1
|
Cruz N, Valente T, Ferreira F, Macedo L, Belisário A, da Silva C, Oliveira N, Gomides A, Velloso-Rodrigues C. CETP gene polymorphisms and haplotypes are explanatory variables for HDL cholesterol level in sickle cell disease. Braz J Med Biol Res 2024; 57:e12879. [PMID: 38265339 PMCID: PMC10802225 DOI: 10.1590/1414-431x2023e12879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 11/18/2023] [Indexed: 01/25/2024] Open
Abstract
Variations in lipid profile have been observed in sickle cell disease (SCD) and understanding their relationship with disease severity is crucial. This study aimed to investigate the association of polymorphisms of the CETP gene and laboratory markers of disease severity with lipid profile in a pediatric population with SCD. Biochemical and anthropometric analyses and CETP and alpha-thalassemia genotyping were performed. The study included 133 children and adolescents with sickle cell anemia (SCA) or hemoglobin SC disease (SCC), in steady-state. The SCA and no hydroxyurea (no HU) groups had higher values of ApoB, total cholesterol, low-density lipoprotein cholesterol (LDL-C), and non-high-density lipoprotein cholesterol (non-HDL-C) compared to the SCC and HU groups. However, there were no significant differences in ApoA1 and HDL-C levels between the groups based on genotype. Furthermore, the groups with altered levels of ApoA1, HDL-C, and the triglyceride/HDL ratio exhibited lower hemoglobin (Hb) levels and higher white blood cell counts. Hb level was associated to HDL-C levels. Analysis of CETP gene variants showed that the minor alleles of rs3764261 (C>A), rs247616 (C>T), and rs183130 (C>T), as well as the TTA haplotype, are explanatory variables for HDL-C levels. These findings suggested that dyslipidemia in SCD, specifically related to HDL-C levels, may be influenced by individual genetic background. Additionally, further investigation is needed to determine if clinical manifestations are impacted by CETP gene variants.
Collapse
Affiliation(s)
- N.R.C. Cruz
- Laboratório de Biologia Celular e Genética Molecular, Departamento de Nutrição, Universidade Federal de Juiz de Fora - Campus Governador Valadares, Governador Valadares, MG, Brasil
| | - T.N.S. Valente
- Laboratório de Biologia Celular e Genética Molecular, Departamento de Ciências Básicas da Vida, Universidade Federal de Juiz de Fora - Campus Governador Valadares, Governador Valadares, MG, Brasil
| | - F.O. Ferreira
- Departamento de Ciências Básicas da Vida, Universidade Federal de Juiz de Fora - Campus Governador Valadares, Governador Valadares, MG, Brasil
| | - L.R. Macedo
- Departamento de Economia, Universidade Federal de Juiz de Fora - Campus Governador Valadares, Governador Valadares, MG, Brasil
| | - A.R. Belisário
- Centro de Tecidos Biológicos, Fundação Hemominas, Lagoa Santa, MG, Brasil
| | - C.M. da Silva
- Faculdade de Ciências Médicas de Minas Gerais, Belo Horizonte, MG, Brasil
| | - N.S. Oliveira
- Laboratório de Biologia Celular e Genética Molecular, Departamento de Nutrição, Universidade Federal de Juiz de Fora - Campus Governador Valadares, Governador Valadares, MG, Brasil
| | - A.F.F. Gomides
- Laboratório de Biologia Celular e Genética Molecular, Departamento de Ciências Básicas da Vida, Universidade Federal de Juiz de Fora - Campus Governador Valadares, Governador Valadares, MG, Brasil
| | - C. Velloso-Rodrigues
- Laboratório de Biologia Celular e Genética Molecular, Departamento de Ciências Básicas da Vida, Universidade Federal de Juiz de Fora - Campus Governador Valadares, Governador Valadares, MG, Brasil
| |
Collapse
|
2
|
Zhang R, Xie Q, Xiao P. Association of the polymorphisms of the cholesteryl ester transfer protein gene with coronary artery disease: a meta-analysis. Front Cardiovasc Med 2023; 10:1260679. [PMID: 38146445 PMCID: PMC10749314 DOI: 10.3389/fcvm.2023.1260679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 11/07/2023] [Indexed: 12/27/2023] Open
Abstract
Aims This meta-analysis aimed to assess the association of the polymorphisms of cholesterol ester transfer protein (CETP) rs708272 (G>A), rs5882 (G>A), rs1800775 (C>A), rs4783961 (G>A), rs247616 (C>T), rs5883 (C>T), rs1800776 (C>A), and rs1532624 (C>A) with coronary artery disease (CAD) and the related underlying mechanisms. Methods A comprehensive search was performed using five databases such as PubMed, EMBASE, Web of Science, Cochrane Library and Scopus to obtain the appropriate articles. The quality of the included studies was assessed by the Newcastle-Ottawa Scale. The statistical analysis of the data was performed using STATA 17.0 software. The association between CETP gene polymorphisms and risk of CAD was estimated using the pooled odds ratio (OR) and 95% confidence interval (95% CI). The association of CETP gene polymorphisms with lipids and with CETP levels was assessed using the pooled standardized mean difference and corresponding 95% CI. P < 0.05 was considered statistically significant. Results A total of 70 case-control studies with 30,619 cases and 31,836 controls from 46 articles were included. The results showed the CETP rs708272 polymorphism was significantly associated with a reduced risk of CAD under the allele model (OR = 0.846, P < 0.001), the dominant model (OR = 0.838, P < 0.001) and the recessive model (OR = 0.758, P < 0.001). AA genotype and GA genotype corresponded to higher high-density lipoprotein cholesterol (HDL-C) concentrations in the blood compared with GG genotype across the studied groups (all P < 0.05). The CETP rs5882 and rs1800775 polymorphisms were not significantly associated with CAD under the allele model (P = 0.802, P = 0.392), the dominant model (P = 0.556, P = 0.183) and the recessive model (P = 0.429, P = 0.551). Similarly, the other mentioned gene polymorphisms were not significantly associated with CAD under the three genetic models. Conclusions The CETP rs708272 polymorphism shows a significant association with CAD, and the carriers of the allele A are associated with a lower risk of CAD and higher HDL-C concentrations in the blood compared to the non-carriers. The CETP rs5882, rs1800775, rs4783961, rs247616, rs5883, rs1800776, and rs1532624 are not significantly associated with CAD. Systematic Review Registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42023432865, identifier: CRD42023432865.
Collapse
Affiliation(s)
- Ruizhe Zhang
- Department of Cardiology, Sir Run Run, Hospital, Nanjing Medical University, Nanjing, China
| | - Qingya Xie
- Department of Cardiology, The Forth Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Pingxi Xiao
- Department of Cardiology, The Forth Affiliated Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
3
|
Poliakova T, Wellington CL. Roles of peripheral lipoproteins and cholesteryl ester transfer protein in the vascular contributions to cognitive impairment and dementia. Mol Neurodegener 2023; 18:86. [PMID: 37974180 PMCID: PMC10652636 DOI: 10.1186/s13024-023-00671-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/24/2023] [Indexed: 11/19/2023] Open
Abstract
This narrative review focuses on the role of cholesteryl ester transfer protein (CETP) and peripheral lipoproteins in the vascular contributions to cognitive impairment and dementia (VCID). Humans have a peripheral lipoprotein profile where low-density lipoproteins (LDL) represent the dominant lipoprotein fraction and high-density lipoproteins (HDL) represent a minor lipoprotein fraction. Elevated LDL-cholesterol (LDL-C) levels are well-established to cause cardiovascular disease and several LDL-C-lowering therapies are clinically available to manage this vascular risk factor. The efficacy of LDL-C-lowering therapies to reduce risk of all-cause dementia and AD is now important to address as recent studies demonstrate a role for LDL in Alzheimer's Disease (AD) as well as in all-cause dementia. The LDL:HDL ratio in humans is set mainly by CETP activity, which exchanges cholesteryl esters for triglycerides across lipoprotein fractions to raise LDL and lower HDL as CETP activity increases. Genetic and pharmacological studies support the hypothesis that CETP inhibition reduces cardiovascular risk by lowering LDL, which, by extension, may also lower VCID. Unlike humans, wild-type mice do not express catalytically active CETP and have HDL as their major lipoprotein fraction. As HDL has potent beneficial effects on endothelial cells, the naturally high HDL levels in mice protect them from vascular disorders, likely including VCID. Genetic restoration of CETP expression in mice to generate a more human-like lipid profile may increase the relevance of murine models for VCID studies. The therapeutic potential of existing and emerging LDL-lowering therapies for VCID will be discussed. Figure Legend. Cholesteryl Ester Transfer Protein in Alzheimer's Disease. CETP is mainly produced by the liver, and exchanges cholesteryl esters for triglycerides across lipoprotein fractions to raise circulating LDL and lower HDL as CETP activity increases. Low CETP activity is associated with better cardiovascular health, due to decreased LDL and increased HDL, which may also improve brain health. Although most peripheral lipoproteins cannot enter the brain parenchyma due to the BBB, it is increasingly appreciated that direct access to the vascular endothelium may enable peripheral lipoproteins to have indirect effects on brain health. Thus, lipoproteins may affect the cerebrovasculature from both sides of the BBB. Recent studies show an association between elevated plasma LDL, a well-known cardiovascular risk factor, and a higher risk of AD, and considerable evidence suggests that high HDL levels are associated with reduced CAA and lower neuroinflammation. Considering the potential detrimental role of LDL in AD and the importance of HDL's beneficial effects on endothelial cells, high CETP activity may lead to compromised BBB integrity, increased CAA deposits and greater neuroinflammation. Abbreviations: CETP - cholesteryl transfer ester protein; LDL - low-density lipoproteins; HDL - high-density lipoproteins; BBB - blood-brain barrier; CAA - cerebral amyloid angiopathy, SMC - smooth muscle cells, PVM - perivascular macrophages, RBC - red blood cells.
Collapse
Affiliation(s)
- Tetiana Poliakova
- Department of Pathology and Laboratory Medicine, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafagian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Cheryl L Wellington
- Department of Pathology and Laboratory Medicine, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- Djavad Mowafagian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
- International Collaboration On Repair Discoveries, Vancouver, BC, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
4
|
Legault MA, Barhdadi A, Gamache I, Lemaçon A, Lemieux Perreault LP, Grenier JC, Sylvestre MP, Hussin JG, Rhainds D, Tardif JC, Dubé MP. Study of effect modifiers of genetically predicted CETP reduction. Genet Epidemiol 2023; 47:198-212. [PMID: 36701426 DOI: 10.1002/gepi.22514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/11/2022] [Accepted: 01/11/2023] [Indexed: 01/27/2023]
Abstract
Genetic variants in drug targets can be used to predict the long-term, on-target effect of drugs. Here, we extend this principle to assess how sex and body mass index may modify the effect of genetically predicted lower CETP levels on biomarkers and cardiovascular outcomes. We found sex and body mass index (BMI) to be modifiers of the association between genetically predicted lower CETP and lipid biomarkers in UK Biobank participants. Female sex and lower BMI were associated with higher high-density lipoprotein cholesterol and lower low-density lipoprotein cholesterol for the same genetically predicted reduction in CETP concentration. We found that sex also modulated the effect of genetically lower CETP on cholesterol efflux capacity in samples from the Montreal Heart Institute Biobank. However, these modifying effects did not extend to sex differences in cardiovascular outcomes in our data. Our results provide insight into the clinical effects of CETP inhibitors in the presence of effect modification based on genetic data. The approach can support precision medicine applications and help assess the external validity of clinical trials.
Collapse
Affiliation(s)
- Marc-André Legault
- Montreal Heart Institute, Montreal, Quebec, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Quebec, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Amina Barhdadi
- Montreal Heart Institute, Montreal, Quebec, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Quebec, Canada
| | - Isabel Gamache
- Montreal Heart Institute, Montreal, Quebec, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Audrey Lemaçon
- Montreal Heart Institute, Montreal, Quebec, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Quebec, Canada
| | - Louis-Philippe Lemieux Perreault
- Montreal Heart Institute, Montreal, Quebec, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Quebec, Canada
| | | | - Marie-Pierre Sylvestre
- Research Centre of the University of Montreal Hospital Centre, Montreal, Quebec, Canada.,Department of Social and Preventive Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Julie G Hussin
- Montreal Heart Institute, Montreal, Quebec, Canada.,Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | | | - Jean-Claude Tardif
- Montreal Heart Institute, Montreal, Quebec, Canada.,Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Marie-Pierre Dubé
- Montreal Heart Institute, Montreal, Quebec, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Quebec, Canada.,Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
5
|
Zihlif M, Otoum S, Al Shhab M, Almadani Z, Momani M, Alhawari H, Esraa Jibrini, Jarrar Y, Al-Ameer H, Imraish A. No association between LDL receptor and CETP genetic variants and atorvastatin response in Jordanian hyperlipidemic patients. Drug Metab Pers Ther 2022; 37:369-374. [PMID: 35447021 DOI: 10.1515/dmpt-2021-0177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 02/04/2022] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Atorvastatin is commonly used medication to achieve low levels of low-density lipoproteins (LDL). Cholesteryl ester transfer protein (CETP) and LDL receptor (LDLR) genetic variants can affect the cholesterol transport and hence may affect on atorvastatin response. This study aimed to investigate the influence of LDLR AvaII, CETP TaqIb, and Rs1532624 on the efficacy of 20 mg atorvastatin among Jordanian hyperlipidemic patients. METHODS One hundred and 50 blood samples were collected from hyperlipidemic patients in the University of Jordan Hospital. Polymerase chain reaction-restriction fragment length polymorphism was used for genotyping of LDLR AvaII and CETP TaqIb genetic variants. The genotyping of CETP Rs1532624 variant was done by Sanger DNA-Sequencing. RESULTS LDLR AvaII and CETP TaqIb and Rs1532624 variants showed a significant (p value < 0.05) association with the baseline of the LDL at the time of diagnoses. On the other hand, none of the tested genetic variants showed a significant (p value>0.05) association with LDL reduction after atorvastatin therapy. CONCLUSIONS Results demonstrated a significant association between the LDLR AvaII and CETP TaqIb, and Rs1532624 genetic variants with the LDL baseline level. However, the atorvastatin therapy among hyperlipidemic patients of Jordanian origin was not affected by any of the tested variants.
Collapse
Affiliation(s)
- Malek Zihlif
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Suhad Otoum
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Mohammad Al Shhab
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Zaid Almadani
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Monther Momani
- Department of Internal Medicine, School of Medicine, The University of Jordan, Amman, Jordan
| | - Hussam Alhawari
- Department of Internal Medicine, School of Medicine, The University of Jordan, Amman, Jordan
| | - Esraa Jibrini
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Yazun Jarrar
- Deprtmant of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Hamzeh Al-Ameer
- Department of Biology and Biotechnology, American University of Madaba, Madaba, Jordan
| | - Amer Imraish
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| |
Collapse
|
6
|
Sex-related differences in single nucleotide polymorphisms associated with dyslipidemia in a Korean population. Lipids Health Dis 2022; 21:124. [PMID: 36419087 PMCID: PMC9685854 DOI: 10.1186/s12944-022-01736-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/14/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND The prevalence of dyslipidemia has increased steadily in Korea, and the incidence of dyslipidemia differs by sex. In this study, we identified single nucleotide polymorphisms (SNPs) related to dyslipidemia in Korean cohorts through genome-wide association study (GWAS) analysis. METHODS Genotyping was conducted to determine the genotypes of 72,298 participants and investigate genotypes for 7,079,946 SNPs. Sex, age, and BMI were set as covariates for GWAS, and significant SNPs were identified in the discovery and replication stages using logistic regression. RESULTS GWAS of the entire cohort revealed a total of five significant SNPs: rs117026536 (LPL), rs651821 (APOA5), rs9804646 (APOA5), rs9926440 (CETP), and rs429358 (APOE). GWAS of the male subjects revealed a total of four significant SNPs. While rs9804646 (APOA5) and rs429358 (APOE) were significant for all the subjects, rs662799 (APOA5) and rs56156922 (CETP) were significant only for the male subjects. GWAS of the female subjects revealed two significant SNPs, rs651821 (APOA5) and rs9804646 (APOA5), both of which were significant in all the subjects. CONCLUSION This is the first study to identify sex-related differences in genetic polymorphisms in Korean populations with dyslipidemia. Further studies considering environmental variables will be needed to elucidate these sex-related genetic differences in dyslipidemia.
Collapse
|
7
|
Wuni R, Kuhnle GGC, Wynn-Jones AA, Vimaleswaran KS. A Nutrigenetic Update on CETP Gene–Diet Interactions on Lipid-Related Outcomes. Curr Atheroscler Rep 2022; 24:119-132. [PMID: 35098451 PMCID: PMC8924099 DOI: 10.1007/s11883-022-00987-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2021] [Indexed: 02/08/2023]
Abstract
Purpose of Review An abnormal lipid profile is considered a main risk factor for cardiovascular diseases and evidence suggests that single nucleotide polymorphisms (SNPs) in the cholesteryl ester transfer protein (CETP) gene contribute to variations in lipid levels in response to dietary intake. The objective of this review was to identify and discuss nutrigenetic studies assessing the interactions between CETP SNPs and dietary factors on blood lipids. Recent Findings Relevant articles were obtained through a literature search of PubMed and Google Scholar through to July 2021. An article was included if it examined an interaction between CETP SNPs and dietary factors on blood lipids. From 49 eligible nutrigenetic studies, 27 studies reported significant interactions between 8 CETP SNPs and 17 dietary factors on blood lipids in 18 ethnicities. The discrepancies in the study findings could be attributed to genetic heterogeneity, and differences in sample size, study design, lifestyle and measurement of dietary intake. The most extensively studied ethnicities were those of Caucasian populations and majority of the studies reported an interaction with dietary fat intake. The rs708272 (TaqIB) was the most widely studied CETP SNP, where ‘B1’ allele was associated with higher CETP activity, resulting in lower high-density lipoprotein cholesterol and higher serum triglycerides under the influence of high dietary fat intake. Summary Overall, the findings suggest that CETP SNPs might alter blood lipid profiles by modifying responses to diet, but further large studies in multiple ethnic groups are warranted to identify individuals at risk of adverse lipid response to diet. Supplementary Information The online version contains supplementary material available at 10.1007/s11883-022-00987-y.
Collapse
|
8
|
Schoch L, Badimon L, Vilahur G. Unraveling the Complexity of HDL Remodeling: On the Hunt to Restore HDL Quality. Biomedicines 2021; 9:805. [PMID: 34356869 PMCID: PMC8301317 DOI: 10.3390/biomedicines9070805] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/26/2022] Open
Abstract
Increasing evidence has cast doubt over the HDL-cholesterol hypothesis. The complexity of the HDL particle and its proven susceptibility to remodel has paved the way for intense molecular investigation. This state-of-the-art review discusses the molecular changes in HDL particles that help to explain the failure of large clinical trials intending to interfere with HDL metabolism, and details the chemical modifications and compositional changes in HDL-forming components, as well as miRNA cargo, that render HDL particles ineffective. Finally, the paper discusses the challenges that need to be overcome to shed a light of hope on HDL-targeted approaches.
Collapse
Affiliation(s)
- Leonie Schoch
- Cardiovascular Program, Institut de Recerca, Hospital Santa Creu i Sant Pau, 08025 Barcelona, Spain; (L.S.); (L.B.)
- Faculty of Medicine, University of Barcelona (UB), 08036 Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Program, Institut de Recerca, Hospital Santa Creu i Sant Pau, 08025 Barcelona, Spain; (L.S.); (L.B.)
- CiberCV, 08025 Barcelona, Spain
- Cardiovascular Research Chair, UAB, 08025 Barcelona, Spain
| | - Gemma Vilahur
- Cardiovascular Program, Institut de Recerca, Hospital Santa Creu i Sant Pau, 08025 Barcelona, Spain; (L.S.); (L.B.)
- CiberCV, 08025 Barcelona, Spain
| |
Collapse
|
9
|
Takaeko Y, Matsui S, Kajikawa M, Maruhashi T, Yamaji T, Harada T, Han Y, Hashimoto H, Kihara Y, Hida E, Chayama K, Goto C, Aibara Y, Yusoff FM, Kishimoto S, Nakashima A, Higashi Y. Relationship between high-density lipoprotein cholesterol levels and endothelial function in women: a cross-sectional study. BMJ Open 2020; 10:e038121. [PMID: 32641366 PMCID: PMC7342861 DOI: 10.1136/bmjopen-2020-038121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES The purpose of this study was to evaluate the relationship between high-density lipoprotein cholesterol (HDL-C) levels and endothelial function in women. DESIGN Cross-sectional study. SETTING 22 university hospitals and affiliated clinics in Japan. PARTICIPANTS 1719 Japanese women aged 17-90 years who were not receiving lipid-lowering therapy. MEASURES We evaluated flow-mediated vasodilation (FMD) and serum levels of HDL-C. All participants were divided into four groups by HDL-C level: low HDL-C (<40 mg/dL), moderate HDL-C (40-59 mg/dL), high HDL-C (60-79 md/dL) and extremely high HDL-C (≥80 mg/dL). RESULTS Univariate regression analysis revealed a significant relationship between FMD and HDL-C (r=0.12, p<0.001). FMD values were significantly smaller in the low HDL-C group (5.2%±3.8%) and moderate HDL-C group (5.2%±3.8%) than in the extremely high HDL-C group (6.7%±3.4%) (p=0.024 and p=0.003, respectively), while there was no significant difference in FMD between the high HDL-C group and the extremely high HDL-C group. Multiple logistic regression analysis did not show a significant association between HDL-C levels and FMD. CONCLUSIONS Endothelial function increased in relation to HDL-C levels. However, there was no association of HDL-C levels with endothelial function after adjustment of traditional cardiovascular risk factors in women. TRIAL REGISTRATION NUMBER UMIN000012950; Results.
Collapse
Affiliation(s)
- Yuji Takaeko
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Shogo Matsui
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Tatsuya Maruhashi
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Takayuki Yamaji
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Takahiro Harada
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yiming Han
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Haruki Hashimoto
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yasuki Kihara
- Department of Cardiovascular Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Eisuke Hida
- Department of Biostatistics and Data Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Chikara Goto
- Department of Physical Therapy, Hiroshima International University, HigashiHiroshima, Hiroshima, Japan
| | - Yoshiki Aibara
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima, Japan
| | - Farina Mohamad Yusoff
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima, Japan
| | - Shinji Kishimoto
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima, Japan
| | - Ayumu Nakashima
- Department of Stem Cell Biology and Medicine, Hiroshima University Faculty of Medicine Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yukihito Higashi
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima, Japan
| |
Collapse
|
10
|
Pawlik J, Wrześniok D. Cholesteryl ester transfer protein: the physiological and molecular characteristics in the pathogenesis of atherosclerosis and Alzheimer’s disease. POSTEP HIG MED DOSW 2019. [DOI: 10.5604/01.3001.0013.3673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cholesteryl ester transfer protein (CETP) is involved in reverse cholesterol transport, mediates the exchange of cholesteryl esters for triglycerides between high-density lipoproteinsand low-density lipoproteins/very low-density lipoproteins. Lipid transfer mechanism by CETP is unknown. Two main models have been proposed for the mechanism of action of CETP: shuttle and tunnel mechanisms. The variants of CETP gene affect activity and level of protein, thus they are associated with lipid profile and risk of many diseases. Some clinical studies reported that polymorphisms of CETP, including TaqIB and I405V, are associated with risk of atherosclerosis and/or Alzheimer’s disease. CETP plays important role an in the metabolism of cholesterol, thus is correlated with pathomechanism of coronary artery disease. Inhibition of CETP can be an effective strategy to improve the lipid profile and reduce risk of cardiovascular diseases. Therefore, new therapeutic strategies to reduce activity of CETP or decrease its level are developed. Effectiveness of following pharmacological methods of modulation of CETP activity was studied: anti-CETP vaccines, antisense oligonucleotide and small molecule inhibitors of CETP. This article presents an overview of the literature on the correlation between cardiovascular diseases and CETP protein/CETP gene. Furthermore, it discusses the impact of CETP on pathogenesis of Alzheimer’s disease.
Collapse
Affiliation(s)
- Justyna Pawlik
- Katedra i Zakład Chemii i Analizy Leków, Wydział Farmaceutyczny z Oddziałem Medycyny Laboratoryjnej w Sosnowcu, Śląski Uniwersytet Medyczny, Katowice, Polska
| | - Dorota Wrześniok
- Katedra i Zakład Chemii i Analizy Leków, Wydział Farmaceutyczny z Oddziałem Medycyny Laboratoryjnej w Sosnowcu, Śląski Uniwersytet Medyczny, Katowice, Polska
| |
Collapse
|
11
|
Prasad G, Bandesh K, Giri AK, Kauser Y, Chanda P, Parekatt V, Mathur S, Madhu SV, Venkatesh P, Bhansali A, Marwaha RK, Basu A, Tandon N, Bharadwaj D. Genome-Wide Association Study of Metabolic Syndrome Reveals Primary Genetic Variants at CETP Locus in Indians. Biomolecules 2019; 9:E321. [PMID: 31366177 PMCID: PMC6723498 DOI: 10.3390/biom9080321] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 07/26/2019] [Accepted: 07/30/2019] [Indexed: 12/11/2022] Open
Abstract
Indians, a rapidly growing population, constitute vast genetic heterogeneity to that of Western population; however they have become a sedentary population in past decades due to rapid urbanization ensuing in the amplified prevalence of metabolic syndrome (MetS). We performed a genome-wide association study (GWAS) of MetS in 10,093 Indian individuals (6,617 MetS and 3,476 controls) of Indo-European origin, that belong to our previous biorepository of The Indian Diabetes Consortium (INDICO). The study was conducted in two stages-discovery phase (N = 2,158) and replication phase (N = 7,935). We discovered two variants within/near the CETP gene-rs1800775 and rs3816117-associated with MetS at genome-wide significance level during replication phase in Indians. Additional CETP loci rs7205804, rs1532624, rs3764261, rs247617, and rs173539 also cropped up as modest signals in Indians. Haplotype association analysis revealed GCCCAGC as the strongest haplotype within the CETP locus constituting all seven CETP signals. In combined analysis, we perceived a novel and functionally relevant sub-GWAS significant locus-rs16890462 in the vicinity of SFRP1 gene. Overlaying gene regulatory data from ENCODE database revealed that single nucleotide polymorphism (SNP) rs16890462 resides in repressive chromatin in human subcutaneous adipose tissue as characterized by the enrichment of H3K27me3 and CTCF marks (repressive gene marks) and diminished H3K36me3 marks (activation gene marks). The variant displayed active DNA methylation marks in adipose tissue, suggesting its likely regulatory activity. Further, the variant also disrupts a potential binding site of a key transcription factor, NRF2, which is known for involvement in obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Gauri Prasad
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics and Integrative Biology Campus, New Delhi 110020, India
| | - Khushdeep Bandesh
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics and Integrative Biology Campus, New Delhi 110020, India
| | - Anil K Giri
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics and Integrative Biology Campus, New Delhi 110020, India
| | - Yasmeen Kauser
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics and Integrative Biology Campus, New Delhi 110020, India
| | - Prakriti Chanda
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Vaisak Parekatt
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India
| | - Sandeep Mathur
- Department of Endocrinology, S.M.S. Medical College, Jaipur, Rajasthan 302004, India
| | - Sri Venkata Madhu
- Division of Endocrinology, University College of Medical Sciences, New Delhi 110095, India
| | - Pradeep Venkatesh
- Department of Endocrinology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Anil Bhansali
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research, Sector-12, Chandigarh 160012, India
| | - Raman K Marwaha
- Department of Endocrinology, International Life Sciences Institute, New Delhi 110024, India
| | - Analabha Basu
- National Institute of Bio Medical Genomics, Netaji Subhas Sanatorium (Tuberculosis Hospital), Kalyani 741251, West Bengal, India
| | - Nikhil Tandon
- Department of Endocrinology, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Dwaipayan Bharadwaj
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics and Integrative Biology Campus, New Delhi 110020, India.
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
12
|
Blauw LL, Li-Gao R, Noordam R, de Mutsert R, Trompet S, Berbée JFP, Wang Y, van Klinken JB, Christen T, van Heemst D, Mook-Kanamori DO, Rosendaal FR, Jukema JW, Rensen PCN, Willems van Dijk K. CETP (Cholesteryl Ester Transfer Protein) Concentration: A Genome-Wide Association Study Followed by Mendelian Randomization on Coronary Artery Disease. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 11:e002034. [PMID: 29728394 DOI: 10.1161/circgen.117.002034] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 02/26/2018] [Indexed: 01/22/2023]
Abstract
BACKGROUND We aimed to identify independent genetic determinants of circulating CETP (cholesteryl ester transfer protein) to assess causal effects of variation in CETP concentration on circulating lipid concentrations and cardiovascular disease risk. METHODS A genome-wide association discovery and replication study on serum CETP concentration were embedded in the NEO study (Netherlands Epidemiology of Obesity). Based on the independent identified variants, Mendelian randomization was conducted on serum lipids (NEO study) and coronary artery disease (CAD; CARDIoGRAMplusC4D consortium). RESULTS In the discovery analysis (n=4248), we identified 3 independent variants (P<5×10-8) that determine CETP concentration. These single-nucleotide polymorphisms were mapped to CETP and replicated in a separate subpopulation (n=1458). Per-allele increase (SE) in serum CETP was 0.32 (0.02) µg/mL for rs247616-C, 0.35 (0.02) µg/mL for rs12720922-A, and 0.12 (0.02) µg/mL for rs1968905-G. Combined, these 3 variants explained 16.4% of the total variation in CETP concentration. One microgram per milliliter increase in genetically determined CETP concentration strongly decreased high-density lipoprotein cholesterol (-0.23 mmol/L; 95% confidence interval, -0.26 to -0.20), moderately increased low-density lipoprotein cholesterol (0.08 mmol/L; 95% confidence interval, 0.00-0.16), and was associated with an odds ratio of 1.08 (95% confidence interval, 0.94-1.23) for CAD risk. CONCLUSIONS This is the first genome-wide association study identifying independent variants that largely determine CETP concentration. Although high-density lipoprotein cholesterol is not a causal risk factor for CAD, it has been unequivocally demonstrated that low-density lipoprotein cholesterol lowering is proportionally associated with a lower CAD risk. Therefore, the results of our study are fully consistent with the notion that CETP concentration is causally associated with CAD through low-density lipoprotein cholesterol.
Collapse
Affiliation(s)
- Lisanne L Blauw
- Department of Internal Medicine, Division of Endocrinology (L.L.B., J.F.P.B., Y.W., P.C.N.R., K.W.v.D.) .,Department of Clinical Epidemiology (L.L.B., R.L.-G., R.d.M., T.C., D.O.M.-K., F.R.R.).,Einthoven Laboratory for Experimental Vascular Medicine (L.L.B., J.F.P.B., Y.W., J.B.v.K., P.C.N.R., K.W.v.D.)
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology (L.L.B., R.L.-G., R.d.M., T.C., D.O.M.-K., F.R.R.)
| | - Raymond Noordam
- Department of Internal Medicine, Division of Gerontology and Geriatrics (R.N., S.T., D.v.H.)
| | - Renée de Mutsert
- Department of Clinical Epidemiology (L.L.B., R.L.-G., R.d.M., T.C., D.O.M.-K., F.R.R.)
| | - Stella Trompet
- Department of Internal Medicine, Division of Gerontology and Geriatrics (R.N., S.T., D.v.H.).,Department of Cardiology (S.T., J.W.J.)
| | - Jimmy F P Berbée
- Department of Internal Medicine, Division of Endocrinology (L.L.B., J.F.P.B., Y.W., P.C.N.R., K.W.v.D.).,Einthoven Laboratory for Experimental Vascular Medicine (L.L.B., J.F.P.B., Y.W., J.B.v.K., P.C.N.R., K.W.v.D.)
| | - Yanan Wang
- Department of Internal Medicine, Division of Endocrinology (L.L.B., J.F.P.B., Y.W., P.C.N.R., K.W.v.D.).,Einthoven Laboratory for Experimental Vascular Medicine (L.L.B., J.F.P.B., Y.W., J.B.v.K., P.C.N.R., K.W.v.D.)
| | - Jan B van Klinken
- Einthoven Laboratory for Experimental Vascular Medicine (L.L.B., J.F.P.B., Y.W., J.B.v.K., P.C.N.R., K.W.v.D.).,Department of Human Genetics (J.B.v.K., K.W.v.D.)
| | - Tim Christen
- Department of Clinical Epidemiology (L.L.B., R.L.-G., R.d.M., T.C., D.O.M.-K., F.R.R.)
| | - Diana van Heemst
- Department of Internal Medicine, Division of Gerontology and Geriatrics (R.N., S.T., D.v.H.)
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology (L.L.B., R.L.-G., R.d.M., T.C., D.O.M.-K., F.R.R.).,and Department of Public Health and Primary Care (D.O.M.-K.) Leiden University Medical Center, The Netherlands
| | - Frits R Rosendaal
- Department of Clinical Epidemiology (L.L.B., R.L.-G., R.d.M., T.C., D.O.M.-K., F.R.R.)
| | | | - Patrick C N Rensen
- Department of Internal Medicine, Division of Endocrinology (L.L.B., J.F.P.B., Y.W., P.C.N.R., K.W.v.D.).,Einthoven Laboratory for Experimental Vascular Medicine (L.L.B., J.F.P.B., Y.W., J.B.v.K., P.C.N.R., K.W.v.D.)
| | - Ko Willems van Dijk
- Department of Internal Medicine, Division of Endocrinology (L.L.B., J.F.P.B., Y.W., P.C.N.R., K.W.v.D.).,Einthoven Laboratory for Experimental Vascular Medicine (L.L.B., J.F.P.B., Y.W., J.B.v.K., P.C.N.R., K.W.v.D.).,Department of Human Genetics (J.B.v.K., K.W.v.D.)
| |
Collapse
|
13
|
Samedy LA, Ryan GJ, Superko RH, Momary KM. CETP genotype and concentrations of HDL and lipoprotein subclasses in African-American men. Future Cardiol 2019; 15:187-195. [PMID: 31148465 DOI: 10.2217/fca-2018-0058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To assess the association between the CETP Taq1B and I405V polymorphisms with levels of lipoprotein subclasses in African-American (AA) men with and without Type 2 diabetes (T2DM). Patients & methods: AA men, over 30 years of age, with (n = 54) or without T2DM (n = 50), and not receiving lipid-lowering agents, underwent advanced lipid analysis and genotyping. Results & conclusion: In the total patient population Taq1B B2-allele carriers had significantly higher levels of large HDL subclasses (HDL-2b [p = 0.017] and HDL-L [p = 0.019]), lower levels of small-HDL subclasses (HDL-3a [p = 0.004] and HDL-3b [p = 0.031]), and lower levels of LDL subclasses (LDL-IVa [p = 0.012] and LDL-IIIb [p = 0.009]). The only significant genotype-diabetes interaction occurred with the HDL-2a subclass (p = 0.015). No statistically significant associations were seen with I405V genotype. Our observations of lower levels of small-HDL and higher levels of large-HDL suggest that a potentially important HDL subclass-CETP relationship exists.
Collapse
Affiliation(s)
- Lesly-Anne Samedy
- Department of Pharmacy Practice, Mercer University, College of Pharmacy, 3001 Mercer University Drive, Atlanta, GA 30341, USA
| | - Gina J Ryan
- Department of Pharmacy Practice, Mercer University, College of Pharmacy, 3001 Mercer University Drive, Atlanta, GA 30341, USA
| | | | - Kathryn M Momary
- Department of Pharmacy Practice, Mercer University, College of Pharmacy, 3001 Mercer University Drive, Atlanta, GA 30341, USA
| |
Collapse
|
14
|
Identifying genetic markers associated with susceptibility to cardiovascular diseases. Future Sci OA 2018; 5:FSO350. [PMID: 30652019 PMCID: PMC6331704 DOI: 10.4155/fsoa-2018-0031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 09/18/2018] [Indexed: 12/25/2022] Open
Abstract
The development of cardiovascular diseases (CVDs) is due to a complex interaction between the genome and the environment. Understanding how genetic differences in individuals contribute to their susceptibility to CVDs can help guide practitioners to give the best advice to achieve a favorable outcome for the patient. As genome technologies evolve, genotyping of individuals could be available to all patients using a simple saliva test. Large-scale genome-wide association studies and meta analyses have provided powerful insights into polymorphisms that may be predictive of disease and an individual's response to certain nutrients, but moving forward it is imperative that these insights can be applied in the medical setting to reduce the incidence and mortality of CVDs. Cardiovascular diseases (CVDs) are the leading cause of death worldwide, and while most CVDs can be prevented by adopting a healthy lifestyle, this is only half the story. Evidence suggests changes in an individual's genes or DNA can cause some form of CVDs, highlighting a complex relationship between genes and the environment. Genotyping, a process used to determine genetic differences within an individual's DNA, can provide doctors with relevant information to identify individuals who are at high risk of developing CVDs. This would allow treatment to begin early and encourage individuals to adopt a healthy lifestyle to reduce their risk.
Collapse
|
15
|
Wang D, Hartmann K, Seweryn M, Sadee W. Interactions Between Regulatory Variants in CYP7A1 (Cholesterol 7α-Hydroxylase) Promoter and Enhancer Regions Regulate CYP7A1 Expression. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2018; 11:e002082. [PMID: 30354296 PMCID: PMC6211808 DOI: 10.1161/circgen.118.002082] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 08/13/2018] [Indexed: 01/11/2023]
Abstract
BACKGROUND CYP7A1 (cholesterol 7α-hydroxylase) catalyzes the rate-limiting step in bile acid biosynthesis from cholesterol-a main pathway for cholesterol removal from the body. CYP7A1 single-nucleotide polymorphisms (SNPs) are associated with total cholesterol and LDL (low-density lipoprotein) levels, risk of cardiovascular diseases, and other phenotypes; however, results are inconsistent, and causative variants remain uncertain, except for a frequent promoter SNP (rs3808607). METHODS We used chromatin conformation capture (4C assay), chromatin immunoprecipitation qPCR assay in hepatocytes, and CRISPR (clustered regularly interspaced short palindromic repeats)-mediated genome editing in hepatocellular carcinoma cell line cells to identify regulatory regions for CYP7A1. We then screened for SNPs located in regulatory regions, testing effects on reporter gene assays and on hepatic CYP7A1 expression by measuring allelic mRNA expression imbalance. RESULTS 4C assays showed several regions interacting with CYP7A1 promoter. CRISPR-mediated genome editing in hepatocellular carcinoma cell line cells revealed a novel CYP7A1 enhancer and a repressor region, located >10 kb downstream of the CYP7A1 promoter. SNP screening with an allelic mRNA expression imbalance in human livers and reporter gene assays identified a frequent functional SNP (rs9297994) located in the downstream CYP7A1 enhancer region. SNP rs9297994 is in high linkage disequilibrium with promoter SNP rs3808607 but has opposite effects on CYP7A1 mRNA expression. Their combined effects using a 2-SNP model robustly associate with hepatic CYP7A1 mRNA expression, ranging >2 orders of magnitude. Moreover, only the 2-SNP model, but not each SNP alone, is significantly associated with LDL levels, risk of coronary artery disease, statin response, and diabetes mellitus in several clinical cohorts, including CATHGEN (Catheterization Genetics) and Framingham. CONCLUSIONS Two interacting regulatory SNPs modulate CYP7A1 expression and are associated with risk of coronary artery disease and diabetes mellitus.
Collapse
Affiliation(s)
- Danxin Wang
- Center for Pharmacogenomics, Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, FL
| | - Kate Hartmann
- Center for Pharmacogenomics, Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH
| | - Michal Seweryn
- Center for Medical Genomics OMICRON, UJ CM, Krakow, Poland
| | - Wolfgang Sadee
- Center for Pharmacogenomics, Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH
| |
Collapse
|
16
|
Iwanicka J, Iwanicki T, Niemiec P, Balcerzyk A, Krauze J, Górczyńska-Kosiorz S, Ochalska-Tyka A, Grzeszczak W, Żak I. Relationship between CETP gene polymorphisms with coronary artery disease in Polish population. Mol Biol Rep 2018; 45:1929-1935. [PMID: 30178218 PMCID: PMC6267260 DOI: 10.1007/s11033-018-4342-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 08/28/2018] [Indexed: 01/27/2023]
Abstract
The cholesteryl ester transfer protein (CETP) gene encodes a hydrophobic glycoprotein that plays a crucial role in the reverse transport of cholesterol. The aim of the present study was to determine whether CETP polymorphisms (rs1532624, rs247616 and rs708272) are associated with coronary artery disease (CAD) in a Polish population. Serum lipid levels and single nucleotide polymorphisms of CETP genes were determined in 494 subjects: 248 patients with premature CAD and 246 blood donors as controls. Selected polymorphisms were examined using TaqMan PCR analysis. We found that CAD risk was significantly higher for CC homozygotes and C allele carriers of the rs247616 polymorphism than for carriers with the T allele (OR 1.89, 95% CI 1.29–2.76, p = 0.001 and OR 1.51, 95% CI 1.14–1.99, p = 0.003) and likewise for the CC genotype of the rs1532624 polymorphism than for those with the A allele (OR 1.59, 95% CI 1.05–2.40, p = 0.026). Moreover, T allele carriers of the rs708272 polymorphism had significantly higher total cholesterol levels compared to CC homozygotes (p < 0.05) in the healthy controls. We also observed an allelic pattern, C(rs2477616)C(rs708272)C(rs1532624), which increased susceptibility to CAD by 43% (OR = 1.43, 95% CI 1.10–1.85, p = 0.006). In conclusion, the rs247616 and rs1532624 polymorphisms of CETP may modulate the risk of CAD in Polish population.
Collapse
Affiliation(s)
- Joanna Iwanicka
- Department of Biochemistry and Medical Genetics, School of Health Sciences in Katowice, Medical University of Silesia, Medyków Street 18, 40-752, Katowice, Poland.
| | - Tomasz Iwanicki
- Department of Biochemistry and Medical Genetics, School of Health Sciences in Katowice, Medical University of Silesia, Medyków Street 18, 40-752, Katowice, Poland
| | - Paweł Niemiec
- Department of Biochemistry and Medical Genetics, School of Health Sciences in Katowice, Medical University of Silesia, Medyków Street 18, 40-752, Katowice, Poland
| | - Anna Balcerzyk
- Department of Biochemistry and Medical Genetics, School of Health Sciences in Katowice, Medical University of Silesia, Medyków Street 18, 40-752, Katowice, Poland
| | - Jolanta Krauze
- 1st Department of Cardiac Surgery/2nd Department of Cardiology, American Heart of Poland, S. A. Armii Krajowej Street 101, 43-316, Bielsko-Biala, Poland
| | - Sylwia Górczyńska-Kosiorz
- Department of Internal Medicine, Diabetes and Nephrology, School of Medicine and Division of Dentistry in Zabrze, Medical University of Silesia, 3 Maja Street 13-15, 41-800, Zabrze, Poland
| | - Anna Ochalska-Tyka
- Regional Centre of Blood Donation and Blood Treatment in Raciborz, Sienkiewicza Street 3, 47-400, Raciborz, Poland
| | - Władysław Grzeszczak
- Department of Internal Medicine, Diabetes and Nephrology, School of Medicine and Division of Dentistry in Zabrze, Medical University of Silesia, 3 Maja Street 13-15, 41-800, Zabrze, Poland
| | - Iwona Żak
- Department of Biochemistry and Medical Genetics, School of Health Sciences in Katowice, Medical University of Silesia, Medyków Street 18, 40-752, Katowice, Poland
| |
Collapse
|
17
|
McCaffery JM, Ordovas JM, Huggins GS, Lai CQ, Espeland MA, Tate DF, Wing RR. Weight gain prevention buffers the impact of CETP rs3764261 on high density lipoprotein cholesterol in young adulthood: The Study of Novel Approaches to Weight Gain Prevention (SNAP). Nutr Metab Cardiovasc Dis 2018; 28:816-821. [PMID: 29699816 PMCID: PMC6127000 DOI: 10.1016/j.numecd.2018.02.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 02/22/2018] [Accepted: 02/27/2018] [Indexed: 11/15/2022]
Abstract
BACKGROUND AND AIMS Two weight gain prevention strategies, one targeting small changes to diet and physical activity and a second targeting large changes, significantly reduced weight gain in young adulthood. We examined whether weight gain prevention blunts genetic risk for body weight increase and/or high density lipoprotein cholesterol (HDL-C) lowering over two years. METHODS AND RESULTS Participants were 524 male and female young adults (mean age = 28.2, SD = 4.3; mean BMI = 25.5, SD = 2.6). Obesity-related SNPs accounting for ≥ 0.04% of the variance were genotyped and combined into a genetic risk score. For HDL-C, SNPs within CETP, LIPC and FADS2 were genotyped. The obesity-related genetic risk score did not predict change in BMI independently or in interaction with treatment arm. However, consistent with the prior literature, each copy of the HDL-C risk, C, allele at CETP rs3764261 was associated with lower HDL-C at baseline. Moreover, significant interaction between SNP and treatment arm for change in HDL-C was observed (p = 0.02). In the control group, HDL-C change was dependent upon rs3764261 (p = 0.004) with C allele carriers showing a continued reduction in HDL-C. In contrast, within the two intervention groups, HDL-C increased on average with no differential effect of rs3764261 (p > 0.24). Notably, even among carriers of the CC genotype, small and large change arms were associated with increased HDL-C and the control arm a reduction (p = 0.013). CONCLUSIONS The C allele at CETP rs3764261 is a strong risk factor for low HDL-C in young adulthood but weight gain prevention may mitigate this risk. CLINICAL TRIAL REGISTRY NUMBER AND WEBSITE: clinicaltrials.gov Identifier: NCT01183689, https://clinicaltrials.gov/.
Collapse
Affiliation(s)
- J M McCaffery
- Department of Psychiatry and Human Behavior, Alpert School of Medicine at Brown University, USA.
| | - J M Ordovas
- JM-USDA-HNRCA at Tufts University, USA; IMDEA Alimentacion, Madrid, Spain
| | - G S Huggins
- Sackler School of Biomedical Science, Tufts University, Madrid, Spain
| | - C-Q Lai
- USDA-ARS, JM-USDA Human Nutrition Research Center on Aging at Tufts University, USA
| | - M A Espeland
- Department of Biostatistical Sciences, Wake Forest School of Medicine, USA
| | - D F Tate
- Gillings School of Global Public Health, University of North Carolina, Carolina, USA
| | - R R Wing
- Department of Psychiatry and Human Behavior, Alpert School of Medicine at Brown University, USA
| |
Collapse
|
18
|
Garcia-Rios A, Alcala-Diaz JF, Gomez-Delgado F, Delgado-Lista J, Marin C, Leon-Acuña A, Camargo A, Rodriguez-Cantalejo F, Blanco-Rojo R, Quintana-Navarro G, Ordovas JM, Perez-Jimenez F, Lopez-Miranda J, Perez-Martinez P. Beneficial effect of CETP gene polymorphism in combination with a Mediterranean diet influencing lipid metabolism in metabolic syndrome patients: CORDIOPREV study. Clin Nutr 2018; 37:229-234. [DOI: 10.1016/j.clnu.2016.12.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 11/09/2016] [Accepted: 12/11/2016] [Indexed: 12/27/2022]
|
19
|
Klingel SL, Roke K, Hidalgo B, Aslibekyan S, Straka RJ, An P, Province MA, Hopkins PN, Arnett DK, Ordovas JM, Lai CQ, Mutch DM. Sex Differences in Blood HDL-c, the Total Cholesterol/HDL-c Ratio, and Palmitoleic Acid are Not Associated with Variants in Common Candidate Genes. Lipids 2017; 52:969-980. [PMID: 29080057 DOI: 10.1007/s11745-017-4307-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 10/04/2017] [Indexed: 01/21/2023]
Abstract
Blood lipids are associated with cardiovascular disease (CVD) risk. Moreover, circulating lipid and fatty acid levels vary between men and women, and evidence demonstrates these traits may be influenced by single nucleotide polymorphisms (SNP). Sex-genotype interactions related to blood lipids and fatty acids have been poorly investigated and may help elucidate sex differences in CVD risk. The goal of this study was to investigate if the influence of SNPs previously associated with blood lipids and fatty acids varies in a sex-specific manner. Lipids and fatty acids were measured in serum and red blood cells (RBC), respectively, in 94 adults (18-30 years) from the GONE FISHIN' cohort and 118 age-matched individuals from the GOLDN cohort. HDL-c levels were higher and the total cholesterol/HDL-c (TC/HDL-c) ratio was lower in women versus men (p < 0.01). RBC palmitoleic acid and the stearoyl-CoA desaturase index were both higher in women (p < 0.01). Fatty acid desaturase (FADS) pathway activity (estimated using the ratio of eicosapentaenoic acid/alpha-linolenic acid) was higher in men (p < 0.01). The AA genotype for rs1800775 in CETP had a lower TC/HDL-c ratio in men, but not women (p int = 0.03). Independent of sex, major alleles for rs174537 in FADS1 (GG) and rs3211956 in CD36 (TT) had higher arachidonic acid, lower dihomo-γ-linoleic acid, and a higher FADS1 activity compared to minor alleles. The current study showed that blood lipid and fatty acid levels vary between healthy young men and women, but that the observed sex differences are not associated with common variants in candidate lipid metabolism genes.
Collapse
Affiliation(s)
- Shannon L Klingel
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Animal Science and Nutrition Building, Guelph, ON, N1G 2W1, Canada
| | - Kaitlin Roke
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Animal Science and Nutrition Building, Guelph, ON, N1G 2W1, Canada
| | - Bertha Hidalgo
- Department of Epidemiology, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | - Stella Aslibekyan
- Department of Epidemiology, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | - Robert J Straka
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Ping An
- Division of Statistical Genomics, Department of Genetics, School of Medicine, Washington University, St. Louis, MO, USA
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, School of Medicine, Washington University, St. Louis, MO, USA
| | - Paul N Hopkins
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Donna K Arnett
- College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Jose M Ordovas
- JM-USDA-Human Nutrition Research Center ON Aging, Tufts University, Medford, MA, USA.,Instituto Madrileno Estudios Avanzados Alimentacion, Madrid, Spain.,Centro Nacional Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - David M Mutch
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Animal Science and Nutrition Building, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
20
|
Liang Y, Kelemen A. Shared polymorphisms and modifiable behavior factors for myocardial infarction and high cholesterol in a retrospective population study. Medicine (Baltimore) 2017; 96:e7683. [PMID: 28906356 PMCID: PMC5604625 DOI: 10.1097/md.0000000000007683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 07/10/2017] [Accepted: 07/12/2017] [Indexed: 01/07/2023] Open
Abstract
Genetic and environmental (behavior, clinical, and demographic) factors are associated with increased risks of both myocardial infarction (MI) and high cholesterol (HC). It is known that HC is major risk factor that may cause MI. However, whether there are common single nucleotide polymorphism (SNPs) associated with both MI and HC is not firmly established, and whether there are modulate and modified effects (interactions of genetic and known environmental factors) on either HC or MI, and whether these joint effects improve the predictions of MI, is understudied.The purpose of this study is to identify novel shared SNPs and modifiable environmental factors on MI and HC. We assess whether SNPs from a metabolic pathway related to MI may relate to HC; whether there are moderate effects among SNPs, lifestyle (smoke and drinking), HC, and MI after controlling other factors [gender, body mass index (BMI), and hypertension (HTN)]; and evaluate prediction power of the joint and modulate genetic and environmental factors influencing the MI and HC.This is a retrospective study with residents of Erie and Niagara counties in New York with a history of MI or with no history of MI. The data set includes environmental variables (demographic, clinical, lifestyle). Thirty-one tagSNPs from a metabolic pathway related to MI are genotyped. Generalized linear models (GLMs) with imputation-based analysis are conducted for examining the common effects of tagSNPs and environmental exposures and their interactions on having a history of HC or MI.MI, BMI, and HTN are significant risk factors for HC. HC shows the strongest effect on risk of MI in addition to HTN; gender and smoking status while drinking status shows protective effect on MI. rs16944 (gene IL-1β) and rs17222772 (gene ALOX) increase the risks of HC, while rs17231896 (gene CETP) has protective effects on HC either with or without the clinical, behavioral, demographic factors with different effect sizes that may indicate the existence of moderate or modifiable effects. Further analysis with the inclusions of gene-gene and gene-environmental interactions shows interactions between rs17231896 (CETP) and rs17222772 (ALOX); rs17231896 (CETP) and gender. rs17237890 (CETP) and rs2070744 (NOS3) are found to be significantly associated with risks of MI adjusted by both SNPs and environmental factors. After multiple testing adjustments, these effects diminished as expected. In addition, an interaction between drinking and smoking status is significant. Overall, the prediction power in successfully classifying MI status is increased to 80% with inclusions of all significant tagSNPs and environmental factors and their interactions compared with environmental factors only (72%).Having a history of either HC or MI has significant effects on each other in both directions, in addition to HTN and gender. Genes/SNPs identified from this analysis that are associated with HC may be potentially linked to MI, which could be further examined and validated through haplotype-pairs analysis with appropriate population stratification corrections, and function/pathway regulation analysis to eliminate the limitations of the current analysis.
Collapse
Affiliation(s)
| | - Arpad Kelemen
- Department of Organizational Systems and Adult Health, University of Maryland, Baltimore, MD
| |
Collapse
|
21
|
Hou H, Ma R, Guo H, He J, Hu Y, Mu L, Yan Y, Ma J, Li S, Zhang J, Ding Y, Zhang M, Niu Q, Liu J, Guo S. Association between Six CETP Polymorphisms and Metabolic Syndrome in Uyghur Adults from Xinjiang, China. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2017; 14:E653. [PMID: 28629169 PMCID: PMC5486339 DOI: 10.3390/ijerph14060653] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 06/15/2017] [Accepted: 06/15/2017] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To explore the association between CETP gene polymorphisms and metabolic syndrome (MS), as well as the relationship between the CETP gene polymorphisms and each component of MS. METHODS A total of 571 individuals which were randomly selected from 5692 Uyghur adults were subdivided into two groups, including 280 patients with MS and 291 control subjects, using the group-matching method after matching for gender. We detected CETP polymorphisms (rs5882, rs1800775, rs3764261, rs12149545, rs711752, and rs708272) by using the Snapshot method. RESULTS (1) Significant differences were found involving the frequency distribution of genotypes and alleles of rs1800775, rs3764261, rs12149545, rs711752, and rs708272 between the control and MS groups (all p < 0.05). (2) rs1800775, rs3764261, rs12149545, rs711752, and rs708272 polymorphisms were significantly related to the risk of MS (all p < 0.05). (3) The rs1800775 polymorphism was associated with high fasting blood glucose levels and low high density lipoprotein cholesterol (HDL-C); rs3764261 and rs12149545 polymorphisms were associated with all components of MS except high blood pressure; rs711752 and rs708272 polymorphisms were associated with low HDL-C (all p < 0.05). (4) Complete linkage disequilibrium (LD) was identified for two pairs of single nucleotide polymorphisms (SNPs) (rs3764261 and rs12149545 (D' = 1.000, r² = 0.931), rs711752 and rs708272 (D' = 1.000, r² = 0.996)). (5) The A-G-G-G-C (p = 0.013, odds ratio [OR] = 0.622, 95% confidence interval [95% CI] = 0.427-0.906) and A-T-A-A-T (p < 0.001, OR = 0.519, 95% CI = 0.386-0.697) haplotypes were more frequent in the control group than in the case group. Conclusions: The rs1800775, rs3764261, rs12149545, rs711752, and rs708272 polymorphisms of CETP were associated with MS and its components among the Uyghur ethnic group. Complete LD was found between two pairs of SNPs (rs3764261 and rs12149545, rs711752, and rs708272). The A-G-G-G-C and A-T-A-A-T haplotypes might be protective factors for MS.
Collapse
Affiliation(s)
- Huixian Hou
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Rulin Ma
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Heng Guo
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Jia He
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Yunhua Hu
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Lati Mu
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Yizhong Yan
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Jiaolong Ma
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Shugang Li
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Jingyu Zhang
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Yusong Ding
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Mei Zhang
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Qiang Niu
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Jiaming Liu
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Shuxia Guo
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| |
Collapse
|
22
|
Classical rather than genetic risk factors account for high cardiovascular disease prevalence in Lithuania: A cross-sectional population study. Adv Med Sci 2017; 62:121-128. [PMID: 28242483 DOI: 10.1016/j.advms.2016.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 08/24/2016] [Accepted: 08/25/2016] [Indexed: 10/20/2022]
Abstract
PURPOSE Cardiovascular disease (CVD) mortality accounts for 54% of all deaths in Lithuania, making it the highest among all of the European Union countries. We evaluated the prevalence of several CVD risk factors, including lifestyle, blood biochemistry and genetic predisposition to determine the reasons behind significantly increased CVD prevalence in Lithuania. MATERIALS AND METHODS In total 435 volunteers of Lithuanian ethnicity and stable geographic settlement for 3 generations, had their anthropometric, biochemical and behavioural risk factors measured. A randomly selected sample of 166 volunteers had their 60 CVD risk alleles genotyped. The prevalence of risk alleles and cumulative CVD genetic risk score were compared with population of North-West European origin (CEU) using data from the phase 3 HapMap project. RESULTS CVD was present in 33.8% of study volunteers, 84% of participants consumed alcohol, 21% were current smokers and only 30% of participants engaged in higher levels of physical activity. Also, the average BMI (males 28.3±4.3kg/m2, females 27.3±5.0kg/m2), total cholesterol (males 6.1±1.2mmol/L, females 6.2±1.0mmol/L) and LDL-cholesterol (males 4.1±1.1mmol/L, females 4.1±1.0mmol/L) were above the normal values. The cumulative genetic susceptibility to develop CVD in Lithuanians was only 1.4% higher than in CEU population. CONCLUSIONS High BMI and poor population plasma lipid profile are the major contributing factors to high CVD mortality and morbidity in Lithuania. Smoking, alcohol consumption and preliminary genetic predisposition results do not explain the difference in CVD mortality between the Lithuanian and wider European populations. CVD prevention programmes in Lithuania should primarily focus on weight loss and improving blood lipid control.
Collapse
|
23
|
Bien SA, Wojcik GL, Zubair N, Gignoux CR, Martin AR, Kocarnik JM, Martin LW, Buyske S, Haessler J, Walker RW, Cheng I, Graff M, Xia L, Franceschini N, Matise T, James R, Hindorff L, Le Marchand L, North KE, Haiman CA, Peters U, Loos RJF, Kooperberg CL, Bustamante CD, Kenny EE, Carlson CS. Strategies for Enriching Variant Coverage in Candidate Disease Loci on a Multiethnic Genotyping Array. PLoS One 2016; 11:e0167758. [PMID: 27973554 PMCID: PMC5156387 DOI: 10.1371/journal.pone.0167758] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 11/18/2016] [Indexed: 11/25/2022] Open
Abstract
Investigating genetic architecture of complex traits in ancestrally diverse populations is imperative to understand the etiology of disease. However, the current paucity of genetic research in people of African and Latin American ancestry, Hispanic and indigenous peoples in the United States is likely to exacerbate existing health disparities for many common diseases. The Population Architecture using Genomics and Epidemiology, Phase II (PAGE II), Study was initiated in 2013 by the National Human Genome Research Institute to expand our understanding of complex trait loci in ethnically diverse and well characterized study populations. To meet this goal, the Multi-Ethnic Genotyping Array (MEGA) was designed to substantially improve fine-mapping and functional discovery by increasing variant coverage across multiple ethnicities at known loci for metabolic, cardiovascular, renal, inflammatory, anthropometric, and a variety of lifestyle traits. Studying the frequency distribution of clinically relevant mutations, putative risk alleles, and known functional variants across multiple populations will provide important insight into the genetic architecture of complex diseases and facilitate the discovery of novel, sometimes population-specific, disease associations. DNA samples from 51,650 self-identified African ancestry (17,328), Hispanic/Latino (22,379), Asian/Pacific Islander (8,640), and American Indian (653) and an additional 2,650 participants of either South Asian or European ancestry, and other reference panels have been genotyped on MEGA by PAGE II. MEGA was designed as a new resource for studying ancestrally diverse populations. Here, we describe the methodology for selecting trait-specific content for use in multi-ethnic populations and how enriching MEGA for this content may contribute to deeper biological understanding of the genetic etiology of complex disease.
Collapse
Affiliation(s)
- Stephanie A. Bien
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- * E-mail: (CSC); (SAB)
| | - Genevieve L. Wojcik
- Department of Genetics, Stanford University, Stanford, California, United States of America
| | - Niha Zubair
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Christopher R. Gignoux
- Department of Genetics, Stanford University, Stanford, California, United States of America
| | - Alicia R. Martin
- Department of Genetics, Stanford University, Stanford, California, United States of America
| | - Jonathan M. Kocarnik
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Lisa W. Martin
- Division of Cardiology, George Washington University School of Medicine and Health Sciences, Washington, DC, United States of America
| | - Steven Buyske
- Department of Genetics, School of Arts and Sciences, Rutgers University, Piscataway, New Jersey, United States of America
| | - Jeffrey Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Ryan W. Walker
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Iona Cheng
- Cancer Prevention Institute of California, Fremont, California, United States of America
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Lucy Xia
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, California, United States of America
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Tara Matise
- Department of Genetics, School of Arts and Sciences, Rutgers University, Piscataway, New Jersey, United States of America
| | - Regina James
- Division of Intramural Research, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lucia Hindorff
- Division of Genomic Medicine, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Loic Le Marchand
- Department of Epidemiology Program, University of Hawai’i Cancer Center, Honolulu, Hawai’i, United States of America
| | - Kari E. North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Christopher A. Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, California, United States of America
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Charles L. Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Carlos D. Bustamante
- Department of Genetics, Stanford University, Stanford, California, United States of America
| | - Eimear E. Kenny
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Christopher S. Carlson
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
- * E-mail: (CSC); (SAB)
| | | |
Collapse
|
24
|
Postmus I, Warren HR, Trompet S, Arsenault BJ, Avery CL, Bis JC, Chasman DI, de Keyser CE, Deshmukh HA, Evans DS, Feng Q, Li X, Smit RAJ, Smith AV, Sun F, Taylor KD, Arnold AM, Barnes MR, Barratt BJ, Betteridge J, Boekholdt SM, Boerwinkle E, Buckley BM, Chen YDI, de Craen AJM, Cummings SR, Denny JC, Dubé MP, Durrington PN, Eiriksdottir G, Ford I, Guo X, Harris TB, Heckbert SR, Hofman A, Hovingh GK, Kastelein JJP, Launer LJ, Liu CT, Liu Y, Lumley T, McKeigue PM, Munroe PB, Neil A, Nickerson DA, Nyberg F, O’Brien E, O’Donnell CJ, Post W, Poulter N, Vasan RS, Rice K, Rich SS, Rivadeneira F, Sattar N, Sever P, Shaw-Hawkins S, Shields DC, Slagboom PE, Smith NL, Smith JD, Sotoodehnia N, Stanton A, Stott DJ, Stricker BH, Stürmer T, Uitterlinden AG, Wei WQ, Westendorp RGJ, Whitsel EA, Wiggins KL, Wilke RA, Ballantyne CM, Colhoun HM, Cupples LA, Franco OH, Gudnason V, Hitman G, Palmer CNA, Psaty BM, Ridker PM, Stafford JM, Stein CM, Tardif JC, Caulfield MJ, Jukema JW, Rotter JI, Krauss RM. Meta-analysis of genome-wide association studies of HDL cholesterol response to statins. J Med Genet 2016; 53:835-845. [PMID: 27587472 PMCID: PMC5309131 DOI: 10.1136/jmedgenet-2016-103966] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/03/2016] [Accepted: 07/26/2016] [Indexed: 11/04/2022]
Abstract
BACKGROUND In addition to lowering low density lipoprotein cholesterol (LDL-C), statin therapy also raises high density lipoprotein cholesterol (HDL-C) levels. Inter-individual variation in HDL-C response to statins may be partially explained by genetic variation. METHODS AND RESULTS We performed a meta-analysis of genome-wide association studies (GWAS) to identify variants with an effect on statin-induced high density lipoprotein cholesterol (HDL-C) changes. The 123 most promising signals with p<1×10-4 from the 16 769 statin-treated participants in the first analysis stage were followed up in an independent group of 10 951 statin-treated individuals, providing a total sample size of 27 720 individuals. The only associations of genome-wide significance (p<5×10-8) were between minor alleles at the CETP locus and greater HDL-C response to statin treatment. CONCLUSIONS Based on results from this study that included a relatively large sample size, we suggest that CETP may be the only detectable locus with common genetic variants that influence HDL-C response to statins substantially in individuals of European descent. Although CETP is known to be associated with HDL-C, we provide evidence that this pharmacogenetic effect is independent of its association with baseline HDL-C levels.
Collapse
Affiliation(s)
- Iris Postmus
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Helen R Warren
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, United Kingdom EC1M6BQ
- Barts NIHR Biomedical Research Unit
| | - Stella Trompet
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
- Department of Cardiology, Leiden University Medical Center, The Netherlands
| | | | - Christy L Avery
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston MA
- Harvard Medical School, Boston, MA
| | | | - Harshal A Deshmukh
- Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Daniel S Evans
- California Pacific Medical Center Research Institute, San Francisco, CA, USA, 94107
| | - QiPing Feng
- Department of Clinical Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Xiaohui Li
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Roelof AJ Smit
- Department of Cardiology, Leiden University Medical Center, The Netherlands
| | - Albert V Smith
- Icelandic Heart Association, Kopavogur, Iceland
- University of Iceland, Reykjavik, Iceland
| | - Fangui Sun
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Alice M Arnold
- Department of Biostatistics, University of Washington, Seattle, WA USA
| | - Michael R Barnes
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, United Kingdom EC1M6BQ
- Barts NIHR Biomedical Research Unit
| | - Bryan J Barratt
- Personalised Healthcare and Biomarkers, AstraZeneca, Alderley Park, UK
| | | | | | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Brendan M Buckley
- Department of Pharmacology and Therapeutics, University College Cork, Ireland
| | - Y-D Ida Chen
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Anton JM de Craen
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Steven R Cummings
- California Pacific Medical Center Research Institute, San Francisco, CA, USA, 94107
| | - Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Vanderbilt University, USA
| | | | - Paul N Durrington
- Cardiovascular Research Group, School of Biosciences, University of Manchester M13 9NT, UK
| | | | - Ian Ford
- Robertson Center for Biostatistics, University of Glasgow, United Kingdom
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Tamara B Harris
- Laboratory of Epidemiology, Demography, Biometry, National Institute on Aging, National Institutes of Health, 7201 Wisconsin Ave, Bethesda, MD 20892, USA
| | - Susan R Heckbert
- Department of Epidemiology, University of Washington, Seattle WA USA
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
- Group Health Research Institute, Group Health Cooperative, Seattle WA USA
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- The Netherlands Consortium for Healthy Ageing, Leiden, the Netherlands
| | - G Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, NL
| | - John JP Kastelein
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, NL
| | - Leonore J Launer
- Laboratory of Epidemiology, Demography, Biometry, National Institute on Aging, National Institutes of Health, 7201 Wisconsin Ave, Bethesda, MD 20892, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Yongmei Liu
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA, 27157
| | - Thomas Lumley
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
- Department of Statistic, University of Auckland, Auckland, New Zealand
| | | | - Patricia B Munroe
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, United Kingdom EC1M6BQ
- Barts NIHR Biomedical Research Unit
| | - Andrew Neil
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LJ UK
| | - Deborah A Nickerson
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Fredrik Nyberg
- Medical Evidence and Observational Research, AstraZeneca Gothenburg, Mölndal, Sweden
- Unit of Occupational and Environmental Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Eoin O’Brien
- The Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Christopher J O’Donnell
- NHLBI Framingham Heart Study, Framingham, MA, USA
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- National Heart, Lung and Blood Institute, Bethesda, MD
| | - Wendy Post
- Department of Cardiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Neil Poulter
- International Centre for Circulatory Health, Imperial College, London UK
| | - Ramachandran S Vasan
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, and the Framingham Heart Study, Framingham, MA, USA
| | - Kenneth Rice
- Department of Biostatistics, University of Washington, Seattle, WA USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, Faculty of Medicine, Glasgow, United Kingdom
| | - Peter Sever
- International Centre for Circulatory Health, Imperial College, London UK
| | - Sue Shaw-Hawkins
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, United Kingdom EC1M6BQ
- Barts NIHR Biomedical Research Unit
| | - Denis C Shields
- The Conway Institute, University College Dublin, Dublin 4, Ireland
- School of Medicine and Medical Sciences, University College Dublin
| | - P Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nicholas L Smith
- Department of Epidemiology, University of Washington, Seattle WA USA
- Group Health Research Institute, Group Health Cooperative, Seattle WA USA
- Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle WA USA
| | - Joshua D Smith
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
- Division of Cardiology, Harborview Medical Center, University of Washington, Seattle, WA USA
| | - Alice Stanton
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Beaumont Hospital, Dublin, Ireland
| | - David J Stott
- Institute of Cardiovascular and Medical Sciences, Faculty of Medicine, University of Glasgow, United Kingdom
| | - Bruno H Stricker
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Health Care Inspectorate. The Hague, The Netherlands
| | - Til Stürmer
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- The Netherlands Consortium for Healthy Ageing, Leiden, the Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| | - Rudi GJ Westendorp
- Department of Public Health, and Center for Healthy Ageing, University of Copenhagen, 1123 Copenhagen, Denmark
| | - Eric A Whitsel
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Kerri L Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | - Russell A Wilke
- Department of Internal Medicine, Sanford Healthcare, Sioux Falls, SD, USA
- Department of Medicine, University of South Dakota, Sioux Falls, SD, USA
| | | | - Helen M Colhoun
- Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
- Department of Public Health, University of Dundee
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- NHLBI Framingham Heart Study, Framingham, MA, USA
| | - Oscar H Franco
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- University of Iceland, Reykjavik, Iceland
| | - Graham Hitman
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London UK
| | - Colin NA Palmer
- Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
- Department of Epidemiology, University of Washington, Seattle WA USA
- Group Health Research Institute, Group Health Cooperative, Seattle WA USA
- Department of Health Services University of Washington, Seattle, WA
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston MA
| | - Jeanette M Stafford
- Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA, 27157
| | - Charles M Stein
- Department of Medicine, Vanderbilt University, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | | | - Mark J Caulfield
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, United Kingdom EC1M6BQ
- Barts NIHR Biomedical Research Unit
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, The Netherlands
- Durrer Center for Cardiogenetic Research, Amsterdam, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Ronald M Krauss
- Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
| |
Collapse
|
25
|
Ganesan M, Nizamuddin S, Katkam SK, Kumaraswami K, Hosad UK, Lobo LL, Kutala VK, Thangaraj K. c.*84G>A Mutation in CETP Is Associated with Coronary Artery Disease in South Indians. PLoS One 2016; 11:e0164151. [PMID: 27768712 PMCID: PMC5074517 DOI: 10.1371/journal.pone.0164151] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 09/20/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Coronary artery disease (CAD) is one of the leading causes of mortality worldwide. It is a multi-factorial disease and several studies have demonstrated that the genetic factors play a major role in CAD. Although variations in cholesteryl ester transfer protein (CETP) gene are reported to be associated with CAD, this gene has not been studied in South Indian populations. Hence we evaluated the CETP gene variations in CAD patients of South Indian origin. METHODS We sequenced all the exons, exon-intron boundaries and UTRs of CETP in 323 CAD patients along with 300 ethnically and age matched controls. Variations observed in CETP were subjected to various statistical analyses. RESULTS AND DISCUSSION Our analysis revealed a total of 13 variations. Of these, one3'UTRvariant rs1801706 (c.*84G>A) was significantly associated with CAD (genotype association test: OR = 2.16, 95% CI: 1.50-3.10, p = 1.88x10-5 and allelic association test: OR = 1.92, 95% CI: 1.40-2.63, p = 2.57x10-5). Mutant allele "A" was observed to influence the higher concentration of mRNA (p = 7.09×10-3, R2 = 0.029 and β = 0.2163). Since expression of CETP has been shown to be positively correlated with the risk of CAD, higher frequency of "A" allele (patients: 22.69% vs.controls: 13%) reveals that c.*84G>A is a risk factor for CAD in South Indians. CONCLUSIONS This is the first report of the CETP gene among South Indians CAD patients. Our results suggest that rs1801706 (c.*84G>A) is a risk factor for CAD in South Indian population.
Collapse
Affiliation(s)
- Mala Ganesan
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | | | - Konda Kumaraswami
- Department of Clinical Pharmacology and Therapeutics, Nizam's Institute of Medical Sciences (NIMS), Hyderabad, India
| | | | | | - Vijay Kumar Kutala
- Department of Clinical Pharmacology and Therapeutics, Nizam's Institute of Medical Sciences (NIMS), Hyderabad, India
| | | |
Collapse
|
26
|
Abstract
Cytochrome P450 3A4 (CYP3A4) metabolizes 30-50% of clinically used drugs. Large interperson variability in CYP3A4 activity affects response to CYP3A4 substrate drugs. We had demonstrated that an intronic single nucleotide polymorphism rs35599367 (CYP3A4*22, located in intron 6) reduces mRNA/protein expression; however, the underlying mechanism remained unknown. Here we show that CYP3A4*22 is associated with a two-fold or greater increase in formation of a nonfunctional CYP3A4 alternative splice variant with partial intron 6 retention in human liver (P=0.006), but not in small intestines. Consistent with this observation, in-vitro transfection experiments with a CYP3A4 minigene (spanning from intron 5 to intron 7) demonstrated that plasmids carrying the rs35599367 minor T allele caused significantly greater intron 6 retention than the C allele in liver derived HepG2 cells, but not in intestine-derived LS-174T cells. These results indicate that tissue-specific increased formation of nonfunctional alternative splice variant causes reduced CYP3A4 mRNA/protein expression in CYP3A4*22 carriers.
Collapse
|
27
|
Kitzmiller JP, Mikulik EB, Dauki AM, Murkherjee C, Luzum JA. Pharmacogenomics of statins: understanding susceptibility to adverse effects. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2016; 9:97-106. [PMID: 27757045 PMCID: PMC5055044 DOI: 10.2147/pgpm.s86013] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Statins are a cornerstone of the pharmacologic treatment and prevention of atherosclerotic cardiovascular disease. Atherosclerotic disease is a predominant cause of mortality and morbidity worldwide. Statins are among the most commonly prescribed classes of medications, and their prescribing indications and target patient populations have been significantly expanded in the official guidelines recently published by the American and European expert panels. Adverse effects of statin pharmacotherapy, however, result in significant cost and morbidity and can lead to nonadherence and discontinuation of therapy. Statin-associated muscle symptoms occur in ~10% of patients on statins and constitute the most commonly reported adverse effect associated with statin pharmacotherapy. Substantial clinical and nonclinical research effort has been dedicated to determining whether genetics can provide meaningful insight regarding an individual patient’s risk of statin adverse effects. This contemporary review of the relevant clinical research on polymorphisms in several key genes that affect statin pharmacokinetics (eg, transporters and metabolizing enzymes), statin efficacy (eg, drug targets and pathways), and end-organ toxicity (eg, myopathy pathways) highlights several promising pharmacogenomic candidates. However, SLCO1B1 521C is currently the only clinically relevant pharmacogenetic test regarding statin toxicity, and its relevance is limited to simvastatin myopathy.
Collapse
Affiliation(s)
| | - Eduard B Mikulik
- Department of Biological Chemistry and Pharmacology, College of Medicine
| | - Anees M Dauki
- College of Pharmacy, The Ohio State University, Columbus, OH
| | | | - Jasmine A Luzum
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| |
Collapse
|
28
|
Guo S, Hu Y, Ding Y, Liu J, Zhang M, Ma R, Guo H, Wang K, He J, Yan Y, Rui D, Sun F, Mu L, Niu Q, Zhang J, Li S. Association between Eight Functional Polymorphisms and Haplotypes in the Cholesterol Ester Transfer Protein (CETP) Gene and Dyslipidemia in National Minority Adults in the Far West Region of China. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2015; 12:15979-92. [PMID: 26694435 PMCID: PMC4690972 DOI: 10.3390/ijerph121215036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 12/03/2015] [Accepted: 12/10/2015] [Indexed: 01/03/2023]
Abstract
We have investigated the relationship between the polymorphisms and haplotypes in the CETP gene, and dyslipidemia among the Xinjiang Kazak and Uyghur populations in China. A total of 712 patients with dyslipidemia and 764 control subjects of CETP gene polymorphism at rs12149545, rs3764261, rs1800775, rs711752, rs708272, rs289714, rs5882, and rs1801706 loci were studied by the Snapshot method, linkage disequilibrium analysis and haplotype construction. The results are as follows: (1) the minor allele of eight loci of frequencies in the two groups were different from other results of similar studies in other countries; (2) In the linear regression analysis, the HDL-C levels of rs708272 TT, rs1800775 AA, rs289714 CC and rs711752 AA genotypes were significantly higher than those of other genotypes, however, the rs3764261 GG and rs12149545 GG genotypes were significantly lower than those of other genotypes in the two ethnic groups. The HDL-C levels of the rs12149545 GG genotype were lower than those of other genotypes; (3) in the control group, the rs708272 CT genotype of TG levels were lower than in the CC genotype, the T genotype of LDL-C levels were lower than in the CC genotype, and the HDL-C levels were higher than in the CT genotype; the rs1800775 AC genotype of TG levels were higher than in the AA genotype, the rs711752 AG genotype of TG levels were lower than in the GG genotype, the AA genotype LDL-C levels were lower than in the GG genotype, and the HDL-C levels were higher than in the AG genotype; the rs1800775 AC genotype of TG levels were higher than in the AA genotype. In the dyslipidemia group, the rs708272 TT genotype of TC and LDL-C levels were higher than in the CT genotype and the rs3764261 TT genotype of TC levels were higher than in the GG genotype. The rs711752 AA genotype of TC and LDL-C levels were higher than in the AG genotype, and the rs12149545 AA genotype of TC and LDL-C levels were higher than in the GG genotype; (4) perfect Linkage Disequilibrium was observed for two sets of two SNPs: rs3764261 and rs12149545; rs711752 and rs708272. (5) Using SHEsis software analysis, the five A/T/A/A/T/C/A/G, A/T/A/A/T/T/G/A, G/G/A/G/C/C/G/G, G/G/C/G/C/C/A/G and G/G/C/G/C/T/G/G haplotypes were between dyslipidemia group and control group statistically significantly different (p < 0.05 in each case). The polymorphism of CETP genes rs708272, rs3764261, rs1800775, rs711752, rs12149545 was closely related to the dyslipidemia in the Xinjiang Uyghur and Kazakh ethnic groups; and the rs708272 T, rs3764261 T, rs711752 A, and rs12149545 A alleles could reduce risk of dyslipidemia in the Uyghur and Kazakh populations, however, the rs1800775 C allele showed risk factors.
Collapse
Affiliation(s)
- Shuxia Guo
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Yunhua Hu
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Yusong Ding
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Jiaming Liu
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Mei Zhang
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Rulin Ma
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Heng Guo
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Kui Wang
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Jia He
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Yizhong Yan
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Dongsheng Rui
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Feng Sun
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Lati Mu
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Qiang Niu
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Jingyu Zhang
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| | - Shugang Li
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases of the Ministry of Education, Shihezi University School of Medicine, Shihezi 832002, China.
| |
Collapse
|
29
|
Leusink M, Onland-Moret NC, de Bakker PIW, de Boer A, Maitland-van der Zee AH. Seventeen years of statin pharmacogenetics: a systematic review. Pharmacogenomics 2015; 17:163-80. [PMID: 26670324 DOI: 10.2217/pgs.15.158] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
AIM We evaluated the evidence of pharmacogenetic associations with statins in a systematic review. METHODS Two separate outcomes were considered of interest: modification of low-density lipoprotein cholesterol (LDL-C) response and modification of risk for cardiovascular events. RESULTS In candidate gene studies, 141 loci were claimed to be associated with LDL-C response. Only 5% of these associations were positively replicated. In addition, six genome-wide association studies of LDL-C response identified common SNPs in APOE, LPA, SLCO1B1, SORT1 and ABCG2 at genome-wide significance. None of the investigated SNPs consistently affected the risk reduction for cardiovascular events. CONCLUSION Only five genetic loci were consistently associated with LDL-C response. However, as effect sizes are modest, there is no evidence for the value of genetic testing in clinical practice.
Collapse
Affiliation(s)
- Maarten Leusink
- Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands.,Julius Center for Health Sciences & Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - N Charlotte Onland-Moret
- Julius Center for Health Sciences & Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Paul I W de Bakker
- Julius Center for Health Sciences & Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Medical Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anthonius de Boer
- Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Anke H Maitland-van der Zee
- Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
30
|
Ding YY, Zhang W, Zhang MQ, Fu K, Chen WP, Ding C, He XL, Zhang XD, Huang L, Yin ZJ. Functional and association studies of the cholesteryl ester transfer protein (CETP) gene in a Wannan Black pig model. Anim Genet 2015; 46:702-6. [PMID: 26477338 DOI: 10.1111/age.12370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2015] [Indexed: 10/22/2022]
Abstract
Some polymorphisms of the human CETP gene are causally and significantly associated with serum lipids levels; however, the information regarding this gene in pigs is sparse. To evaluate the effects of CETP on blood lipid traits and fat deposition in pig, porcine CETP tissue expression patterns were observed by quantitative real-time polymerase chain reaction (qPCR) first. High expression was detected in liver, spleen, gluteus medius (GM) muscle and backfat. A de novo polymorphism (AF333037:g.795C>T) in the intron 1 region of porcine CETP was identified. This polymorphism was further genotyped by direct sequencing of the PCR products of 390 Wannan Black pigs, a Chinese native breed population. Association analyses at 45 and 300 days of age revealed highly significant associations between CETP genotypes and serum lipid traits. Furthermore, this polymorphism was proved to be associated with differences in liver CETP mRNA levels: pigs at 300 days of age with the TT genotype had higher levels than did those with other genotypes (P = 0.021). Additionally, analysis at 300 days of age showed that GM CETP mRNA expression correlated positively with serum lipids levels as well as with carcass backfat thickness and intramuscular fat content in GM. These results indicate that CETP is involved in serum, adipose and muscle lipid metabolism in pigs. The mechanisms underlying such relationships and their functional implications are worthy of further research.
Collapse
Affiliation(s)
- Y Y Ding
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, Department of Animal Science, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, 230036, P. R., China
| | - W Zhang
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, Department of Animal Science, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, 230036, P. R., China
| | - M Q Zhang
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, Department of Animal Science, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, 230036, P. R., China
| | - K Fu
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, Department of Animal Science, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, 230036, P. R., China
| | - W P Chen
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, Department of Animal Science, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, 230036, P. R., China
| | - C Ding
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, Department of Animal Science, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, 230036, P. R., China
| | - X L He
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, Department of Animal Science, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, 230036, P. R., China
| | - X D Zhang
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, Department of Animal Science, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, 230036, P. R., China
| | - L Huang
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, Department of Animal Science, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, 230036, P. R., China
| | - Z J Yin
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, Department of Animal Science, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, 230036, P. R., China
| |
Collapse
|
31
|
Hepatic Long Intergenic Noncoding RNAs: High Promoter Conservation and Dynamic, Sex-Dependent Transcriptional Regulation by Growth Hormone. Mol Cell Biol 2015; 36:50-69. [PMID: 26459762 DOI: 10.1128/mcb.00861-15] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 10/02/2015] [Indexed: 01/04/2023] Open
Abstract
Long intergenic noncoding RNAs (lincRNAs) are increasingly recognized as key chromatin regulators, yet few studies have characterized lincRNAs in a single tissue under diverse conditions. Here, we analyzed 45 mouse liver RNA sequencing (RNA-Seq) data sets collected under diverse conditions to systematically characterize 4,961 liver lincRNAs, 59% of them novel, with regard to gene structures, species conservation, chromatin accessibility, transcription factor binding, and epigenetic states. To investigate the potential for functionality, we focused on the responses of the liver lincRNAs to growth hormone stimulation, which imparts clinically relevant sex differences to hepatic metabolism and liver disease susceptibility. Sex-biased expression characterized 247 liver lincRNAs, with many being nuclear RNA enriched and regulated by growth hormone. The sex-biased lincRNA genes are enriched for nearby and correspondingly sex-biased accessible chromatin regions, as well as sex-biased binding sites for growth hormone-regulated transcriptional activators (STAT5, hepatocyte nuclear factor 6 [HNF6], FOXA1, and FOXA2) and transcriptional repressors (CUX2 and BCL6). Repression of female-specific lincRNAs in male liver, but not that of male-specific lincRNAs in female liver, was associated with enrichment of H3K27me3-associated inactive states and poised (bivalent) enhancer states. Strikingly, we found that liver-specific lincRNA gene promoters are more highly species conserved and have a significantly higher frequency of proximal binding by liver transcription factors than liver-specific protein-coding gene promoters. Orthologs for many liver lincRNAs were identified in one or more supraprimates, including two rat lincRNAs showing the same growth hormone-regulated, sex-biased expression as their mouse counterparts. This integrative analysis of liver lincRNA chromatin states, transcription factor occupancy, and growth hormone regulation provides novel insights into the expression of sex-specific lincRNAs and their potential for regulation of sex differences in liver physiology and disease.
Collapse
|
32
|
Koopal C, van der Graaf Y, Asselbergs FW, Westerink J, Visseren FLJ. Association between CETP gene polymorphism, insulin resistance and risk of diabetes mellitus in patients with vascular disease. Atherosclerosis 2015; 242:605-10. [PMID: 26318399 DOI: 10.1016/j.atherosclerosis.2015.08.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 06/24/2015] [Accepted: 08/10/2015] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Genetic inhibition of Cholesteryl Ester Transfer Protein (CETP) might be associated with insulin resistance and incident type 2 diabetes mellitus (T2DM). This study investigated the relation between a genetic variant in the CETP gene and measures of insulin resistance and incident T2DM in patients with manifest cardiovascular disease (CVD). Furthermore the effect on risk of recurrent cardiovascular events was investigated. METHODS SMART is a prospective cohort study performed in 5601 patients with clinically manifest CVD. We selected a variant (rs3764261) associated with reduced CETP activity and increased levels of HDL cholesterol (HDL-C). Patients were divided in three groups: 2640 wild type patients (GG), 2420 heterozygotes for rs3764261 (GT) and 541 homozygotes for rs3764261 (TT). Regression analyses were performed using an additive model. RESULTS The study population consisted of 4656 patients without T2DM and 945 patients with T2DM at baseline. Presence of rs3764261 was associated with increased HDL-C in patients without T2DM (β 0.106, 95%CI 0.083-0.128) and with T2DM (β 0.043, 95%CI 0.007-0.078). During a median follow up of 7.2 years (IQR 4.7-10.2) 427 incident T2DM occurred. Presence of rs3764261 was not related to incident T2DM (HR 0.96, 95%CI 0.83-1.11) in patients without T2DM at baseline. Furthermore, presence of rs3764261 was not related to insulin resistance (glucose, insulin, HOMA-IR, HbA1c) or recurrent CVD (HR 0.92, 95%CI 0.84-1.02). CONCLUSION Presence of CETP SNP rs3764261 is not associated with insulin resistance and incident T2DM in patients with clinically manifest vascular disease. Furthermore, no effect of rs3764261 on the risk of recurrent CVD was observed.
Collapse
Affiliation(s)
- Charlotte Koopal
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Yolanda van der Graaf
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Folkert W Asselbergs
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands; Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, The Netherlands; Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, United Kingdom
| | - Jan Westerink
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frank L J Visseren
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | |
Collapse
|
33
|
Lu R, Smith RM, Seweryn M, Wang D, Hartmann K, Webb A, Sadee W, Rempala GA. Analyzing allele specific RNA expression using mixture models. BMC Genomics 2015; 16:566. [PMID: 26231172 PMCID: PMC4521363 DOI: 10.1186/s12864-015-1749-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 07/03/2015] [Indexed: 11/10/2022] Open
Abstract
Background Measuring allele-specific RNA expression provides valuable insights into cis-acting genetic and epigenetic regulation of gene expression. Widespread adoption of high-throughput sequencing technologies for studying RNA expression (RNA-Seq) permits measurement of allelic RNA expression imbalance (AEI) at heterozygous single nucleotide polymorphisms (SNPs) across the entire transcriptome, and this approach has become especially popular with the emergence of large databases, such as GTEx. However, the existing binomial-type methods used to model allelic expression from RNA-seq assume a strong negative correlation between reference and variant allele reads, which may not be reasonable biologically. Results Here we propose a new strategy for AEI analysis using RNA-seq data. Under the null hypothesis of no AEI, a group of SNPs (possibly across multiple genes) is considered comparable if their respective total sums of the allelic reads are of similar magnitude. Within each group of “comparable” SNPs, we identify SNPs with AEI signal by fitting a mixture of folded Skellam distributions to the absolute values of read differences. By applying this methodology to RNA-Seq data from human autopsy brain tissues, we identified numerous instances of moderate to strong imbalanced allelic RNA expression at heterozygous SNPs. Findings with SLC1A3 mRNA exhibiting known expression differences are discussed as examples. Conclusion The folded Skellam mixture model searches for SNPs with significant difference between reference and variant allele reads (adjusted for different library sizes), using information from a group of “comparable” SNPs across multiple genes. This model is particularly suitable for performing AEI analysis on genes with few heterozygous SNPs available from RNA-seq, and it can fit over-dispersed read counts without specifying the direction of the correlation between reference and variant alleles. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1749-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rong Lu
- Division of Biostatistics, College of Public Health, The Ohio State University, Columbus, OH, 43210, USA
| | - Ryan M Smith
- Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Michal Seweryn
- Division of Biostatistics, College of Public Health, The Ohio State University, Columbus, OH, 43210, USA.,Mathematical Biosciences Institute, The Ohio State University, Columbus, OH, 43201, USA
| | - Danxin Wang
- Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Katherine Hartmann
- Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Amy Webb
- Department of Biomedical Informatics, Program in Pharmacogenomics, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Wolfgang Sadee
- Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Grzegorz A Rempala
- Division of Biostatistics, College of Public Health, The Ohio State University, Columbus, OH, 43210, USA. .,Mathematical Biosciences Institute, The Ohio State University, Columbus, OH, 43201, USA.
| |
Collapse
|
34
|
Suhy A, Hartmann K, Papp AC, Wang D, Sadee W. Regulation of cholesteryl ester transfer protein expression by upstream polymorphisms: reduced expression associated with rs247616. Pharmacogenet Genomics 2015; 25:394-401. [PMID: 26061659 PMCID: PMC4499003 DOI: 10.1097/fpc.0000000000000151] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Cholesteryl ester transfer protein (CETP) is involved in reverse cholesterol transport by exchanging cholesteryl esters for triglycerides between high-density lipoprotein and low-density lipoprotein particles, effectively decreasing high-density lipoprotein cholesterol levels. Variants within a large haplotype block upstream of CETP (rs247616, rs173539) have been shown to be significantly associated with reduced expression; however, the underlying mechanism has not been identified. METHODS We analyzed the linkage structure of our top candidate single-nucleotide polymorphism (SNP), rs247616, and assessed each SNP of the haplotype block for potential interactions with transcription factor binding sites. We then used a reporter gene assay to assess the effect of three SNPs (rs247616, rs173539, and rs1723150) on expression in vitro. RESULTS Several variants in the upstream haplotype, including rs247616, rs173539, and rs1723150, disrupt or generate transcription factor binding sites. In reporter gene assays, rs247616 and rs173539 were found to significantly affected expression in HepG2 cells, whereas rs17231506 had no effect. rs247616 decreased expression by 1.7-fold (P<0.0001), whereas rs173539 increased expression by 2.2-fold (P=0.0006). CONCLUSION SNPs rs247616 and rs173539 are in high linkage disequilibrium (R=0.96, D'=1.00) and have the potential to regulate CETP expression. Although opposing effects suggest that regulation of CETP expression could vary between tissues, the minor allele of rs247616 and SNPs in high linkage with it were found to be associated with reduced expression across all tissues.
Collapse
Affiliation(s)
- Adam Suhy
- Department of Pharmacology, Center for Pharmacogenomics, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | | | | | | | | |
Collapse
|
35
|
Qi Q, Durst R, Schwarzfuchs D, Leitersdorf E, Shpitzen S, Li Y, Wu H, Champagne CM, Hu FB, Stampfer MJ, Bray GA, Sacks FM, Shai I, Qi L. CETP genotype and changes in lipid levels in response to weight-loss diet intervention in the POUNDS LOST and DIRECT randomized trials. J Lipid Res 2014; 56:713-721. [PMID: 25548261 DOI: 10.1194/jlr.p055715] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Little is known about whether cholesteryl ester transfer protein (CETP) genetic variation may modify the effect of weight-loss diets varying in fat content on changes in lipid levels. We analyzed the interaction between the CETP variant rs3764261 and dietary interventions on changes in lipid levels among 732 overweight/obese adults from a 2 year randomized weight-loss trial [Preventing Overweight Using Novel Dietary Strategies (POUNDS LOST)], and replicated the findings in 171 overweight/obese adults from an independent 2 year weight-loss trial [Dietary Intervention Randomized Controlled Trial (DIRECT)]. In the POUNDS LOST, participants with the CETP rs3764261 CC genotype on the high-fat diet had larger increases in HDL cholesterol (P = 0.001) and decreases in triglycerides (P = 0.007) than those on the low-fat diet at 6 months, while no significant difference between these two diets was observed among participants carrying other genotypes. The gene-diet interactions on changes in HDL-cholesterol and tri-glyc-erides were replicated in the DIRECT (pooled P for interaction ≤ 0.01). Similar results on trajectory of changes in HDL cholesterol and triglycerides over the 2 year intervention were observed in both trials. Our study provides replicable evidence that individuals with the CETP rs3764261 CC genotype might derive greater effects on raising HDL cholesterol and lowering triglycerides by choosing a low-carbohydrate/high-fat weight-loss diet instead of a low-fat diet.
Collapse
Affiliation(s)
- Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY; Departments of Nutrition Harvard School of Public Health, Boston, MA
| | - Ronen Durst
- Cardiology Department Hadassah Hebrew University Medical Center, Jerusalem, Israel; Center for Research, Prevention, and Treatment of Atherosclerosis Internal Medicine Department, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | | - Eran Leitersdorf
- Center for Research, Prevention, and Treatment of Atherosclerosis Internal Medicine Department, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Shoshi Shpitzen
- Center for Research, Prevention, and Treatment of Atherosclerosis Internal Medicine Department, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Yanping Li
- Departments of Nutrition Harvard School of Public Health, Boston, MA
| | - Hongyu Wu
- Departments of Nutrition Harvard School of Public Health, Boston, MA
| | - Catherine M Champagne
- Pennington Biomedical Research Center of the Louisiana State University System, Baton Rouge, LA
| | - Frank B Hu
- Departments of Nutrition Harvard School of Public Health, Boston, MA; Epidemiology, Harvard School of Public Health, Boston, MA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Meir J Stampfer
- Departments of Nutrition Harvard School of Public Health, Boston, MA; Epidemiology, Harvard School of Public Health, Boston, MA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - George A Bray
- Pennington Biomedical Research Center of the Louisiana State University System, Baton Rouge, LA
| | - Frank M Sacks
- Departments of Nutrition Harvard School of Public Health, Boston, MA
| | - Iris Shai
- Department of Public Health, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Lu Qi
- Departments of Nutrition Harvard School of Public Health, Boston, MA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.
| |
Collapse
|
36
|
Barrie ES, Weinshenker D, Verma A, Pendergrass SA, Lange LA, Ritchie MD, Wilson JG, Kuivaniemi H, Tromp G, Carey DJ, Gerhard GS, Brilliant MH, Hebbring SJ, Cubells JF, Pinsonneault JK, Norman GJ, Sadee W. Regulatory polymorphisms in human DBH affect peripheral gene expression and sympathetic activity. Circ Res 2014; 115:1017-25. [PMID: 25326128 DOI: 10.1161/circresaha.116.304398] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
RATIONALE Dopamine β-hydroxylase (DBH) catalyzes the conversion of dopamine to norepinephrine in the central nervous system and peripherally. DBH variants are associated with large changes in circulating DBH and implicated in multiple disorders; yet causal relationships and tissue-specific effects remain unresolved. OBJECTIVE To characterize regulatory variants in DBH, effect on mRNA expression, and role in modulating sympathetic tone and disease risk. METHODS AND RESULTS Analysis of DBH mRNA in human tissues confirmed high expression in the locus coeruleus and adrenal gland, but also in sympathetically innervated organs (liver>lung>heart). Allele-specific mRNA assays revealed pronounced allelic expression differences in the liver (2- to 11-fold) attributable to promoter rs1611115 and exon 2 rs1108580, but only small differences in locus coeruleus and adrenals. These alleles were also associated with significantly reduced mRNA expression in liver and lung. Although DBH protein is expressed in other sympathetically innervated organs, mRNA levels were too low for analysis. In mice, hepatic Dbh mRNA levels correlated with cardiovascular risk phenotypes. The minor alleles of rs1611115 and rs1108580 were associated with sympathetic phenotypes, including angina pectoris. Testing combined effects of these variants suggested protection against myocardial infarction in 3 separate clinical cohorts. CONCLUSIONS We demonstrate profound effects of DBH variants on expression in 2 sympathetically innervated organs, liver and lung, but not in adrenals and brain. Preliminary results demonstrate an association of these variants with clinical phenotypes responsive to peripheral sympathetic tone. We hypothesize that in addition to endocrine effects via circulating DBH and norepinephrine, the variants act in sympathetically innervated target organs.
Collapse
Affiliation(s)
- Elizabeth S Barrie
- From the Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus (E.S.B., J.K.P., W.S.); Department of Human Genetics, Emory University School of Medicine, Atlanta, GA (D.W., J.F.C.); Center for Systems Genomics, Pennsylvania State University, University Park (A.V., S.A.P., M.D.R.); Department of Genetics, University of North Carolina School of Medicine, Chapel Hill (L.A.L.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.G.W.); The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA (H.K., G.T., D.J.C.); Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey (G.S.G.); Center for Human Genetics, Marshfield Clinic Research Foundation, WI (M.H.B., S.J.H.); and Department of Psychology, The University of Chicago, IL (G.J.N.)
| | - David Weinshenker
- From the Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus (E.S.B., J.K.P., W.S.); Department of Human Genetics, Emory University School of Medicine, Atlanta, GA (D.W., J.F.C.); Center for Systems Genomics, Pennsylvania State University, University Park (A.V., S.A.P., M.D.R.); Department of Genetics, University of North Carolina School of Medicine, Chapel Hill (L.A.L.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.G.W.); The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA (H.K., G.T., D.J.C.); Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey (G.S.G.); Center for Human Genetics, Marshfield Clinic Research Foundation, WI (M.H.B., S.J.H.); and Department of Psychology, The University of Chicago, IL (G.J.N.)
| | - Anurag Verma
- From the Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus (E.S.B., J.K.P., W.S.); Department of Human Genetics, Emory University School of Medicine, Atlanta, GA (D.W., J.F.C.); Center for Systems Genomics, Pennsylvania State University, University Park (A.V., S.A.P., M.D.R.); Department of Genetics, University of North Carolina School of Medicine, Chapel Hill (L.A.L.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.G.W.); The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA (H.K., G.T., D.J.C.); Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey (G.S.G.); Center for Human Genetics, Marshfield Clinic Research Foundation, WI (M.H.B., S.J.H.); and Department of Psychology, The University of Chicago, IL (G.J.N.)
| | - Sarah A Pendergrass
- From the Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus (E.S.B., J.K.P., W.S.); Department of Human Genetics, Emory University School of Medicine, Atlanta, GA (D.W., J.F.C.); Center for Systems Genomics, Pennsylvania State University, University Park (A.V., S.A.P., M.D.R.); Department of Genetics, University of North Carolina School of Medicine, Chapel Hill (L.A.L.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.G.W.); The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA (H.K., G.T., D.J.C.); Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey (G.S.G.); Center for Human Genetics, Marshfield Clinic Research Foundation, WI (M.H.B., S.J.H.); and Department of Psychology, The University of Chicago, IL (G.J.N.)
| | - Leslie A Lange
- From the Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus (E.S.B., J.K.P., W.S.); Department of Human Genetics, Emory University School of Medicine, Atlanta, GA (D.W., J.F.C.); Center for Systems Genomics, Pennsylvania State University, University Park (A.V., S.A.P., M.D.R.); Department of Genetics, University of North Carolina School of Medicine, Chapel Hill (L.A.L.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.G.W.); The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA (H.K., G.T., D.J.C.); Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey (G.S.G.); Center for Human Genetics, Marshfield Clinic Research Foundation, WI (M.H.B., S.J.H.); and Department of Psychology, The University of Chicago, IL (G.J.N.)
| | - Marylyn D Ritchie
- From the Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus (E.S.B., J.K.P., W.S.); Department of Human Genetics, Emory University School of Medicine, Atlanta, GA (D.W., J.F.C.); Center for Systems Genomics, Pennsylvania State University, University Park (A.V., S.A.P., M.D.R.); Department of Genetics, University of North Carolina School of Medicine, Chapel Hill (L.A.L.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.G.W.); The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA (H.K., G.T., D.J.C.); Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey (G.S.G.); Center for Human Genetics, Marshfield Clinic Research Foundation, WI (M.H.B., S.J.H.); and Department of Psychology, The University of Chicago, IL (G.J.N.)
| | - James G Wilson
- From the Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus (E.S.B., J.K.P., W.S.); Department of Human Genetics, Emory University School of Medicine, Atlanta, GA (D.W., J.F.C.); Center for Systems Genomics, Pennsylvania State University, University Park (A.V., S.A.P., M.D.R.); Department of Genetics, University of North Carolina School of Medicine, Chapel Hill (L.A.L.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.G.W.); The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA (H.K., G.T., D.J.C.); Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey (G.S.G.); Center for Human Genetics, Marshfield Clinic Research Foundation, WI (M.H.B., S.J.H.); and Department of Psychology, The University of Chicago, IL (G.J.N.)
| | - Helena Kuivaniemi
- From the Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus (E.S.B., J.K.P., W.S.); Department of Human Genetics, Emory University School of Medicine, Atlanta, GA (D.W., J.F.C.); Center for Systems Genomics, Pennsylvania State University, University Park (A.V., S.A.P., M.D.R.); Department of Genetics, University of North Carolina School of Medicine, Chapel Hill (L.A.L.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.G.W.); The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA (H.K., G.T., D.J.C.); Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey (G.S.G.); Center for Human Genetics, Marshfield Clinic Research Foundation, WI (M.H.B., S.J.H.); and Department of Psychology, The University of Chicago, IL (G.J.N.)
| | - Gerard Tromp
- From the Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus (E.S.B., J.K.P., W.S.); Department of Human Genetics, Emory University School of Medicine, Atlanta, GA (D.W., J.F.C.); Center for Systems Genomics, Pennsylvania State University, University Park (A.V., S.A.P., M.D.R.); Department of Genetics, University of North Carolina School of Medicine, Chapel Hill (L.A.L.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.G.W.); The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA (H.K., G.T., D.J.C.); Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey (G.S.G.); Center for Human Genetics, Marshfield Clinic Research Foundation, WI (M.H.B., S.J.H.); and Department of Psychology, The University of Chicago, IL (G.J.N.)
| | - David J Carey
- From the Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus (E.S.B., J.K.P., W.S.); Department of Human Genetics, Emory University School of Medicine, Atlanta, GA (D.W., J.F.C.); Center for Systems Genomics, Pennsylvania State University, University Park (A.V., S.A.P., M.D.R.); Department of Genetics, University of North Carolina School of Medicine, Chapel Hill (L.A.L.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.G.W.); The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA (H.K., G.T., D.J.C.); Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey (G.S.G.); Center for Human Genetics, Marshfield Clinic Research Foundation, WI (M.H.B., S.J.H.); and Department of Psychology, The University of Chicago, IL (G.J.N.)
| | - Glenn S Gerhard
- From the Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus (E.S.B., J.K.P., W.S.); Department of Human Genetics, Emory University School of Medicine, Atlanta, GA (D.W., J.F.C.); Center for Systems Genomics, Pennsylvania State University, University Park (A.V., S.A.P., M.D.R.); Department of Genetics, University of North Carolina School of Medicine, Chapel Hill (L.A.L.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.G.W.); The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA (H.K., G.T., D.J.C.); Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey (G.S.G.); Center for Human Genetics, Marshfield Clinic Research Foundation, WI (M.H.B., S.J.H.); and Department of Psychology, The University of Chicago, IL (G.J.N.)
| | - Murray H Brilliant
- From the Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus (E.S.B., J.K.P., W.S.); Department of Human Genetics, Emory University School of Medicine, Atlanta, GA (D.W., J.F.C.); Center for Systems Genomics, Pennsylvania State University, University Park (A.V., S.A.P., M.D.R.); Department of Genetics, University of North Carolina School of Medicine, Chapel Hill (L.A.L.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.G.W.); The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA (H.K., G.T., D.J.C.); Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey (G.S.G.); Center for Human Genetics, Marshfield Clinic Research Foundation, WI (M.H.B., S.J.H.); and Department of Psychology, The University of Chicago, IL (G.J.N.)
| | - Scott J Hebbring
- From the Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus (E.S.B., J.K.P., W.S.); Department of Human Genetics, Emory University School of Medicine, Atlanta, GA (D.W., J.F.C.); Center for Systems Genomics, Pennsylvania State University, University Park (A.V., S.A.P., M.D.R.); Department of Genetics, University of North Carolina School of Medicine, Chapel Hill (L.A.L.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.G.W.); The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA (H.K., G.T., D.J.C.); Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey (G.S.G.); Center for Human Genetics, Marshfield Clinic Research Foundation, WI (M.H.B., S.J.H.); and Department of Psychology, The University of Chicago, IL (G.J.N.)
| | - Joseph F Cubells
- From the Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus (E.S.B., J.K.P., W.S.); Department of Human Genetics, Emory University School of Medicine, Atlanta, GA (D.W., J.F.C.); Center for Systems Genomics, Pennsylvania State University, University Park (A.V., S.A.P., M.D.R.); Department of Genetics, University of North Carolina School of Medicine, Chapel Hill (L.A.L.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.G.W.); The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA (H.K., G.T., D.J.C.); Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey (G.S.G.); Center for Human Genetics, Marshfield Clinic Research Foundation, WI (M.H.B., S.J.H.); and Department of Psychology, The University of Chicago, IL (G.J.N.)
| | - Julia K Pinsonneault
- From the Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus (E.S.B., J.K.P., W.S.); Department of Human Genetics, Emory University School of Medicine, Atlanta, GA (D.W., J.F.C.); Center for Systems Genomics, Pennsylvania State University, University Park (A.V., S.A.P., M.D.R.); Department of Genetics, University of North Carolina School of Medicine, Chapel Hill (L.A.L.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.G.W.); The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA (H.K., G.T., D.J.C.); Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey (G.S.G.); Center for Human Genetics, Marshfield Clinic Research Foundation, WI (M.H.B., S.J.H.); and Department of Psychology, The University of Chicago, IL (G.J.N.)
| | - Greg J Norman
- From the Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus (E.S.B., J.K.P., W.S.); Department of Human Genetics, Emory University School of Medicine, Atlanta, GA (D.W., J.F.C.); Center for Systems Genomics, Pennsylvania State University, University Park (A.V., S.A.P., M.D.R.); Department of Genetics, University of North Carolina School of Medicine, Chapel Hill (L.A.L.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.G.W.); The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA (H.K., G.T., D.J.C.); Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey (G.S.G.); Center for Human Genetics, Marshfield Clinic Research Foundation, WI (M.H.B., S.J.H.); and Department of Psychology, The University of Chicago, IL (G.J.N.)
| | - Wolfgang Sadee
- From the Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus (E.S.B., J.K.P., W.S.); Department of Human Genetics, Emory University School of Medicine, Atlanta, GA (D.W., J.F.C.); Center for Systems Genomics, Pennsylvania State University, University Park (A.V., S.A.P., M.D.R.); Department of Genetics, University of North Carolina School of Medicine, Chapel Hill (L.A.L.); Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.G.W.); The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA (H.K., G.T., D.J.C.); Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey (G.S.G.); Center for Human Genetics, Marshfield Clinic Research Foundation, WI (M.H.B., S.J.H.); and Department of Psychology, The University of Chicago, IL (G.J.N.).
| |
Collapse
|
37
|
Sadee W, Hartmann K, Seweryn M, Pietrzak M, Handelman SK, Rempala GA. Missing heritability of common diseases and treatments outside the protein-coding exome. Hum Genet 2014; 133:1199-215. [PMID: 25107510 PMCID: PMC4169001 DOI: 10.1007/s00439-014-1476-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 07/23/2014] [Indexed: 02/07/2023]
Abstract
Genetic factors strongly influence risk of common human diseases and treatment outcomes but the causative variants remain largely unknown; this gap has been called the 'missing heritability'. We propose several hypotheses that in combination have the potential to narrow the gap. First, given a multi-stage path from wellness to disease, we propose that common variants under positive evolutionary selection represent normal variation and gate the transition between wellness and an 'off-well' state, revealing adaptations to changing environmental conditions. In contrast, genome-wide association studies (GWAS) focus on deleterious variants conveying disease risk, accelerating the path from off-well to illness and finally specific diseases, while common 'normal' variants remain hidden in the noise. Second, epistasis (dynamic gene-gene interactions) likely assumes a central role in adaptations and evolution; yet, GWAS analyses currently are poorly designed to reveal epistasis. As gene regulation is germane to adaptation, we propose that epistasis among common normal regulatory variants, or between common variants and less frequent deleterious variants, can have strong protective or deleterious phenotypic effects. These gene-gene interactions can be highly sensitive to environmental stimuli and could account for large differences in drug response between individuals. Residing largely outside the protein-coding exome, common regulatory variants affect either transcription of coding and non-coding RNAs (regulatory SNPs, or rSNPs) or RNA functions and processing (structural RNA SNPs, or srSNPs). Third, with the vast majority of causative variants yet to be discovered, GWAS rely on surrogate markers, a confounding factor aggravated by the presence of more than one causative variant per gene and by epistasis. We propose that the confluence of these factors may be responsible to large extent for the observed heritability gap.
Collapse
Affiliation(s)
- Wolfgang Sadee
- Department of Pharmacology, Center for Pharmacogenomics, College of Medicine, The Ohio State University Wexner Medical Center, 5184A Graves Hall, 333 West 10th Avenue, Columbus, OH, 43210, USA,
| | | | | | | | | | | |
Collapse
|
38
|
Abstract
Inhibition of cholesteryl ester transfer protein (CETP) lowers plasma low-density lipoprotein cholesterol concentration and raises high-density lipoprotein (HDL) cholesterol, suggesting it might prevent cardiovascular disease (CVD). From the outset, however, the concept has been controversial owing to uncertainty about its effects on HDL function and reverse cholesterol transport (RCT). Although there has long been good evidence that CETP inhibition reduces atherosclerosis in rabbits, the first information on CETP as a CVD risk factor in a prospectively followed cohort was not published until after the first Phase 3 trial of a CETP inhibitor had begun. The worrying finding that CVD incidence was related inversely to plasma CETP has since been reproduced in each of five further prospective cohort studies. Similar results were obtained in subjects on or off statin therapy, for first and second CVD events, and for mortality as well as CVD morbidity. Additionally, two recent studies have found alleles of the CETP gene that lower hepatic CETP secretion to be associated with an increased risk of myocardial infarction. Meanwhile, CETP gene transfer in mice was found to increase RCT from peripheral macrophages in vivo, and human plasma with high CETP activity was shown to have a greater capacity to remove cholesterol from cultured cells than plasma with low activity. This mounting evidence for a protective function of CETP has been given remarkably little attention, and indeed was not mentioned in several recent reviews. It appears to show that CETP inhibition does not test the HDL hypothesis as originally hoped, and raises a pressing ethical issue regarding two Phase 3 trials of inhibitors, involving more than forty thousand subjects, which are currently in progress. As the weight of evidence now clearly supports an adverse effect of CETP inhibition on CVD, an urgent review is needed to determine if these trials should be discontinued.
Collapse
|
39
|
Hellwege JN, Palmer ND, Raffield LM, Ng MCY, Hawkins GA, Long J, Lorenzo C, Norris JM, Ida Chen YD, Speliotes EK, Rotter JI, Langefeld CD, Wagenknecht LE, Bowden DW. Genome-wide family-based linkage analysis of exome chip variants and cardiometabolic risk. Genet Epidemiol 2014; 38:345-52. [PMID: 24719370 DOI: 10.1002/gepi.21801] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 02/14/2014] [Accepted: 02/28/2014] [Indexed: 01/31/2023]
Abstract
Linkage analysis of complex traits has had limited success in identifying trait-influencing loci. Recently, coding variants have been implicated as the basis for some biomedical associations. We tested whether coding variants are the basis for linkage peaks of complex traits in 42 African-American (n = 596) and 90 Hispanic (n = 1,414) families in the Insulin Resistance Atherosclerosis Family Study (IRASFS) using Illumina HumanExome Beadchips. A total of 92,157 variants in African Americans (34%) and 81,559 (31%) in Hispanics were polymorphic and tested using two-point linkage and association analyses with 37 cardiometabolic phenotypes. In African Americans 77 LOD scores greater than 3 were observed. The highest LOD score was 4.91 with the APOE SNP rs7412 (MAF = 0.13) with plasma apolipoprotein B (ApoB). This SNP was associated with ApoB (P-value = 4 × 10(-19)) and accounted for 16.2% of the variance in African Americans. In Hispanic families, 104 LOD scores were greater than 3. The strongest evidence of linkage (LOD = 4.29) was with rs5882 (MAF = 0.46) in CETP with HDL. CETP variants were strongly associated with HDL (0.00049 < P-value <4.6 × 10(-12)), accounting for up to 4.5% of the variance. These loci have previously been shown to have effects on the biomedical traits evaluated here. Thus, evidence of strong linkage in this genome wide survey of primarily coding variants was uncommon. Loci with strong evidence of linkage was characterized by large contributions to the variance, and, in these cases, are common variants. Less compelling evidence of linkage and association was observed with additional loci that may require larger family sets to confirm.
Collapse
Affiliation(s)
- Jacklyn N Hellwege
- Molecular Genetics and Genomics Program, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America; Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America; Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Wang D, Zhou J, Hou X, Nguyen DH, Cao G, Li G, Qiu G, Zhang K, Zhang M, Su Z. CETP Gene may be Associated with Advanced Age-Related Macular Degeneration in the Chinese Population. Ophthalmic Genet 2014; 36:303-8. [PMID: 24498989 DOI: 10.3109/13816810.2014.881506] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES This study aims to investigate whether variations in LIPC, CETP, ABCA1 and LPL, which are involved in high-density lipoprotein (HDL) metabolism, are associated with advanced age-related macular degeneration (AMD) in the Chinese population. DESIGN AND METHODS A total of 119 Chinese patients with advanced AMD and 99 control individuals were recruited. Genomic DNA was extracted from peripheral blood leukocytes. Genotypes of seven single nucleotide polymorphisms (SNPs) including rs1061170 and rs1410996 in CFH, rs10490924 in HTRA1, rs10468017 in LIPC, rs3764261 in CETP, rs1883025 in ABCA1 and rs12678919 near LPL were determined by polymerase chain reaction (PCR) followed by allele-specific restriction enzyme digestion or SNaPshot. Unconditional logistic regression analyses were performed to generate a risk predictive model. RESULTS We observed the frequency of allele A of rs3764261 in CETP to be significantly lower in advanced AMD after Bonferroni correction (15.5% in patients with AMD and 20.7% in controls; OR = 0.49, 95% CI: 0.29-0.85; p = 0.011). Furthermore, we found that it was also associated with reduced risk of both unilateral AMD (OR = 0.52, 95% CI: 0.28-0.98; p = 0.043) and bilateral AMD (OR = 0.45, 95% CI: 0.22-0.91; p = 0.026). Rs10468017 in LIPC, rs12678919 near LPL and rs1883025 in ABCA1 were not found to be associated with advanced AMD (all p > 0.05). CONCLUSION Our data suggested that the allele A in rs3764261 in CETP gene may be associated with a decreased risk of advanced AMD in Chinese population.
Collapse
Affiliation(s)
- Dan Wang
- a Molecular Medicine Research Center, State Key Laboratory of Biotherapy .,b Department of Ophthalmology , West China Hospital, Sichuan University , Chengdu , China , and
| | - Jie Zhou
- a Molecular Medicine Research Center, State Key Laboratory of Biotherapy .,b Department of Ophthalmology , West China Hospital, Sichuan University , Chengdu , China , and
| | - Xiaoming Hou
- a Molecular Medicine Research Center, State Key Laboratory of Biotherapy
| | - Duy H Nguyen
- c Department of Ophthalmology , University of California , San Diego , CA , USA
| | - Guiqun Cao
- a Molecular Medicine Research Center, State Key Laboratory of Biotherapy
| | - Gen Li
- a Molecular Medicine Research Center, State Key Laboratory of Biotherapy
| | - Guoxian Qiu
- b Department of Ophthalmology , West China Hospital, Sichuan University , Chengdu , China , and
| | - Kang Zhang
- a Molecular Medicine Research Center, State Key Laboratory of Biotherapy .,c Department of Ophthalmology , University of California , San Diego , CA , USA
| | - Ming Zhang
- b Department of Ophthalmology , West China Hospital, Sichuan University , Chengdu , China , and
| | - Zhiguang Su
- a Molecular Medicine Research Center, State Key Laboratory of Biotherapy
| |
Collapse
|
41
|
Suhy A, Hartmann K, Newman L, Papp A, Toneff T, Hook V, Sadee W. Genetic variants affecting alternative splicing of human cholesteryl ester transfer protein. Biochem Biophys Res Commun 2014; 443:1270-4. [PMID: 24393849 DOI: 10.1016/j.bbrc.2013.12.127] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 12/24/2013] [Indexed: 11/25/2022]
Abstract
Cholesteryl ester transfer protein (CETP) plays an important role in reverse cholesterol transport, with decreased CETP activity increasing HDL levels. Formation of an alternative splice form lacking exon 9 (Δ9-CETP) has been associated with two single nucleotide polymorphisms (SNPs) in high linkage disequilibrium with each other, namely rs9930761 T>C located in intron 8 in a putative splicing branch site and rs5883 C>T in a possible exonic splicing enhancer (ESE) site in exon 9. To assess the relative effect of rs9930761 and rs5883 on splicing, mini-gene constructs spanning CETP exons 8 to 10, carrying all four possible allele combinations, were transfected into HEK293 and HepG2 cells. The minor T allele of rs5883 enhanced splicing significantly in both cell lines whereas the minor C allele of rs9930761 did not. In combination, the two alleles did not yield greater splicing than the rs5883 T allele alone in HepG2 cells. These results indicate that the genetic effect on CETP splicing is largely attributable to rs5883. We also confirm that Δ9-CETP protein is expressed in the liver but fails to circulate in the blood.
Collapse
Affiliation(s)
- Adam Suhy
- Center for Pharmacogenomics, Department of Pharmacology, College of Medicine, The Ohio State University Wexner Medical Center, 5184A Graves Hall, 333 West 10th Avenue, Columbus, OH 43210, USA.
| | - Katherine Hartmann
- Center for Pharmacogenomics, Department of Pharmacology, College of Medicine, The Ohio State University Wexner Medical Center, 5184A Graves Hall, 333 West 10th Avenue, Columbus, OH 43210, USA
| | - Leslie Newman
- Center for Pharmacogenomics, Department of Pharmacology, College of Medicine, The Ohio State University Wexner Medical Center, 5184A Graves Hall, 333 West 10th Avenue, Columbus, OH 43210, USA
| | - Audrey Papp
- Center for Pharmacogenomics, Department of Pharmacology, College of Medicine, The Ohio State University Wexner Medical Center, 5184A Graves Hall, 333 West 10th Avenue, Columbus, OH 43210, USA
| | - Thomas Toneff
- Skaggs School of Pharmacy and Pharmaceutical Sciences, School of Medicine, Departments of Neuroscience and Pharmacology, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Vivian Hook
- Skaggs School of Pharmacy and Pharmaceutical Sciences, School of Medicine, Departments of Neuroscience and Pharmacology, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Wolfgang Sadee
- Center for Pharmacogenomics, Department of Pharmacology, College of Medicine, The Ohio State University Wexner Medical Center, 5184A Graves Hall, 333 West 10th Avenue, Columbus, OH 43210, USA
| |
Collapse
|
42
|
Wang D, Poi MJ, Sun X, Gaedigk A, Leeder JS, Sadee W. Common CYP2D6 polymorphisms affecting alternative splicing and transcription: long-range haplotypes with two regulatory variants modulate CYP2D6 activity. Hum Mol Genet 2014; 23:268-78. [PMID: 23985325 PMCID: PMC3857955 DOI: 10.1093/hmg/ddt417] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 08/01/2013] [Accepted: 08/22/2013] [Indexed: 01/20/2023] Open
Abstract
Cytochrome P450 2D6 (CYP2D6) is involved in the metabolism of 25% of clinically used drugs. Genetic polymorphisms cause substantial variation in CYP2D6 activity and serve as biomarkers guiding drug therapy. However, genotype-phenotype relationships remain ambiguous except for poor metabolizers carrying null alleles, suggesting the presence of yet unknown genetic variants. Searching for regulatory CYP2D6 polymorphisms, we find that a SNP defining the CYP2D6*2 allele, rs16947 [R296C, 17-60% minor allele frequency (MAF)], previously thought to convey normal activity, alters exon 6 splicing, thereby reducing CYP2D6 expression at least 2-fold. In addition, two completely linked SNPs (rs5758550/rs133333, MAF 13-42%) increase CYP2D6 transcription more than 2-fold, located in a distant downstream enhancer region (>100 kb) that interacts with the CYP2D6 promoter. In high linkage disequilibrium (LD) with each other, rs16947 and the enhancer SNPs form haplotypes that affect CYP2D6 enzyme activity in vivo. In a pediatric cohort of 164 individuals, rs16947 alone (minor haplotype frequency 28%) was associated with reduced CYP2D6 metabolic activity (measured as dextromethorphan/metabolite ratios), whereas rs5758550/rs133333 alone (frequency 3%) resulted in increased CYP2D6 activity, while haplotypes containing both rs16947 and rs5758550/rs133333 were similar to the wild-type. Other alleles used in biomarker panels carrying these variants such as CYP2D6*41 require re-evaluation of independent effects on CYP2D6 activity. The occurrence of two regulatory variants of high frequency and in high LD, residing on a long haplotype, highlights the importance of gene architecture, likely shaped by evolutionary selection pressures, in determining activity of encoded proteins.
Collapse
Affiliation(s)
- Danxin Wang
- Program in Pharmacogenomics, Department of Pharmacology, School of Medicine and
| | - Ming J. Poi
- Department of Pharmacy, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, USA and
| | - Xiaochun Sun
- Program in Pharmacogenomics, Department of Pharmacology, School of Medicine and
| | - Andrea Gaedigk
- Division of Clinical Pharmacology and Therapeutic Innovation, Children's Mercy Hospital, Kansas City, MO, USA
| | - J. Steven Leeder
- Division of Clinical Pharmacology and Therapeutic Innovation, Children's Mercy Hospital, Kansas City, MO, USA
| | - Wolfgang Sadee
- Program in Pharmacogenomics, Department of Pharmacology, School of Medicine and
| |
Collapse
|
43
|
Baldassarre D, Amato M, Frigerio B, Columbo G, Binkley PF, Pandey SR, Suhy AM, Hartmann K, Kitzmiller JP. [Pharmacogenomic Biomarkers for the Prediction of Statin Efficacy and Safety]. GIORNALE ITALIANO DELL'ARTERIOSCLEROSI 2013; 4:23-35. [PMID: 25763360 PMCID: PMC4353572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Affiliation(s)
- Damiano Baldassarre
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italia ; Centro Cardiologico Monzino, Instituto Di Ricovero e Cura a Carattera Scientificao, Milano, Italia
| | - Mauro Amato
- Centro Cardiologico Monzino, Instituto Di Ricovero e Cura a Carattera Scientificao, Milano, Italia
| | - Beatrice Frigerio
- Centro Cardiologico Monzino, Instituto Di Ricovero e Cura a Carattera Scientificao, Milano, Italia
| | - Gualtiero Columbo
- Centro Cardiologico Monzino, Instituto Di Ricovero e Cura a Carattera Scientificao, Milano, Italia
| | - Philip F Binkley
- College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Saurabh R Pandey
- College of Pharmacy, University of Charleston, Charleston, West Virginia 25304, USA
| | - Adam M Suhy
- College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | | | | |
Collapse
|
44
|
Leusink M, Onland-Moret NC, Asselbergs FW, Ding B, Kotti S, van Zuydam NR, Papp AC, Danchin N, Donnelly L, Morris AD, Chasman DI, Doevendans PAFM, Klungel OH, Ridker PM, van Gilst WH, Simon T, Nyberg F, Palmer CNA, Sadee W, van der Harst P, de Bakker PIW, de Boer A, Verstuyft C, Maitland-van der Zee AH. Cholesteryl ester transfer protein polymorphisms, statin use, and their impact on cholesterol levels and cardiovascular events. Clin Pharmacol Ther 2013; 95:314-20. [PMID: 24080640 DOI: 10.1038/clpt.2013.194] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 09/16/2013] [Indexed: 11/09/2022]
Abstract
The association of nonfunctional variants of the cholesteryl ester transfer protein (CETP) with efficacy of statins has been a subject of debate. We evaluated whether three functional CETP variants influence statin efficacy. The effect of CETP genotype on achieved levels of high-density lipoprotein cholesterol (HDLc), low-density lipoprotein cholesterol (LDLc), and total cholesterol during statin treatment was estimated by meta-analysis of the linear regression outcomes of three studies (11,021 individuals). The effect of these single-nucleotide polymorphisms (SNPs) on statin response in protecting against myocardial infarction (MI) was estimated by meta-analysis of statin × SNP interaction terms from logistic regression in five studies (16,570 individuals). The enhancer SNP rs3764261 significantly increased HDLc by 0.02 mmol/l per T allele (P = 6 × 10(-5)) and reduced protection against MI by statins (interaction odds ratio (OR) = 1.19 per T allele; P = 0.04). Focusing on functional CETP variants, we showed that in carriers of the rs3764261 T variant, HDLc increased more during statin treatment, and protection against MI by statins appeared to be reduced as compared with those in noncarriers.
Collapse
Affiliation(s)
- M Leusink
- 1] Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands [2] Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - N C Onland-Moret
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - F W Asselbergs
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - B Ding
- Global Epidemiology, AstraZeneca R&D, Mölndal, Sweden
| | - S Kotti
- Assistance Publique-Hopitaux de Paris, Hopital St. Antoine, URC-EST, Paris, France
| | - N R van Zuydam
- Centre for Pharmacogenomics, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - A C Papp
- Program in Pharmacogenomics, Department of Pharmacology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - N Danchin
- 1] Assistance Publique-Hopitaux de Paris, Hopital Europeen Georges Pompidou, Paris, France [2] Universite Paris-Descartes, Paris, France
| | - L Donnelly
- Centre for Pharmacogenomics, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - A D Morris
- Centre for Pharmacogenomics, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - D I Chasman
- 1] Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA [2] Harvard Medical School, Boston, Massachusetts, USA
| | - P A F M Doevendans
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - O H Klungel
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - P M Ridker
- 1] Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA [2] Harvard Medical School, Boston, Massachusetts, USA
| | - W H van Gilst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - T Simon
- 1] Assistance Publique-Hopitaux de Paris, Hopital St. Antoine, URC-EST, Paris, France [2] Department of Clinical Pharmacology, Universite Pierre et Marie Curie (Paris 6), Paris, France
| | - F Nyberg
- 1] Global Epidemiology, AstraZeneca R&D, Mölndal, Sweden [2] Occupational and Environmental Medicine, Department of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - C N A Palmer
- Centre for Pharmacogenomics, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - W Sadee
- Program in Pharmacogenomics, Department of Pharmacology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - P van der Harst
- 1] Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands [2] Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - P I W de Bakker
- 1] Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands [2] Harvard Medical School, Boston, Massachusetts, USA [3] Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - A de Boer
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - C Verstuyft
- 1] Assistance Publique-Hopitaux de Paris, Hopital Bicetre, Service de Genetique Moleculaire, Pharmacogenetique et Hormonologie, Le Kremlin Bicetre, France [2] Universite Paris-Sud, Le Kremlin-Bicetre, France
| | - A H Maitland-van der Zee
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
45
|
Kitzmiller JP, Binkley PF, Pandey SR, Suhy AM, Baldassarre D, Hartmann K. Statin pharmacogenomics: pursuing biomarkers for predicting clinical outcomes. DISCOVERY MEDICINE 2013; 16:45-51. [PMID: 23911231 PMCID: PMC4039562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Indicated for treating hyperlipidemias and for the prevention of cardiovascular disease (CVD), statins rank among the most commonly prescribed drug classes. While statins are considered to be highly effective in preventing atherosclerotic events, a substantial portion of treated patients still progress to overt CVD. Genetic factors are thought to contribute substantially to treatment outcome. Several candidate genes have been associated with statin dose requirements and treatment outcomes, but a clinically relevant pharmacogenomics test to guide statin therapy has not yet emerged. Here we define basic pharmacogenomics terminology, present strong candidate genes (CETP, HMGCR, SLCO1B1, ABCB1, and CYP3A4/5), and discuss the challenges in developing much-needed statin pharmacogenomics biomarkers for predicting treatment outcomes.
Collapse
|
46
|
Morrison AC, Voorman A, Johnson AD, Liu X, Yu J, Li A, Muzny D, Yu F, Rice K, Zhu C, Bis J, Heiss G, O'Donnell CJ, Psaty BM, Cupples LA, Gibbs R, Boerwinkle E. Whole-genome sequence-based analysis of high-density lipoprotein cholesterol. Nat Genet 2013; 45:899-901. [PMID: 23770607 PMCID: PMC4030301 DOI: 10.1038/ng.2671] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 05/24/2013] [Indexed: 12/15/2022]
Abstract
We describe initial steps for interrogating whole genome sequence (WGS) data to characterize the genetic architecture of a complex trait, such as high density lipoprotein cholesterol (HDL-C). We estimate that common variation contributes more to HDL-C heritability than rare variation, and screening for Mendelian dyslipidemia variants identified individuals with extreme HDL-C. WGS analyses highlight the value of regulatory and non-protein coding regions of the genome in addition to protein coding regions.
Collapse
Affiliation(s)
- Alanna C Morrison
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Xu L, Zhou J, Huang S, Huang Y, LE Y, Jiang D, Wang F, Yang X, Xu W, Huang X, Dong C, Zhang L, Ye M, Lian J, Duan S. An association study between genetic polymorphisms related to lipoprotein-associated phospholipase A(2) and coronary heart disease. Exp Ther Med 2013; 5:742-750. [PMID: 23404648 PMCID: PMC3570076 DOI: 10.3892/etm.2013.911] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Accepted: 01/14/2013] [Indexed: 01/06/2023] Open
Abstract
Previous genome-wide association studies (GWAS) have revealed seven single nucleotide polymorphisms (SNPs) that affect lipoprotein-associated phospholipase A2 (Lp-PLA2) activity or levels in American and European individuals. A total of 290 coronary heart disease (CHD) patients, 198 non-CHD patients and 331 unrelated healthy volunteers were recruited for the present case-control study of Han Chinese. Four SNPs (rs964184 of ZNF259, rs7528419 of CELSR2 and rs7756935 and rs1805017 of PLA2G7) were shown to be significantly associated with CHD. The rs964184-G allele of the ZNF259 gene was identified as a risk factor of CHD in females (odds ratio (OR) =1.49, 95% confidence interval (CI) =1.00–2.22, P=0.05). The rs7528419-G allele of the CELSR2 gene was protective against CHD in males (OR=0.48, 95% CI=0.25–0.93, P=0.04). The other two alleles (rs7756935-C and rs1805017-A) of the PLA2G7 gene acted as protective factors against CHD in females (rs7756935-C: OR=0.59, 95% CI=0.35–1.00, P=0.05; rs1805017-A: OR=0.51, 95% CI=0.28–0.93, P=0.03). Moreover, rs1805017 of the PLA2G7 gene was associated with the severity of CHD only in females (r2=0.02, P=0.04). We identified four Lp-PLA2-associated SNPs significantly associated with CHD in a Han Chinese population. Specifically, rs7528419 was protective factor against CHD in males, while the other two SNPs (rs7756935 and rs1805017 of the PLA2G7 gene) were protective factors against CHD in females and rs964184 of the ZNF259 gene was regarded as a risk factor for CHD in females.
Collapse
Affiliation(s)
- Limin Xu
- School of Medicine, The Affiliated Hospital, Ningbo University, Ningbo, Zhejiang 315211
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|