Next Article in Journal
Age and Serum Adipocyte Fatty-Acid-Binding Protein Level Are Associated with Aortic Stiffness in Coronary Artery Bypass Graft Patients
Previous Article in Journal
Clinical Presentation and Management of Methicillin-Resistant Staphylococcus aureus Pericarditis—Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genome-Wide Association of New-Onset Hypertension According to Renin Concentration: The Korean Genome and Epidemiology Cohort Study

1
Severance Check-up, Yonsei University Health System, Yongin-si 16995, Korea
2
Department of Medicine, Graduate School, Yonsei University Wonju College of Medicine, Wonju-si 26426, Korea
3
Department of Family Medicine, Yongin Severance Hosptal, Yongin-si 16995, Korea
4
Healthcare R&D Division, Theragen Bio Co., Ltd., Ganggyo-ro 145, Suwon-si 16229, Korea
5
Department of Family Medicine, Yonsei University College of Medicine, Seoul 03722, Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Cardiovasc. Dev. Dis. 2022, 9(4), 104; https://doi.org/10.3390/jcdd9040104
Submission received: 10 February 2022 / Revised: 21 March 2022 / Accepted: 29 March 2022 / Published: 30 March 2022
(This article belongs to the Section Genetics)

Abstract

:
The renin-angiotensin system (RAS) is a crucial regulator of vascular resistance and blood volume in the body. This study aimed to examine the genetic predisposition of the plasma renin concentration influencing future hypertension incidence. Based on the Korean Genome and Epidemiology Cohort dataset, 5211 normotensive individuals at enrollment were observed over 12 years, categorized into the low-renin and high-renin groups. We conducted genome-wide association studies for the total, low-renin, and high-renin groups. Among the significant SNPs, the lead SNPs of each locus were focused on for further interpretation. The effect of genotypes was determined by logistic regression analysis between controls and new-onset hypertension, after adjusting for potential confounding variables. During a mean follow-up period of 7.6 years, 1704 participants (32.7%) developed hypertension. The low-renin group showed more incidence rates of new-onset hypertension (35.3%) than the high-renin group (26.5%). Among 153 SNPs in renin-related gene regions, two SNPs (rs11726091 and rs8137145) showed an association in the high-renin group, four SNPs (rs17038966, rs145286444, rs2118663, and rs12336898) in the low-renin group, and three SNPs (rs1938859, rs7968218, and rs117246401) in the total population. Most significantly, the low-renin SNP rs12336898 in the SPTAN1 gene, closely related to vascular wall remodeling, was associated with the development of hypertension (p-value = 1.3 × 10−6). We found the candidate genetic polymorphisms according to blood renin concentration. Our results might be a valuable indicator for hypertension risk prediction and preventive measure, considering renin concentration with genetic susceptibility.

1. Introduction

Hypertension, the primary cause of cardiovascular disease and all-cause mortality, is a leading disease worldwide [1]. Previous estimates suggest that 31.1% (1.39 billion) of adults worldwide had hypertension in 2010 [2]. The prevalence of hypertension has increased globally due to the aging population, exposure to unhealthy foods (i.e., low potassium and high sodium intake), and lack of physical activity. In Korea, hypertension in 2017 was prevalent in 27% of adults over 30 and 60% of adults over 60 [3].
Renin, part of the renin-angiotensin system (RAS), is an enzyme from the kidney’s juxtaglomerular cells, and its secretion mainly responds to renal perfusion pressure [4,5]. Renin hydrolyzes angiotensinogen secreted from the liver into angiotensin I, while circulating through the blood stream. Angiotensin I is converted into angiotensin II by the angiotensin-converting enzyme (ACE). Finally, as a central effector molecule of the RAS, angiotensin II stimulates the release of catecholamines in the adrenal medulla and constricts vessels, resulting in increased blood pressure [6].
RAS activity is often increased in hypertensive patients, and low-renin hypertension has been reported to be around 20–30% [7]. However, epidemiological studies have shown that the degree of RAS dependence on hypertension varies, depending on ethnicity or salt intake [8], which has raised the possibility that genetic susceptibility is one of the underlying mechanisms. Thus, we investigated the genetic predisposition of the blood renin concentration, influencing future hypertension incidence, using a community-based Korean cohort, observed over 12 years.

2. Materials and Methods

2.1. Study Population and Participants

This study used data from the Korean Genome and Epidemiology Study (KoGES) conducted by the National Research Institute of Health (NIH), Centers for Disease Control and Prevention, Ministry of Health and Welfare, Republic of Korea. The KoGES aims to investigate the genetic and environmental etiologies of non-communicable diseases, such as hypertension and type 2 diabetes. A total of 10,030 participants between the ages of 40 and 69 was recruited through two population-based prospective cohort studies conducted in the Ansung (n = 5018) and Ansan (n = 5012) regions of South Korea. Of the 10,030 participants, genotype data were available for 8840. After excluding hypertensive patients at enrollment, we included 5211 normotensive individuals in the final analysis (2440 men and 2771 women). The KoGES is ongoing, with biannual repeated surveys beginning in 2001–2002. Data from the baseline study to the fifth examination, in 2011–2012, were used in the current study. Detailed information about the KoGES is described in a previous report [9]. The dataset used in this study (Ansan–Ansung cohort) was provided after reviewing and evaluating the Korea Centers for Disease Control and Prevention (http://www.cdc.go.kr/CDC/eng/main.jsp (accessed on 14 March 2022)).

2.2. Data Collection

Baseline and follow-up assessment and recruitment were conducted after obtaining informed consent from all participants. Trained physicians examined study subjects according to a standard protocol [9]. Blood pressure was measured in a sitting position with arms supported at the heart level using mercury sphygmomanometers after at least 5 min of rest in a quiet room (Baumanometer, Baum Co. Inc., Copiague, NY, USA). Systolic blood pressure and diastolic blood pressure were defined as the average of both arm readings. Smoking status was determined based on self-questionnaires and classified into two categories: non-smokers (<100 cigarettes in their lifetime) and current smokers (≥100 cigarettes in their lifetime or currently smoking). Blood samples were collected after fasting overnight or for more than 12 h. Serum concentrations of fasting plasma glucose, total cholesterol, triglycerides, and high-density lipoprotein (HDL) cholesterol were measured with an autoanalyzer (Advia 1650, Siemens, Tarrytown, NY, USA). Plasma renin concentration was measured by using Gamma Counter Cobra in a supine position after more than 15 min between 8 and 10 a.m. (Packard, Meriden, CT, USA) [10]. Furthermore, we subdivided renin concentration into low- or high-renin groups based on 3.3 ng/mL, which is the highest tertile value of plasma renin concentration [11] We used the World Health Organization (WHO) diagnostic criteria for type 2 diabetes mellitus: 8-h fasting plasma glucose ≥ 126 mg/dL or HbA1c ≥ 6.5% [12]. In addition, participants who stated that they were receiving hypoglycaemic agents were included in the diabetic group. Newly developed and baseline hypertension were defined as Systolic blood pressure ≥ 140 mmHg, diastolic blood pressure ≥ 90 mmHg, or current use of antihypertensive medication [13].

2.3. Genotype Analysis

Genotypes were obtained from the National Institutes of Health (NIH), and the details of the study design have been described previously [14]. Single nucleotide polymorphism (SNP) genotypes of subjects were extracted from the Affymetrix 5.0 SNP microarray. SNPs with missing genotype call rate > 0.1, minor allele frequency < 0.01, or Hardy–Weinberg equilibrium (HWE) p-value < 1 × 10−6 were excluded. Finally, 333,651 experimentally determined SNPs and 1,000,000 SNPs imputed based on the 100 genome haplotype phase of the Asian panel or the Korean HapMap were used in this study [15].

2.4. Statistical Analysis

We conducted genome-wide association studies (GWAS) for the total, low-renin, and high-renin groups. Among 153 SNPs in renin-related gene regions, the effect of genotype was determined by logistic regression analysis between incident hypertension cases and controls after adjusting for confounding variables. The effect of genotype was determined by logistic regression analysis between controls and new-onset hypertension. The thresholds for association criteria were defined as the genome-wide significant level (p-value < 5 × 10−8) and the genome-wide suggestive level (p-value < 1 × 10−5).

3. Results

3.1. Baseline Characteristics of the Study Population

Table 1 shows the characteristics of the study population. The mean follow-up period was 7.6 years, and a total of 1704 individuals (32.7%, 1704/5211) were diagnosed with hypertension. The mean age was 49.1 ± 7.9 years in the normotensive group and 53.1 ± 8.8 years in the new-onset hypertension group (p-value < 0.001). We divided the sample by renin level into two groups, a high-renin group and a low-renin group, and the low-renin group showed more incidence rates of new-onset hypertension (35.3%) than the high-renin group (26.5%). Body mass index, fasting plasma glucose, and triglycerides were significantly higher in the new-onset hypertension group. HDL cholesterol and type 2 diabetes were lower and less frequent in the new-onset hypertension group. However, total cholesterol, systolic blood pressure, diastolic blood pressure, and current smoker status were not significantly different between groups. The incidence rates of hypertension between males and females were similar, 0.34 and 0.32, respectively.

3.2. Association between SNPs and New-Onset Hypertension

Performing the case-control analysis, we found a significant association between SNPs and new-onset hypertension cases, as listed in Supplementary Table S1. A total of 153 SNPs in renin-related gene regions showed association in all three GWAS. Furthermore, we selected a cluster pick region to represent the more reliable association and summarized the top SNPs of that region in Table 2. An additional column reveals associations by group (L: low-renin group; H: high-renin group; M: both groups). The next column outlines the top significant SNPs among the associated loci, and the final column shows the cluster picked in each locus, with at least three SNPs associated with hypertension with p-value < 1 × 10−5. Among 153 SNPs in renin-related gene regions, two SNPs (rs11726091 and rs8137145) showed an association in the high-renin group, four SNPs (rs17038966, rs145286444, rs2118663, and rs12336898) in the low-renin group, and three SNPs (rs1938859, rs7968218, and rs117246401) in the total population. Among these SNPs, rs12336898 showed the most significant p-value and is located in the 3′ downstream region.

3.3. In Silico Annotation of Linked Genes and Functional Relevance

In silico annotation of the positions of SNPs in gene regions is described in Table 3. Five SNPs were located near the functional gene regions, and four were located in intergenic regions. The most significant SNP, rs12336898, was located downstream of the SPTAN1 gene in the L group. Interestingly, SPTAN1 (αII spectrin) is one of two α spectrin genes, and its activation is associated with the compensation of vessel fibrosis. In silico annotation of the SNP function showed that rs1938859 is located in the 5′ untranslated region of the TRPC6 gene and was reported as an expression quantitative trait locus (eQTL) of the same gene, which is related to glomerulosclerosis [16]. Among the SNPs, rs11726091 is associated with the artery and aorta and is an eQTL of the MSX1 gene.

4. Discussion

This large-scale cohort study, over 12 years, found that hypertension occurred more in the low-renin group, among community-dwelling Koreans, and the genetic predisposition influenced the future hypertension incidence, according to renin concentration. Koreans have significantly higher salt intake than Westerners, and the frequency of low-renin hypertension may be higher than commonly known [7,42,43]. In a previous four-year follow-up study conducted in Korea, the low-renin group had a higher incidence of hypertension than the high-renin group, which was noticeable in the individuals with high salt intake [8]. Furthermore, it suggests that different genetic predispositions may affect the occurrence of hypertension, depending on the blood renin concentration.
In this study, there are two SNPs (rs11726091 and rs8137145) in regions associated with high renin level and hypertension. The first SNP (rs11726091) showed a p-value of 4.2 × 10−6, suggesting an association with increased hypertension incidence. The SNP was located on chromosome region 4p16.2, and there were no functional genes within ±50 kbp, with the nearest genes being MSX1 and CYTL1. The rs11726091 SNP region was associated with body mass index, lung function, and auto-immune diseases [17,18,19]. The eQTL was located in the MSX1 gene, expressed in artery and muscle tissues (GETX Portal), an indispensable homeodomain transcription factor for cardiac valve morphogenesis [44]. During embryogenesis, the encoded protein functions as a transcriptional repressor through interactions with the core transcription complex components and other homeoproteins. It also plays roles in the limb-pattern formation, craniofacial development, particularly odontogenesis, and tumor growth inhibition. Mutations in this gene, which was once known as homeobox 7, have been associated with non-syndromic cleft lip, with or without cleft palate, Witkop syndrome, Wolf–Hirschhorn syndrome, and autosomal dominant hypodontia [45,46,47,48]. The second SNP (rs8137145) showed a p-value of 6.4 × 10−6, suggesting an association with protection against hypertension. The SNP is located on chromosome 22q11.23, in an intron of the SGSM1 gene, which encodes small G protein signaling modulator 1. Diseases associated with SGSM1 include autosomal dominant adult-onset proximal spinal muscular atrophy and spinal muscular atrophy, with lower extremity predominance [49].
Interestingly, the most significant SNP was rs12336898 (p-value = 1.3 × 10−6), suggestive of the location of the incidence of hypertension in the low-renin group. The SNP was located on chromosome region 9q34.11, associated with the SPTAN1 gene. This study identifies a relationship between rs12336898 in the SPTAN1 gene and hypertension in low-renin subjects. Moreover, our results indicate that polymorphisms in the downstream region of the SPTAN1 gene contribute to the incidence of hypertension. Pathogenesis of the SPTAN1 gene and hypertension via the RAS support these findings.
Angiotensin activates calpains, which are calcium-activated proteases, associated with cardiovascular remodeling and inflammatory kidney diseases [50]. These proteins promote fibrosis through several types of pathogenesis. Calpains degrade IκBα in a specific sequence, leading to NF-κB activation and pro-inflammatory cytokines production [51]. They limit glucocorticoids by degrading HSP-90, reducing anti-inflammatory activity [52,53]. They also promote interleukin-1α maturation and activation and induce the onset of inflammatory processes and fibrosis, in the renal and cardiovascular systems [54,55]. Moreover, through AT-1 receptor signaling, angiotensin II induces vascular aging [56]. Therefore, angiotensin can promote vascular and kidney aging because of the breakdown products of spectrin in the kidney cortex and cardiomyocytes [57].
Spectrins play an essential role between the actin-based cytoskeleton and membrane and maintain the cellular structure [56,58]. αII spectrin (SPTAN1) is one of two α spectrin genes. Dysfunction of this gene is associated with alterations in axon initial segment formation and cortical lamination. II spectrin-deficient mice present significant structural and cellular phenotypes that result in structural remodeling and fibrosis [59]. If sodium uptake increases, the blood volume expands, and preload increases. Increased preload can result in vessel injury and cardiac remodeling. Therefore, the RAS is down-regulated, and the SPTAN1 gene is activated for compensation for vessel fibrosis [60].
Building upon this evidence, we describe a significant association between rs12336898 and hypertension in a community-based Korean cohort study. Furthermore, rs12336898 was related to the incidence of hypertension in the baseline low-renin group. Generally, new-onset hypertension increases if renin increases. However, low-renin activity can be an independent predictor of hypertension in Korean individuals [8]. A high level of sodium intake can decrease renin activity due to an increase in blood pressure [43]. Most patients with low-renin hypertension have sodium-volume-dependent hypertension, suggesting that high sodium uptake leads to fluid retention and decreased renin activity to compensate [61,62,63].
In the current study, rs1938859 in the transient receptor potential channel C6 (TRPC6) gene was also associated with hypertension. Polymorphisms in the upstream region of the TRPC6 gene could also result in new-onset hypertension through the RAS. The glomerular filtration barrier consists of podocytes, linked by the slit diaphragm, glomerular basement membrane, and endothelial cells. The slit diaphragm provides physical linkage to the podocytes by connecting podocyte foot processes [64,65]. Mutations in TRPC6 cause hereditary focal segmental glomerulosclerosis (FSGS), enhancing podocyte expression of wild-type [66,67]. TRPC6 is related to the slit diaphragm proteins nephrin and podocin, suggesting that TRPC6 signals the slit diaphragm [68]. Glomerular TRPC6 expression is elevated in proteinuric diseases, such as FSGS and membranous glomerulopathy [66]. Several TRPC6 mutations have been identified in the TRPC6-encoding gene [69,70].
TRPC6 is a receptor-operated channel activated by angiotensin II [71,72]. Angiotensin II is an essential contributor to the pathogenesis of the glomerular disease, and it is well known that ACE inhibition and angiotensin type 1 receptor (AT1R) block have antiproteinuric effects [73,74]. In nonrenal cells, Angiotensin II can activate TRPC6 [75,76]. Podocytes in the glomerulus can express both AT1R and AT2R, and Angiotensin II has detrimental effects on podocytes [77,78]. Therefore, TRPC6 influences podocytes in the glomerulus via angiotensin. When AT1R is overexpressed in podocytes, podocyte damage and glomerulosclerosis are induced in a rat model [79]. Furthermore, overexpression of renin in mice leads to podocyte damage and proteinuria. Such detrimental effects can be improved by treating experimental animals with angiotensin receptor blockers [80].
Some strengths and limitations require careful consideration and may affect the interpretation of the results of the present study. A major strength was that this study was conducted in a large population-based cohort. Furthermore, our findings are drawn from two replicate sample cohorts to validate the associations between relevant SNPs and hypertension. Replication is the gold standard for decreasing false-positive errors in genotype studies [81]. This study had some limitations that should also be acknowledged. First, we did not consider the effects of changes in renin level or the incidence of chronic diseases, such as hepatitis and cancer, which could affect the incidence of hypertension during the follow-up period. Second, the baseline study in KoGES did not contain information on diet, stress, physical activity in detail, which can affect new-onset hypertension. Comprehensively, the interactions between genes, epigenetics, and the environment are complex and influence gene expression [82,83]. Moreover, the data did not include information on which type of antihypertensive medications had been taken. The medications, such as angiotensin II receptor blocker and angiotensin converting enzyme inhibitor, can affect genetic polymorphism, depending on renin levels. Further prospective studies, including changes in renin level and other comorbidities, which can affect the incident hypertension, are needed to demonstrate the genetic polymorphisms as a predictive marker of hypertension more clearly.

5. Conclusions

In summary, we revealed an association between rs12336898 in SPTAN1 and hypertension in the low-renin group. Low-renin individuals could be at high risk of hypertension among Koreans, due to the genetic predisposition with high sodium uptake. Our results might be a valuable indicator for hypertension risk prediction and preventive measures, considering renin levels with genetic susceptibility.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcdd9040104/s1, Table S1: Series of SNPs for association analysis of hypertension and renin-related SNPs.

Author Contributions

D.-H.J. and K.-W.H.: study concept and design. All authors: acquisition, analysis, and interpretation of data and critical revision of the manuscript for important intellectual content. B.P. and S.-B.L.: drafting of the manuscript. All authors approved the submitted version. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

This study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Institutional Review Board of Yonsei University College of Medicine, Yongin Severance Hospital (IRB No: 2020-0040-002). The studies involving human participants were reviewed and approved by The Institutional Review Board of Yonsei University College of Medicine.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Data in this study were from the Korean Genome and Epidemiology Study (KoGES; 4851-302). These data are available through an online sharing service, under the permission of the division of epidemiology and health index in the Korea Centers for Disease Control and Prevention (KCDC), at http://www.nih.go.kr/NIH/eng/contents/ (accessed on 14 March 2022).

Acknowledgments

The authors would like to express their heartfelt gratitude to the participants and survey staff of the Korean Genome and Epidemiology Study (KoGES) for contributing to the present study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Roth, G.A.; Abate, D.; Abate, K.H.; Abay, S.M.; Abbafati, C.; Abbasi, N.; Abbastabar, H.; Abd-Allah, F.; Abdela, J.; Abdelalim, A. Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980–2017: A systematic analysis for the global burden of disease study 2017. Lancet 2018, 392, 1736–1788. [Google Scholar] [CrossRef] [Green Version]
  2. Mills, K.T.; Stefanescu, A.; He, J. The global epidemiology of hypertension. Nat. Rev. Nephrol. 2020, 16, 223–237. [Google Scholar] [CrossRef]
  3. Mills, K.T.; Bundy, J.D.; Kelly, T.N.; Reed, J.E.; Kearney, P.M.; Reynolds, K.; Chen, J.; He, J. Global disparities of hypertension prevalence and control: A systematic analysis of population-based studies from 90 countries. Circulation 2016, 134, 441–450. [Google Scholar] [CrossRef] [PubMed]
  4. Harrison-Bernard, L.M. The renal renin-angiotensin system. Adv. Physiol. Educ. 2009, 33, 270–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Ames, M.K.; Atkins, C.E.; Pitt, B. The renin-angiotensin-aldosterone system and its suppression. J. Vet. Intern. Med. 2019, 33, 363–382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Kotchen, T.A.; Guthrie, G.P., Jr. Renin-angiotensin-aldosterone and hypertension. Endocr. Rev. 1980, 1, 78–99. [Google Scholar] [CrossRef] [PubMed]
  7. Mulatero, P.; Verhovez, A.; Morello, F.; Veglio, F. Diagnosis and treatment of low-renin hypertension. Clin. Endocrinol. 2007, 67, 324–334. [Google Scholar] [CrossRef]
  8. Choo, E.H.; Ha, K.H.; Lee, S.-W.; Kim, H.C.; Park, S.; Lee, H.Y.; Ihm, S.-H. Low plasma renin activity is an independent predictor of near-term incidence of hypertension in asian populations. Clin. Exp. Hypertens. 2019, 41, 330–335. [Google Scholar] [CrossRef]
  9. Kim, Y.; Han, B.-G.; Group, K. Cohort profile: The Korean genome and epidemiology study (KoGES) consortium. Int. J. Epidemiol. 2017, 46, e20. [Google Scholar] [CrossRef] [PubMed]
  10. Kim, E.H.; Lim, J.H.; Kim, Y.H.; Park, Y.W. The relationship between aldosterone to renin ratio and ri value of the uterine artery in the preeclamptic patient vs. Normal pregnancy. Yonsei Med. J. 2008, 49, 138–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Sealey, J.E.; Gordon, R.D.; Mantero, F. Plasma renin and aldosterone measurements in low renin hypertensive states. Trends Endocrinol. Metab. 2005, 16, 86–91. [Google Scholar] [CrossRef] [PubMed]
  12. Kerner, W.; Brückel, J. Definition, classification and diagnosis of diabetes mellitus. Exp. Clin. Endocrinol. Diabetes 2014, 122, 384–386. [Google Scholar] [CrossRef] [Green Version]
  13. Carretero, O.A.; Oparil, S. Essential hypertension: Part i: Definition and etiology. Circulation 2000, 101, 329–335. [Google Scholar] [CrossRef]
  14. Cho, Y.S.; Go, M.J.; Kim, Y.J.; Heo, J.Y.; Oh, J.H.; Ban, H.-J.; Yoon, D.; Lee, M.H.; Kim, D.-J.; Park, M. A large-scale genome-wide association study of asian populations uncovers genetic factors influencing eight quantitative traits. Nat. Genet. 2009, 41, 527–534. [Google Scholar] [CrossRef] [PubMed]
  15. Consortium, H.P.-A.S.; Abdulla, M.A.; Ahmed, I.; Assawamakin, A.; Bhak, J.; Brahmachari, S.K.; Calacal, G.C.; Chaurasia, A.; Chen, C.-H.; Chen, J. Mapping human genetic diversity in asia. Science 2009, 326, 1541–1545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Winn, M.P.; Conlon, P.J.; Lynn, K.L.; Farrington, M.K.; Creazzo, T.; Hawkins, A.F.; Daskalakis, N.; Kwan, S.Y.; Ebersviller, S.; Burchette, J.L. A mutation in the trpc6 cation channel causes familial focal segmental glomerulosclerosis. Science 2005, 308, 1801–1804. [Google Scholar] [CrossRef] [Green Version]
  17. Pulit, S.L.; Stoneman, C.; Morris, A.P.; Wood, A.R.; Glastonbury, C.A.; Tyrrell, J.; Yengo, L.; Ferreira, T.; Marouli, E.; Ji, Y. Meta-analysis of genome-wide association studies for body fat distribution in 694 649 individuals of european ancestry. Hum. Mol. Genet. 2019, 28, 166–174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Lutz, S.M.; Cho, M.H.; Young, K.; Hersh, C.P.; Castaldi, P.J.; McDonald, M.-L.; Regan, E.; Mattheisen, M.; DeMeo, D.L.; Parker, M. A genome-wide association study identifies risk loci for spirometric measures among smokers of european and african ancestry. BMC Genet. 2015, 16, 138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Li, Y.R.; Li, J.; Zhao, S.D.; Bradfield, J.P.; Mentch, F.D.; Maggadottir, S.M.; Hou, C.; Abrams, D.J.; Chang, D.; Gao, F. Meta-analysis of shared genetic architecture across ten pediatric autoimmune diseases. Nat. Med. 2015, 21, 1018–1027. [Google Scholar] [CrossRef]
  20. Wilk, J.B.; Shrine, N.R.; Loehr, L.R.; Zhao, J.H.; Manichaikul, A.; Lopez, L.M.; Smith, A.V.; Heckbert, S.R.; Smolonska, J.; Tang, W. Genome-wide association studies identify chrna5/3 and htr4 in the development of airflow obstruction. Am. J. Respir. Crit. Care Med. 2012, 186, 622–632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Kichaev, G.; Bhatia, G.; Loh, P.-R.; Gazal, S.; Burch, K.; Freund, M.K.; Schoech, A.; Pasaniuc, B.; Price, A.L. Leveraging polygenic functional enrichment to improve gwas power. Am. J. Hum. Genet. 2019, 104, 65–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Jonsson, L.; Magnusson, T.; Thordarson, A.; Jonsson, T.; Geller, F.; Feenstra, B.; Melbye, M.; Nohr, E.; Vucic, S.; Dhamo, B. Rare and common variants conferring risk of tooth agenesis. J. Dent. Res. 2018, 97, 515–522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Jacob, J.; Vanholme, B.; Haegeman, A.; Gheysen, G. Four transthyretin-like genes of the migratory plant-parasitic nematode radopholus similis: Members of an extensive nematode-specific family. Gene 2007, 402, 9–19. [Google Scholar] [CrossRef]
  24. Gericke, B.; Raila, J.; Sehouli, J.; Haebel, S.; Könsgen, D.; Mustea, A.; Schweigert, F.J. Microheterogeneity of transthyretin in serum and ascitic fluid of ovarian cancer patients. BMC Cancer 2005, 5, 133. [Google Scholar] [CrossRef] [Green Version]
  25. Schweigert, F.J.; Sehouli, J. Transthyretin, a biomarker for nutritional status and ovarian cancer. Cancer Res. 2005, 65, 1114. [Google Scholar]
  26. Rietveld, C.A.; Medland, S.E.; Derringer, J.; Yang, J.; Esko, T.; Martin, N.W.; Westra, H.-J.; Shakhbazov, K.; Abdellaoui, A.; Agrawal, A. Gwas of 126,559 individuals identifies genetic variants associated with educational attainment. Science 2013, 340, 1467–1471. [Google Scholar] [CrossRef] [Green Version]
  27. Pilling, L.C.; Atkins, J.L.; Duff, M.O.; Beaumont, R.N.; Jones, S.E.; Tyrrell, J.; Kuo, C.-L.; Ruth, K.S.; Tuke, M.A.; Yaghootkar, H. Red blood cell distribution width: Genetic evidence for aging pathways in 116,666 volunteers. PLoS ONE 2017, 12, e0185083. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Briggs, F.; Shao, X.; Goldstein, B.A.; Oksenberg, J.R.; Barcellos, L.F.; De Jager, P.L. Genome-wide association study of severity in multiple sclerosis. Genes Immun. 2011, 12, 615–625. [Google Scholar]
  29. Evangelou, E.; Warren, H.R.; Mosen-Ansorena, D.; Mifsud, B.; Pazoki, R.; Gao, H.; Ntritsos, G.; Dimou, N.; Cabrera, C.P.; Karaman, I. Genetic analysis of over 1 million people identifies 535 new loci associated with blood pressure traits. Nat. Genet. 2018, 50, 1412–1425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Dashti, H.S.; Jones, S.E.; Wood, A.R.; Lane, J.M.; Van Hees, V.T.; Wang, H.; Rhodes, J.A.; Song, Y.; Patel, K.; Anderson, S.G. Genome-wide association study identifies genetic loci for self-reported habitual sleep duration supported by accelerometer-derived estimates. Nat. Commun. 2019, 10, 1100. [Google Scholar] [CrossRef] [Green Version]
  31. Karlsson Linnér, R.; Biroli, P.; Kong, E.; Meddens, S.F.W.; Wedow, R.; Fontana, M.A.; Lebreton, M.; Tino, S.P.; Abdellaoui, A.; Hammerschlag, A.R. Genome-wide association analyses of risk tolerance and risky behaviors in over 1 million individuals identify hundreds of loci and shared genetic influences. Nat. Genet. 2019, 51, 245–257. [Google Scholar] [CrossRef]
  32. Serie, D.J.; Crook, J.E.; Necela, B.M.; Dockter, T.J.; Wang, X.; Asmann, Y.W.; Fairweather, D.; Bruno, K.A.; Colon-Otero, G.; Perez, E.A. Genome-wide association study of cardiotoxicity in ncctg n9831 (alliance) adjuvant trastuzumab trial. Pharm. Genom. 2017, 27, 378. [Google Scholar] [CrossRef]
  33. Shrine, N.; Guyatt, A.L.; Erzurumluoglu, A.M.; Jackson, V.E.; Hobbs, B.D.; Melbourne, C.A.; Batini, C.; Fawcett, K.A.; Song, K.; Sakornsakolpat, P. New genetic signals for lung function highlight pathways and chronic obstructive pulmonary disease associations across multiple ancestries. Nat. Genet. 2019, 51, 481–493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Lotta, L.A.; Wittemans, L.B.; Zuber, V.; Stewart, I.D.; Sharp, S.J.; Luan, J.A.; Day, F.R.; Li, C.; Bowker, N.; Cai, L. Association of genetic variants related to gluteofemoral vs abdominal fat distribution with type 2 diabetes, coronary disease, and cardiovascular risk factors. JAMA 2018, 320, 2553–2563. [Google Scholar] [CrossRef]
  35. Sherva, R.; Tripodis, Y.; Bennett, D.A.; Chibnik, L.B.; Crane, P.K.; de Jager, P.L.; Farrer, L.A.; Saykin, A.J.; Shulman, J.M.; Naj, A. Genome-wide association study of the rate of cognitive decline in alzheimer’s disease. Alzheimer’s Dement. 2014, 10, 45–52. [Google Scholar] [CrossRef] [Green Version]
  36. Lee, J.J.; Wedow, R.; Okbay, A.; Kong, E.; Maghzian, O.; Zacher, M.; Nguyen-Viet, T.A.; Bowers, P.; Sidorenko, J.; Karlsson Linnér, R. Gene discovery and polygenic prediction from a genome-wide association study of educational attainment in 1.1 million individuals. Nat. Genet. 2018, 50, 1112–1121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Baselmans, B.M.; Jansen, R.; Ip, H.F.; van Dongen, J.; Abdellaoui, A.; van de Weijer, M.P.; Bao, Y.; Smart, M.; Kumari, M.; Willemsen, G. Multivariate genome-wide analyses of the well-being spectrum. Nat. Genet. 2019, 51, 445–451. [Google Scholar] [CrossRef]
  38. Deng, X.; Sabino, E.C.; Cunha-Neto, E.; Ribeiro, A.L.; Ianni, B.; Mady, C.; Busch, M.P.; Seielstad, M.; the REDSII Chagas study group from the NHLBI Retrovirus Epidemiology Donor Study-II (REDS-II); International Component. Genome wide association study (gwas) of chagas cardiomyopathy in trypanosoma cruzi seropositive subjects. PLoS ONE 2013, 8, e79629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Ahola-Olli, A.V.; Würtz, P.; Havulinna, A.S.; Aalto, K.; Pitkänen, N.; Lehtimäki, T.; Kähönen, M.; Lyytikäinen, L.-P.; Raitoharju, E.; Seppälä, I. Genome-wide association study identifies 27 loci influencing concentrations of circulating cytokines and growth factors. Am. J. Hum. Genet. 2017, 100, 40–50. [Google Scholar] [CrossRef] [Green Version]
  40. Zhou, H.; Cheng, Z.; Bass, N.; Krystal, J.H.; Farrer, L.A.; Kranzler, H.R.; Gelernter, J. Genome-wide association study identifies glutamate ionotropic receptor gria4 as a risk gene for comorbid nicotine dependence and major depression. Transl. Psychiatry 2018, 8, 208. [Google Scholar] [CrossRef] [Green Version]
  41. Jawinski, P.; Kirsten, H.; Sander, C.; Spada, J.; Ulke, C.; Huang, J.; Burkhardt, R.; Scholz, M.; Hensch, T.; Hegerl, U. Human brain arousal in the resting state: A genome-wide association study. Mol. Psychiatry 2019, 24, 1599–1609. [Google Scholar] [CrossRef]
  42. Lee, H.-S.; Duffey, K.J.; Popkin, B.M. Sodium and potassium intake patterns and trends in south korea. J. Hum. Hypertens. 2013, 27, 298–303. [Google Scholar] [CrossRef]
  43. Group, I.C.R. Intersalt: An international study of electrolyte excretion and blood pressure. Results for 24 h urinary sodium and potassium excretion. BMJ Br. Med. J. 1988, 297, 319–328. [Google Scholar]
  44. Chen, Y.-H.; Ishii, M.; Sucov, H.M.; Maxson, R.E. Msx1 and msx2 are required for endothelial-mesenchymal transformation of the atrioventricular cushions and patterning of the atrioventricular myocardium. BMC Dev. Biol. 2008, 8, 75. [Google Scholar] [CrossRef] [Green Version]
  45. Kim, N.Y.; Kim, Y.H.; Park, J.W.; Baek, S.-H. Association between msx1 snps and nonsyndromic cleft lip with or without cleft palate in the korean population. J. Korean Med. Sci. 2013, 28, 522–526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Jumlongras, D.; Bei, M.; Stimson, J.M.; Wang, W.-F.; DePalma, S.R.; Seidman, C.E.; Felbor, U.; Maas, R.; Seidman, J.G.; Olsen, B.R. A nonsense mutation in msx1 causes witkop syndrome. Am. J. Hum. Genet. 2001, 69, 67–74. [Google Scholar] [CrossRef] [Green Version]
  47. Rjiba, K.; Ayech, H.; Kraiem, O.; Slimani, W.; Jelloul, A.; Ben Hadj Hmida, I.; Mahdhaoui, N.; Saad, A.; Mougou-Zerelli, S. Disorders of sex development in wolf–hirschhorn syndrome: A genotype–phenotype correlation and msx1 as candidate gene. Mol. Cytogenet. 2021, 14, 12. [Google Scholar] [CrossRef]
  48. Zheng, J.; Yu, M.; Liu, H.; Cai, T.; Feng, H.; Liu, Y.; Han, D. Novel msx1 variants identified in families with nonsyndromic oligodontia. Int. J. Oral Sci. 2021, 13, 12. [Google Scholar] [CrossRef] [PubMed]
  49. Penttilä, S.; Jokela, M.; Hackman, P.; Maija Saukkonen, A.; Toivanen, J.; Udd, B. Autosomal dominant late-onset spinal motor neuronopathy is linked to a new locus on chromosome 22q11.2-q13.2. Eur. J. Hum. Genet. 2012, 20, 1193–1196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Goll, D.E.; Thompson, V.F.; Li, H.; Wei, W.; Cong, J. The calpain system. Physiol. Rev. 2003, 83, 731–801. [Google Scholar] [CrossRef]
  51. Lin, Y.-C.; Brown, K.; Siebenlist, U. Activation of nf-kappa b requires proteolysis of the inhibitor i kappa b-alpha: Signal-induced phosphorylation of i kappa b-alpha alone does not release active nf-kappa b. Proc. Natl. Acad. Sci. USA 1995, 92, 552–556. [Google Scholar] [CrossRef] [Green Version]
  52. Nuzzi, P.A.; Senetar, M.A.; Huttenlocher, A. Asymmetric localization of calpain 2 during neutrophil chemotaxis. Mol. Biol. Cell 2007, 18, 795–805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Bellocq, A.; Doublier, S.; Suberville, S.; Perez, J.; Escoubet, B.; Fouqueray, B.; Puyol, D.R.g.; Baud, L. Somatostatin increases glucocorticoid binding and signaling in macrophages by blocking the calpain-specific cleavage of hsp 90. J. Biol. Chem. 1999, 274, 36891–36896. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kobayashi, Y.; Yamamoto, K.; Saido, T.; Kawasaki, H.; Oppenheim, J.J.; Matsushima, K. Identification of calcium-activated neutral protease as a processing enzyme of human interleukin 1 alpha. Proc. Natl. Acad. Sci. USA 1990, 87, 5548–5552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Letavernier, E.; Perez, J.; Bellocq, A.; Mesnard, L.; de Castro Keller, A.; Haymann, J.-P.; Baud, L. Targeting the calpain/calpastatin system as a new strategy to prevent cardiovascular remodeling in angiotensin ii–induced hypertension. Circ. Res. 2008, 102, 720–728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Machnicka, B.; Czogalla, A.; Hryniewicz-Jankowska, A.; Bogusławska, D.M.; Grochowalska, R.; Heger, E.; Sikorski, A.F. Spectrins: A structural platform for stabilization and activation of membrane channels, receptors and transporters. Biochim. et Biophys. Acta (BBA)-Biomembr. 2014, 1838, 620–634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Hanouna, G.; Mesnard, L.; Vandermeersch, S.; Perez, J.; Placier, S.; Haymann, J.-P.; Campagne, F.; Moroch, J.; Bataille, A.; Baud, L. Specific calpain inhibition protects kidney against inflammaging. Sci. Rep. 2017, 7, 8016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Tohyama, J.; Nakashima, M.; Nabatame, S.; Miyata, R.; Rener-Primec, Z.; Kato, M.; Matsumoto, N.; Saitsu, H. Sptan1 encephalopathy: Distinct phenotypes and genotypes. J. Hum. Genet. 2015, 60, 167–173. [Google Scholar] [CrossRef]
  59. Lubbers, E.R.; Murphy, N.P.; Musa, H.; Huang, C.Y.-M.; Gupta, R.; Price, M.V.; Han, M.; Daoud, G.; Gratz, D.; El Refaey, M. Defining new mechanistic roles for αii spectrin in cardiac function. J. Biol. Chem. 2019, 294, 9576–9591. [Google Scholar] [CrossRef]
  60. Wang, X.; Gao, G.; Liu, J.; Guo, R.; Lin, Y.; Chu, Y.; Han, F.; Zhang, W.; Bai, Y. Identification of differentially expressed genes induced by angiotensin ii in rat cardiac fibroblasts. Clin. Exp. Pharmacol. Physiol. 2006, 33, 41–46. [Google Scholar] [CrossRef]
  61. Levy, S.B.; Lilley, J.J.; Frigon, R.P.; Stone, R.A. Urinary kallikrein and plasma renin activity as determinants of renal blood flow. The influence of race and dietary sodium intake. J. Clin. Investig. 1977, 60, 129–138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Luft, F.; Grim, C.; Higgins, J., Jr.; Weinberger, M. Differences in response to sodium administration in normotensive white and black subjects. J. Lab. Clin. Med. 1977, 90, 555–562. [Google Scholar] [PubMed]
  63. Alderman, M.H.; Madhavan, S.; Ooi, W.L.; Cohen, H.; Sealey, J.E.; Laragh, J.H. Association of the renin-sodium profile with the risk of myocardial infarction in patients with hypertension. N. Engl. J. Med. 1991, 324, 1098–1104. [Google Scholar] [CrossRef] [PubMed]
  64. Faul, C.; Asanuma, K.; Yanagida-Asanuma, E.; Kim, K.; Mundel, P. Actin up: Regulation of podocyte structure and function by components of the actin cytoskeleton. Trends Cell Biol. 2007, 17, 428–437. [Google Scholar] [CrossRef]
  65. Reiser, J.; Polu, K.R.; Möller, C.C.; Kenlan, P.; Altintas, M.M.; Wei, C.; Faul, C.; Herbert, S.; Villegas, I.; Avila-Casado, C. Trpc6 is a glomerular slit diaphragm-associated channel required for normal renal function. Nat. Genet. 2005, 37, 739–744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Möller, C.C.; Wei, C.; Altintas, M.M.; Li, J.; Greka, A.; Ohse, T.; Pippin, J.W.; Rastaldi, M.P.; Wawersik, S.; Schiavi, S. Induction of trpc6 channel in acquired forms of proteinuric kidney disease. J. Am. Soc. Nephrol. 2007, 18, 29–36. [Google Scholar] [CrossRef]
  67. Zheng, S.; Carlson, E.C.; Yang, L.; Kralik, P.M.; Huang, Y.; Epstein, P.N. Podocyte-specific overexpression of the antioxidant metallothionein reduces diabetic nephropathy. J. Am. Soc. Nephrol. 2008, 19, 2077–2085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Möller, C.C.; Flesche, J.; Reiser, J. Sensitizing the slit diaphragm with trpc6 ion channels. J. Am. Soc. Nephrol. 2009, 20, 950–953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Zhu, B.; Chen, N.; Wang, Z.-h.; Pan, X.-x.; Ren, H.; Zhang, W.; Wang, W.-m. Identification and functional analysis of a novel trpc6 mutation associated with late onset familial focal segmental glomerulosclerosis in chinese patients. Mutat. Res./Fundam. Mol. Mech. Mutagenesis 2009, 664, 84–90. [Google Scholar] [CrossRef]
  70. Santín, S.; Ars, E.; Rossetti, S.; Salido, E.; Silva, I.; García-Maset, R.; Giménez, I.; Ruíz, P.; Mendizábal, S.; Luciano Nieto, J. Trpc6 mutational analysis in a large cohort of patients with focal segmental glomerulosclerosis. Nephrol. Dial. Transplant. 2009, 24, 3089–3096. [Google Scholar] [CrossRef] [Green Version]
  71. Hofmann, T.; Obukhov, A.G.; Schaefer, M.; Harteneck, C.; Gudermann, T.; Schultz, G. Direct activation of human trpc6 and trpc3 channels by diacylglycerol. Nature 1999, 397, 259–263. [Google Scholar] [CrossRef] [PubMed]
  72. Kuwahara, K.; Wang, Y.; McAnally, J.; Richardson, J.A.; Bassel-Duby, R.; Hill, J.A.; Olson, E.N. Trpc6 fulfills a calcineurin signaling circuit during pathologic cardiac remodeling. J. Clin. Investig. 2006, 116, 3114–3126. [Google Scholar] [CrossRef] [PubMed]
  73. Ruggenenti, P.; Perna, A.; Gherardi, G.; Garini, G.; Zoccali, C.; Salvadori, M.; Scolari, F.; Schena, F.P.; Remuzzi, G. Renoprotective properties of ace-inhibition in non-diabetic nephropathies with non-nephrotic proteinuria. Lancet 1999, 354, 359–364. [Google Scholar] [CrossRef]
  74. Ruggenenti, P.; Perna, A.; Loriga, G.; Ganeva, M.; Ene-Iordache, B.; Turturro, M.; Lesti, M.; Perticucci, E.; Chakarski, I.N.; Leonardis, D. Blood-pressure control for renoprotection in patients with non-diabetic chronic renal disease (rein-2): Multicentre, randomised controlled trial. Lancet 2005, 365, 939–946. [Google Scholar] [CrossRef]
  75. Saleh, S.; Albert, A.P.; Peppiatt, C.; Large, W. Angiotensin ii activates two cation conductances with distinct trpc1 and trpc6 channel properties in rabbit mesenteric artery myocytes. J. Physiol. 2006, 577, 479–495. [Google Scholar] [CrossRef] [PubMed]
  76. Onohara, N.; Nishida, M.; Inoue, R.; Kobayashi, H.; Sumimoto, H.; Sato, Y.; Mori, Y.; Nagao, T.; Kurose, H. Trpc3 and trpc6 are essential for angiotensin ii-induced cardiac hypertrophy. EMBO J. 2006, 25, 5305–5316. [Google Scholar] [CrossRef]
  77. Van den Hoven, M.J.; Waanders, F.; Rops, A.L.; Kramer, A.B.; Van Goor, H.; Berden, J.H.; Navis, G.; Van der Vlag, J. Regulation of glomerular heparanase expression by aldosterone, angiotensin ii and reactive oxygen species. Nephrol. Dial. Transplant. 2009, 24, 2637–2645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Harrison-Bernard, L.M.; Navar, L.G.; Ho, M.; Vinson, G.; El-Dahr, S. Immunohistochemical localization of ang ii at1 receptor in adult rat kidney using a monoclonal antibody. Am. J. Physiol.-Ren. Physiol. 1997, 273, F170–F177. [Google Scholar] [CrossRef] [PubMed]
  79. Hoffmann, S.; Podlich, D.; Hähnel, B.; Kriz, W.; Gretz, N. Angiotensin ii type 1 receptor overexpression in podocytes induces glomerulosclerosis in transgenic rats. J. Am. Soc. Nephrol. 2004, 15, 1475–1487. [Google Scholar] [CrossRef] [Green Version]
  80. Huby, A.-C.; Rastaldi, M.-P.; Caron, K.; Smithies, O.; Dussaule, J.-C.; Chatziantoniou, C. Restoration of podocyte structure and improvement of chronic renal disease in transgenic mice overexpressing renin. PLoS ONE 2009, 4, e6721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Chanock, S.J.; Manolio, T.; Boehnke, M.; Boerwinkle, E.; Hunter, D.J.; Thomas, G.; Hirschhorn, J.N.; Abecasis, G.; Altshuler, D.; Bailey-Wilson, J.E. Replicating genotype–phenotype associations. Nature 2007, 447, 655. [Google Scholar]
  82. Rönn, T.; Volkov, P.; Davegårdh, C.; Dayeh, T.; Hall, E.; Olsson, A.H.; Nilsson, E.; Tornberg, Å.; Nitert, M.D.; Eriksson, K.-F. A six months exercise intervention influences the genome-wide DNA methylation pattern in human adipose tissue. PLoS Genet. 2013, 9, e1003572. [Google Scholar] [CrossRef] [PubMed]
  83. Lindholm, M.E.; Marabita, F.; Gomez-Cabrero, D.; Rundqvist, H.; Ekström, T.J.; Tegnér, J.; Sundberg, C.J. An integrative analysis reveals coordinated reprogramming of the epigenome and the transcriptome in human skeletal muscle after training. Epigenetics 2014, 9, 1557–1569. [Google Scholar] [CrossRef] [PubMed]
Table 1. Baseline characteristics of the study population.
Table 1. Baseline characteristics of the study population.
Low Renin GroupHigh Renin Group
NormotensiveNew-Onset Hypertensionp-ValueNormotensiveNew-Onset Hypertensionp-Value
No. subjects (%)23721294 1135410
Age (years)49.2 ± 7.953.9 ± 8.8<0.00149.1 ± 8.251.9 ± 8.8<0.001
Sex, n (%) 0.069 0.009
Male944 (39.8)555 (42.9) 667 (58.8)271 (66.1)
Female1428 (60.2)739 (57.1) 468 (41.2)139 (33.9)
Body mass index (kg/m2)24.0 ± 2.924.9 ± 3.0<0.00123.8 ± 2.924.5 ± 3.2<0.001
Fasting plasma glucose (mg/dL)88.9 ± 16.691.8 ± 20.7<0.00192.2 ± 23.196.5 ± 35.00.007
Total cholesterol (mg/dL)193.0 ± 34.7196.1 ± 34.80.016199.6 ± 34.5202.4 ± 38.20.173
Triglycerides (mg/dL)132.4 ± 92.0150.9 ± 99.7<0.001141.1 ± 97.9168.9 ± 117.8<0.001
HDL cholesterol (mg/dL)50.2 ± 11.748.4 ± 11.2<0.00150.3 ± 11.949.7 ± 12.20.379
Systolic blood pressure (mmHg)120.8 ± 19.7121.1 ± 19.30.663119.8 ± 18.0120.6 ± 18.90.449
Diastolic blood pressure (mmHg)88.9 ± 16.679.9 ± 12.40.33779.8 ± 11.479.3 ± 11.90.450
Current smoker, n (%)509 (21.4)323 (24.9)0.015361 (31.8)143 (34.9)0.256
Diabetes, n (%)165 (7.0)167 (13.0)<0.001103 (9.1)64 (15.6)<0.001
Data are presented as mean ± SD, proportion, or median (interquartile range) for skewed variables. p-values were calculated by one-way ANOVA for continuous variables and chi-square test for categorical variables.
Table 2. Top significant SNP of three hypertension case-control GWASs in the association cluster locus.
Table 2. Top significant SNP of three hypertension case-control GWASs in the association cluster locus.
CHRSNPBPA1Total Hypertension Cases
(n= 1704) and Controls (n = 3507)
Low Renin Group Cases
(n = 1294) and Controls (n = 2372)
High Renin Group Cases
(n = 410) and Controls (n = 1135)
Associations by Renin GroupLocus TopCluster Pick
ORL95U95pORL95U95PORL95U95p
4rs117260914,952,412C1.141.041.243.3 × 10−31.030.931.145.5 × 10−11.481.251.754.2 × 10−6HTC
4rs17038966109,296,214A0.720.600.862.1 × 10−40.620.500.766.0 × 10−61.050.761.447.7 × 10−1LTC
4rs145286444167,264,137A1.331.151.552.1 × 10−41.541.281.842.9 × 10−60.970.721.308.2 × 10−1LTC
5rs2118663153,340,145C1.261.101.441.0 × 10−31.461.241.713.8 × 10−60.810.611.081.5 × 10−1LTC
9rs12336898131,305,177T0.850.780.934.8 × 10−40.770.690.861.3 × 10−61.090.931.292.9 × 10−1LTC
11rs1938859101,547,723T1.381.201.584.6 × 10−61.401.191.644.7 × 10−51.351.041.752.3 × 10−2MTC
12rs796821894,189,185C0.740.650.844.1 × 10−60.750.650.882.3 × 10−40.680.530.883.9 × 10−3MTC
18rs11724640132,975,483T1.711.372.141.8 × 10−61.571.212.046.6 × 10−42.091.383.165.1 × 10−4MTC
22rs813714525,213,480T0.830.760.916.6 × 10−50.910.821.017.7 × 10−20.670.570.806.4 × 10−6HTC
CHR, chromosome; SNP, single nucleotide polymorphism; BP, base pair; A1, minor allele; OR, odds ratio; L95, lower 95% confidence interval; U95, upper 95% confidence interval; p, p-value; L, low-renin group; H, high-renin group; M, both groups; T, top significant SNP in the locus; C, cluster more than three SNPs in each locus with p-values < 1 × 10−5, adjusted for sex, age, and BMI.
Table 3. Interpretations of significant SNPs.
Table 3. Interpretations of significant SNPs.
CHRSNPBPA1Renin GroupLocusNearby Genes (±100 kbp)FunctionPrevious GWAS (±100 kbp)eQTL
4rs117260914,952,412CH4p16.2MSX1
CYTL1
IntergenicBody mass index, waist-hip ratio, post bronchodilator FEV1/FVC ratio, lung function (FVC), pediatric autoimmune diseases [17,18,19]MSX1 (artery-aorta, muscle-skeletal)
4rs17038966109,296,214AL4q25No geneIntergenicLung function (FVC), hair colour, tooth agenesis, airflow obstruction [20,21,22]
4rs145286444167,264,137AL4q32.3TTL1IntergenicVelopharyngeal dysfunction, serous borderline ovarian cancer, obesity-related traits [23,24,25]
5rs2118663153,340,145CL5q33.2FAM114A2
MFAP3
3′ downstreamEducational attainment (MTAG), red cell distribution width, multiple sclerosis [26,27,28]FAM114A2 (muscle-skeletal, whole-blood, heart-atrial appendage, brain-frontal-cortex, adipose-subcutaneous, cultured-fibroblasts)
9rs12336898131,305,177TL9q34.11ODF2
GLE1
SPTAN1
3′ downstreamAxial length, pulse pressure [29]
11rs1938859101,547,723TM11q22.1TRPC65′ upstreamSleep duration, general risk tolerance (MTAG), change in LVEF in response to paclitaxel and trastuzumab in HER2+ breast cancer [30,31,32]TRPC6 (colon-sigmoid, pancreas, heart-left-ventricle, nerve-tibial)
12rs796821894,189,185CM12q22CRADDIntronLung function (FVC), waist-to-hip ratio, Alzheimer’s disease (cognitive decline) [33,34,35]
18rs11724640132,975,483TM18q12.2ZNF271P, ZNF396, INO80CIntergenicHighest math class taken (MTAG), Parkinson’s disease, well-being spectrum (multivariate analysis), neuroticism, life satisfaction, parasitaemia in Tripanosoma cruzi seropositivity [36,37,38]
22rs813714525,213,480TH22q11.23PIWIL3
SGSM1
Intron of SGSM1Macrophage migration inhibitory factor levels, nicotine dependence and major depression (severity of comorbidity), resting-state electroencephalogram vigilance [39,40,41]SGSM1 (muscle-skeletal, lung, adipose-visceral, spleen, pancreas, whole-blood)
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lee, S.-B.; Park, B.; Hong, K.-W.; Jung, D.-H. Genome-Wide Association of New-Onset Hypertension According to Renin Concentration: The Korean Genome and Epidemiology Cohort Study. J. Cardiovasc. Dev. Dis. 2022, 9, 104. https://doi.org/10.3390/jcdd9040104

AMA Style

Lee S-B, Park B, Hong K-W, Jung D-H. Genome-Wide Association of New-Onset Hypertension According to Renin Concentration: The Korean Genome and Epidemiology Cohort Study. Journal of Cardiovascular Development and Disease. 2022; 9(4):104. https://doi.org/10.3390/jcdd9040104

Chicago/Turabian Style

Lee, Sung-Bum, Byoungjin Park, Kyung-Won Hong, and Dong-Hyuk Jung. 2022. "Genome-Wide Association of New-Onset Hypertension According to Renin Concentration: The Korean Genome and Epidemiology Cohort Study" Journal of Cardiovascular Development and Disease 9, no. 4: 104. https://doi.org/10.3390/jcdd9040104

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop