1
|
Sarakul M, Elzo MA, Koonawootrittriron S, Suwanasopee T, Jattawa D, Laodim T. A comparison of five sets of overlapping and non-overlapping sliding windows for semen production traits in the Thai multibreed dairy population. Anim Biosci 2024; 37:428-436. [PMID: 37946424 PMCID: PMC10915195 DOI: 10.5713/ab.23.0230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/03/2023] [Accepted: 10/02/2023] [Indexed: 11/12/2023] Open
Abstract
OBJECTIVE This study compared five distinct sets of biological pathways and associated genes related to semen volume (VOL), number of sperm (NS), and sperm motility (MOT) in the Thai multibreed dairy population. METHODS The phenotypic data included 13,533 VOL records, 12,773 NS records, and 12,660 MOT records from 131 bulls. The genotypic data consisted of 76,519 imputed and actual single nucleotide polymorphisms (SNPs) from 72 animals. The SNP additive genetic variances for VOL, NS, and MOT were estimated for SNP windows of one SNP (SW1), ten SNP (SW10), 30 SNP (SW30), 50 SNP (SW50), and 100 SNP (SW100) using a single-step genomic best linear unbiased prediction approach. The fixed effects in the model were contemporary group, ejaculate order, bull age, ambient temperature, and heterosis. The random effects accounted for animal additive genetic effects, permanent environment effects, and residual. The SNPs explaining at least 0.001% of the additive genetic variance in SW1, 0.01% in SW10, 0.03% in SW30, 0.05% in SW50, and 0.1% in SW100 were selected for gene identification through the NCBI database. The pathway analysis utilized genes associated with the identified SNP windows. RESULTS Comparison of overlapping and non-overlapping SNP windows revealed notable differences among the identified pathways and genes associated with the studied traits. Overlapping windows consistently yielded a larger number of shared biological pathways and genes than non-overlapping windows. In particular, overlapping SW30 and SW50 identified the largest number of shared pathways and genes in the Thai multibreed dairy population. CONCLUSION This study yielded valuable insights into the genetic architecture of VOL, NS, and MOT. It also highlighted the importance of assessing overlapping and non-overlapping SNP windows of various sizes for their effectiveness to identify shared pathways and genes influencing multiple traits.
Collapse
Affiliation(s)
- Mattaneeya Sarakul
- Department of Animal Science, Nakhon Phanom University, Nakhon Phanom, 48000,
Thailand
| | - Mauricio A. Elzo
- Department of Animal Sciences, University of Florida, Gainesville, FL 32611-0910,
USA
| | | | | | - Danai Jattawa
- Department of Animal Science, Kasetsart University, Bangkok 10900,
Thailand
| | - Thawee Laodim
- Department of Animal Science, Kasetsart University, Kamphaeng Saen Campus, Nakhon Pathom 73140,
Thailand
| |
Collapse
|
2
|
Pharmacometabolomic study of drug response to antihypertensive medications for hypertension marker identification in Han Chinese individuals in Taiwan. Comput Struct Biotechnol J 2022; 20:6458-6466. [DOI: 10.1016/j.csbj.2022.11.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 11/13/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022] Open
|
3
|
Singh B, Kosuru R, Lakshmikanthan S, Sorci-Thomas M, Zhang D, Sparapani R, Vasquez-Vivar J, Chrzanowska M. Endothelial Rap1 (Ras-Association Proximate 1) Restricts Inflammatory Signaling to Protect From the Progression of Atherosclerosis. Arterioscler Thromb Vasc Biol 2021; 41:638-650. [PMID: 33267664 PMCID: PMC8105264 DOI: 10.1161/atvbaha.120.315401] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Small GTPase Rap1 (Ras-association proximate 1) is a novel, positive regulator of NO release and endothelial function with a potentially key role in mechanosensing of atheroprotective, laminar flow. Our objective was to delineate the role of Rap1 in the progression of atherosclerosis and its specific functions in the presence and absence of laminar flow, to better define its role in endothelial mechanisms contributing to plaque formation and atherogenesis. Approach and Results: In a mouse atherosclerosis model, endothelial Rap1B deletion exacerbates atherosclerotic plaque formation. In the thoracic aorta, where laminar shear stress-induced NO is otherwise atheroprotective, plaque area is increased in Athero-Rap1BiΔEC (atherogenic endothelial cell-specific, tamoxifen-inducible Rap1A+Rap1B knockout) mice. Endothelial Rap1 deficiency also leads to increased plaque size, leukocyte accumulation, and increased CAM (cell adhesion molecule) expression in atheroprone areas, whereas vascular permeability is unchanged. In endothelial cells, in the absence of protective laminar flow, Rap1 deficiency leads to an increased proinflammatory TNF-α (tumor necrosis factor alpha) signaling and increased NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) activation and elevated inflammatory receptor expression. Interestingly, this increased signaling to NF-κB activation is corrected by AKTVIII-an inhibitor of Akt (protein kinase B) translocation to the membrane. Together, these data implicate Rap1 in restricting Akt-dependent signaling, preventing excessive cytokine receptor signaling and proinflammatory NF-κB activation. CONCLUSIONS Via 2 distinct mechanisms, endothelial Rap1 protects from the atherosclerosis progression in the presence and absence of laminar flow; Rap1-stimulated NO release predominates in laminar flow, and restriction of proinflammatory signaling predominates in the absence of laminar flow. Our studies provide novel insights into the mechanisms underlying endothelial homeostasis and reveal the importance of Rap1 signaling in cardiovascular disease.
Collapse
Affiliation(s)
- Bandana Singh
- Blood Research Institute, Versiti, Milwaukee, Wisconsin
| | - Ramoji Kosuru
- Blood Research Institute, Versiti, Milwaukee, Wisconsin
| | | | - Mary Sorci-Thomas
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Division of Endocrinology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - David Zhang
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Rodney Sparapani
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jeannette Vasquez-Vivar
- Department of Biophysics and Redox Biology Program, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Magdalena Chrzanowska
- Blood Research Institute, Versiti, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
4
|
Abstract
PURPOSE OF REVIEW To review the current evidence on research related to age of hypertension onset-its definition, correlates, heritability, and association with adverse outcomes. We also propose a framework for implementing assessment of hypertension onset age into clinical practice. RECENT FINDINGS Prior studies have used both objective measurements and self-report to determine age of hypertension onset or early-onset hypertension. Yet, no criterion for standard definition currently exists for either. Data from epidemiological and clinical studies demonstrate that early-onset hypertension is a highly heritable trait that confers an increased risk for cardiovascular death and end-organ damage compared with late-onset hypertension. Literature to date suggests that (parental) age of hypertension onset can be feasibly assessed for estimating (1) risk of future hypertension in non-hypertensive persons; and (2) the propensity for cardiovascular disease in individuals with established hypertension.
Collapse
Affiliation(s)
- Karri Suvila
- Division of Medicine, Turku University Hospital, Turku, Finland.
- Department of Internal Medicine, University of Turku, Turku, Finland.
| | - Ville Langén
- Division of Medicine, Turku University Hospital, Turku, Finland
- Department of Geriatrics, University of Turku, Turku, Finland
| | - Susan Cheng
- The Framingham Heart Study, Framingham, MA, USA
- Barbra Streisand Women's Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Teemu J Niiranen
- Department of Internal Medicine, University of Turku, Turku, Finland
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Turku, Finland
| |
Collapse
|
5
|
Integration of Rap1 and Calcium Signaling. Int J Mol Sci 2020; 21:ijms21051616. [PMID: 32120817 PMCID: PMC7084553 DOI: 10.3390/ijms21051616] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 02/07/2023] Open
Abstract
Ca2+ is a universal intracellular signal. The modulation of cytoplasmic Ca2+ concentration regulates a plethora of cellular processes, such as: synaptic plasticity, neuronal survival, chemotaxis of immune cells, platelet aggregation, vasodilation, and cardiac excitation–contraction coupling. Rap1 GTPases are ubiquitously expressed binary switches that alternate between active and inactive states and are regulated by diverse families of guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). Active Rap1 couples extracellular stimulation with intracellular signaling through secondary messengers—cyclic adenosine monophosphate (cAMP), Ca2+, and diacylglycerol (DAG). Much evidence indicates that Rap1 signaling intersects with Ca2+ signaling pathways to control the important cellular functions of platelet activation or neuronal plasticity. Rap1 acts as an effector of Ca2+ signaling when activated by mechanisms involving Ca2+ and DAG-activated (CalDAG-) GEFs. Conversely, activated by other GEFs, such as cAMP-dependent GEF Epac, Rap1 controls cytoplasmic Ca2+ levels. It does so by regulating the activity of Ca2+ signaling proteins such as sarcoendoplasmic reticulum Ca2+-ATPase (SERCA). In this review, we focus on the physiological significance of the links between Rap1 and Ca2+ signaling and emphasize the molecular interactions that may offer new targets for the therapy of Alzheimer’s disease, hypertension, and atherosclerosis, among other diseases.
Collapse
|
6
|
Chiang KM, Chang HC, Yang HC, Chen CH, Chen HH, Lee WJ, Pan WH. Genome-wide association study of morbid obesity in Han Chinese. BMC Genet 2019; 20:97. [PMID: 31852448 PMCID: PMC6921553 DOI: 10.1186/s12863-019-0797-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 11/28/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND As obesity is becoming pandemic, morbid obesity (MO), an extreme type of obesity, is an emerging issue worldwide. It is imperative to understand the factors responsible for huge weight gain in certain populations in the modern society. Very few genome-wide association studies (GWAS) have been conducted on MO patients. This study is the first MO-GWAS study in the Han-Chinese population in Asia. METHODS We conducted a two-stage GWAS with 1110 MO bariatric patients (body mass index [BMI] ≥ 35 kg/m2) from Min-Sheng General Hospital, Taiwan. The first stage involved 575 patients, and 1729 sex- and age-matched controls from the Taiwan Han Chinese Cell and Genome Bank. In the second stage, another 535 patients from the same hospital were genotyped for 52 single nucleotide polymorphisms (SNPs) discovered in the first stage, and 9145 matched controls from Taiwan Biobank were matched for confirmation analysis. RESULTS The results of the joint analysis for the second stage revealed six top ranking SNPs, including rs8050136 (p-value = 7.80 × 10- 10), rs9939609 (p-value = 1.32 × 10- 9), rs1421085 (p-value = 1.54 × 10- 8), rs9941349 (p-value = 9.05 × 10- 8), rs1121980 (p-value = 7.27 × 10- 7), and rs9937354 (p-value = 6.65 × 10- 7), which were all located in FTO gene. Significant associations were also observed between MO and RBFOX1, RP11-638 L3.1, TMTC1, CBLN4, CSMD3, and ERBB4, respectively, using the Bonferroni correction criteria for 52 SNPs (p < 9.6 × 10- 4). CONCLUSION The most significantly associated locus of MO in the Han-Chinese population was the well-known FTO gene. These SNPs located in intron 1, may include the leptin receptor modulator. Other significant loci, showing weak associations with MO, also suggested the potential mechanism underlying the disorders with eating behaviors or brain/neural development.
Collapse
Affiliation(s)
- Kuang-Mao Chiang
- Institute of Biomedical Sciences, Academia Sinica, Taipei City, Taiwan
| | - Heng-Cheng Chang
- Department of Gynecology and Obstetrics, School of Medicine, Taipei Medical University, Taipei City, Taiwan
| | - Hsin-Chou Yang
- Institute of Statistical Science, Academia Sinica, Taipei City, Taiwan
| | - Chien-Hsiun Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei City, Taiwan
| | - Hsin-Hung Chen
- Department of Nutrition and Health Science, Chang Jung Christian University, Tainan City, Taiwan
| | - Wei-Jei Lee
- Department of Surgery, Min-Sheng General Hospital, Taoyuan City, Taiwan
| | - Wen-Harn Pan
- Institute of Biomedical Sciences, Academia Sinica, Taipei City, Taiwan
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli County, Taiwan
| |
Collapse
|
7
|
Kolifarhood G, Daneshpour MS, Khayat BS, Saadati HM, Guity K, Khosravi N, Akbarzadeh M, Sabour S. Generality of genomic findings on blood pressure traits and its usefulness in precision medicine in diverse populations: A systematic review. Clin Genet 2019; 96:17-27. [PMID: 30820929 DOI: 10.1111/cge.13527] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/14/2019] [Accepted: 02/21/2019] [Indexed: 01/01/2023]
Abstract
Remarkable findings from genome-wide association studies (GWAS) on blood pressure (BP) traits have made new insights for developing precision medicine toward more effective screening measures. However, generality of GWAS findings in diverse populations is hampered by some technical limitations. There is no comprehensive study to evaluate source(s) of the non-generality of GWAS results on BP traits, so to fill the gap, this systematic review study was carried out. Using MeSH terms, 1545 records were detected through searching in five databases and 49 relevant full-text articles were included in our review. Overall, 749 unique variants were reported, of those, majority of variants have been detected in Europeans and were associated to systolic and diastolic BP traits. Frequency of genetic variants with same position was low in European and non-European populations (n = 38). However, more than 200 (>25%) single nucleotide polymorphisms were found on same loci or linkage disequilibrium blocks (r2 ≥ 80%). Investigating for locus position and linkage disequilibrium of infrequent unique variants showed modest to high reproducibility of findings in Europeans that in some extent was generalizable in other populations. Beyond theoretical limitations, our study addressed other possible sources of non-generality of GWAS findings for BP traits in the same and different origins.
Collapse
Affiliation(s)
- Goodarz Kolifarhood
- Department of Epidemiology, School of Public Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam S Daneshpour
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahareh S Khayat
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein M Saadati
- Department of Epidemiology, School of Public Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamran Guity
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasim Khosravi
- Department of Community Health Nursing, School of Nursing and Midwifery, Iran University of Medical Sciences, Tehran, Iran
| | - Mahdi Akbarzadeh
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Siamak Sabour
- Department of Epidemiology, School of Public Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Safety Promotion and Injury Prevention Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Guerrero-Preston R, Lawson F, Rodriguez-Torres S, Noordhuis MG, Pirini F, Manuel L, Valle BL, Hadar T, Rivera B, Folawiyo O, Baez A, Marchionni L, Koch WM, Westra WH, Kim YJ, Eshleman JR, Sidransky D. JAK3 Variant, Immune Signatures, DNA Methylation, and Social Determinants Linked to Survival Racial Disparities in Head and Neck Cancer Patients. Cancer Prev Res (Phila) 2019; 12:255-270. [PMID: 30777857 DOI: 10.1158/1940-6207.capr-17-0356] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 10/30/2018] [Accepted: 02/11/2019] [Indexed: 02/06/2023]
Abstract
To inform novel personalized medicine approaches for race and socioeconomic disparities in head and neck cancer, we examined germline and somatic mutations, immune signatures, and epigenetic alterations linked to neighborhood determinants of health in Black and non-Latino White (NLW) patients with head and neck cancer. Cox proportional hazards revealed that Black patients with squamous cell carcinoma of head and neck (HNSCC) with PAX5 (P = 0.06) and PAX1 (P = 0.017) promoter methylation had worse survival than NLW patients, after controlling for education, zipcode, and tumor-node-metastasis stage (n = 118). We also found that promoter methylation of PAX1 and PAX5 (n = 78), was correlated with neighborhood characteristics at the zip-code level (P < 0.05). Analyses also showed differences in the frequency of TP53 mutations (n = 32) and tumor-infiltrating lymphocyte (TIL) counts (n = 24), and the presence of a specific C → A germline mutation in JAK3, chr19:17954215 (protein P132T), in Black patients with HNSCC (n = 73; P < 0.05), when compared with NLW (n = 37) patients. TIL counts are associated (P = 0.035) with long-term (>5 years), when compared with short-term survival (<2 years). We show bio-social determinants of health associated with survival in Black patients with HNSCC, which together with racial differences shown in germline mutations, somatic mutations, and TIL counts, suggests that contextual factors may significantly inform precision oncology services for diverse populations.
Collapse
Affiliation(s)
- Rafael Guerrero-Preston
- Department of Otolaryngology and Head and Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, Maryland. .,Department of Obstetrics and Gynecology, University of Puerto Rico, School of Medicine, San Juan, Puerto Rico
| | - Fahcina Lawson
- Department of Otolaryngology and Head and Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Sebastian Rodriguez-Torres
- Department of Otolaryngology and Head and Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, Maryland.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Maartje G Noordhuis
- Department of Otolaryngology and Head and Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, Maryland.,Department of Otorhinolaryngology/Head and Neck Surgery, University of Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Francesca Pirini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Laura Manuel
- Department of Epidemiology and Biostatistics, The University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Blanca L Valle
- Department of Otolaryngology and Head and Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Tal Hadar
- Breast Health Unit, Department of General Surgery, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Bianca Rivera
- Department of Otolaryngology, University of Puerto Rico, School of Medicine, San Juan, Puerto Rico
| | - Oluwasina Folawiyo
- Department of Otolaryngology and Head and Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Adriana Baez
- Department of Otolaryngology, University of Puerto Rico, School of Medicine, San Juan, Puerto Rico
| | - Luigi Marchionni
- Department of Oncology, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Wayne M Koch
- Department of Otolaryngology and Head and Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - William H Westra
- Department of Pathology, The Icahn School of Medicine at Mount Sinai Hospital, New York, New York
| | - Young J Kim
- Department of Otolaryngology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - James R Eshleman
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - David Sidransky
- Department of Otolaryngology and Head and Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| |
Collapse
|
9
|
A Two-Stage Whole-Genome Gene Expression Association Study of Young-Onset Hypertension in Han Chinese Population of Taiwan. Sci Rep 2018; 8:1800. [PMID: 29379041 PMCID: PMC5789005 DOI: 10.1038/s41598-018-19520-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 01/03/2018] [Indexed: 12/31/2022] Open
Abstract
Hypertension is an important public health problem in the world. Since the intermediate position of the gene expression between genotype and phenotype makes it suitable to link genotype to phenotype, we carried out a two-stage whole-genome gene expression association study to find differentially expressed genes and pathways for hypertension. In the first stage, 126 cases and 149 controls were used to find out the differentially expressed genes. In the second stage, an independent set of samples (127 cases and 150 controls) was used to validate the results. Additionally, we conducted a gene set enrichment analysis (GSEA) to search for differentially affected pathways. A total of nine genes were implicated in the first stage (Bonferroni-corrected p-value < 0.05). Among these genes, ZRANB1, FAM110A, PREP, ANKRD9 and LAMB2 were also differentially expressed in an existing database of hypertensive mouse model (GSE19817). A total of 16 pathways were identified by the GSEA. ZRANB1 and six pathways identified are related to TNF-α. Three pathways are related to interleukin, one to metabolic syndrome, and one to Hedgehog signaling. Identification of these genes and pathways suggest the importance of 1. inflammation, 2. visceral fat metabolism, and 3. adipocytes and osteocytes homeostasis in hypertension mechanisms and complications.
Collapse
|
10
|
The role of a FADS1 polymorphism in the association of fatty acid blood levels, BMI and blood pressure in young children-Analyses based on path models. PLoS One 2017; 12:e0181485. [PMID: 28732058 PMCID: PMC5521833 DOI: 10.1371/journal.pone.0181485] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 06/30/2017] [Indexed: 01/26/2023] Open
Abstract
Background The recent obesity epidemic in children also showed an increase in the prevalence of hypertension. As blood pressure (BP) is associated with (long-chain) polyunsaturated fatty acids (LC PUFA), genetic variation in desaturase enzymes being involved in the synthesis of LC PUFA may be associated with BP. This study aimed to investigate the direct effects (independent of mediating variables) and indirect effects (mediated through intermediate variables) of a common variant in the FADS1 gene, rs174546, known to affect delta-5 desaturase (D5D) activity on PUFA level, body mass index (BMI) and BP. Methods A subsample of the IDEFICS (Identification and prevention of dietary- and lifestyle-induced health effects in children and infants) baseline survey including 520 children aged 2 to <10 years from six European countries was included. The association between rs174546 (T<C) and BP z-score as well as the mediating effects of selected key PUFA levels (dihomo-gamma-linolenic acid, DGLA; arachidonic acid, ARA; eicosapentaenoic acid, EPA) or estimated D5D activity (D5D index) and BMI z-score were investigated through path model analyses, adjusting for sex, age, educational level of parents, family history of hypertension, lifestyle factors and blood levels of saturated and monounsaturated fatty acids, triglycerides and low density lipoprotein cholesterol. Whole blood fatty acids were measured by a validated gas chromatographic method and recorded as percentage of weight of all fatty acids detected. Results Minor allele carriers of the SNP rs174546 had significantly higher DGLA and lower ARA and EPA levels as well as a lower D5D index. Via ARA and BMI z-score, the polymorphism had an indirect lowering effect on systolic BP z-score for each additional T allele (standardized effect estimate -0.057, p = 0.007). For DGLA, EPA and D5D index, the indirect effects of rs174546 on systolic BP were also negative but did not reach significance. DGLA and EPA had an increasing indirect effect on systolic BP via BMI. Results for diastolic BP were in general similar but effect estimates were lower compared to systolic BP. Conclusion Genetic variation in FADS1 influences BP via ARA and BMI indicating a favorable effect of the minor allele in SNP rs174546. Thus, polymorphisms with an impact on the D5D activity may play a role for the BP level mediated through PUFA and BMI. Therefore, health effects of dietary n-6 and n-3 PUFA may vary depending on genetic FADS1 variants.
Collapse
|
11
|
Abstract
Hypertension tends to perpetuate in families and the heritability of hypertension is estimated to be around 20-60%. So far, the main proportion of this heritability has not been found by single-locus genome-wide association studies. Therefore, the current study explored gene-gene interactions that have the potential to partially fill in the missing heritability. A two-stage discovery-confirmatory analysis was carried out in the Framingham Heart Study cohorts. The first stage was an exhaustive pairwise search performed in 2320 early-onset hypertensive cases with matched normotensive controls from the offspring cohort. Then, identified gene-gene interactions were assessed in an independent set of 694 subjects from the original cohort. Four unique gene-gene interactions were found to be related to hypertension. Three detected genes were recognized by previous studies, and the other 5 loci/genes (MAN1A1, LMO3, NPAP1/SNRPN, DNAL4, and RNA5SP455/KRT8P5) were novel findings, which had no strong main effect on hypertension and could not be easily identified by single-locus genome-wide studies. Also, by including the identified gene-gene interactions, more variance was explained in hypertension. Overall, our study provides evidence that the genome-wide gene-gene interaction analysis has the possibility to identify new susceptibility genes, which can provide more insights into the genetic background of blood pressure regulation.
Collapse
|
12
|
Fowdar JY, Grealy R, Lu Y, Griffiths LR. A genome-wide association study of essential hypertension in an Australian population using a DNA pooling approach. Mol Genet Genomics 2016; 292:307-324. [PMID: 27866268 DOI: 10.1007/s00438-016-1274-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 11/10/2016] [Indexed: 01/11/2023]
Abstract
Despite the success of genome-wide association studies (GWAS) in detecting genetic loci involved in complex traits, few susceptibility genes have been detected for essential hypertension (EH). We aimed to use pooled DNA GWAS approach to identify and validate novel genomic loci underlying EH susceptibility in an Australian case-control population. Blood samples and questionnaires detailing medical history, blood pressure, and prescribed medications were collected for 409 hypertensives and 409 age-, sex- and ethnicity-matched normotensive controls. Case and control DNA were pooled in quadruplicate and hybridized to Illumina 1 M-Duo arrays. Allele frequencies agreed with those reported in reference data and known EH association signals were represented in the top-ranked SNPs more frequently than expected by chance. Validation showed that pooled DNA GWAS gave reliable estimates of case and control allele frequencies. Although no markers reached Bonferroni-corrected genome-wide significance levels (5.0 × 10-8), the top marker rs34870220 near ASGR1 approached significance (p = 4.32 × 10-7), as did several candidate loci (p < 1 × 10-6) on chromosomes 2, 4, 6, 9, 12, and 17. Four markers (located in or near genes NHSL1, NKFB1, GLI2, and LRRC10) from the top ten ranked SNPs were individually genotyped in pool samples and were tested for association between cases and controls using the χ 2 test. Of these, rs1599961 (NFKB1) and rs12711538 (GLI2) showed significant difference between cases and controls (p < 0.01). Additionally, four top-ranking markers within NFKB1 were found to be in LD, suggesting a single strong association signal for this gene.
Collapse
Affiliation(s)
- Javed Y Fowdar
- School of Medical Science, Griffith University, Gold Coast, Australia
| | - Rebecca Grealy
- School of Medical Science, Griffith University, Gold Coast, Australia
| | - Yi Lu
- Genetic Epidemiology Department, Queensland Institute of Medical Research, Brisbane, Australia
| | - Lyn R Griffiths
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, 60 Musk Ave, Kelvin Grove, Brisbane, QLD, 4059, Australia.
| |
Collapse
|
13
|
Igarashi R, Fujihara K, Heianza Y, Ishizawa M, Kodama S, Saito K, Hara S, Hanyu O, Honda R, Tsuji H, Arase Y, Sone H. Impact of individual components and their combinations within a family history of hypertension on the incidence of hypertension: Toranomon hospital health management center study 22. Medicine (Baltimore) 2016; 95:e4564. [PMID: 27661014 PMCID: PMC5044884 DOI: 10.1097/md.0000000000004564] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Although a family history (FH) of hypertension is a risk factor for the development of hypertension, only a few studies have investigated in detail the impact of individual components of an FH on incident hypertension. We investigated the impact of individual components and their combinations on the presence or development of hypertension considering obesity, smoking habits, physical activity, and other metabolic parameters.Studied were 12,222 Japanese individuals without hypertension (n = 9,766) and with hypertension (n = 2,456) at the baseline examination. The presence or incidence of hypertension during 5 years after a baseline examination was assessed by the presence of systolic blood pressure ≥140 mmHg and/or diastolic blood pressure ≥90 mmHg or a self-reported history of clinician-diagnosed hypertension. In this prospective study, the odds ratio for incident hypertension was 1.39 (95% confidence interval [CI], 1.22, 1.59) for individuals with any FH of hypertension compared with those without such an FH. Individuals with an FH of hypertension in both parents and one or more grandparents had an odds ratio of 3.05 (95% CI 1.74, 5.36) for hypertension compared with those without an FH of hypertension. FH was associated with incident hypertension independently of other modifiable risk factors such as obesity, smoking, physical inactivity, hyperglycemia, hyperuricemia, and hypertriglyceridemia.A parental history of hypertension was an essential component within an FH for incident hypertension. FH of hypertension over two generations with both parents affected was the most important risk factor for incident hypertension. Although an FH is not a modifiable risk factor, modifying other risk factors could contribute to reducing the risk of hypertension even among individuals with a family history of hypertension.
Collapse
Affiliation(s)
- Risa Igarashi
- Department of Internal Medicine, Niigata University Faculty of Medicine, Niigata
| | - Kazuya Fujihara
- Department of Internal Medicine, Niigata University Faculty of Medicine, Niigata
| | - Yoriko Heianza
- Department of Internal Medicine, Niigata University Faculty of Medicine, Niigata
| | - Masahiro Ishizawa
- Department of Internal Medicine, Niigata University Faculty of Medicine, Niigata
| | - Satoru Kodama
- Department of Internal Medicine, Niigata University Faculty of Medicine, Niigata
- Health Management Center, Toranomon Hospital
| | - Kazumi Saito
- Department of Internal Medicine, Niigata University Faculty of Medicine, Niigata
- Health Management Center, Toranomon Hospital
| | - Shigeko Hara
- Health Management Center, Toranomon Hospital
- Okinaka Memorial Institute for Medical Research, Tokyo, Japan
| | - Osamu Hanyu
- Department of Internal Medicine, Niigata University Faculty of Medicine, Niigata
| | | | - Hiroshi Tsuji
- Health Management Center, Toranomon Hospital
- Okinaka Memorial Institute for Medical Research, Tokyo, Japan
| | - Yasuji Arase
- Health Management Center, Toranomon Hospital
- Okinaka Memorial Institute for Medical Research, Tokyo, Japan
| | - Hirohito Sone
- Department of Internal Medicine, Niigata University Faculty of Medicine, Niigata
- Health Management Center, Toranomon Hospital
- Correspondence: Hirohito Sone, Health Management Center, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo 105-8470, Japan (e-mail: )
| |
Collapse
|
14
|
Liang YJ, Lin YT, Chen CW, Lin CW, Chao KM, Pan WH, Yang HC. SMART: Statistical Metabolomics Analysis—An R Tool. Anal Chem 2016; 88:6334-41. [DOI: 10.1021/acs.analchem.6b00603] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Yu-Jen Liang
- Graduate
Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan
- Institute
of Statistical Science, Academia Sinica, Taipei 115, Taiwan
| | - Yu-Ting Lin
- Institute
of Statistical Science, Academia Sinica, Taipei 115, Taiwan
| | - Chia-Wei Chen
- Institute
of Statistical Science, Academia Sinica, Taipei 115, Taiwan
| | - Chien-Wei Lin
- Institute
of Statistical Science, Academia Sinica, Taipei 115, Taiwan
| | - Kun-Mao Chao
- Graduate
Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan
- Department
of Computer Science and Information Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Wen-Harn Pan
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Hsin-Chou Yang
- Institute
of Statistical Science, Academia Sinica, Taipei 115, Taiwan
- Institute
of Public Health, National Yang Ming University, Taipei 11221, Taiwan
- Department
of Statistics, National Cheng Kung University, Tainan 701, Taiwan
- Institute
of Statistics, National Tsing Hua University, Hsinchu 30013, Taiwan
- School
of Public Health, National Defense Medical Center, Taipei 114, Taiwan
| |
Collapse
|
15
|
Han Y, Li L, Zhang Y, Yuan H, Ye L, Zhao J, Duan DD. Phenomics of Vascular Disease: The Systematic Approach to the Combination Therapy. Curr Vasc Pharmacol 2016; 13:433-40. [PMID: 25313004 PMCID: PMC4397150 DOI: 10.2174/1570161112666141014144829] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 02/15/2014] [Accepted: 05/21/2014] [Indexed: 12/28/2022]
Abstract
Vascular diseases are usually caused by multifactorial pathogeneses involving genetic and environmental factors. Our current understanding of vascular disease is, however, based on the focused genotype/phenotype studies driven by the “one-gene/one-phenotype” hypothesis. Drugs with “pure target” at individual molecules involved in the pathophysiological pathways are the mainstream of current clinical treatments and the basis of combination therapy of vascular diseases. Recently, the combination of genomics, proteomics, and metabolomics has unraveled the etiology and pathophysiology of vascular disease in a big-data fashion and also revealed unmatched relationships between the omic variability and the much narrower definition of various clinical phenotypes of vascular disease in individual patients. Here, we introduce the phenomics strategy that will change the conventional focused phenotype/genotype/genome study to a new systematic phenome/genome/proteome approach to the understanding of pathophysiology and combination therapy of vascular disease. A phenome is the sum total of an organism’s phenotypic traits that signify the expression of genome and specific environmental influence. Phenomics is the study of phenome to quantitatively correlate complex traits to variability not only in genome, but also in transcriptome, proteome, metabolome, interactome, and environmental factors by exploring the systems biology that links the genomic and phenomic spaces. The application of phenomics and the phenome-wide associated study (PheWAS) will not only identify a systemically-integrated set of biomarkers for diagnosis and prognosis of vascular disease but also provide novel treatment targets for combination therapy and thus make a revolutionary paradigm shift in the clinical treatment of these devastating diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dayue Darrel Duan
- Laboratory of Cardiovascular Phenomics, Department of Pharmacology, University of Nevada School of Medicine, Center for Molecular Medicine 303F, 1664 N Virginia Street/MS 318, Reno, Nevada 89557-0318, USA.
| |
Collapse
|
16
|
Recent Advances in the Genetics of Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 956:561-581. [PMID: 27957710 DOI: 10.1007/5584_2016_75] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Hypertension is a silent killer worldwide, caused by both genetic and environmental factors. Until now, genetic and genomic association studies of hypertension are reporting different degree of association on hypertension. Hence, it is essential to gather all the available information on the reported genetic loci and to determine if any biomarker(s) is/are significantly associated with hypertension. Current review concluded the potential biomarkers for hypertension, with regards to electrolyte and fluid transports, as well as sodium/potassium ions homeostasis, which are supported by the results of case-controls and meta-analyses.
Collapse
|
17
|
Xu K, Ma L, Li Y, Wang F, Zheng GY, Sun Z, Jiang F, Chen Y, Liu H, Dang A, Chen X, Chun J, Tian XL. Genetic and Functional Evidence Supports LPAR1 as a Susceptibility Gene for Hypertension. Hypertension 2015; 66:641-6. [PMID: 26123684 DOI: 10.1161/hypertensionaha.115.05515] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 06/08/2015] [Indexed: 01/11/2023]
Abstract
Essential hypertension is a complex disease affected by genetic and environmental factors and serves as a major risk factor for cardiovascular diseases. Serum lysophosphatidic acid correlates with an elevated blood pressure in rats, and lysophosphatidic acid interacts with 6 subtypes of receptors. In this study, we assessed the genetic association of lysophosphatidic acid receptors with essential hypertension by genotyping 28 single-nucleotide polymorphisms from genes encoding for lysophosphatidic acid receptors, LPAR1, LPAR2, LPAR3, LPAR4, LPAR5, and LPAR6 and their flanking sequences, in 3 Han Chinese cohorts consisting of 2630 patients and 3171 controls in total. We identified a single-nucleotide polymorphism, rs531003 in the 3'-flanking genomic region of LPAR1, associated with hypertension (the Bonferroni corrected P=1.09×10(-5), odds ratio [95% confidence interval]=1.23 [1.13-1.33]). The risk allele C of rs531003 is associated with the increased expression of LPAR1 and the susceptibility of hypertension, particularly in those with a shortage of sleep (P=4.73×10(-5), odds ratio [95% confidence interval]=1.75 [1.34-2.28]). We further demonstrated that blood pressure elevation caused by sleep deprivation and phenylephrine-induced vasoconstriction was both diminished in LPAR1-deficient mice. Together, we show that LPAR1 is a novel susceptibility gene for human essential hypertension and that stress, such as shortage of sleep, increases the susceptibility of patients with risk allele to essential hypertension.
Collapse
Affiliation(s)
- Ke Xu
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Lu Ma
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Yang Li
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Fang Wang
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Gu-Yan Zheng
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Zhijun Sun
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Feng Jiang
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Yundai Chen
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Huirong Liu
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Aimin Dang
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Xi Chen
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Jerold Chun
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.)
| | - Xiao-Li Tian
- From the Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, Beijing, PR China (K.X., Y.L., G.Y.Z., X.L.T.); Department of Physiology and Pathophysiology, School of Basic Medical Sciences (L.M., H.L.) and Department of Cardiology, Beijing Chaoyang Hospital (F.J.), Capital Medical University, Beijing, PR China; State Key Laboratory of Cardiovascular Diseases (F.W., X.C.) and Department of Cardiology (A.D.), Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, PR China; Cardiovascular Department, PLA General Hospital, Beijing, PR China (Z.S., Y.C.); and Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA (J.C.).
| |
Collapse
|
18
|
Lim NK, Lee JY, Lee JY, Park HY, Cho MC. The Role of Genetic Risk Score in Predicting the Risk of Hypertension in the Korean population: Korean Genome and Epidemiology Study. PLoS One 2015; 10:e0131603. [PMID: 26110887 PMCID: PMC4482533 DOI: 10.1371/journal.pone.0131603] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 06/03/2015] [Indexed: 01/11/2023] Open
Abstract
Hypertension is regarded as a multifactorial disease with a modest contribution of genetic factors and strongly affected by environmental factors. Recent genome-wide association studies have identified specific loci associated with high blood pressure (BP) and hypertension. This study aimed to examine the association between the genetic risk score (GRS), a linear function of multiple single nucleotide polymorphisms (SNPs) associated with hypertension, and high BP and prevalent hypertension at baseline examination and to evaluate the efficacy of the GRS for predicting incident hypertension with longitudinal data in Korean subjects. Data for 8,556 participants, aged 40 to 69, in a community-based cohort study were analyzed. Unweighted GRS (cGRS) and weighted GRS (wGRS) were constructed from 4 SNPs related to high BP or hypertension in previous genome-wide association and its replication studies for the Korean middle-aged population. Cross-sectional analysis (n=8,556) revealed that cGRS was significantly associated with prevalent hypertension (odds ratio=1.15 per risk allele; 95%CI, 1.09-1.20). Additionally, the odds ratios (ORs) of prevalent hypertension for those who in medium and the highest tertile compared with those who in the lowest tertile of wGRS were 1.31 (95% CI, 1.15-1.50) and 1.59 (95%CI, 1.38-1.82), respectively. In a longitudinal analysis (n=5,632), participants in the highest tertile of wGRS had a 1.22-fold (OR=1.22, 95%CI, 1.02‒1.46) greater risk of incident hypertension relative to those in the lowest tertile, after adjusting for a number of confounding factors. However, wGRS topped with traditional risk factors had no significant effect on discrimination ability (c-statistics with and without wGRS were 0.811 and 0.810, P=0.1057). But, reclassification analysis showed that the addition of GRS to the model with conventional risk factors led to about 9% significant increment in category-free net reclassification improvement. GRSs based on 4 SNPs were independently associated with hypertension and may provide a statistically significant improvement over the existing model for prediction of incident hypertension.
Collapse
Affiliation(s)
- Nam-Kyoo Lim
- Division of Cardiovascular and Rare Diseases, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Ji-Young Lee
- Division of Structural and Functional Genomics, Korea National Institute of Health, Cheongju, Republic of Korea
- Cardiovascular Research Institute and Cardiovascular Genome Center, Yonsei University Health System, Seoul, Republic of Korea
| | - Jong-Young Lee
- Division of Structural and Functional Genomics, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Hyun-Young Park
- Division of Cardiovascular and Rare Diseases, Korea National Institute of Health, Cheongju, Republic of Korea
- * E-mail:
| | - Myeong-Chan Cho
- Department of Cardiology, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
19
|
Gu D, Gomez-Redondo R, Dupre ME. Studying Disability Trends in Aging Populations. J Cross Cult Gerontol 2014; 30:21-49. [DOI: 10.1007/s10823-014-9245-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
20
|
Chen J, Zhao X, Wang H, Chen Y, Wang W, Zhou W, Wang X, Tang J, Zhao Y, Lu X, Chen S, Wang L, Shen C, Yang S. Common variants in TGFBR2 and miR-518 genes are associated with hypertension in the Chinese population. Am J Hypertens 2014; 27:1268-76. [PMID: 24687999 DOI: 10.1093/ajh/hpu047] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND An animal study reported that TGF-β1 maturation was linked to the homeostasis of blood pressure and elastogenesis of essential hypertension (EH). Recent advances require further research of TGF-β1 receptor in EH. METHODS A case-control study comprised of 2,012 adult hypertension case patients and 2,210 adult control subjects was conducted, and the association with blood pressure was further tested in children. Logistic regression and calculated genetic risk score were used to evaluate the effects of one single nucleotide polymorphism (SNP) and multiple SNPs on EH, respectively. RESULTS The genetic risk score of 10 SNPs showed a significant association with hypertension; the odds ratio of the upper quartile vs. the lower quartile was 1.282 (P = 4.67 × 10(-3)). rs7256241 in miR-518 was significantly associated with diastolic blood pressure (DBP) change in control subjects (P = 0.002), and this association was also observed in children (P = 0.04). The systolic blood pressure (SBP) and DBP of female patients taking reserpine were higher with the C and G alleles of rs3773661 (P = 0.004) and rs7256241 (P = 0.002), respectively. In patients taking Zhen Ju Jiang Ya tablets, SBP and DBP decreased linearly with rs749794 (P = 0.004 and P = 0.048, respectively). SBP decreased linearly with rs1155705 (P = 0.007) and rs11709624 (P = 0.04), but increased with rs1036096 (P = 0.03) in male patients. In male patients taking Jiang Ya tablets, SBP increased linearly with rs11709624 (P = 0.007), DBP increased linearly with rs1155705 (P = 0.03) whereas decreased with rs7256241 (P = 0.04). CONCLUSIONS Our results suggest that TGFBR2 and miR-518 harbor variants that increase the risk of EH and affect blood pressure homeostasis as well as efficacy of antihypertensive agents.
Collapse
Affiliation(s)
- Jinfeng Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China; Jiangsu Province Institute of Geriatrics, Nanjing, China
| | - Xianghai Zhao
- Department of Cardiology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, China
| | - Hairu Wang
- Nanjing Institute of Industry Technology, Nanjing, China
| | - Yanchun Chen
- Department of Cardiology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, China
| | - Wen Wang
- Department of Cardiology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, China
| | - Wei Zhou
- Department of Cardiology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, China
| | - Xuecai Wang
- Department of Clinical Laboratory, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, China
| | - Junming Tang
- Department of Clinical Laboratory, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, China
| | - Yanping Zhao
- Department of Neurology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, China
| | - Xiangfeng Lu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shufeng Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Laiyuan Wang
- National Human Genome Center at Beijing, Beijing, China
| | - Chong Shen
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China;
| | - Song Yang
- Department of Cardiology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, China
| |
Collapse
|
21
|
Quilter CR, Cooper WN, Cliffe KM, Skinner BM, Prentice PM, Nelson L, Bauer J, Ong KK, Constância M, Lowe WL, Affara NA, Dunger DB. Impact on offspring methylation patterns of maternal gestational diabetes mellitus and intrauterine growth restraint suggest common genes and pathways linked to subsequent type 2 diabetes risk. FASEB J 2014; 28:4868-79. [PMID: 25145626 DOI: 10.1096/fj.14-255240] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Size at birth, postnatal weight gain, and adult risk for type 2 diabetes may reflect environmental exposures during developmental plasticity and may be mediated by epigenetics. Both low birth weight (BW), as a marker of fetal growth restraint, and high birth weight (BW), especially after gestational diabetes mellitus (GDM), have been linked to increased risk of adult type 2 diabetes. We assessed DNA methylation patterns using a bead chip in cord blood samples from infants of mothers with GDM (group 1) and infants with prenatal growth restraint indicated by rapid postnatal catch-up growth (group 2), compared with infants with normal postnatal growth (group 3). Seventy-five CpG loci were differentially methylated in groups 1 and 2 compared with the controls (group 3), representing 72 genes, many relevant to growth and diabetes. In replication studies using similar methodology, many of these differentially methylated regions were associated with levels of maternal glucose exposure below that defined by GDM [the Hyperglycemia and Adverse Pregnancy Outcome (HAPO) study] or were identified as changes observed after randomized periconceptional nutritional supplementation in a Gambian cohort characterized by maternal deprivation. These studies provide support for the concept that similar epigenetic modifications may underpin different prenatal exposures and potentially increase long-term risk for diseases such as type 2 diabetes.
Collapse
Affiliation(s)
| | - Wendy N Cooper
- Metabolic Research Laboratories, Medical Research Council (MRC) Metabolic Diseases Unit, Department of Obstetrics and Gynaecology, National Institute for Health Research Cambridge Biomedical Research Centre, Centre for Trophoblast Research, and
| | - Kerry M Cliffe
- Mammalian Molecular Genetics Group, Department of Pathology
| | | | - Philippa M Prentice
- National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Department of Paediatrics, University of Cambridge, Cambridge, UK; and
| | - LaTasha Nelson
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine Chicago, Chicago, Illinois, USA
| | - Julien Bauer
- Mammalian Molecular Genetics Group, Department of Pathology
| | - Ken K Ong
- National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Department of Paediatrics, University of Cambridge, Cambridge, UK; and
| | - Miguel Constância
- Metabolic Research Laboratories, Medical Research Council (MRC) Metabolic Diseases Unit, Department of Obstetrics and Gynaecology, National Institute for Health Research Cambridge Biomedical Research Centre, Centre for Trophoblast Research, and
| | - William L Lowe
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine Chicago, Chicago, Illinois, USA
| | | | - David B Dunger
- National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Department of Paediatrics, University of Cambridge, Cambridge, UK; and
| |
Collapse
|
22
|
Simino J, Shi G, Bis JC, Chasman DI, Ehret GB, Gu X, Guo X, Hwang SJ, Sijbrands E, Smith AV, Verwoert GC, Bragg-Gresham JL, Cadby G, Chen P, Cheng CY, Corre T, de Boer RA, Goel A, Johnson T, Khor CC, Lluís-Ganella C, Luan J, Lyytikäinen LP, Nolte IM, Sim X, Sõber S, van der Most PJ, Verweij N, Zhao JH, Amin N, Boerwinkle E, Bouchard C, Dehghan A, Eiriksdottir G, Elosua R, Franco OH, Gieger C, Harris TB, Hercberg S, Hofman A, James AL, Johnson AD, Kähönen M, Khaw KT, Kutalik Z, Larson MG, Launer LJ, Li G, Liu J, Liu K, Morrison AC, Navis G, Ong RTH, Papanicolau GJ, Penninx BW, Psaty BM, Raffel LJ, Raitakari OT, Rice K, Rivadeneira F, Rose LM, Sanna S, Scott RA, Siscovick DS, Stolk RP, Uitterlinden AG, Vaidya D, van der Klauw MM, Vasan RS, Vithana EN, Völker U, Völzke H, Watkins H, Young TL, Aung T, Bochud M, Farrall M, Hartman CA, Laan M, Lakatta EG, Lehtimäki T, Loos RJF, Lucas G, Meneton P, Palmer LJ, Rettig R, Snieder H, Tai ES, Teo YY, van der Harst P, Wareham NJ, Wijmenga C, Wong TY, Fornage M, Gudnason V, Levy D, Palmas W, Ridker PM, Rotter JI, van Duijn CM, et alSimino J, Shi G, Bis JC, Chasman DI, Ehret GB, Gu X, Guo X, Hwang SJ, Sijbrands E, Smith AV, Verwoert GC, Bragg-Gresham JL, Cadby G, Chen P, Cheng CY, Corre T, de Boer RA, Goel A, Johnson T, Khor CC, Lluís-Ganella C, Luan J, Lyytikäinen LP, Nolte IM, Sim X, Sõber S, van der Most PJ, Verweij N, Zhao JH, Amin N, Boerwinkle E, Bouchard C, Dehghan A, Eiriksdottir G, Elosua R, Franco OH, Gieger C, Harris TB, Hercberg S, Hofman A, James AL, Johnson AD, Kähönen M, Khaw KT, Kutalik Z, Larson MG, Launer LJ, Li G, Liu J, Liu K, Morrison AC, Navis G, Ong RTH, Papanicolau GJ, Penninx BW, Psaty BM, Raffel LJ, Raitakari OT, Rice K, Rivadeneira F, Rose LM, Sanna S, Scott RA, Siscovick DS, Stolk RP, Uitterlinden AG, Vaidya D, van der Klauw MM, Vasan RS, Vithana EN, Völker U, Völzke H, Watkins H, Young TL, Aung T, Bochud M, Farrall M, Hartman CA, Laan M, Lakatta EG, Lehtimäki T, Loos RJF, Lucas G, Meneton P, Palmer LJ, Rettig R, Snieder H, Tai ES, Teo YY, van der Harst P, Wareham NJ, Wijmenga C, Wong TY, Fornage M, Gudnason V, Levy D, Palmas W, Ridker PM, Rotter JI, van Duijn CM, Witteman JCM, Chakravarti A, Rao DC. Gene-age interactions in blood pressure regulation: a large-scale investigation with the CHARGE, Global BPgen, and ICBP Consortia. Am J Hum Genet 2014; 95:24-38. [PMID: 24954895 DOI: 10.1016/j.ajhg.2014.05.010] [Show More Authors] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Accepted: 05/20/2014] [Indexed: 01/11/2023] Open
Abstract
Although age-dependent effects on blood pressure (BP) have been reported, they have not been systematically investigated in large-scale genome-wide association studies (GWASs). We leveraged the infrastructure of three well-established consortia (CHARGE, GBPgen, and ICBP) and a nonstandard approach (age stratification and metaregression) to conduct a genome-wide search of common variants with age-dependent effects on systolic (SBP), diastolic (DBP), mean arterial (MAP), and pulse (PP) pressure. In a two-staged design using 99,241 individuals of European ancestry, we identified 20 genome-wide significant (p ≤ 5 × 10(-8)) loci by using joint tests of the SNP main effect and SNP-age interaction. Nine of the significant loci demonstrated nominal evidence of age-dependent effects on BP by tests of the interactions alone. Index SNPs in the EHBP1L1 (DBP and MAP), CASZ1 (SBP and MAP), and GOSR2 (PP) loci exhibited the largest age interactions, with opposite directions of effect in the young versus the old. The changes in the genetic effects over time were small but nonnegligible (up to 1.58 mm Hg over 60 years). The EHBP1L1 locus was discovered through gene-age interactions only in whites but had DBP main effects replicated (p = 8.3 × 10(-4)) in 8,682 Asians from Singapore, indicating potential interethnic heterogeneity. A secondary analysis revealed 22 loci with evidence of age-specific effects (e.g., only in 20 to 29-year-olds). Age can be used to select samples with larger genetic effect sizes and more homogenous phenotypes, which may increase statistical power. Age-dependent effects identified through novel statistical approaches can provide insight into the biology and temporal regulation underlying BP associations.
Collapse
Affiliation(s)
- Jeannette Simino
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Gang Shi
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Georg B Ehret
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva 1211, Switzerland
| | - Xiangjun Gu
- Research Center for Human Genetics, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Shih-Jen Hwang
- Framingham Heart Study, Framingham, MA 01702, USA; Center for Population Studies, National Heart, Lung, and Blood Institute, Framingham, MA 01702, USA
| | - Eric Sijbrands
- Department of Internal Medicine, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands
| | - Albert V Smith
- Icelandic Heart Association, 201 Kopavogur, Iceland; Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Germaine C Verwoert
- Department of Internal Medicine, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands; Department of Epidemiology, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands
| | | | - Gemma Cadby
- Centre for Genetic Origins of Health and Disease, University of Western Australia, Nedlands, WA 6009, Australia; Genetic Epidemiology and Biostatistics Platform, Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada; Samuel Lunenfeld Research Institute, Toronto, ON M5T 3L9, Canada
| | - Peng Chen
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117597, Singapore; Saw Swee Hock School of Public Health, National University Health System, Singapore 117597, Singapore
| | - Ching-Yu Cheng
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117597, Singapore; Saw Swee Hock School of Public Health, National University Health System, Singapore 117597, Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; Department of Ophthalmology, National University Health System, Singapore 119228, Singapore; Singapore Eye Research Institute, Singapore 168751, Singapore; Centre for Quantitative Medicine, Office of Clinical Sciences, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Tanguy Corre
- Department of Medical Genetics, University of Lausanne, 1005 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Rudolf A de Boer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Anuj Goel
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Toby Johnson
- Clinical Pharmacology, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Chiea-Chuen Khor
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117597, Singapore; Saw Swee Hock School of Public Health, National University Health System, Singapore 117597, Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; Department of Ophthalmology, National University Health System, Singapore 119228, Singapore; Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; Department of Paediatrics, National University Health System, Singapore 119074, Singapore
| | - Carla Lluís-Ganella
- Cardiovascular Epidemiology and Genetics, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain
| | - Jian'an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 30101, Finland; Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere 33101, Finland
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Xueling Sim
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA; Centre for Molecular Epidemiology, National University of Singapore, Singapore 119260, Singapore
| | - Siim Sõber
- Human Molecular Genetics Group, Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia
| | - Peter J van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Niek Verweij
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Jing Hua Zhao
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Najaf Amin
- Department of Epidemiology, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Sciences Center, Houston, TX 77225, USA
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands
| | | | - Roberto Elosua
- Cardiovascular Epidemiology and Genetics, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain; Epidemiology and Public Health Network (CIBERESP), 08036 Barcelona, Spain
| | - Oscar H Franco
- Department of Epidemiology, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - Tamara B Harris
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, NIH, Bethesda, MD 20892, USA
| | - Serge Hercberg
- U557 Institut National de la Santé et de la Recherche Médicale, U1125 Institut National de la Recherche Agronomique, Université Paris 13, 93000 Bobigny, France
| | - Albert Hofman
- Department of Epidemiology, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands
| | - Alan L James
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia; School of Medicine and Pharmacology, University of Western Australia, Nedlands, WA 6009, Australia
| | - Andrew D Johnson
- Framingham Heart Study, Framingham, MA 01702, USA; Cardiovascular Epidemiology and Human Genomics Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere 33521, Finland; Department of Clinical Physiology, University of Tampere School of Medicine, Tampere 33521, Finland
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, Cambridge CB2 2SR, UK
| | - Zoltan Kutalik
- Department of Medical Genetics, University of Lausanne, 1005 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Martin G Larson
- Framingham Heart Study, Framingham, MA 01702, USA; Department of Mathematics, Boston University, Boston, MA 02215, USA
| | - Lenore J Launer
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, NIH, Bethesda, MD 20892, USA
| | - Guo Li
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101, USA
| | - Jianjun Liu
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117597, Singapore; Saw Swee Hock School of Public Health, National University Health System, Singapore 117597, Singapore; Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Kiang Liu
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Alanna C Morrison
- Human Genetics Center, University of Texas Health Sciences Center, Houston, TX 77225, USA
| | - Gerjan Navis
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Rick Twee-Hee Ong
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117597, Singapore; Saw Swee Hock School of Public Health, National University Health System, Singapore 117597, Singapore
| | - George J Papanicolau
- Division of Cardiovascular Sciences, National Heart, Lung, & Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Brenda W Penninx
- Department of Psychiatry/EMGO Institute/Neuroscience Campus, VU University Medical Centre, 1081 BT Amsterdam, the Netherlands; Department of Psychiatry, Leiden University Medical Centre, 2333 ZD Leiden, the Netherlands; Department of Psychiatry, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101, USA; Department of Epidemiology, University of Washington, Seattle, WA 98195, USA; Department of Health Services, University of Washington, Seattle, WA 98195, USA; Group Health Research Institute, Group Health Cooperative, Seattle, WA 98101, USA
| | - Leslie J Raffel
- Medical Genetics Institute, Cedars-Sinai Medical Center, Pacific Theatres, Los Angeles, CA 90048, USA
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku 20521, Finland; Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku 20521, Finland
| | - Kenneth Rice
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands; Department of Epidemiology, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands
| | - Lynda M Rose
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA
| | - Serena Sanna
- Istituto di Ricerca Genetica e Biomedica, CNR, Monserrato 09042, Italy
| | - Robert A Scott
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - David S Siscovick
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101, USA; Department of Epidemiology, University of Washington, Seattle, WA 98195, USA
| | - Ronald P Stolk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Andre G Uitterlinden
- Department of Internal Medicine, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands; Department of Epidemiology, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands; Netherland Genomics Inititiative, Netherlands Center for Healthy Aging, The Hague 2509, the Netherlands
| | - Dhananjay Vaidya
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21202, USA
| | - Melanie M van der Klauw
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Ramachandran S Vasan
- Framingham Heart Study, Framingham, MA 01702, USA; Divisions of Epidemiology and Cardiology, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Eranga Nishanthie Vithana
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; Department of Ophthalmology, National University Health System, Singapore 119228, Singapore; Singapore Eye Research Institute, Singapore 168751, Singapore; Neuroscience and Behavioural Disorders (NBD) Program, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, 17487 Greifswald, Germany
| | - Henry Völzke
- Institute for Community Medicine, University of Greifswald, 17487 Greifswald, Germany
| | - Hugh Watkins
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Terri L Young
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710, USA; Division of Neuroscience, Duke-National University of Singapore, Singapore 169857, Singapore
| | - Tin Aung
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; Department of Ophthalmology, National University Health System, Singapore 119228, Singapore; Singapore Eye Research Institute, Singapore 168751, Singapore
| | - Murielle Bochud
- Institute of Social and Preventive Medicine, Lausanne University Hospital, 1010 Lausanne, Switzerland
| | - Martin Farrall
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Catharina A Hartman
- Interdisciplinary Center for Pathology of Emotions, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Maris Laan
- Human Molecular Genetics Group, Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Bethesda, MD 21224, USA
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 30101, Finland; Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere 33101, Finland
| | - Ruth J F Loos
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK; The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Genetics of Obesity and Related Metabolic Traits Program, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Gavin Lucas
- Cardiovascular Epidemiology and Genetics, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain
| | - Pierre Meneton
- U872 Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Paris 75006, France
| | - Lyle J Palmer
- Genetic Epidemiology and Biostatistics Platform, Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada; Samuel Lunenfeld Research Institute, Toronto, ON M5T 3L9, Canada
| | - Rainer Rettig
- Institute of Physiology, University of Greifswald, 17495 Karlsburg, Germany
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - E Shyong Tai
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117597, Singapore; Saw Swee Hock School of Public Health, National University Health System, Singapore 117597, Singapore; Department of Medicine, National University Health System and Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; Duke-National University of Singapore Graduate Medical School, Singapore 169857, Singapore
| | - Yik-Ying Teo
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117597, Singapore; Saw Swee Hock School of Public Health, National University Health System, Singapore 117597, Singapore; Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore; Department of Statistics and Applied Probability, National University of Singapore, Singapore 117543, Singapore; Genome Institute of Singapore, A(∗)STAR, Singapore 138672, Singapore
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands; Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands; Durrer Center for Cardiogenetic Research, 3501 DG Utrecht, the Netherlands
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Tien Yin Wong
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; Department of Ophthalmology, National University Health System, Singapore 119228, Singapore; Singapore Eye Research Institute, Singapore 168751, Singapore
| | - Myriam Fornage
- Research Center for Human Genetics, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA; Human Genetics Center, University of Texas Health Sciences Center, Houston, TX 77225, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, 201 Kopavogur, Iceland; Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Daniel Levy
- Framingham Heart Study, Framingham, MA 01702, USA; Center for Population Studies, National Heart, Lung, and Blood Institute, Framingham, MA 01702, USA; Boston University School of Medicine, Boston, MA 02118, USA
| | - Walter Palmas
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands; Netherland Genomics Inititiative, Netherlands Center for Healthy Aging, The Hague 2509, the Netherlands; Netherland Genomics Initiative, Centre for Medical Systems Biology, 2300 RC Leiden, the Netherlands
| | - Jacqueline C M Witteman
- Department of Epidemiology, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands
| | - Aravinda Chakravarti
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Dabeeru C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA; Departments of Psychiatry, Genetics, and Mathematics, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
23
|
Leu HB, Chung CM, Lin SJ, Lu TM, Yang HC, Ho HY, Ting CT, Lin TH, Sheu SH, Tsai WC, Chen JH, Yin WH, Chiu TY, Chen CI, Pan WH, Chen JW. A novel SNP associated with nighttime pulse pressure in young-onset hypertension patients could be a genetic prognostic factor for cardiovascular events in a general cohort in Taiwan. PLoS One 2014; 9:e97919. [PMID: 24892410 PMCID: PMC4043733 DOI: 10.1371/journal.pone.0097919] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 04/26/2014] [Indexed: 01/11/2023] Open
Abstract
Background Pulse pressure (PP) is a risk factor for cardiovascular disease. It has been reported that ambulatory blood pressure (BP) and nighttime BP parameters are heritable traits. However, the genetic association of pulse pressure and its clinical impact remain undetermined. Method and Results We conducted a genome-wide association study of PP using ambulatory BP monitoring in young-onset hypertensive patients and found a significant association between nighttime PP and SNP rs897876 (p = 0.009) at chromosome 2p14, which contains the predicted gene FLJ16124. Young-onset hypertension patients carrying TT genotypes at rs897876 had higher nighttime PP than those with CT and CC genotypes (TT, 41.6±7.3 mm Hg; CT, 39.1±6.0 mm Hg; CC, 38.9±6.3 mm Hg; p<0.05,). The T risk allele resulted in a cumulative increase in nighttime PP (β = 1.036 mm Hg, se. = 0.298, p<0.001 per T allele). An independent community-based cohort containing 3325 Taiwanese individuals (mean age, 50.2 years) was studied to investigate the genetic impact of rs897876 polymorphisms in determining future cardiovascular events. After an average 7.79±0.28 years of follow-up, the TT genotype of rs897876 was independently associated with an increased risk (in a recessive model) of coronary artery disease (HR, 2.20; 95% CI, 1.20–4.03; p = 0.01) and total cardiovascular events (HR, 1.99; 95% CI, 1.29–3.06; p = 0.002), suggesting that the TT genotype of rs897876C, which is associated with nighttime pulse pressure in young-onset hypertension patients, could be a genetic prognostic factor of cardiovascular events in the general cohort. Conclusion The TT genotype of rs897876C at 2p14 identified in young-onset hypertensive had higher nighttime PP and could be a genetic prognostic factor of cardiovascular events in the general cohort in Taiwan.
Collapse
Affiliation(s)
- Hsin-Bang Leu
- Institute of Clinical Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
- Heath Care and Management Center, Taipei Veterans General Hospital, Taipei, Taiwan
- Divison of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chia-Min Chung
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Shing-Jong Lin
- Institute of Clinical Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
- Divison of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tse-Min Lu
- Institute of Clinical Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
- Divison of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hsin-Chou Yang
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Hung-Yun Ho
- Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chih-Tai Ting
- Taichung Veterans General Hospital, Taichung, Taiwan
| | - Tsung-Hsien Lin
- Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung, Taiwan
| | - Sheng-Hsiung Sheu
- Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung, Taiwan
| | | | - Jyh-Hong Chen
- National Cheng Kung University Hospital, Tainan, Taiwan
| | - Wei-Hsian Yin
- Cheng Hsin Rehabilitation Medical Center, Taipei, Taiwan
| | | | | | - Wen-Harn Pan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- * E-mail: (JC); (WP)
| | - Jaw-Wen Chen
- Institute of Clinical Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
- Divison of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
- * E-mail: (JC); (WP)
| |
Collapse
|
24
|
Chiang KM, Yang HC, Liang YJ, Chen JW, Hwang SM, Ho HY, Ting CT, Lin TH, Sheu SH, Tsai WC, Chen JH, Leu HB, Yin WH, Chiu TY, Chen CI, Lin SJ, Thomas GN, Tomlinson B, Guo Y, Gui HS, Sham PC, Lam TH, Pan WH. A three-stage genome-wide association study combining multilocus test and gene expression analysis for young-onset hypertension in Taiwan Han Chinese. Am J Hypertens 2014; 27:819-27. [PMID: 24413707 DOI: 10.1093/ajh/hpt239] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Although many large-scale genome-wide association studies (GWASs) have been performed, only a few studies have successfully identified replicable, large-impact hypertension loci; even fewer studies have been done on Chinese subjects. Young-onset hypertension (YOH) is considered to be a more promising target disorder to investigate than late-onset hypertension because of its stronger genetic component. METHODS To map YOH genetic variants, we performed a 3-stage study combining 1st-stage multilocus GWASs, 2nd-stage gene expression analysis, and 3rd-stage multilocus confirmatory study. RESULTS In the 1st stage, Illumina550K data from 400 case-control pairs were used, and 22 genes flanked by 14 single nucleotide polymorphism (SNP) septets (P values adjusted for false discovery rate (pFDR) < 3.16×10(-7)) were identified. In the 2nd stage, differential gene expression analysis was carried out for these genes, and 5 genes were selected (pFDR < 0.05). In the 3rd stage, we re-examined the finding with an independent set of 592 case-control pairs and with the joint samples (n = 992 case-control pairs). A total of 6 SNP septets flanking C1orf135, GSN, LARS, and ACTN4 remained significant in all 3 stages. Among them, the same septet flanking ACTN4 was also associated with blood pressure traits in the Hong Kong Hypertension Study (HKHS) and in the Wellcome Trust Case-Control Consortium Hypertension Study (WTCCCHS). LARS was detected in the HKHS, but not in the WTCCCHS. GSN may be specific to Taiwanese individuals because it was not found by either the HKHS or the WTCCCHS. CONCLUSIONS Our study identified 4 previously unknown YOH loci in Han Chinese. Identification of these genes enriches the hypertension susceptibility gene list, thereby shedding light on the etiology of hypertension in Han Chinese.
Collapse
Affiliation(s)
- Kuang-Mao Chiang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Nguyen KDH, Pihur V, Ganesh SK, Rakha A, Cooper RS, Hunt SC, Freedman BI, Coresh J, Kao WHL, Morrison AC, Boerwinkle E, Ehret GB, Chakravarti A. Effects of rare and common blood pressure gene variants on essential hypertension: results from the Family Blood Pressure Program, CLUE, and Atherosclerosis Risk in Communities studies. Circ Res 2013; 112:318-26. [PMID: 23149595 PMCID: PMC3548950 DOI: 10.1161/circresaha.112.276725] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 11/12/2012] [Indexed: 12/16/2022]
Abstract
RATIONALE Hypertension affects ≈30% of adults in industrialized countries and is the major risk factor for cardiovascular disease. OBJECTIVE We sought to study the genetic effect of coding and conserved noncoding variants in syndromic hypertension genes on systolic blood pressure (BP) and diastolic BP to assess their overall impact on essential hypertension. METHODS AND RESULTS We resequenced 11 genes (AGT, CYP11B1, CYP17A1, HSD11B2, NR3C1, NR3C2, SCNN1A, SCNN1B, SCNN1G, WNK1, and WNK4) in 560 European American (EA) and African American ancestry GenNet participants with extreme systolic BP. We investigated genetic associations of 2535 variants with BP in 19997 EAs and in 6069 African Americans in 3 types of analyses. First, we studied the combined effects of all variants in GenNet. Second, we studied 1000 Genomes imputed polymorphic variants in 9747 EA and 3207 African American Atherosclerosis Risk in Communities subjects. Finally, we genotyped 37 missense and common noncoding variants in 6591 EAs and in 6521 individuals (3659 EA/2862 African American) from the CLUE and Family Blood Pressure Program studies, respectively. None of the variants individually reached significant false-discovery rates ≤0.05 for systolic BP and diastolic BP. However, on pooling all coding and noncoding variants, we identified at least 5 loci (AGT, CYP11B1, NR3C2, SCNN1G, and WNK1) with higher association at evolutionary conserved sites. CONCLUSIONS Both rare and common variants at these genes affect BP in the general population with modest effects sizes (<0.05 standard deviation units), and much larger sample sizes are required to assess the impact of individual genes. Collectively, conserved noncoding variants affect BP to a greater extent than missense mutations.
Collapse
Affiliation(s)
- Khanh-Dung H. Nguyen
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Pre-doctoral Training Program in Human Genetics and Molecular Biology, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vasyl Pihur
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Santhi K. Ganesh
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ankit Rakha
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Richard S. Cooper
- Department of Community Medicine, Loyola University School of Medicine, Maywood, IL, USA
| | - Steven C. Hunt
- Cardiovascular Genetics Division, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Barry I. Freedman
- Department of Internal Medicine, Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Joe Coresh
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Wen H. L. Kao
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Alanna C. Morrison
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Georg B. Ehret
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Aravinda Chakravarti
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
26
|
Fava C, Sjögren M, Montagnana M, Danese E, Almgren P, Engström G, Nilsson P, Hedblad B, Guidi GC, Minuz P, Melander O. Prediction of blood pressure changes over time and incidence of hypertension by a genetic risk score in Swedes. Hypertension 2012; 61:319-26. [PMID: 23232644 DOI: 10.1161/hypertensionaha.112.202655] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Recent Genome-Wide Association Studies (GWAS) have pinpointed different single nucleotide polymorphisms consistently associated with blood pressure (BP) and hypertension prevalence. However, little data exist regarding single nucleotide polymorphisms predicting BP variation over time and hypertension incidence. The aim of this study was to confirm the association of a genetic risk score (GRS), based on 29 independent single nucleotide polymorphisms, with cross-sectional BP and hypertension prevalence and to challenge its prediction of BP change over time and hypertension incidence in >17 000 middle-aged Swedes participating in a prospective study, the Malmö Preventive Project, investigated at baseline and over a 23-year average period of follow-up. The GRS was associated with higher systolic and diastolic BP values both at baseline (β ± SEM, 0.968 ± 0.102 mm Hg and 0.585 ± 0.064 mm Hg; P<1E-19 for both) and at reinvestigation (β ± SEM, 1.333 ± 0.161 mm Hg and 0.724 ± 0.086 mm Hg; P<1E-15 for both) and with increased hypertension prevalence (odds ratio [95% CI], 1.192 [1.140-1.245] and 1.144 [1.107-1.183]; P<1E-15 for both). The GRS was positively associated with change (Δ) in BP (β ± SEM, 0.033 ± 0.008 mm Hg/y and 0.023 ± 0.004 mm Hg/y; P<1E-04 for both) and hypertension incidence (odds ratio [95% CI], 1.110 [1.065-1.156]; P=6.7 E-07), independently from traditional risk factors. The relative weight of the GRS was lower in magnitude than obesity or prehypertension, but comparable with diabetes mellitus or a positive family history of hypertension. A C-statistics analysis does not show any improvement in the prediction of incident hypertension on top of traditional risk factors. Our data from a large cohort study show that a GRS is independently associated with BP increase and incidence of hypertension.
Collapse
Affiliation(s)
- Cristiano Fava
- Department of Clinical Sciences, Lund University, University Hospital of Malmö, Sweden.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Lane HY, Tsai GE, Lin E. Assessing Gene-Gene Interactions in Pharmacogenomics. Mol Diagn Ther 2012; 16:15-27. [DOI: 10.1007/bf03256426] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
28
|
A two-stage matched case-control study on multiple hypertensive candidate genes in Han Chinese. Am J Hypertens 2012; 25:804-11. [PMID: 22534794 DOI: 10.1038/ajh.2012.44] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Hypertension affects about 1/3 of adults worldwide, ~3.8 million in Taiwan, 160 million in China, and 1 billion worldwide. It is a major risk factor leading to stroke, cardiovascular disease, and end-stage renal disease. In each year, more than 13.5 million deaths are due to hypertension-related diseases worldwide. METHODS We performed a two-stage association study of hypertension using genotype data of single-nucleotide polymorphisms (SNPs) from 992 young-onset hypertensive cases and 992 matched controls of Han Chinese in Taiwan. A total of 238 SNPs of 36 highly replicated hypertension candidate genes with functional importance were investigated. Association analysis was carried out using conditional logistic regression. RESULTS We identified two SNPs that were strongly associated with hypertension in both the first and the second stages. The first SNP (rs2301339) is located at guanine nucleotide-binding protein β3 subunit (GNB3) and the other one (rs17254521) is located at insulin receptor (INSR). CONCLUSIONS SNP rs2301339 is perfectly linked in linkage disequilibrium (LD) with C825T (rs5443) which has been associated with hypertension in Caucasian, but inconsistent in Asian populations. However, we found that in our sample this SNP has an opposite effect with the previous findings. In summary, this study identified one novel SNP in GNB3 and one novel SNP in INSR that are strongly associated with young-onset hypertension. Due to relatively small sample size, the results should still be interpreted with caution and need to be replicated in other studies.
Collapse
|
29
|
|
30
|
Between candidate genes and whole genomes: time for alternative approaches in blood pressure genetics. Curr Hypertens Rep 2012; 14:46-61. [PMID: 22161147 DOI: 10.1007/s11906-011-0241-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Blood pressure has a significant genetic component, but less than 3% of the observed variance has been attributed to genetic variants identified to date. Candidate gene studies of rare, monogenic hypertensive syndromes have conclusively implicated several genes altering renal sodium balance, and studies of essential hypertension have inconsistently implicated over 50 genes in pathways affecting renal sodium balance and other functions. Genome-wide linkage scans have replicated numerous quantitative trait loci throughout the genome, and over 50 single nucleotide polymorphisms (SNPs) have been replicated in multiple genome-wide association studies. These studies provide considerable evidence that epistasis and other interactions play a role in the genetic architecture of blood pressure regulation, but candidate gene studies have limited scope to test for epistasis, and genome-wide studies have low power for both main effects and interactions. This review summarizes the genetic findings to date for blood pressure, and it proposes focused, pathway-based approaches involving epistasis, gene-environment interactions, and next-generation sequencing to further the genetic dissection of blood pressure and hypertension.
Collapse
|
31
|
Yang HC, Liang YJ, Chen JW, Chiang KM, Chung CM, Ho HY, Ting CT, Lin TH, Sheu SH, Tsai WC, Chen JH, Leu HB, Yin WH, Chiu TY, Chern CI, Lin SJ, Tomlinson B, Guo Y, Sham PC, Cherny SS, Lam TH, Thomas GN, Pan WH. Identification of IGF1, SLC4A4, WWOX, and SFMBT1 as hypertension susceptibility genes in Han Chinese with a genome-wide gene-based association study. PLoS One 2012; 7:e32907. [PMID: 22479346 PMCID: PMC3315540 DOI: 10.1371/journal.pone.0032907] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 02/07/2012] [Indexed: 01/11/2023] Open
Abstract
Hypertension is a complex disorder with high prevalence rates all over the world. We conducted the first genome-wide gene-based association scan for hypertension in a Han Chinese population. By analyzing genome-wide single-nucleotide-polymorphism data of 400 matched pairs of young-onset hypertensive patients and normotensive controls genotyped with the Illumina HumanHap550-Duo BeadChip, 100 susceptibility genes for hypertension were identified and also validated with permutation tests. Seventeen of the 100 genes exhibited differential allelic and expression distributions between patient and control groups. These genes provided a good molecular signature for classifying hypertensive patients and normotensive controls. Among the 17 genes, IGF1, SLC4A4, WWOX, and SFMBT1 were not only identified by our gene-based association scan and gene expression analysis but were also replicated by a gene-based association analysis of the Hong Kong Hypertension Study. Moreover, cis-acting expression quantitative trait loci associated with the differentially expressed genes were found and linked to hypertension. IGF1, which encodes insulin-like growth factor 1, is associated with cardiovascular disorders, metabolic syndrome, decreased body weight/size, and changes of insulin levels in mice. SLC4A4, which encodes the electrogenic sodium bicarbonate cotransporter 1, is associated with decreased body weight/size and abnormal ion homeostasis in mice. WWOX, which encodes the WW domain-containing protein, is related to hypoglycemia and hyperphosphatemia. SFMBT1, which encodes the scm-like with four MBT domains protein 1, is a novel hypertension gene. GRB14, TMEM56 and KIAA1797 exhibited highly significant differential allelic and expressed distributions between hypertensive patients and normotensive controls. GRB14 was also found relevant to blood pressure in a previous genetic association study in East Asian populations. TMEM56 and KIAA1797 may be specific to Taiwanese populations, because they were not validated by the two replication studies. Identification of these genes enriches the collection of hypertension susceptibility genes, thereby shedding light on the etiology of hypertension in Han Chinese populations.
Collapse
Affiliation(s)
- Hsin-Chou Yang
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Yu-Jen Liang
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Jaw-Wen Chen
- National Yang-Ming University School of Medicine and Taipei Veterans General Hospital, Taipei, Taiwan
| | - Kuang-Mao Chiang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- School of Public Health, National Medical Defense Center, Taipei, Taiwan
| | - Chia-Min Chung
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hung-Yun Ho
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chih-Tai Ting
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Tsung-Hsien Lin
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Sheng-Hsiung Sheu
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Wei-Chuan Tsai
- Department of Internal Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jyh-Hong Chen
- Department of Internal Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsin-Bang Leu
- National Yang-Ming University School of Medicine and Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wei-Hsian Yin
- Division of Cardiology, Cheng-Hsin Rehabilitation Medical Center, Taipei, Taiwan
| | - Ting-Yu Chiu
- Division of Cardiology, Min-Sheng General Hospital, Taoyuan, Taiwan
| | - Ching-Iuan Chern
- Division of Cardiology, Min-Sheng General Hospital, Taoyuan, Taiwan
| | - Shing-Jong Lin
- National Yang-Ming University School of Medicine and Taipei Veterans General Hospital, Taipei, Taiwan
| | - Brian Tomlinson
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Youling Guo
- Department of Psychiatry, The University of Hong Kong, Hong Kong, China
| | - Pak C. Sham
- Department of Psychiatry, The University of Hong Kong, Hong Kong, China
- The State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| | - Stacey S. Cherny
- Department of Psychiatry, The University of Hong Kong, Hong Kong, China
- The State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| | - Tai Hing Lam
- School of Public Health, The University of Hong Kong, Hong Kong, China
| | - G. Neil Thomas
- Public Health, Epidemiology and Biostatistics, School of Health and Population Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Wen-Harn Pan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Division of Preventive Medicine and Health Services Research, Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan
- * E-mail:
| |
Collapse
|
32
|
Reevaluation of the association of seven candidate genes with blood pressure and hypertension: a replication study and meta-analysis with a larger sample size. Hypertens Res 2012; 35:825-31. [DOI: 10.1038/hr.2012.43] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
33
|
Guo Y, Tomlinson B, Chu T, Fang YJ, Gui H, Tang CS, Yip BH, Cherny SS, Hur YM, Sham PC, Lam TH, Thomas NG. A genome-wide linkage and association scan reveals novel loci for hypertension and blood pressure traits. PLoS One 2012; 7:e31489. [PMID: 22384028 PMCID: PMC3286457 DOI: 10.1371/journal.pone.0031489] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 01/11/2012] [Indexed: 01/11/2023] Open
Abstract
Hypertension is caused by the interaction of environmental and genetic factors. The condition which is very common, with about 18% of the adult Hong Kong Chinese population and over 50% of older individuals affected, is responsible for considerable morbidity and mortality. To identify genes influencing hypertension and blood pressure, we conducted a combined linkage and association study using over 500,000 single nucleotide polymorphisms (SNPs) genotyped in 328 individuals comprising 111 hypertensive probands and their siblings. Using a family-based association test, we found an association with SNPs on chromosome 5q31.1 (rs6596140; P<9×10−8) for hypertension. One candidate gene, PDC, was replicated, with rs3817586 on 1q31.1 attaining P = 2.5×10−4 and 2.9×10−5 in the within-family tests for DBP and MAP, respectively. We also identified regions of significant linkage for systolic and diastolic blood pressure on chromosomes 2q22 and 5p13, respectively. Further family-based association analysis of the linkage peak on chromosome 5 yielded a significant association (rs1605685, P<7×10−5) for DBP. This is the first combined linkage and association study of hypertension and its related quantitative traits with Chinese ancestry. The associations reported here account for the action of common variants whereas the discovery of linkage regions may point to novel targets for rare variant screening.
Collapse
Affiliation(s)
- Youling Guo
- The State Key Laboratory of Brain and Cognitive Sciences, Department of Psychiatry, The University of Hong Kong, Hong Kong, China
| | - Brian Tomlinson
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong, China
| | - Tanya Chu
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong, China
| | - Yu Jing Fang
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong, China
| | - Hongsheng Gui
- The State Key Laboratory of Brain and Cognitive Sciences, Department of Psychiatry, The University of Hong Kong, Hong Kong, China
| | - Clara S. Tang
- The State Key Laboratory of Brain and Cognitive Sciences, Department of Psychiatry, The University of Hong Kong, Hong Kong, China
| | - Benjamin H. Yip
- The State Key Laboratory of Brain and Cognitive Sciences, Department of Psychiatry, The University of Hong Kong, Hong Kong, China
| | - Stacey S. Cherny
- The State Key Laboratory of Brain and Cognitive Sciences, Department of Psychiatry, The University of Hong Kong, Hong Kong, China
- * E-mail: (SSC); (THL); (NGT)
| | - Yoon-Mi Hur
- Mokpo National University, Seoul, South Korea
| | - Pak Chung Sham
- The State Key Laboratory of Brain and Cognitive Sciences, Department of Psychiatry, The University of Hong Kong, Hong Kong, China
| | - Tai Hing Lam
- School of Public Health, The University of Hong Kong, Hong Kong, China
- * E-mail: (SSC); (THL); (NGT)
| | - Neil G. Thomas
- Unit of Public Health, Epidemiology and Biostatistics, University of Birmingham, Birmingham, United Kingdom
- * E-mail: (SSC); (THL); (NGT)
| |
Collapse
|
34
|
Abstract
Cardiovascular diseases are the leading cause of death worldwide. Essential hypertension is a major risk factor for the development of other cardiovascular diseases and is caused by a combination of environmental and genetic factors, with up to 50% of blood pressure variance currently attributed to an individual's genetic makeup. By studying genes that cause monogenic forms of hypertension and pathways relevant to blood pressure control, a number of polymorphisms have been identified that increase an individual's risk of developing high blood pressure. We report on candidate gene association studies and genome-wide association studies that have been performed to date in the field of hypertension research. It is becoming clear that for the majority of people there is no single gene polymorphism that causes hypertension, but rather a number of common genetic variants, each having a small effect. Using pharmacogenomics to personalize the treatment of hypertension holds promise for achieving and sustaining normotensive pressures quickly, while minimizing the risk of adverse reactions and unwanted side-effects. This will decrease the risk of stroke and myocardial infarction in individuals and lead to a reduced burden of disease upon society as a whole.
Collapse
|
35
|
Lane HY, Tsai GE, Lin E. Assessing gene-gene interactions in pharmacogenomics. Mol Diagn Ther 2012; 16:15-27. [PMID: 22352452 DOI: 10.2165/11597270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
In pharmacogenomics studies, gene-gene interactions play an important role in characterizing a trait that involves complex pharmacokinetic and pharmacodynamic mechanisms, particularly when each involved feature only demonstrates a minor effect. In addition to the candidate gene approach, genome-wide association studies (GWAS) are widely utilized to identify common variants that are associated with treatment response. In the wake of recent advances in scientific research, a paradigm shift from GWAS to whole-genome sequencing is expected, because of the reduced cost and the increased throughput of next-generation sequencing technologies. This review first outlines several promising methods for addressing gene-gene interactions in pharmacogenomics studies. We then summarize some candidate gene studies for various treatments with consideration of gene-gene interactions. Furthermore, we give a brief overview for the pharmacogenomics studies with the GWAS approach and describe the limitations of these GWAS in terms of gene-gene interactions. Future research in translational medicine promises to lead to mechanistic findings related to drug responsiveness in light of complex gene-gene interactions and will probably make major contributions to individualized medicine and therapeutic decision-making.
Collapse
Affiliation(s)
- Hsien-Yuan Lane
- Department of Psychiatry, China Medical University Hospital, Taichung, Taiwan
| | | | | |
Collapse
|
36
|
Ji LD, Zhang LN, Xu J. Genome-wide association studies of hypertension: Achievements, difficulties and strategies. World J Hypertens 2011; 1:10-14. [DOI: 10.5494/wjh.v1.i1.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Estimated from family studies, the heritability of hypertension ranges from 31% to 68%. Linkage studies and candidate gene association studies were once widely used to investigate the genetic mechanisms of hypertension. However, results from these studies could only explain 1%-2% heritability. With the technological advances and subsequently the accomplishment of the Human Genome Project, genome-wide association studies (GWA studies) have been applied to find genome-wide significant signals for many common diseases. Current GWA studies of hypertension have identified dozens of hypertension or blood pressure associated variants. However, different GWA study identified different variants and the results could hardly be replicated in other studies. Therefore, a debate took place on whether GWA studies will unlock the genetic basis of hypertension and whether we shall continue throwing millions of dollars on GWA studies. This review gives a short introduction to the history of genetic study on hypertension and summarizes the current findings for GWA studies of hypertension or blood pressure. Finally, we will discuss that debate and try to find alternative strategies and technologies that may hold a greater chance to make progress in understanding the genetic risk factors of hypertension and blood pressure regulation.
Collapse
|
37
|
Lin Y, Lai X, Chen B, Xu Y, Huang B, Chen Z, Zhu S, Yao J, Jiang Q, Huang H, Wen J, Chen G. Genetic variations in CYP17A1, CACNB2 and PLEKHA7 are associated with blood pressure and/or hypertension in She ethnic minority of China. Atherosclerosis 2011; 219:709-14. [PMID: 21963141 DOI: 10.1016/j.atherosclerosis.2011.09.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 09/03/2011] [Accepted: 09/05/2011] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Two large-scale genome-wide association studies (GWAs) have identified multiple variants associated with blood pressure (BP) or hypertension. The present study was to investigate whether some variations were associated with BP traits and hypertension or even prehypertension in adult She ethnic minority of China. METHODS The population of the present study comprised 4460 (1979 males and 2481 females, respectively) unrelated she ethnic minority based on a cross-sectional study from Ningde City in Fujian province of China. There were 1692 hypertensives, 1600 prehypertensives and 1168 normotensive controls, respectively. We genotyped 7 variants in CYP17A1, PLEKHA7, CACNB2, ATP2B1, TBX3-TBX5, CSK-ULK3 and SH2B3 reported by the previous GWAs on Europeans. All analyses were performed in an additive genetic model. RESULTS As the minor allele of rs653178 in/near SH2B3 was very rare with the frequency of 0.018, we excluded this single nucleotide polymorphism (SNP) in the further analyses. Of the other 6 loci, linear regression analyses revealed that rs11191548 in CYP17A1 and rs11014166 in CACNB2 were significantly associated with systolic BP (β = -1.17, P = 0.002 and β = -0.50, P = 0.006, respectively), while only SNP rs11191548 was significantly associated with diastolic BP (β = -0.56, P=0.002) after adjusted by age, sex and BMI. Two variants in CACNB2 and PLEKHA7 were found to be significantly related to hypertension (odds ratios [OR] and (95% confidence interval [CI]): 0.79 (0.65-0.97) and 1.19 (1.01-1.41), respectively) in logistic regression analyses after adjusted by age, sex and BMI. In addition, we found that combined risk alleles of the 6 SNPs increased risk of hypertension in a stepwise fashion (P for trend < 0.001). However, none of the 6 SNPs was significantly associated with BMI or prehypertension status. While logistic analysis showed that subjects with cumulative risk alleles more than 9 had significantly higher risk for prehypertension (adjusted OR: 3.10, P < 0.001) compared with those with risk alleles less than 4. CONCLUSIONS We replicated that variations in CYP17A1, CACNB2 and PLEKHA7 were related to BP traits and/or hypertension in She population. In addition, although we failed to observe single gene associated with prehypertension, we first found that conjoint effect of multiple risk alleles on BP might increase the risk of progressing to prehypertension.
Collapse
Affiliation(s)
- Yinghua Lin
- Department of Endocrinology, Ningde Municipal Hospital, Ningde 352100, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Jeemon P, Pettigrew K, Sainsbury C, Prabhakaran D, Padmanabhan S. Implications of discoveries from genome-wide association studies in current cardiovascular practice. World J Cardiol 2011; 3:230-47. [PMID: 21860704 PMCID: PMC3158871 DOI: 10.4330/wjc.v3.i7.230] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 07/02/2011] [Accepted: 07/10/2011] [Indexed: 02/06/2023] Open
Abstract
Genome-wide association studies (GWAS) have identified several genetic variants associated with coronary heart disease (CHD), and variations in plasma lipoproteins and blood pressure (BP). Loci corresponding to CDKN2A/CDKN2B/ANRIL, MTHFD1L, CELSR2, PSRC1 and SORT1 genes have been associated with CHD, and TMEM57, DOCK7, CELSR2, APOB, ABCG5, HMGCR, TRIB1, FADS2/S3, LDLR, NCAN and TOMM40-APOE with total cholesterol. Similarly, CELSR2-PSRC1-SORT1, PCSK9, APOB, HMGCR, NCAN-CILP2-PBX4, LDLR, TOMM40-APOE, and APOC1-APOE are associated with variations in low-density lipoprotein cholesterol levels. Altogether, forty, forty three and twenty loci have been associated with high-density lipoprotein cholesterol, triglycerides and BP phenotypes, respectively. Some of these identified loci are common for all the traits, some do not map to functional genes, and some are located in genes that encode for proteins not previously known to be involved in the biological pathway of the trait. GWAS have been successful at identifying new and unexpected genetic loci common to diseases and traits, thus rapidly providing key novel insights into disease biology. Since genotype information is fixed, with minimum biological variability, it is useful in early life risk prediction. However, these variants explain only a small proportion of the observed variance of these traits. Therefore, the utility of genetic determinants in assessing risk at later stages of life has limited immediate clinical impact. The future application of genetic screening will be in identifying risk groups early in life to direct targeted preventive measures.
Collapse
Affiliation(s)
- Panniyammakal Jeemon
- Panniyammakal Jeemon, Kerry Pettigrew, Christopher Sainsbury, Sandosh Padmanabhan, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, G12 8TA, United Kingdom
| | | | | | | | | |
Collapse
|
39
|
Kraja AT, Hunt SC, Rao DC, Dávila-Román VG, Arnett DK, Province MA. Genetics of hypertension and cardiovascular disease and their interconnected pathways: lessons from large studies. Curr Hypertens Rep 2011; 13:46-54. [PMID: 21128019 DOI: 10.1007/s11906-010-0174-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Blood pressure (BP), hypertension (HT) and cardiovascular disease (CVD) are common complex phenotypes, which are affected by multiple genetic and environmental factors. This article describes recent genome-wide association studies (GWAS) that have reported causative variants for BP/HT and CVD/heart traits and analyzes the overlapping associated gene polymorphisms. It also examines potential replication of findings from the HyperGEN data on African Americans and whites. Several genes involved in BP/HT regulation also appear to be involved in CVD. A better picture is emerging, with overlapping hot-spot regions and with interconnected pathways between BP/HT and CVD. A systemic approach to full understanding of BP/HT and CVD development and their progression to disease may lead to the identification of gene targets and pathways for the development of novel therapeutic interventions.
Collapse
Affiliation(s)
- Aldi T Kraja
- Division of Statistical Genomics, Washington University School of Medicine, 4444 Forest Park Avenue, St. Louis, MO 63108, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Yang HC, Lin HC, Kang M, Chen CH, Lin CW, Li LH, Wu JY, Chen YT, Pan WH. SAQC: SNP array quality control. BMC Bioinformatics 2011; 12:100. [PMID: 21501472 PMCID: PMC3101186 DOI: 10.1186/1471-2105-12-100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Accepted: 04/18/2011] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Genome-wide single-nucleotide polymorphism (SNP) arrays containing hundreds of thousands of SNPs from the human genome have proven useful for studying important human genome questions. Data quality of SNP arrays plays a key role in the accuracy and precision of downstream data analyses. However, good indices for assessing data quality of SNP arrays have not yet been developed. RESULTS We developed new quality indices to measure the quality of SNP arrays and/or DNA samples and investigated their statistical properties. The indices quantify a departure of estimated individual-level allele frequencies (AFs) from expected frequencies via standardized distances. The proposed quality indices followed lognormal distributions in several large genomic studies that we empirically evaluated. AF reference data and quality index reference data for different SNP array platforms were established based on samples from various reference populations. Furthermore, a confidence interval method based on the underlying empirical distributions of quality indices was developed to identify poor-quality SNP arrays and/or DNA samples. Analyses of authentic biological data and simulated data show that this new method is sensitive and specific for the detection of poor-quality SNP arrays and/or DNA samples. CONCLUSIONS This study introduces new quality indices, establishes references for AFs and quality indices, and develops a detection method for poor-quality SNP arrays and/or DNA samples. We have developed a new computer program that utilizes these methods called SNP Array Quality Control (SAQC). SAQC software is written in R and R-GUI and was developed as a user-friendly tool for the visualization and evaluation of data quality of genome-wide SNP arrays. The program is available online (http://www.stat.sinica.edu.tw/hsinchou/genetics/quality/SAQC.htm).
Collapse
Affiliation(s)
- Hsin-Chou Yang
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Mick E, McGough JJ, Middleton FA, Neale B, Faraone SV. Genome-wide association study of blood pressure response to methylphenidate treatment of attention-deficit/hyperactivity disorder. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:466-72. [PMID: 21130132 DOI: 10.1016/j.pnpbp.2010.11.037] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 11/23/2010] [Accepted: 11/23/2010] [Indexed: 12/31/2022]
Abstract
OBJECTIVE We conducted a genome-wide association study of blood pressure in an open-label study of the methylphenidate transdermal system (MTS) for the treatment of attention-deficit/hyperactivity disorder (ADHD). METHOD Genotyping was conducted with the Affymetrix Genome-Wide Human SNP Array 6.0. Multivariate association analyses were conducted using the software package PLINK. After data cleaning and quality control we tested 316,934 SNPs in 140 children with ADHD. RESULTS We observed no genome-wide statistically significant findings, but a SNP in a K(+)-dependent Na(+)/Ca(2+) exchanger expressed in vascular smooth muscle (SLC24A3) was included in our top associations at p<1E-04. Genetic enrichment analyses of genes with ≥1 SNP significant at p<0.01, implicated several functional categories (FERM domain, p=5.0E-07; immunoglobulin domain, p=8.1E-06; the transmembrane region, p=4.4E-05; channel activity, p=2.0E-04; and type-III fibronectins, p=2.7E-05) harboring genes previously associated with related cardiovascular phenotypes. CONCLUSIONS The hypothesis generating results from this study suggests that polymorphisms in several genes consistently associated with cardiovascular diseases may impact changes in blood pressure observed with methylphenidate pharmacotherapy in children with ADHD.
Collapse
Affiliation(s)
- Eric Mick
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States.
| | | | | | | | | |
Collapse
|
42
|
Puppala S, Coletta DK, Schneider J, Hu SL, Farook VS, Dyer TD, Arya R, Blangero J, Duggirala R, DeFronzo RA, Jenkinson CP. Genome-Wide Linkage Screen for Systolic Blood Pressure in the Veterans Administration Genetic Epidemiology Study (VAGES) of Mexican-Americans and Confirmation of a Major Susceptibility Locus on Chromosome 6q14.1. Hum Hered 2011; 71:1-10. [DOI: 10.1159/000323143] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Accepted: 11/22/2010] [Indexed: 01/11/2023] Open
|
43
|
Validation of genetic association in apelin-AGTRL1 system with hypertension in a larger Han Chinese population. J Hypertens 2010; 28:1854-61. [PMID: 20485192 DOI: 10.1097/hjh.0b013e32833b1fad] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND OBJECTIVE We have recently resequenced apelin and AGTRL1 genes to identify candidate polymorphisms in family-based association with hypertension and related phenotypes. In this study, we aimed to determine and replicate these polymorphisms via a larger, independent case-control study in Shanghai Han Chinese. METHODS Two polymorphisms [rs3761581 (A/C) and T-1860C] in apelin gene and two [rs7119375 (G/A), rs10501367 (G/A)] in AGTRL1 gene were genotyped using the TaqMan assay among 969 patients diagnosed with essential hypertension and 980 age and sex-matched controls. Data were analyzed using Haplo.stats program. RESULTS In single-locus analysis, significant differences were observed in allele distribution of rs3761581 (P = 0.0156) in men and in rs7119375 (P = 0.0488) genotype distribution in women between patients and controls. Haplotype analysis indicated that haplotypes C-C-G-G (in order of T-1860C, rs3761581, rs7119375 and rs10501367) [adjusted odds ratio (ORadjusted) = 1.67, P = 0.0061] and T-A-A-A (ORadjusted = 1.62, P = 0.0008) conferred an increased risk for hypertension after adjustment for age, onset age, body mass index (BMI) and waist-to-hip ratio, whereas haplotype C-C-A-A (ORadjusted = 0.33, P = 0.0048) conferred a protective effect. In women, increased risk for hypertension was seen for haplotypes T-A-G-G (ORadjusted = 1.30, P = 0.0051), C-C-G-G (ORadjusted = 2.86, P < 0.0001), T-A-A-A (ORadjusted = 1.66, P = 0.0003) and C-C-A-A (ORadjusted = 2.65, P < 0.0001), whereas decreased risk was seen for haplotypes C-A-G-G (ORadjusted = 0.48, P < 0.0001) and T-C-G-G (ORadjusted = 0.40, P < 0.0001). Further haplotype-phenotype analyses indicated the robustness of these associations. For example, haplotype T-A-A-A was significantly associated with obesity index (BMI and waist-to-hip ratio) in both sexes and blood pressures in men (P-sim < 0.05). CONCLUSION In this exploratory pilot case-control study, we found robust haplotype-based and haplotype-phenotype associations of four well characterized polymorphisms in apelin-AGTRL1 system with hypertension and related phenotypes.
Collapse
|
44
|
Niu W, Zhang Y, Ji K, Gu M, Gao P, Zhu D. Confirmation of top polymorphisms in hypertension genome wide association study among Han Chinese. Clin Chim Acta 2010; 411:1491-5. [DOI: 10.1016/j.cca.2010.06.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Revised: 06/03/2010] [Accepted: 06/03/2010] [Indexed: 01/11/2023]
|
45
|
Kao YT, Huang CC, Leu HB, Wu TC, Huang PH, Lin SJ, Chen JW. Ambulatory pulse pressure as a novel predictor for long-term prognosis in essential hypertensive patients. J Hum Hypertens 2010; 25:444-50. [DOI: 10.1038/jhh.2010.80] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
46
|
Abstract
Blood pressure and hypertension have significant genetic underpinnings that may be age-dependent. The age-dependency, significant contributions from environmental factors such as diet and exercise, and inherent moment-to-moment variability complicate the identification of the genes contributing to the development of hypertension. Although genetic abnormalities may have moderate effects, the physiologic pathways involving these genes have redundant compensating mechanisms to bring the system back into equilibrium. This has the effect of reducing or completely masking the initial genetic defects, one of the hypothesized reasons for the small genetic effects found by the recent genome-wide association studies. This review article discusses the concept of initiators versus compensators in the context of finding genes related to hypertension development. A brief review is provided of some key genes found to be associated with hypertension, including the genes identified from the nine genome-wide association studies published to date.
Collapse
Affiliation(s)
- Steven C Hunt
- Cardiovascular Genetics Division, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84108, USA.
| |
Collapse
|
47
|
Yang HC, Lin HC, Huang MC, Li LH, Pan WH, Wu JY, Chen YT. A new analysis tool for individual-level allele frequency for genomic studies. BMC Genomics 2010; 11:415. [PMID: 20602748 PMCID: PMC2996943 DOI: 10.1186/1471-2164-11-415] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Accepted: 07/05/2010] [Indexed: 01/23/2023] Open
Abstract
Background Allele frequency is one of the most important population indices and has been broadly applied to genetic/genomic studies. Estimation of allele frequency using genotypes is convenient but may lose data information and be sensitive to genotyping errors. Results This study utilizes a unified intensity-measuring approach to estimating individual-level allele frequencies for 1,104 and 1,270 samples genotyped with the single-nucleotide-polymorphism arrays of the Affymetrix Human Mapping 100K and 500K Sets, respectively. Allele frequencies of all samples are estimated and adjusted by coefficients of preferential amplification/hybridization (CPA), and large ethnicity-specific and cross-ethnicity databases of CPA and allele frequency are established. The results show that using the CPA significantly improves the accuracy of allele frequency estimates; moreover, this paramount factor is insensitive to the time of data acquisition, effect of laboratory site, type of gene chip, and phenotypic status. Based on accurate allele frequency estimates, analytic methods based on individual-level allele frequencies are developed and successfully applied to discover genomic patterns of allele frequencies, detect chromosomal abnormalities, classify sample groups, identify outlier samples, and estimate the purity of tumor samples. The methods are packaged into a new analysis tool, ALOHA (Allele-frequency/Loss-of-heterozygosity/Allele-imbalance). Conclusions This is the first time that these important genetic/genomic applications have been simultaneously conducted by the analyses of individual-level allele frequencies estimated by a unified intensity-measuring approach. We expect that additional practical applications for allele frequency analysis will be found. The developed databases and tools provide useful resources for human genome analysis via high-throughput single-nucleotide-polymorphism arrays. The ALOHA software was written in R and R GUI and can be downloaded at http://www.stat.sinica.edu.tw/hsinchou/genetics/aloha/ALOHA.htm.
Collapse
Affiliation(s)
- Hsin-Chou Yang
- Institute of Statistical Science, Academia Sinica, Taipei 115, Taiwan.
| | | | | | | | | | | | | |
Collapse
|
48
|
Kitsios GD, Zintzaras E. Synopsis and data synthesis of genetic association studies in hypertension for the adrenergic receptor family genes: the CUMAGAS-HYPERT database. Am J Hypertens 2010; 23:305-13. [PMID: 20044737 PMCID: PMC2962566 DOI: 10.1038/ajh.2009.251] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The adrenergic receptor (adrenoceptor) family genes have been extensively studied as candidate genes in hypertension but the results of individual genetic association studies (GAS) are controversial and inconclusive. To clarify these data, a systematic assessment of GAS for adrenoceptor family genes in hypertension was conducted. METHODS Data from 163 GAS involving 7 genes and 37 distinct genetic variants were analyzed and cataloged in CUMAGAS-HYPERT (Cumulative Meta-analysis of Genetic Association Studies-HYPERTension; a web-based information system, which allows the retrieval and synthesis of data from GAS in hypertension, available at http://biomath.med.uth.gr). Data from genome-wide association studies involving the adrenoceptor family genes were also systematically searched. RESULTS Individual GAS reported inconsistent associations and had limited power to detect modest genetic effects, with only 1.2% having power >80%. Thirteen variants were investigated by three or more studies and their results were subject to meta-analysis. In the main meta-analyses, significant results were shown for five variants (ADRB1 p.Arg389Gly, ADRB1 p.Ser49Gly, ADRB2 g.9368308A>G, ADRB3 p.Trp64Arg, and ADRA1A p.Cys347Arg) under the allelic contrast and/or the dominant model. Subgroup analyses by ethnicity and gender detected significant associations for three variants (ADRB1 p.Arg389Gly in east Asians, ADRB2 p.Gln27Glu in whites, and ADRB3 p.Trp64Arg in whites and in males). Heterogeneity ranged from none to high. No significant associations were recorded from genome-wide studies. CONCLUSIONS There is evidence to implicate adrenoceptor genes in hypertension, although future studies designed to investigate epistatic and gene-environment interactions would allow more solid conclusions to be drawn about the role of these genes in hypertension.
Collapse
Affiliation(s)
- Georgios D Kitsios
- Department of Biomathematics, University of Thessaly School of Medicine, Larissa, Greece
| | | |
Collapse
|
49
|
Ehret GB. Genome-wide association studies: contribution of genomics to understanding blood pressure and essential hypertension. Curr Hypertens Rep 2010; 12:17-25. [PMID: 20425154 PMCID: PMC2865585 DOI: 10.1007/s11906-009-0086-6] [Citation(s) in RCA: 156] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Contemporary genomic tools now allow the fast and reliable genotyping of hundreds of thousands of variants and permit an unbiased interrogation of the common variability across the human genome. These technical advances have been the basis of numerous recent investigations of genes underlying complex genetic traits, and the results for blood pressure and hypertension have been of particular interest. The pathophysiology of the complex genetic trait blood pressure and hypertension is unclear. The heritability of essential hypertension is high and insights can be gained by finding associated genes. Current genome-wide association studies (GWAS) have identified 10 to 20 loci in or near genes that generally were not expected to be associated with blood pressure or essential hypertension; more significant variants will be discovered when even larger and more refined studies become available. This article gives a short introduction to GWAS and summarizes the current findings for blood pressure and hypertension.
Collapse
Affiliation(s)
- Georg B Ehret
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, 733 N. Broadway, Baltimore, MD 21205, USA.
| |
Collapse
|
50
|
Kitsios GD, Zintzaras E. AnNOS3Haplotype Is Protective against Hypertension in a Caucasian Population. Int J Hypertens 2010; 2010:865031. [PMID: 20981307 PMCID: PMC2958494 DOI: 10.4061/2010/865031] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Accepted: 02/13/2010] [Indexed: 11/22/2022] Open
Abstract
The endothelial nitric oxide synthase gene (NOS3) has been implicated in the development of hypertension, although the specific role of variants and haplotypes has not been clarified. In this study, the association of three polymorphisms (promoter T786C, intronic 4a/b, and nonsynonymous G894T) was tested in a case-control sample of 230 patients with essential hypertension and 306 healthy controls. Haplotype analysis was also performed. The mutant allele a∗ of the 4a/b polymorphism showed a protective effect against hypertension under a dominant model (odds ratio = 0.64, 95% confidence interval (0.44–0.93)), although this effect was not significant after the adjustment for covariates (P = 0.06). The estimated frequency of the haplotype composed of the T786∗, 4a∗, and G894∗ alleles was significantly higher in controls (5.5%) compared to cases (2%). These results indicate that although individual NOS3 polymorphisms are not associated with hypertension, a rare haplotype of the gene might be protective against the development of hypertension.
Collapse
Affiliation(s)
- Georgios D. Kitsios
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Tufts University School of Medicine, 800 Washington Street, Tufts MC no. 63, Boston, MA 02111, USA
- Department of Biomathematics, University of Thessaly School of Medicine, 2 Panepistimiou, Biopolis, Larissa 41100, Greece
| | - Elias Zintzaras
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Tufts University School of Medicine, 800 Washington Street, Tufts MC no. 63, Boston, MA 02111, USA
- Department of Biomathematics, University of Thessaly School of Medicine, 2 Panepistimiou, Biopolis, Larissa 41100, Greece
| |
Collapse
|