201
|
Zhang M, Chen J, Yin Z, Wang L, Peng L. The association between depression and metabolic syndrome and its components: a bidirectional two-sample Mendelian randomization study. Transl Psychiatry 2021; 11:633. [PMID: 34903730 PMCID: PMC8668963 DOI: 10.1038/s41398-021-01759-z] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 12/31/2022] Open
Abstract
Observational studies suggested a bidirectional correlation between depression and metabolic syndrome (MetS) and its components. However, the causal associations between them remained unclear. We aimed to investigate whether genetically predicted depression is related to the risk of MetS and its components, and vice versa. We performed a bidirectional two-sample Mendelian randomization (MR) study using summary-level data from the most comprehensive genome-wide association studies (GWAS) of depression (n = 2,113,907), MetS (n = 291,107), waist circumference (n = 462,166), hypertension (n = 463,010) fasting blood glucose (FBG, n = 281,416), triglycerides (n = 441,016), high-density lipoprotein cholesterol (HDL-C, n = 403,943). The random-effects inverse-variance weighted (IVW) method was applied as the primary method. The results identified that genetically predicted depression was significantly positive associated with risk of MetS (OR: 1.224, 95% CI: 1.091-1.374, p = 5.58 × 10-4), waist circumference (OR: 1.083, 95% CI: 1.027-1.143, p = 0.003), hypertension (OR: 1.028, 95% CI: 1.016-1.039, p = 1.34 × 10-6) and triglycerides (OR: 1.111, 95% CI: 1.060-1.163, p = 9.35 × 10-6) while negative associated with HDL-C (OR: 0.932, 95% CI: 0.885-0.981, p = 0.007) but not FBG (OR: 1.010, 95% CI: 0.986-1.034, p = 1.34). No causal relationships were identified for MetS and its components on depression risk. The present MR analysis strength the evidence that depression is a risk factor for MetS and its components (waist circumference, hypertension, FBG, triglycerides, and HDL-C). Early diagnosis and prevention of depression are crucial in the management of MetS and its components.
Collapse
Affiliation(s)
- Min Zhang
- grid.203458.80000 0000 8653 0555School of Public Health and Management, Chongqing Medical University, Chongqing, 400016 China
| | - Jing Chen
- grid.452206.70000 0004 1758 417XDepartment of Anesthesia and Pain Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Zhiqun Yin
- Department of Psychiatry and Psychology, No.964 Hospital of People’s Liberation Army, Changchun City, 130026 Jilin Province China
| | - Lanbing Wang
- Division of medical affairs, The First Affiliated Hospital of Army Military Medical University, Chongqing, 400038 China
| | - Lihua Peng
- Department of Anesthesia and Pain Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
202
|
Olczak KJ, Taylor-Bateman V, Nicholls HL, Traylor M, Cabrera CP, Munroe PB. Hypertension genetics past, present and future applications. J Intern Med 2021; 290:1130-1152. [PMID: 34166551 DOI: 10.1111/joim.13352] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Essential hypertension is a complex trait where the underlying aetiology is not completely understood. Left untreated it increases the risk of severe health complications including cardiovascular and renal disease. It is almost 15 years since the first genome-wide association study for hypertension, and after a slow start there are now over 1000 blood pressure (BP) loci explaining ∼6% of the single nucleotide polymorphism-based heritability. Success in discovery of hypertension genes has provided new pathological insights and drug discovery opportunities and translated to the development of BP genetic risk scores (GRSs), facilitating population disease risk stratification. Comparing highest and lowest risk groups shows differences of 12.9 mm Hg in systolic-BP with significant differences in risk of hypertension, stroke, cardiovascular disease and myocardial infarction. GRSs are also being trialled in antihypertensive drug responses. Drug targets identified include NPR1, for which an agonist drug is currently in clinical trials. Identification of variants at the PHACTR1 locus provided insights into regulation of EDN1 in the endothelin pathway, which is aiding the development of endothelin receptor EDNRA antagonists. Drug re-purposing opportunities, including SLC5A1 and canagliflozin (a type-2 diabetes drug), are also being identified. In this review, we present key studies from the past, highlight current avenues of research and look to the future focusing on gene discovery, epigenetics, gene-environment interactions, GRSs and drug discovery. We evaluate limitations affecting BP genetics, including ancestry bias and discuss streamlining of drug target discovery and applications for treating and preventing hypertension, which will contribute to tailored precision medicine for patients.
Collapse
Affiliation(s)
- Kaya J Olczak
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Victoria Taylor-Bateman
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Hannah L Nicholls
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Centre for Translational Bioinformatics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Matthew Traylor
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Claudia P Cabrera
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Centre for Translational Bioinformatics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,NIHR Barts Biomedical Centre, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Patricia B Munroe
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,NIHR Barts Biomedical Centre, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
203
|
Kaur H, Crawford DC, Liang J, Benchek P, COGENT BP Consortium, Zhu X, Kallianpur AR, Bush WS. Replication of European hypertension associations in a case-control study of 9,534 African Americans. PLoS One 2021; 16:e0259962. [PMID: 34793544 PMCID: PMC8601554 DOI: 10.1371/journal.pone.0259962] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 10/29/2021] [Indexed: 12/01/2022] Open
Abstract
OBJECTIVE Hypertension is more prevalent in African Americans (AA) than other ethnic groups. Genome-wide association studies (GWAS) have identified loci associated with hypertension and other cardio-metabolic traits like type 2 diabetes, coronary artery disease, and body mass index (BMI), however the AA population is underrepresented in these studies. In this study, we examined a large AA cohort for the generalizability of 14 Metabochip array SNPs with previously reported European hypertension associations. METHODS To evaluate associations, we analyzed genotype data of 14 SNPs for their associations with a diagnosis of hypertension, systolic blood pressure (SBP), and diastolic blood pressure (DBP) in a case-control study of an AA population (N = 9,534). We also performed an age-stratified analysis (>30, 30≥59 and ≥60 years) following the hypertension definition described by the 8th Joint National Committee (JNC). Associations were adjusted for BMI, age, age2, sex, clinical confounders, and genetic ancestry using multivariable regression models to estimate odds ratios (ORs) and beta-coefficients. Analyses stratified by sex were also conducted. Meta-analyses (including both BioVU and COGENT-BP cohorts) were performed using a random-effects model. RESULTS We found rs880315 to be associated with systolic hypertension (SBP≥140 mmHg) in the entire cohort (OR = 1.14, p = 0.003) and within women only (OR = 1.16, p = 0.012). Variant rs17080093 associated with lower SBP and DBP (β = -2.99, p = 0.0352 and - β = 1.69, p = 0.0184) among younger individuals, particularly in younger women (β = -3.92, p = 0.0025 and β = -1.87, p = 0.0241 for SBP and DBP respectively). SNP rs1530440 associated with higher SBP and DBP measurements (younger individuals β = 4.1, p = 0.039 and β = 2.5, p = 0.043 for SBP and DBP; (younger women β = 4.5, p = 0.025 and β = 2.9, p = 0.028 for SBP and DBP), and hypertension risk in older women (OR = 1.4, p = 0.050). rs16948048 increases hypertension risk in younger individuals (OR = 1.31, p = 0.011). Among mid-age women rs880315 associated with higher risk of hypertension (OR = 1.20, p = 0.027). rs1361831 associated with DBP (β = -1.96, p = 0.02) among individuals older than 60 years. rs3096277 increases hypertension risk among older individuals (OR = 1.26 p = 0.0015), however, this variant also reduces SBP among younger women (β = -2.63, p = 0.0102). CONCLUSION These findings suggest that European-descent and AA populations share genetic loci that contribute to blood pressure traits and hypertension. However, the OR and beta-coefficient estimates differ, and some are age-dependent. Additional genetic studies of hypertension in AA are warranted to identify new loci associated with hypertension and blood pressure traits in this population.
Collapse
Affiliation(s)
- Harpreet Kaur
- Genomic Medicine Institute, Cleveland Clinic/Lerner Research Institute, Cleveland, OH, United States of America
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, United States of America
| | - Dana C. Crawford
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, United States of America
| | - Jingjing Liang
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, United States of America
| | - Penelope Benchek
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, United States of America
| | | | - Xiaofeng Zhu
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, United States of America
| | - Asha R. Kallianpur
- Genomic Medicine Institute, Cleveland Clinic/Lerner Research Institute, Cleveland, OH, United States of America
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, United States of America
| | - William S. Bush
- Genomic Medicine Institute, Cleveland Clinic/Lerner Research Institute, Cleveland, OH, United States of America
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, United States of America
| |
Collapse
|
204
|
Li ZH, Huang QM, Gao X, Chung VCH, Zhang PD, Shen D, Zhang XR, Zhong WF, Liu D, Chen PL, Chen Q, Cai MC, Cheng X, Yang HL, Song WQ, Wu XB, Kraus VB, Mao C. Healthy Sleep Associated With Lower Risk of Hypertension Regardless of Genetic Risk: A Population-Based Cohort Study. Front Cardiovasc Med 2021; 8:769130. [PMID: 34869684 PMCID: PMC8637045 DOI: 10.3389/fcvm.2021.769130] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/18/2021] [Indexed: 12/02/2022] Open
Abstract
Background: Hypertension is a leading contributor to the global burden of disease and to mortality. The combined effects of sleep factors on the risk of hypertension are unclear. We aimed to evaluate the effect of combined sleep factors on the risk of hypertension and to explore whether this association is independent of genetic risk. Methods: This population-based prospective cohort study included 170,378 participants from the UK Biobank study. We conducted a healthy sleep score based on a combination of major five sleep factors and a genetic risk score based on 118 risk variants. Cox proportional hazard regression models were used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs). Results: A total of 170,378 participants were included. Compared to participants with a healthy sleep score of 0-1, those with healthy sleep scores of 2 (HR, 0.90; 95% CI, 0.83-0.98), 3 (HR, 0.81; 95% CI, 0.75-0.88), 4 (HR, 0.74; 95% CI, 0.68-0.81), or 5 (HR, 0.67; 95% CI, 0.59-0.77) had increasingly lower risks of hypertension (P for trend <0.001). Participants with high genetic risk and an unfavorable sleep pattern had a 1.80-fold greater risk of hypertension than participants with low genetic risk and a favorable sleep pattern. The association between sleep patterns and hypertension persisted in subgroup analysis, stratified by the genetic risk. Nearly 18.2% of hypertension events in this cohort could be attributed to unfavorable sleep pattern. Conclusions: Favorable sleep pattern was associated with a low risk of hypertension, regardless of genetic risk. These findings highlight the potential of sleep interventions to reduce risk of hypertension across entire populations.
Collapse
Affiliation(s)
- Zhi-Hao Li
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Qing-Mei Huang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiang Gao
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, United States
| | - Vincent C. H. Chung
- Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Pei-Dong Zhang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Dong Shen
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xi-Ru Zhang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wen-Fang Zhong
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Dan Liu
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Pei-Liang Chen
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Qing Chen
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Miao-Chun Cai
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xin Cheng
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Hai-Lian Yang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wei-Qi Song
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xian-Bo Wu
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Virginia Byers Kraus
- Division of Rheumatology, Department of Medicine, Duke University School of Medicine, Duke Molecular Physiology Institute, Durham, NC, United States
| | - Chen Mao
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
- Division of Laboratory Medicine, Microbiome Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
205
|
Cappadona C, Paraboschi EM, Ziliotto N, Bottaro S, Rimoldi V, Gerussi A, Azimonti A, Brenna D, Brunati A, Cameroni C, Campanaro G, Carloni F, Cavadini G, Ciravegna M, Composto A, Converso G, Corbella P, D’Eugenio D, Dal Rì G, Di Giorgio SM, Grondelli MC, Guerrera L, Laffoucriere G, Lando B, Lopedote L, Maizza B, Marconi E, Mariola C, Matronola GM, Menga LM, Montorsi G, Papatolo A, Patti R, Profeta L, Rebasti V, Smidili A, Tarchi SM, Tartaglia FC, Tettamanzi G, Tinelli E, Stuani R, Bolchini C, Pattini L, Invernizzi P, Degenhardt F, Franke A, Duga S, Asselta R. MEDTEC Students against Coronavirus: Investigating the Role of Hemostatic Genes in the Predisposition to COVID-19 Severity. J Pers Med 2021; 11:1166. [PMID: 34834519 PMCID: PMC8622845 DOI: 10.3390/jpm11111166] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/04/2021] [Accepted: 11/06/2021] [Indexed: 12/23/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiologic agent of the coronavirus disease 2019 (COVID-19) pandemic. Besides virus intrinsic characteristics, the host genetic makeup is predicted to account for the extreme clinical heterogeneity of the disease, which is characterized, among other manifestations, by a derangement of hemostasis associated with thromboembolic events. To date, large-scale studies confirmed that genetic predisposition plays a role in COVID-19 severity, pinpointing several susceptibility genes, often characterized by immunologic functions. With these premises, we performed an association study of common variants in 32 hemostatic genes with COVID-19 severity. We investigated 49,845 single-nucleotide polymorphism in a cohort of 332 Italian severe COVID-19 patients and 1668 controls from the general population. The study was conducted engaging a class of students attending the second year of the MEDTEC school (a six-year program, held in collaboration between Humanitas University and the Politecnico of Milan, allowing students to gain an MD in Medicine and a Bachelor's Degree in Biomedical Engineering). Thanks to their willingness to participate in the fight against the pandemic, we evidenced several suggestive hits (p < 0.001), involving the PROC, MTHFR, MTR, ADAMTS13, and THBS2 genes (top signal in PROC: chr2:127192625:G:A, OR = 2.23, 95%CI = 1.50-3.34, p = 8.77 × 10-5). The top signals in PROC, MTHFR, MTR, ADAMTS13 were instrumental for the construction of a polygenic risk score, whose distribution was significantly different between cases and controls (p = 1.62 × 10-8 for difference in median levels). Finally, a meta-analysis performed using data from the Regeneron database confirmed the contribution of the MTHFR variant chr1:11753033:G:A to the predisposition to severe COVID-19 (pooled OR = 1.21, 95%CI = 1.09-1.33, p = 4.34 × 10-14 in the weighted analysis).
Collapse
Affiliation(s)
- Claudio Cappadona
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Elvezia Maria Paraboschi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
- Humanitas Clinical and Research Center, IRCCS, Via Manzoni 56, 20089 Rozzano, Italy
| | - Nicole Ziliotto
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Sandro Bottaro
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
- Humanitas Clinical and Research Center, IRCCS, Via Manzoni 56, 20089 Rozzano, Italy
| | - Valeria Rimoldi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Alessio Gerussi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (A.G.); (P.I.)
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, 20900 Monza, Italy
| | - Andrea Azimonti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Daniele Brenna
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Andrea Brunati
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Charlotte Cameroni
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Giovanni Campanaro
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Francesca Carloni
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Giacomo Cavadini
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Martina Ciravegna
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Antonio Composto
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Giuseppe Converso
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Pierluigi Corbella
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Davide D’Eugenio
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Giovanna Dal Rì
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Sofia Maria Di Giorgio
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Maria Chiara Grondelli
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Lorenza Guerrera
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Georges Laffoucriere
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Beatrice Lando
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Leandro Lopedote
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Benedetta Maizza
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Elettra Marconi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Carlotta Mariola
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Guia Margherita Matronola
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Luca Maria Menga
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Giulia Montorsi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Antonio Papatolo
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Riccardo Patti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Lorenzo Profeta
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Vera Rebasti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Alice Smidili
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Sofia Maria Tarchi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Francesco Carlo Tartaglia
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Gaia Tettamanzi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Elena Tinelli
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Riccardo Stuani
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
| | - Cristiana Bolchini
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133 Milan, Italy; (C.B.); (L.P.)
| | - Linda Pattini
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133 Milan, Italy; (C.B.); (L.P.)
| | - Pietro Invernizzi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (A.G.); (P.I.)
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, 20900 Monza, Italy
| | - Frauke Degenhardt
- Institute of Clinical Molecular Biology, Christian-Albrechts-University Kiel, 24105 Kiel, Germany; (F.D.); (A.F.)
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University Kiel, 24105 Kiel, Germany; (F.D.); (A.F.)
- University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany
| | - Stefano Duga
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
- Humanitas Clinical and Research Center, IRCCS, Via Manzoni 56, 20089 Rozzano, Italy
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (C.C.); (E.M.P.); (N.Z.); (S.B.); (V.R.); (A.A.); (D.B.); (A.B.); (C.C.); (G.C.); (F.C.); (G.C.); (M.C.); (A.C.); (G.C.); (P.C.); (D.D.); (G.D.R.); (S.M.D.G.); (M.C.G.); (L.G.); (G.L.); (B.L.); (L.L.); (B.M.); (E.M.); (C.M.); (G.M.M.); (L.M.M.); (G.M.); (A.P.); (R.P.); (L.P.); (V.R.); (A.S.); (S.M.T.); (F.C.T.); (G.T.); (E.T.); (R.S.); (S.D.)
- Humanitas Clinical and Research Center, IRCCS, Via Manzoni 56, 20089 Rozzano, Italy
| |
Collapse
|
206
|
Xu B, Xu Z, Xu D, Tan X. Effect of n-3 polyunsaturated fatty acids on ischemic heart disease and cardiometabolic risk factors: a two-sample Mendelian randomization study. BMC Cardiovasc Disord 2021; 21:532. [PMID: 34749668 PMCID: PMC8576934 DOI: 10.1186/s12872-021-02342-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 10/22/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The cardioprotective ability of n-3 polyunsaturated fatty acids (PUFAs) is controversial. Most studies suggest a specific role for PUFAs in cardioprotection from ischemic heart disease (IHD). However, few studies have used genetic biomarkers of n-3 PUFAs to examine their potential relationships with IHD. This study aimed to use Mendelian randomization to evaluate whether genetically-predicted n-3 PUFAs affect IHD and cardiometabolic risk factors (CRFs). METHODS Genetic variants strongly (p < 5 × 10-8) and independently (r2 > 0.1) associated with n-3 PUFAs were derived from the CHARGE Consortium (including 8,866 subjects of European ancestry) and were used as instrumental variables (IVs) for evaluating the effect of n-3 PUFAs, including α-linolenic acid (ALA), docosapentaenoic acid (DPA), docosahexaenoic acid (DHA), and eicosapentaenoic acid (EPA). Data on the associations between the IVs and IHD, myocardial infarction, and CRFs (including diabetes, lipids, blood pressure, body mass index, and waist-to-hip ratio (WHR)) were obtained from the UK Biobank SOFT CAD GWAS with the CARDIoGRAMplusC4D 1000 Genomes-based GWAS (113,937 IHD cases and 339,115 controls), the Myocardial Infarction Genetics and CARDIoGRAM Exome consortia (42,335 MI cases and 78,240 controls), the DIAbetes Genetics Replication And Meta-analysis consortium (26,676 diabetes mellitus cases and 132,532 controls), the Global Lipids Genetics Consortium (n = 196,475), the International Consortium for Blood Pressure (n = 69,395), and the meta-analysis of GWAS for body fat distribution in the UK Biobank and Genetic Investigation of Anthropometric Traits (n = 694,649). RESULTS Genetically-predicted higher ALA was associated with lower risk of IHD, type 2 diabetes (T2D), and lower serum lipids. The effect size per 0.05-unit increase (about 1 standard deviation) in plasma ALA level) was - 1.173 (95% confidence interval - 2.214 to - 0.133) for IHD. DPA and EPA had no association with IHD but were associated with a higher risk of T2D, higher levels of lipids or WHR. DHA had no association with IHD or CRFs. CONCLUSIONS Our study suggests a benefit of ALA for IHD and its main risk factors. DHA, DPA, and EPA had no association with IHD but were partly associated with increasing cardiometabolic risk factors.
Collapse
Affiliation(s)
- Bayi Xu
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Zhixia Xu
- Department of Medical Service, Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Duanmin Xu
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xuerui Tan
- Clinical Research Center, First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China.
| |
Collapse
|
207
|
Wang H, Noordam R, Cade BE, Schwander K, Winkler TW, Lee J, Sung YJ, Bentley AR, Manning AK, Aschard H, Kilpeläinen TO, Ilkov M, Brown MR, Horimoto AR, Richard M, Bartz TM, Vojinovic D, Lim E, Nierenberg JL, Liu Y, Chitrala K, Rankinen T, Musani SK, Franceschini N, Rauramaa R, Alver M, Zee PC, Harris SE, van der Most PJ, Nolte IM, Munroe PB, Palmer ND, Kühnel B, Weiss S, Wen W, Hall KA, Lyytikäinen LP, O'Connell J, Eiriksdottir G, Launer LJ, de Vries PS, Arking DE, Chen H, Boerwinkle E, Krieger JE, Schreiner PJ, Sidney S, Shikany JM, Rice K, Chen YDI, Gharib SA, Bis JC, Luik AI, Ikram MA, Uitterlinden AG, Amin N, Xu H, Levy D, He J, Lohman KK, Zonderman AB, Rice TK, Sims M, Wilson G, Sofer T, Rich SS, Palmas W, Yao J, Guo X, Rotter JI, Biermasz NR, Mook-Kanamori DO, Martin LW, Barac A, Wallace RB, Gottlieb DJ, Komulainen P, Heikkinen S, Mägi R, Milani L, Metspalu A, Starr JM, Milaneschi Y, Waken RJ, Gao C, Waldenberger M, Peters A, Strauch K, Meitinger T, Roenneberg T, Völker U, Dörr M, Shu XO, Mukherjee S, Hillman DR, Kähönen M, Wagenknecht LE, Gieger C, Grabe HJ, Zheng W, et alWang H, Noordam R, Cade BE, Schwander K, Winkler TW, Lee J, Sung YJ, Bentley AR, Manning AK, Aschard H, Kilpeläinen TO, Ilkov M, Brown MR, Horimoto AR, Richard M, Bartz TM, Vojinovic D, Lim E, Nierenberg JL, Liu Y, Chitrala K, Rankinen T, Musani SK, Franceschini N, Rauramaa R, Alver M, Zee PC, Harris SE, van der Most PJ, Nolte IM, Munroe PB, Palmer ND, Kühnel B, Weiss S, Wen W, Hall KA, Lyytikäinen LP, O'Connell J, Eiriksdottir G, Launer LJ, de Vries PS, Arking DE, Chen H, Boerwinkle E, Krieger JE, Schreiner PJ, Sidney S, Shikany JM, Rice K, Chen YDI, Gharib SA, Bis JC, Luik AI, Ikram MA, Uitterlinden AG, Amin N, Xu H, Levy D, He J, Lohman KK, Zonderman AB, Rice TK, Sims M, Wilson G, Sofer T, Rich SS, Palmas W, Yao J, Guo X, Rotter JI, Biermasz NR, Mook-Kanamori DO, Martin LW, Barac A, Wallace RB, Gottlieb DJ, Komulainen P, Heikkinen S, Mägi R, Milani L, Metspalu A, Starr JM, Milaneschi Y, Waken RJ, Gao C, Waldenberger M, Peters A, Strauch K, Meitinger T, Roenneberg T, Völker U, Dörr M, Shu XO, Mukherjee S, Hillman DR, Kähönen M, Wagenknecht LE, Gieger C, Grabe HJ, Zheng W, Palmer LJ, Lehtimäki T, Gudnason V, Morrison AC, Pereira AC, Fornage M, Psaty BM, van Duijn CM, Liu CT, Kelly TN, Evans MK, Bouchard C, Fox ER, Kooperberg C, Zhu X, Lakka TA, Esko T, North KE, Deary IJ, Snieder H, Penninx BWJH, Gauderman WJ, Rao DC, Redline S, van Heemst D. Multi-ancestry genome-wide gene-sleep interactions identify novel loci for blood pressure. Mol Psychiatry 2021; 26:6293-6304. [PMID: 33859359 PMCID: PMC8517040 DOI: 10.1038/s41380-021-01087-0] [Show More Authors] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 03/18/2021] [Accepted: 03/29/2021] [Indexed: 02/02/2023]
Abstract
Long and short sleep duration are associated with elevated blood pressure (BP), possibly through effects on molecular pathways that influence neuroendocrine and vascular systems. To gain new insights into the genetic basis of sleep-related BP variation, we performed genome-wide gene by short or long sleep duration interaction analyses on four BP traits (systolic BP, diastolic BP, mean arterial pressure, and pulse pressure) across five ancestry groups in two stages using 2 degree of freedom (df) joint test followed by 1df test of interaction effects. Primary multi-ancestry analysis in 62,969 individuals in stage 1 identified three novel gene by sleep interactions that were replicated in an additional 59,296 individuals in stage 2 (stage 1 + 2 Pjoint < 5 × 10-8), including rs7955964 (FIGNL2/ANKRD33) that increases BP among long sleepers, and rs73493041 (SNORA26/C9orf170) and rs10406644 (KCTD15/LSM14A) that increase BP among short sleepers (Pint < 5 × 10-8). Secondary ancestry-specific analysis identified another novel gene by long sleep interaction at rs111887471 (TRPC3/KIAA1109) in individuals of African ancestry (Pint = 2 × 10-6). Combined stage 1 and 2 analyses additionally identified significant gene by long sleep interactions at 10 loci including MKLN1 and RGL3/ELAVL3 previously associated with BP, and significant gene by short sleep interactions at 10 loci including C2orf43 previously associated with BP (Pint < 10-3). 2df test also identified novel loci for BP after modeling sleep that has known functions in sleep-wake regulation, nervous and cardiometabolic systems. This study indicates that sleep and primary mechanisms regulating BP may interact to elevate BP level, suggesting novel insights into sleep-related BP regulation.
Collapse
Affiliation(s)
- Heming Wang
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA.
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Brian E Cade
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Karen Schwander
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Thomas W Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Jiwon Lee
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Joint Carnegie Mellon University-University of Pittsburgh PhD Program in Computational Biology, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Pittsburgh Center for Evolutionary Biology and Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alisa K Manning
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hugues Aschard
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
- Centre de Bioinformatique, Biostatistique et Biologie Intégrative (C3BI), Institut Pasteur, Paris, France
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Environmental Medicine and Public Health, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Andrea R Horimoto
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Melissa Richard
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Dina Vojinovic
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Elise Lim
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Jovia L Nierenberg
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Yongmei Liu
- Division of Cardiology, Department of Medicine, Duke Molecular Physiology Institute Duke University School of Medicine, Durham, NC, USA
| | - Kumaraswamynaidu Chitrala
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Solomon K Musani
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Rainer Rauramaa
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Maris Alver
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Phyllis C Zee
- Division of Sleep Medicine, Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Sarah E Harris
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Peter J van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Patricia B Munroe
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, London, London, UK
| | | | - Brigitte Kühnel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- German Center for Cardiovascular Research (DZHK), partner site Greifswald, Greifswald, Germany
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Kelly A Hall
- School of Public Health, The University of Adelaide, Adelaide, SA, Australia
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jeff O'Connell
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
- Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Dan E Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Han Chen
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Precision Health, School of Public Health & School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Jose E Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Pamela J Schreiner
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | | | - James M Shikany
- Division of Preventive Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kenneth Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Sina A Gharib
- Computational Medicine Core, Center for Lung Biology, UW Medicine Sleep Center, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Annemarie I Luik
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Hanfei Xu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Daniel Levy
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Kurt K Lohman
- Division of Cardiology, Department of Medicine, Duke Molecular Physiology Institute Duke University School of Medicine, Durham, NC, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Treva K Rice
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Mario Sims
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Gregory Wilson
- JHS Graduate Training and Education Center, Jackson State University, Jackson, MS, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Walter Palmas
- Division of General Medicine, Department of Medicine, Columbia University, New York, NY, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Nienke R Biermasz
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, The Netherlands
| | - Lisa W Martin
- George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Ana Barac
- MedStar Heart and Vascular Institute, Washington, DC, USA
| | - Robert B Wallace
- Department of Epidemiology, University of Iowa College of Public Health, Iowa City, IA, USA
| | - Daniel J Gottlieb
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Pirjo Komulainen
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Sami Heikkinen
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Finland
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Reedik Mägi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Lili Milani
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Andres Metspalu
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - John M Starr
- Alzheimer Scotland Dementia Research Centre, The University of Edinburgh, Edinburgh, UK
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit, Amsterdam, HJ, The Netherlands
| | - R J Waken
- Division of Cardiology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Chuan Gao
- Molecular Genetics and Genomics Program, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Neuherberg, Germany
| | - Konstantin Strauch
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Till Roenneberg
- Institute and Polyclinic for Occupational-, Social- and Environmental Medicine, LMU Munich, Munich, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- German Center for Cardiovascular Research (DZHK), partner site Greifswald, Greifswald, Germany
| | - Marcus Dörr
- German Center for Cardiovascular Research (DZHK), partner site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Sutapa Mukherjee
- Sleep Health Service, Respiratory and Sleep Services, Southern Adelaide Local Health Network, Adelaide, SA, Australia
- Adelaide Institute for Sleep Health, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - David R Hillman
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
- Department of Clinical Physiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Lynne E Wagenknecht
- Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Hans J Grabe
- Department Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Lyle J Palmer
- School of Public Health, The University of Adelaide, Adelaide, SA, Australia
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alexandre C Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Cardiovascular Health Research Unit, Departments of Epidemiology and Health Services, University of Washington, Seattle, WA, USA
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Tanika N Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Ervin R Fox
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Timo A Lakka
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Tõnu Esko
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Ian J Deary
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit, Amsterdam, HJ, The Netherlands
| | - W James Gauderman
- Division of Biostatistics, Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Dabeeru C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
208
|
Niu M, Wang Y, Zhang L, Tu R, Liu X, Hou J, Huo W, Mao Z, Wang C, Bie R. Identifying the predictive effectiveness of a genetic risk score for incident hypertension using machine learning methods among populations in rural China. Hypertens Res 2021; 44:1483-1491. [PMID: 34480134 DOI: 10.1038/s41440-021-00738-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/31/2021] [Accepted: 08/04/2021] [Indexed: 12/17/2022]
Abstract
Current studies have shown the controversial effect of genetic risk scores (GRSs) in hypertension prediction. Machine learning methods are used extensively in the medical field but rarely in the mining of genetic information. This study aims to determine whether genetic information can improve the prediction of incident hypertension using machine learning approaches in a prospective study. The study recruited 4592 subjects without hypertension at baseline from a cohort study conducted in rural China. A polygenic risk score (PGGRS) was calculated using 13 SNPs. According to a ratio of 7:3, subjects were randomly allocated to the train and test datasets. Models with and without the PGGRS were established using the train dataset with Cox regression, artificial neural network (ANN), random forest (RF), and gradient boosting machine (GBM) methods. The discrimination and reclassification of models were estimated using the test dataset. The PGGRS showed a significant association with the risk of incident hypertension (HR (95% CI), 1.046 (1.004, 1.090), P = 0.031) irrespective of baseline blood pressure. Models that did not include the PGGRS achieved AUCs (95% CI) of 0.785 (0.763, 0.807), 0.790 (0.768, 0.811), 0.838 (0.817, 0.857), and 0.854 (0.835, 0.873) for the Cox, ANN, RF, and GBM methods, respectively. The addition of the PGGRS led to the improvement of the AUC by 0.001, 0.008, 0.023, and 0.017; IDI by 1.39%, 2.86%, 4.73%, and 4.68%; and NRI by 25.05%, 13.01%, 44.87%, and 22.94%, respectively. Incident hypertension risk was better predicted by the traditional+PGGRS model, especially when machine learning approaches were used, suggesting that genetic information may have the potential to identify new hypertension cases using machine learning methods in resource-limited areas. CLINICAL TRIAL REGISTRATION: The Henan Rural Cohort Study has been registered at the Chinese Clinical Trial Register (Registration number: ChiCTR-OOC-15006699). http://www.chictr.org.cn/showproj.aspx?proj=11375 .
Collapse
Affiliation(s)
- Miaomiao Niu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Yikang Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Liying Zhang
- School of Information Engineering, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Runqi Tu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Xiaotian Liu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Jian Hou
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Wenqian Huo
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Zhenxing Mao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Chongjian Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China.
| | - Ronghai Bie
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China.
| |
Collapse
|
209
|
Jin Q, Shi G. Meta-Analysis of Joint Test of SNP and SNP-Environment Interaction with Heterogeneity. Hum Hered 2021; 86:1-9. [PMID: 34700323 DOI: 10.1159/000519098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 07/29/2021] [Indexed: 12/13/2022] Open
Abstract
Many complex diseases are caused by single nucleotide polymorphisms (SNPs), environmental factors, and the interaction between SNPs and environment. Joint tests of the SNP and SNP-environment interaction effects (JMA) and meta-regression (MR) are commonly used to evaluate these SNP-environment interactions. However, these two methods do not consider genetic heterogeneity. We previously presented a random-effect MR, which provided higher power than the MR in datasets with high heterogeneity. However, this method requires group-level data, which sometimes are not available. Given this, we designed this study to evaluate the introduction of the random effects of SNP and SNP-environment interaction into the JMA, and then extended this to the random effect model. Likelihood ratio statistic is applied to test the JMA and the new method we proposed in this paper. We evaluated the null distributions of these tests, and the powers for this method. This method was verified by simulation and was shown to provide similar powers to the random effect meta-regression method (RMR). However, this method only requires study-level data which relaxed the condition of the RMR. Our study suggests that this method is more suitable for finding the association between SNP and diseases in the absence of group-level data.
Collapse
Affiliation(s)
- Qinqin Jin
- State Key Laboratory of Integrated Services Networks, Xidian University, Xi'an, China.,Applied Science College, Taiyuan University of Science and Technology, Taiyuan, China
| | - Gang Shi
- State Key Laboratory of Integrated Services Networks, Xidian University, Xi'an, China
| |
Collapse
|
210
|
Wang X, Fang X, Zheng W, Zhou J, Song Z, Xu M, Min J, Wang F. Genetic Support of A Causal Relationship Between Iron Status and Type 2 Diabetes: A Mendelian Randomization Study. J Clin Endocrinol Metab 2021; 106:e4641-e4651. [PMID: 34147035 PMCID: PMC8530720 DOI: 10.1210/clinem/dgab454] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Indexed: 12/15/2022]
Abstract
CONTEXT Iron overload is a known risk factor for type 2 diabetes (T2D); however, iron overload and iron deficiency have both been associated with metabolic disorders in observational studies. OBJECTIVE Using mendelian randomization (MR), we assessed how genetically predicted systemic iron status affected T2D risk. METHODS A 2-sample MR analysis was used to obtain a causal estimate. We selected genetic variants strongly associated (P < 5 × 10-8) with 4 biomarkers of systemic iron status from a study involving 48 972 individuals performed by the Genetics of Iron Status consortium and applied these biomarkers to the T2D case-control study (74 124 cases and 824 006 controls) performed by the Diabetes Genetics Replication and Meta-analysis consortium. The simple median, weighted median, MR-Egger, MR analysis using mixture-model, weighted allele scores, and MR based on a Bayesian model averaging approaches were used for the sensitivity analysis. RESULTS Genetically instrumented serum iron (odds ratio [OR]: 1.07; 95% CI, 1.02-1.12), ferritin (OR: 1.19; 95% CI, 1.08-1.32), and transferrin saturation (OR: 1.06; 95% CI, 1.02-1.09) were positively associated with T2D. In contrast, genetically instrumented transferrin, a marker of reduced iron status, was inversely associated with T2D (OR: 0.91; 95% CI, 0.87-0.96). CONCLUSION Genetic evidence supports a causal link between increased systemic iron status and increased T2D risk. Further studies involving various ethnic backgrounds based on individual-level data and studies regarding the underlying mechanism are warranted for reducing the risk of T2D.
Collapse
Affiliation(s)
- Xinhui Wang
- The Fourth Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xuexian Fang
- The Fourth Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wanru Zheng
- The Fourth Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jiahui Zhou
- The Fourth Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zijun Song
- The Fourth Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Mingqing Xu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
| | - Junxia Min
- The Fourth Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Fudi Wang
- The Fourth Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
211
|
Yu J, Liu L, Li Z, Wang Y, Zhang W, Jin Y, He L, Chen Y, Yao Y. Association of single nucleotide polymorphisms in ADIPOQ gene with risk of hypertension: a systematic review and meta-analysis. INTERNATIONAL JOURNAL OF MOLECULAR EPIDEMIOLOGY AND GENETICS 2021; 12:90-101. [PMID: 34853633 PMCID: PMC8611228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 09/14/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Hypertension has been continuing to be a major contributor to the global burden of disease and to the global mortality, leading to over 10 million deaths each year. The purpose of this study was to investigate the association between Adiponectin gene polymorphism with Essential hypertension (EH). METHODS PubMed, EMbase, the Cochrane Library, and China National Knowledge Infrastructure (CNKI) were searched independently by two investigators. Pooled odds ratios and 95% confidence intervals were calculated to estimate the associations of Adiponectin polymorphism with EH. RESULTS Thirteen studies with 3198 cases and 3076 controls for meta-analysis (MA) were included in present study. Pooled results showed that rs2241766 polymorphism is associated with the risk of EH in the allelic model (G vs. T: OR=1.10; 95% CI, 1.01-1.21). In the <40 years subgroup, rs2241766 polymorphism is associated with the risk of EH in allele model (G vs. T: OR=1.43; 95% CI, 1.06-1.94), recessive model (GG vs. GT + TT: OR=5.26, 95% CI=1.47-18.76), homozygous model of GG (GG vs.TT: OR=5.27, 95% CI=1.47-18.95), and rs266729 in recessive model (GG vs. GT + TT: OR=2.33, 95% CI=1.33-4.08). CONCLUSIONS Our meta-analysis results show that the rs2241766 polymorphism is associated with the risk of hypertension. There still need a larger sample with better design to verify.
Collapse
Affiliation(s)
- Jiegen Yu
- School of Humanities Management, Wannan Medical CollegeWuhu 241002, Anhui, China
| | - Ling Liu
- School of Public Health, Wannan Medical CollegeWuhu 241002, Anhui, China
- Institute of Chronic Disease Control and Prevention, Wannan Medical CollegeWuhu 241002, Anhui China
| | - Zhipeng Li
- School of Medicine, Taizhou UniversityTaizhou 318000, Zhejiang, China
| | - Yanqiu Wang
- School of Public Health, Wannan Medical CollegeWuhu 241002, Anhui, China
- Institute of Chronic Disease Control and Prevention, Wannan Medical CollegeWuhu 241002, Anhui China
| | - Wanjun Zhang
- School of Public Health, Wannan Medical CollegeWuhu 241002, Anhui, China
- Institute of Chronic Disease Control and Prevention, Wannan Medical CollegeWuhu 241002, Anhui China
| | - Yuelong Jin
- School of Public Health, Wannan Medical CollegeWuhu 241002, Anhui, China
- Institute of Chronic Disease Control and Prevention, Wannan Medical CollegeWuhu 241002, Anhui China
| | - Liangping He
- School of Medicine, Taizhou UniversityTaizhou 318000, Zhejiang, China
| | - Yan Chen
- School of Public Health, Wannan Medical CollegeWuhu 241002, Anhui, China
- Institute of Chronic Disease Control and Prevention, Wannan Medical CollegeWuhu 241002, Anhui China
| | - Yingshui Yao
- School of Public Health, Wannan Medical CollegeWuhu 241002, Anhui, China
- Institute of Chronic Disease Control and Prevention, Wannan Medical CollegeWuhu 241002, Anhui China
- Anhui College of Traditional Chinese MedicineWuhu 241002, Anhui, China
| |
Collapse
|
212
|
Mompeo O, Berry SE, Spector TD, Menni C, Mangino M, Gibson R. Differential associations between a priori diet quality scores and markers of cardiovascular health in women: cross-sectional analyses from TwinsUK. Br J Nutr 2021; 126:1017-1027. [PMID: 33298202 DOI: 10.1017/s000711452000495x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
CVD is the leading cause of death worldwide and, after dementia, is the second biggest cause of death for women. In England, it accounts for one in four of all deaths. Lifestyle modifications represent the primary route both to reduce CVD risk factors and prevent CVD outcomes. Diet constitutes one of the key modifiable risk factors in the aetiology of CVD. We investigated the relationship between nine main dietary indices and a comprehensive range of CVD risk factors in 2590 women from TwinsUK. After adjustment for multiple testing, we found that the Dietary Approaches to Stop Hypertension (DASH) diet was inversely correlated with some of the most common CVD risk factors (BMI, visceral fat (VF), TAG, insulin, homoeostasis model assessment of insulin resistance (HOMA2-IR) and atherosclerotic CVD (ASCVD) risk) with PFDR ranging from 6·28 × 10-7 to 5·63 × 10-4. Similar association patterns were detected across most of the dietary indices analysed. In our post hoc investigation, to determine if any specific food groups were driving associations between the DASH score and markers of cardiometabolic risk, we found that increased BMI, VF, HOMA2-IR, ASCVD risk, insulin and TAG levels were directly correlated with red meat consumption (PFDR ranging from 4·65 × 10-9 to 7·98 × 10-3) and inversely correlated with whole-grain cereal consumption (PFDR ranging from 1·26 × 10-6 to 8·28 × 10-3). Our findings revealed that the DASH diet is associated with a more favourable CVD risk profile, suggesting that this diet may be a candidate dietary pattern to supplement current UK dietary recommendations for CVD prevention.
Collapse
Affiliation(s)
- Olatz Mompeo
- Department of Twin Research and Genetic Epidemiology, King's College London, LondonSE1 7EH, UK
| | - Sarah E Berry
- Department of Nutritional Sciences, King's College London, LondonSE1 9NH, UK
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, LondonSE1 7EH, UK
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King's College London, LondonSE1 7EH, UK
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, LondonSE1 7EH, UK
- NIHR Biomedical Research Centre, Guy's and St Thomas' Foundation Trust, LondonSE1 9RT, UK
| | - Rachel Gibson
- Department of Nutritional Sciences, King's College London, LondonSE1 9NH, UK
| |
Collapse
|
213
|
Distler O, Ludwig RJ, Niemann S, Riemekasten G, Schreiber S. Editorial: Precision Medicine in Chronic Inflammation. Front Immunol 2021; 12:770462. [PMID: 34630441 PMCID: PMC8495129 DOI: 10.3389/fimmu.2021.770462] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 01/12/2023] Open
Affiliation(s)
- Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ralf J Ludwig
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Zurich, Germany
| | - Stefan Niemann
- Research Center Borstel, Molecular and Experimental Mycobacteriology, Borstel, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
| | | | - Stefan Schreiber
- Department of Medicine I, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
214
|
Mozaffari H, Askari M, Bellissimo N, Azadbakht L. Associations between dietary intake of B vitamins and cardiovascular risk factors in elderly men: A cross-sectional study. Int J Clin Pract 2021; 75:e14691. [PMID: 34331825 DOI: 10.1111/ijcp.14691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 07/28/2021] [Indexed: 10/20/2022] Open
Abstract
OBJECTIVE Low intake of B vitamins can lead to hyperhomocysteinaemia, which is reported as a risk factor for cardiovascular disease (CVD). However, little is known about this relationship. Therefore, we decided to examine the association between dietary intake of B vitamins and cardiovascular risk factors in elderly men. METHODS The present cross-sectional study consisted of 357 elderly men. Dietary intake was assessed using a validated and reliable food frequency questionnaire. All biochemical factors [fasting blood sugar (FBS), triglyceride (TG), high-sensitivity C-reactive protein (hs-CRP), interleukin 6 (IL-6) and tumour necrosis factor-α (TNF-α)], waist circumference (WC) and blood pressure (BP) were assessed using standard methods. RESULTS A significant inverse association was observed between high dietary intake of total B vitamins (ORTotal B vitamins : 0.30; 95% CI: 0.10, 0.86; P = .01) and vitamin B9 (ORvitamin B9 : 0.20; 95% CI: 0.02, 0.52; P = .002) with high WC. Furthermore, an inverse association was observed between high dietary intake of vitamin B6 (ORvitamin B6 : 0.28; 95% CI: 0.08, 1.00; P = .05) and vitamin B9 (ORvitamin B9 : 0.20; 95% CI: 0.06, 0.70; P = .01) with hs-CRP level. CONCLUSIONS In elderly men, a high dietary intake of total B vitamins and vitamin B9 was associated with lower odds of having a high WC. Similarly, high dietary intake of vitamin B6 and B9 was associated with lower odds of having a high hs-CRP level.
Collapse
Affiliation(s)
- Hadis Mozaffari
- Faculty of Land and Food Systems, University of British Columbia, Vancouver, BC, Canada
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Askari
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Nick Bellissimo
- School of Nutrition, Ryerson University, Toronto, ON, Canada
| | - Leila Azadbakht
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Science, Isfahan, Iran
| |
Collapse
|
215
|
Abstract
Chronic cardiovascular diseases are associated with inflammatory responses within the blood vessels and end organs. The origin of this inflammation has not been certain, and neither is its relationship to disease clear. There is a need to determine whether this association is causal or coincidental to the processes leading to cardiovascular disease. These processes are themselves complex: many cardiovascular diseases arise in conjunction with the presence of sustained elevation of blood pressure. Inflammatory processes have been linked to hypertension, and causality has been suggested. Evidence of causality poses the difficult challenge of linking the integrated and multifaceted biology of blood pressure regulation with vascular function and complex elements of immune system function. These include both, innate and adaptive immunity, as well as interactions between the host immune system and the omnipresent microorganisms that are encountered in the environment and that colonize and exist in commensal relationship with the host. Progress has been made in this task and has drawn on experimental approaches in animals, much of which have focused on hypertension occurring with prolonged infusion of angiotensin II. These laboratory studies are complemented by studies that seek to inform disease mechanism by examining the genomic basis of heritable disease susceptibility in human populations. In this realm too, evidence has emerged that implicates genetic variation affecting immunity in disease pathogenesis. In this article, we survey the genetic and genomic evidence linking high blood pressure and its end-organ injuries to immune system function and examine evidence that genomic factors can influence disease risk. © 2021 American Physiological Society. Compr Physiol 11:1-22, 2021.
Collapse
Affiliation(s)
- Isha S Dhande
- Center for Human Genetics, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Peter A Doris
- Center for Human Genetics, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
216
|
You R, Chen L, Xu L, Zhang D, Li H, Shi X, Zheng Y, Chen L. High Level of Uromodulin Increases the Risk of Hypertension: A Mendelian Randomization Study. Front Cardiovasc Med 2021; 8:736001. [PMID: 34540925 PMCID: PMC8440862 DOI: 10.3389/fcvm.2021.736001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 08/09/2021] [Indexed: 12/21/2022] Open
Abstract
Background: The association of uromodulin and hypertension has been observed in clinical studies, but not proven by a causal relationship. We conducted a two-sample Mendelian randomization (MR) analysis to investigate the causal relationship between uromodulin and blood pressure. Methods: We selected single nucleotide polymorphisms (SNPs) related to urinary uromodulin (uUMOD) and serum uromodulin (sUMOD) from a large Genome-Wide Association Studies (GWAS) meta-analysis study and research in PubMed. Six datasets based on the UK Biobank and the International Consortium for Blood Pressure (ICBP) served as outcomes with a large sample of hypertension (n = 46,188), systolic blood pressure (SBP, n = 1,194,020), and diastolic blood pressure (DBP, n = 1,194,020). The inverse variance weighted (IVW) method was performed in uUMOD MR analysis, while methods of IVW, MR-Egger, Weighted median, and Mendelian Randomization Pleiotropy RESidual Sum and Outlier (MR-PRESSO) were utilized on sUMOD MR analysis. Results: MR analysis of IVM showed the odds ratio (OR) of the uUMOD to hypertension (“ukb-b-14057” and “ukb-b-14177”) is 1.04 (95% Confidence Interval (CI), 1.03-1.04, P < 0.001); the effect sizes of the uUMOD to SBP are 1.10 (Standard error (SE) = 0.25, P = 8.92E-06) and 0.03 (SE = 0.01, P = 2.70E-04) in “ieu-b-38” and “ukb-b-20175”, respectively. The β coefficient of the uUMOD to DBP is 0.88 (SE = 0.19, P = 4.38E-06) in “ieu-b-39” and 0.05 (SE = 0.01, P = 2.13E-10) in “ukb-b-7992”. As for the sUMOD, the OR of hypertension (“ukb-b-14057” and “ukb-b-14177”) is 1.01 (95% CI 1.01–1.02, all P < 0.001). The β coefficient of the SBP is 0.37 (SE = 0.07, P = 1.26E-07) in “ieu-b-38” and 0.01 (SE = 0.003, P = 1.04E-04) in “ukb-b-20175”. The sUMOD is causally associated with elevated DBP (“ieu-b-39”: β = 0.313, SE = 0.050, P = 3.43E-10; “ukb-b-7992”: β = 0.018, SE = 0.003, P = 8.41E-09). Conclusion: Our results indicated that high urinary and serum uromodulin levels are potentially detrimental in elevating blood pressure, and serve as a causal risk factor for hypertension.
Collapse
Affiliation(s)
- Ruilian You
- Department of Nephrology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Lanlan Chen
- First Clinical Medical College of Norman Bethune Health Science Center, Jilin University, Changchun, China
| | - Lubin Xu
- Department of Nephrology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Dingding Zhang
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haitao Li
- China-Japan Friendship Hospital, Jilin University, Changchun, China
| | - Xiaoxiao Shi
- Department of Nephrology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yali Zheng
- Department of Nephrology, Affiliated Ningxia People's Hospital of Ningxia Medical University, Yinchuan, China
| | - Limeng Chen
- Department of Nephrology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
217
|
Nakaya N, Xie T, Scheerder B, Tsuchiya N, Narita A, Nakamura T, Metoki H, Obara T, Ishikuro M, Hozawa A, Snieder H, Kuriyama S. Spousal similarities in cardiometabolic risk factors: A cross-sectional comparison between Dutch and Japanese data from two large biobank studies. Atherosclerosis 2021; 334:85-92. [PMID: 34492521 DOI: 10.1016/j.atherosclerosis.2021.08.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 08/18/2021] [Accepted: 08/25/2021] [Indexed: 01/25/2023]
Abstract
BACKGROUND AND AIMS Few studies have examined and compared spousal concordance in different populations. This study aimed to quantify and compare spousal similarities in cardiometabolic risk factors and diseases between Dutch and Japanese populations. METHODS This cross-sectional study included 28,265 Dutch Lifelines Cohort Study spouse pairs (2006-2013) and 5,391 Japanese Tohoku Medical Megabank Organization (ToMMo) Cohort Study pairs (2013-2016). Spousal similarities in cardiometabolic risk factors were evaluated using Pearson's correlation or logistic regression analyses adjusted for spousal age. RESULTS The husbands' and wives' average ages in the Lifelines and ToMMo cohorts were 50.0 and 47.7 years and 63.2 and 60.4 years, respectively. Significant spousal similarities occurred with all cardiometabolic risk factors and diseases of interest in both cohorts. The age-adjusted correlation coefficients ranged from 0.032 to 0.263, with the strongest correlations observed in anthropometric traits. Spousal odds ratios [95% confidence interval] for the Lifelines vs. ToMMo cohort ranged from 1.45 (1.36-1.55) vs. 1.20 (1.05-1.38) for hypertension to 6.86 (6.30-7.48) vs. 4.60 (3.52-6.02) for current smoking. An increasing trend in spousal concordance with age was observed for sufficient physical activity in both cohorts. For current smoking, those aged 20-39 years showed the strongest concordance between pairs in both cohorts. The Dutch pairs showed stronger similarities in anthropometric traits and lifestyle habits (smoking and drinking) than their Japanese counterparts. CONCLUSIONS Spouses showed similarities in several cardiometabolic risk factors among Dutch and Japanese populations, with regional and cultural influences on spousal similarities.
Collapse
Affiliation(s)
- Naoki Nakaya
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Health Science, Saitama Prefectural University, Japan.
| | - Tian Xie
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Bart Scheerder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands; Center for Development & Innovation, University Medical Center Groningen, Groningen, the Netherlands
| | - Naho Tsuchiya
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akira Narita
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Tomohiro Nakamura
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hirohito Metoki
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Division of Public Health, Hygiene and Epidemiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Taku Obara
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan
| | - Mami Ishikuro
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Atsushi Hozawa
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Shinichi Kuriyama
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Disaster Public Health, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
| |
Collapse
|
218
|
Gupta R, Karczewski KJ, Howrigan D, Neale BM, Mootha VK. Human genetic analyses of organelles highlight the nucleus in age-related trait heritability. eLife 2021; 10:68610. [PMID: 34467851 PMCID: PMC8476128 DOI: 10.7554/elife.68610] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022] Open
Abstract
Most age-related human diseases are accompanied by a decline in cellular organelle integrity, including impaired lysosomal proteostasis and defective mitochondrial oxidative phosphorylation. An open question, however, is the degree to which inherited variation in or near genes encoding each organelle contributes to age-related disease pathogenesis. Here, we evaluate if genetic loci encoding organelle proteomes confer greater-than-expected age-related disease risk. As mitochondrial dysfunction is a 'hallmark' of aging, we begin by assessing nuclear and mitochondrial DNA loci near genes encoding the mitochondrial proteome and surprisingly observe a lack of enrichment across 24 age-related traits. Within nine other organelles, we find no enrichment with one exception: the nucleus, where enrichment emanates from nuclear transcription factors. In agreement, we find that genes encoding several organelles tend to be 'haplosufficient,' while we observe strong purifying selection against heterozygous protein-truncating variants impacting the nucleus. Our work identifies common variation near transcription factors as having outsize influence on age-related trait risk, motivating future efforts to determine if and how this inherited variation then contributes to observed age-related organelle deterioration.
Collapse
Affiliation(s)
- Rahul Gupta
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, United States.,Broad Institute of MIT and Harvard, Cambridge, United States.,Analytic and Translational Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, United States
| | - Konrad J Karczewski
- Broad Institute of MIT and Harvard, Cambridge, United States.,Analytic and Translational Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, United States
| | - Daniel Howrigan
- Broad Institute of MIT and Harvard, Cambridge, United States.,Analytic and Translational Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, United States
| | - Benjamin M Neale
- Broad Institute of MIT and Harvard, Cambridge, United States.,Analytic and Translational Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, United States
| | - Vamsi K Mootha
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, United States.,Broad Institute of MIT and Harvard, Cambridge, United States
| |
Collapse
|
219
|
Curtis D. Analysis of 200,000 Exome-Sequenced UK Biobank Subjects Implicates Genes Involved in Increased and Decreased Risk of Hypertension. Pulse (Basel) 2021; 9:17-29. [PMID: 34722352 PMCID: PMC8527905 DOI: 10.1159/000517419] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/10/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Previous analyses have identified common variants along with some specific genes and rare variants which are associated with risk of hypertension, but much remains to be discovered. METHODS AND RESULTS Exome-sequenced UK Biobank participants were phenotyped based on having a diagnosis of hypertension or taking anti-hypertensive medication to produce a sample of 66,123 cases and 134,504 controls. Variants with minor allele frequency (MAF) <0.01 were subjected to a gene-wise weighted burden analysis, with higher weights assigned to variants which are rarer and/or predicted to have more severe effects. Of 20,384 genes analysed, 2 genes were exome-wide significant, DNMT3A and FES. Also strongly implicated were GUCY1A1 and GUCY1B1, which code for the subunits of soluble guanylate cyclase. There was further support for the previously reported effects of variants in NPR1 and protective effects of variants in DBH. An inframe deletion in CACNA1D with MAF = 0.005, rs72556363, is associated with modestly increased risk of hypertension. Other biologically plausible genes highlighted consist of CSK, AGTR1, ZYX, and PREP. All variants implicated were rare, and cumulatively they are not predicted to make a large contribution to the population risk of hypertension. CONCLUSIONS This approach confirms and clarifies previously reported findings and also offers novel insights into biological processes influencing hypertension risk, potentially facilitating the development of improved therapeutic interventions. This research has been conducted using the UK Biobank Resource.
Collapse
Affiliation(s)
- David Curtis
- UCL Genetics Institute, University College London, London, United Kingdom
- Centre for Psychiatry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
220
|
Boumitri M, Rai NK, Drawz PE. Genetic Risk Scores and Blood Pressure - The Heart is What Matters. KIDNEY360 2021; 2:1209-1211. [PMID: 35369658 PMCID: PMC8676381 DOI: 10.34067/kid.0003272021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/02/2021] [Indexed: 06/14/2023]
Affiliation(s)
- Mirna Boumitri
- Division of Nephrology and Hypertension, University of Minnesota, Minneapolis, Minnesota
| | - Nayanjot K. Rai
- Division of Nephrology and Hypertension, University of Minnesota, Minneapolis, Minnesota
| | - Paul E. Drawz
- Division of Nephrology and Hypertension, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
221
|
Kim K, Kim S, Myung W, Shim I, Lee H, Kim B, Cho SK, Yoon J, Kim DK, Won HH. Shared Genetic Background between Parkinson's Disease and Schizophrenia: A Two-Sample Mendelian Randomization Study. Brain Sci 2021; 11:1042. [PMID: 34439661 PMCID: PMC8393703 DOI: 10.3390/brainsci11081042] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 12/25/2022] Open
Abstract
Background and objectives: Parkinson's disease (PD) and schizophrenia often share symptomatology. Psychotic symptoms are prevalent in patients with PD, and similar motor symptoms with extrapyramidal signs are frequently observed in antipsychotic-naïve patients with schizophrenia as well as premorbid families. However, few studies have examined the relationship between PD and schizophrenia. We performed this study to evaluate whether genetic variants which increase PD risk influence the risk of developing schizophrenia, and vice versa. Materials and Methods: Two-sample Mendelian randomization (TSMR) with summary statistics from large-scale genome-wide association studies (GWAS) was applied. Summary statistics were extracted for these instruments from GWAS of PD and schizophrenia; Results: We found an increase in the risk of schizophrenia per one-standard deviation (SD) increase in the genetically-predicted PD risk (inverse-variance weighted method, odds ratio = 1.10; 95% confidence interval, 1.05-1.15; p = 3.49 × 10-5). The association was consistent in sensitivity analyses, including multiple TSMR methods, analysis after removing outlier variants with potential pleiotropic effects, and analysis after applying multiple GWAS subthresholds. No relationships were evident between PD and smoking or other psychiatric disorders, including attention deficit hyperactivity disorder, autism spectrum disorder, bipolar affective disorder, major depressive disorder, Alzheimer's disease, or alcohol dependence. However, we did not find a reverse relationship; genetic variants increasing schizophrenia risk did not alter the risk of PD; Conclusions: Overall, our findings suggest that increased genetic risk of PD can be associated with increased risk of schizophrenia. This association supports the intrinsic nature of the psychotic symptom in PD rather than medication or environmental effects. Future studies for possible comorbidities and shared genetic structure between the two diseases are warranted.
Collapse
Affiliation(s)
- Kiwon Kim
- Department of Psychiatry, Kangdong Sacred Heart Hospital, College of Medicine, Hallym University, Sungan-ro, Kangdong-gu, Seoul 05355, Korea;
| | - Soyeon Kim
- Samsung Medical Center, Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Korea; (S.K.); (I.S.); (B.K.)
| | - Woojae Myung
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam 13620, Korea;
| | - Injeong Shim
- Samsung Medical Center, Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Korea; (S.K.); (I.S.); (B.K.)
| | - Hyewon Lee
- Department of Health Administration and Management, College of Medical Sciences, Soonchunhyang University, Asan 31538, Korea;
- Department of Software Convergence, Graduate School, Soonchunhyang University, Asan 31538, Korea
| | - Beomsu Kim
- Samsung Medical Center, Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Korea; (S.K.); (I.S.); (B.K.)
| | - Sung Kweon Cho
- Department of Pharmacology, School of Medicine, Ajou University, Worldcup-ro, Yeongtong-gu, Suwon 16499, Korea;
| | - Joohyun Yoon
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam 13620, Korea;
| | - Doh Kwan Kim
- Samsung Medical Center, Department of Psychiatry, School of Medicine, Sungkyunkwan University, Seoul 06351, Korea;
| | - Hong-Hee Won
- Samsung Medical Center, Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Korea; (S.K.); (I.S.); (B.K.)
| |
Collapse
|
222
|
Gemfibrozil derivatives as activators of soluble guanylyl cyclase - A structure-activity study. Eur J Med Chem 2021; 224:113729. [PMID: 34365128 DOI: 10.1016/j.ejmech.2021.113729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 01/02/2023]
Abstract
Previous studies demonstrated that anti-hyperlipidemic drug gemfibrozil acts as NO- and heme-independent activator of NO receptor soluble guanylyl cyclase. A series of new gemfibrozil derivatives were synthesized and evaluated for sGC activation. The structure-activity relationship study identified the positions in gemfibrozil's scaffold that are detrimental for sGC activation and those that are amendable for optimizing modifications. Compared with gemfibrozil, compounds 7c and 15b were more potent activators of cGMP-forming activity of purified sGC and exhibited enhanced relaxation of preconstricted mouse thoracic aorta rings. These studies established the overall framework needed for futher improvement of sGC activators based on gemfibrozil scaffold.
Collapse
|
223
|
Nawaz A, Shilikbay T, Skariah G, Ceman S. Unwinding the roles of RNA helicase MOV10. WILEY INTERDISCIPLINARY REVIEWS-RNA 2021; 13:e1682. [PMID: 34327836 PMCID: PMC8799784 DOI: 10.1002/wrna.1682] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/15/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022]
Abstract
MOV10 is an RNA helicase that associates with the RNA‐induced silencing complex component Argonaute (AGO), likely resolving RNA secondary structures. MOV10 also binds the Fragile X mental retardation protein to block AGO2 binding at some sites and associates with UPF1, a principal component of the nonsense‐mediated RNA decay pathway. MOV10 is widely expressed and has a key role in the cellular response to viral infection and in suppressing retrotransposition. Posttranslational modifications of MOV10 include ubiquitination, which leads to stimulation‐dependent degradation, and phosphorylation, which has an unknown function. MOV10 localizes to the nucleus and/or cytoplasm in a cell type‐specific and developmental stage‐specific manner. Knockout of Mov10 leads to embryonic lethality, underscoring an important role in development where it is required for the completion of gastrulation. MOV10 is expressed throughout the organism; however, most studies have focused on germline cells and neurons. In the testes, the knockdown of Mov10 disrupts proliferation of spermatogonial progenitor cells. In brain, MOV10 is significantly elevated postnatally and binds mRNAs encoding cytoskeleton and neuron projection proteins, suggesting an important role in neuronal architecture. Heterozygous Mov10 mutant mice are hyperactive and anxious and their cultured hippocampal neurons have reduced dendritic arborization. Zygotic knockdown of Mov10 in Xenopus laevis causes abnormal head and eye development and mislocalization of neuronal precursors in the brain. Thus, MOV10 plays a vital role during development, defense against viral infection and in neuronal development and function: its many roles and regulation are only beginning to be unraveled. This article is categorized under:RNA Interactions with Proteins and Other Molecules > RNA‐Protein Complexes RNA Interactions with Proteins and Other Molecules > Protein‐RNA Interactions: Functional Implications
Collapse
Affiliation(s)
- Aatiqa Nawaz
- Department of Cell and Developmental Biology, University of Illinois-Urbana Champaign, Champaign, Illinois, USA
| | - Temirlan Shilikbay
- Department of Cell and Developmental Biology, University of Illinois-Urbana Champaign, Champaign, Illinois, USA
| | - Geena Skariah
- Neuroscience Program, University of Illinois-Urbana Champaign, Champaign, Illinois, USA.,Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Stephanie Ceman
- Department of Cell and Developmental Biology, Neuroscience Program, University of Illinois-Urbana Champaign, Champaign, Illinois, USA
| |
Collapse
|
224
|
Fang X, Liu Y, Xiao W, Zhao N, Zhu C, Yu D, Zhao Y. Prognostic SLC family genes promote cell proliferation, migration, and invasion in hepatocellular carcinoma. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1065-1075. [PMID: 34128989 DOI: 10.1093/abbs/gmab076] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Indexed: 12/13/2022] Open
Abstract
The solute carrier (SLC) superfamily genes encode more than 300 members that are responsible for the transmembrane transportation of many essential endogenous and exogenous compounds ranging from nutrients to drugs. SLCs are highly expressed in metabolic organs such as the liver, regulating the homeostasis of metabolites and the disposition of drugs. In contrast to their well-studied roles in physiological and pharmacological processes, little is known about the relationship between SLCs and cancer progression. Here, we aimed to explore the potential role of SLCs in progression and prognosis of hepatocellular carcinoma (HCC), one of the most commonly diagnosed cancers and leading causes of death worldwide. By performing bioinformatics analyses of HCC dataset from The Cancer Genome Atlas database, we identified three novel signature SLCs (SLC51B, SLC22A15, and SLC2A1) that are indicative of poor prognosis. Further functional analyses suggested the potential regulation of the three prognostic SLCs on cell proliferation and metastasis. Subsequent knockdown experiments performed in HCC cell lines showed that all three prognostic SLCs positively regulated the proliferation of HCC cells, among which SLC22A15 and SLC2A1 were required for migration and invasion of the cells, demonstrating remarkable consistency with the roles identified by bioinformatics methods in HCC. Therefore, our study provides a novel prognostic biomarker for HCC and reveals the significant roles of SLCs in HCC progression, which might have been undervalued in the past.
Collapse
Affiliation(s)
- Xiao Fang
- Jiangsu Key Laboratory of Experimental and Translational Non-coding RNA Research, Medical College, Yangzhou University, Yangzhou 225009, China
- Clinical Medical College, Yangzhou University, Yangzhou 225009, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonosis, Medical College, Yangzhou University, Yangzhou 225009, China
| | - Ying Liu
- Jiangsu Key Laboratory of Experimental and Translational Non-coding RNA Research, Medical College, Yangzhou University, Yangzhou 225009, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
| | - Wangwen Xiao
- Jiangsu Key Laboratory of Experimental and Translational Non-coding RNA Research, Medical College, Yangzhou University, Yangzhou 225009, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
| | - Nan Zhao
- Jiangsu Key Laboratory of Experimental and Translational Non-coding RNA Research, Medical College, Yangzhou University, Yangzhou 225009, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
| | - Chunmiao Zhu
- Jiangsu Key Laboratory of Experimental and Translational Non-coding RNA Research, Medical College, Yangzhou University, Yangzhou 225009, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
| | - Duonan Yu
- Jiangsu Key Laboratory of Experimental and Translational Non-coding RNA Research, Medical College, Yangzhou University, Yangzhou 225009, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonosis, Medical College, Yangzhou University, Yangzhou 225009, China
| | - Ya Zhao
- Jiangsu Key Laboratory of Experimental and Translational Non-coding RNA Research, Medical College, Yangzhou University, Yangzhou 225009, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonosis, Medical College, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
225
|
Song W, Cao H, Zhang D, Xu H, Zhang Q, Wang Z, Li S, Wang W, Hu W, Wang B, Duan H. Association between NR3C1 gene polymorphisms and age-related hearing impairment in Qingdao Chinese elderly. BMC Med Genomics 2021; 14:193. [PMID: 34320993 PMCID: PMC8320226 DOI: 10.1186/s12920-021-01044-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/22/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Age-related hearing impairment (ARHI) has attracted increasing attention recently. It is caused by genetic and environmental factors. A number of ARHI-related genes have been found. This study aimed to detect the potential association between NR3C1 gene polymorphisms and ARHI by means of weighted allele score. METHODS A total of 861 participants from Qingdao city were selected by means of cluster random sampling. We statistically evaluated the characteristics of individuals and used the Mann-Whitney U test or chi-square test for comparison. The publicly available expression quantitative trait locus (eQTL) was queried on the website of the Genotype-Tissue Expression (GTEx). We used the weighted allele score and logistic regression analysis to explore the association between NR3C1 gene polymorphisms and ARHI. Finally, the prediction model was constructed by logistic regression and receiver operating characteristic (ROC) curve. RESULTS All individuals over 60 years of age were enrolled in this study. The allele of rs61757411, rs41423247 and rs6877893 were significantly different between the ARHI group and the normal hearing group (P < 0.01). Though eQTL analysis, rs6877893 and rs33388 might affect the occurrence of ARHI by affecting the expression of NR3C1 gene in artery aorta. Then we performed two models: one without adding any covariates into model and the other adjusting for demographic characteristic, smoking and drinking, diet and exercise, and physical conditions. In the multivariate-adjusted model 2, the odds ratio with 95% confidence interval for weighted allele score (NR3C1) was 0.841 (0.710-0.995, P = 0.043). The area under the ROC curve was 0.755, indicating that the model had good predictability. CONCLUSIONS Our study suggests that NR3C1 gene polymorphisms was significantly associated with ARHI.
Collapse
Affiliation(s)
- Wanxue Song
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 38 Dengzhou Road, Shibei District, Qingdao, 266021, Shandong Province, People's Republic of China
| | - Hainan Cao
- Department of Otorhinolaryngology, Qingdao Municipal Hospital, Qingdao, 266011, Shandong Province, People's Republic of China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 38 Dengzhou Road, Shibei District, Qingdao, 266021, Shandong Province, People's Republic of China
| | - Haiyan Xu
- Chengyang Street Community Health Service Center, No. 137 Wenyang Road, Chengyang District, Qingdao, 266109, Shandong Province, People's Republic of China
| | - Qianqian Zhang
- Zaozhuang Municipal Center for Disease Control and Prevention, No. 223 Jiefang North Road, Shizhong District, Zaozhuang, 277100, Shandong Province, People's Republic of China
| | - Zhaoguo Wang
- Qingdao Municipal Center for Disease Control and Prevention, No. 175 Shandong Road, Shibei District, Qingdao, 266033, Shandong Province, People's Republic of China
- Qingdao Institute of Preventive Medicine, No. 175 Shandong Road, Shibei District, Qingdao, 266033, Shandong Province, People's Republic of China
| | - Suyun Li
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 38 Dengzhou Road, Shibei District, Qingdao, 266021, Shandong Province, People's Republic of China
| | - Weijing Wang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 38 Dengzhou Road, Shibei District, Qingdao, 266021, Shandong Province, People's Republic of China
| | - Wenchao Hu
- Department of Endocrinology, Qilu Hospital of Shandong University (Qingdao), No. 758 Hefei Road, Shibei District, Qingdao, 266033, Shandong Province, People's Republic of China
| | - Bingling Wang
- Qingdao Municipal Center for Disease Control and Prevention, No. 175 Shandong Road, Shibei District, Qingdao, 266033, Shandong Province, People's Republic of China
- Qingdao Institute of Preventive Medicine, No. 175 Shandong Road, Shibei District, Qingdao, 266033, Shandong Province, People's Republic of China
| | - Haiping Duan
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 38 Dengzhou Road, Shibei District, Qingdao, 266021, Shandong Province, People's Republic of China.
- Qingdao Municipal Center for Disease Control and Prevention, No. 175 Shandong Road, Shibei District, Qingdao, 266033, Shandong Province, People's Republic of China.
- Qingdao Institute of Preventive Medicine, No. 175 Shandong Road, Shibei District, Qingdao, 266033, Shandong Province, People's Republic of China.
| |
Collapse
|
226
|
Mineharu Y, Miyamoto S. RNF213 and GUCY1A3 in Moyamoya Disease: Key Regulators of Metabolism, Inflammation, and Vascular Stability. Front Neurol 2021; 12:687088. [PMID: 34381413 PMCID: PMC8350054 DOI: 10.3389/fneur.2021.687088] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
Moyamoya disease is an idiopathic chronically progressive cerebrovascular disease, which causes both ischemic and hemorrhagic stroke. Genetic studies identified RNF213/Mysterin and GUCY1A3 as disease-causing genes. They were also known to be associated with non-moyamoya intracranial large artery disease, coronary artery disease and pulmonary artery hypertension. This review focused on these two molecules and their strong linker, calcineurin/NFAT signaling and caveolin to understand the pathophysiology of moyamoya disease and related vascular diseases. They are important regulators of lipid metabolism especially lipotoxicity, NF-κB mediated inflammation, and nitric oxide-mediated vascular protection. Although intimal thickening with fibrosis and damaged vascular smooth muscle cells are the distinguishing features of moyamoya disease, origin of the fibrous tissue and the mechanism of smooth muscle cell damages remains not fully elucidated. Endothelial cells and smooth muscle cells have long been a focus of interest, but other vascular components such as immune cells and extracellular matrix also need to be investigated in future studies. Molecular research on moyamoya disease would give us a clue to understand the mechanism preserving vascular stability.
Collapse
Affiliation(s)
- Yohei Mineharu
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | |
Collapse
|
227
|
Clergue-Duval V, Nicolas-Sacy L, Karsinti E, Zerdazi EH, Laplanche JL, Brousse G, Marees AT, Derks EM, Henry P, Bellivier F, Vorspan F, Bloch V. Risk and Protective Factors of Lifetime Cocaine-Associated Chest Pain. Front Psychiatry 2021; 12:704276. [PMID: 34366936 PMCID: PMC8335401 DOI: 10.3389/fpsyt.2021.704276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/25/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction: Cocaine users often present with repetitive events of cocaine-associated chest pain (CACP), clinically resembling acute coronary syndromes. The aim of the study is to describe the specific risk factors for CACP. Method: Cocaine users (n = 316) were recruited for a multicenter cross-sectional study. Lifetime CACP history, sociodemographic factors, and lifetime use of cocaine and other substances were assessed. Thirty single nucleotide polymorphisms (SNPs) of NOS3, ROCK2, EDN1, GUCY1A3, and ALDH2 genes, suggested by the literature on coronary spasms, were selected. The associations with CACP history were tested using the chi-square test, Student's t-test and logistic regression. Results: Among the 316 subjects [78.5% men, mean age 37.5 years, (standard-deviation ±8.7)], 190 (60.1%) were daily cocaine users and 103 (32.6%) reported a lifetime CACP history. Among those with a lifetime CACP history, the median was 10 events per individual. In multivariate analysis, lifetime CACP history was associated with daily cocaine use [odds-ratio (OR) 3.24; 95% confidence intervals (1.29-9.33)], rapid route of cocaine use [OR 2.33 (1.20-4.64) vs. intranasal use], and lifetime amphetamine use [daily amphetamine use: OR 2.80 (1.25-6.32) and non-daily amphetamine use: OR 2.14 (1.15-4.04) vs. never used]. Patients with lifetime opioid maintenance treatment (OMT) reported significantly less lifetime CACP history [OR 0.35 (0.16-0.76)]. None of the selected SNPs was associated with CACP history after multiple testing corrections. Conclusions: Clinical variables describing the intensity of stimulant use were positively associated with lifetime CACP history, while OMT was negatively associated with it. Specific harm reduction strategies can target these risk factors.
Collapse
Affiliation(s)
- Virgile Clergue-Duval
- APHP, Département de Psychiatrie et de Médecine Addictologique, Site Lariboisière Fernand-Widal, Groupe hospitalier universitaire APHP Nord - Université de Paris, Paris, France
- Inserm UMRS-1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Université de Paris, Paris, France
- Fédération Hospitalo-Universitaire NOR-SUD Network of Research in Substance Use Disorders, Ile-de-France, France
- Faculté de Médecine, Université de Paris, Paris, France
| | - Louise Nicolas-Sacy
- APHP, Pharmacie, Site Lariboisière Fernand-Widal, Groupe Hospitalier Universitaire APHP Nord - Université de Paris, Paris, France
| | - Emily Karsinti
- APHP, Département de Psychiatrie et de Médecine Addictologique, Site Lariboisière Fernand-Widal, Groupe hospitalier universitaire APHP Nord - Université de Paris, Paris, France
- Inserm UMRS-1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Université de Paris, Paris, France
- Fédération Hospitalo-Universitaire NOR-SUD Network of Research in Substance Use Disorders, Ile-de-France, France
- Laboratoire ClipsyD, Université Paris Nanterre, Nanterre, France
| | - El-Hadi Zerdazi
- Inserm UMRS-1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Université de Paris, Paris, France
- APHP, Service d'addictologie, DMU IMPACT, GHU Mondor, Hôpital Emile ROUX, Limeil Brévannes, France
| | - Jean-Louis Laplanche
- Inserm UMRS-1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Université de Paris, Paris, France
- APHP, Département de Biochimie et Biologie Moléculaire, Site Lariboisière Fernand-Widal, Groupe Hospitalier Universitaire APHP Nord - Université de Paris, Paris, France
- Faculté de Pharmacie, Université de Paris, Paris, France
| | - Georges Brousse
- Service de psychiatrie-addictologie, CHU de Clermont-Ferrand, Université Clermont-Auvergne, Clermont-Ferrand, France
| | - Andries T. Marees
- Department of Economics, School of Business and Economics, VU University Amsterdam, Amsterdam, Netherlands
| | - Eske M. Derks
- Queensland Institute of Medical Research Berghofer, Translational Neurogenomics Group, Brisbane, QLD, Australia
| | - Patrick Henry
- Faculté de Médecine, Université de Paris, Paris, France
- APHP, Département de Cardiologie, Site Lariboisière Fernand-Widal, Groupe Hospitalier Universitaire APHP Nord - Université de Paris, Paris, France
| | - Frank Bellivier
- APHP, Département de Psychiatrie et de Médecine Addictologique, Site Lariboisière Fernand-Widal, Groupe hospitalier universitaire APHP Nord - Université de Paris, Paris, France
- Inserm UMRS-1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Université de Paris, Paris, France
- Fédération Hospitalo-Universitaire NOR-SUD Network of Research in Substance Use Disorders, Ile-de-France, France
- Faculté de Médecine, Université de Paris, Paris, France
| | - Florence Vorspan
- APHP, Département de Psychiatrie et de Médecine Addictologique, Site Lariboisière Fernand-Widal, Groupe hospitalier universitaire APHP Nord - Université de Paris, Paris, France
- Inserm UMRS-1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Université de Paris, Paris, France
- Fédération Hospitalo-Universitaire NOR-SUD Network of Research in Substance Use Disorders, Ile-de-France, France
- Faculté de Médecine, Université de Paris, Paris, France
| | - Vanessa Bloch
- Inserm UMRS-1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Université de Paris, Paris, France
- Fédération Hospitalo-Universitaire NOR-SUD Network of Research in Substance Use Disorders, Ile-de-France, France
- APHP, Pharmacie, Site Lariboisière Fernand-Widal, Groupe Hospitalier Universitaire APHP Nord - Université de Paris, Paris, France
- Faculté de Pharmacie, Université de Paris, Paris, France
| |
Collapse
|
228
|
Brouwers S, Sudano I, Kokubo Y, Sulaica EM. Arterial hypertension. Lancet 2021; 398:249-261. [PMID: 34019821 DOI: 10.1016/s0140-6736(21)00221-x] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/15/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023]
Abstract
Arterial hypertension is the most important contributor to the global burden of disease; however, disease control remains poor. Although the diagnosis of hypertension is still based on office blood pressure, confirmation with out-of-office blood pressure measurements (ie, ambulatory or home monitoring) is strongly recommended. The definition of hypertension differs throughout various guidelines, but the indications for antihypertensive therapy are relatively similar. Lifestyle adaptation is absolutely key in non-pharmacological treatment. Pharmacologically, angiotensin-converting enzyme inhibitors or angiotensin receptor blockers, calcium channel blockers, and diuretics are the first-line agents, with advice for the use of single-pill combination therapy by most guidelines. As a fourth-line agent, spironolactone should be considered. The rapidly evolving field of device-based therapy, especially renal denervation, will further broaden therapeutic options. Despite being a largely controllable condition, the actual rates of awareness, treatment, and control of hypertension are disappointingly low. Further improvements throughout the process of patient screening, diagnosis, treatment, and follow-up need to be urgently addressed.
Collapse
Affiliation(s)
- Sofie Brouwers
- Department of Cardiology, Cardiovascular Center Aalst, OLV Hospital Aalst, Aalst, Belgium; Department of Experimental Pharmacology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium.
| | - Isabella Sudano
- University Heart Center, Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Yoshihiro Kokubo
- Department of Preventive Cardiology, National Cerebral and Cardiovascular Center, Suita, Japan; Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Elisabeth M Sulaica
- Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, TX, USA
| |
Collapse
|
229
|
Nemoto Y, Satoh T, Takahashi T, Hattori T, Konno S, Suzuki S, Sakihara S, Munakata M. Effects of Isometric Handgrip Training on Home Blood Pressure Measurements in Hypertensive Patients: A Randomized Crossover Study. Intern Med 2021; 60:2181-2188. [PMID: 33583887 PMCID: PMC8355381 DOI: 10.2169/internalmedicine.5865-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Objective We aimed to examine the effects of isometric handgrip (IHG) training on home blood pressure (BP) levels in hypertensive Japanese patients undergoing treatment. Methods Fifty-three hypertensive patients (mean age, 61.7 years; 56.6% men) with a home systolic BP ≥135 mmHg and/or a home diastolic BP ≥85 mmHg were randomly assigned to either group A or B. As per the crossover design, group A performed 8 weeks of IHG training, followed by an equivalent training-free, control period, while the reverse protocol was performed by group B. The baseline characteristics were similar between both groups. The individualized daily IHG training comprised four sets of 2-min isometric contractions at 30% of the individual's maximum voluntary contraction capacity, including 1 min of rest between sets, for ≥3 days/week. The outcome measure was morning and evening home BP readings taken over the last 2 weeks of the training and control periods. Results A combined data analysis for both groups showed that IHG training was significantly associated with the lowering of both systolic and diastolic BP in the morning (137.9±9.3 vs. 135.3±9.5 mmHg, p=0.007 and 83.0±9.5 vs. 81.2±9.3 mmHg, p<0.001, respectively) and evening (130.0±10.7 vs. 127.6±10.1 mmHg, p=0.003 and 75.8±10.4 vs. 73.8±9.2 mmHg, p<0.001, respectively), while no significant change was observed after the control period. A larger increase in the maximum grip strength due to IHG training was associated with greater BP reductions. Conclusion An 8-week period of IHG training significantly lowered both the morning and evening home BP in hypertensive Japanese patients undergoing treatment.
Collapse
Affiliation(s)
- Yuki Nemoto
- Research Center for the Promotion of Health and Employment Support, Tohoku Rosai Hospital, Japan
| | - Tomonori Satoh
- Research Center for the Promotion of Health and Employment Support, Tohoku Rosai Hospital, Japan
| | - Takako Takahashi
- Research Center for the Promotion of Health and Employment Support, Tohoku Rosai Hospital, Japan
| | - Tomomi Hattori
- Research Center for Lifestyle-related Disease, Tohoku Rosai Hospital, Japan
| | - Satoshi Konno
- Research Center for Lifestyle-related Disease, Tohoku Rosai Hospital, Japan
- Division of Hypertension, Tohoku Rosai Hospital, Japan
| | | | - Satoru Sakihara
- Division of Diabetes and Endocrinology, Aomori Rosai Hospital, Japan
| | - Masanori Munakata
- Research Center for the Promotion of Health and Employment Support, Tohoku Rosai Hospital, Japan
- Research Center for Lifestyle-related Disease, Tohoku Rosai Hospital, Japan
- Division of Hypertension, Tohoku Rosai Hospital, Japan
| |
Collapse
|
230
|
Li Q, Dibus M, Casey A, Yee CSK, Vargas SO, Luo S, Rosen SM, Madden JA, Genetti CA, Brabek J, Brownstein CA, Kazerounian S, Raby BA, Schmitz-Abe K, Kennedy JC, Fishman MP, Mullen MP, Taylor JM, Rosel D, Agrawal PB. A homozygous stop-gain variant in ARHGAP42 is associated with childhood interstitial lung disease, systemic hypertension, and immunological findings. PLoS Genet 2021; 17:e1009639. [PMID: 34232960 PMCID: PMC8289122 DOI: 10.1371/journal.pgen.1009639] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/19/2021] [Accepted: 06/02/2021] [Indexed: 11/18/2022] Open
Abstract
ARHGAP42 encodes Rho GTPase activating protein 42 that belongs to a member of the GTPase Regulator Associated with Focal Adhesion Kinase (GRAF) family. ARHGAP42 is involved in blood pressure control by regulating vascular tone. Despite these findings, disorders of human variants in the coding part of ARHGAP42 have not been reported. Here, we describe an 8-year-old girl with childhood interstitial lung disease (chILD), systemic hypertension, and immunological findings who carries a homozygous stop-gain variant (c.469G>T, p.(Glu157Ter)) in the ARHGAP42 gene. The family history is notable for both parents with hypertension. Histopathological examination of the proband lung biopsy showed increased mural smooth muscle in small airways and alveolar septa, and concentric medial hypertrophy in pulmonary arteries. ARHGAP42 stop-gain variant in the proband leads to exon 5 skipping, and reduced ARHGAP42 levels, which was associated with enhanced RhoA and Cdc42 expression. This is the first report linking a homozygous stop-gain variant in ARHGAP42 with a chILD disorder, systemic hypertension, and immunological findings in human patient. Evidence of smooth muscle hypertrophy on lung biopsy and an increase in RhoA/ROCK signaling in patient cells suggests the potential mechanistic link between ARHGAP42 deficiency and the development of chILD disorder.
Collapse
Affiliation(s)
- Qifei Li
- Division of Newborn Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Genetics and Genomics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Michal Dibus
- Department of Cell Biology, Charles University in Prague, Viničná 7, Prague, Czech Republic
- Department of Cell Biology, Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, Vestec u Prahy, Czech Republic
| | - Alicia Casey
- Division of Pulmonary Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Christina S. K. Yee
- Division of Immunology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sara O. Vargas
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Shiyu Luo
- Division of Newborn Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Genetics and Genomics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Samantha M. Rosen
- Division of Newborn Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Genetics and Genomics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jill A. Madden
- Division of Genetics and Genomics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Casie A. Genetti
- Division of Genetics and Genomics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jan Brabek
- Department of Cell Biology, Charles University in Prague, Viničná 7, Prague, Czech Republic
- Department of Cell Biology, Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, Vestec u Prahy, Czech Republic
| | - Catherine A. Brownstein
- Division of Genetics and Genomics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Shideh Kazerounian
- Division of Newborn Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Genetics and Genomics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Benjamin A. Raby
- Division of Pulmonary Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Klaus Schmitz-Abe
- Division of Newborn Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Genetics and Genomics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - John C. Kennedy
- Division of Pulmonary Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Martha P. Fishman
- Division of Pulmonary Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mary P. Mullen
- Department of Cardiology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Joan M. Taylor
- Dept. Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Daniel Rosel
- Department of Cell Biology, Charles University in Prague, Viničná 7, Prague, Czech Republic
- Department of Cell Biology, Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, Vestec u Prahy, Czech Republic
| | - Pankaj B. Agrawal
- Division of Newborn Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Genetics and Genomics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
231
|
Geelhoed B, Börschel CS, Niiranen T, Palosaari T, Havulinna AS, Fouodo CJK, Scheinhardt MO, Blankenberg S, Jousilahti P, Kuulasmaa K, Zeller T, Salomaa V, Schnabel RB. Assessment of causality of natriuretic peptides and atrial fibrillation and heart failure: a Mendelian randomization study in the FINRISK cohort. Europace 2021; 22:1463-1469. [PMID: 32830215 PMCID: PMC7544535 DOI: 10.1093/europace/euaa158] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/08/2020] [Indexed: 12/11/2022] Open
Abstract
AIMS Natriuretic peptides are extensively studied biomarkers for atrial fibrillation (AF) and heart failure (HF). Their role in the pathogenesis of both diseases is not entirely understood and previous studies several single-nucleotide polymorphisms (SNPs) at the NPPA-NPPB locus associated with natriuretic peptides have been identified. We investigated the causal relationship between natriuretic peptides and AF as well as HF using a Mendelian randomization approach. METHODS AND RESULTS N-terminal pro B-type natriuretic peptide (NT-proBNP) (N = 6669), B-type natriuretic peptide (BNP) (N = 6674), and mid-regional pro atrial natriuretic peptide (MR-proANP) (N = 6813) were measured in the FINRISK 1997 cohort. N = 30 common SNPs related to NT-proBNP, BNP, and MR-proANP were selected from studies. We performed six Mendelian randomizations for all three natriuretic peptide biomarkers and for both outcomes, AF and HF, separately. Polygenic risk scores (PRSs) based on multiple SNPs were used as genetic instrumental variable in Mendelian randomizations. Polygenic risk scores were significantly associated with the three natriuretic peptides. Polygenic risk scores were not significantly associated with incident AF nor HF. Most cardiovascular risk factors showed significant confounding percentages, but no association with PRS. A causal relation except for small causal betas is unlikely. CONCLUSION In our Mendelian randomization approach, we confirmed an association between common genetic variation at the NPPA-NPPB locus and natriuretic peptides. A strong causal relationship between natriuretic peptides and incidence of AF as well as HF at the community-level was ruled out. Therapeutic approaches targeting natriuretic peptides will therefore very likely work through indirect mechanisms.
Collapse
Affiliation(s)
- Bastiaan Geelhoed
- Department of General and Interventional Cardiology, University Heart Centre Hamburg, Martinistr. 52, 20246 Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Christin S Börschel
- Department of General and Interventional Cardiology, University Heart Centre Hamburg, Martinistr. 52, 20246 Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Teemu Niiranen
- Finnish Institute for Health and Welfare, THL, Helsinki, Finland
| | - Tarja Palosaari
- Finnish Institute for Health and Welfare, THL, Helsinki, Finland
| | - Aki S Havulinna
- Finnish Institute for Health and Welfare, THL, Helsinki, Finland.,Institute for Molecular Medicine Finland (FIMM), Helsinki, Finland
| | - Césaire J K Fouodo
- Institute of Medical Biometry and Statistics, University of Lübeck, University Medical Centre Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Markus O Scheinhardt
- Institute of Medical Biometry and Statistics, University of Lübeck, University Medical Centre Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Stefan Blankenberg
- Department of General and Interventional Cardiology, University Heart Centre Hamburg, Martinistr. 52, 20246 Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Pekka Jousilahti
- Finnish Institute for Health and Welfare, THL, Helsinki, Finland
| | - Kari Kuulasmaa
- Finnish Institute for Health and Welfare, THL, Helsinki, Finland
| | - Tanja Zeller
- Department of General and Interventional Cardiology, University Heart Centre Hamburg, Martinistr. 52, 20246 Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Veikko Salomaa
- Finnish Institute for Health and Welfare, THL, Helsinki, Finland
| | - Renate B Schnabel
- Department of General and Interventional Cardiology, University Heart Centre Hamburg, Martinistr. 52, 20246 Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| |
Collapse
|
232
|
Cheng CF, Hsieh AR, Liang WM, Chen CC, Chen CH, Wu JY, Lin TH, Liao CC, Huang SM, Huang YC, Ban B, Lin YJ, Tsai FJ. Genome-Wide and Candidate Gene Association Analyses Identify a 14-SNP Combination for Hypertension in Patients With Type 2 Diabetes. Am J Hypertens 2021; 34:651-661. [PMID: 33276381 DOI: 10.1093/ajh/hpaa203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/19/2020] [Accepted: 12/02/2020] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND High blood pressure is common and comorbid with type 2 diabetes (T2D). Almost 50% of patients with T2D have high blood pressure. Patients with both conditions of hypertension (HTN) and T2D are at risk for cardiovascular diseases and mortality. The study aim was to investigate genetic risk factors for HTN in T2D patients. METHODS This study included 999 T2D (cohort 1) patients for the first genome scan stage and 922 T2D (cohort 2) patients for the replication stage. Here, we investigated the genetic susceptibility and cumulative weighted genetic risk score for HTN in T2D patients of Han Chinese descent in Taiwan. RESULTS Thirty novel genetic single nucleotide polymorphisms (SNPs) were associated with HTN in T2D after adjusting for age and body mass index (P value <1 × 10-4). Eight blood pressure-related and/or HTN-related genetic SNPs were associated with HTN in T2D after adjusting for age and body mass index (P value <0.05). Linkage disequilibrium and cumulative weighted genetic risk score analyses showed that 14 of the 38 SNPs were associated with risk of HTN in a dose-dependent manner in T2D (Cochran-Armitage trend test: P value <0.0001). The 14-SNP cumulative weighted genetic risk score was also associated with increased regression tendency of systolic blood pressure in T2D (SBP = 122.05 + 0.8 × weighted genetic risk score; P value = 0.0001). CONCLUSIONS A cumulative weighted genetic risk score composed of 14 SNPs is important for HTN, increased tendency of systolic blood pressure, and may contribute to HTN risk in T2D in Taiwan.
Collapse
Affiliation(s)
- Chi-Fung Cheng
- Graduate Institute of Biostatistics, School of Public Health, China Medical University, Taichung, Taiwan
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Ai-Ru Hsieh
- Department of Statistics, Tamkang University, New Taipei City, Taiwan
| | - Wen-Miin Liang
- Graduate Institute of Biostatistics, School of Public Health, China Medical University, Taichung, Taiwan
| | - Ching-Chu Chen
- Division of Endocrinology and Metabolism, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Chien-Hsiun Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Jer-Yuarn Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Ting-Hsu Lin
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Chiu-Chu Liao
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Shao-Mei Huang
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Yu-Chuen Huang
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Bo Ban
- Chinese Research Center for Behavior Medicine in Growth and Development, Jining, Shandong, China
| | - Ying-Ju Lin
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Fuu-Jen Tsai
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Biotechnology and Bioinformatics, Asia University, Taichung, Taiwan
| |
Collapse
|
233
|
Lu J, Wang Y, Hou L, Zuo Z, Zhang N, Wei A. Multimorbidity patterns in old adults and their associated multi-layered factors: a cross-sectional study. BMC Geriatr 2021; 21:372. [PMID: 34147073 PMCID: PMC8214251 DOI: 10.1186/s12877-021-02292-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 05/20/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Influenced by various factors such as socio-demographic characteristics, behavioral lifestyles and socio-cultural environment, the multimorbidity patterns in old adults remain complex. This study aims to identify their characteristics and associated multi-layered factors based on health ecological model. METHODS In 2019, we surveyed a total of 7480 participants aged 60+ by using a multi-stage random cluster sampling method in Shanxi province, China. Latent class analysis was used to discriminate the multimorbidity patterns in old adults, and hierarchical regression was performed to determine the multi-layered factors associated with their various multimorbidity patterns. RESULTS The prevalence of multimorbidity was 34.70% among the old patients with chronic disease. Over half (60.59%) of the patients with multimorbidity had two co-existing chronic diseases. "Degenerative/digestive diseases", "metabolic diseases" and "cardiovascular diseases" were three specific multimorbidity patterns. Behavioral lifestyles-layered factors had the most explanatory power for the three patterns, whose proportions of explanatory power were 54.00, 43.90 and 48.15% individually. But the contributions of other multi-layered factors were different in different patterns; balanced diet, medication adherence, the size of family and friendship network, and different types of basic medical insurance might have the opposite effect on the three multimorbidity patterns (p < 0.05). CONCLUSIONS In management of old patients with multimorbidity, we should prioritize both the "lifestyle change"-centered systematic management strategy and group-customized intervention programs.
Collapse
Affiliation(s)
- Jiao Lu
- School of Management, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi Province, China.
| | - Yuan Wang
- School of Management, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi Province, China
| | - Lihong Hou
- The Second Affiliated Hospital, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Zhenxing Zuo
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Na Zhang
- School of Management, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi Province, China
| | - Anle Wei
- School of Management, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi Province, China
| |
Collapse
|
234
|
Luca CT, Crisan S, Cozma D, Negru A, Lazar MA, Vacarescu C, Trofenciuc M, Rachieru C, Craciun LM, Gaita D, Petrescu L, Mischie A, Iurciuc S. Arterial Hypertension: Individual Therapeutic Approaches-From DNA Sequencing to Gender Differentiation and New Therapeutic Targets. Pharmaceutics 2021; 13:pharmaceutics13060856. [PMID: 34207606 PMCID: PMC8229802 DOI: 10.3390/pharmaceutics13060856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
The aim of this paper is to provide an accurate overview regarding the current recommended approach for antihypertensive treatment. The importance of DNA sequencing in understanding the complex implication of genetics in hypertension could represent an important step in understanding antihypertensive treatment as well as in developing new medical strategies. Despite a pool of data from studies regarding cardiovascular risk factors emphasizing a worse prognosis for female patients rather than male patients, there are also results indicating that women are more likely to be predisposed to the use of antihypertensive medication and less likely to develop uncontrolled hypertension. Moreover, lower systolic blood pressure values are associated with increased cardiovascular risk in women compared to men. The prevalence, awareness and, most importantly, treatment of hypertension is variable in male and female patients, since the mechanisms responsible for this pathology may be different and closely related to gender factors such as the renin–angiotensin system, sympathetic nervous activity, endothelin-1, sex hormones, aldosterone, and the immune system. Thus, gender-related antihypertensive treatment individualization may be a valuable tool in improving female patients’ prognosis.
Collapse
Affiliation(s)
- Constantin-Tudor Luca
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (C.-T.L.); (D.C.); (A.N.); (M.-A.L.); (C.V.); (C.R.); (L.M.C.); (D.G.); (L.P.); (S.I.)
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Simina Crisan
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (C.-T.L.); (D.C.); (A.N.); (M.-A.L.); (C.V.); (C.R.); (L.M.C.); (D.G.); (L.P.); (S.I.)
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Correspondence: (S.C.); (M.T.)
| | - Dragos Cozma
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (C.-T.L.); (D.C.); (A.N.); (M.-A.L.); (C.V.); (C.R.); (L.M.C.); (D.G.); (L.P.); (S.I.)
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Alina Negru
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (C.-T.L.); (D.C.); (A.N.); (M.-A.L.); (C.V.); (C.R.); (L.M.C.); (D.G.); (L.P.); (S.I.)
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Mihai-Andrei Lazar
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (C.-T.L.); (D.C.); (A.N.); (M.-A.L.); (C.V.); (C.R.); (L.M.C.); (D.G.); (L.P.); (S.I.)
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Cristina Vacarescu
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (C.-T.L.); (D.C.); (A.N.); (M.-A.L.); (C.V.); (C.R.); (L.M.C.); (D.G.); (L.P.); (S.I.)
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Mihai Trofenciuc
- Department of Cardiology, “Vasile Goldis” Western University of Arad, Bulevardul Revoluției 94, 310025 Arad, Romania
- Multidisciplinary Heart Research Center, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Correspondence: (S.C.); (M.T.)
| | - Ciprian Rachieru
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (C.-T.L.); (D.C.); (A.N.); (M.-A.L.); (C.V.); (C.R.); (L.M.C.); (D.G.); (L.P.); (S.I.)
- Internal Medicine Department, County Emergency Hospital, 5 Gheorghe Dima Street, 300079 Timisoara, Romania
- Advanced Research Center in Cardiovascular Pathology and Hemostaseology, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Laura Maria Craciun
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (C.-T.L.); (D.C.); (A.N.); (M.-A.L.); (C.V.); (C.R.); (L.M.C.); (D.G.); (L.P.); (S.I.)
| | - Dan Gaita
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (C.-T.L.); (D.C.); (A.N.); (M.-A.L.); (C.V.); (C.R.); (L.M.C.); (D.G.); (L.P.); (S.I.)
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Lucian Petrescu
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (C.-T.L.); (D.C.); (A.N.); (M.-A.L.); (C.V.); (C.R.); (L.M.C.); (D.G.); (L.P.); (S.I.)
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Alexandru Mischie
- Invasive Cardiology Unit, Centre Hospitalier de Montluçon, 03100 Montluçon, France;
| | - Stela Iurciuc
- Department of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (C.-T.L.); (D.C.); (A.N.); (M.-A.L.); (C.V.); (C.R.); (L.M.C.); (D.G.); (L.P.); (S.I.)
- Angiogenesis Research Center, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Multidisciplinary Center for Research, Evaluation, Diagnosis and Therapies in Oral Medicine, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| |
Collapse
|
235
|
Stehle D, Xu MZ, Schomber T, Hahn MG, Schweda F, Feil S, Kraehling JR, Eitner F, Patzak A, Sandner P, Feil R, Bénardeau A. Novel soluble guanylyl cyclase activators increase glomerular cGMP, induce vasodilation and improve blood flow in the murine kidney. Br J Pharmacol 2021; 179:2476-2489. [PMID: 34096053 PMCID: PMC9292672 DOI: 10.1111/bph.15586] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/07/2021] [Accepted: 05/23/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Generation of cGMP via NO-sensitive soluble guanylyl cyclase (sGC) has been implicated in the regulation of renal functions. Chronic kidney disease (CKD) is associated with decreased NO bioavailability, increased oxidative stress and oxidation of sGC to its haem-free form, apo-sGC. Apo-sGC cannot be activated by NO, resulting in impaired cGMP signalling that is associated with chronic kidney disease progression. We hypothesised that sGC activators, which activate apo-sGC independently of NO, increase renal cGMP production under conditions of oxidative stress, thereby improving renal blood flow (RBF) and kidney function. EXPERIMENTAL APPROACH Two novel sGC activators, runcaciguat and BAY-543, were tested on murine kidney. We measured cGMP levels in real time in kidney slices of cGMP sensor mice, vasodilation of pre-constricted glomerular arterioles and RBF in isolated perfused kidneys. Experiments were performed at baseline conditions, under L-NAME-induced NO deficiency, and in the presence of oxidative stress induced by ODQ. KEY RESULTS Mouse glomeruli showed NO-induced cGMP increases. Under baseline conditions, sGC activator did not alter glomerular cGMP concentration or NO-induced cGMP generation. In the presence of ODQ, NO-induced glomerular cGMP signals were markedly reduced, whereas sGC activator induced strong cGMP increases. L-NAME and ODQ pretreated isolated glomerular arterioles were strongly dilated by sGC activator. sGC activator also increased cGMP and RBF in ODQ-perfused kidneys. CONCLUSION AND IMPLICATION sGC activators increase glomerular cGMP, dilate glomerular arterioles and improve RBF under disease-relevant oxidative stress conditions. Therefore, sGC activators represent a promising class of drugs for chronic kidney disease treatment.
Collapse
Affiliation(s)
- Daniel Stehle
- Interfakultäres Institut für Biochemie (IFIB), University of Tübingen, Tübingen, Germany
| | - Min Ze Xu
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tibor Schomber
- Bayer AG, Cardiovascular Research, Pharma Research Center, Wuppertal, Germany
| | - Michael G Hahn
- Bayer AG, Cardiovascular Research, Pharma Research Center, Wuppertal, Germany
| | - Frank Schweda
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | - Susanne Feil
- Interfakultäres Institut für Biochemie (IFIB), University of Tübingen, Tübingen, Germany
| | - Jan R Kraehling
- Bayer AG, Cardiovascular Research, Pharma Research Center, Wuppertal, Germany
| | - Frank Eitner
- Bayer AG, Cardiovascular Research, Pharma Research Center, Wuppertal, Germany.,Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Andreas Patzak
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Peter Sandner
- Bayer AG, Cardiovascular Research, Pharma Research Center, Wuppertal, Germany.,Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| | - Robert Feil
- Interfakultäres Institut für Biochemie (IFIB), University of Tübingen, Tübingen, Germany
| | - Agnès Bénardeau
- Bayer AG, Cardiovascular Research, Pharma Research Center, Wuppertal, Germany.,Novo Nordisk A/S, Cardio-Renal Biology, Måløv, Denmark
| |
Collapse
|
236
|
Foley CN, Mason AM, Kirk PDW, Burgess S. MR-Clust: clustering of genetic variants in Mendelian randomization with similar causal estimates. Bioinformatics 2021; 37:531-541. [PMID: 32915962 PMCID: PMC8088327 DOI: 10.1093/bioinformatics/btaa778] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 08/06/2020] [Accepted: 09/01/2020] [Indexed: 01/22/2023] Open
Abstract
Motivation Mendelian randomization is an epidemiological technique that uses genetic variants as instrumental variables to estimate the causal effect of a risk factor on an outcome. We consider a scenario in which causal estimates based on each variant in turn differ more strongly than expected by chance alone, but the variants can be divided into distinct clusters, such that all variants in the cluster have similar causal estimates. This scenario is likely to occur when there are several distinct causal mechanisms by which a risk factor influences an outcome with different magnitudes of causal effect. We have developed an algorithm MR-Clust that finds such clusters of variants, and so can identify variants that reflect distinct causal mechanisms. Two features of our clustering algorithm are that it accounts for differential uncertainty in the causal estimates, and it includes ‘null’ and ‘junk’ clusters, to provide protection against the detection of spurious clusters. Results Our algorithm correctly detected the number of clusters in a simulation analysis, outperforming methods that either do not account for uncertainty or do not include null and junk clusters. In an applied example considering the effect of blood pressure on coronary artery disease risk, the method detected four clusters of genetic variants. A post hoc hypothesis-generating search suggested that variants in the cluster with a negative effect of blood pressure on coronary artery disease risk were more strongly related to trunk fat percentage and other adiposity measures than variants not in this cluster. Availability and implementation MR-Clust can be downloaded from https://github.com/cnfoley/mrclust. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Christopher N Foley
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SR, UK
| | - Amy M Mason
- Department of Public Health and Primary Care, Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge CB1 8RN, UK
| | - Paul D W Kirk
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SR, UK.,Cambridge Institute of Therapeutic Immunology & Infectious Disease, University of Cambridge, Cambridge CB2 0AW, UK
| | - Stephen Burgess
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SR, UK.,Department of Public Health and Primary Care, Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge CB1 8RN, UK
| |
Collapse
|
237
|
Kolifarhood G, Sabour S, Akbarzadeh M, Sedaghati-Khayat B, Guity K, Rasekhi Dehkordi S, Amiri Roudbar M, Hadaegh F, Azizi F, Daneshpour MS. Genome-wide association study on blood pressure traits in the Iranian population suggests ZBED9 as a new locus for hypertension. Sci Rep 2021; 11:11699. [PMID: 34083597 PMCID: PMC8175429 DOI: 10.1038/s41598-021-90925-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
High blood pressure is the heritable risk factor for cardiovascular and kidney diseases. Genome-wide association studies(GWAS) on blood pressure traits increase our understanding of its underlying genetic basis. However, a large proportion of GWAS was conducted in Europeans, and some roadblocks deprive other populations to benefit from their results. Iranians population with a high degree of genomic specificity has not been represented in international databases to date, so to fill the gap, we explored the effects of 652,919 genomic variants on Systolic Blood Pressure (SBP), Diastolic Blood Pressure (DBP), and Hypertension (HTN) in 7694 Iranian adults aged 18 and over from Tehran Cardiometabolic Genetic Study (TCGS). We identified consistent signals on ZBED9 associated with HTN in the genome-wide borderline threshold after adjusting for different sets of environmental predictors. Moreover, strong signals on ABHD17C and suggestive signals on FBN1 were detected for DBP and SBP, respectively, while these signals were not consistent in different GWA analysis. Our finding on ZBED9 was confirmed for all BP traits by linkage analysis in an independent sample. We found significant associations with similar direction of effects and allele frequency of genetic variants on ZBED9 with DBP (genome-wide threshold) and HTN (nominal threshold) in GWAS summary data of UK Biobank. Although there is no strong evidence to support the function of ZBED9 in blood pressure regulation, it provides new insight into the pleiotropic effects of hypertension and other cardiovascular diseases.
Collapse
Affiliation(s)
- Goodarz Kolifarhood
- Department of Epidemiology, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Siamak Sabour
- Department of Epidemiology, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Akbarzadeh
- Cellular and Molecular Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahareh Sedaghati-Khayat
- Cellular and Molecular Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamran Guity
- Cellular and Molecular Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeid Rasekhi Dehkordi
- Cellular and Molecular Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmoud Amiri Roudbar
- Department of Animal Science, Safiabad-Dezful Agricultural and Natural Resources Research and Education Center, Agricultural Research, Education and Extension Organization (AREEO), Dezful, Iran
| | - Farzad Hadaegh
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam S Daneshpour
- Cellular and Molecular Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
238
|
Gong M, Liu P, Sciurba FC, Stojanov P, Tao D, Tseng GC, Zhang K, Batmanghelich K. Unpaired data empowers association tests. Bioinformatics 2021; 37:785-792. [PMID: 33070196 PMCID: PMC8098021 DOI: 10.1093/bioinformatics/btaa886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 09/07/2020] [Accepted: 10/05/2020] [Indexed: 11/25/2022] Open
Abstract
Motivation There is growing interest in the biomedical research community to incorporate retrospective data, available in healthcare systems, to shed light on associations between different biomarkers. Understanding the association between various types of biomedical data, such as genetic, blood biomarkers, imaging, etc. can provide a holistic understanding of human diseases. To formally test a hypothesized association between two types of data in Electronic Health Records (EHRs), one requires a substantial sample size with both data modalities to achieve a reasonable power. Current association test methods only allow using data from individuals who have both data modalities. Hence, researchers cannot take advantage of much larger EHR samples that includes individuals with at least one of the data types, which limits the power of the association test. Results We present a new method called the Semi-paired Association Test (SAT) that makes use of both paired and unpaired data. In contrast to classical approaches, incorporating unpaired data allows SAT to produce better control of false discovery and to improve the power of the association test. We study the properties of the new test theoretically and empirically, through a series of simulations and by applying our method on real studies in the context of Chronic Obstructive Pulmonary Disease. We are able to identify an association between the high-dimensional characterization of Computed Tomography chest images and several blood biomarkers as well as the expression of dozens of genes involved in the immune system. Availability and implementation Code is available on https://github.com/batmanlab/Semi-paired-Association-Test. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Mingming Gong
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA.,Department of Philosophy, Carnegie Mellon University, Pittsburgh, PA 15213, USA.,School of Mathematics and Statistics, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Peng Liu
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| | - Frank C Sciurba
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| | - Petar Stojanov
- Department of Philosophy, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Dacheng Tao
- Australia School of Computer Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - George C Tseng
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| | - Kun Zhang
- Department of Philosophy, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Kayhan Batmanghelich
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| |
Collapse
|
239
|
de las Fuentes L, Sung YJ, Noordam R, Winkler T, Feitosa MF, Schwander K, Bentley AR, Brown MR, Guo X, Manning A, Chasman DI, Aschard H, Bartz TM, Bielak LF, Campbell A, Cheng CY, Dorajoo R, Hartwig FP, Horimoto ARVR, Li C, Li-Gao R, Liu Y, Marten J, Musani SK, Ntalla I, Rankinen T, Richard M, Sim X, Smith AV, Tajuddin SM, Tayo BO, Vojinovic D, Warren HR, Xuan D, Alver M, Boissel M, Chai JF, Chen X, Christensen K, Divers J, Evangelou E, Gao C, Girotto G, Harris SE, He M, Hsu FC, Kühnel B, Laguzzi F, Li X, Lyytikäinen LP, Nolte IM, Poveda A, Rauramaa R, Riaz M, Rueedi R, Shu XO, Snieder H, Sofer T, Takeuchi F, Verweij N, Ware EB, Weiss S, Yanek LR, Amin N, Arking DE, Arnett DK, Bergmann S, Boerwinkle E, Brody JA, Broeckel U, Brumat M, Burke G, Cabrera CP, Canouil M, Chee ML, Chen YDI, Cocca M, Connell J, de Silva HJ, de Vries PS, Eiriksdottir G, Faul JD, Fisher V, Forrester T, Fox EF, Friedlander Y, Gao H, Gigante B, Giulianini F, Gu CC, Gu D, Harris TB, He J, Heikkinen S, Heng CK, Hunt S, Ikram MA, Irvin MR, Kähönen M, Kavousi M, et alde las Fuentes L, Sung YJ, Noordam R, Winkler T, Feitosa MF, Schwander K, Bentley AR, Brown MR, Guo X, Manning A, Chasman DI, Aschard H, Bartz TM, Bielak LF, Campbell A, Cheng CY, Dorajoo R, Hartwig FP, Horimoto ARVR, Li C, Li-Gao R, Liu Y, Marten J, Musani SK, Ntalla I, Rankinen T, Richard M, Sim X, Smith AV, Tajuddin SM, Tayo BO, Vojinovic D, Warren HR, Xuan D, Alver M, Boissel M, Chai JF, Chen X, Christensen K, Divers J, Evangelou E, Gao C, Girotto G, Harris SE, He M, Hsu FC, Kühnel B, Laguzzi F, Li X, Lyytikäinen LP, Nolte IM, Poveda A, Rauramaa R, Riaz M, Rueedi R, Shu XO, Snieder H, Sofer T, Takeuchi F, Verweij N, Ware EB, Weiss S, Yanek LR, Amin N, Arking DE, Arnett DK, Bergmann S, Boerwinkle E, Brody JA, Broeckel U, Brumat M, Burke G, Cabrera CP, Canouil M, Chee ML, Chen YDI, Cocca M, Connell J, de Silva HJ, de Vries PS, Eiriksdottir G, Faul JD, Fisher V, Forrester T, Fox EF, Friedlander Y, Gao H, Gigante B, Giulianini F, Gu CC, Gu D, Harris TB, He J, Heikkinen S, Heng CK, Hunt S, Ikram MA, Irvin MR, Kähönen M, Kavousi M, Khor CC, Kilpeläinen TO, Koh WP, Komulainen P, Kraja AT, Krieger JE, Langefeld CD, Li Y, Liang J, Liewald DCM, Liu CT, Liu J, Lohman KK, Mägi R, McKenzie CA, Meitinger T, Metspalu A, Milaneschi Y, Milani L, Mook-Kanamori DO, Nalls MA, Nelson CP, Norris JM, O'Connell J, Ogunniyi A, Padmanabhan S, Palmer ND, Pedersen NL, Perls T, Peters A, Petersmann A, Peyser PA, Polasek O, Porteous DJ, Raffel LJ, Rice TK, Rotter JI, Rudan I, Rueda-Ochoa OL, Sabanayagam C, Salako BL, Schreiner PJ, Shikany JM, Sidney SS, Sims M, Sitlani CM, Smith JA, Starr JM, Strauch K, Swertz MA, Teumer A, Tham YC, Uitterlinden AG, Vaidya D, van der Ende MY, Waldenberger M, Wang L, Wang YX, Wei WB, Weir DR, Wen W, Yao J, Yu B, Yu C, Yuan JM, Zhao W, Zonderman AB, Becker DM, Bowden DW, Deary IJ, Dörr M, Esko T, Freedman BI, Froguel P, Gasparini P, Gieger C, Jonas JB, Kammerer CM, Kato N, Lakka TA, Leander K, Lehtimäki T, Magnusson PKE, Marques-Vidal P, Penninx BWJH, Samani NJ, van der Harst P, Wagenknecht LE, Wu T, Zheng W, Zhu X, Bouchard C, Cooper RS, Correa A, Evans MK, Gudnason V, Hayward C, Horta BL, Kelly TN, Kritchevsky SB, Levy D, Palmas WR, Pereira AC, Province MM, Psaty BM, Ridker PM, Rotimi CN, Tai ES, van Dam RM, van Duijn CM, Wong TY, Rice K, Gauderman WJ, Morrison AC, North KE, Kardia SLR, Caulfield MJ, Elliott P, Munroe PB, Franks PW, Rao DC, Fornage M. Gene-educational attainment interactions in a multi-ancestry genome-wide meta-analysis identify novel blood pressure loci. Mol Psychiatry 2021; 26:2111-2125. [PMID: 32372009 PMCID: PMC7641978 DOI: 10.1038/s41380-020-0719-3] [Show More Authors] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 03/19/2020] [Accepted: 03/24/2020] [Indexed: 02/07/2023]
Abstract
Educational attainment is widely used as a surrogate for socioeconomic status (SES). Low SES is a risk factor for hypertension and high blood pressure (BP). To identify novel BP loci, we performed multi-ancestry meta-analyses accounting for gene-educational attainment interactions using two variables, "Some College" (yes/no) and "Graduated College" (yes/no). Interactions were evaluated using both a 1 degree of freedom (DF) interaction term and a 2DF joint test of genetic and interaction effects. Analyses were performed for systolic BP, diastolic BP, mean arterial pressure, and pulse pressure. We pursued genome-wide interrogation in Stage 1 studies (N = 117 438) and follow-up on promising variants in Stage 2 studies (N = 293 787) in five ancestry groups. Through combined meta-analyses of Stages 1 and 2, we identified 84 known and 18 novel BP loci at genome-wide significance level (P < 5 × 10-8). Two novel loci were identified based on the 1DF test of interaction with educational attainment, while the remaining 16 loci were identified through the 2DF joint test of genetic and interaction effects. Ten novel loci were identified in individuals of African ancestry. Several novel loci show strong biological plausibility since they involve physiologic systems implicated in BP regulation. They include genes involved in the central nervous system-adrenal signaling axis (ZDHHC17, CADPS, PIK3C2G), vascular structure and function (GNB3, CDON), and renal function (HAS2 and HAS2-AS1, SLIT3). Collectively, these findings suggest a role of educational attainment or SES in further dissection of the genetic architecture of BP.
Collapse
Affiliation(s)
- Lisa de las Fuentes
- Cardiovascular Division, Department of Medicine, Washington University, St. Louis, MO, 63110, USA.
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Raymond Noordam
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, 2333ZA, The Netherlands
| | - Thomas Winkler
- Department of Genetic Epidemiology, University of Regensburg, 93051, Regensburg, Germany
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63108, USA
| | - Karen Schwander
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Division of Genomic Outcomes, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Alisa Manning
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Hugues Aschard
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, 02115, USA
- Centre de Bioinformatique, Biostatistique et Biologie Intégrative (C3BI), Institut Pasteur, Paris, 75724, France
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Biostatistics and Medicine, University of Washington, Seattle, WA, 98101, USA
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Archie Campbell
- Centre for Genomic & Experimental Medicine, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Ching-Yu Cheng
- Ocular Epidemiology, Singapore Eye Research Institute, Singapore National Ecy Centre, Singapore, 169856, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Rajkumar Dorajoo
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, 138672, Singapore
| | - Fernando P Hartwig
- Postgraduate Programme in Epidemiology, Federal University of Pelotas, Pelotas, RS, 96020-220, Brazil
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK
| | - A R V R Horimoto
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, SP, 5403000, Brazil
| | - Changwei Li
- Epidemiology and Biostatistics, University of Georgia at Athens College of Public Health, Athens, GA, 30602, USA
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, 2333ZA, Netherlands
| | - Yongmei Liu
- Public Health Sciences, Epidemiology and Prevention, Wake Forest University Health Sciences, Winston-Salem, NC, 27157, USA
| | - Jonathan Marten
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Solomon K Musani
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, 39213, USA
| | - Ioanna Ntalla
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Melissa Richard
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 70808, USA
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore, 117549, Singapore
| | - Albert V Smith
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, 48109, USA
- Icelandic Heart Association, Kopavogur, 201, Iceland
| | - Salman M Tajuddin
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Bamidele O Tayo
- Department of Public Health Sciences, Loyola University Chicago, Maywood, IL, 60153, USA
| | - Dina Vojinovic
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Helen R Warren
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
- NIHR Barts Cardiovascular Biomedical Research Unit, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, London, EC1M 6BQ, UK
| | - Deng Xuan
- Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Maris Alver
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, 51010, Estonia
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, 51010, Estonia
| | - Mathilde Boissel
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille, 59000, France
| | - Jin-Fang Chai
- Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore, 117549, Singapore
| | - Xu Chen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Stockholm, 17177, Sweden
| | - Kaare Christensen
- Unit of Epidemiology, Biostatistics and Biodemography, Department of Public Health, Southern Denmark University, Odense, 5000, Denmark
| | - Jasmin Divers
- Biostatistical Sciences, Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, W2 1PG, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Chuan Gao
- Molecular Genetics and Genomics Program, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Giorgia Girotto
- Medical Genetics, Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, 34100, Italy
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", Trieste, 34100, Italy
| | - Sarah E Harris
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Meian He
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fang-Chi Hsu
- Biostatistical Sciences, Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Brigitte Kühnel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Federica Laguzzi
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Xiaoyin Li
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
- Department of Mathematics and Statistics, University of Minnesota, Duluth, MN, 55812, USA
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, 33520, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, University of Tampere, Tampere, 33014, Finland
| | - Ilja M Nolte
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, 9700RB, The Netherlands
| | - Alaitz Poveda
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Skåne, 205 02, Sweden
| | - Rainer Rauramaa
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, 70100, Finland
| | - Muhammad Riaz
- College of Medicine, Biological Sciences and Psychology, Health Sciences, The Infant Mortality and Morbidity Studies (TIMMS), Leicester, LE1 7RH, UK
| | - Rico Rueedi
- Department of Computational Biology, University of Lausanne, Lausanne, 1011, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37203, USA
| | - Harold Snieder
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, 9700RB, The Netherlands
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Fumihiko Takeuchi
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, 1628655, Japan
| | - Niek Verweij
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, 9700, The Netherlands
| | - Erin B Ware
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, 48104, USA
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 9713GZ, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Health), Partner Site Greifswald, 17475, Greifswald, Germany
| | - Lisa R Yanek
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Najaf Amin
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dan E Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Donna K Arnett
- Dean's Office, University of Kentucky College of Public Health, Lexington, KY, 40536, USA
| | - Sven Bergmann
- Department of Computational Biology, University of Lausanne, Lausanne, 1011, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Medicine, University of Washington, Seattle, WA, 98101, USA
| | - Ulrich Broeckel
- Section of Genomic Pediatrics, Department of Pediatrics, Medicine and Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Marco Brumat
- Medical Genetics, Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, 34100, Italy
| | - Gregory Burke
- Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27109, USA
| | - Claudia P Cabrera
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
- NIHR Barts Cardiovascular Biomedical Research Unit, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, London, EC1M 6BQ, UK
| | - Mickaël Canouil
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille, 59000, France
| | - Miao Li Chee
- Statistics Unit, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, 169856, Singapore
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Division of Genomic Outcomes, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Massimiliano Cocca
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", Trieste, 34100, Italy
| | - John Connell
- Ninewells Hospital & Medical School, University of Dundee, Dundee, Scotland, DD1 9SY, UK
| | - H Janaka de Silva
- Department of Medicine, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | | | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, 48104, USA
| | - Virginia Fisher
- Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Terrence Forrester
- Tropical Metabolism Research Unit, Tropical Medicine Research Institute, University of the West Indies, Mona, JMAAW15, Jamaica
| | - Ervin F Fox
- Cardiology, Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Yechiel Friedlander
- Braun School of Public Health, Hebrew University-Hadassah Medical Center, Jerusalem, 91120, Israel
| | - He Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, W2 1PG, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, W2 1PG, UK
| | - Bruna Gigante
- Cardiovascular Unit, Bioclinicum, Department of Medicine, Karolinska Hospital, Stockholm, 17164, Sweden
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd University Hospital, Stockholm, 18288, Sweden
| | | | - Chi Charles Gu
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Dongfeng Gu
- Department of Epidemiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jiang He
- Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, 70112, USA
- Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Sami Heikkinen
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Kuopio, 70211, Finland
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, 70211, Finland
| | - Chew-Kiat Heng
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Khoo Teck Puat - National University Children's Medical Institute, National University Health System, Singapore, 119228, Singapore
| | - Steven Hunt
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, UT, 84108, USA
- Weill Cornell Medicine in Qatar, Doha, Qatar
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marguerite R Irvin
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere, 33521, Finland
- Department of Clinical Physiology, Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, University of Tampere, Tampere, 33014, Finland
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Chiea Chuen Khor
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, 138672, Singapore
- Department of Biochemistry, National University of Singapore, Singapore, 117596, Singapore
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2200, Denmark
- Department of Environmental Medicine and Public Health, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Woon-Puay Koh
- Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore, 117549, Singapore
- Health Services and Systems Research, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Pirjo Komulainen
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, 70100, Finland
| | - Aldi T Kraja
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63108, USA
| | - J E Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, SP, 5403000, Brazil
| | - Carl D Langefeld
- Biostatistical Sciences, Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Yize Li
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jingjing Liang
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - David C M Liewald
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Ching-Ti Liu
- Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Jianjun Liu
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, 138672, Singapore
- Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore, 117549, Singapore
| | - Kurt K Lohman
- Public Health Sciences, Biostatistics and Data Science, Wake Forest University Health Sciences, Winston-Salem, NC, 27157, USA
| | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, 51010, Estonia
| | - Colin A McKenzie
- Tropical Metabolism Research Unit, Tropical Medicine Research Institute, University of the West Indies, Mona, JMAAW15, Jamaica
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
- Institute of Human Genetics, Technische Universität München, 80333, Munich, Germany
| | - Andres Metspalu
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, 51010, Estonia
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, 51010, Estonia
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, VU University Medical Center, Amsterdam, 1081 BT, The Netherlands
| | - Lili Milani
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, 51010, Estonia
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, 2333ZA, Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, 2333ZA, Netherlands
| | - Mike A Nalls
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, 20895, USA
- Data Tecnica International, Glen Echo, MD, 20812, USA
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester, Leicester, LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Jill M Norris
- Department of Epidemiology, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Jeff O'Connell
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
- Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Adesola Ogunniyi
- Department of Medicine, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Sandosh Padmanabhan
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | | | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Stockholm, 17177, Sweden
| | - Thomas Perls
- Department of Medicine, Geriatrics Section, Boston Medical Center, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 85764, Neuherberg, Germany
| | - Astrid Petersmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, 17475, Greifswald, Germany
| | - Patricia A Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ozren Polasek
- University of Split School of Medicine, Split, Croatia
- University Hospital Split, Split, Croatia
- Psychiatric Hospital "Sveti Ivan", Zagreb, Croatia
| | - David J Porteous
- Centre for Genomic & Experimental Medicine, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Leslie J Raffel
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of California, Irvine, CA, 92868, USA
| | - Treva K Rice
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Division of Genomic Outcomes, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Igor Rudan
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH8 9AG, UK
| | | | - Charumathi Sabanayagam
- Ocular Epidemiology, Singapore Eye Research Institute, Singapore National Ecy Centre, Singapore, 169856, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, 169857, Singapore
| | | | - Pamela J Schreiner
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, 55454, USA
| | - James M Shikany
- Division of Preventive Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 25249, USA
| | - Stephen S Sidney
- Division of Research, Kaiser Permanente of Northern California, Oakland, CA, USA
| | - Mario Sims
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, 39213, USA
| | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Medicine, University of Washington, Seattle, WA, 98101, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, 48104, USA
| | - John M Starr
- Alzheimer Scotland Dementia Research Centre, The University of Edinburgh, Edinburgh, EH8 9AZ, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, UK
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
- Institute of Medical Informatics Biometry and Epidemiology, Ludwig-Maximilians-Universitat Munchen, 80539, Munich, Germany
| | - Morris A Swertz
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, 9700RB, The Netherlands
| | - Alexander Teumer
- DZHK (German Centre for Cardiovascular Health), Partner Site Greifswald, 17475, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, 17475, Greifswald, Germany
| | - Yih Chung Tham
- Ocular Epidemiology, Singapore Eye Research Institute, Singapore National Ecy Centre, Singapore, 169856, Singapore
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dhananjay Vaidya
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - M Yldau van der Ende
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, 9700, The Netherlands
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 85764, Neuherberg, Germany
| | - Lihua Wang
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63108, USA
| | - Ya-Xing Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Ophthalmology and Visual Science Key Lab, Beijing Tongren Hospital, Capital Medical University, 100730, Beijing, China
| | - Wen-Bin Wei
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, 100730, Beijing, China
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, 48104, USA
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37203, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Division of Genomic Outcomes, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Bing Yu
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Caizheng Yu
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jian-Min Yuan
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer, , University of Pittsburgh, Pittsburgh, PA, 15232, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Alan B Zonderman
- Behavioral Epidemiology Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Diane M Becker
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Donald W Bowden
- Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Ian J Deary
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Marcus Dörr
- DZHK (German Centre for Cardiovascular Health), Partner Site Greifswald, 17475, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, 17475, Greifswald, Germany
| | - Tõnu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, 51010, Estonia
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Boston, MA, 02142, USA
| | - Barry I Freedman
- Section on Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-, Salem, NC, 27157, USA
| | - Philippe Froguel
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille, 59000, France
- Department of Genomics of Common Disease, Imperial College London, London, W12 0NN, UK
| | - Paolo Gasparini
- Medical Genetics, Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, 34100, Italy
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", Trieste, 34100, Italy
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD e.V.), 85764, Neuherberg, Germany
| | - Jost Bruno Jonas
- Department of Ophthalmology, Medical Faculty Mannheim, University Heidelberg, 68167, Mannheim, Germany
- Beijing Institute of Ophthalmology, Beijing Ophthalmology and Visual Science Key Lab, Beijing Tongren Eye Center, Capital Medical University, 100730, Beijing, China
| | - Candace M Kammerer
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Norihiro Kato
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, 1628655, Japan
| | - Timo A Lakka
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, 70100, Finland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Kuopio, 70211, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, 70211, Finland
| | - Karin Leander
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, 33520, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, University of Tampere, Tampere, 33014, Finland
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Stockholm, 17177, Sweden
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, 1011, Switzerland
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, VU University Medical Center, Amsterdam, 1081 BT, The Netherlands
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Pim van der Harst
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, 9700, The Netherlands
- Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Ultrecht, The Netherlands
| | - Lynne E Wagenknecht
- Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27109, USA
| | - Tangchun Wu
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37203, USA
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Richard S Cooper
- Department of Public Health Sciences, Loyola University Chicago, Maywood, IL, 60153, USA
| | - Adolfo Correa
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, 39213, USA
| | - Michele K Evans
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Bernardo L Horta
- Postgraduate Programme in Epidemiology, Federal University of Pelotas, Pelotas, RS, 96020-220, Brazil
| | - Tanika N Kelly
- Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, 70112, USA
| | - Stephen B Kritchevsky
- Sticht Center for Health Aging and Alzheimer's Prevention, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Daniel Levy
- NHLBI Framingham Heart Study, Framingham, MA, 01702, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, 20892, USA
| | - Walter R Palmas
- Division of General Medicine, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - A C Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, SP, 5403000, Brazil
| | - Michael M Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63108, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Epidemiology, Medicine and Health Services, University of Washington, Seattle, WA, 98101, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, 98101, USA
| | - Paul M Ridker
- Harvard Medical School, Boston, MA, 02115, USA
- Brigham and Women's Hospital, Boston, MA, 02215, USA
| | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - E Shyong Tai
- Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore, 117549, Singapore
- Health Services and Systems Research, Duke-NUS Medical School, Singapore, 169857, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Rob M van Dam
- Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore, 117549, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Tien Yin Wong
- Ocular Epidemiology, Singapore Eye Research Institute, Singapore National Ecy Centre, Singapore, 169856, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Kenneth Rice
- Department of Biostatistics, University of Washington, Seattle, WA, 98195, USA
| | - W James Gauderman
- Biostatistics, Preventive Medicine, University of Southern California, Los Angeles, CA, 90032, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Kari E North
- Epidemiology, University of North Carolina Gilling School of Global Public Health, Chapel Hill, NC, 27514, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Mark J Caulfield
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
- NIHR Barts Cardiovascular Biomedical Research Unit, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, London, EC1M 6BQ, UK
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, W2 1PG, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, W2 1PG, UK
| | - Patricia B Munroe
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
- NIHR Barts Cardiovascular Biomedical Research Unit, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, London, EC1M 6BQ, UK
| | - Paul W Franks
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Skåne, 205 02, Sweden
- Department of Public Health & Clinical Medicine, Umeå University, Umeå, Västerbotten, 901 85, Sweden
| | - Dabeeru C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Myriam Fornage
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 70808, USA
| |
Collapse
|
240
|
Betrie AH, Brock JA, Harraz OF, Bush AI, He GW, Nelson MT, Angus JA, Wright CE, Ayton S. Zinc drives vasorelaxation by acting in sensory nerves, endothelium and smooth muscle. Nat Commun 2021; 12:3296. [PMID: 34075043 PMCID: PMC8169932 DOI: 10.1038/s41467-021-23198-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 04/16/2021] [Indexed: 02/05/2023] Open
Abstract
Zinc, an abundant transition metal, serves as a signalling molecule in several biological systems. Zinc transporters are genetically associated with cardiovascular diseases but the function of zinc in vascular tone regulation is unknown. We found that elevating cytoplasmic zinc using ionophores relaxed rat and human isolated blood vessels and caused hyperpolarization of smooth muscle membrane. Furthermore, zinc ionophores lowered blood pressure in anaesthetized rats and increased blood flow without affecting heart rate. Conversely, intracellular zinc chelation induced contraction of selected vessels from rats and humans and depolarized vascular smooth muscle membrane potential. We demonstrate three mechanisms for zinc-induced vasorelaxation: (1) activation of transient receptor potential ankyrin 1 to increase calcitonin gene-related peptide signalling from perivascular sensory nerves; (2) enhancement of cyclooxygenase-sensitive vasodilatory prostanoid signalling in the endothelium; and (3) inhibition of voltage-gated calcium channels in the smooth muscle. These data introduce zinc as a new target for vascular therapeutics.
Collapse
Affiliation(s)
- Ashenafi H Betrie
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria, Australia
- Cardiovascular Therapeutics Unit, Department of Biochemistry and Pharmacology, The University of Melbourne, Victoria, Australia
- Department of Cardiovascular Surgery & Center for Basic Medical Research, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences; The Institute of Cardiovascular Diseases, Tianjin University, Tianjin; Center for Drug Development, Wannan Medical College, Wuhu, Anhui, China
| | - James A Brock
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Osama F Harraz
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
- Vermont Center for Cardiovascular and Brain Health, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria, Australia
| | - Guo-Wei He
- Department of Cardiovascular Surgery & Center for Basic Medical Research, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences; The Institute of Cardiovascular Diseases, Tianjin University, Tianjin; Center for Drug Development, Wannan Medical College, Wuhu, Anhui, China
| | - Mark T Nelson
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
- Vermont Center for Cardiovascular and Brain Health, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - James A Angus
- Cardiovascular Therapeutics Unit, Department of Biochemistry and Pharmacology, The University of Melbourne, Victoria, Australia
| | - Christine E Wright
- Cardiovascular Therapeutics Unit, Department of Biochemistry and Pharmacology, The University of Melbourne, Victoria, Australia.
| | - Scott Ayton
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria, Australia.
| |
Collapse
|
241
|
Dissecting polygenic signals from genome-wide association studies on human behaviour. Nat Hum Behav 2021; 5:686-694. [PMID: 33986517 DOI: 10.1038/s41562-021-01110-y] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 03/31/2021] [Indexed: 02/03/2023]
Abstract
Genome-wide association studies on human behavioural traits are producing large amounts of polygenic signals with significant predictive power and potentially useful biological clues. Behavioural traits are more distal and are less directly under biological control compared with physical characteristics, which makes the associated genetic effects harder to interpret. The results of genome-wide association studies for human behaviour are likely made up of a composite of signals from different sources. While sample sizes continue to increase, we outline additional steps that need to be taken to better delineate the origin of the increasingly stronger polygenic signals. In addition to genetic effects on the traits themselves, the major sources of polygenic signals are those that are associated with correlated traits, environmental effects and ascertainment bias. Advances in statistical approaches that disentangle polygenic effects from different traits as well as extending data collection to families and social circles with better geographical coverage will probably contribute to filling the gap of knowledge between genetic effects and behavioural outcomes.
Collapse
|
242
|
Abstract
The kexin-like proprotein convertases perform the initial proteolytic cleavages that ultimately generate a variety of different mature peptide and proteins, ranging from brain neuropeptides to endocrine peptide hormones, to structural proteins, among others. In this review, we present a general introduction to proprotein convertase structure and biochemistry, followed by a comprehensive discussion of each member of the kexin-like subfamily of proprotein convertases. We summarize current knowledge of human proprotein convertase insufficiency syndromes, including genome-wide analyses of convertase polymorphisms, and compare these to convertase null and mutant mouse models. These mouse models have illuminated our understanding of the roles specific convertases play in human disease and have led to the identification of convertase-specific substrates; for example, the identification of procorin as a specific PACE4 substrate in the heart. We also discuss the limitations of mouse null models in interpreting human disease, such as differential precursor cleavage due to species-specific sequence differences, and the challenges presented by functional redundancy among convertases in attempting to assign specific cleavages and/or physiological roles. However, in most cases, knockout mouse models have added substantively both to our knowledge of diseases caused by human proprotein convertase insufficiency and to our appreciation of their normal physiological roles, as clearly seen in the case of the furin, proprotein convertase 1/3, and proprotein convertase 5/6 mouse models. The creation of more sophisticated mouse models with tissue- or temporally-restricted expression of specific convertases will improve our understanding of human proprotein convertase insufficiency and potentially provide support for the emerging concept of therapeutic inhibition of convertases.
Collapse
Affiliation(s)
- Manita Shakya
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Iris Lindberg
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
243
|
Sun YV, Liu C, Staimez L, Ali MK, Chang H, Kondal D, Patel S, Jones D, Mohan V, Tandon N, Prabhakaran D, Quyyumi AA, Narayan KMV, Agrawal A. Cardiovascular disease risk and pathophysiology in South Asians: can longitudinal multi-omics shed light? Wellcome Open Res 2021; 5:255. [PMID: 34136649 PMCID: PMC8176264 DOI: 10.12688/wellcomeopenres.16336.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality in South Asia, with rapidly increasing prevalence of hypertension, type 2 diabetes (T2DM) and hyperlipidemia over the last two decades. Atherosclerotic CVD (ASCVD) affects South Asians earlier in life and at lower body weights, which is not fully explained by differential burden of conventional risk factors. Heart failure (HF) is a complex clinical syndrome of heterogeneous structural phenotypes including two major clinical subtypes, HF with preserved (HFpEF) and reduced ejection fraction (HFrEF). The prevalence of HF in South Asians is also rising with other metabolic diseases, and HFpEF develops at younger age and leaner body mass index in South Asians than in Whites. Recent genome-wide association studies, epigenome-wide association studies and metabolomic studies of ASCVD and HF have identified genes, metabolites and pathways associated with CVD traits. However, these findings were mostly driven by samples of European ancestry, which may not accurately represent the CVD risk at the molecular level, and the unique risk profile of CVD in South Asians. Such bias, while formulating hypothesis-driven research studies, risks missing important causal or predictive factors unique to South Asians. Importantly, a longitudinal design of multi-omic markers can capture the life-course risk and natural history related to CVD, and partially disentangle putative causal relationship between risk factors, multi-omic markers and subclinical and clinical ASCVD and HF. In conclusion, combining high-resolution untargeted metabolomics with epigenomics of rigorous, longitudinal design will provide comprehensive unbiased molecular characterization of subclinical and clinical CVD among South Asians. A thorough understanding of CVD-associated metabolomic profiles, together with advances in epigenomics and genomics, will lead to more accurate estimates of CVD progression and stimulate new strategies for improving cardiovascular health.
Collapse
Affiliation(s)
- Yan V. Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
- Department of Biomedical Informatics, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Chang Liu
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Lisa Staimez
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Mohammed K. Ali
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
- Department of Family and Preventive Medicine, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Howard Chang
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | | | - Shivani Patel
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Dean Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | | | - Nikhil Tandon
- All India Institute of Medical Sciences, New Delhi, India
| | | | - Arshed A. Quyyumi
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - K. M. Venkat Narayan
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Anurag Agrawal
- Institute of Genomics and Integrative Biology, Council of Scientific and Industrial Research, New Delhi, India
| |
Collapse
|
244
|
Association between dietary sodium intake and blood pressure variability in Chinese patients with hypertension. Chin Med J (Engl) 2021; 133:1066-1072. [PMID: 32301758 PMCID: PMC7213635 DOI: 10.1097/cm9.0000000000000740] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background The association between dietary sodium intake and blood pressure variability (BPV) in hypertensive patients remains unclear. The objective of this study was to demonstrate whether dietary sodium intake is a predictor of elevated BPV in Chinese patients with hypertension. Methods A total of 235 patients with essential hypertension were enrolled in the Department of Cardiology, Chinese People's Liberation Army (PLA) General Hospital in 2018 to 2019, all of whom underwent 24-h ambulatory blood pressure monitoring. BPV was calculated as the standard deviation (SD), coefficient of variation (CV), variation independent of mean (VIM) of blood pressure measurements, respectively, and divided into diurnal systolic BPV (SBPV), diurnal diastolic BPV (DBPV), nocturnal SBPV, and nocturnal DBPV. 24-h urine samples were collected to measure 24-h urine sodium excretion, which represents dietary sodium intake. The relationship between dietary sodium intake and BPV was analyzed by using Spearman correlations and multiple linear regression analysis. Results Nocturnal SBPV-SD, CV, VIM, and nocturnal DBPV-SD in the high urine sodium excretion group were significantly higher than those in the medium and low urine sodium excretion groups, whereas diurnal SBPV-SD, CV, VIM, diurnal DBPV-SD, CV, VIM, and nocturnal DBPV-CV, VIM were not. Using the Spearman correlation analysis, we found a linear correlation between 24-h urine sodium excretion and nocturnal SBPV-SD, CV, VIM (SD, r = 0.22, P = 0.001; CV, r = 0.17, P = 0.009; VIM, r = 0.16, P = 0.020), nocturnal DBPV-SD (r = 0.21, P = 0.001), respectively. After further adjusting for confounding factors by multiple linear regression, the positive correlations remained between 24-h urine sodium excretion and nocturnal SBPV-SD, CV, VIM (SD, β = 0.224, P < 0.001; CV, β = 0.211, P = 0.001; VIM, β = 0.213, P = 0.001), nocturnal DBPV (SD, β = 0.215, P = 0.001), respectively. Conclusions Dietary sodium intake is associated with nocturnal SBPV in Chinese patients with hypertension.
Collapse
|
245
|
The Road to Better Management in Resistant Hypertension-Diagnostic and Therapeutic Insights. Pharmaceutics 2021; 13:pharmaceutics13050714. [PMID: 34068168 PMCID: PMC8153016 DOI: 10.3390/pharmaceutics13050714] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/07/2021] [Accepted: 05/07/2021] [Indexed: 11/29/2022] Open
Abstract
Resistant hypertension (R-HTN) implies a higher mortality and morbidity compared to non-R-HTN due to increased cardiovascular risk and associated adverse outcomes—greater risk of developing chronic kidney disease, heart failure, stroke and myocardial infarction. R-HTN is considered when failing to lower blood pressure below 140/90 mmHg despite adequate lifestyle measures and optimal treatment with at least three medications, including a diuretic, and usually a blocker of the renin-angiotensin system and a calcium channel blocker, at maximally tolerated doses. Hereby, we discuss the diagnostic and therapeutic approach to a better management of R-HTN. Excluding pseudoresistance, secondary hypertension, white-coat hypertension and medication non-adherence is an important step when diagnosing R-HTN. Most recently different phenotypes associated to R-HTN have been described, specifically refractory and controlled R-HTN and masked uncontrolled hypertension. Optimizing the three-drug regimen, including the diuretic treatment, adding a mineralocorticoid receptor antagonist as the fourth drug, a β-blocker as the fifth drug and an α1-blocker or a peripheral vasodilator as a final option when failing to achieve target blood pressure values are current recommendations regarding the correct management of R-HTN.
Collapse
|
246
|
Zhu N, Swietlik EM, Welch CL, Pauciulo MW, Hagen JJ, Zhou X, Guo Y, Karten J, Pandya D, Tilly T, Lutz KA, Martin JM, Treacy CM, Rosenzweig EB, Krishnan U, Coleman AW, Gonzaga-Jauregui C, Lawrie A, Trembath RC, Wilkins MR, Morrell NW, Shen Y, Gräf S, Nichols WC, Chung WK. Rare variant analysis of 4241 pulmonary arterial hypertension cases from an international consortium implicates FBLN2, PDGFD, and rare de novo variants in PAH. Genome Med 2021; 13:80. [PMID: 33971972 PMCID: PMC8112021 DOI: 10.1186/s13073-021-00891-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 04/19/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a lethal vasculopathy characterized by pathogenic remodeling of pulmonary arterioles leading to increased pulmonary pressures, right ventricular hypertrophy, and heart failure. PAH can be associated with other diseases (APAH: connective tissue diseases, congenital heart disease, and others) but often the etiology is idiopathic (IPAH). Mutations in bone morphogenetic protein receptor 2 (BMPR2) are the cause of most heritable cases but the vast majority of other cases are genetically undefined. METHODS To identify new risk genes, we utilized an international consortium of 4241 PAH cases with exome or genome sequencing data from the National Biological Sample and Data Repository for PAH, Columbia University Irving Medical Center, and the UK NIHR BioResource - Rare Diseases Study. The strength of this combined cohort is a doubling of the number of IPAH cases compared to either national cohort alone. We identified protein-coding variants and performed rare variant association analyses in unrelated participants of European ancestry, including 1647 IPAH cases and 18,819 controls. We also analyzed de novo variants in 124 pediatric trios enriched for IPAH and APAH-CHD. RESULTS Seven genes with rare deleterious variants were associated with IPAH with false discovery rate smaller than 0.1: three known genes (BMPR2, GDF2, and TBX4), two recently identified candidate genes (SOX17, KDR), and two new candidate genes (fibulin 2, FBLN2; platelet-derived growth factor D, PDGFD). The new genes were identified based solely on rare deleterious missense variants, a variant type that could not be adequately assessed in either cohort alone. The candidate genes exhibit expression patterns in lung and heart similar to that of known PAH risk genes, and most variants occur in conserved protein domains. For pediatric PAH, predicted deleterious de novo variants exhibited a significant burden compared to the background mutation rate (2.45×, p = 2.5e-5). At least eight novel pediatric candidate genes carrying de novo variants have plausible roles in lung/heart development. CONCLUSIONS Rare variant analysis of a large international consortium identified two new candidate genes-FBLN2 and PDGFD. The new genes have known functions in vasculogenesis and remodeling. Trio analysis predicted that ~ 15% of pediatric IPAH may be explained by de novo variants.
Collapse
Affiliation(s)
- Na Zhu
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Emilia M Swietlik
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Carrie L Welch
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA
| | - Michael W Pauciulo
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jacob J Hagen
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Xueya Zhou
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Yicheng Guo
- Department of Systems Biology, Columbia University, New York, NY, USA
| | | | - Divya Pandya
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Tobias Tilly
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Katie A Lutz
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jennifer M Martin
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource for Translational Research, Cambridge Biomedical Campus, Cambridge, UK
| | - Carmen M Treacy
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Erika B Rosenzweig
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA
| | - Usha Krishnan
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA
| | - Anna W Coleman
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | - Allan Lawrie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Richard C Trembath
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | - Martin R Wilkins
- National Heart & Lung Institute, Imperial College London, London, UK
| | | | | | | | | | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource for Translational Research, Cambridge Biomedical Campus, Cambridge, UK
- Addenbrooke's Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
- Royal Papworth Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
| | - Yufeng Shen
- Department of Systems Biology, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource for Translational Research, Cambridge Biomedical Campus, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - William C Nichols
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Irving Medical Center, 1150 St. Nicholas Avenue, Room 620, New York, NY, 10032, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
247
|
Eales JM, Jiang X, Xu X, Saluja S, Akbarov A, Cano-Gamez E, McNulty MT, Finan C, Guo H, Wystrychowski W, Szulinska M, Thomas HB, Pramanik S, Chopade S, Prestes PR, Wise I, Evangelou E, Salehi M, Shakanti Y, Ekholm M, Denniff M, Nazgiewicz A, Eichinger F, Godfrey B, Antczak A, Glyda M, Król R, Eyre S, Brown J, Berzuini C, Bowes J, Caulfield M, Zukowska-Szczechowska E, Zywiec J, Bogdanski P, Kretzler M, Woolf AS, Talavera D, Keavney B, Maffia P, Guzik TJ, O'Keefe RT, Trynka G, Samani NJ, Hingorani A, Sampson MG, Morris AP, Charchar FJ, Tomaszewski M. Uncovering genetic mechanisms of hypertension through multi-omic analysis of the kidney. Nat Genet 2021; 53:630-637. [PMID: 33958779 DOI: 10.1038/s41588-021-00835-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/04/2021] [Indexed: 02/02/2023]
Abstract
The kidney is an organ of key relevance to blood pressure (BP) regulation, hypertension and antihypertensive treatment. However, genetically mediated renal mechanisms underlying susceptibility to hypertension remain poorly understood. We integrated genotype, gene expression, alternative splicing and DNA methylation profiles of up to 430 human kidneys to characterize the effects of BP index variants from genome-wide association studies (GWASs) on renal transcriptome and epigenome. We uncovered kidney targets for 479 (58.3%) BP-GWAS variants and paired 49 BP-GWAS kidney genes with 210 licensed drugs. Our colocalization and Mendelian randomization analyses identified 179 unique kidney genes with evidence of putatively causal effects on BP. Through Mendelian randomization, we also uncovered effects of BP on renal outcomes commonly affecting patients with hypertension. Collectively, our studies identified genetic variants, kidney genes, molecular mechanisms and biological pathways of key relevance to the genetic regulation of BP and inherited susceptibility to hypertension.
Collapse
Affiliation(s)
- James M Eales
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Xiao Jiang
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Xiaoguang Xu
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Sushant Saluja
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Artur Akbarov
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Eddie Cano-Gamez
- Department of Human Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - Michelle T McNulty
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA
- The Broad Institute, Cambridge, MA, USA
| | - Christopher Finan
- Institute of Cardiovascular Science, University College London, London, UK
| | - Hui Guo
- Centre for Biostatistics, School of Health Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Wojciech Wystrychowski
- Department of General, Vascular and Transplant Surgery, Medical University of Silesia, Katowice, Poland
| | - Monika Szulinska
- Department of Obesity, Metabolic Disorders Treatment and Clinical Dietetics, Karol Marcinkowski University of Medical Sciences, Poznan, Poland
| | - Huw B Thomas
- Division of Evolution and Genomic Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Sanjeev Pramanik
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
- East Lancashire Hospitals NHS Trust, Blackburn, UK
| | - Sandesh Chopade
- Institute of Cardiovascular Science, University College London, London, UK
| | - Priscilla R Prestes
- Health Innovation and Transformation Centre, School of Science, Psychology and Sport, Federation University Australia, Ballarat, Victoria, Australia
| | - Ingrid Wise
- Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, Queensland, Australia
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Mahan Salehi
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Yusif Shakanti
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Mikael Ekholm
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Matthew Denniff
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Alicja Nazgiewicz
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Felix Eichinger
- Department of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Bradley Godfrey
- Department of Urology and Uro-oncology, Karol Marcinkowski University of Medical Sciences, Poznan, Poland
| | - Andrzej Antczak
- Department of Urology and Uro-oncology, Karol Marcinkowski University of Medical Sciences, Poznan, Poland
| | - Maciej Glyda
- Department of Transplantology and General Surgery Poznan, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Robert Król
- Department of General, Vascular and Transplant Surgery, Medical University of Silesia, Katowice, Poland
| | - Stephen Eyre
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Jason Brown
- Division of Research and Innovation, Manchester University NHS Foundation Trust, Manchester, UK
| | - Carlo Berzuini
- Centre for Biostatistics, School of Health Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - John Bowes
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Mark Caulfield
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research, Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK
| | | | - Joanna Zywiec
- Department of Internal Medicine, Diabetology and Nephrology, Zabrze, Medical University of Silesia, Katowice, Poland
| | - Pawel Bogdanski
- Department of Obesity, Metabolic Disorders Treatment and Clinical Dietetics, Karol Marcinkowski University of Medical Sciences, Poznan, Poland
| | | | - Adrian S Woolf
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Royal Manchester Children's Hospital and Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - David Talavera
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Bernard Keavney
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
- Division of Cardiology and Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Pasquale Maffia
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Internal and Agricultural Medicine, Jagiellonian University College of Medicine, Kraków, Poland
| | - Raymond T O'Keefe
- Division of Evolution and Genomic Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Gosia Trynka
- Department of Human Genetics, Wellcome Sanger Institute, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- National Institute for Health Research, Leicester Biomedical Research Centre, Leicester, UK
| | - Aroon Hingorani
- Institute of Cardiovascular Science, University College London, London, UK
| | - Matthew G Sampson
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA
- The Broad Institute, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Andrew P Morris
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
- Department of Biostatistics, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Fadi J Charchar
- Health Innovation and Transformation Centre, School of Science, Psychology and Sport, Federation University Australia, Ballarat, Victoria, Australia
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- Department of Physiology, University of Melbourne, Parkville, Victoria, Australia
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK.
- Manchester Heart Centre and Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester, UK.
| |
Collapse
|
248
|
Traylor M, Persyn E, Tomppo L, Klasson S, Abedi V, Bakker MK, Torres N, Li L, Bell S, Rutten-Jacobs L, Tozer DJ, Griessenauer CJ, Zhang Y, Pedersen A, Sharma P, Jimenez-Conde J, Rundek T, Grewal RP, Lindgren A, Meschia JF, Salomaa V, Havulinna A, Kourkoulis C, Crawford K, Marini S, Mitchell BD, Kittner SJ, Rosand J, Dichgans M, Jern C, Strbian D, Fernandez-Cadenas I, Zand R, Ruigrok Y, Rost N, Lemmens R, Rothwell PM, Anderson CD, Wardlaw J, Lewis CM, Markus HS. Genetic basis of lacunar stroke: a pooled analysis of individual patient data and genome-wide association studies. Lancet Neurol 2021; 20:351-361. [PMID: 33773637 PMCID: PMC8062914 DOI: 10.1016/s1474-4422(21)00031-4] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 11/06/2020] [Accepted: 01/15/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND The genetic basis of lacunar stroke is poorly understood, with a single locus on 16q24 identified to date. We sought to identify novel associations and provide mechanistic insights into the disease. METHODS We did a pooled analysis of data from newly recruited patients with an MRI-confirmed diagnosis of lacunar stroke and existing genome-wide association studies (GWAS). Patients were recruited from hospitals in the UK as part of the UK DNA Lacunar Stroke studies 1 and 2 and from collaborators within the International Stroke Genetics Consortium. Cases and controls were stratified by ancestry and two meta-analyses were done: a European ancestry analysis, and a transethnic analysis that included all ancestry groups. We also did a multi-trait analysis of GWAS, in a joint analysis with a study of cerebral white matter hyperintensities (an aetiologically related radiological trait), to find additional genetic associations. We did a transcriptome-wide association study (TWAS) to detect genes for which expression is associated with lacunar stroke; identified significantly enriched pathways using multi-marker analysis of genomic annotation; and evaluated cardiovascular risk factors causally associated with the disease using mendelian randomisation. FINDINGS Our meta-analysis comprised studies from Europe, the USA, and Australia, including 7338 cases and 254 798 controls, of which 2987 cases (matched with 29 540 controls) were confirmed using MRI. Five loci (ICA1L-WDR12-CARF-NBEAL1, ULK4, SPI1-SLC39A13-PSMC3-RAPSN, ZCCHC14, ZBTB14-EPB41L3) were found to be associated with lacunar stroke in the European or transethnic meta-analyses. A further seven loci (SLC25A44-PMF1-BGLAP, LOX-ZNF474-LOC100505841, FOXF2-FOXQ1, VTA1-GPR126, SH3PXD2A, HTRA1-ARMS2, COL4A2) were found to be associated in the multi-trait analysis with cerebral white matter hyperintensities (n=42 310). Two of the identified loci contain genes (COL4A2 and HTRA1) that are involved in monogenic lacunar stroke. The TWAS identified associations between the expression of six genes (SCL25A44, ULK4, CARF, FAM117B, ICA1L, NBEAL1) and lacunar stroke. Pathway analyses implicated disruption of the extracellular matrix, phosphatidylinositol 5 phosphate binding, and roundabout binding (false discovery rate <0·05). Mendelian randomisation analyses identified positive associations of elevated blood pressure, history of smoking, and type 2 diabetes with lacunar stroke. INTERPRETATION Lacunar stroke has a substantial heritable component, with 12 loci now identified that could represent future treatment targets. These loci provide insights into lacunar stroke pathogenesis, highlighting disruption of the vascular extracellular matrix (COL4A2, LOX, SH3PXD2A, GPR126, HTRA1), pericyte differentiation (FOXF2, GPR126), TGF-β signalling (HTRA1), and myelination (ULK4, GPR126) in disease risk. FUNDING British Heart Foundation.
Collapse
Affiliation(s)
- Matthew Traylor
- Clinical Pharmacology and The Barts Heart Centre and NIHR Barts Biomedical Research Centre, Barts Health NHS Trust, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Elodie Persyn
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | - Liisa Tomppo
- Department of Neurology, Helsinki University Hospital, Helsinki, Finland
| | - Sofia Klasson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Vida Abedi
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Health System, Danville, PA, USA
| | - Mark K Bakker
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - Nuria Torres
- Stroke Pharmacogenomics and Genetics, Sant Pau Institute of Research, Hospital de la Santa Creu I Sant Pau, Barcelona, Spain
| | - Linxin Li
- Centre for the Prevention of Stroke and Dementia, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Steven Bell
- Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Loes Rutten-Jacobs
- Product Development Personalized Health Care, F Hoffmann-La Roche, Basel, Switzerland
| | - Daniel J Tozer
- Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Christoph J Griessenauer
- Neuroscience Institute, Geisinger Health System, Danville, PA, USA; Institute of Neurointervention, Paracelsus Medical University, Salzburg, Austria
| | - Yanfei Zhang
- Genomic Medicine Institute, Geisinger Health System, Danville, PA, USA
| | - Annie Pedersen
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Pankaj Sharma
- Institute of Cardiovascular Research, Royal Holloway University of London, London, UK
| | - Jordi Jimenez-Conde
- Neurovascular Research Group, Department of Neurology of Hospital del Mar-IMIM (Institut Hospital del Mar d'Investigacions Mediques), Universitat Autonoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
| | - Tatjana Rundek
- Evelyn F McKnight Brain Institute, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Raji P Grewal
- Neuroscience Institute, Saint Francis Medical Center, School of Health and Medical Sciences, Seton Hall University, South Orange, NJ, USA
| | - Arne Lindgren
- Department of Neurology, Skane University Hospital, Lund, Sweden; Department of Clinical Sciences Lund, Neurology, Lund University, Lund, Sweden
| | | | - Veikko Salomaa
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Aki Havulinna
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland; Institute for Molecular Medicine Finland (FIMM HiLIFE), Helsinki, Finland
| | - Christina Kourkoulis
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA; Program in Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Katherine Crawford
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Program in Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Sandro Marini
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Program in Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Braxton D Mitchell
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA; Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Steven J Kittner
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA; Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Jonathan Rosand
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Program in Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christina Jern
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Daniel Strbian
- Department of Neurology, Helsinki University Hospital, Helsinki, Finland; Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| | - Israel Fernandez-Cadenas
- Stroke Pharmacogenomics and Genetics, Sant Pau Institute of Research, Hospital de la Santa Creu I Sant Pau, Barcelona, Spain; Neurovascular Research Laboratory and Neurovascular Unit, Institut de Recerca, Hospital Vall d'Hebron, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Ramin Zand
- Neuroscience Institute, Geisinger Health System, Danville, PA, USA
| | - Ynte Ruigrok
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - Natalia Rost
- J Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Robin Lemmens
- Experimental Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium; VIB Center for Brain & Disease Research, Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Peter M Rothwell
- Centre for the Prevention of Stroke and Dementia, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Christopher D Anderson
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Program in Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Joanna Wardlaw
- Centre for Clinical Brain Sciences, UK Dementia Research Institute and Row Fogo Centre for Research into the Ageing Brain, University of Edinburgh, Edinburgh, UK
| | - Cathryn M Lewis
- Department of Medical and Molecular Genetics, King's College London, London, UK; Social, Genetic, and Developmental Psychiatry Centre, King's College London, London, UK
| | - Hugh S Markus
- Clinical Neurosciences, University of Cambridge, Cambridge, UK.
| |
Collapse
|
249
|
Strojny W, Kwiecińska K, Fijorek K, Korostyński M, Piechota M, Balwierz W, Skoczeń S. Comparison of blood pressure values and expression of genes associated with hypertension in children before and after hematopoietic cell transplantation. Sci Rep 2021; 11:9303. [PMID: 33927307 PMCID: PMC8085120 DOI: 10.1038/s41598-021-88848-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 04/05/2021] [Indexed: 01/19/2023] Open
Abstract
Hypertension is a well-known late effect of hematopoietic cell transplantation (HCT), but no markers predicting its development are known. Our aim was to assess short-term blood pressure (BP) values and expressions of hypertension-associated genes as possible markers of hypertension in children treated with HCT. We measured systolic blood pressure (SBP) and diastolic blood pressure (DBP), using both office procedure and ambulatory BP monitoring (ABPM) in children before HCT and after a median of 6 months after HCT. We compared the results with two control groups, one of healthy children and another of children with simple obesity. We also performed microarray analysis of hypertension-associated genes in patients treated with HCT and children with obesity. We found no significant differences in SBP and DBP in patients before and after HCT. We found significant differences in expressions of certain genes in patients treated with HCT compared with children with obesity. We concluded that BP values in short-term follow-up after HCT do not seem to be useful predictors of hypertension as a late effect of HCT. However, over expressions of certain hypertension-associated genes might be used as markers of hypertension as a late effect of HCT if this is confirmed in larger long-term studies.
Collapse
Affiliation(s)
- Wojciech Strojny
- Department of Oncology and Hematology, University Children's Hospital, Krakow, Poland
| | - Kinga Kwiecińska
- Department of Oncology and Hematology, Institute of Pediatrics, Jagiellonian University Medical College, Wielicka 265, 30-662, Kraków, Poland
| | - Kamil Fijorek
- Department of Statistics, Cracow University of Economics, Kraków, Poland
| | - Michał Korostyński
- Department of Molecular Neuropharmacology, Institute of Pharmacology PAS, Kraków, Poland
| | - Marcin Piechota
- Department of Molecular Neuropharmacology, Institute of Pharmacology PAS, Kraków, Poland
| | - Walentyna Balwierz
- Department of Oncology and Hematology, Institute of Pediatrics, Jagiellonian University Medical College, Wielicka 265, 30-662, Kraków, Poland
| | - Szymon Skoczeń
- Department of Oncology and Hematology, Institute of Pediatrics, Jagiellonian University Medical College, Wielicka 265, 30-662, Kraków, Poland.
| |
Collapse
|
250
|
Association of cytochrome P450 1B1 gene polymorphisms and environmental biomarkers with hypertension in Slovak midlife women. ACTA ACUST UNITED AC 2021; 27:1287-1294. [PMID: 33110045 DOI: 10.1097/gme.0000000000001605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE This study investigated the association of the Leu432Val and Asn453Ser CYP1B1 polymorphisms and selected environmental biomarkers with hypertension (HT) in Slovak midlife women. METHODS We studied 575 women. Divided according to their blood pressure status: 255 with HT and 320 without HT. All data was obtained by using standard anthropometric, genetic methods and analyzed by regression models to adjust for HT risk factors such as age, obesity, smoking, and level of education. RESULTS Our findings revealed that CYP1B1 Leu432Val polymorphism was associated with HT, whereas no association was found between Asn453Ser polymorphism and HT. Women with at least one Val allele had significantly higher odds of HT compared to women with the Leu/Leu genotype in the total sample (Exp(B) = 1.82, CI 1.16-2.84, P = 0.009). After dividing women by menopausal status and the presence of HT environmental risk factor, the association between CYP1B1 polymorphism and HT was observed in pre/perimenopausal women (Exp(B), 2.36; 95% CI 1.13-4.92; P = 0.02), smokers (Exp(B), 3.40; 95% CI 1.48-7.82; P = 0.004), abdominal obesity (Exp(B), 2.41; 95% CI 1.23-4.75; P = 0.01) and in women with only basic education (Exp(B), 4.20, 95% CI 1.12-15.71; P = 0.03). However, general linear models did not reveal a statistically significant interactions between CYP1B1, menopausal status, and HT risk factors and their common association with HT (P > 0.05). CONCLUSIONS In this pilot study, we have provided novel data that supports the significant association of CYP1B1 Leu432Val gene polymorphism with HT in Slovak midlife women.
Collapse
|