, University of Helsinki, Helsinki, Finland (E.T., T.T., E.W., L.P., S.R.); National Institute for Health and Welfare, Helsinki, Finland (E.T., T.T., L.P., S.R.); the Department of Clinical Chemistry, Tampere University Hospital and University of Tampere, Tampere, Finland (T.L.); the Department of Medicine, University of Turku and Turku University Hospital, Turku, Finland (J.V.); the Department of Clinical Physiology, Tampere University Hospital
| | - Jorma Viikari
- From the Institute for Molecular Medicine, Finland FIMM, University of Helsinki, Helsinki, Finland (E.T., T.T., E.W., L.P., S.R.); National Institute for Health and Welfare, Helsinki, Finland (E.T., T.T., L.P., S.R.); the Department of Clinical Chemistry, Tampere University Hospital and University of Tampere, Tampere, Finland (T.L.); the Department of Medicine, University of Turku and Turku University Hospital, Turku, Finland (J.V.); the Department of Clinical Physiology, Tampere University Hospital
| | - Mika Kähönen
- From the Institute for Molecular Medicine, Finland FIMM, University of Helsinki, Helsinki, Finland (E.T., T.T., E.W., L.P., S.R.); National Institute for Health and Welfare, Helsinki, Finland (E.T., T.T., L.P., S.R.); the Department of Clinical Chemistry, Tampere University Hospital and University of Tampere, Tampere, Finland (T.L.); the Department of Medicine, University of Turku and Turku University Hospital, Turku, Finland (J.V.); the Department of Clinical Physiology, Tampere University Hospital
| | - Leena Peltonen
- From the Institute for Molecular Medicine, Finland FIMM, University of Helsinki, Helsinki, Finland (E.T., T.T., E.W., L.P., S.R.); National Institute for Health and Welfare, Helsinki, Finland (E.T., T.T., L.P., S.R.); the Department of Clinical Chemistry, Tampere University Hospital and University of Tampere, Tampere, Finland (T.L.); the Department of Medicine, University of Turku and Turku University Hospital, Turku, Finland (J.V.); the Department of Clinical Physiology, Tampere University Hospital
| | - Olli T. Raitakari
- From the Institute for Molecular Medicine, Finland FIMM, University of Helsinki, Helsinki, Finland (E.T., T.T., E.W., L.P., S.R.); National Institute for Health and Welfare, Helsinki, Finland (E.T., T.T., L.P., S.R.); the Department of Clinical Chemistry, Tampere University Hospital and University of Tampere, Tampere, Finland (T.L.); the Department of Medicine, University of Turku and Turku University Hospital, Turku, Finland (J.V.); the Department of Clinical Physiology, Tampere University Hospital
| | - Samuli Ripatti
- From the Institute for Molecular Medicine, Finland FIMM, University of Helsinki, Helsinki, Finland (E.T., T.T., E.W., L.P., S.R.); National Institute for Health and Welfare, Helsinki, Finland (E.T., T.T., L.P., S.R.); the Department of Clinical Chemistry, Tampere University Hospital and University of Tampere, Tampere, Finland (T.L.); the Department of Medicine, University of Turku and Turku University Hospital, Turku, Finland (J.V.); the Department of Clinical Physiology, Tampere University Hospital
| |
Collapse
|
43
|
Stender S, Frikke-Schmidt R, Anestis A, Kardassis D, Sethi AA, Nordestgaard BG, Tybjærg-Hansen A. Genetic Variation in Liver X Receptor Alpha and Risk of Ischemic Vascular Disease in the General Population. Arterioscler Thromb Vasc Biol 2011; 31:2990-6. [DOI: 10.1161/atvbaha.111.223867] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Objective—
Although animal studies indicate that
liver X receptor alpha
(
LXR
α) might influence risk of atherosclerosis, data in humans remain scarce. We tested the hypothesis that genetic variation in
LXR
α associates with risk of ischemic vascular disease and/or plasma lipid and lipoprotein levels in the general population.
Methods and Results—
We studied 10,281 white persons of Danish ancestry from a general population cohort, including 1,986 in whom ischemic heart disease (IHD) developed, and 989 in whom ischemic cerebrovascular disease developed. We examined another 51,429 white persons of Danish ancestry from a general population study, including 3,789 with IHD. We genotyped 10 genetic variants identified by resequencing
LXR
α. Homozygosity for −840AA/−115AA(=2.7%) predicted hazard ratios of 1.3 (95% confidence interval, 1.0–1.7) for IHD, 1.6 (1.2–2.2) for myocardial infarction, and 1.7 (1.3–2.4) for ischemic cerebrovascular disease. The corresponding odds ratios in the second cohort were 1.1 (0.9–1.4) for IHD and 1.5 (1.1–2.0) for myocardial infarction. In the combined studies, odds ratios were 1.2 (1.0–1.4) for IHD and 1.5 (1.2–1.9) for myocardial infarction. Homozygosity for −840AA/−115AA did not associate with lipid or lipoprotein levels.
LXR
α −1830T>C (tagging the haplotype −1830C/−840A/−115A, all r
2
≥0.97) associated with 91% increased transcriptional activity.
Conclusion—
This study suggests that functional genetic variation in
LXR
α predicts risk of ischemic vascular disease in the general population.
Collapse
Affiliation(s)
- Stefan Stender
- From the Department of Clinical Biochemistry (S.S., R.F.-S., A.A.S., A.T.-H.), Rigshospitalet, Copenhagen University Hospital and Faculty of Health Sciences, University of Copenhagen, Denmark; Department of Biochemistry (A.A., D.K.), University of Crete Medical School, Heraklion, Greece; Pacific Biometrics, Inc (A.A.S.), Seattle, WA; Department of Clinical Biochemistry (A.A.S., B.G.N.) and The Copenhagen General Population Study (R.F.-S., B.G.N., A.T.-H.), Herlev Hospital, and The Copenhagen City
| | - Ruth Frikke-Schmidt
- From the Department of Clinical Biochemistry (S.S., R.F.-S., A.A.S., A.T.-H.), Rigshospitalet, Copenhagen University Hospital and Faculty of Health Sciences, University of Copenhagen, Denmark; Department of Biochemistry (A.A., D.K.), University of Crete Medical School, Heraklion, Greece; Pacific Biometrics, Inc (A.A.S.), Seattle, WA; Department of Clinical Biochemistry (A.A.S., B.G.N.) and The Copenhagen General Population Study (R.F.-S., B.G.N., A.T.-H.), Herlev Hospital, and The Copenhagen City
| | - Aristomenis Anestis
- From the Department of Clinical Biochemistry (S.S., R.F.-S., A.A.S., A.T.-H.), Rigshospitalet, Copenhagen University Hospital and Faculty of Health Sciences, University of Copenhagen, Denmark; Department of Biochemistry (A.A., D.K.), University of Crete Medical School, Heraklion, Greece; Pacific Biometrics, Inc (A.A.S.), Seattle, WA; Department of Clinical Biochemistry (A.A.S., B.G.N.) and The Copenhagen General Population Study (R.F.-S., B.G.N., A.T.-H.), Herlev Hospital, and The Copenhagen City
| | - Dimitris Kardassis
- From the Department of Clinical Biochemistry (S.S., R.F.-S., A.A.S., A.T.-H.), Rigshospitalet, Copenhagen University Hospital and Faculty of Health Sciences, University of Copenhagen, Denmark; Department of Biochemistry (A.A., D.K.), University of Crete Medical School, Heraklion, Greece; Pacific Biometrics, Inc (A.A.S.), Seattle, WA; Department of Clinical Biochemistry (A.A.S., B.G.N.) and The Copenhagen General Population Study (R.F.-S., B.G.N., A.T.-H.), Herlev Hospital, and The Copenhagen City
| | - Amar A. Sethi
- From the Department of Clinical Biochemistry (S.S., R.F.-S., A.A.S., A.T.-H.), Rigshospitalet, Copenhagen University Hospital and Faculty of Health Sciences, University of Copenhagen, Denmark; Department of Biochemistry (A.A., D.K.), University of Crete Medical School, Heraklion, Greece; Pacific Biometrics, Inc (A.A.S.), Seattle, WA; Department of Clinical Biochemistry (A.A.S., B.G.N.) and The Copenhagen General Population Study (R.F.-S., B.G.N., A.T.-H.), Herlev Hospital, and The Copenhagen City
| | - Børge G. Nordestgaard
- From the Department of Clinical Biochemistry (S.S., R.F.-S., A.A.S., A.T.-H.), Rigshospitalet, Copenhagen University Hospital and Faculty of Health Sciences, University of Copenhagen, Denmark; Department of Biochemistry (A.A., D.K.), University of Crete Medical School, Heraklion, Greece; Pacific Biometrics, Inc (A.A.S.), Seattle, WA; Department of Clinical Biochemistry (A.A.S., B.G.N.) and The Copenhagen General Population Study (R.F.-S., B.G.N., A.T.-H.), Herlev Hospital, and The Copenhagen City
| | - Anne Tybjærg-Hansen
- From the Department of Clinical Biochemistry (S.S., R.F.-S., A.A.S., A.T.-H.), Rigshospitalet, Copenhagen University Hospital and Faculty of Health Sciences, University of Copenhagen, Denmark; Department of Biochemistry (A.A., D.K.), University of Crete Medical School, Heraklion, Greece; Pacific Biometrics, Inc (A.A.S.), Seattle, WA; Department of Clinical Biochemistry (A.A.S., B.G.N.) and The Copenhagen General Population Study (R.F.-S., B.G.N., A.T.-H.), Herlev Hospital, and The Copenhagen City
| |
Collapse
|
44
|
Sivapalaratnam S, Motazacker MM, Maiwald S, Hovingh GK, Kastelein JJP, Levi M, Trip MD, Dallinga-Thie GM. Genome-wide association studies in atherosclerosis. Curr Atheroscler Rep 2011; 13:225-32. [PMID: 21369780 PMCID: PMC3085784 DOI: 10.1007/s11883-011-0173-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease remains the major cause of worldwide morbidity and mortality. Its pathophysiology is complex and multifactorial. Because the phenotype of cardiovascular disease often shows a marked heritable pattern, it is likely that genetic factors play an important role. In recent years, large genome-wide association studies have been conducted to decipher the molecular mechanisms underlying this heritable and prevalent phenotype. The emphasis of this review is on the recently identified 17 susceptibility loci for coronary artery disease. Implications of their discovery for biology and clinical medicine are discussed. A description of the landscape of human genetics in the near future in the context of next-generation sequence technologies is provided at the conclusion of this review.
Collapse
Affiliation(s)
- S Sivapalaratnam
- Department of Vascular Medicine, Academic Medical Center Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Brunzell JD, Zambon A, Deeb SS. The effect of hepatic lipase on coronary artery disease in humans is influenced by the underlying lipoprotein phenotype. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1821:365-72. [PMID: 21986251 DOI: 10.1016/j.bbalip.2011.09.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 09/13/2011] [Accepted: 09/20/2011] [Indexed: 12/31/2022]
Abstract
Increased or decreased hepatic lipase (HL) activity has been associated with coronary artery disease (CAD). This is consistent with the findings that gene variants that influence HL activity were associated with increased CAD risk in some population studies but not in others. In this review, we will explain the conditions that influence the effects of HL on CAD. Increased HL is associated with smaller and denser LDL (sdLDL) and HDL (HDL(3)) particles, while decreased HL is associated with larger and more buoyant LDL and HDL particles. The effect of HL activity on CAD risk is dependent on the underlying lipoprotein phenotype or disorder. Central obesity with hypertriglyceridemia (HTG) is associated with high HL activity that leads to the formation of sdLDL that is pro-atherogenic. In the absence of HTG, where large buoyant cholesteryl ester-enriched LDL is prominent, elevation of HL does not raise the risk for CAD. In HTG patients, drug therapy that decreases HL activity selectively decreases sdLDL particles, an anti-atherogenic effect. Drug therapy that raises HDL(2) cholesterol has not decreased the risk for CAD. In trials where inhibition of cholesterol ester transfer protein (CETP) or HL occurs, the increase in HDL(2) most likely is due to inhibition of catabolism of HDL(2) and impairment of reverse cholesterol transport (RCT). In patients with isolated hypercholesterolemia, but with normal triglyceride levels and big-buoyant LDL particles, an increase in HL activity is beneficial; possibly because it increases RCT. Drugs that lower HL activity might decrease the risk for CAD only in hypertriglyceridemic patients with sdLDL by selectively clearing sdLDL particles from plasma, which would override the potentially pro-atherogenic effect on RCT. This article is part of a Special Issue entitled Advances in High Density Lipoprotein Formation and Metabolism: A Tribute to John F. Oram (1945-2010).
Collapse
Affiliation(s)
- John D Brunzell
- University of Washington, Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, Box 356426, 1959 NE Pacific Avenue, Seattle, Washington 98195, USA.
| | | | | |
Collapse
|
46
|
Guella I, Duga S, Ardissino D, Merlini PA, Peyvandi F, Mannucci PM, Asselta R. Common variants in the haemostatic gene pathway contribute to risk of early-onset myocardial infarction in the Italian population. Thromb Haemost 2011; 106:655-64. [PMID: 21901231 DOI: 10.1160/th11-04-0247] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Accepted: 07/18/2011] [Indexed: 12/13/2022]
Abstract
Occlusive coronary thrombus formation superimposed on an atherosclerotic plaque is the ultimate event leading to myocardial infarction (MI). Therefore, haemostatic proteins may represent important players in the pathogenesis of MI. It was the objective of this study to evaluate, in a comprehensive way, the role of haemostatic gene polymorphisms in predisposition to premature MI. A total of 810 single nucleotide polymorphisms (SNPs) in 37 genes were assessed for association with MI in a large cohort (1,670 males, 210 females) of Italian patients who suffered from an MI event before the age of 45, and an equal number of controls. Thirty-eight SNPs selected from the literature were genotyped using the SNPlex technology, whereas genotypes for the remaining 772 SNPs were extracted from a previous genome-wide association study. Genotypes were analysed by a standard case-control analysis corrected for classical cardiovascular risk factors, and by haplotype analysis. A weighted Genetic Risk Score (GRS) was calculated. Evidence for association with MI after covariate correction was found for 35 SNPs in 12 loci: F5, PROS1, F11, ITGA2, F12, F13A1, SERPINE1, PLAT, VWF, THBD, PROCR, and F9. The weighted GRS was constructed by including the top SNP for each of the 12 associated loci. The GRS distribution was significantly different between cases and controls, and subjects in the highest quintile had a 2.69-fold increased risk for MI compared with those in the lowest quintile. Our results suggest that a GRS, based on the combined effect of several risk alleles in different haemostatic genes, is associated with an increased risk of MI.
Collapse
Affiliation(s)
- Ilaria Guella
- Department of Biology and Genetics for Medical Sciences, University of Milan, Milano, Italy
| | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Predictive models for future risk of coronary heart disease (CHD) based on traditional risk factors, such as age, male gender, LDL cholesterol, HDL cholesterol, diabetes mellitus, hypertension, smoking and family history of premature CHD, are quite robust but leave room for further improvement. Thus, efforts are being made to assess additional biomarkers for CHD, such as, lipoprotein (a), C-reactive protein, fibrinogen, lipoprotein-associated phospholipase A2, homocysteine and others. However, none of the novel biomarkers has demonstrated improved prediction beyond traditional risk factor models in a consistent fashion across multiple cohorts. Many criteria have to be fulfilled before a biomarker can be considered clinically relevant. Another way is to develop new models predicting long-term or life-time risk of CHD. Further research using novel biomarkers and long-term predictive models has the potential to improve CHD risk prediction.
Collapse
Affiliation(s)
- Abhimanyu Garg
- Division of Nutrition and Metabolic Diseases, Department of Internal Medicine, Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
48
|
Shared genetic architecture in the relationship between adult stature and subclinical coronary artery atherosclerosis. Atherosclerosis 2011; 219:679-83. [PMID: 21937044 DOI: 10.1016/j.atherosclerosis.2011.08.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 08/16/2011] [Accepted: 08/17/2011] [Indexed: 11/21/2022]
Abstract
BACKGROUND Short stature is associated with increased risk of coronary heart disease (CHD); although the mechanisms for this relationship are unknown, shared genetic factors have been proposed. Subclinical atherosclerosis, measured by coronary artery calcification (CAC), is associated with CHD events and represents part of the biological continuum to overt CHD. Many molecular mechanisms of CAC development are shared with bone growth. Thus, we examined whether there was evidence of shared genes (pleiotropy) between adult stature and CAC. METHODS 877 Asymptomatic white adults (46% men) from 625 families in a community-based sample had computed tomography measures of CAC. Pleiotropy between height and CAC was determined using maximum-likelihood estimation implemented in SOLAR. RESULTS Adult height was significantly and inversely associated with CAC score (P = 0.01). After adjusting for age, sex and CHD risk factors, the estimated genetic correlation between height and CAC score was -0.37 and was significantly different than 0 (P = 0.001) and -1 (P < 0.001). The environmental correlation between height and CAC score was 0.60 and was significantly different than 0 (P = 0.024). CONCLUSIONS Further studies of shared genetic factors between height and CAC may provide important insight into the complex genetic architecture of CHD, in part through increased understanding of the molecular pathways underlying the process of both normal growth and disease development. Bivariate genetic linkage analysis may provide a powerful mechanism for identifying specific genomic regions associated with both height and CAC.
Collapse
|
49
|
Wu G, Liu L, Huang J, Pang S, Wei G, Cui Y, Yan B. Alterations of autophagic-lysosomal system in the peripheral leukocytes of patients with myocardial infarction. Clin Chim Acta 2011; 412:1567-71. [PMID: 21575615 DOI: 10.1016/j.cca.2011.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 05/02/2011] [Accepted: 05/02/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND Myocardial infarction (MI) is a common and multifactorial disease. To date, causal genes and underlying mechanisms remain largely unknown. Autophagic-lysosomal system, a highly conserved degradative process in cells, has been implicated in lipid metabolism. In this study, we explored the alterations of the autophagic-lysosomal system in patients with acute MI. METHODS Gene expression of lysosomal associated membrane protein 2 (LAMP-2), a lysosomal marker gene, and microtubule-associated protein 1 light chain 3 (LC3), an autophagy marker gene, in the peripheral leukocytes of MI patients were examined at transcription and protein levels by RT-PCR assay and western blot analysis, respectively. RESULTS Compared to age- and sex-matched healthy controls (n=146), levels of LC3 gene expression and LC3-II protein, a cleaved form of LC3 protein, were significantly decreased in MI patients (n=81). LAMP-2 gene expression and protein levels were significantly increased. Decreased LC3 gene expression (OR, 2.150, 95%CI, 1.050-4.405, P=0.036) or increased LAMP-2 gene expression (OR, 3.317, 95%CI, 1.588-6.931, P<0.001) levels were associated with MI. CONCLUSIONS Our findings indicated that in the peripheral leukocytes of MI patients, autophagy activity is reduced and lysosomal accumulation is increased, which may contribute to the MI pathogenesis. Further genetic analyses of autophagic-lysosomal genes are warranted.
Collapse
Affiliation(s)
- Guanghua Wu
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Jining Medical College Affiliated Hospital, Jining Medical College, Jining, Shandong, PR China
| | | | | | | | | | | | | |
Collapse
|
50
|
Ashokkumar M, Anbarasan C, Saibabu R, Kuram S, Raman SC, Cherian KM. An association study of thrombospondin 1 and 2 SNPs with coronary artery disease and myocardial infarction among South Indians. Thromb Res 2011; 128:e49-53. [PMID: 21762961 DOI: 10.1016/j.thromres.2011.05.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 04/27/2011] [Accepted: 05/30/2011] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Thrombospondin 1 and 2 are multidomain calcium-binding extracellular glycoproteins and they play a role in platelet aggregation, inflammatory response and assembly of connective tissue extracellular matrix. The association of thrombospondins (TSP) in the pathogenesis of coronary artery disease (CAD) and myocardial infarction (MI) is well established. The association of the TSP-1 (Asn700Ser, 2210A → G, rs2228262) and TSP-2 un-translated region (UTR) (3949T → G, rs8089) gene variations among South Indian CAD and MI patients has been examined in the present study. MATERIALS AND METHODS We analyzed the thrombospondin polymorphisms in unrelated CAD patients (n = 511) and a subgroup with an event of MI (n = 173) compared with controls (n = 522). The polymorphisms were assessed using polymerase chain reaction, restriction fragment length analysis and the circulating TSP concentration were measured using enzyme linked immune-sorbent assay. RESULTS The prevalence of TSP-1 and TSP-2 alleles did not show any significant difference statistically, when compared controls against CAD/MI patients. The rare GG genotype of the N700S polymorphism was not observed among the studied population. Further, multiple regression analysis revealed that there was no significant risk for CAD (OR = 1.68; 95% CI 0.927 - 3.055; p = 0.087) or MI (OR = 1.84; 95% CI 0.846 - 4.007; p = 0.124) for the GA genotype. The GA genotype showed no impact on clinical characteristics of the CAD patients and their circulating TSP-1 levels. A similar non-association was observed for the TSP-2 in 3949T → G polymorphism (GG genotype) for CAD (OR = 0.64; 95% CI 0.278 - 1.455; p = 0.636) and MI (OR = 0.53; 95% CI 0.166 - 1.675; p = 0.278). CONCLUSIONS Our data suggests that the presence of thrombospondin-1 (rs2228262) and thrombospondin-2 (rs8089) variants need not be considered a risk for coronary artery disease or myocardial infarction among South Indians.
Collapse
|