1
|
Jan A, Alanzi AR, Mothana RA, Kaimori JY, Ali SS, Muhammad T, Saeed M, Akbar R, Khan M. Pharmacogenomic Study of Selected Genes Affecting Amlodipine Blood Pressure Response in Patients with Hypertension. Pharmgenomics Pers Med 2024; 17:473-486. [PMID: 39492848 PMCID: PMC11531276 DOI: 10.2147/pgpm.s481068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024] Open
Abstract
Introduction Despite the availability of various antihypertensive medications, the response to these medications varies among individuals. Understanding how individual genetic variations affect drugs treatment outcomes is a key area of focus in precision medicine. This study investigated the correlation between single nucleotide polymorphisms (SNPs) in selected genes (CACNA1C, CACNA1D, ABCB1, ACE, ADBR2, and NOS1AP) and the blood pressure (BP) control by amlodipine. Methods Four hundred individuals of Pashtun ethnicity undergoing amlodipine treatment for hypertension were included in the present study and divided into the controlled (BP less than 140/90 mmHg) and uncontrolled (BP greater than 140/90 mmHg) hypertension groups. Blood samples (3 mL) were collected from each participant, and DNA was extracted using the Kit method. Ten SNPs in amlodipine pharmacogenes were selected and genotyped using real-time PCR with the TaqMan® system. Logistic regression model was used to determine the association between SNPs and the amlodipine BP response. Results Notable association were observed between SNP rs2239050/CACNA1C and amlodipine blood pressure response, with GG genotype carriers demonstrating a better response (P=0.004) than individuals carrying CC or CG genotypes. SNP rs312481/CACNA1D also exhibited a positive pharmacogenetic association, Individuals with the GG genotype showing a considerable reduction in BP (P=0.021) compared to participants with AA or GA genotypes. In case of SNP rs429/ACE individuals carrying TA genotype were less likely to achieve BP control (P=0.002) than AA genotype carriers. Conclusion Our finding suggests that the SNPs rs2239050/CACNA1C, rs312481/CACNA1D and rs429/ACE influence amlodipine blood pressure response in patients with hypertension. It is recommended that prior knowledge of amlodipine associated pharmacogenetic variants is important that could improve its treatment outcomes in hypertensive patients.
Collapse
Affiliation(s)
- Asif Jan
- Department of Pharmacy, University of Peshawar, Peshawar, 25000, Pakistan
- District Headquarter Hospital (DHQH) Charsadda, Charsadda, 24430, Pakistan
| | - Abdullah R Alanzi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, 1151, Saudi Arabia
| | - Ramzi A Mothana
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, 1151, Saudi Arabia
| | - Jun-Ya Kaimori
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Syed Shaukat Ali
- Department of Pharmacy, University of Malakand, Malakand, Pakistan
| | - Tahir Muhammad
- Molecular Neuropsychiatry & Development (Mind) Lab, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, 43964, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, 43964, Canada
| | - Muhammad Saeed
- Department of Pharmacy, Qurtaba University of Science and Technology, Peshawar, 25000, Pakistan
| | - Rani Akbar
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan
| | - Mehtab Khan
- Department of Biology, Faculty of Science, University of Moncton, Moncton, NB, E1A 3E9, Canada
| |
Collapse
|
2
|
da Cunha Agostini L, de Paula W, Melo AS, Silva NNT, Faria Lopes AC, de Almeida Belo V, Coura-Vital W, de Medeiros Teixeira LF, Lima AA, da Silva GN. Single nucleotide polymorphism (SNP) rs4291 of the angiotensin-converting enzyme (ACE) gene is associated with the response to losartan treatment in hypertensive patients. Mol Biol Rep 2024; 51:458. [PMID: 38551694 DOI: 10.1007/s11033-024-09437-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/11/2024] [Indexed: 04/02/2024]
Abstract
Arterial hypertension is characterized by systolic pressure ≥ 140 mmHg and/or diastolic pressure ≥ 90 mmHg and its treatment consists of the use of antihypertensive drugs, as losartan and hydrochlorothiazide. Blood pressure is regulated by angiotensin-converting enzyme (ACE) and polymorphisms in the ACE gene are associated to a greater predisposition to hypertension and response to treatment. The aim of this study was to evaluate the association of genetic polymorphisms of ACE rs4363, rs4291 and rs4335 and the response to antihypertensive drugs in hypertensive patients from Ouro Preto/MG, Brazil. A case-control study was carried out with 87 hypertensive patients being treated with losartan and 75 with hydrochlorothiazide, who answered a questionnaire and had blood samples collected. Biochemical analyzes were performed on serum using UV/Vis spectrophotometry and identification of ACE variants rs4363, rs4291 and rs4335 was performed by real-time PCR using the TaqMan® system. Univariate logistic regression test was performed to compare categorical data in STATA 13.0 software. The results showed that there was an influence of ACE polymorphisms on the response to losartan, demonstrating that AT or TT genotypes of rs4291 were more frequent in the group of controlled AH (54.9%), indicating that these individuals are 2.8 times more likely to of being controlled AH (95% CI 1.12-6.80, p. =0.026) compared to those with AA genotype. In contrast, no influence of ACE polymorphisms on the response to hydrochlorothiazide was observed. In conclusion, the presence of the T allele of the rs4291 variant was associated to controled blood pressure when losartan was used as an antihypertensive agent. These results show the importance of pharmacogenetic studies to detect genetic characteristics, enabling therapeutic individuality and reducing costs for the healthcare system.
Collapse
Affiliation(s)
- Lívia da Cunha Agostini
- Programa de Pós-Graduação em Ciências Farmacêuticas (CiPharma), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Waléria de Paula
- Programa de Pós-Graduação em Ciências Farmacêuticas (CiPharma), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - André Sacramento Melo
- Departamento de Análises Clínicas (DEACL), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | | | - Ana Cláudia Faria Lopes
- Departamento de Análises Clínicas (DEACL), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Vanessa de Almeida Belo
- Programa de Pós-Graduação em Ciências Farmacêuticas (CiPharma), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
- Departamento de Farmácia (DEFAR), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Wendel Coura-Vital
- Programa de Pós-Graduação em Ciências Farmacêuticas (CiPharma), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
- Departamento de Análises Clínicas (DEACL), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | | | - Angélica Alves Lima
- Programa de Pós-Graduação em Ciências Farmacêuticas (CiPharma), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
- Departamento de Análises Clínicas (DEACL), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Glenda Nicioli da Silva
- Programa de Pós-Graduação em Ciências Farmacêuticas (CiPharma), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Brazil.
- Departamento de Análises Clínicas (DEACL), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Brazil.
- Departamento de Análises Clínicas (DEACL), Escola de Farmácia, Universidade Federal de Ouro Preto, Morro do Cruzeiro, s/n, Ouro Preto, MG, CEP 35402-163, Brazil.
| |
Collapse
|
3
|
Dua P, Seth S, Prasher B, Mukerji M, Maulik SK, Reeta KH. Pharmacogenomic biomarkers in coronary artery disease: a narrative review. Biomark Med 2024; 18:191-202. [PMID: 38456296 DOI: 10.2217/bmm-2023-0476] [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] [Indexed: 03/09/2024] Open
Abstract
Coronary artery disease (CAD) has a high mortality rate. Despite various therapeutic targets, non-responsiveness to drugs remains a prevalent issue. Pharmacogenomics assesses the way an individual's genetic attributes affect their likely response to drug therapy. Single-nucleotide polymorphisms play a crucial role in determining these outcomes. This review offers an overview of single-nucleotide polymorphisms investigated in clinical studies and their associations with drug response/nonresponse in the treatment of CAD. A total of 104 studies of whole sets of chromosomes and several genes were explored. A total of 161 polymorphisms exhibited associations with drug response/nonresponse in CAD across diverse ethnic populations. This pool can serve as a pharmacogenomic biomarker for predicting response to drug therapy in patients with CAD.
Collapse
Affiliation(s)
- Pamila Dua
- All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Sandeep Seth
- All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | | | - Mitali Mukerji
- Indian Institute of Technology, Jodhpur, Rajasthan, India
| | | | - K H Reeta
- All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| |
Collapse
|
4
|
Chekanova V, Vaucher J, Marques-Vidal P. No association between genetic markers and hypertension control in multiple cross-sectional studies. Sci Rep 2023; 13:11811. [PMID: 37479854 PMCID: PMC10362004 DOI: 10.1038/s41598-023-39103-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 07/20/2023] [Indexed: 07/23/2023] Open
Abstract
We aimed to assess whether genetic markers are associated with hypertension control using two cross-sectional surveys conducted in Lausanne, Switzerland. Management of hypertension was assessed as per ESC guidelines using the 140/90 or the 130/80 mm Hg thresholds. One genetic risk score (GRS) for hypertension (18 SNPs) and 133 individual SNPs related to response to specific antihypertensive drugs were tested. We included 1073 (first) and 1157 (second survey) participants treated for hypertension. The prevalence of controlled participants using the 140/90 threshold was 58.8% and 63.6% in the first and second follow-up, respectively. On multivariable analysis, only older age was consistently and negatively associated with hypertension control. No consistent associations were found between GRS and hypertension control (140/90 threshold) for both surveys: Odds ratio and (95% confidence interval) for the highest vs. the lowest quartile of the GRS: 1.06 (0.71-1.58) p = 0.788, and 1.11 (0.71-1.72) p = 0.657, in the first and second survey, respectively. Similar findings were obtained using the 130/80 threshold: 1.23 (0.79-1.90) p = 0.360 and 1.09 (0.69-1.73) p = 0.717, in the first and second survey, respectively. No association between individual SNPs and hypertension control was found. We conclude that control of hypertension is poor in Switzerland. No association between GRS or SNPs and hypertension control was found.
Collapse
Affiliation(s)
| | - Julien Vaucher
- Department of Medicine, Internal Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, Office BH10-642, Rue du Bugnon 46, 1011, Lausanne, Switzerland
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, Office BH10-642, Rue du Bugnon 46, 1011, Lausanne, Switzerland.
| |
Collapse
|
5
|
Ganekal P, Vastrad B, Kavatagimath S, Vastrad C, Kotrashetti S. Bioinformatics and Next-Generation Data Analysis for Identification of Genes and Molecular Pathways Involved in Subjects with Diabetes and Obesity. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59020309. [PMID: 36837510 PMCID: PMC9967176 DOI: 10.3390/medicina59020309] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/19/2023] [Accepted: 01/29/2023] [Indexed: 02/10/2023]
Abstract
Background and Objectives: A subject with diabetes and obesity is a class of the metabolic disorder. The current investigation aimed to elucidate the potential biomarker and prognostic targets in subjects with diabetes and obesity. Materials and Methods: The next-generation sequencing (NGS) data of GSE132831 was downloaded from Gene Expression Omnibus (GEO) database. Functional enrichment analysis of DEGs was conducted with ToppGene. The protein-protein interactions network, module analysis, target gene-miRNA regulatory network and target gene-TF regulatory network were constructed and analyzed. Furthermore, hub genes were validated by receiver operating characteristic (ROC) analysis. A total of 872 DEGs, including 439 up-regulated genes and 433 down-regulated genes were observed. Results: Second, functional enrichment analysis showed that these DEGs are mainly involved in the axon guidance, neutrophil degranulation, plasma membrane bounded cell projection organization and cell activation. The top ten hub genes (MYH9, FLNA, DCTN1, CLTC, ERBB2, TCF4, VIM, LRRK2, IFI16 and CAV1) could be utilized as potential diagnostic indicators for subjects with diabetes and obesity. The hub genes were validated in subjects with diabetes and obesity. Conclusion: This investigation found effective and reliable molecular biomarkers for diagnosis and prognosis by integrated bioinformatics analysis, suggesting new and key therapeutic targets for subjects with diabetes and obesity.
Collapse
Affiliation(s)
- Prashanth Ganekal
- Department of General Medicine, Basaveshwara Medical College, Chitradurga 577501, Karnataka, India
| | - Basavaraj Vastrad
- Department of Pharmaceutical Chemistry, K.L.E. College of Pharmacy, Gadag 582101, Karnataka, India
| | - Satish Kavatagimath
- Department of Pharmacognosy, K.L.E. College of Pharmacy, Belagavi 590010, Karnataka, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karnataka, India
- Correspondence: ; Tel.: +91-9480073398
| | - Shivakumar Kotrashetti
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karnataka, India
| |
Collapse
|
6
|
Chen Y, Han Y, Wu Y, Hui R, Yang Y, Zhong Y, Zhang S, Zhang W. Pharmacogenetic association of the NR1H3 promoter variant with antihypertensive response among patients with hypertension: A longitudinal study. Front Pharmacol 2023; 14:1083134. [PMID: 36950018 PMCID: PMC10025344 DOI: 10.3389/fphar.2023.1083134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Background: The genetic factors in assessing therapeutic efficacy and predicting antihypertensive drug response are unclear. Therefore, this study aims to identify the associations between variants and antihypertensive drug response. Methods: A longitudinal study including 1837 hypertensive patients was conducted in Northern China and followed up for a median 2.24 years. The associations of 11 candidate variants with blood pressure changes in response to antihypertensive drugs and with the risk of cardiovascular events during the follow-up were examined. The dual-luciferase assay was carried out to assess the effect of genetic variants on gene transcriptional activity. Results: The variant rs11039149A>G in the promoter of nuclear receptor subfamily 1 group H member 3 (NR1H3) was associated with the change in systolic blood pressure (ΔSBP) in response to calcium channel blockers (CCBs) monotherapy. Patients carrying rs11039149AG genotype showed a significant increase of systolic blood pressure (SBP) at follow-up compared with AA carriers, and the difference of ΔSBP between AG and AA carriers was 5.94 mm Hg (95%CI: 2.09-9.78, p = 0.002). In 1,184 patients with CCBs therapy, SBP levels decreased in AA carriers, but increased in AG carriers, the difference of ΔSBP between AG and AA carriers was 8.04 mm Hg (95%CI: 3.28-12.81, p = 0.001). Further analysis in 359 patients with CCBs monotherapy, the difference of ΔSBP between AG and AA carriers was 15.25 mm Hg (95%CI: 6.48-24.02, p = 0.001). However, there was no significant difference in ΔSBP between AG and AA carriers with CCBs multitherapy. The rs11039149A>G was not associated with the cardiovascular events incidence during the follow-up. Additionally, transcriptional factor forkhead box C1 (FOXC1) bound to the NR1H3 promoter containing rs11039149A and significantly increased the transcriptional activity, while rs11039149 A to G change led to a loss-of-function and disabled FOXC1 binding. For the other 10 variants, associations with blood pressure changes or risk of cardiovascular events were not observed. Conclusion: Hypertensive patients with rs11039149AG genotype in the NR1H3 gene have a significant worse SBP control in response to CCBs monotherapy compared with AA carriers. Our findings suggest that the NR1H3 gene might act as a promising genetic factor to affect individual sensitivity to antihypertensive drugs.
Collapse
Affiliation(s)
- Yu Chen
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
| | - Yuqing Han
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
| | - Yiyi Wu
- The First Affiliated Hospital of Anhui University of Science and Technology, The First People’s Hospital of Huainan City, Huainan, Anhui, China
| | - Rutai Hui
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
| | - Yunyun Yang
- Clinical Laboratory, Xiamen Key Laboratory of Genetic Testing, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Yixuan Zhong
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
| | - Shuyuan Zhang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
| | - Weili Zhang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
- Central-China Branch of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Hospital, Zhengzhou, China
- *Correspondence: Weili Zhang,
| |
Collapse
|
7
|
Single Nucleotide Polymorphisms in Amlodipine-Associated Genes and Their Correlation with Blood Pressure Control among South African Adults with Hypertension. Genes (Basel) 2022; 13:genes13081394. [PMID: 36011305 PMCID: PMC9407577 DOI: 10.3390/genes13081394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/31/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022] Open
Abstract
Objective: This study describes the single nucleotide polymorphisms (SNPs) in amlodipine-associated genes and assesses their correlation with blood pressure control among South African adults with hypertension. Methods: In total, 304 hypertensive patients on amlodipine treatment belonging to the indigenous Swati, Xhosa and Zulu population groups of South Africa were recruited between June 2017 and June 2019. Participants were categorized into: controlled (blood pressure < 140/90 mmHg) and uncontrolled (blood pressure ≥ 140/90 mmHg) hypertension. Thirteen SNPs in amlodipine pharmacogenes with a high PharmGKB evidence base were selected and genotyped using MassArray (Agena BioscienceTM). Logistic regression was fitted to identify significant associations between the SNPs and blood pressure control with amlodipine. Results: The majority of the participants were females (76.6%), older than 45 years (89.1%) and had uncontrolled hypertension (52.3%). Of the 13 SNPs genotyped, five SNPs, rs1042713 (minor allele frequency = 45.9%), rs10494366 (minor allele frequency = 35.3%), rs2239050 (minor allele frequency = 28.7%), rs2246709 (minor allele frequency = 51.6%) and rs4291 (minor allele frequency = 34.4%), were detected among the Xhosa participants, while none were detected among the Swati and Zulu tribal groups. Variants rs1042713 and rs10494366 demonstrated an expression frequency of 97.5% and 79.5%, respectively. Variant TA genotype of rs4291 was significantly associated with uncontrolled hypertension. No association was established between blood pressure response to amlodipine and the remaining four SNPs. Conclusions: This study reports the discovery of five SNPs in amlodipine genes (rs2239050, rs2246709, rs4291, rs1042713 and rs10494366) among the indigenous Xhosa-speaking tribe of South Africa. In addition, the TA genotype of rs4291 was associated with blood pressure control in this cohort. These findings might open doors for more pharmacogenomic studies, which could inform innovations to personalised anti-hypertensive treatment in the ethnically diverse population of South Africa.
Collapse
|
8
|
Abstract
Hypertension is a frequent finding in elderly patients. Hypertension in older age can be both associated with frailty and represent a risk factor for frailty. Hypertension is recognized as a main risk factor for cardiovascular diseases such as heart failure, atrial fibrillation, and stroke and the occurrence of these diseases may provoke a decline in health status and/or worsen the degree of frailty. Blood pressure targets in hypertensive older and frail patients are not completely defined. However, specific evaluations of individual patients and their co-morbidities and assessment of domains and components of frailty, together with weighted consideration of drug use, may help in finding the appropriate therapy.
Collapse
|
9
|
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
|
10
|
Polymorphisms in common antihypertensive targets: Pharmacogenomic implications for the treatment of cardiovascular disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 94:141-182. [PMID: 35659371 DOI: 10.1016/bs.apha.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The idea of personalized medicine came to fruition with sequencing the human genome; however, aside from a few cases, the genetic revolution has yet to materialize. Cardiovascular diseases are the leading cause of death globally, and hypertension is a common prelude to nearly all cardiovascular diseases. Thus, hypertension is an ideal candidate disease to apply tenants of personalized medicine to lessen cardiovascular disease. Herein is a survey that visually depicts the polymorphisms in the top eight antihypertensive targets. Although there are numerous genome-wide association studies regarding cardiovascular disease, few studies look at the effects of receptor polymorphisms on drug treatment. With 17,000+ polymorphisms in the combined target proteins examined, it is expected that some of the clinical variability in the treatment of hypertension is due to polymorphisms in the drug targets. Recent advances in techniques and technology, such as high throughput examination of single mutations, structure prediction, computational power for modeling, and CRISPR models of point mutations, allow for a relatively rapid and comprehensive examination of the effects of known and future polymorphisms on drug affinity and effects. As hypertension is easy to measure and has a plethora of clinically viable ligands, hypertension makes an excellent disease to study pharmacogenomics in the lab and the clinic. If the promises of personalized medicine are to materialize, a concerted effort to examine the effects polymorphisms have on drugs is required. A clinician with the knowledge of a patient's genotype can then prescribe drugs that are optimal for treating that specific patient.
Collapse
|
11
|
McDonough CW, Warren HR, Jack JR, Motsinger-Reif AA, Armstrong ND, Bis JC, House JS, Singh S, El Rouby NM, Gong Y, Mychaleckyj JC, Rotroff DM, Benavente OR, Caulfield MJ, Doria A, Pepine CJ, Psaty BM, Glorioso V, Glorioso N, Hiltunen TP, Kontula KK, Arnett DK, Buse JB, Irvin MR, Johnson JA, Munroe PB, Wagner MJ, Cooper-DeHoff RM. Adverse Cardiovascular Outcomes and Antihypertensive Treatment: A Genome-Wide Interaction Meta-Analysis in the International Consortium for Antihypertensive Pharmacogenomics Studies. Clin Pharmacol Ther 2021; 110:723-732. [PMID: 34231218 PMCID: PMC8672325 DOI: 10.1002/cpt.2355] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 06/11/2021] [Indexed: 01/01/2023]
Abstract
We sought to identify genome-wide variants influencing antihypertensive drug response and adverse cardiovascular outcomes, utilizing data from four randomized controlled trials in the International Consortium for Antihypertensive Pharmacogenomics Studies (ICAPS). Genome-wide antihypertensive drug-single nucleotide polymorphism (SNP) interaction tests for four drug classes (β-blockers, n = 9,195; calcium channel blockers (CCBs), n = 10,511; thiazide/thiazide-like diuretics, n = 3,516; ACE-inhibitors/ARBs, n = 2,559) and cardiovascular outcomes (incident myocardial infarction, stroke, or death) were analyzed among patients with hypertension of European ancestry. Top SNPs from the meta-analyses were tested for replication of cardiovascular outcomes in an independent Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) study (n = 21,267), blood pressure (BP) response in independent ICAPS studies (n = 1,552), and ethnic validation in African Americans from the Genetics of Hypertension Associated Treatment study (GenHAT; n = 5,115). One signal reached genome-wide significance in the β-blocker-SNP interaction analysis (rs139945292, Interaction P = 1.56 × 10-8 ). rs139945292 was validated through BP response to β-blockers, with the T-allele associated with less BP reduction (systolic BP response P = 6 × 10-4 , Beta = 3.09, diastolic BP response P = 5 × 10-3 , Beta = 1.53). The T-allele was also associated with increased adverse cardiovascular risk within the β-blocker treated patients' subgroup (P = 2.35 × 10-4 , odds ratio = 1.57, 95% confidence interval = 1.23-1.99). The locus showed nominal replication in CHARGE, and consistent directional trends in β-blocker treated African Americans. rs139945292 is an expression quantitative trait locus for the 50 kb upstream gene NTM (neurotrimin). No SNPs attained genome-wide significance for any other drugs classes. Top SNPs were located near CALB1 (CCB), FLJ367777 (ACE-inhibitor), and CES5AP1 (thiazide). The NTM region is associated with increased risk for adverse cardiovascular outcomes and less BP reduction in β-blocker treated patients. Further investigation into this region is warranted.
Collapse
Affiliation(s)
- Caitrin W. McDonough
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Helen R. Warren
- Clinical Pharmacology Department, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- NIHR Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - John R. Jack
- Bioinformatics Research Center, Department of Statistics, North Carolina State University, Raleigh, North Carolina, USA
| | - Alison A. Motsinger-Reif
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | - Nicole D. Armstrong
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - John S. House
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | - Sonal Singh
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Nihal M. El Rouby
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Yan Gong
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Joesyf C. Mychaleckyj
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Daniel M. Rotroff
- Bioinformatics Research Center, Department of Statistics, North Carolina State University, Raleigh, North Carolina, USA
| | - Oscar R. Benavente
- Department of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mark J. Caulfield
- National Institute for Health Research, Barts Cardiovascular Biomedical Research Center, Queen Mary University of London, London, UK
| | - Alessandrio Doria
- Research Division, Joslin Diabetes Center; and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Carl J. Pepine
- Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Valeria Glorioso
- Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milano, Italy
| | - Nicola Glorioso
- Department of Clinical, Surgical and Experimental Science, University of Sassari, Medical School, Sassari, Italy
| | - Timo P. Hiltunen
- Department of Medicine and Research Program for Clinical and Molecular Metabolism, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Kimmo K. Kontula
- Department of Medicine and Research Program for Clinical and Molecular Metabolism, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Donna K. Arnett
- College of Public Health, Dean’s Office, University of Kentucky, Lexington, Kentucky, USA
| | - John B. Buse
- Division of Endocrinology, Department of Medicine, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Marguerite R. Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Julie A. Johnson
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
- Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Patricia B. Munroe
- Clinical Pharmacology Department, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- NIHR Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michael J. Wagner
- Center for Pharmacogenomics and Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Rhonda M. Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
- Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
12
|
Rahman F, Muthaiah N, Kumaramanickavel G. Current concepts and molecular mechanisms in pharmacogenetics of essential hypertension. Indian J Pharmacol 2021; 53:301-309. [PMID: 34414909 PMCID: PMC8411967 DOI: 10.4103/ijp.ijp_593_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Hypertension is a leading age-related disease in our society and if left untreated, leads to fatal cardiovascular complications. The prevalence of hypertension has increased and becomes a significant global health economic burden, particularly in lower-income societies. Many loci associated with blood pressure and hypertension have been reported by genome-wide association studies that provided potential targets for pharmacotherapy. Pharmacogenetic research had shown interindividual variations in drug efficacy, safety, and tolerability. This could be due to genetic polymorphisms in the pharmacokinetics (genes involved in a transporter, plasma protein binding, and metabolism) or pharmacodynamic pathway (receptors, ion channels, enzymes). Pharmacogenetics promises great hope toward targeted therapy, but challenges remain in implementing pharmacogenetic aided antihypertensive therapy in clinical practice. Using various databases, we analyzed the underlying mechanisms between the candidate gene polymorphisms and antihypertensive drug interactions and the challenges of implementing precision medicine. We review the emergence of pharmacogenetics and its relevance to clinical pharmacological practice.
Collapse
Affiliation(s)
- Farhana Rahman
- Department of Pharmacology, Sree Balaji Medical College and Hospital, Bharat University, Chennai, Tamil Nadu, India
| | - Nagasundaram Muthaiah
- Department of Pharmacology, Sree Balaji Medical College and Hospital, Bharat University, Chennai, Tamil Nadu, India
| | - Govindasamy Kumaramanickavel
- Genomic Research Centre, Sree Balaji Medical College and Hospital, Bharat University, Chennai, Tamil Nadu, India
| |
Collapse
|
13
|
McDonough CW. Pharmacogenomics in Cardiovascular Diseases. Curr Protoc 2021; 1:e189. [PMID: 34232575 DOI: 10.1002/cpz1.189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cardiovascular pharmacogenomics is the study and identification of genomic markers that are associated with variability in cardiovascular drug response, cardiovascular drug-related outcomes, or cardiovascular drug-related adverse events. This overview presents an introduction and historical background to cardiovascular pharmacogenomics, and a protocol for designing a cardiovascular pharmacogenomics study. Important considerations are also included for constructing a cardiovascular pharmacogenomics phenotype, designing the replication or validation strategy, common statistical approaches, and how to put the results in context with the cardiovascular drug or cardiovascular disease under investigation. © 2021 Wiley Periodicals LLC. Basic Protocol: Designing a cardiovascular pharmacogenomics study.
Collapse
Affiliation(s)
- Caitrin W McDonough
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida
| |
Collapse
|
14
|
Prashanth G, Vastrad B, Tengli A, Vastrad C, Kotturshetti I. Investigation of candidate genes and mechanisms underlying obesity associated type 2 diabetes mellitus using bioinformatics analysis and screening of small drug molecules. BMC Endocr Disord 2021; 21:80. [PMID: 33902539 PMCID: PMC8074411 DOI: 10.1186/s12902-021-00718-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Obesity associated type 2 diabetes mellitus is a metabolic disorder ; however, the etiology of obesity associated type 2 diabetes mellitus remains largely unknown. There is an urgent need to further broaden the understanding of the molecular mechanism associated in obesity associated type 2 diabetes mellitus. METHODS To screen the differentially expressed genes (DEGs) that might play essential roles in obesity associated type 2 diabetes mellitus, the publicly available expression profiling by high throughput sequencing data (GSE143319) was downloaded and screened for DEGs. Then, Gene Ontology (GO) and REACTOME pathway enrichment analysis were performed. The protein - protein interaction network, miRNA - target genes regulatory network and TF-target gene regulatory network were constructed and analyzed for identification of hub and target genes. The hub genes were validated by receiver operating characteristic (ROC) curve analysis and RT- PCR analysis. Finally, a molecular docking study was performed on over expressed proteins to predict the target small drug molecules. RESULTS A total of 820 DEGs were identified between healthy obese and metabolically unhealthy obese, among 409 up regulated and 411 down regulated genes. The GO enrichment analysis results showed that these DEGs were significantly enriched in ion transmembrane transport, intrinsic component of plasma membrane, transferase activity, transferring phosphorus-containing groups, cell adhesion, integral component of plasma membrane and signaling receptor binding, whereas, the REACTOME pathway enrichment analysis results showed that these DEGs were significantly enriched in integration of energy metabolism and extracellular matrix organization. The hub genes CEBPD, TP73, ESR2, TAB1, MAP 3K5, FN1, UBD, RUNX1, PIK3R2 and TNF, which might play an essential role in obesity associated type 2 diabetes mellitus was further screened. CONCLUSIONS The present study could deepen the understanding of the molecular mechanism of obesity associated type 2 diabetes mellitus, which could be useful in developing therapeutic targets for obesity associated type 2 diabetes mellitus.
Collapse
Affiliation(s)
- G Prashanth
- Department of General Medicine, Basaveshwara Medical College, Chitradurga, Karnataka, 577501, India
| | - Basavaraj Vastrad
- Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka, 582103, India
| | - Anandkumar Tengli
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru and JSS Academy of Higher Education & Research, Mysuru, Karnataka, 570015, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad, Karnataka, 580001, India.
| | - Iranna Kotturshetti
- Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, Karnataka, 582209, India
| |
Collapse
|
15
|
Alghamdi MA, Al-Eitan L, Alkhatib R, Al-Assi A, Almasri A, Aljamal H, Aman H, Khasawneh R. Variants in CDHR3, CACNAC1, and LTA Genes Predisposing Sensitivity and Response to Warfarin in Patients with Cardiovascular Disease. Int J Gen Med 2021; 14:1093-1100. [PMID: 33790638 PMCID: PMC8006967 DOI: 10.2147/ijgm.s298597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/08/2021] [Indexed: 11/23/2022] Open
Abstract
Introduction Warfarin has been in use for more than 60 years; however, it has serious side effects including major bleeding. The high interpatient variability in the required dose impacts the sensitivity and responsiveness to warfarin in different patients. This study aims to assess the influence of CDHR3, CACNAC1, and LTA gene polymorphisms on the variability of warfarin dose requirements and susceptibility to coronary heart disease in the Jordanian population. Methods This study was conducted in the anti-coagulation clinic in Queen Alia Heart Institute in Amman, with 212 patients in total. Three SNPs were genotyped within CDHR3 (rs10270308), CACNAC1 (rs216013), and LTA (rs1041981) genes. Results Our findings revealed that patients with LTA polymorphism are more prone to warfarin sensitivity than others. Furthermore, carriers of the LTA polymorphism needed a lower initial dose of warfarin and are associated with less variation in doses required to achieve target INR. Conclusion The current study could help in understanding the role of genetic variability in warfarin dosing and matching patients to different treatment options. Clinical applications of these findings for warfarin treatment may also contribute to improving the efficacy and safety of warfarin treatment in Jordanian patients with cardiovascular disease.
Collapse
Affiliation(s)
- Mansour A Alghamdi
- Department of Anatomy, College of Medicine, King Khalid University, Abha, 61421, Saudi Arabia.,Genomics and Personalized Medicine Unit, College of Medicine, King Khalid University, Abha, 61421, Saudi Arabia
| | - Laith Al-Eitan
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Rami Alkhatib
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Ahmad Al-Assi
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Ayah Almasri
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Hanan Aljamal
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Hatem Aman
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Rame Khasawneh
- Department of Hematopathology, King Hussein Medical Center (KHMC), Royal Medical Services (RMS), Amman, 11118, Jordan
| |
Collapse
|
16
|
Rysz J, Franczyk B, Rysz-Górzyńska M, Gluba-Brzózka A. Pharmacogenomics of Hypertension Treatment. Int J Mol Sci 2020; 21:ijms21134709. [PMID: 32630286 PMCID: PMC7369859 DOI: 10.3390/ijms21134709] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/21/2020] [Accepted: 06/26/2020] [Indexed: 12/12/2022] Open
Abstract
Hypertension is one of the strongest modifiable cardiovascular risk factors, affecting an increasing number of people worldwide. Apart from poor medication adherence, the low efficacy of some therapies could also be related to inter-individual genetic variability. Genetic studies of families revealed that heritability accounts for 30% to 50% of inter-individual variation in blood pressure (BP). Genetic factors not only affect blood pressure (BP) elevation but also contribute to inter-individual variability in response to antihypertensive treatment. This article reviews the recent pharmacogenomics literature concerning the key classes of antihypertensive drugs currently in use (i.e., diuretics, β-blockers, ACE inhibitors, ARB, and CCB). Due to the numerous studies on this topic and the sometimes-contradictory results within them, the presented data are limited to several selected SNPs that alter drug response. Genetic polymorphisms can influence drug responses through genes engaged in the pathogenesis of hypertension that are able to modify the effects of drugs, modifications in drug–gene mechanistic interactions, polymorphisms within drug-metabolizing enzymes, genes related to drug transporters, and genes participating in complex cascades and metabolic reactions. The results of numerous studies confirm that genotype-based antihypertension therapies are the most effective and may help to avoid the occurrence of major adverse events, as well as decrease the costs of treatment. However, the genetic heritability of drug response phenotypes seems to remain hidden in multigenic and multifactorial complex traits. Therefore, further studies are required to analyze all associations and formulate final genome-based treatment recommendations.
Collapse
Affiliation(s)
- Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.)
| | - Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.)
| | - Magdalena Rysz-Górzyńska
- Department of Ophthalmology and Visual Rehabilitation, Medical University of Lodz, 90-549 Lodz, Poland;
| | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.)
- Correspondence:
| |
Collapse
|
17
|
Lam CK, Tian L, Belbachir N, Wnorowski A, Shrestha R, Ma N, Kitani T, Rhee JW, Wu JC. Identifying the Transcriptome Signatures of Calcium Channel Blockers in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Circ Res 2019; 125:212-222. [PMID: 31079550 DOI: 10.1161/circresaha.118.314202] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
RATIONALE Calcium channel blockers (CCBs) are an important class of drugs in managing cardiovascular diseases. Patients usually rely on these medications for the remainder of their lives after diagnosis. Although the acute pharmacological actions of CCBs in the hearts are well-defined, little is known about the drug-specific effects on human cardiomyocyte transcriptomes and physiological alterations after long-term exposure. OBJECTIVE This study aimed to simulate chronic CCB treatment and to examine both the functional and transcriptomic changes in human cardiomyocytes. METHODS AND RESULTS We differentiated cardiomyocytes and generated engineered heart tissues from 3 human induced pluripotent stem cell lines and exposed them to 4 different CCBs-nifedipine, amlodipine, diltiazem, and verapamil-at their physiological serum concentrations for 2 weeks. Without inducing cell death and damage to myofilament structure, CCBs elicited line-specific inhibition on calcium kinetics and contractility. While all 4 CCBs exerted similar inhibition on calcium kinetics, verapamil applied the strongest inhibition on cardiomyocyte contractile function. By profiling cardiomyocyte transcriptome after CCB treatment, we identified little overlap in their transcriptome signatures. Verapamil is the only inhibitor that reduced the expression of contraction-related genes, such as MYH (myosin heavy chain) and troponin I, consistent with its depressive effects on contractile function. The reduction of these contraction-related genes may also explain the responsiveness of patients with hypertrophic cardiomyopathy to verapamil in managing left ventricular outflow tract obstruction. CONCLUSIONS This is the first study to identify the transcriptome signatures of different CCBs in human cardiomyocytes. The distinct gene expression patterns suggest that although the 4 inhibitors act on the same target, they may have distinct effects on normal cardiac cell physiology.
Collapse
Affiliation(s)
- Chi Keung Lam
- From the Stanford Cardiovascular Institute, CA (C.K.L., L.T., N.B., A.W., R.S., N.M., T.K., J.-W.R., J.C.W.).,Department of Medicine, Division of Cardiology (C.K.L., L.T., N.B., A.W., R.S., N.M., T.K., J.-W.R., J.C.W.), Stanford University School of Medicine, CA
| | - Lei Tian
- From the Stanford Cardiovascular Institute, CA (C.K.L., L.T., N.B., A.W., R.S., N.M., T.K., J.-W.R., J.C.W.).,Department of Medicine, Division of Cardiology (C.K.L., L.T., N.B., A.W., R.S., N.M., T.K., J.-W.R., J.C.W.), Stanford University School of Medicine, CA
| | - Nadjet Belbachir
- From the Stanford Cardiovascular Institute, CA (C.K.L., L.T., N.B., A.W., R.S., N.M., T.K., J.-W.R., J.C.W.).,Department of Medicine, Division of Cardiology (C.K.L., L.T., N.B., A.W., R.S., N.M., T.K., J.-W.R., J.C.W.), Stanford University School of Medicine, CA
| | - Alexa Wnorowski
- From the Stanford Cardiovascular Institute, CA (C.K.L., L.T., N.B., A.W., R.S., N.M., T.K., J.-W.R., J.C.W.).,Department of Medicine, Division of Cardiology (C.K.L., L.T., N.B., A.W., R.S., N.M., T.K., J.-W.R., J.C.W.), Stanford University School of Medicine, CA
| | - Rajani Shrestha
- From the Stanford Cardiovascular Institute, CA (C.K.L., L.T., N.B., A.W., R.S., N.M., T.K., J.-W.R., J.C.W.).,Department of Medicine, Division of Cardiology (C.K.L., L.T., N.B., A.W., R.S., N.M., T.K., J.-W.R., J.C.W.), Stanford University School of Medicine, CA
| | - Ning Ma
- From the Stanford Cardiovascular Institute, CA (C.K.L., L.T., N.B., A.W., R.S., N.M., T.K., J.-W.R., J.C.W.).,Department of Medicine, Division of Cardiology (C.K.L., L.T., N.B., A.W., R.S., N.M., T.K., J.-W.R., J.C.W.), Stanford University School of Medicine, CA
| | - Tomoya Kitani
- From the Stanford Cardiovascular Institute, CA (C.K.L., L.T., N.B., A.W., R.S., N.M., T.K., J.-W.R., J.C.W.).,Department of Medicine, Division of Cardiology (C.K.L., L.T., N.B., A.W., R.S., N.M., T.K., J.-W.R., J.C.W.), Stanford University School of Medicine, CA
| | - June-Wha Rhee
- From the Stanford Cardiovascular Institute, CA (C.K.L., L.T., N.B., A.W., R.S., N.M., T.K., J.-W.R., J.C.W.).,Department of Medicine, Division of Cardiology (C.K.L., L.T., N.B., A.W., R.S., N.M., T.K., J.-W.R., J.C.W.), Stanford University School of Medicine, CA
| | - Joseph C Wu
- From the Stanford Cardiovascular Institute, CA (C.K.L., L.T., N.B., A.W., R.S., N.M., T.K., J.-W.R., J.C.W.).,Department of Medicine, Division of Cardiology (C.K.L., L.T., N.B., A.W., R.S., N.M., T.K., J.-W.R., J.C.W.), Stanford University School of Medicine, CA.,Deparment of Radiology (J.C.W.), Stanford University School of Medicine, CA
| |
Collapse
|
18
|
Abstract
Pharmacogenetics is an emerging area of medicine, and more work is needed to fully integrate it into a clinical setting for the benefit of patients. Genetic markers can influence the action of many drugs, including those that prevent and treat cardiovascular conditions. Genotyping is not yet commonplace, but guidelines are being put in place to help practitioners determine the effect a genetic marker may have on certain drugs. With advancements in genetic technology and falling costs, genotyping could be available to all patients via a simple saliva test. This would be a cost-effective way for practitioners to determine the most effective treatment for individuals, reducing "trial and error," adverse effects, and rehospitalization rates and increasing patient compliance. Cardiovascular diseases are the leading causes of death worldwide, so using the most effective medication to treat or prevent them is of utmost importance in reducing incidence and mortality.
Collapse
|
19
|
Association of single nucleotide polymorphisms in CACNA 1A/CACNA 1C/CACNA 1H calcium channel genes with diabetic peripheral neuropathy in Chinese population. Biosci Rep 2018; 38:BSR20171670. [PMID: 29581247 PMCID: PMC6435562 DOI: 10.1042/bsr20171670] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/20/2018] [Accepted: 03/26/2018] [Indexed: 12/22/2022] Open
Abstract
The present study was conducted to explore the correlations between single nucleotide polymorphisms (SNPs) in the calcium channel CACNA 1A, CACNA 1C, and CACNA 1H genes and diabetic peripheral neuropathy (DPN) amongst the Chinese population. In total, 281 patients diagnosed with type 2 diabetes participated in the present study. These patients were divided into the case group, which was subdivided into the DPN (143 cases) and the non-DPN groups (138 cases). Subsequently, 180 healthy individuals that had undergone routine health examinations were also recruited and assigned to the control group. PCR-restriction fragment length polymorphism (PCR-RFLP) was used to detect the genotype and allele frequencies of CACNA 1A, CACNA 1C, and CACNA 1H genes; logistic regression analysis to investigate the association of gene polymorphisms with DNP. Gene–gene interactions were then detected by generalized multifactor dimensionality reduction (GMDR). The results revealed that CACNA 1A rs2248069 and rsl6030, CACNA 1C rs216008 and rs2239050, and CACNA 1H rs3794619, and rs7191246 SNPs were all associated with DPN, while rs2248069, rsl6030, rs2239050, and rs7191246 polymorphisms were attributed to the susceptibility to DPN. It was also observed that the optimal models were three-, four- and five-dimensional models with a prediction accuracy of 61.05% and the greatest consistency of cross-validation was 10/10. In summary, these findings demonstrated that the SNPs in the CACNA 1A, CACNA 1C, and CACNA 1H genes were involved in the pathophysiology of DPN. In addition, polymorphisms in the CACNA 1A, CACNA 1C, and CACNA 1H genes and their interactions also had effects on DPN.
Collapse
|
20
|
Eadon MT, Kanuri SH, Chapman AB. Pharmacogenomic studies of hypertension: paving the way for personalized antihypertensive treatment. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2018; 3:33-47. [PMID: 29888336 DOI: 10.1080/23808993.2018.1420419] [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] [Indexed: 01/02/2023]
Abstract
Introduction Increasing clinical evidence supports the implementation of genotyping for anti-hypertensive drug dosing and selection. Despite robust evidence gleaned from clinical trials, the translation of genotype guided therapy into clinical practice faces significant challenges. Challenges to implementation include the small effect size of individual variants and the polygenetic nature of antihypertensive drug response, a lack of expert consensus on dosing guidelines even without genetic information, and proper definition of major antihypertensive drug toxicities. Balancing clinical benefit with cost, while overcoming these challenges, remains crucial. Areas covered This review presents the most impactful clinical trials and cohorts which continue to inform and guide future investigation. Variants were selected from among those identified in the Pharmacogenomic Evaluation of Antihypertensive Responses (PEAR), the Genetic Epidemiology of Responses to Antihypertensives study (GERA), the Genetics of Drug Responsiveness in Essential Hypertension (GENRES) study, the SOPHIA study, the Milan Hypertension Pharmacogenomics of hydro-chlorothiazide (MIHYPHCTZ), the Campania Salute Network, the International Verapamil SR Trandolapril Study (INVEST), the Nordic Diltiazem (NORDIL) Study, GenHAT, and others. Expert Commentary The polygenic nature of antihypertensive drug response is a major barrier to clinical implementation. Further studies examining clinical effectiveness are required to support broad-based implementation of genotype-based prescribing in medical practice.
Collapse
Affiliation(s)
- Michael T Eadon
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sri H Kanuri
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | |
Collapse
|
21
|
Amato RJ, Boland J, Myer N, Few L, Dowd D. Pharmacogenomics and Psychiatric Clinical Care. J Psychosoc Nurs Ment Health Serv 2018; 56:22-31. [DOI: 10.3928/02793695-20170928-01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/14/2017] [Indexed: 12/28/2022]
|
22
|
Lin K, Xu G, Shi L, Lu W, Guan L, Ouyang H, Chen K, Dang Y, Zhou L, So KF. CACNA1C polymorphisms Impact Cognitive Recovery in Patients with Bipolar Disorder in a Six-week Open-label Trial. Sci Rep 2017; 7:7022. [PMID: 28765577 PMCID: PMC5539285 DOI: 10.1038/s41598-017-07368-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/28/2017] [Indexed: 11/21/2022] Open
Abstract
Cognitive impairments in bipolar patients deteriorate as the disorder progresses. Little is known about whether genetic risks impact cognitive recovery during the course from depression to remission. In this six-week open-label trial, we shed light on the impacts of six single nucleotide polymorphisms (SNPs) in the calcium voltage-gated channel subunit alpha1 C (CACNA1C) gene on cognitive recovery in 192 bipolar patients suffering a major depressive episode (MDE). The primary outcome measures were changes in a battery of neuropsychological tests following 6-week treatment. Carriers with rs10466907 GT genotype did not significantly improve their executive function total scores on the Wisconsin Card Sorting Test after six weeks of treatment compared to the TT genotypes (β = −0.944, 95% Confidence Interval (CI) = −1.482–−0.405). Moreover, during a MDE carriers with rs58619945 GG and GA genotypes performed significantly worse than those with AA genotype on the categories completed (p = 0.013 and p = 0.001), total errors (p = 0.039 and p = 0.009), and random errors (p = 0.055 and p = 0.014, respectively). Our data suggest that the tested CACNA1C SNPs may have impacts on cognitive recovery from depression.
Collapse
Affiliation(s)
- Kangguang Lin
- Department of Affective Disorders, The Affiliated Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China.,Laboratory of Emotion and Cognition, The Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,GMH Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Guiyun Xu
- Department of Affective Disorders, The Affiliated Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China. .,Laboratory of Emotion and Cognition, The Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Lingling Shi
- GMH Institute of CNS Regeneration, Jinan University, Guangzhou, China.,Ministry of Education Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou, China.,Co-Innovation Center for Neuroregeneration, Nantong University, Nantong, China
| | - Weicong Lu
- Department of Affective Disorders, The Affiliated Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China.,Laboratory of Emotion and Cognition, The Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lijie Guan
- Department of Affective Disorders, The Affiliated Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Huiyi Ouyang
- Department of Affective Disorders, The Affiliated Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Kun Chen
- Department of Affective Disorders, The Affiliated Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China.,Laboratory of Emotion and Cognition, The Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yamei Dang
- Department of Affective Disorders, The Affiliated Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Libing Zhou
- GMH Institute of CNS Regeneration, Jinan University, Guangzhou, China.,Ministry of Education Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou, China.,Co-Innovation Center for Neuroregeneration, Nantong University, Nantong, China
| | - Kwok-Fai So
- GMH Institute of CNS Regeneration, Jinan University, Guangzhou, China. .,Ministry of Education Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou, China. .,Co-Innovation Center for Neuroregeneration, Nantong University, Nantong, China. .,The State Key Laboratory of Brain and Cognitive Sciences and Department of Ophthalmology, The University of Hong Kong, Hong Kong, Hong Kong.
| |
Collapse
|
23
|
Phan NN, Wang CY, Chen CF, Sun Z, Lai MD, Lin YC. Voltage-gated calcium channels: Novel targets for cancer therapy. Oncol Lett 2017; 14:2059-2074. [PMID: 28781648 PMCID: PMC5530219 DOI: 10.3892/ol.2017.6457] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 04/13/2017] [Indexed: 01/11/2023] Open
Abstract
Voltage-gated calcium channels (VGCCs) comprise five subtypes: The L-type; R-type; N-type; P/Q-type; and T-type, which are encoded by α1 subunit genes. Calcium ion channels also have confirmed roles in cellular functions, including mitogenesis, proliferation, differentiation, apoptosis and metastasis. An association between VGCCs, a reduction in proliferation and an increase in apoptosis in prostate cancer cells has also been reported. Therefore, in the present study, the online clinical database Oncomine was used to identify the alterations in the mRNA expression level of VGCCs in 19 cancer subtypes. Overall, VGCC family genes exhibited under-expression in numerous types of cancer, including brain, breast, kidney and lung cancers. Notably, the majority of VGCC family members (CACNA1C, CACNA1D, CACNA1A, CACNA1B, CACNA1E, CACNA1H and CACNA1I) exhibited low expression in brain tumors, with mRNA expression levels in the top 1–9% of downregulated gene rankings. A total of 5 VGCC family members (CACNA1A, CACNA1B, CACNA1E, CACNA1G and CACNA1I) were under-expressed in breast cancer, with a gene ranking in the top 1–10% of the low-expressed genes compared with normal tissue. In kidney and lung cancers, CACNA1S, CACNA1C, CACNA1D, CACNA1A and CACNA1H exhibited low expression, with gene rankings in the top 1–8% of downregulated genes. In conclusion, the present findings may contribute to the development of new cancer treatment approaches by identifying target genes involved in specific types of cancer.
Collapse
Affiliation(s)
- Nam Nhut Phan
- Faculty of Applied Sciences, Ton Duc Thang University, Tan Phong Ward, Ho Chi Minh 700000, Vietnam
| | - Chih-Yang Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, R.O.C.,Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, R.O.C.,Department of Anatomy, University of California, San Francisco, CA 94143, USA
| | - Chien-Fu Chen
- School of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung 84001, Taiwan, R.O.C
| | - Zhengda Sun
- Department of Radiology, University of California, San Francisco, CA 94143, USA
| | - Ming-Derg Lai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, R.O.C.,Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, R.O.C
| | - Yen-Chang Lin
- Graduate Institute of Biotechnology, Chinese Culture University, Taipei 1114, Taiwan, R.O.C
| |
Collapse
|
24
|
Wild PS, Felix JF, Schillert A, Teumer A, Chen MH, Leening MJ, Völker U, Großmann V, Brody JA, Irvin MR, Shah SJ, Pramana S, Lieb W, Schmidt R, Stanton AV, Malzahn D, Smith AV, Sundström J, Minelli C, Ruggiero D, Lyytikäinen LP, Tiller D, Smith JG, Monnereau C, Di Tullio MR, Musani SK, Morrison AC, Pers TH, Morley M, Kleber ME, Aragam J, Benjamin EJ, Bis JC, Bisping E, Broeckel U, Cheng S, Deckers JW, Del Greco M F, Edelmann F, Fornage M, Franke L, Friedrich N, Harris TB, Hofer E, Hofman A, Huang J, Hughes AD, Kähönen M, investigators KNHI, Kruppa J, Lackner KJ, Lannfelt L, Laskowski R, Launer LJ, Leosdottir M, Lin H, Lindgren CM, Loley C, MacRae CA, Mascalzoni D, Mayet J, Medenwald D, Morris AP, Müller C, Müller-Nurasyid M, Nappo S, Nilsson PM, Nuding S, Nutile T, Peters A, Pfeufer A, Pietzner D, Pramstaller PP, Raitakari OT, Rice KM, Rivadeneira F, Rotter JI, Ruohonen ST, Sacco RL, Samdarshi TE, Schmidt H, Sharp AS, Shields DC, Sorice R, Sotoodehnia N, Stricker BH, Surendran P, Thom S, Töglhofer AM, Uitterlinden AG, Wachter R, Völzke H, Ziegler A, Münzel T, März W, Cappola TP, Hirschhorn JN, Mitchell GF, Smith NL, Fox ER, Dueker ND, Jaddoe VW, Melander O, Russ M, Lehtimäki T, Ciullo M, Hicks AA, Lind L, Gudnason V, Pieske B, Barron AJ, Zweiker R, Schunkert H, Ingelsson E, Liu K, Arnett DK, Psaty BM, Blankenberg S, Larson MG, Felix SB, Franco OH, Zeller T, Vasan RS, Dörr M. Large-scale genome-wide analysis identifies genetic variants associated with cardiac structure and function. J Clin Invest 2017; 127:1798-1812. [PMID: 28394258 PMCID: PMC5409098 DOI: 10.1172/jci84840] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 02/16/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Understanding the genetic architecture of cardiac structure and function may help to prevent and treat heart disease. This investigation sought to identify common genetic variations associated with inter-individual variability in cardiac structure and function. METHODS A GWAS meta-analysis of echocardiographic traits was performed, including 46,533 individuals from 30 studies (EchoGen consortium). The analysis included 16 traits of left ventricular (LV) structure, and systolic and diastolic function. RESULTS The discovery analysis included 21 cohorts for structural and systolic function traits (n = 32,212) and 17 cohorts for diastolic function traits (n = 21,852). Replication was performed in 5 cohorts (n = 14,321) and 6 cohorts (n = 16,308), respectively. Besides 5 previously reported loci, the combined meta-analysis identified 10 additional genome-wide significant SNPs: rs12541595 near MTSS1 and rs10774625 in ATXN2 for LV end-diastolic internal dimension; rs806322 near KCNRG, rs4765663 in CACNA1C, rs6702619 near PALMD, rs7127129 in TMEM16A, rs11207426 near FGGY, rs17608766 in GOSR2, and rs17696696 in CFDP1 for aortic root diameter; and rs12440869 in IQCH for Doppler transmitral A-wave peak velocity. Findings were in part validated in other cohorts and in GWAS of related disease traits. The genetic loci showed associations with putative signaling pathways, and with gene expression in whole blood, monocytes, and myocardial tissue. CONCLUSION The additional genetic loci identified in this large meta-analysis of cardiac structure and function provide insights into the underlying genetic architecture of cardiac structure and warrant follow-up in future functional studies. FUNDING For detailed information per study, see Acknowledgments.
Collapse
Affiliation(s)
- Philipp S. Wild
- Preventive Cardiology and Preventive Medicine, Department of Medicine 2, and
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- DZHK (German Centre for Cardiovascular Research), partner site RhineMain, Mainz, Germany
| | - Janine F. Felix
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Arne Schillert
- Institute for Medical Biometry and Statistics, University Lübeck, University Medical Center Schleswig-Holstein, Lübeck, Germany
- DZHK, partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK, partner site Greifswald, Greifswald, Germany
| | - Ming-Huei Chen
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Maarten J.G. Leening
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Uwe Völker
- DZHK, partner site Greifswald, Greifswald, Germany
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Vera Großmann
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Marguerite R. Irvin
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sanjiv J. Shah
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Setia Pramana
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Wolfgang Lieb
- Institute of Epidemiology and Popgen Biobank, Christian-Albrechts University of Kiel, Kiel, Germany
| | - Reinhold Schmidt
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University Graz, Graz, Austria
| | - Alice V. Stanton
- Blood Pressure Unit, Beaumont Hospital, Dublin, Ireland
- Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Dörthe Malzahn
- Department of Genetic Epidemiology, University Medical Center, Georg-August University, Göttingen, Germany
| | - Albert Vernon Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Johan Sundström
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Cosetta Minelli
- Population Health and Occupational Disease, National Heart and Lung Institute (NHLI), Imperial College London, London, United Kingdom
| | - Daniela Ruggiero
- Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, Naples, Italy
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Daniel Tiller
- Institute of Medical Epidemiology, Biostatistics, and Informatics, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - J. Gustav Smith
- Department of Cardiology, Lund University and Skåne University Hospital, Lund, Sweden
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA
- Center for Human Genetic Research and Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Claire Monnereau
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- The Generation R Study Group and
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Marco R. Di Tullio
- Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Solomon K. Musani
- Jackson Heart Study, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Alanna C. Morrison
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Tune H. Pers
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children’s Hospital, Boston, Massachusetts, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Statens Serum Institut, Department of Epidemiology Research, Copenhagen, Denmark
| | - Michael Morley
- Penn Cardiovascular Institute and Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marcus E. Kleber
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - AortaGen Consortium
- Members of the AortaGen Consortium and their affiliations are detailed in the Supplemental Acknowledgments
| | - Jayashri Aragam
- Harvard Medical School, Boston, Massachusetts, USA
- Veteran’s Administration Hospital, West Roxbury, Boston, Massachusetts, USA
| | - Emelia J. Benjamin
- National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, Massachusetts, USA
- Sections of Cardiology, Preventive Medicine and Epidemiology, Department of Medicine, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, USA
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Egbert Bisping
- Department of Cardiology, Medical University Graz, Graz, Austria
| | | | | | - Susan Cheng
- National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, Massachusetts, USA
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jaap W. Deckers
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Fabiola Del Greco M
- Center for Biomedicine, European Academy of Bolzano/Bozen, Bolzano, Italy – Affiliated institute of the University of Lübeck, Lübeck, Germany
| | - Frank Edelmann
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Myriam Fornage
- University of Texas Health Science Center, Houston, Texas, USA
| | - Lude Franke
- Department of Genetics, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Nele Friedrich
- DZHK, partner site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Tamara B. Harris
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, NIH, Bethesda, Maryland, USA
| | - Edith Hofer
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University Graz, Graz, Austria
- Institute for Medical Informatics, Statistics and Documentation, Medical University Graz, Graz, Austria
| | - Albert Hofman
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Jie Huang
- Boston VA Research Institute, Boston, Massachusetts, USA
- Brigham and Women’s Hospital Division of Aging, Harvard Medical School, Boston, Massachusetts, USA
| | - Alun D. Hughes
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
- Department of Clinical Physiology, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - KNHI investigators
- KNHI investigators and their affiliations are detailed in the Supplemental Acknowledgments
| | - Jochen Kruppa
- Institute for Medical Biometry and Statistics, University Lübeck, University Medical Center Schleswig-Holstein, Lübeck, Germany
- University of Veterinary Medicine, Foundation Institute of Veterinary Medicine and Genetics, Hannover, Germany
| | - Karl J. Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - Lars Lannfelt
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Rafael Laskowski
- Department of Medicine 2, University Medical Center Mainz, Mainz, Germany
| | - Lenore J. Launer
- Neuroepidemiology Section, National Institute on Aging, NIH, Bethesda, Maryland, USA
| | - Margrét Leosdottir
- Department of Cardiology, Lund University, and Skåne University Hospital, Malmö, Sweden
| | - Honghuang Lin
- National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, Massachusetts, USA
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Cecilia M. Lindgren
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA
| | - Christina Loley
- Institute for Medical Biometry and Statistics, University Lübeck, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Calum A. MacRae
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Deborah Mascalzoni
- Center for Biomedicine, European Academy of Bolzano/Bozen, Bolzano, Italy – Affiliated institute of the University of Lübeck, Lübeck, Germany
| | - Jamil Mayet
- International Centre for Circulatory Health, Hammersmith Hospital, London, United Kingdom
- NHLI, Imperial College London, London, United Kingdom
| | - Daniel Medenwald
- Institute of Medical Epidemiology, Biostatistics, and Informatics, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Department of Biostatistics, University of Liverpool, Liverpool, United Kingdom
| | - Christian Müller
- Department of General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany
| | - Martina Müller-Nurasyid
- Department of Medicine I, Ludwig-Maximilians-University Munich, Munich, Germany
- DZHK, partner site Munich Heart Alliance, Munich, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Stefania Nappo
- Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, Naples, Italy
| | - Peter M. Nilsson
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Sebastian Nuding
- Department of Medicine III, University Clinics Halle (Saale), Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Teresa Nutile
- Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, Naples, Italy
| | - Annette Peters
- DZHK, partner site Munich Heart Alliance, Munich, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Arne Pfeufer
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Diana Pietzner
- Institute of Medical Epidemiology, Biostatistics, and Informatics, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Peter P. Pramstaller
- Center for Biomedicine, European Academy of Bolzano/Bozen, Bolzano, Italy – Affiliated institute of the University of Lübeck, Lübeck, Germany
- Department of Neurology, General Central Hospital, Bolzano, Italy
- Department of Neurology, University of Lübeck, Lübeck, Germany
| | - Olli T. Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Kenneth M. Rice
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- The Generation R Study Group and
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Saku T. Ruohonen
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Ralph L. Sacco
- Department of Neurology and
- McKnight Brain Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA
- Departments of Public Health Sciences and Human Genomics, University of Miami, Miami, Florida, USA
| | - Tandaw E. Samdarshi
- Division of Cardiology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Helena Schmidt
- Institute of Molecular Biology and Biochemistry, Medical University Graz, Graz, Austria
| | - Andrew S.P. Sharp
- Department of Cardiology, Royal Devon and Exeter Hospital and University of Exeter, Exeter, United Kingdom
| | - Denis C. Shields
- UCD Conway Institute of Biomolecular and Biomedical Research and
- School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Rossella Sorice
- Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, Naples, Italy
- IRCCS Neuromed, Pozzilli, Isernia, Italy
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
- Division of Cardiology, University of Washington, Seattle, Washington, USA
| | - Bruno H. Stricker
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Inspectorate for Health Care, Utrecht, Netherlands
| | - Praveen Surendran
- Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
- School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Simon Thom
- International Centre for Circulatory Health, Hammersmith Hospital, London, United Kingdom
- NHLI, Imperial College London, London, United Kingdom
| | - Anna M. Töglhofer
- Institute of Molecular Biology and Biochemistry, Medical University Graz, Graz, Austria
| | - André G. Uitterlinden
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Rolf Wachter
- Department of Cardiology and Pneumology, University Medical Center of Göttingen, Georg-August University, Göttingen, Germany
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK, partner site Greifswald, Greifswald, Germany
| | - Andreas Ziegler
- Institute for Medical Biometry and Statistics, University Lübeck, University Medical Center Schleswig-Holstein, Lübeck, Germany
- DZHK, partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
- School of Mathematics, Statistics and Computer Science, University of KwaZulu-Natal, Durban, South Africa
- Zentrum für Klinische Studien, Universität Lübeck, Lübeck, Germany
| | - Thomas Münzel
- DZHK (German Centre for Cardiovascular Research), partner site RhineMain, Mainz, Germany
- Department of Medicine 2, University Medical Center Mainz, Mainz, Germany
| | - Winfried März
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Synlab Academy, Synlab Services GmbH, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Thomas P. Cappola
- Penn Cardiovascular Institute and Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joel N. Hirschhorn
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Nicholas L. Smith
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
- Group Health Research Institute, Group Health Cooperative, Seattle, Washington, USA
- Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle, Washington, USA
| | - Ervin R. Fox
- Division of Cardiology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Nicole D. Dueker
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Vincent W.V. Jaddoe
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- The Generation R Study Group and
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Martin Russ
- Department of Medicine III, University Clinics Halle (Saale), Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
- Helios-Amperklinikum Dachau, Dachau, Germany
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Marina Ciullo
- Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, Naples, Italy
- IRCCS Neuromed, Pozzilli, Isernia, Italy
| | - Andrew A. Hicks
- Center for Biomedicine, European Academy of Bolzano/Bozen, Bolzano, Italy – Affiliated institute of the University of Lübeck, Lübeck, Germany
| | - Lars Lind
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Burkert Pieske
- Department of Cardiology, Medical University Graz, Graz, Austria
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
- German Heart Institute Berlin DHZB, Department of Internal Medicine/Cardiology, Berlin, Germany
| | - Anthony J. Barron
- International Centre for Circulatory Health, Hammersmith Hospital, London, United Kingdom
- NHLI, Imperial College London, London, United Kingdom
| | - Robert Zweiker
- Department of Cardiology, Medical University Graz, Graz, Austria
| | - Heribert Schunkert
- DZHK, partner site Munich Heart Alliance, Munich, Germany
- Deutsches Herzzentrum, Technische Universität München, Munich, Germany
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Kiang Liu
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Donna K. Arnett
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Bruce M. Psaty
- Group Health Research Institute, Group Health Cooperative, Seattle, Washington, USA
- Cardiovacular Health Research Unit, Departments of Medicine, Epidemiology, and Health Services, University of Washington, Seattle, Washington, USA
| | - Stefan Blankenberg
- DZHK, partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany
| | - Martin G. Larson
- Biostatistics Department, Boston University School of Public Health, Boston, Massachusetts, USA
- Department of Mathematics and Statistics, Boston University, Boston, Massachusetts, USA
| | - Stephan B. Felix
- DZHK, partner site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Oscar H. Franco
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Tanja Zeller
- DZHK, partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany
| | - Ramachandran S. Vasan
- National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, Massachusetts, USA
- Sections of Cardiology, Preventive Medicine and Epidemiology, Department of Medicine, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, USA
| | - Marcus Dörr
- DZHK, partner site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
25
|
Baek MY, Hwang HS, Park JY, Chung JE, Lee KE, Lee GY, Seong JW, Yee J, Kim YJ, Gwak HS. Association between CACNA1C gene polymorphisms and ritodrine-induced adverse events in preterm labor patients. Eur J Clin Pharmacol 2017; 73:837-842. [PMID: 28391406 DOI: 10.1007/s00228-017-2222-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 02/17/2017] [Indexed: 12/19/2022]
Abstract
PURPOSE As a tocolytic agent, ritodrine has been used in European and Asian countries but has lost popularity due to safety concerns. This study aimed to investigate the relationship between adverse drug events caused by ritodrine and the CACNA1C polymorphisms in preterm labor patients. METHODS Data were collected from medical records including maternal age, gestational age, body mass index, dilation score, effacement score, modified Bishop score, maximum infusion rate, and adverse drug events. Five single-nucleotide polymorphisms of the CACNA1C gene (rs10774053, rs215994, rs215976, rs2239128, and rs2041135) were analyzed. RESULTS One hundred eighty-six patients were included, 33 of whom had adverse drug events. A allele carriers of rs10774053 showed about 0.293-fold lower adverse drug events than GG genotype carriers (p = 0.012, absolute risk reduction = 16.5%) after adjusting for other confounding variables; the number needed to genotype for preventing one patient with GG genotype from suffering higher incidence of adverse drug events was calculated to be 14.6. Increase in maximum infusion rate of 1 mL/h was associated with a 1.03-fold (95% CI 1.01~1.06, p = 0.005) increased risk of adverse drug events. None of the patients with a CC genotype of rs215994 had adverse drug events, whereas 22.1% of the T allele carriers had adverse drug events. CONCLUSION This study showed that CACNA1C gene polymorphisms could alter the probability of adverse drug event risk when ritodrine is used in preterm labor.
Collapse
Affiliation(s)
- Min Young Baek
- College of Pharmacy & Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Han Sung Hwang
- Department of Obstetrics and Gynecology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, 05030, Republic of Korea
| | - Jin Young Park
- College of Pharmacy & Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Jee Eun Chung
- College of Pharmacy, Sungkyunkwan University, Suwon-si, Kyunggi-do, 16419, Republic of Korea
| | - Kyung Eun Lee
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Gwan Yung Lee
- College of Pharmacy & Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Jin Won Seong
- College of Pharmacy & Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Jeong Yee
- College of Pharmacy & Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Young Ju Kim
- Department of Obstetrics and Gynecology, Ewha Womans University School of Medicine, Seoul, 07985, Republic of Korea.
| | - Hye Sun Gwak
- College of Pharmacy & Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea.
| |
Collapse
|
26
|
Smooth Muscle Phenotypic Diversity: Effect on Vascular Function and Drug Responses. ADVANCES IN PHARMACOLOGY 2017. [PMID: 28212802 DOI: 10.1016/bs.apha.2016.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
At its simplest resistance to blood flow is regulated by changes in the state of contraction of the vascular smooth muscle (VSM), a function of the competing activities of the myosin kinase and phosphatase determining the phosphorylation and activity of the myosin ATPase motor protein. In contrast, the vascular system of humans and other mammals is incredibly complex and highly regulated. Much of this complexity derives from phenotypic diversity within the smooth muscle, reflected in very differing power outputs and responses to signaling pathways that regulate vessel tone, presumably having evolved over the millennia to optimize vascular function and its control. The highly regulated nature of VSM tone, described as pharmacomechanical coupling, likely underlies the many classes of drugs in clinical use to alter vascular tone through activation or inhibition of these signaling pathways. This review will first describe the phenotypic diversity within VSM, followed by presentation of specific examples of how molecular diversity in signaling, myofilament, and calcium cycling proteins impacts arterial smooth muscle function and drug responses.
Collapse
|
27
|
Zaiou M, El Amri H. Cardiovascular pharmacogenetics: a promise for genomically‐guided therapy and personalized medicine. Clin Genet 2016; 91:355-370. [DOI: 10.1111/cge.12881] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 09/30/2016] [Accepted: 10/03/2016] [Indexed: 12/28/2022]
Affiliation(s)
- M. Zaiou
- Faculté de PharmacieUniversité de Lorraine Nancy France
| | - H. El Amri
- Laboratoire de Génétique de la Gendarmerie RoyaleAvenue Ibn Sina Rabat Maroc
| |
Collapse
|
28
|
Chang SW, McDonough CW, Gong Y, Johnson TA, Tsunoda T, Gamazon ER, Perera MA, Takahashi A, Tanaka T, Kubo M, Pepine CJ, Johnson JA, Cooper-DeHoff RM. Genome-wide association study identifies pharmacogenomic loci linked with specific antihypertensive drug treatment and new-onset diabetes. THE PHARMACOGENOMICS JOURNAL 2016; 18:106-112. [PMID: 27670767 PMCID: PMC5368017 DOI: 10.1038/tpj.2016.67] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 07/11/2016] [Accepted: 08/25/2016] [Indexed: 01/14/2023]
Abstract
We conducted a discovery genome-wide association study with expression quantitative trait loci (eQTL) annotation of new-onset diabetes (NOD) among European Americans, who were exposed to a calcium channel blocker-based strategy (CCB strategy) or a β-blocker-based strategy (β-blocker strategy) in the INternational VErapamil SR Trandolapril STudy. Replication of the top signal from the SNP*treatment interaction analysis was attempted in Hispanic and African Americans, and a joint meta-analysis was performed (total 334 NOD cases and 806 matched controls). PLEKHH2 rs11124945 at 2p21 interacted with antihypertensive exposure for NOD (meta-analysis p=5.3×10−8). rs11124945 G allele carriers had lower odds for NOD when exposed to the β-blocker strategy compared with the CCB strategy [OR=0.38 (0.24-0.60), p=4.0×10−5], while A/A homozygotes exposed to the β-blocker strategy had increased odds for NOD compared with the CCB strategy [OR=2.02 (1.39-2.92), p=2.0×10−4]. eQTL annotation of the 2p21 locus provides functional support for regulating gene expression.
Collapse
Affiliation(s)
- S-W Chang
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - C W McDonough
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Y Gong
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - T A Johnson
- Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - T Tsunoda
- Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan.,Department of Medical Science Mathematics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - E R Gamazon
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - M A Perera
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - A Takahashi
- Laboratory for Statistical Analysis, SNP Research Center, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - T Tanaka
- Laboratory for Cardiovascular Diseases, SNP Research Center, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - M Kubo
- Laboratory for Genotyping Development, SNP Research Center, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - C J Pepine
- Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - J A Johnson
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, FL, USA.,Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - R M Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, FL, USA.,Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
29
|
Eadon MT, Chapman AB. A Physiologic Approach to the Pharmacogenomics of Hypertension. Adv Chronic Kidney Dis 2016; 23:91-105. [PMID: 26979148 DOI: 10.1053/j.ackd.2016.02.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Hypertension is a multifactorial condition with diverse physiological systems contributing to its pathogenesis. Individuals exhibit significant variation in their response to antihypertensive agents. Traditional markers, such as age, gender, diet, plasma renin level, and ethnicity, aid in drug selection. However, this review explores the contribution of genetics to facilitate antihypertensive agent selection and predict treatment efficacy. The findings, reproducibility, and limitations of published studies are examined, with emphasis placed on candidate genetic variants affecting drug metabolism, the renin-angiotensin system, adrenergic signalling, and renal sodium reabsorption. Single-nucleotide polymorphisms identified and replicated in unbiased genome-wide association studies of hypertension treatment are reviewed to illustrate the evolving understanding of the disease's complex and polygenic pathophysiology. Implementation efforts at academic centers seek to overcome barriers to the broad adoption of pharmacogenomics in the treatment of hypertension. The level of evidence required to support the implementation of pharmacogenomics in clinical practice is considered.
Collapse
|
30
|
Goldstein BI, Carnethon MR, Matthews KA, McIntyre RS, Miller GE, Raghuveer G, Stoney CM, Wasiak H, McCrindle BW. Major Depressive Disorder and Bipolar Disorder Predispose Youth to Accelerated Atherosclerosis and Early Cardiovascular Disease: A Scientific Statement From the American Heart Association. Circulation 2015; 132:965-86. [PMID: 26260736 DOI: 10.1161/cir.0000000000000229] [Citation(s) in RCA: 340] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the 2011 "Expert Panel on Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents," several medical conditions among youth were identified that predispose to accelerated atherosclerosis and early cardiovascular disease (CVD), and risk stratification and management strategies for youth with these conditions were elaborated. Major depressive disorder (MDD) and bipolar disorder (BD) among youth satisfy the criteria set for, and therefore merit inclusion among, Expert Panel tier II moderate-risk conditions. The combined prevalence of MDD and BD among adolescents in the United States is ≈10%, at least 10 times greater than the prevalence of the existing moderate-risk conditions combined. The high prevalence of MDD and BD underscores the importance of positioning these diseases alongside other pediatric diseases previously identified as moderate risk for CVD. The overall objective of this statement is to increase awareness and recognition of MDD and BD among youth as moderate-risk conditions for early CVD. To achieve this objective, the primary specific aims of this statement are to (1) summarize evidence that MDD and BD are tier II moderate-risk conditions associated with accelerated atherosclerosis and early CVD and (2) position MDD and BD as tier II moderate-risk conditions that require the application of risk stratification and management strategies in accordance with Expert Panel recommendations. In this scientific statement, there is an integration of the various factors that putatively underlie the association of MDD and BD with CVD, including pathophysiological mechanisms, traditional CVD risk factors, behavioral and environmental factors, and psychiatric medications.
Collapse
|
31
|
Abstract
Personalized medicine seeks to identify the right dose of the right drug for the right patient at the right time. Typically, individualization of therapy is based on the pharmacogenomic makeup of the individual and environmental factors that alter drug disposition and response. In addition to these factors, during pregnancy, a woman's body undergoes many changes that can impact the therapeutic efficacy of medications. Yet, there is minimal research regarding personalized medicine in obstetrics. Adoption of pharmacogenetic testing into the obstetrical care is dependent on evidence of analytical validity, clinical validity, and clinical utility. Here, we briefly present information regarding the potential utility of personalized medicine for treating the obstetric patient for pain with narcotics, hypertension, and preterm labor, and discuss the impediments of bringing personalized medicine to the obstetrical clinic.
Collapse
Affiliation(s)
- Sara K Quinney
- Department of Obstetrics and Gynecology and Department of Medicine, Indiana University School of Medicine, Indianapolis, IN.
| | - David A Flockhart
- Departments of Medicine, Medical Genetics, and Pharmacology; Indiana Institute of Personalized Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Avinash S Patil
- Departments of Obstetrics & Gynecology and Medicine; Indiana Institute for Personalized Medicine, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
32
|
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
|
33
|
The neurobiology of bipolar disorder: identifying targets for specific agents and synergies for combination treatment. Int J Neuropsychopharmacol 2014; 17:1039-52. [PMID: 23449044 DOI: 10.1017/s1461145713000096] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Bipolar disorder (BD) is a chronic psychiatric illness described by severe changes in mood. Extensive research has been carried out to understand the aetiology and pathophysiology of BD. Several hypotheses have been postulated, including alteration in genetic factors, protein expression, calcium signalling, neuropathological alteration, mitochondrial dysfunction and oxidative stress in BD. In the following paper, we will attempt to integrate these data in a manner which is to understand targets of treatment and how they may be, in particular, relevant to combination treatment. In summary, the data suggested that BD might be associated with neuronal and glial cellular impairment in specific brain areas, including the prefrontal cortex. From molecular and genetics: (1) alterations in dopaminergic system, through catechol-O-aminotransferase; (2) decreased expression and polymorphism on brain-derived neurotrophic factor; (3) alterations cyclic-AMP responsive element binding; (4) dysregulation of calcium signalling, including genome-wide finding for voltage-dependent calcium channel α-1 subunit are relevant findings in BD. Future studies are now necessary to understand how these molecular pathways interact and their connection to the complex clinical manifestations observed in BD.
Collapse
|
34
|
Zhang J, Cai J, Zhang X, Ni J, Guo Z, Zhang Y, Lu W, Zhang C. Does the bipolar disorder-associated CACNA1C gene confer susceptibility to schizophrenia in Han Chinese? J Mol Neurosci 2013; 51:474-7. [PMID: 23900723 DOI: 10.1007/s12031-013-0079-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 07/16/2013] [Indexed: 10/26/2022]
Abstract
The gene coding for the calcium channel, voltage-dependent, L type, alpha 1C subunit (CACNA1C) has been reported to be associated with bipolar disorder (BD) in our previous study. Broad evidence suggests some degree of shared genetic susceptibility between BD and schizophrenia. In this study, we aimed to determine whether the BD-associated gene CACNA1C confers susceptibility to schizophrenia. The association was assessed using a cross section study of 696 schizophrenia patients and 1,114 matched control subjects of Han Chinese origin. Between-group comparisons of genotypic or allelic frequencies showed no statistically significant differences. CACNA1C is unlikely to play a major role in the pathophysiology of schizophrenia in Han Chinese. Further large-scale replication studies using a haplotype-tagging single-nucleotide polymorphism selection approach are required to obtain more conclusive results of any potential association.
Collapse
Affiliation(s)
- Jiangtao Zhang
- First Geriatric Department, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, China
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Bochud M, Guessous I. Gene-environment interactions of selected pharmacogenes in arterial hypertension. Expert Rev Clin Pharmacol 2013; 5:677-86. [PMID: 23234325 DOI: 10.1586/ecp.12.58] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Hypertension affects approximately 1 billion people worldwide. Owing to population aging, hypertension-related cardiovascular burden is expected to rise in the near future. In addition to genetic variants influencing the blood pressure response to antihypertensive drugs, several genes encoding for drug-metabolizing or -transporting enzymes have been associated with blood pressure and/or hypertension in humans (e.g., ACE, CYP1A2, CYP3A5, ABCB1 and MTHFR) regardless of drug treatment. These genes are also involved in the metabolism and transport of endogenous substances and their effects may be modified by selected environmental factors, such as diet or lifestyle. However, little is currently known on the complex interplay between environmental factors, endogenous factors, genetic variants and drugs on blood pressure control. This review will discuss the respective role of population-based primary prevention and personalized medicine for arterial hypertension, taking a pharmacogenomics' perspective focusing on selected pharmacogenes.
Collapse
Affiliation(s)
- Murielle Bochud
- Institute of Social and Preventive Medicine, Lausanne University Hospital, Switzerland.
| | | |
Collapse
|
36
|
Davis JT, Rao F, Naqshbandi D, Fung MM, Zhang K, Schork AJ, Nievergelt CM, Ziegler MG, O'Connor DT. Autonomic and hemodynamic origins of pre-hypertension: central role of heredity. J Am Coll Cardiol 2012; 59:2206-16. [PMID: 22676942 DOI: 10.1016/j.jacc.2012.02.040] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2011] [Revised: 02/13/2012] [Accepted: 02/14/2012] [Indexed: 01/11/2023]
Abstract
OBJECTIVES The purpose of this study is to better understand the origins and progression of pre-hypertension. BACKGROUND Pre-hypertension is a risk factor for progression to hypertension, cardiovascular disease, and increased mortality. We used a cross-sectional twin study design to examine the role of heredity in likely pathophysiological events (autonomic or hemodynamic) in pre-hypertension. METHODS Eight hundred twelve individuals (337 normotensive, 340 pre-hypertensive, 135 hypertensive) were evaluated in a sample of twin pairs, their siblings, and other family members. They underwent noninvasive hemodynamic, autonomic, and biochemical testing, as well as estimates of trait heritability (the percentage of trait variance accounted for by heredity) and pleiotropy (the genetic covariance or shared genetic determination of traits) by variance components. RESULTS In the hemodynamic realm, an elevation of cardiac contractility prompted increased stroke volume, in turn increasing cardiac output, which elevated blood pressure into the pre-hypertension range. Autonomic monitoring detected an elevation of norepinephrine secretion plus a decline in cardiac parasympathetic tone. Twin pair variance components documented substantial heritability as well as joint genetic determination for blood pressure and the contributory autonomic and hemodynamic traits. Genetic variation at a pathway locus also indicated pleiotropic effects on contractility and blood pressure. CONCLUSIONS Elevated blood pressure in pre-hypertension results from increased cardiac output, driven by contractility as well as heart rate, which may reflect both diminished parasympathetic and increased sympathetic tone. In the face of increased cardiac output, systemic vascular resistance fails to decline homeostatically. Such traits display substantial heritability and shared genetic determination, although by loci not yet elucidated. These findings clarify the role of heredity in the origin of pre-hypertension and its autonomic and hemodynamic pathogenesis. The results also establish pathways that suggest new therapeutic targets for pre-hypertension, or approaches to its prevention.
Collapse
Affiliation(s)
- Jason T Davis
- Department of Medicine, University of California-San Diego, and V.A. San Diego Healthcare System, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kandoi G, Nanda A, Scaria V, Sivasubbu S. A case for pharmacogenomics in management of cardiac arrhythmias. Indian Pacing Electrophysiol J 2012; 12:54-64. [PMID: 22557843 PMCID: PMC3337369 DOI: 10.1016/s0972-6292(16)30480-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Disorders of the cardiac rhythm are quite prevalent in clinical practice. Though the variability in drug response between individuals has been extensively studied, this information has not been widely used in clinical practice. Rapid advances in the field of pharmacogenomics have provided us with crucial insights on inter-individual genetic variability and its impact on drug metabolism and action. Technologies for faster and cheaper genetic testing and even personal genome sequencing would enable clinicians to optimize prescription based on the genetic makeup of the individual, which would open up new avenues in the area of personalized medicine. We have systematically looked at literature evidence on pharmacogenomics markers for anti-arrhythmic agents from the OpenPGx consortium collection and reason the applicability of genetics in the management of arrhythmia. We also discuss potential issues that need to be resolved before personalized pharmacogenomics becomes a reality in regular clinical practice.
Collapse
Affiliation(s)
- Gaurav Kandoi
- Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Main Bawana Road, Delhi, India
| | | | | | | |
Collapse
|
38
|
Williams PJ, Morgan L. The role of genetics in pre-eclampsia and potential pharmacogenomic interventions. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2012; 5:37-51. [PMID: 23226061 PMCID: PMC3513227 DOI: 10.2147/pgpm.s23141] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Indexed: 01/23/2023]
Abstract
The pregnancy-specific condition pre-eclampsia not only affects the health of mother and baby during pregnancy but also has long-term consequences, increasing the chances of cardiovascular disease in later life. It is accepted that pre-eclampsia has a placental origin, but the pathogenic mechanisms leading to the systemic endothelial dysfunction characteristic of the disorder remain to be determined. In this review we discuss some key factors regarded as important in the development of pre-eclampsia, including immune maladaptation, inadequate placentation, oxidative stress, and thrombosis. Genetic factors influence all of these proposed pathophysiological mechanisms. The inherited nature of pre-eclampsia has been known for many years, and extensive genetic studies have been undertaken in this area. Genetic research offers an attractive strategy for studying the pathogenesis of pre-eclampsia as it avoids the ethical and practical difficulties of conducting basic science research during the preclinical phase of pre-eclampsia when the underlying pathological changes occur. Although pharmacogenomic studies have not yet been conducted in pre-eclampsia, a number of studies investigating treatment for essential hypertension are of relevance to therapies used in pre-eclampsia. The pharmacogenomics of antiplatelet agents, alpha and beta blockers, calcium channel blockers, and magnesium sulfate are discussed in relation to the treatment and prevention of pre-eclampsia. Pharmacogenomics offers the prospect of individualized patient treatment, ensuring swift introduction of optimal treatment whilst minimizing the use of inappropriate or ineffective drugs, thereby reducing the risk of harmful effects to both mother and baby.
Collapse
|
39
|
Abstract
Patients vary in their responses to drug therapy, and some of that variability is genetically determined. This review outlines general approaches used to identify genetic variation that influences drug response. Examples from specific therapeutic areas are presented, such as cholesterol management, arrhythmias, heart failure, hypertension, warfarin anticoagulation, and antiplatelet agents. A brief view of potential pathways to implementation is presented.
Collapse
Affiliation(s)
- Dan M Roden
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232-0575, USA.
| | | | | | | | | | | | | |
Collapse
|
40
|
Pan S, Dewey FE, Perez MV, Knowles JW, Chen R, Butte AJ, Ashley EA. Personalized Medicine and Cardiovascular Disease: From Genome to Bedside. CURRENT CARDIOVASCULAR RISK REPORTS 2011. [DOI: 10.1007/s12170-011-0202-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
41
|
Johnson JA, Cavallari LH, Beitelshees AL, Lewis JP, Shuldiner AR, Roden DM. Pharmacogenomics: application to the management of cardiovascular disease. Clin Pharmacol Ther 2011; 90:519-31. [PMID: 21918509 PMCID: PMC3190669 DOI: 10.1038/clpt.2011.179] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The past decade has seen substantial advances in cardiovascular pharmacogenomics. Genetic determinants of response to clopidogrel and warfarin have been defined, resulting in changes to the product labels for these drugs that suggest the use of genetic information as a guide for therapy. Genetic tests are available, as are guidelines for incorporation of genetic information into patient-care decisions. These guidelines and the literature supporting them are reviewed herein. Significant advances have also been made in the pharmacogenomics of statin-induced myopathy and the response to β-blockers in heart failure, although the clinical applications of these findings are less clear. Other areas hold promise, including the pharmacogenomics of antihypertensive drugs, aspirin, and drug-induced long-QT syndrome (diLQTS). The potential value of pharmacogenomics in the discovery and development of new drugs is also described. In summary, pharmacogenomics has current applications in the management of cardiovascular disease, with clinically relevant data continuing to mount.
Collapse
Affiliation(s)
- J A Johnson
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, Florida, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Price ET, Pacanowski MA, Martin MA, Cooper-DeHoff RM, Pepine CJ, Zineh I, Johnson JA. Liver X receptor α gene polymorphisms and variable cardiovascular outcomes in patients treated with antihypertensive therapy: results from the INVEST-GENES study. Pharmacogenet Genomics 2011; 21:333-40. [PMID: 21562465 DOI: 10.1097/fpc.0b013e3283452fec] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND/AIMS Liver X receptor-α (LXRA) is a nuclear receptor that regulates genes important in cholesterol homeostasis and inflammation. Several single nucleotide polymorphisms (SNPs) in the LXRA gene (NR1H3) have been earlier associated with metabolic phenotypes (dyslipidemia and elevated body mass index). Metabolic dysregulation is a major contributor to coronary disease; therefore, we assessed LXRA in International Verapamil Sustained Release SR Trandolapril Study Genetic Substudy (INVEST-GENES), a genetic-substudy of a large clinical trial in patients with hypertension and coronary artery disease. METHODS Seven tag SNPs in the LXRA gene region (NR1H3) were selected for study: rs11039149, rs12221497, rs2279238, rs7120118, rs326213, rs11039159, and rs10501321. One thousand fifty-nine patients were genotyped from the INVEST-GENES case-control set (verapamil-sustained release-based or atenolol-based treatment strategies) that comprised of 297 cases frequency matched (approximately 2.5:1) with that of event-free controls by sex and race. The primary outcome was defined as first occurrence of all-cause death, nonfatal myocardial infarction, or nonfatal stroke. Adjusted odds ratios (ORs) were calculated using logistic regression. RESULTS Three of the seven SNPs were associated with significant effects on the primary outcome in nonBlacks. The variant G allele of rs11039149 and the variant A allele of rs12221497 were associated with reduced risk of experiencing the primary outcome [OR: 0.62, confidence interval (CI): 0.45-0.85, P=0.003 and OR: 0.60, CI: 0.39-0.91, P=0.016, respectively]. The rs2279238 genotype was associated with a significant increase in risk for the primary outcome (OR: 1.42, CI: 1.03-1.95, P=0.03). Furthermore, there was a significant genotype-treatment strategy interaction for carriers of the variant T allele of rs2279238 (OR for verapamil-sustained release strategy compared with atenolol strategy: 2.86, CI: 1.50-5.46, P=0.0015). Diplotype analyses showed that the SNPs are rarely coinherited and support the directionally opposite effects of the SNPs on the primary outcome. CONCLUSION LXRA genotypes were associated with variable risk for cardiovascular outcomes and pharmacogenetic effect in INVEST-GENES. These novel findings suggest that LXRA is a genetic/pharmacogenetic target that should be further explored.
Collapse
Affiliation(s)
- Elvin Tyrone Price
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida College of Medicine, Gainesville, FL, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Structural and functional differences between L-type calcium channels: crucial issues for future selective targeting. Trends Pharmacol Sci 2011; 32:366-75. [DOI: 10.1016/j.tips.2011.02.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 02/07/2011] [Accepted: 02/17/2011] [Indexed: 11/21/2022]
|
44
|
Jung C, Gené GG, Tomás M, Plata C, Selent J, Pastor M, Fandos C, Senti M, Lucas G, Elosua R, Valverde MA. A gain-of-function SNP in TRPC4 cation channel protects against myocardial infarction. Cardiovasc Res 2011; 91:465-71. [PMID: 21427121 DOI: 10.1093/cvr/cvr083] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS The TRPC4 non-selective cation channel is widely expressed in the endothelium, where it generates Ca(2+) signals that participate in the endothelium-mediated vasodilatory response. This study sought to identify single-nucleotide polymorphisms (SNPs) in the TRPC4 gene that are associated with myocardial infarction (MI). METHODS AND RESULTS Our candidate-gene association studies identified a missense SNP (TRPC4-I957V) associated with a reduced risk of MI in diabetic patients [odds ratio (OR) = 0.61; confidence interval (CI), 0.40-0.95, P= 0.02]. TRPC4 was also associated with MI in the Wellcome Trust Case-Control Consortium's genome-wide data: an intronic SNP (rs7319926) within the same linkage disequilibrium block as TRPC4-I957V showed an OR of 0.86 (CI, 0.81-0.94; P =10(-4)). Functional studies of the missense SNP were carried out in HEK293 and CHO cells expressing wild-type or mutant channels. Patch-clamp studies and measurement of intracellular [Ca(2+)] in response to muscarinic agonists and direct G-protein activation showed increased channel activity in TRPC4-I957V-transfected cells compared with TRPC4-WT. Site-directed mutagenesis and molecular modelling of TRPC4-I957V suggested that the gain of function was due to the presence of a less bulky Val-957. This permits a firmer interaction between the TRPC4 and the catalytic site of the tyrosine kinase that phosphorylates TRPC4 at Tyr-959 and facilitates channel insertion into the plasma membrane. CONCLUSION We provide evidence for the association of a TRPC4 SNP with MI in population-based genetic studies. The higher Ca(2+) signals generated by TRPC4-I957V may ultimately facilitate the generation of endothelium- and nitric oxide-dependent vasorelaxation, thereby explaining its protective effect at the vasculature.
Collapse
Affiliation(s)
- Carole Jung
- Laboratory of Molecular Physiology and Channelopathies, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona, C/ Dr. Aiguader 88, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Franke B, Vasquez AA, Veltman JA, Brunner HG, Rijpkema M, Fernández G. Genetic variation in CACNA1C, a gene associated with bipolar disorder, influences brainstem rather than gray matter volume in healthy individuals. Biol Psychiatry 2010; 68:586-8. [PMID: 20638048 DOI: 10.1016/j.biopsych.2010.05.037] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Revised: 05/07/2010] [Accepted: 05/25/2010] [Indexed: 12/22/2022]
Abstract
BACKGROUND Genetic variation in CACNA1C has been repeatedly shown to increase risk for psychiatric disorders, with the strongest evidence for involvement in bipolar disorder. To elucidate the mechanisms by which such effects on psychiatric disease are brought about by genetic factors, we investigated the influence of CACNA1C polymorphisms on brain structure. METHODS In 585 healthy volunteers, for whom magnetic resonance imaging data at 1.5 T (n = 282) or 3 T (n = 304) were available, we tested 193 single nucleotide polymorphisms (SNPs) in or near CACNA1C for association with FSL FIRST-segmented subcortical brain structures and hippocampus as well as SPM5-derived total brain volume and global gray and white matter volume using PLINK. RESULTS A study-wide significant association of SNPs in intron 3 of the CACNA1C gene was found for brainstem volume (lowest p value = 3.62E-05) and was confirmed by voxel-based morphometry. An effect on gray matter volume of the bipolar disorder-associated SNP rs1006737, as reported earlier in a sample of 77 healthy adults, could not be confirmed. CONCLUSIONS Genetic variation in the pleiotropic psychiatric disease gene CACNA1C is associated with brainstem volume. Modulation of this structure, with its central control over motor, cognitive, affective, and arousal functions, constitutes an interesting novel potential mode of action of psychiatric risk factors.
Collapse
Affiliation(s)
- Barbara Franke
- Department of Human Genetics, Institute for Genetic and Metabolic Disorders, Center for Neuroscience, Radboud University Nijmegen Medical Center, the Netherlands.
| | | | | | | | | | | |
Collapse
|