1
|
Armstrong ND, Srinivasasainagendra V, Chekka LMS, Nguyen NHK, Nahid NA, Jones AC, Tanner RM, Hidalgo BA, Limdi NA, Claas SA, Gong Y, McDonough CW, Cooper-DeHoff RM, Johnson JA, Tiwari HK, Arnett DK, Irvin MR. Genetic Contributors of Efficacy and Adverse Metabolic Effects of Chlorthalidone in African Americans from the Genetics of Hypertension Associated Treatments (GenHAT) Study. Genes (Basel) 2022; 13:1260. [PMID: 35886043 PMCID: PMC9319619 DOI: 10.3390/genes13071260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/01/2022] [Accepted: 07/07/2022] [Indexed: 02/05/2023] Open
Abstract
Hypertension is a leading risk factor for cardiovascular disease mortality. African Americans (AAs) have the highest prevalence of hypertension in the United States, and to alleviate the burden of hypertension in this population, better control of blood pressure (BP) is needed. Previous studies have shown considerable interpersonal differences in BP response to antihypertensive treatment, suggesting a genetic component. Utilizing data from 4297 AA participants randomized to chlorthalidone from the Genetics of Hypertension Associated Treatments (GenHAT) study, we aimed to identify variants associated with the efficacy of chlorthalidone. An additional aim was to find variants that contributed to changes in fasting glucose (FG) in these individuals. We performed genome-wide association analyses on the change of systolic and diastolic BP (SBP and DBP) over six months and FG levels over 24 months of treatment. We sought replication in the International Consortia of Pharmacogenomics Studies. We identified eight variants statistically associated with BP response and nine variants associated with FG response. One suggestive LINC02211-CDH9 intergenic variant was marginally replicated with the same direction of effect. Given the impact of hypertension in AAs, this study implies that understanding the genetic background for BP control and glucose changes during chlorthalidone treatment may help prevent adverse cardiovascular events in this population.
Collapse
Affiliation(s)
- Nicole D. Armstrong
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (N.D.A.); (A.C.J.); (R.M.T.); (B.A.H.)
| | - Vinodh Srinivasasainagendra
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (V.S.); (H.K.T.)
| | - Lakshmi Manasa S. Chekka
- Division of Applied Regulatory Sciences, Center for Drug Evaluation and Research, Silver Spring, MD 20903, USA;
| | - Nam H. K. Nguyen
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA; (N.H.K.N.); (N.A.N.); (Y.G.); (C.W.M.); (R.M.C.-D.); (J.A.J.)
| | - Noor A. Nahid
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA; (N.H.K.N.); (N.A.N.); (Y.G.); (C.W.M.); (R.M.C.-D.); (J.A.J.)
| | - Alana C. Jones
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (N.D.A.); (A.C.J.); (R.M.T.); (B.A.H.)
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Rikki M. Tanner
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (N.D.A.); (A.C.J.); (R.M.T.); (B.A.H.)
| | - Bertha A. Hidalgo
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (N.D.A.); (A.C.J.); (R.M.T.); (B.A.H.)
| | - Nita A. Limdi
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Steven A. Claas
- Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY 40506, USA; (S.A.C.); (D.K.A.)
| | - Yan Gong
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA; (N.H.K.N.); (N.A.N.); (Y.G.); (C.W.M.); (R.M.C.-D.); (J.A.J.)
| | - Caitrin W. McDonough
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA; (N.H.K.N.); (N.A.N.); (Y.G.); (C.W.M.); (R.M.C.-D.); (J.A.J.)
| | - Rhonda M. Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA; (N.H.K.N.); (N.A.N.); (Y.G.); (C.W.M.); (R.M.C.-D.); (J.A.J.)
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Julie A. Johnson
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA; (N.H.K.N.); (N.A.N.); (Y.G.); (C.W.M.); (R.M.C.-D.); (J.A.J.)
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Hemant K. Tiwari
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (V.S.); (H.K.T.)
| | - Donna K. Arnett
- Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY 40506, USA; (S.A.C.); (D.K.A.)
- Deans Office, College of Public Health, University of Kentucky, Lexington, KY 40506, USA
| | - Marguerite R. Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (N.D.A.); (A.C.J.); (R.M.T.); (B.A.H.)
| |
Collapse
|
2
|
Protective effect of Xin-Ji-Er-Kang on cardiovascular remodeling in high salt-induced hypertensive mice. Exp Ther Med 2018; 17:1551-1562. [PMID: 30783421 PMCID: PMC6364186 DOI: 10.3892/etm.2018.7105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 11/14/2018] [Indexed: 12/29/2022] Open
Abstract
The aim of the present study was to investigate the effects of Xin-Ji-Er-Kang (XJEK) on high salt-induced hypertensive mice. Mice with high-salt diet-induced hypertension were divided into four groups: Control (standard diet alone for 8 weeks), model (diet containing 8% NaCl for 8 weeks and intragastric administration of distilled water for the last 4 weeks), XJEK + high-salt-treated (diet containing 8% NaCl for 8 weeks and intragastric administration of XJEK for the last 4 weeks) and irbesartan + high-salt-treated (diet containing 8% NaCl for 8 weeks with intragastric administration of irbesartan for the last 4 weeks). The hemodynamic index and cardiac pathological changes in the hypertensive mice were then examined. An aortic ring apparatus was used to detect acetylcholine-dependent endothelium relaxation function. Colorimetric analysis was applied to determine serum nitric oxide (NO), superoxide dismutase activity and malondialdehyde content; ELISA was employed to measure brain natriuretic peptide, serum angiotensin II (Ang II), endothelin-1 content and aldosterone; and immunohistochemistry was used to detect the expression of endothelial nitric oxide synthase (eNOS), interleukin (IL)-1β, IL-10 and tumor necrosis factor (TNF)-α in cardiac tissues. XJEK improved the heart systolic and diastolic function, ameliorated hemodynamic parameters and cardiovascular remodeling indices, blunted the cardiac pathological changes and improved endothelial dysfunction (ED) via boosting eNOS activity, promoting NO bioavailability and decreasing serum Ang II content. Furthermore, treatment with XJEK inhibited the increase of IL-1β and TNF-α expression and the decrease of IL-10 expression in cardiac tissues, and ameliorated oxidative stress status. Therefore, XJEK exerted protective effects against high salt-induced hypertension and cardiovascular remodeling in mice via improving ED, restoring pro- and anti-inflammatory factor balance and decreasing oxidative stress.
Collapse
|
3
|
Polonikov AV, Ponomarenko IV, Bykanova MA, Sirotina SS, Bocharova AV, Vagaytseva KV, Stepanov VA, Azarova IE, Churnosov MI, Solodilova MA. A comprehensive study revealed SNP-SNP interactions and a sex-dependent relationship between polymorphisms of the CYP2J2 gene and hypertension risk. Hypertens Res 2018; 42:257-272. [PMID: 30518987 DOI: 10.1038/s41440-018-0142-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 06/18/2018] [Accepted: 07/12/2018] [Indexed: 11/09/2022]
Abstract
This study investigated whether common polymorphisms of cytochrome P450 2J2 (CYP2J2), a major enzyme that controls the biosynthesis of vasoactive epoxyeicosatrienoic acids, are collectively involved in the molecular basis of essential hypertension (EH). A total of 2314 unrelated Russian subjects from the Kursk (discovery sample: 913 EH patients and 645 controls) and Belgorod (replication sample: 345 EH patients and 411 controls) regions were recruited for this study. Eight single nucleotide polymorphisms (SNPs), including rs890293, rs11572182, rs10493270, rs1155002, rs2280275, rs7515289, rs11572325, and rs10889162, of CYP2J2 were genotyped using the MassARRAY 4 system and TaqMan-based assays. Significant associations were identified among the SNPs rs890293 (OR = 2.17, 95%CI 1.30-3.65), rs2280275 (OR = 1.59, 95%CI 1.10-2.37) and rs11572325 (OR = 1.89, 95%CI 1.22-2.95) and the risk of EH in females from the Kursk population. Sixteen CYP2J2 genotype combinations only showed significant associations with EH risk only in females. A common haplotype, T-T-G-C-C-C-T-A, increased the risk of EH in females. The bioinformatic analysis enabled identification of the SNPs that possess regulatory potential and/or are located within the binding sites for multiple transcription factors that play roles in the pathways involved in hypertension pathogenesis. Moreover, the polymorphisms rs890293, rs2280275, and rs11572325 were found to be significantly associated with hypertension risk in the Belgorod population. In conclusion, the rs2280275 and rs11572325 SNPs of CYP2J2 may be considered novel genetic markers of hypertension, at least in Russian women. However, sex-specific associations between CYP2J2 gene polymorphisms and hypertension require further investigation to clarify the specific genetic and/or environmental factors that are responsible for the increased disease susceptibility of women compared to that of men.
Collapse
Affiliation(s)
- Alexey V Polonikov
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx St., Kursk, 305041, Russian Federation. .,Laboratory of Statistical Genetics and Bioinformatics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk, 305041, Russian Federation.
| | - Irina V Ponomarenko
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx St., Kursk, 305041, Russian Federation
| | - Marina A Bykanova
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk, 305041, Russian Federation
| | - Svetlana S Sirotina
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx St., Kursk, 305041, Russian Federation
| | - Anna V Bocharova
- Evolutionary Genetics Laboratory, Research Institute of Medical Genetics, Tomsk National Medical Research Center, 10 Nabereznaya Ushaiki, Tomsk, 634050, Russian Federation
| | - Kseniya V Vagaytseva
- Evolutionary Genetics Laboratory, Research Institute of Medical Genetics, Tomsk National Medical Research Center, 10 Nabereznaya Ushaiki, Tomsk, 634050, Russian Federation
| | - Vadim A Stepanov
- Evolutionary Genetics Laboratory, Research Institute of Medical Genetics, Tomsk National Medical Research Center, 10 Nabereznaya Ushaiki, Tomsk, 634050, Russian Federation
| | - Iuliia E Azarova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk, 305041, Russian Federation
| | - Mikhail I Churnosov
- Department of Medical Biological Disciplines, Belgorod State University, 85 Pobeda St., Belgorod, 308015, Russian Federation
| | - Maria A Solodilova
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx St., Kursk, 305041, Russian Federation
| |
Collapse
|
4
|
Hart EC. Human hypertension, sympathetic activity and the selfish brain. Exp Physiol 2018; 101:1451-1462. [PMID: 27519960 DOI: 10.1113/ep085775] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/10/2016] [Indexed: 12/19/2022]
Abstract
NEW FINDINGS What is the topic of this review? This review article revisits an historical hypothesis that cerebral hypoperfusion, caused by elevated cerebral vascular resistances, causes the onset of high sympathetic nerve activity and hypertension in humans. What advances does it highlight? The review article highlights new evidence indicating that congenital cerebrovascular abnormalities, namely vertebral artery hypoplasia and an incomplete posterior circle of Willis, may play a role in the onset of hypertension. Despite the harmful consequences of high blood pressure (hypertension; e.g. stroke, renal failure, dementia and even death), the underlying physiological mechanisms that cause the onset of hypertension are poorly understood. The most established finding is that hypertension occurs alongside activation of the sympathetic nervous system, yet exactly what triggers this in humans is ambiguous. This review discusses evidence for elevated sympathetic nerve activity, particularly in human hypertension, and revisits an historical theory regarding the aetiology underlying human hypertension that was proposed by Seymour Kety and John Dickinson in the 1940s-1950s. My research group hypothesizes that elevated sympathetic nerve activity and hypertension develop as a fundamental mechanism to maintain adequate cerebral blood flow, which is now termed Cushing's mechanism or the selfish brain hypothesis. Moreover, it goes against the traditional belief that high cerebrovascular resistance is a consequence of hypertension; we propose that this elevated resistance drives hypertension. This review discusses historical and new evidence in animals and humans supporting this hypothesis. In particular, unique human data indicating a higher prevalence of congenital cerebral vascular abnormalities in hypertension are considered.
Collapse
Affiliation(s)
- Emma C Hart
- School of Physiology, Pharmacology and Neuroscience, Clinical Research and Imaging Centre, University of Bristol, Bristol, UK
| |
Collapse
|
5
|
Cheng X, Waghulde H, Mell B, Morgan EE, Pruett-Miller SM, Joe B. Positional cloning of quantitative trait nucleotides for blood pressure and cardiac QT-interval by targeted CRISPR/Cas9 editing of a novel long non-coding RNA. PLoS Genet 2017; 13:e1006961. [PMID: 28827789 PMCID: PMC5578691 DOI: 10.1371/journal.pgen.1006961] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/31/2017] [Accepted: 08/07/2017] [Indexed: 01/11/2023] Open
Abstract
Multiple GWAS studies have reported strong association of cardiac QT-interval to a region on HSA17. Interestingly, a rat locus homologous to this region is also linked to QT-intervals. The high resolution positional mapping study located the rat QT-interval locus to a <42.5kb region on RNO10. This region contained no variants in protein-coding sequences, but a prominent contiguous 19bp indel polymorphism was noted within a novel predicted long non-coding RNA (lncRNA), which we named as Rffl-lnc1. To assess the candidacy of this novel lncRNA on QT-interval, targeted CRISPR/Cas9 based genome-engineering approaches were applied on the rat strains used to map this locus. Targeted disruption of the rat Rffl-lnc1 locus caused aberrant, short QT-intervals and elevated blood pressure. Further, to specifically examine the significance of the 19bp polymorphism within the Rffl-lnc1 locus, a CRISPR/Cas9 based targeted knock-in rescue model was constructed by inserting the 19bp into the strain which contained the deletion polymorphism. The knock-in alleles successfully rescued the aberrant QT-interval and blood pressure phenotypes. Further studies revealed that the 19bp polymorphism was necessary and sufficient to recapitulate the phenotypic effect of the previously mapped <42.5kb rat locus. To our knowledge, this study is the first demonstration of a combination of both CRISPR/Cas9 based targeted disruption as well as CRISPR/Cas9 based targeted knock-in rescue approaches applied for a mammalian positional cloning study, which defines the quantitative trait nucleotides (QTNs) within a rat long non-coding RNA as being important for the pleiotropic regulation of both cardiac QT-intervals and blood pressure. Diseases of the cardiovascular system such as essential hypertension do not have a clear cause, but are known to run in families. The inheritance patterns of essential hypertension and other cardiac diseases suggest that they are not due to a single defective gene but instead are caused by multiple genetic defects that are inherited together in a patient. This complex inheritance makes it difficult to pinpoint the underlying defects. Here, we describe a panel of genetically-engineered rats, using which we have discovered a novel gene, which does not code for any protein, as a gene required for maintenance of normal blood pressure. Structural defects within this non-coding RNA cause hypertension and cardiac short-QT interval. Further, by performing genome surgery to correct the gene defect, we demonstrate the precise error in nucleotides that was inherited and caused hypertension and cardiac short-QT interval syndrome.
Collapse
Affiliation(s)
- Xi Cheng
- Program in Physiological Genomics, Center for Hypertension and Personalized Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States of America
| | - Harshal Waghulde
- Program in Physiological Genomics, Center for Hypertension and Personalized Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States of America
| | - Blair Mell
- Program in Physiological Genomics, Center for Hypertension and Personalized Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States of America
| | - Eric E. Morgan
- Program in Physiological Genomics, Center for Hypertension and Personalized Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States of America
- Department of Radiology, University of Toledo Medical Center, Toledo, OH, United States of America
| | - Shondra M. Pruett-Miller
- Department of Cell & Molecular Biology, Center for Advanced Genome Engineering, St. Jude Children’s Research Hospital, Memphis, TN, United States of America
| | - Bina Joe
- Program in Physiological Genomics, Center for Hypertension and Personalized Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States of America
- * E-mail:
| |
Collapse
|
6
|
Polonikov A, Bykanova M, Ponomarenko I, Sirotina S, Bocharova A, Vagaytseva K, Stepanov V, Churnosov M, Bushueva O, Solodilova M, Shvetsov Y, Ivanov V. The contribution of CYP2C gene subfamily involved in epoxygenase pathway of arachidonic acids metabolism to hypertension susceptibility in Russian population. Clin Exp Hypertens 2017; 39:306-311. [PMID: 28513222 DOI: 10.1080/10641963.2016.1246562] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Alexey Polonikov
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, Kursk, Russian Federation
- Laboratory of Statistical Genetics and Bioinformatics, Research Institute for Genetic and Molecular Epidemiology, Kursk, Russian Federation
| | - Marina Bykanova
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, Kursk, Russian Federation
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk, Russian Federation
| | - Irina Ponomarenko
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, Kursk, Russian Federation
| | - Svetlana Sirotina
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, Kursk, Russian Federation
| | - Anna Bocharova
- Evolutionary Genetics Laboratory, Research Institute for Medical Genetics, Tomsk, Russian Federation
| | - Kseniya Vagaytseva
- Evolutionary Genetics Laboratory, Research Institute for Medical Genetics, Tomsk, Russian Federation
| | - Vadim Stepanov
- Evolutionary Genetics Laboratory, Research Institute for Medical Genetics, Tomsk, Russian Federation
| | - Mikhail Churnosov
- Department of Medical Biological Disciplines, Belgorod State University, Belgorod, Russian Federation
| | - Olga Bushueva
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, Kursk, Russian Federation
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk, Russian Federation
| | - Maria Solodilova
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, Kursk, Russian Federation
| | - Yaroslav Shvetsov
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, Kursk, Russian Federation
| | - Vladimir Ivanov
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, Kursk, Russian Federation
| |
Collapse
|
7
|
Impact and influence of “omics” technology on hyper tension studies. Int J Cardiol 2017; 228:1022-1034. [DOI: 10.1016/j.ijcard.2016.11.179] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/06/2016] [Indexed: 12/14/2022]
|
8
|
A comprehensive contribution of genes for aryl hydrocarbon receptor signaling pathway to hypertension susceptibility. Pharmacogenet Genomics 2017; 27:57-69. [PMID: 27977510 DOI: 10.1097/fpc.0000000000000261] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
9
|
Li R, Gao X, Liu B, Ge H, Ning L, Zhao J, Liu J. Prospective Cohort Study to Elucidate the Correlation between Occupational Stress and Hypertension Risk in Oil Workers from Kelamayi City in the Xinjiang Uygur Autonomous Region of China. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 14:ijerph14010001. [PMID: 28025517 PMCID: PMC5295252 DOI: 10.3390/ijerph14010001] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 12/14/2016] [Accepted: 12/15/2016] [Indexed: 12/18/2022]
Abstract
The purpose of this study was to assess the major risk factors for hypertension in oil workers, and investigate the effect of occupational stress on the incidence of hypertension after controlling for other risk factors. A prospective cohort approach was used following enrollment of 1354 oil workers. The occupational stress experienced by oil workers was higher than for the general population in China. By the end of the cohort study, 231 new cases of hypertension among the oil workers had been diagnosed. The cumulative incidence of hypertension was 17.06%. There were 44, 112, and 75 workers who developed hypertension in the low, intermediate, and high occupational stress groups, which represented a 12.0%, 15.6%, and 20.3% cumulative incidence, respectively (chi-square value = 9.812, p < 0.01). Multivariate Cox proportional hazard model analysis showed that type of work, cigarette smoking, excess body weight, and obesity were risk factors for hypertension (p < 0.05). After risk factors such as type of work, cigarette smoking, alcohol consumption, and body mass index (BMI) were controlled, the hypertension risk (hazard ratio, HR) in the high occupational stress group was 1.549 (1.072-2.236) compared to the low exposure group, and 2.337 (1.191-4.585) in female subjects. Our study indicated that an increase in occupational stress was associated with an increased risk of hypertension after other factors were adjusted.
Collapse
Affiliation(s)
- Rong Li
- Department of Public Health, Xinjiang Medical University, Urumqi 830011, China.
| | - Xiaoyan Gao
- Department of Public Health, Xinjiang Medical University, Urumqi 830011, China.
| | - Bo Liu
- Department of Public Health, Xinjiang Medical University, Urumqi 830011, China.
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China.
| | - Hua Ge
- Department of Public Health, Xinjiang Medical University, Urumqi 830011, China.
| | - Li Ning
- Department of Public Health, Xinjiang Medical University, Urumqi 830011, China.
| | - Junling Zhao
- Department of Public Health, Xinjiang Medical University, Urumqi 830011, China.
| | - Jiwen Liu
- Department of Public Health, Xinjiang Medical University, Urumqi 830011, China.
| |
Collapse
|
10
|
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: 1.4] [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
|
11
|
Petry CJ, Sanz Marcos N, Pimentel G, Hayes MG, Nodzenski M, Scholtens DM, Hughes IA, Acerini CL, Ong KK, Lowe WL, Dunger DB. Associations Between Fetal Imprinted Genes and Maternal Blood Pressure in Pregnancy. Hypertension 2016; 68:1459-1466. [PMID: 27777362 DOI: 10.1161/hypertensionaha.116.08261] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 08/19/2016] [Accepted: 09/28/2016] [Indexed: 12/31/2022]
Abstract
In addition to maternal genes and environmental exposures, variation in fetal imprinted genes could also affect maternal blood pressure during pregnancy. Our objective was to test the associations between polymorphic variants in 16 imprinted genes and maternal mean arterial blood pressures in 1160 DNA trios from 2 established birth cohorts (the Cambridge Baby Growth and Wellbeing Studies) and seek replication in 1367 Hyperglycemia and Adverse Pregnancy Outcome Study participants. Significant univariate associations, all independent of fetal sex, were observed in the Cambridge cohorts, including FAM99A rs1489945 transmitted from the mother (P=2×10-4), DLK1 rs10139403 (mother; P=9×10-4), DLK1 rs12147008 (mother; P=1×10-3), H19 rs217222 (father; P=1×10-3), SNRPN rs1453556 (father; P=1×10-3), IGF2 rs6356 (father; P=1×10-3), and NNAT rs6066671 (father; P=1×10-3). In meta-analysis including additional independent Hyperglycemia and Adverse Pregnancy Outcome Study data, the association with maternally transmitted fetal DLK1 rs10139403 reached genome-wide significance (P=6.3×10-10). With the exception of fetal rs1489945 and rs217222, all of other associations were unidirectional and most were statistically significant. To further explore the significance of these relationships, we developed an allele score based on the univariate findings. The score was strongly associated with maternal blood pressure at 31 weeks (P=4.1×10-8; adjusted r2=5.6%) and 37 weeks of pregnancy (P=1.1×10-4; r2=3.6%), and during the last 2 weeks before parturition (P=1.1×10-10; r2=8.7%). It was also associated with gestational hypertension (odds ratio, 1.54 [range, 1.14-2.09] per allele; P=0.005; 45 cases and 549 controls). These data support the concept that fetal imprinted genes are related to the development of gestational hypertension.
Collapse
Affiliation(s)
- Clive J Petry
- From the Department of Paediatrics (C.J.P., N.S.M., G.P., I.A.H., C.L.A., K.K.O., D.B.D.), Medical Research Council Epidemiology Unit (K.K.O.), and Institute of Metabolic Science (D.B.D.), University of Cambridge, United Kingdom; Hospital Sant Joan de Déu, Servicio de Pediatría, Barcelona, Spain (N.S.M.); Divisão de Endocrinologia Pediátrica, Departamento de Pediatria, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil (G.P.); and Division of Endocrinology, Metabolism and Molecular Medicine, Department of Medicine (M.G.H., W.L.L.) and Division of Biostatistics, Department of Preventive Medicine (M.N., D.M.S.), Northwestern University Feinberg School of Medicine, Chicago, IL.
| | - Nuria Sanz Marcos
- From the Department of Paediatrics (C.J.P., N.S.M., G.P., I.A.H., C.L.A., K.K.O., D.B.D.), Medical Research Council Epidemiology Unit (K.K.O.), and Institute of Metabolic Science (D.B.D.), University of Cambridge, United Kingdom; Hospital Sant Joan de Déu, Servicio de Pediatría, Barcelona, Spain (N.S.M.); Divisão de Endocrinologia Pediátrica, Departamento de Pediatria, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil (G.P.); and Division of Endocrinology, Metabolism and Molecular Medicine, Department of Medicine (M.G.H., W.L.L.) and Division of Biostatistics, Department of Preventive Medicine (M.N., D.M.S.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Gracielle Pimentel
- From the Department of Paediatrics (C.J.P., N.S.M., G.P., I.A.H., C.L.A., K.K.O., D.B.D.), Medical Research Council Epidemiology Unit (K.K.O.), and Institute of Metabolic Science (D.B.D.), University of Cambridge, United Kingdom; Hospital Sant Joan de Déu, Servicio de Pediatría, Barcelona, Spain (N.S.M.); Divisão de Endocrinologia Pediátrica, Departamento de Pediatria, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil (G.P.); and Division of Endocrinology, Metabolism and Molecular Medicine, Department of Medicine (M.G.H., W.L.L.) and Division of Biostatistics, Department of Preventive Medicine (M.N., D.M.S.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - M Geoffrey Hayes
- From the Department of Paediatrics (C.J.P., N.S.M., G.P., I.A.H., C.L.A., K.K.O., D.B.D.), Medical Research Council Epidemiology Unit (K.K.O.), and Institute of Metabolic Science (D.B.D.), University of Cambridge, United Kingdom; Hospital Sant Joan de Déu, Servicio de Pediatría, Barcelona, Spain (N.S.M.); Divisão de Endocrinologia Pediátrica, Departamento de Pediatria, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil (G.P.); and Division of Endocrinology, Metabolism and Molecular Medicine, Department of Medicine (M.G.H., W.L.L.) and Division of Biostatistics, Department of Preventive Medicine (M.N., D.M.S.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Michael Nodzenski
- From the Department of Paediatrics (C.J.P., N.S.M., G.P., I.A.H., C.L.A., K.K.O., D.B.D.), Medical Research Council Epidemiology Unit (K.K.O.), and Institute of Metabolic Science (D.B.D.), University of Cambridge, United Kingdom; Hospital Sant Joan de Déu, Servicio de Pediatría, Barcelona, Spain (N.S.M.); Divisão de Endocrinologia Pediátrica, Departamento de Pediatria, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil (G.P.); and Division of Endocrinology, Metabolism and Molecular Medicine, Department of Medicine (M.G.H., W.L.L.) and Division of Biostatistics, Department of Preventive Medicine (M.N., D.M.S.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Denise M Scholtens
- From the Department of Paediatrics (C.J.P., N.S.M., G.P., I.A.H., C.L.A., K.K.O., D.B.D.), Medical Research Council Epidemiology Unit (K.K.O.), and Institute of Metabolic Science (D.B.D.), University of Cambridge, United Kingdom; Hospital Sant Joan de Déu, Servicio de Pediatría, Barcelona, Spain (N.S.M.); Divisão de Endocrinologia Pediátrica, Departamento de Pediatria, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil (G.P.); and Division of Endocrinology, Metabolism and Molecular Medicine, Department of Medicine (M.G.H., W.L.L.) and Division of Biostatistics, Department of Preventive Medicine (M.N., D.M.S.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Ieuan A Hughes
- From the Department of Paediatrics (C.J.P., N.S.M., G.P., I.A.H., C.L.A., K.K.O., D.B.D.), Medical Research Council Epidemiology Unit (K.K.O.), and Institute of Metabolic Science (D.B.D.), University of Cambridge, United Kingdom; Hospital Sant Joan de Déu, Servicio de Pediatría, Barcelona, Spain (N.S.M.); Divisão de Endocrinologia Pediátrica, Departamento de Pediatria, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil (G.P.); and Division of Endocrinology, Metabolism and Molecular Medicine, Department of Medicine (M.G.H., W.L.L.) and Division of Biostatistics, Department of Preventive Medicine (M.N., D.M.S.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Carlo L Acerini
- From the Department of Paediatrics (C.J.P., N.S.M., G.P., I.A.H., C.L.A., K.K.O., D.B.D.), Medical Research Council Epidemiology Unit (K.K.O.), and Institute of Metabolic Science (D.B.D.), University of Cambridge, United Kingdom; Hospital Sant Joan de Déu, Servicio de Pediatría, Barcelona, Spain (N.S.M.); Divisão de Endocrinologia Pediátrica, Departamento de Pediatria, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil (G.P.); and Division of Endocrinology, Metabolism and Molecular Medicine, Department of Medicine (M.G.H., W.L.L.) and Division of Biostatistics, Department of Preventive Medicine (M.N., D.M.S.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Ken K Ong
- From the Department of Paediatrics (C.J.P., N.S.M., G.P., I.A.H., C.L.A., K.K.O., D.B.D.), Medical Research Council Epidemiology Unit (K.K.O.), and Institute of Metabolic Science (D.B.D.), University of Cambridge, United Kingdom; Hospital Sant Joan de Déu, Servicio de Pediatría, Barcelona, Spain (N.S.M.); Divisão de Endocrinologia Pediátrica, Departamento de Pediatria, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil (G.P.); and Division of Endocrinology, Metabolism and Molecular Medicine, Department of Medicine (M.G.H., W.L.L.) and Division of Biostatistics, Department of Preventive Medicine (M.N., D.M.S.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - William L Lowe
- From the Department of Paediatrics (C.J.P., N.S.M., G.P., I.A.H., C.L.A., K.K.O., D.B.D.), Medical Research Council Epidemiology Unit (K.K.O.), and Institute of Metabolic Science (D.B.D.), University of Cambridge, United Kingdom; Hospital Sant Joan de Déu, Servicio de Pediatría, Barcelona, Spain (N.S.M.); Divisão de Endocrinologia Pediátrica, Departamento de Pediatria, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil (G.P.); and Division of Endocrinology, Metabolism and Molecular Medicine, Department of Medicine (M.G.H., W.L.L.) and Division of Biostatistics, Department of Preventive Medicine (M.N., D.M.S.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - David B Dunger
- From the Department of Paediatrics (C.J.P., N.S.M., G.P., I.A.H., C.L.A., K.K.O., D.B.D.), Medical Research Council Epidemiology Unit (K.K.O.), and Institute of Metabolic Science (D.B.D.), University of Cambridge, United Kingdom; Hospital Sant Joan de Déu, Servicio de Pediatría, Barcelona, Spain (N.S.M.); Divisão de Endocrinologia Pediátrica, Departamento de Pediatria, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil (G.P.); and Division of Endocrinology, Metabolism and Molecular Medicine, Department of Medicine (M.G.H., W.L.L.) and Division of Biostatistics, Department of Preventive Medicine (M.N., D.M.S.), Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
12
|
Abstract
BACKGROUND New technologies for acquisition of genomic data, while offering unprecedented opportunities for genetic discovery, also impose severe burdens of interpretation and penalties for multiple testing. METHODS The Pathway-based Analyses Group of the Genetic Analysis Workshop 19 (GAW19) sought reduction of multiple-testing burden through various approaches to aggregation of highdimensional data in pathways informed by prior biological knowledge. RESULTS Experimental methods testedincluded the use of "synthetic pathways" (random sets of genes) to estimate power and false-positive error rate of methods applied to simulated data; data reduction via independent components analysis, single-nucleotide polymorphism (SNP)-SNP interaction, and use of gene sets to estimate genetic similarity; and general assessment of the efficacy of prior biological knowledge to reduce the dimensionality of complex genomic data. CONCLUSIONS The work of this group explored several promising approaches to managing high-dimensional data, with the caveat that these methods are necessarily constrained by the quality of external bioinformatic annotation.
Collapse
Affiliation(s)
- Jack W Kent
- Department of Genetics, Texas Biomedical Research Institute, PO Box 760549, San Antonio, TX, 78245-0549, USA.
| |
Collapse
|
13
|
Zheng J, Rao DC, Shi G. An update on genome-wide association studies of hypertension. ACTA ACUST UNITED AC 2015. [DOI: 10.1186/s40535-015-0013-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
14
|
Liu X, Wang W, Chen W, Jiang X, Zhang Y, Wang Z, Yang J, Jones JE, Jose PA, Yang Z. Regulation of blood pressure, oxidative stress and AT1R by high salt diet in mutant human dopamine D5 receptor transgenic mice. Hypertens Res 2015; 38:394-9. [PMID: 25716648 PMCID: PMC6400478 DOI: 10.1038/hr.2015.17] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 01/12/2015] [Accepted: 01/25/2015] [Indexed: 12/20/2022]
Abstract
Humans have dopamine D5 receptors (hD5R) with single-nucleotide polymorphisms and a diminished function. We generated hD5(F173L) cDNA that has a decreased response to D5R agonist-mediated increase in cAMP production and increased production of reactive oxygen species, relative to wild-type hD5R (hD5(WT)) cDNA expressed in Chinese hamster ovary cells. To investigate the role of hD5(F173L) in the pathogenesis of salt-sensitive hypertension, we generated transgenic mice overexpressing hD5(F173L) or hD5(WT) and fed them normal (0.8% NaCl) or high (4% NaCl) salt diet. On normal salt diet, the blood pressure, and renal NADPH oxidase activity and angiotensin type 1 receptor (AT1R) expression were higher in hD5(F173L) than hD5(WT) transgenic mice. After 2 weeks on high salt diet, the blood pressure and renal NADPH oxidase activity, but not AT1R expression, were increased in hD5(F173L) but not in hD5(WT) transgenic mice. Candesartan, an AT1R antagonist, decreased the blood pressure and NADPH oxidase activity in hD5(F173L) but not in hD5(WT) transgenic mice. We suggest that the ability of the hD5R to negatively regulate the renal NADPH oxidase activity and AT1R function may have important implications in the pathogenesis of salt-sensitive blood pressure. However, the mechanisms involved in regulating the balance of renal D5R and AT1R function in the oxidative stress-mediated salt-sensitive blood pressure remain to be determined.
Collapse
Affiliation(s)
- Xing Liu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, People's Republic China
| | - Wenjie Wang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, People's Republic China
| | - Wei Chen
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, People's Republic China
| | - Xiaoliang Jiang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, People's Republic China
| | - Yanrong Zhang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, People's Republic China
| | - Zihao Wang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, People's Republic China
| | - Jian Yang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - John E Jones
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Pedro A Jose
- 1] Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA [2] Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Zhiwei Yang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, People's Republic China
| |
Collapse
|
15
|
Fox CS, Hall JL, Arnett DK, Ashley EA, Delles C, Engler MB, Freeman MW, Johnson JA, Lanfear DE, Liggett SB, Lusis AJ, Loscalzo J, MacRae CA, Musunuru K, Newby LK, O'Donnell CJ, Rich SS, Terzic A. Future translational applications from the contemporary genomics era: a scientific statement from the American Heart Association. Circulation 2015; 131:1715-36. [PMID: 25882488 DOI: 10.1161/cir.0000000000000211] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The field of genetics and genomics has advanced considerably with the achievement of recent milestones encompassing the identification of many loci for cardiovascular disease and variable drug responses. Despite this achievement, a gap exists in the understanding and advancement to meaningful translation that directly affects disease prevention and clinical care. The purpose of this scientific statement is to address the gap between genetic discoveries and their practical application to cardiovascular clinical care. In brief, this scientific statement assesses the current timeline for effective translation of basic discoveries to clinical advances, highlighting past successes. Current discoveries in the area of genetics and genomics are covered next, followed by future expectations, tools, and competencies for achieving the goal of improving clinical care.
Collapse
|
16
|
The potential impact of the fetal genotype on maternal blood pressure during pregnancy. J Hypertens 2015; 32:1553-61; discussion 1561. [PMID: 24842698 DOI: 10.1097/hjh.0000000000000212] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The heritability of pregnancy-induced hypertension (encompassing both gestational hypertension and preeclampsia) is around 0.47, suggesting that there is a genetic component to its development. However, the maternal genetic risk variants discovered so far only account for a small proportion of the heritability. Other genetic variants that may affect maternal blood pressure in pregnancy arise from the fetal genome, for example wild-type pregnant mice carrying offspring with Cdkn1c or Stox1 disrupted develop hypertension and proteinuria. In humans, there is a higher risk for preeclampsia in women carrying fetuses with Beckwith-Wiedemann syndrome (including those fetuses with CDKN1C mutations) and a lower risk for women carrying babies with trisomy 21. Other risk may be associated with imprinted fetal growth genes and genes that are highly expressed in the placenta such as GCM1. This article reviews the current state of knowledge linking the fetal genotype with maternal blood pressure in pregnancy.
Collapse
|
17
|
Basson JJ, de las Fuentes L, Rao DC. Single nucleotide polymorphism-single nucleotide polymorphism interactions among inflammation genes in the genetic architecture of blood pressure in the Framingham Heart Study. Am J Hypertens 2015; 28:248-55. [PMID: 25063733 PMCID: PMC4400411 DOI: 10.1093/ajh/hpu132] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 01/08/2014] [Accepted: 05/25/2014] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Hypertension is a major global health burden, but, although systolic and diastolic blood pressure (BP) each have estimated heritability of at least 30%, <3% of their variance has been attributed to particular genetic variants. Few studies have shown interactions between pairs of single nucleotide polymorphisms (SNPs) to be associated with BP. Although many studies use a Bonferroni correction for multiple testing to control type I error, thereby potentially reducing power, false discovery rate (FDR) approaches are also used in genome-wide studies. Renal ion balance genes have been associated with BP regulation, but, although inflammation has been studied in connection with BP, few studies have reported associations between inflammation genes and BP. METHODS We analyzed SNP-SNP interactions among 31 SNPs from genes involved in renal ion balance and 30 SNPs from genes involved in inflammation using data from the Framingham Heart Study. RESULTS No evidence of association was found for interactions among renal ion balance SNPs for either systolic or diastolic BP. A group of 3 interactions involving 6 inflammation genes (IKBKB-NFKBIA, IKBKE-CHUK, and ADIPOR2-RETN) showed evidence of association with diastolic BP with an FDR of 4.2%; no single interaction reached experiment-wide significance. CONCLUSIONS This study identified promising and biologically plausible candidates for interactions between inflammation genes that may be associated with DBP. Analysis using the FDR may allow detection of signals in the presence of modest noise (false positives) that a stringent approach based on Bonferroni-corrected P value thresholds may miss.
Collapse
Affiliation(s)
- Jacob J Basson
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, USA;
| | - Lisa de las Fuentes
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, USA; Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dabeeru C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
18
|
Yang X, He J, Gu D, Hixson JE, Huang J, Rao DC, Shimmin LC, Chen J, Rice TK, Li J, Schwander K, Kelly TN. Associations of epithelial sodium channel genes with blood pressure changes and hypertension incidence: the GenSalt study. Am J Hypertens 2014; 27:1370-6. [PMID: 24735600 DOI: 10.1093/ajh/hpu060] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND We examined the associations of epithelial sodium channel (ENaC) genes with blood pressure (BP) changes and hypertension incidence in a longitudinal family study. METHODS A total of 2,755 Han Chinese participants of the Genetic Epidemiology Network of Salt Sensitivity (GenSalt) baseline examination were eligible for this study. The associations of 43 tag single nucleotide polymorphisms (SNPs) in ENaC genes with BP changes and hypertension incidence were assessed using mixed models to account for the correlations of repeated measures among individuals and within families. A genotype by time interaction term was used to model differences in longitudinal BP change according to genotype over time. Gene-based analyses were conducted using the truncated product method. The Bonferroni method was used to adjust for multiple testing in all analyses. RESULTS During an average of 7.4 years follow-up, systolic BP (SBP) and diastolic BP (DBP) increased, and approximately 33% of participants developed hypertension. SCNN1A SNP rs11064153 and SCNN1G SNP rs4401050 were significantly associated with longitudinal changes in SBP after adjustment for multiple testing (P interaction = 5.8×10(-4) and 0.001, respectively). Similar but nonsignificant trends were observed for the associations between both rs11064153 and rs4401050 and DBP changes (P interaction = 0.024 and 0.005, respectively) and between rs11604153 and hypertension incidence (P = 0.02). Gene-based analyses also supported the overall association of SCNN1G with longitudinal changes in SBP (P = 2.0×10(-4)). CONCLUSIONS Our findings indicated that SCNN1A and SCNN1G may contribute to BP changes over time in the Han Chinese population. Replication of these findings is warranted.
Collapse
Affiliation(s)
- Xueli Yang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana; 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
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana; Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Dongfeng Gu
- 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;
| | - James E Hixson
- Department of Epidemiology, Human Genetics and Environmental Sciences, University of Texas School of Public Health, Houston, Texas
| | - Jianfeng Huang
- 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
| | - Dabeeru C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Lawrence C Shimmin
- Department of Epidemiology, Human Genetics and Environmental Sciences, University of Texas School of Public Health, Houston, Texas
| | - Jichun 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
| | - Treva K Rice
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Jianxin Li
- 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
| | - Karen Schwander
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Tanika N Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| |
Collapse
|
19
|
An update on the pharmacogenetics of treating hypertension. J Hum Hypertens 2014; 29:283-91. [PMID: 25355012 DOI: 10.1038/jhh.2014.76] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 06/24/2014] [Accepted: 07/10/2014] [Indexed: 02/08/2023]
Abstract
Hypertension is a leading cause of cardiovascular mortality, but only one third of patients achieve blood pressure goals despite antihypertensive therapy. Genetic polymorphisms may partially account for the interindividual variability and abnormal response to antihypertensive drugs. Candidate gene and genome-wide approaches have identified common genetic variants associated with response to antihypertensive drugs. However, there is no currently available pharmacogenetic test to guide hypertension treatment in clinical practice. In this review, we aimed to summarize the recent findings on pharmacogenetics of the most commonly used antihypertensive drugs in clinical practice, including diuretics, angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers, beta-blockers and calcium channel blockers. Notably, only a small percentage of the genetic variability on response to antihypertensive drugs has been explained, and the vast majority of the genetic variants associated with antihypertensives efficacy and toxicity remains to be identified. Despite some genetic variants with evidence of association with the variable response related to these most commonly used antihypertensive drug classes, further replication is needed to confirm these associations in different populations. Further studies on epigenetics and regulatory pathways involved in the responsiveness to antihypertensive drugs might provide a deeper understanding of the physiology of hypertension, which may favor the identification of new targets for hypertension treatment and genetic predictors of antihypertensive response.
Collapse
|
20
|
Gudo B, Nussberger J, Bohlender J. [Variability of plasma angiotensinogen levels and risk of hypertension in a transgenic rat model]. Ann Cardiol Angeiol (Paris) 2014; 63:124-127. [PMID: 24836939 DOI: 10.1016/j.ancard.2014.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 04/15/2014] [Indexed: 06/03/2023]
Abstract
AIM Genetic polymorphisms of the human angiotensinogen gene are frequent and may induce up to 30% increase of plasma angiotensinogen concentrations with a blood pressure increase of up to 5mmHg. Their role for the pathogenesis of human arterial hypertension remains unclear. High plasma angiotensinogen levels could increase the sensitivity to other blood pressure stressors. METHODS Male transgenic rats with a 9-fold increase of plasma angiotensinogen concentrations and male non-transgenic rats aged 10 weeks were treated or not with NG-Nitro-L-arginine-methyl ester for 3 weeks in their drinking water (n=3/group). Systolic blood pressure and body weight were measured at baseline and at the end of the study when left ventricular weight and ventricular expression of angiotensin I-converting enzyme and procollagen Iα1 were determined (polymerase chain reaction). RESULTS At baseline, transgenic rats had +18mmHg higher bood pressure and -8% lower body weight compared to non-transgenic rats (P<0.05) without significant changes for the vehicle groups throughout the study (P>0.05). NG-Nitro-L-arginine-methyl ester increased blood pressure, left ventricular weight and left ventricular weight indexed for body weight by +41%, +17.6% and +18.6% (P<0.05) in transgenic and +25%, +5.3% and +6.7% (P>0.05) in non-transgenic rats compared to untreated animals, respectively. Cardiac gene expression showed no differences between groups (P>0.05). CONCLUSION Increased plasma angiotensinogen levels may sensitize to additional blood pressure stressors. Our preliminary results point towards an independent role of angiotensinogen in the pathogenesis of human hypertension and associated end-organ damage.
Collapse
Affiliation(s)
- B Gudo
- Faculté de médecine, université Otto-von-Guericke, Leipziger Str. 44, 39120 Magdebourg, Allemagne
| | - J Nussberger
- Département de médecine, centre hospitalier universitaire vaudois (CHUV), avenue Pierre-Decker 5, 1011 Lausanne, Suisse
| | - J Bohlender
- Faculté de médecine, université Otto-von-Guericke, Leipziger Str. 44, 39120 Magdebourg, Allemagne; KMG Klinikum Havelberg, Domherrnstr. 10, 39350 Havelberg, Allemagne; Institut de biologie cellulaire, université de Berne, Baltzerstrasse 4, 3012 Berne, Suisse.
| |
Collapse
|
21
|
Natekar A, Olds RL, Lau MW, Min K, Imoto K, Slavin TP. Elevated blood pressure: Our family's fault? The genetics of essential hypertension. World J Cardiol 2014; 6:327-37. [PMID: 24944762 PMCID: PMC4062117 DOI: 10.4330/wjc.v6.i5.327] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 02/10/2014] [Accepted: 04/16/2014] [Indexed: 02/06/2023] Open
Abstract
AIM To provide an updated review on current genetic aspects possibly affecting essential hypertension (EH), and to further elucidate their role in EH. METHODS We searched for genetic and epigenetic factors in major studies associated with EH between Jan 2008-Oct 2013 using PubMed. We limited our search to reviews that discussed mostly human studies, and were accessible through the university online resource. We found 11 genome wide association studies (GWAS), as well as five methylation and three miRNA studies that fit our search criteria. A distinction was not made between genes with protective effects or negative effects, as this article is only meant to be a summary of genes associated with any aspect of EH. RESULTS We found 130 genes from the studies that met our inclusion/exclusion criteria. Of note, genes with multiple study references include: STK39, CYP17A1, MTHFR-NPPA, MTHFR-NPPB, ATP2B1, CSK, ZNF652, UMOD, CACNB2, PLEKHA7, SH2B3, TBX3-TBX5, ULK4, CSK-ULK3, CYP1A2, NT5C2, CYP171A, PLCD3, SH2B3, ATXN2, CACNB2, PLEKHA7, SH2B3, TBX3-TBX5, ULK4, and HFE. The following genes overlapped between the genetic studies and epigenetic studies: WNK4 and BDKRB2. Several of the identified genes were found to have functions associated with EH. Many epigenetic factors were also correlated with EH. Of the epigenetic factors, there were no articles discussing siRNA and its effects on EH that met the search criteria, thus the topic was not included in this review. Among the miRNA targets found to be associated with EH, many of the genes involved were also identified in the GWAS studies. CONCLUSION Genetic hypertension risk algorithms could be developed in the future but may be of limited benefit due to the multi-factorial nature of EH. With emerging technologies, like next-generation sequencing, more direct causal relationships between genetic and epigenetic factors affecting EH will likely be discovered creating a tremendous potential for personalized medicine using pharmacogenomics.
Collapse
Affiliation(s)
- Aniket Natekar
- Aniket Natekar, Randi L Olds, Meghann W Lau, Kathleen Min, Karra Imoto, Thomas P Slavin, The John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, United States
| | - Randi L Olds
- Aniket Natekar, Randi L Olds, Meghann W Lau, Kathleen Min, Karra Imoto, Thomas P Slavin, The John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, United States
| | - Meghann W Lau
- Aniket Natekar, Randi L Olds, Meghann W Lau, Kathleen Min, Karra Imoto, Thomas P Slavin, The John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, United States
| | - Kathleen Min
- Aniket Natekar, Randi L Olds, Meghann W Lau, Kathleen Min, Karra Imoto, Thomas P Slavin, The John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, United States
| | - Karra Imoto
- Aniket Natekar, Randi L Olds, Meghann W Lau, Kathleen Min, Karra Imoto, Thomas P Slavin, The John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, United States
| | - Thomas P Slavin
- Aniket Natekar, Randi L Olds, Meghann W Lau, Kathleen Min, Karra Imoto, Thomas P Slavin, The John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, United States
| |
Collapse
|
22
|
Montasser ME, Shimmin LC, Gu D, Chen J, Gu C, Kelly TN, Jaquish CE, Rice TK, Rao DC, Cao J, Chen J, Liu DP, Whelton PK, Hamm LL, He J, Hixson JE. Variation in genes that regulate blood pressure are associated with glomerular filtration rate in Chinese. PLoS One 2014; 9:e92468. [PMID: 24658007 PMCID: PMC3962404 DOI: 10.1371/journal.pone.0092468] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 02/21/2014] [Indexed: 01/13/2023] Open
Abstract
Chronic kidney disease (CKD) can be a consequence of diabetes, hypertension, immunologic disorders, and other exposures, as well as genetic factors that are still largely unknown. Glomerular filtration rate (GFR), which is widely used to measure kidney function, has a heritability ranging from 25% to 75%, but only 1.5% of this heritability is explained by genetic loci that have been identified to date. In this study we tested for associations between GFR and 234 SNPs in 26 genes from pathways of blood pressure regulation in 3,025 rural Chinese participants of the "Genetic Epidemiology Network of Salt Sensitivity" (GenSalt) study. We estimated GFR (eGFR) using baseline serum creatinine measurements obtained prior to dietary intervention. We identified significant associations between eGFR and 12 SNPs in 6 genes (ACE, ADD1, AGT, GRK4, HSD11B1, and SCNN1G). The cumulative effect of the protective alleles was an increase in mean eGFR of 4 mL/min per 1.73 m2, while the cumulative effect of the risk alleles was a decrease in mean eGFR of 3 mL/min per 1.73 m2. In addition, we identified a significant interaction between SNPs in CYP11B1 and ADRB2. We have identified common variants in genes from pathways that regulate blood pressure and influence kidney function as measured by eGFR, providing new insights into the genetic determinants of kidney function. Complex genetic effects on kidney function likely involve interactions among genes as we observed for CYP11B1 and ADRB2.
Collapse
Affiliation(s)
- May E. Montasser
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- * E-mail:
| | - Lawrence C. Shimmin
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Dongfeng Gu
- Cardiovascular Institute and Fu Wai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Chen
- Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Charles Gu
- Washington University in School of Medicine, St. Louis, Missouri, United States of America
| | - Tanika N. Kelly
- Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Cashell E. Jaquish
- National Heart, Lung and Blood Institute, National Institute of Health, Bethesda, Maryland, United States of America
| | - Treva K. Rice
- Washington University in School of Medicine, St. Louis, Missouri, United States of America
| | - Dabeeru C. Rao
- Washington University in School of Medicine, St. Louis, Missouri, United States of America
| | - Jie Cao
- Cardiovascular Institute and Fu Wai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Jichun Chen
- Cardiovascular Institute and Fu Wai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - De-Pei Liu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Paul K. Whelton
- Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Lotuce Lee Hamm
- Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Jiang He
- Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - James E. Hixson
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| |
Collapse
|
23
|
Ji L, Cai X, Zhang L, Fei L, Wang L, Su J, Lazar L, Xu J, Zhang Y. Association between polymorphisms in the renin-angiotensin-aldosterone system genes and essential hypertension in the Han Chinese population. PLoS One 2013; 8:e72701. [PMID: 24015270 PMCID: PMC3756014 DOI: 10.1371/journal.pone.0072701] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 07/12/2013] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Renin-angiotensin-aldosterone system (RAAS) is the most important endocrine blood pressure control mechanism in our body, genes encoding components of this system have been strong candidates for the investigation of the genetic basis of hypertension. However, previous studies mainly focused on limited polymorphisms, thus we carried out a case-control study in the Han Chinese population to systemically investigate the association between polymorphisms in the RAAS genes and essential hypertension. METHODS 905 essential hypertensive cases and 905 normotensive controls were recruited based on stringent inclusion and exclusion criteria. All 41 tagSNPs within RAAS genes were retrieved from HapMap, and the genotyping was performed using the GenomeLab SNPstream Genotyping System. Logistic regression analysis, Multifactor dimensionality reduction (MDR), stratified analysis and crossover analysis were used to identify and characterize interactions among the SNPs and the non-genetic factors. RESULTS Serum levels of total cholesterol (TC) and triglyceride (TG), and body mass index (BMI) were significantly higher in the hypertensive group than in the control group. Of 41 SNPs genotyped, rs3789678 and rs2493132 within AGT, rs4305 within ACE, rs275645 within AGTR1, rs3802230 and rs10086846 within CYP11B2 were shown to associate with hypertension. The MDR analysis demonstrated that the interaction between BMI and rs4305 increased the susceptibility to hypertension. Crossover analysis and stratified analysis further indicated that BMI has a major effect, and rs4305 has a minor effect. CONCLUSION These novel findings indicated that together with non-genetic factors, these genetic variants in the RAAS may play an important role in determining an individual's susceptibility to hypertension in the Han Chinese.
Collapse
Affiliation(s)
- Lindan Ji
- Department of Biochemistry, School of Medicine, Ningbo University, Ningbo, China
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Xiaobo Cai
- Department of Biochemistry, School of Medicine, Ningbo University, Ningbo, China
| | - Lina Zhang
- Department of Preventive Medicine, School of Medicine, Ningbo University, Ningbo, China
| | - Lijuan Fei
- Department of Preventive Medicine, School of Medicine, Ningbo University, Ningbo, China
| | - Lin Wang
- Department of Pathology, School of Medicine, Ningbo University, Ningbo, China
| | - Jia Su
- Department of Cardiology, The Affiliated Ningbo No.1 Hospital, School of Medicine, Ningbo University, Ningbo, China
| | - Lissy Lazar
- Department of Preventive Medicine, School of Medicine, Ningbo University, Ningbo, China
| | - Jin Xu
- Department of Preventive Medicine, School of Medicine, Ningbo University, Ningbo, China
- * E-mail: (JX); (YZ)
| | - Yaping Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- * E-mail: (JX); (YZ)
| |
Collapse
|
24
|
Taylor JY, Kraja AT, de las Fuentes L, Stanfill AG, Clark A, Cashion A. An overview of the genomics of metabolic syndrome. J Nurs Scholarsh 2013; 45:52-9. [PMID: 23368731 PMCID: PMC3594572 DOI: 10.1111/j.1547-5069.2012.01484.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE This article provides a brief overview of the diagnostic criteria and genomic risk factors for the components of metabolic syndrome (MetS). ORGANIZING CONSTRUCTS Contributions of cardiovascular, obesity, and diabetes genomic risk factors to the development of MetS as reported in the literature have been reviewed. FINDINGS The genomic risk factors for the development of MetS are strongly linked to the genomic risk factors that make up the components of the disease. Many of the cardiovascular and renal genomic risk factors for MetS development are similar to those found in the development of hypertension and dyslipidemia. Obesity may act as a master trigger to turn on the gene expression changes necessary for the other components of the disease. Studies in the genomics of type 2 diabetes show a number of overlapping genes and polymorphisms that influence both the development of diabetes and MetS. CONCLUSIONS Although health practitioners now have some insights into the genomics of risk factors associated with MetS, the overall understanding of MetS remains inadequate. Clinical applications based on some of the discussed genomic risk factors are being developed but are not yet available for the diagnosis and treatment of MetS. CLINICAL RELEVANCE A broad knowledge of the genomic contributions to disease processes will enable the clinician to better utilize genomics to assess and tailor management of patients.
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Modern molecular techniques are identifying pathways and genes involved in the pathogenesis of the complex disorder essential hypertension. This review provides an overview of genetic methodologies and recent results in the study of high blood pressure (BP), hypertension-attributed nephropathy, and related intermediate phenotypes. RECENT FINDINGS Candidate gene studies have implicated aberrations in ion channels, ion channel regulation, aldosterone signaling, vasoconstriction and inflammation in essential hypertension; genome-wide association studies (GWAS) have detected more than 50 BP loci, most previously unsuspected in essential hypertension. Mapping by admixture linkage disequilibrium (MALD; or admixture mapping) recently led to a major breakthrough in hypertension-attributed kidney disease in African Americans, demonstrating the role of the apolipoprotein L1 (APOL1) and nonmuscle myosin heavy chain 9 (MYH9) genes in this primary kidney disease residing in the spectrum of focal segmental glomerulosclerosis. GWAS have detected associations between kidney function and UMOD and SHROOM3. SUMMARY Genetic studies confirm that 'essential hypertension' consists of disparate mechanisms that ultimately lead to elevations in systemic BP. The cause of hypertension in the majority of cases remains unknown. It is anticipated that epigenetic phenomena, rare exonic mutations, and interactions with environmental factors make additional contributions.
Collapse
|
26
|
Blackett PR, Sanghera DK. Genetic determinants of cardiometabolic risk: a proposed model for phenotype association and interaction. J Clin Lipidol 2013; 7:65-81. [PMID: 23351585 PMCID: PMC3559023 DOI: 10.1016/j.jacl.2012.04.079] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 03/16/2012] [Accepted: 04/16/2012] [Indexed: 12/15/2022]
Abstract
This review provides a translational and unifying summary of metabolic syndrome genetics and highlights evidence that genetic studies are starting to unravel and untangle origins of the complex and challenging cluster of disease phenotypes. The associated genes effectively express in the brain, liver, kidney, arterial endothelium, adipocytes, myocytes, and β cells. Progression of syndrome traits has been associated with ectopic lipid accumulation in the arterial wall, visceral adipocytes, myocytes, and liver. Thus, it follows that the genetics of dyslipidemia, obesity, and nonalcoholic fatty liver disease are central in triggering progression of the syndrome to overt expression of disease traits and have become a key focus of interest for early detection and for designing prevention and treatments. To support the "birds' eye view" approach, we provide a road-map depicting commonality and interrelationships between the traits and their genetic and environmental determinants based on known risk factors, metabolic pathways, pharmacologic targets, treatment responses, gene networks, pleiotropy, and association with circadian rhythm. Although only a small portion of the known heritability is accounted for and there is insufficient support for clinical application of gene-based prediction models, there is direction and encouraging progress in a rapidly moving field that is beginning to show clinical relevance.
Collapse
Affiliation(s)
- Piers R Blackett
- Department of Pediatrics, 940 NE 13St., University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Dharambir K Sanghera
- Department of Pediatrics, 940 NE 13St., University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| |
Collapse
|
27
|
Searching for genes involved in hypertension development in special populations: children and pre-eclamptic women. Where are we standing now? Clin Chem Lab Med 2013; 51:2253-69. [DOI: 10.1515/cclm-2013-0405] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 07/23/2013] [Indexed: 01/02/2023]
|
28
|
Muñoz Contreras AM, Bedoya Berrío G, Velásquez R CM. An approach to the etiology of metabolic syndrome. Colomb Med (Cali) 2013; 44:57-63. [PMID: 24892324 PMCID: PMC4002015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Revised: 07/21/2012] [Accepted: 11/29/2012] [Indexed: 11/24/2022] Open
Abstract
Increased prevalence of obesity in the world, especially accumulation of abnormal amounts of visceral fat predisposes to insulin resistance, which is the central role of metabolic syndrome (MS). Obesity can deregulate the intracellular signaling of insulin due to the production of inflammatory substances, chemoattractant proteins, adipokines and molecules that trigger hormonal mediator potentials for destabilization of signal transduction, leading to metabolic disorders such as hyperglycemia, hypertension, and dyslipidemia. The complexity of the MS and of the genetic mechanisms involved in its etiology derives from the combination of variants on genes involved and environmental factors that predispose it. The purpose of this paper is to review the effects of obesity in molecular and biochemical responses that trigger insulin resistance and its relation to some candidate genes and the ancestral component of the population.
Collapse
Affiliation(s)
| | - Gabriel Bedoya Berrío
- Faculty of Exact and Natural Sciences, Group of Molecular Genetics -GENMOL. Universidad de Antioquia, E-mail:
| | - Claudia M Velásquez R
- School of Nutrition and Diet, Research Group on Food and Human Nutrition. Universidad de Antioquia, E-mail:
| |
Collapse
|
29
|
Rose AM, Bell LCK. Epistasis and immunity: the role of genetic interactions in autoimmune diseases. Immunology 2012; 137:131-8. [PMID: 22804709 DOI: 10.1111/j.1365-2567.2012.03623.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Autoimmune disorders are a complex and varied group of diseases that are caused by breakdown of self-tolerance. The aetiology of autoimmunity is multi-factorial, with both environmental triggers and genetically determined risk factors. In recent years, it has been increasingly recognized that genetic risk factors do not act in isolation, but rather the combination of individual additive effects, gene-gene interactions and gene-environment interactions determine overall risk of autoimmunity. The importance of gene-gene interactions, or epistasis, has been recently brought into focus, with research demonstrating that many autoimmune diseases, including rheumatic arthritis, autoimmune glomerulonephritis, systemic lupus erythematosus and multiple sclerosis, are influenced by epistatic interactions. This review sets out to examine the basic mechanisms of epistasis, how epistasis influences the immune system and the role of epistasis in two major autoimmune conditions, systemic lupus erythematosus and multiple sclerosis.
Collapse
Affiliation(s)
- Anna M Rose
- Department of Genetics, UCL Institute of Ophthalmology, London, UK.
| | | |
Collapse
|