1
|
Jones SK, Alberg AJ, Wallace K, Froeliger B, Carpenter MJ, Wolf BJ. CHRNA5-A3-B4, CYP2A6, and DBH Genetic Associations With Smoking Cessation Throughout Adulthood Within Two Longitudinal Studies of Women. Nicotine Tob Res 2025; 27:970-979. [PMID: 39673390 DOI: 10.1093/ntr/ntae284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 11/09/2024] [Accepted: 11/27/2024] [Indexed: 12/16/2024]
Abstract
INTRODUCTION Genetic studies of smoking cessation have been limited by short-term follow-up or cross-sectional design. Within seven genes (CHRNA3, CHRNA5, CHRNB2, CHRNB4, DRD2, DBH, and CYP2A6) influencing biological mechanisms relevant to smoking, this study aimed to identify single nucleotide polymorphisms (SNPs) associated with smoking cessation throughout up to 38 years of follow-up. AIMS AND METHODS Participants were from two all-female cohort studies, Nurses' Health Study (NHS) (n = 10 017) and NHS-2 (n = 2793). For 132 SNPs providing coverage of these genes, genotype associations with the probability of quitting smoking over time were evaluated using generalized estimating equations models. For SNPs reaching nominal statistical significance (p < .05) within NHS, NHS-2 was used as the replication cohort to control for multiple testing (false discovery rate [FDR] < 0.05). SNP genotype by smoking intensity (lifetime light vs. non-light smoking) interactions were also evaluated. RESULTS Five SNPs identified in NHS were replicated in NHS-2 with FDR < 0.05. Women with the minor alleles of CHRNA5 SNPs rs637137 (odd ratio [OR] = 1.21) and rs503464 (OR = 1.24) had increased odds of cessation. Women with the minor alleles of CYP2A6 SNPs rs56113850 (OR = 0.81) and rs56267346 (OR = 0.82) and DBH SNP rs6479643 (OR = 0.78) had lower odds of cessation throughout adulthood. An interaction with smoking intensity was indicated for three SNPs, CHRNB4 rs4887074, CHRNA3 SNP rs77438700, and CHRNA5 SNP rs76474922. CONCLUSION Genetic associations with smoking cessation over decades of follow-up were observed and may guide targeted approaches for smokers most at risk for long-term relapse. IMPLICATIONS This study identified single nucleotide polymorphisms within CHRNA5-A3-B4, CYP2A6, and DBH that were associated with smoking cessation in women over decades of follow-up. This study is the first to examine these genetic associations over years of follow-up. Some associations were novel while others replicated previous findings from short-term studies for the first time. Potential differences in some associations between light and non-light smokers were also observed. Genetic factors associated with long-term smoking behavior may help inform interventions modeled on long-term chronic disease management approaches; specifically, targeted maintenance interventions to sustain abstinence could be implemented among high-risk smokers.
Collapse
Affiliation(s)
| | - Anthony J Alberg
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Kristin Wallace
- Department of Public Health, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Brett Froeliger
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Matthew J Carpenter
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Bethany J Wolf
- Department of Public Health, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
2
|
Hanyuda A, Zeleznik OA, Raita Y, Negishi K, Pasquale LR, Lasky-Su J, Wiggs JL, Kang JH. Machine Learning on Prediagnostic Metabolite Data Identifies Etiologic Endotypes of Exfoliation Glaucoma in United States Health Professionals. OPHTHALMOLOGY SCIENCE 2025; 5:100678. [PMID: 40161462 PMCID: PMC11950771 DOI: 10.1016/j.xops.2024.100678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 04/02/2025]
Abstract
Purpose Exfoliation glaucoma (XFG) etiology is poorly understood. Metabolomics-based etiologic endotypes of XFG may provide novel etiologic insights. We aimed to use unsupervised machine learning on prediagnostic plasma metabolites to characterize etiologic XFG endotypes. Design Prospective case-only analysis. Participants Among Nurses' Health Study and Health Professionals Follow-up Study participants, 205 (174 female and 31 male) incident XFG cases diagnosed with an average of 11.8 years following blood collection (1989-1995) were included. Methods We identified and confirmed incident cases of XFG or XFG suspect (collectively called "XFG" henceforth) through 2016 with medical record review. Liquid chromatography-mass spectrometry was used to profile 341 plasma metabolites. After preprocessing prediagnostic metabolites with adjustment for season, time of blood draw, and fasting status, we computed a distance matrix using Pearson distance and computed gap statistics to identify distinct endotypes. Main Outcome Measures Metabolomics-based XFG etiologic endotypes. Metabolomic profiles were compared across endotypes; false discovery rate (FDR) was used to account for multiple comparisons in Metabolite Set Enrichment Analyses. Exfoliation glaucoma environmental risk factors (e.g., lifetime ultraviolet (UV) exposure, folate consumption), a genetic risk score incorporating 8 major single nucleotide polymorphisms for exfoliation syndrome, and clinical presentations were compared across endotypes. Results We identified 3 distinct XFG metabolomic endotypes. Compared with the most common endotype 2 (reference group [n = 90; 43.9%]), endotype 1 (n = 56; 27.3%) tended to include more male southern US residents with greater UV exposure and were the least likely to have cardiovascular disease; among women, a higher percentage were postmenopausal. Endotype 3 (n = 59; 28.8%) was associated with being a male northern US resident; a higher prevalence of cardiovascular disease and risk factors such as higher body mass index, diabetes, hypertension, and dyslipidemia; and the lowest genetic susceptibility score. There were no differences in ophthalmic characteristics (e.g., maximum intraocular pressure, bilaterality, age at diagnosis) across endotypes (P ≥ 0.6). In metabolite class analyses, compared with endotype 2, organic acids and carnitines were positively associated with endotype 1, whereas diacylglycerols and triacylglycerols were positively associated with endotype 3 (FDR <0.05). Conclusions Integrated metabolomic profiling can identify distinct XFG etiologic endotypes, suggesting different pathobiological mechanisms. Financial Disclosures Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Akiko Hanyuda
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Oana A. Zeleznik
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Yoshihiko Raita
- Department of Nephrology, Okinawa Prefectural Chubu Hospital, Naha, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Louis R. Pasquale
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Janey L. Wiggs
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| | - Jae H. Kang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
3
|
Zhang Z, Zhang Y, Stopsack KH, Kibel AS, Giovannucci EL, Penney KL, Wang A, Vijai J, Kantoff PW, Pomerantz MM, Offit K, Mucci LA, Plym A. Identifying Patients at Risk of Early Lethal Prostate Cancer by Integrating Family History, Polygenic Risk Score, Rare Variants in DNA Repair Genes, and Lifestyle Factors. Eur Urol Oncol 2025:S2588-9311(25)00083-5. [PMID: 40187970 DOI: 10.1016/j.euo.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/13/2025] [Accepted: 03/12/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND AND OBJECTIVE In men with prostate cancer, one-third of deaths occur before the age of 75 yr. There remains a need to characterize heritable and environmental risk factors for these early deaths. This study aims to improve risk stratification for early lethal outcomes among prostate cancer patients with genetic factors beyond family history and with modifiable factors. METHODS This study included 966 prostate cancer patients, enriched for high-risk localized disease and with germline genetic data, in two prospective cohorts. Three genetic factors (family history of prostate cancer, polygenic risk score [PRS] in the top 20%, and rare variants in DNA repair genes) and a lifestyle score were examined for their association with early lethal (metastases/prostate cancer death before the age of 75 yr) compared with nonlethal cases using logistic regression and by calculating 10-yr lethal disease risks. KEY FINDINGS AND LIMITATIONS In total, 289 lethal, including 77 early lethal, cases were observed (median age at the end follow-up: 84.3 yr). Early lethal cases had higher percentages of men with a family history (23% vs 15%), a high PRS (47% vs 36%), and rare variants (14% vs 7.8%). Having two or more genetic factors was strongly associated with increased odds of early lethal disease (odds ratio [OR], 3.5; 95% confidence interval [CI], 1.8-7.0) and linked to higher 10-yr lethal disease risks in high-risk localized patients diagnosed before the age of 75 yr. Healthy men with none of the genetic factors had the lowest odds of early lethal disease (OR, 0.3: 95% CI, 0.1-0.7), compared with unhealthy men with any genetic factor. The pattterns were similar for early fatal disease. The study had limited data for more detailed analyses. CONCLUSIONS AND CLINICAL IMPLICATIONS The combination of family history with rare variants, a PRS, and lifestyle factors may improve the identification of prostate cancer patients at risk of early lethal and fatal disease.
Collapse
Affiliation(s)
- Zhizhu Zhang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Yiwen Zhang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Konrad H Stopsack
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Adam S Kibel
- Department of Urology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Edward L Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kathryn L Penney
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Anqi Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Joseph Vijai
- Niehaus Center for Inherited Cancer Genomics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Philip W Kantoff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Convergent Therapeutics, Inc., Cambridge, MA, USA
| | - Mark M Pomerantz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kenneth Offit
- Niehaus Center for Inherited Cancer Genomics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Anna Plym
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Urology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
4
|
Wu X, Jiang S, Ge A, Turman C, Colditz GA, Tamimi R, Kraft P. Investigating the relationship between breast cancer risk factors and an AI-generated mammographic texture feature in the Nurses' Health Study II. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.18.25322419. [PMID: 40034795 PMCID: PMC11875271 DOI: 10.1101/2025.02.18.25322419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
INTRODUCTION The mammogram risk score (MRS), an AI-driven texture feature derived from digital mammograms, strongly predicts breast cancer risk independently of breast density, though underlying mechanisms remain unclear. This study investigated relationships between established breast cancer risk factors, covering anthropometrics, reproductive factors, family history, and mammographic density metrics, and MRS. METHODS Using data from the Nurses' Health Study II (292 cases, 561 controls), we validated MRS's association with breast cancer using logistic regression and evaluated its relationships with risk factors through: linear regressions of MRS on observed risk factors and polygenic scores associated with risk factors, and Mendelian randomization (MR) analysis via two-stage least squares regression. We conducted two-sample MR of MRS using summary statistics from genome-wide association studies of risk factors. RESULTS MRS was significantly associated with breast cancer risk before adjustment for BI-RADS density (OR=1.92 per SD increase in MRS; 95%CI:1.57-2.33; AUC=0.69) and after (OR=1.85; 95%CI:1.49-2.30). Early life body size and adult body mass index (BMI) were inversely associated with MRS, while history of benign breast disease and BI-RADS density showed positive associations; after adjusting for BI-RADS density, associations between MRS and the other three risk factors attenuated. Higher polygenic score for dense area was associated with increased MRS (ß=0.16 SD increase in MRS per SD increase in polygenic score; 95%CI: 0.06-0.25), as was percent density (ß=0.14; 95%CI:0.05-0.23). Two-sample MR identified associations between genetically predicted dense area (ß=0.83 SD increase in MRS per SD increase in dense area; 95%CI:0.39-1.27) and percent density (ß=1.14; 95%CI:0.55-1.74) with MRS. After adjusting for BI-RADS density and BMI, higher waist-to-hip ratio was significantly associated with increased MRS in polygenic score and two-sample MR analyses. No significant associations were observed with other risk factors. CONCLUSION We validated MRS's association with breast cancer risk in cases diagnosed 0.5-10.1 years (median 2.6) after mammogram acquisition. Our findings reveal robust associations between breast density measures and MRS and suggest a potential impact of central obesity on MRS. Future larger-scale studies are crucial to validate these results and explore their potential to enhance our understanding of breast cancer etiology and refine risk prediction models.
Collapse
|
5
|
Li Y, Li Y, Gu X, Liu Y, Dong D, Kang JH, Wang M, Eliassen H, Willett WC, Stampfer MJ, Wang D. Long-Term Intake of Red Meat in Relation to Dementia Risk and Cognitive Function in US Adults. Neurology 2025; 104:e210286. [PMID: 39813632 PMCID: PMC11735148 DOI: 10.1212/wnl.0000000000210286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/14/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Previous studies have shown inconsistent associations between red meat intake and cognitive health. Our objective was to examine the association between red meat intake and multiple cognitive outcomes. METHODS In this prospective cohort study, we included participants free of dementia at baseline from 2 nationwide cohort studies in the United States: the Nurses' Health Study (NHS) and the Health Professionals Follow-Up Study (HPFS). Diets were assessed using a validated semiquantitative food frequency questionnaire. We ascertained incident dementia cases from both NHS participants (1980-2023) and HPFS participants (1986-2023). Objective cognitive function was assessed using the Telephone Interview for Cognitive Status (1995-2008) among a subset of NHS participants. Subjective cognitive decline (SCD) was self-reported by NHS participants (2012, 2014) and HPFS participants (2012, 2016). Cox proportional hazards models, general linear regression, and Poisson regression models were applied to assess the associations between red meat intake and different cognitive outcomes. RESULTS The dementia analysis included 133,771 participants (65.4% female) with a mean baseline age of 48.9 years, the objective cognitive function analysis included 17,458 female participants with a mean baseline age of 74.3 years, and SCD analysis included 43,966 participants (77.1% female) with a mean baseline age of 77.9 years. Participants with processed red meat intake ≥0.25 serving per day, compared with <0.10 serving per day, had a 13% higher risk of dementia (hazard ratio [HR] 1.13; 95% CI 1.08-1.19; plinearity < 0.001) and a 14% higher risk of SCD (relative risk [RR] 1.14; 95% CI 1.04-1.25; plinearity = 0.004). Higher processed red meat intake was associated with accelerated aging in global cognition (1.61 years per 1 serving per day increment [95% CI 0.20-3.03]) and in verbal memory (1.69 years per 1 serving per day increment [95% CI 0.13-3.25], both plinearity = 0.03). Unprocessed red meat intake of ≥1.00 serving per day, compared with <0.50 serving per day, was associated with a 16% higher risk of SCD (RR 1.16; 95% CI 1.03-1.30; plinearity = 0.04). Replacing 1 serving per day of nuts and legumes for processed red meat was associated with a 19% lower risk of dementia (HR 0.81, 95% CI 0.75-0.86), 1.37 fewer years of cognitive aging (95% CI -2.49 to -0.25), and a 21% lower risk of SCD (RR 0.79, 95% CI 0.68-0.92). DISCUSSION Higher intake of red meat, particularly processed red meat, was associated with a higher risk of developing dementia and worse cognition. Reducing red meat consumption could be included in dietary guidelines to promote cognitive health. Further research is needed to assess the generalizability of these findings to populations with diverse ethnic backgrounds.
Collapse
Affiliation(s)
- Yuhan Li
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Yanping Li
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Xiao Gu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Yuxi Liu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA; and
| | - Danyue Dong
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA; and
| | - Jae Hee Kang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Molin Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Heather Eliassen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Walter C Willett
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Meir J Stampfer
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Dong Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA; and
| |
Collapse
|
6
|
Huang Y, Zhang W, Chen J, Qiu S, Xue C, Wu H. Skimmed milk intake reduces the risk of ER- breast cancer: a Mendelian randomization analysis. Discov Oncol 2024; 15:612. [PMID: 39487869 PMCID: PMC11531460 DOI: 10.1007/s12672-024-01448-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/11/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND In prior observational investigations, it has been demonstrated that the consumption of milk is associated with the incidence of breast cancer (BC). Despite the existence of a two-sample Mendelian randomization (MR) that suggests a causal relationship between milk intake and breast cancer risk, the outcomes still lack a definitive conclusion. This ambiguity may be attributed to variables such as the variety of milk ingested, estrogen levels, the specific type of BC, and potential confounding factors. Therefore, our principal objective is to establish the causal association between the consumption of skimmed milk and full cream milk and the risk of different types of BC through the utilization of two-sample and two-step MR analyses. METHODS In this study, we analyzed single nucleotide polymorphisms associated with skimmed and full-cream milk consumption in a cohort of 360,806 individuals from European populations through genome-wide association studies. We conducted a two-sample MR analysis using three different methods: inverse variance weighting (IVW) was used as the main analysis, and MR-Egger and Weighted median were used as supplementary analyses to IVW. We also performed sensitivity analysis, which included leave-one-out analysis, Cochran's Q test to detect heterogeneity, and MR-Egger intercept analysis to detect potential biases caused by pleiotropy. We used two-step MR analysis to evaluate potential mediators of associations. RESULTS In the two-sample MR analysis, IVW analysis suggests a potential inverse causal relationship between skimmed milk and BC [OR 0.34, 95% CI (0.12-1.00), P = 0.05]. Subgroup analysis revealed that skimmed milk reduces the risk of estrogen receptor-negative (ER-) breast cancer [OR 0.18, 95% CI (0.04-0.90), P = 0.04], but not estrogen receptor-positive (ER+) breast cancer [OR 0.42, 95% CI (0.15-1.22), P = 0.11]. MR Egger reached similar results, that is, skimmed milk reduces the risk of ER- breast cancer [OR 0.006, 95% CI (0.00-0.70), P = 0.04], but not BC [OR 0.16, 95% CI (0.01-4.66), P = 0.30] and ER+ breast cancer [OR 0.50, 95% CI (0.02-12.61), P = 0.65]. Additionally, we found no causal relationship between full cream milk and BC (P > 0.05). In two-step MR analysis, we found evidence for a mediating role of BMI in the relationship between skimmed milk intake and ER-breast cancer risk. CONCLUSION Our findings strengthen the evidence for a protective effect of skimmed milk consumption on ER-breast cancer risk. Further two-step MR analyses suggest that this protective effect may partly result from body mass index (BMI). There is no evidence that full cream milk consumption affects the risk of BC.
Collapse
Affiliation(s)
- Yingdan Huang
- Department of Lymphoma Medicine, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, China
| | - Wangjin Zhang
- Department of Ultrasound, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, 364000, China
| | - Jinghui Chen
- Department of Ultrasound, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, 364000, China
| | - Sihua Qiu
- Department of Ultrasound, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, 364000, China
| | - Chang Xue
- Department of Lymphoma Medicine, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, China.
| | - Huijing Wu
- Department of Lymphoma Medicine, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, China.
| |
Collapse
|
7
|
Major TJ, Takei R, Matsuo H, Leask MP, Sumpter NA, Topless RK, Shirai Y, Wang W, Cadzow MJ, Phipps-Green AJ, Li Z, Ji A, Merriman ME, Morice E, Kelley EE, Wei WH, McCormick SPA, Bixley MJ, Reynolds RJ, Saag KG, Fadason T, Golovina E, O'Sullivan JM, Stamp LK, Dalbeth N, Abhishek A, Doherty M, Roddy E, Jacobsson LTH, Kapetanovic MC, Melander O, Andrés M, Pérez-Ruiz F, Torres RJ, Radstake T, Jansen TL, Janssen M, Joosten LAB, Liu R, Gaal OI, Crişan TO, Rednic S, Kurreeman F, Huizinga TWJ, Toes R, Lioté F, Richette P, Bardin T, Ea HK, Pascart T, McCarthy GM, Helbert L, Stibůrková B, Tausche AK, Uhlig T, Vitart V, Boutin TS, Hayward C, Riches PL, Ralston SH, Campbell A, MacDonald TM, Nakayama A, Takada T, Nakatochi M, Shimizu S, Kawamura Y, Toyoda Y, Nakaoka H, Yamamoto K, Matsuo K, Shinomiya N, Ichida K, Lee C, Bradbury LA, Brown MA, Robinson PC, Buchanan RRC, Hill CL, Lester S, Smith MD, Rischmueller M, Choi HK, Stahl EA, Miner JN, Solomon DH, Cui J, Giacomini KM, Brackman DJ, Jorgenson EM, Liu H, Susztak K, Shringarpure S, So A, Okada Y, Li C, Shi Y, Merriman TR. A genome-wide association analysis reveals new pathogenic pathways in gout. Nat Genet 2024; 56:2392-2406. [PMID: 39406924 DOI: 10.1038/s41588-024-01921-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 08/21/2024] [Indexed: 10/18/2024]
Abstract
Gout is a chronic disease that is caused by an innate immune response to deposited monosodium urate crystals in the setting of hyperuricemia. Here, we provide insights into the molecular mechanism of the poorly understood inflammatory component of gout from a genome-wide association study (GWAS) of 2.6 million people, including 120,295 people with prevalent gout. We detected 377 loci and 410 genetically independent signals (149 previously unreported loci in urate and gout). An additional 65 loci with signals in urate (from a GWAS of 630,117 individuals) but not gout were identified. A prioritization scheme identified candidate genes in the inflammatory process of gout, including genes involved in epigenetic remodeling, cell osmolarity and regulation of NOD-like receptor protein 3 (NLRP3) inflammasome activity. Mendelian randomization analysis provided evidence for a causal role of clonal hematopoiesis of indeterminate potential in gout. Our study identifies candidate genes and molecular processes in the inflammatory pathogenesis of gout suitable for follow-up studies.
Collapse
Affiliation(s)
- Tanya J Major
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Riku Takei
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
- Department of Biomedical Information Management, National Defense Medical College Research Institute, National Defense Medical College, Saitama, Japan
| | - Megan P Leask
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nicholas A Sumpter
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ruth K Topless
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Yuya Shirai
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Wei Wang
- Genomics R&D, 23andMe, Inc, Sunnyvale, CA, USA
| | - Murray J Cadzow
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | | | - Zhiqiang Li
- The Biomedical Sciences Institute and The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
| | - Aichang Ji
- Shandong Provincial Key Laboratory of Metabolic Diseases, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- The Institute of Metabolic Diseases, Qingdao University, Qingdao, Shandong, China
| | - Marilyn E Merriman
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Emily Morice
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eric E Kelley
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - Wen-Hua Wei
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
- Department of Women's and Children's Health, University of Otago, Dunedin, New Zealand
| | | | - Matthew J Bixley
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Richard J Reynolds
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kenneth G Saag
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tayaza Fadason
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Evgenia Golovina
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Justin M O'Sullivan
- Liggins Institute, University of Auckland, Auckland, New Zealand
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, United Kingdom
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore, Singapore
- Australian Parkinsons Mission, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Lisa K Stamp
- Department of Medicine, University of Otago, Christchurch, Christchurch, New Zealand
| | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Abhishek Abhishek
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Michael Doherty
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Edward Roddy
- School of Medicine, Keele University, Keele, Staffordshire, United Kingdom
- Haywood Academic Rheumatology Centre, Midlands Partnership University NHS Foundation Trust, Stoke-on-Trent, UK
| | - Lennart T H Jacobsson
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Meliha C Kapetanovic
- Department of Clinical Sciences Lund, Section of Rheumatology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Emergency and Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Mariano Andrés
- Rheumatology Department, Dr Balmis General University Hospital-ISABIAL, Alicante, Spain
- Department of Clinical Medicine, Miguel Hernandez University, Alicante, Spain
| | - Fernando Pérez-Ruiz
- Osakidetza, OSI-EE-Cruces, BIOBizkaia Health Research Institute and Medicine Department of Medicine and Nursery School, University of the Basque Country, Biskay, Spain
| | - Rosa J Torres
- Department of Biochemistry, Hospital La Paz Institute for Health Research (IdiPaz), Madrid, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Timothy Radstake
- Department of Rheumatology and Clinical Immunology, University Medical Center, Utrecht, The Netherlands
| | - Timothy L Jansen
- Department of Rheumatology, VieCuri Medical Centre, Venlo, The Netherlands
| | - Matthijs Janssen
- Department of Rheumatology, VieCuri Medical Centre, Venlo, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Institute of Molecular Life Science, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ruiqi Liu
- Department of Internal Medicine and Radboud Institute of Molecular Life Science, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Orsolya I Gaal
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Tania O Crişan
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Simona Rednic
- Department of Rheumatology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Cluj, Romania
| | - Fina Kurreeman
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom W J Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - René Toes
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Frédéric Lioté
- Rheumatology Department, Feel'Gout, GH Paris Saint Joseph, Paris, France
- Rheumatology Department, INSERM U1132, BIOSCAR, University Paris Cité, Lariboisière Hospital, Paris, France
| | - Pascal Richette
- Rheumatology Department, INSERM U1132, BIOSCAR, University Paris Cité, Lariboisière Hospital, Paris, France
| | - Thomas Bardin
- Rheumatology Department, INSERM U1132, BIOSCAR, University Paris Cité, Lariboisière Hospital, Paris, France
| | - Hang Korng Ea
- Rheumatology Department, INSERM U1132, BIOSCAR, University Paris Cité, Lariboisière Hospital, Paris, France
| | - Tristan Pascart
- Department of Rheumatology, Hopital Saint-Philibert, Lille Catholic University, Lille, France
| | - Geraldine M McCarthy
- Department of Rheumatology, Mater Misericordiae University Hospital and School of Medicine, University College, Dublin, Ireland
| | - Laura Helbert
- Department of Rheumatology, Mater Misericordiae University Hospital and School of Medicine, University College, Dublin, Ireland
| | - Blanka Stibůrková
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- Institute of Rheumatology, Prague, Czech Republic
| | - Anne-K Tausche
- Department of Rheumatology, University Clinic 'Carl Gustav Carus' at the Technical University, Dresden, Germany
| | - Till Uhlig
- Center for Treatment of Rheumatic and Musculoskeletal Diseases, Diakonhjemmet Hospital, Oslo, Norway
| | - Véronique Vitart
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Thibaud S Boutin
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Philip L Riches
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Stuart H Ralston
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Thomas M MacDonald
- MEMO Research, Division of Molecular and Clinical Medicine, University of Dundee Medical School, Ninewells Hospital, Dundee, United Kingdom
| | - Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Seiko Shimizu
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Yusuke Kawamura
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
- Department of Cancer Genome Research, Sasaki Institute, Sasaki Foundation, Tokyo, Japan
| | - Yu Toyoda
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Hirofumi Nakaoka
- Department of Cancer Genome Research, Sasaki Institute, Sasaki Foundation, Tokyo, Japan
| | - Ken Yamamoto
- Department of Medical Biochemistry, Kurume University School of Medicine, Fukuoka, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology & Prevention, Aichi Cancer Center, Aichi, Japan
- Division of Cancer Epidemiology, Nagoya University Graduate School of Medicine, Aichi, Japan
- The Japan Multi-Institutional Collaborative Cohort (J-MICC) Study, Tokyo, Japan
| | - Nariyoshi Shinomiya
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Kimiyoshi Ichida
- Department of Pathophysiology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Chaeyoung Lee
- Department of Bioinformatics and Life Science, Soongsil University, Seoul, South Korea
| | - Linda A Bradbury
- Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Matthew A Brown
- Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Philip C Robinson
- School of Clinical Medicine, Faculty of Medicine, University of Queensland, Brisbane, Australia
| | | | - Catherine L Hill
- Rheumatology Department, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | - Susan Lester
- Rheumatology Department, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | | | - Maureen Rischmueller
- Rheumatology Department, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | - Hyon K Choi
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eli A Stahl
- Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeff N Miner
- Viscient Biosciences, 5752 Oberlin Dr., Suite 111, San Diego, CA, 92121, USA
| | - Daniel H Solomon
- Division of Rheumatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jing Cui
- Division of Rheumatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences and Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Deanna J Brackman
- Department of Bioengineering and Therapeutic Sciences and Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Eric M Jorgenson
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Hongbo Liu
- Penn / The Children's Hospital of Pennsylvania Kidney Innovation Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19101, USA
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19101, USA
| | - Katalin Susztak
- Penn / The Children's Hospital of Pennsylvania Kidney Innovation Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19101, USA
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19101, USA
| | | | - Alexander So
- Service of Rheumatology, Center Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- University of Lausanne, Lausanne, Switzerland
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Changgui Li
- Shandong Provincial Key Laboratory of Metabolic Diseases, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- The Institute of Metabolic Diseases, Qingdao University, Qingdao, Shandong, China
| | - Yongyong Shi
- Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Tony R Merriman
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA.
- The Institute of Metabolic Diseases, Qingdao University, Qingdao, Shandong, China.
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
8
|
Bae JH, Kang H. Longitudinal Analysis of Sweet Taste Preference Through Genetic and Phenotypic Data Integration. Foods 2024; 13:3370. [PMID: 39517154 PMCID: PMC11545761 DOI: 10.3390/foods13213370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/09/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Understanding the genetic basis of sweet taste preference is crucial for potential implications in diet-related health outcomes, such as obesity. This study identified genes and single nucleotide polymorphisms (SNPs) associated with sweet taste preferences over time. Data from the American Nurses' Health Study (NHS1) and Health Professionals Follow-up Study (HPFS) cohorts were analyzed. Using tools like PLINK and METAL for genetic associations and FUMA for functional annotation, the study identified eight SNPs associated with sweet taste preferences. Notably, rs80115239 and rs12878143 were identified as key determinants of the highest and lowest associations with sweet taste preferences, respectively. Individuals with the rs80115239 (AA) genotype displayed a higher preference for sweet tastes, including chocolate and cake, but a lower preference for physical activity, fruits, and vegetables, particularly in females from the NHS1 cohort, linking this genotype to a higher obesity risk. Conversely, those with the rs12878143 (CC) genotype preferred fruits, vegetables, coffee, and tea, with a lower preference for sweetened beverages, but the correlation with obesity risk was less clear due to inconsistent data. In conclusion, these findings highlight the genetic influences on sweet taste preference and their potential role in personalized dietary recommendations and obesity management strategies.
Collapse
Affiliation(s)
| | - Hyunju Kang
- Department of Food Science and Nutrition, Keimyung University, Daegu 42601, Republic of Korea;
| |
Collapse
|
9
|
Liu Y, Peng C, Brorson IS, O'Mahony DG, Kelly RL, Heng YJ, Baker GM, Grenaker Alnæs GI, Bodelon C, Stover DG, Van Allen EM, Eliassen AH, Kristensen VN, Tamimi RM, Kraft P. Germline polygenic risk scores are associated with immune gene expression signature and immune cell infiltration in breast cancer. Am J Hum Genet 2024; 111:2150-2163. [PMID: 39270649 PMCID: PMC11480808 DOI: 10.1016/j.ajhg.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 09/15/2024] Open
Abstract
The tumor immune microenvironment (TIME) plays key roles in tumor progression and response to immunotherapy. Previous studies have identified individual germline variants associated with differences in TIME. Here, we hypothesize that common variants associated with breast cancer risk or cancer-related traits, represented by polygenic risk scores (PRSs), may jointly influence immune features in TIME. We derived 154 immune traits from bulk gene expression profiles of 764 breast tumors and 598 adjacent normal tissue samples from 825 individuals with breast cancer in the Nurses' Health Study (NHS) and NHSII. Immunohistochemical staining of four immune cell markers were available for a subset of 205 individuals. Germline PRSs were calculated for 16 different traits including breast cancer, autoimmune diseases, type 2 diabetes, ages at menarche and menopause, body mass index (BMI), BMI-adjusted waist-to-hip ratio, alcohol intake, and tobacco smoking. Overall, we identified 44 associations between germline PRSs and immune traits at false discovery rate q < 0.25, including 3 associations with q < 0.05. We observed consistent inverse associations of inflammatory bowel disease (IBD) and Crohn disease (CD) PRSs with interferon signaling and STAT1 scores in breast tumor and adjacent normal tissue; these associations were replicated in a Norwegian cohort. Inverse associations were also consistently observed for IBD PRS and B cell abundance in normal tissue. We also observed positive associations between CD PRS and endothelial cell abundance in tumor. Our findings suggest that the genetic mechanisms that influence immune-related diseases are also associated with TIME in breast cancer.
Collapse
Affiliation(s)
- Yuxi Liu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Cheng Peng
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Ina S Brorson
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Denise G O'Mahony
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Rebecca L Kelly
- Cancer Prevention Fellowship Program, National Cancer Institute, Rockville, MD, USA; Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Yujing J Heng
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Gabrielle M Baker
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Grethe I Grenaker Alnæs
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Clara Bodelon
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Daniel G Stover
- Division of Medical Oncology, Stefanie Spielman Comprehensive Breast Center, Columbus, OH, USA; Department of Biomedical Informatics, Ohio State University, Columbus, OH, USA
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Cancer Program, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - A Heather Eliassen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Vessela N Kristensen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Rulla M Tamimi
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
10
|
Bae JH, Kang H. Identification of Sweetness Preference-Related Single-Nucleotide Polymorphisms for Polygenic Risk Scores Associated with Obesity. Nutrients 2024; 16:2972. [PMID: 39275286 PMCID: PMC11397467 DOI: 10.3390/nu16172972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/09/2024] [Accepted: 08/31/2024] [Indexed: 09/16/2024] Open
Abstract
Our study aimed to identify sweetness preference-associated single-nucleotide polymorphisms (SNPs), characterize the related genetic loci, and develop SNP-based polygenic risk scores (PRS) to analyze their associations with obesity. For genotyping, we utilized a pooled genome-wide association study (GWAS) dataset of 18,499 females and 10,878 males. We conducted genome-wide association analyses, functional annotation, and employed the weighted method to calculate the levels of PRS from 677 sweetness preference-related SNPs. We used Cox proportional hazards modeling with time-varying covariates to estimate age-adjusted and multivariable hazard ratios (HRs) and 95% confidence intervals (CIs) for obesity incidence. We also tested the correlation between PRS and environmental factors, including smoking and dietary components, on obesity. Our results showed that in males, the TT genotype of rs4861982 significantly increased obesity risk compared to the GG genotype in the Health Professionals Follow-up Study (HPFS) cohort (HR = 1.565; 95% CI, 1.122-2.184; p = 0.008) and in the pooled analysis (HR = 1.259; 95% CI, 1.030-1.540; p = 0.025). Protein tyrosine phosphatase receptor type O (PTPRO) was identified as strongly associated with sweetness preference, indicating a positive correlation between sweetness preference and obesity risk. Moreover, each 10 pack-year increment in smoking was significantly associated with an increased risk of obesity in the HPFS cohort (HR = 1.024; 95% CI, 1.000-1.048) in males but not in females. In conclusion, significant associations between rs4861982, sweetness preference, and obesity were identified, particularly among males, where environmental factors like smoking are also correlated with obesity risk.
Collapse
Affiliation(s)
- Ji Hyun Bae
- Department of Food Science and Nutrition, Keimyung University, Daegu 42601, Republic of Korea
| | - Hyunju Kang
- Department of Food Science and Nutrition, Keimyung University, Daegu 42601, Republic of Korea
| |
Collapse
|
11
|
Plym A, Zhang Y, Stopsack KH, Ugalde-Morales E, Seibert TM, Conti DV, Haiman CA, Baras A, Stocks T, Drake I, Penney KL, Giovannucci E, Kibel AS, Wiklund F, Mucci LA. Early Prostate Cancer Deaths Among Men With Higher vs Lower Genetic Risk. JAMA Netw Open 2024; 7:e2420034. [PMID: 38958976 PMCID: PMC11222990 DOI: 10.1001/jamanetworkopen.2024.20034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/23/2024] [Indexed: 07/04/2024] Open
Abstract
Importance Prostate cancer, a leading cause of cancer death among men, urgently requires new prevention strategies, which may involve targeting men with an underlying genetic susceptibility. Objective To explore differences in risk of early prostate cancer death among men with higher vs lower genetic risk to inform prevention efforts. Design, Setting, and Participants This cohort study used a combined analysis of genotyped men without prostate cancer at inclusion and with lifestyle data in 2 prospective cohort studies in Sweden and the US, the Malmö Diet and Cancer Study (MDCS) and the Health Professionals Follow-Up Study (HPFS), followed up from 1991 to 2019. Data were analyzed between April 2023 and April 2024. Exposures Men were categorized according to modifiable lifestyle behaviors and genetic risk. A polygenic risk score above the median or a family history of cancer defined men at higher genetic risk (67% of the study population); the remaining men were categorized as being at lower genetic risk. Main Outcomes and Measures Prostate cancer death analyzed using time-to-event analysis estimating hazard ratios (HR), absolute risks, and preventable deaths by age. Results Among the 19 607 men included for analysis, the median (IQR) age at inclusion was 59.0 (53.0-64.7) years (MDCS) and 65.1 (58.0-71.8) years (HPFS). During follow-up, 107 early (by age 75 years) and 337 late (after age 75 years) prostate cancer deaths were observed. Compared with men at lower genetic risk, men at higher genetic risk had increased rates of both early (HR, 3.26; 95% CI, 1.82-5.84) and late (HR, 2.26; 95% CI, 1.70-3.01) prostate cancer death, and higher lifetime risks of prostate cancer death (3.1% vs 1.3% [MDCS] and 2.3% vs 0.6% [HPFS]). Men at higher genetic risk accounted for 94 of 107 early prostate cancer deaths (88%), of which 36% (95% CI, 12%-60%) were estimated to be preventable through adherence to behaviors associated with a healthy lifestyle (not smoking, healthy weight, high physical activity, and a healthy diet). Conclusions and Relevance In this 20-year follow-up study, men with a genetic predisposition accounted for the vast majority of early prostate cancer deaths, of which one-third were estimated to be preventable. This suggests that men at increased genetic risk should be targeted in prostate cancer prevention strategies.
Collapse
Affiliation(s)
- Anna Plym
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Department of Urology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yiwen Zhang
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Konrad H. Stopsack
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Emilio Ugalde-Morales
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Tyler M. Seibert
- Department of Radiation Medicine and Applied Sciences, Department of Radiology, and Department of Bioengineering, University of California San Diego, La Jolla
| | - David V. Conti
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles
| | - Christopher A. Haiman
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles
| | - Aris Baras
- Regeneron Genetics Center, Tarrytown, New York
| | - Tanja Stocks
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Isabel Drake
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
- Skåne University Hospital, Malmö, Sweden
| | - Kathryn L. Penney
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Edward Giovannucci
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Adam S. Kibel
- Department of Urology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Fredrik Wiklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Lorelei A. Mucci
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
12
|
Zhang Y, Stopsack KH, Song M, Mucci LA, Liu B, Penney KL, Tabung FK, Giovannucci E, Plym A. Healthy dietary patterns and risk of prostate cancer in men at high genetic risk. Int J Cancer 2024; 155:71-80. [PMID: 38429859 PMCID: PMC11068494 DOI: 10.1002/ijc.34898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/30/2024] [Accepted: 02/08/2024] [Indexed: 03/03/2024]
Abstract
Prostate cancer has high heritability. Healthy lifestyle has been associated with lower lethal prostate cancer risk among men at increased genetic susceptibility, but the role of healthy dietary patterns remains unknown. We prospectively followed 10,269 genotyped men in the Health Professionals Follow-up Study (1993-2019). Genetic risk was quantified using an established polygenic risk score (PRS). Five dietary patterns were investigated: healthy eating index, Mediterranean, diabetes risk-reducing, hyperinsulinemic and inflammatory diet. Overall and lethal prostate cancer rates (metastatic disease/prostate cancer-specific death) were analyzed using multivariable Cox proportional hazards models. During 26 years of follow-up, 2133 overall and 253 lethal prostate cancer events were documented. In the highest PRS quartile, higher adherence to a diabetes risk-reducing diet was associated with lower rates of overall (top vs. bottom quintile HR [95% CI], 0.74 [0.58-0.94]) and lethal prostate cancer (0.43 [0.21-0.88]). A low insulinemic diet was associated with similar lower rates (overall, 0.76 [0.60-0.95]; lethal, 0.46 [0.23-0.94]). Other dietary patterns showed weaker, but similar associations. In the highest PRS quartile, men with healthy lifestyles based on body weight, physical activity, and low insulinemic diet had a substantially lower rate (0.26 [0.13-0.49]) of lethal prostate cancer compared with men with unhealthy lifestyles, translating to a lifetime risk of 3.4% (95% CI, 2.3%-5.0%) among those with healthy lifestyles and 9.5% (5.3%-16.7%) among those with unhealthy lifestyles. Our findings indicate that lifestyle modifications lowering insulin resistance and chronic hyperinsulinemia could be relevant in preventing aggressive prostate cancer among men genetically predisposed to prostate cancer.
Collapse
Affiliation(s)
- Yiwen Zhang
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Konrad H. Stopsack
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Mingyang Song
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Lorelei A. Mucci
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Discovery Science, American Cancer Society, Atlanta GA
| | - Binkai Liu
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Kathryn L. Penney
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Fred K. Tabung
- Division of Medicine Oncology, Department of Internal Medicine, The Ohio State University College of Medicine and Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Edward Giovannucci
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Anna Plym
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Urology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Rumpf F, Plym A, Vaselkiv JB, Penney KL, Preston MA, Kibel AS, Mucci LA, Salari K. Impact of Family History and Germline Genetic Risk Single Nucleotide Polymorphisms on Long-Term Outcomes of Favorable-Risk Prostate Cancer. J Urol 2024; 211:754-764. [PMID: 38598641 PMCID: PMC11251859 DOI: 10.1097/ju.0000000000003927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
PURPOSE Family history and germline genetic risk single nucleotide polymorphisms (SNPs) have been separately shown to stratify lifetime risk of prostate cancer. Here, we evaluate the combined prognostic value of family history of prostate and other related cancers and germline risk SNPs among patients with favorable-risk prostate cancer. MATERIALS AND METHODS A total of 1367 participants from the prospective Health Professionals Follow-up Study diagnosed with low- or favorable intermediate-risk prostate cancer from 1986 to 2017 underwent genome-wide SNP genotyping. Multivariable Cox regression was used to estimate the association between family history, specific germline risk variants, and a 269 SNP polygenic risk score with prostate cancer‒specific death. RESULTS Family history of prostate, breast, and/or pancreatic cancer was observed in 489 (36%) participants. With median follow-up from diagnosis of 14.9 years, participants with favorable-risk prostate cancer with a positive family history had a significantly higher risk of prostate cancer‒specific death (HR 1.95, 95% CI 1.15-3.32, P = .014) compared to those without any family history. The rs2735839 (19q13) risk allele was associated with prostate cancer‒specific death (HR 1.81 per risk allele, 95% CI 1.04-3.17, P = .037), whereas the polygenic risk score was not. Combined family history and rs2735839 risk allele were each associated with an additive risk of prostate cancer‒specific death (HR 1.78 per risk factor, 95% CI 1.25-2.53, P = .001). CONCLUSIONS Family history of prostate, breast, or pancreatic cancer and/or a 19q13 germline risk allele are associated with an elevated risk of prostate cancer‒specific death among favorable-risk patients. These findings have implications for how family history and germline genetic risk SNPs should be factored into clinical decision-making around favorable-risk prostate cancer.
Collapse
Affiliation(s)
- Florian Rumpf
- Department of Urology, Massachusetts General Hospital, Boston, MA
- Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, D-97080 Wuerzburg, Germany
| | - Anna Plym
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Division of Urology, Department of Surgery, Brigham and Women’s Hospital, Boston, MA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jane B. Vaselkiv
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Kathryn L. Penney
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Mark A. Preston
- Division of Urology, Department of Surgery, Brigham and Women’s Hospital, Boston, MA
| | - Adam S. Kibel
- Division of Urology, Department of Surgery, Brigham and Women’s Hospital, Boston, MA
| | - Lorelei A. Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Keyan Salari
- Department of Urology, Massachusetts General Hospital, Boston, MA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
| |
Collapse
|
14
|
Tessier AJ, Cortese M, Yuan C, Bjornevik K, Ascherio A, Wang DD, Chavarro JE, Stampfer MJ, Hu FB, Willett WC, Guasch-Ferré M. Consumption of Olive Oil and Diet Quality and Risk of Dementia-Related Death. JAMA Netw Open 2024; 7:e2410021. [PMID: 38709531 PMCID: PMC11074805 DOI: 10.1001/jamanetworkopen.2024.10021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 03/06/2024] [Indexed: 05/07/2024] Open
Abstract
Importance Age-standardized dementia mortality rates are on the rise. Whether long-term consumption of olive oil and diet quality are associated with dementia-related death is unknown. Objective To examine the association of olive oil intake with the subsequent risk of dementia-related death and assess the joint association with diet quality and substitution for other fats. Design, Setting, and Participants This prospective cohort study examined data from the Nurses' Health Study (NHS; 1990-2018) and Health Professionals Follow-Up Study (HPFS; 1990-2018). The population included women from the NHS and men from the HPFS who were free of cardiovascular disease and cancer at baseline. Data were analyzed from May 2022 to July 2023. Exposures Olive oil intake was assessed every 4 years using a food frequency questionnaire and categorized as (1) never or less than once per month, (2) greater than 0 to less than or equal to 4.5 g/d, (3) greater than 4.5 g/d to less than or equal to 7 g/d, and (4) greater than 7 g/d. Diet quality was based on the Alternative Healthy Eating Index and Mediterranean Diet score. Main Outcome and Measure Dementia death was ascertained from death records. Multivariable Cox proportional hazards regressions were used to estimate hazard ratios (HRs) and 95% CIs adjusted for confounders including genetic, sociodemographic, and lifestyle factors. Results Of 92 383 participants, 60 582 (65.6%) were women and the mean (SD) age was 56.4 (8.0) years. During 28 years of follow-up (2 183 095 person-years), 4751 dementia-related deaths occurred. Individuals who were homozygous for the apolipoprotein ε4 (APOE ε4) allele were 5 to 9 times more likely to die with dementia. Consuming at least 7 g/d of olive oil was associated with a 28% lower risk of dementia-related death (adjusted pooled HR, 0.72 [95% CI, 0.64-0.81]) compared with never or rarely consuming olive oil (P for trend < .001); results were consistent after further adjustment for APOE ε4. No interaction by diet quality scores was found. In modeled substitution analyses, replacing 5 g/d of margarine and mayonnaise with the equivalent amount of olive oil was associated with an 8% (95% CI, 4%-12%) to 14% (95% CI, 7%-20%) lower risk of dementia mortality. Substitutions for other vegetable oils or butter were not significant. Conclusions and Relevance In US adults, higher olive oil intake was associated with a lower risk of dementia-related mortality, irrespective of diet quality. Beyond heart health, the findings extend the current dietary recommendations of choosing olive oil and other vegetable oils for cognitive-related health.
Collapse
Affiliation(s)
- Anne-Julie Tessier
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Marianna Cortese
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Changzheng Yuan
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kjetil Bjornevik
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Alberto Ascherio
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Daniel D. Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Jorge E. Chavarro
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Meir J. Stampfer
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Frank B. Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Walter C. Willett
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Public Health and Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Wu X, Hou W, Zhao Z, Huang L, Sheng N, Yang Q, Zhang S, Wang Y. MMGAT: a graph attention network framework for ATAC-seq motifs finding. BMC Bioinformatics 2024; 25:158. [PMID: 38643066 PMCID: PMC11031952 DOI: 10.1186/s12859-024-05774-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/10/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND Motif finding in Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq) data is essential to reveal the intricacies of transcription factor binding sites (TFBSs) and their pivotal roles in gene regulation. Deep learning technologies including convolutional neural networks (CNNs) and graph neural networks (GNNs), have achieved success in finding ATAC-seq motifs. However, CNN-based methods are limited by the fixed width of the convolutional kernel, which makes it difficult to find multiple transcription factor binding sites with different lengths. GNN-based methods has the limitation of using the edge weight information directly, makes it difficult to aggregate the neighboring nodes' information more efficiently when representing node embedding. RESULTS To address this challenge, we developed a novel graph attention network framework named MMGAT, which employs an attention mechanism to adjust the attention coefficients among different nodes. And then MMGAT finds multiple ATAC-seq motifs based on the attention coefficients of sequence nodes and k-mer nodes as well as the coexisting probability of k-mers. Our approach achieved better performance on the human ATAC-seq datasets compared to existing tools, as evidenced the highest scores on the precision, recall, F1_score, ACC, AUC, and PRC metrics, as well as finding 389 higher quality motifs. To validate the performance of MMGAT in predicting TFBSs and finding motifs on more datasets, we enlarged the number of the human ATAC-seq datasets to 180 and newly integrated 80 mouse ATAC-seq datasets for multi-species experimental validation. Specifically on the mouse ATAC-seq dataset, MMGAT also achieved the highest scores on six metrics and found 356 higher-quality motifs. To facilitate researchers in utilizing MMGAT, we have also developed a user-friendly web server named MMGAT-S that hosts the MMGAT method and ATAC-seq motif finding results. CONCLUSIONS The advanced methodology MMGAT provides a robust tool for finding ATAC-seq motifs, and the comprehensive server MMGAT-S makes a significant contribution to genomics research. The open-source code of MMGAT can be found at https://github.com/xiaotianr/MMGAT , and MMGAT-S is freely available at https://www.mmgraphws.com/MMGAT-S/ .
Collapse
Affiliation(s)
- Xiaotian Wu
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, 130012, China
- School of Artificial Intelligence, Jilin University, Changchun, 130012, China
| | - Wenju Hou
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, 130012, China
| | - Ziqi Zhao
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, 130012, China
| | - Lan Huang
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, 130012, China
| | - Nan Sheng
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, 130012, China
| | - Qixing Yang
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, 130012, China
| | - Shuangquan Zhang
- School of Cyber Science and Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China.
| | - Yan Wang
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, 130012, China.
- School of Artificial Intelligence, Jilin University, Changchun, 130012, China.
| |
Collapse
|
16
|
Rajanala SH, Plym A, Vaselkiv JB, Ebot EM, Matsoukas K, Lin Z, Chakraborty G, Markt SC, Penney KL, Lee GSM, Mucci LA, Kantoff PW, Stopsack KH. SLCO1B3 and SLCO2B1 genotypes, androgen deprivation therapy, and prostate cancer outcomes: a prospective cohort study and meta-analysis. Carcinogenesis 2024; 45:35-44. [PMID: 37856781 PMCID: PMC10859730 DOI: 10.1093/carcin/bgad075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 10/09/2023] [Accepted: 10/18/2023] [Indexed: 10/21/2023] Open
Abstract
Solute carrier organic anion (SLCO) transporters (OATP transporters) are involved in cellular uptake of drugs and hormones. Germline variants in SLCO1B3 and SLCO2B1 have been implicated in prostate cancer progression and therapy response, including to androgen deprivation and statin medications, but results have appeared heterogeneous. We conducted a cohort study of five single-nucleotide polymorphisms (SNPs) in SLCO1B3 and SLCO2B1 with prior evidence among 3208 men with prostate cancer who participated in the Health Professionals Follow-up Study or the Physicians' Health Study, following participants prospectively after diagnosis over 32 years (median, 14 years) for development of metastases and cancer-specific death (lethal disease, 382 events). Results were suggestive of, but not conclusive for, associations between some SNPs and lethal disease and differences by androgen deprivation and statin use. All candidate SNPs were associated with SLCO mRNA expression in tumor-adjacent prostate tissue. We also conducted a systematic review and harmonized estimates for a dose-response meta-analysis of all available data, including 9 further studies, for a total of 5598 patients and 1473 clinical events. The A allele of the exonic SNP rs12422149 (14% prevalence), which leads to lower cellular testosterone precursor uptake via SLCO2B1, was associated with lower rates of prostate cancer progression (hazard ratio per A allele, 0.80; 95% confidence interval, 0.69-0.93), with little heterogeneity between studies (I2, 0.27). Collectively, the totality of evidence suggests a strong association between inherited genetic variation in SLCO2B1 and prostate cancer prognosis, with potential clinical use in risk stratification related to androgen deprivation therapy.
Collapse
Affiliation(s)
- Sai Harisha Rajanala
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anna Plym
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Urology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Jane B Vaselkiv
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ericka M Ebot
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Konstantina Matsoukas
- Technology Division, Library Services, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhike Lin
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Goutam Chakraborty
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Urology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah C Markt
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Kathryn L Penney
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Gwo-Shu M Lee
- Lank Center for Genitourinary Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Philip W Kantoff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Convergent Therapeutics Inc., Boston, MA, USA
| | - Konrad H Stopsack
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Huang T, Goodman M, Wang H, Sofer T, Tworoger SS, Stampfer MJ, Saxena R, Redline S. Genetic Predisposition to Elevated C-Reactive Protein and Risk of Obstructive Sleep Apnea. Am J Respir Crit Care Med 2024; 209:329-331. [PMID: 37883203 PMCID: PMC10840766 DOI: 10.1164/rccm.202307-1159le] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/26/2023] [Indexed: 10/27/2023] Open
Affiliation(s)
- Tianyi Huang
- Channing Division of Network Medicine and
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts
| | - Matthew Goodman
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts
| | - Heming Wang
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts
| | - Shelley S. Tworoger
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Meir J. Stampfer
- Channing Division of Network Medicine and
- Department of Epidemiology, T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Richa Saxena
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts
- Department of Epidemiology, T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| |
Collapse
|
18
|
Ma C, Li Y, Mei Z, Yuan C, Kang JH, Grodstein F, Ascherio A, Willett WC, Chan AT, Huttenhower C, Stampfer MJ, Wang DD. Association Between Bowel Movement Pattern and Cognitive Function: Prospective Cohort Study and a Metagenomic Analysis of the Gut Microbiome. Neurology 2023; 101:e2014-e2025. [PMID: 37775319 PMCID: PMC10662989 DOI: 10.1212/wnl.0000000000207849] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/03/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Little is known regarding the association between intestinal motility patterns and cognitive function in individuals who are baseline cognitively healthy. The gut microbiome may contribute to the association. We examined the association between bowel movement (BM) pattern and cognitive function and explored the role of the gut microbiome in explaining this association. METHODS In this prospective study, we leveraged 3 cohort studies, Nurses' Health Study (NHS), NHSII, and Health Professionals Follow-Up Study (HPFS). Participants reported BM frequency and subjective cognitive function. In a subset of NHSII participants, we assessed cognitive function using an objective neuropsychological battery. We profiled the gut microbiome in a subset of participants using whole-genome shotgun metagenomics. General linear models, Poisson regression, and logistic regression were used to quantify the association of BM frequency with different cognitive measurements. RESULTS We followed 112,753 men and women (women: 87.6%) with a mean age of 67.2 years at baseline (NHS: 76 years, NHSII: 59 years, HPFS: 75 years) for a median follow-up of 4 years (NHSII and HPFS: 4 years, NHS: 2 years). Compared with those with BM once daily, participants with BM frequency every 3+ days had significantly worse objective cognitive function, equivalent to 3.0 (95% confidence interval [CI],1.2-4.7) years of chronological cognitive aging. We observed similar J-shape dose-response relationships of BM frequency with the odds of subjective cognitive decline and the likelihood of having more subsequent subjective cognitive complaints (both p nonlinearity < 0.001). BM frequencies of every 3+ days and ≥twice/day, compared with once daily, were associated with the odds ratios of subjective cognitive decline of 1.73 (95% CI 1.60-1.86) and 1.37 (95% CI 1.33-1.44), respectively. BM frequency and subjective cognitive decline were significantly associated with the overall gut microbiome configuration (both p < 0.005) and specific microbial species in the 515 participants with microbiome data. Butyrate-producing microbial species were depleted in those with less frequent BM and worse cognition, whereas a higher abundance of proinflammatory species was associated with BM frequency of ≥twice/day and worse cognition. DISCUSSION Lower BM frequency was associated with worse cognitive function. The gut microbial dysbiosis may be a mechanistic link underlying the association.
Collapse
Affiliation(s)
- Chaoran Ma
- From the Channing Division of Network Medicine (C.M., Z.M., J.H.K., A.A., M.J.S., D.D.W.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Nutrition (C.M.), University of Massachusetts Amherst; Departments of Nutrition (Y.L., A.A., W.C.W., M.J.S., D.D.W.), Epidemiology (A.A., W.C.W., A.T.C., M.J.S.), and Biostatistics (C.H.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Medicine (C.Y.), Zhejiang University, Hangzhou, China; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Gastroenterology (A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Broad Institute of MIT and Harvard (A.T.C., C.H., D.D.W), Cambridge, MA..
| | - Yanping Li
- From the Channing Division of Network Medicine (C.M., Z.M., J.H.K., A.A., M.J.S., D.D.W.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Nutrition (C.M.), University of Massachusetts Amherst; Departments of Nutrition (Y.L., A.A., W.C.W., M.J.S., D.D.W.), Epidemiology (A.A., W.C.W., A.T.C., M.J.S.), and Biostatistics (C.H.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Medicine (C.Y.), Zhejiang University, Hangzhou, China; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Gastroenterology (A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Broad Institute of MIT and Harvard (A.T.C., C.H., D.D.W), Cambridge, MA
| | - Zhendong Mei
- From the Channing Division of Network Medicine (C.M., Z.M., J.H.K., A.A., M.J.S., D.D.W.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Nutrition (C.M.), University of Massachusetts Amherst; Departments of Nutrition (Y.L., A.A., W.C.W., M.J.S., D.D.W.), Epidemiology (A.A., W.C.W., A.T.C., M.J.S.), and Biostatistics (C.H.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Medicine (C.Y.), Zhejiang University, Hangzhou, China; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Gastroenterology (A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Broad Institute of MIT and Harvard (A.T.C., C.H., D.D.W), Cambridge, MA
| | - Changzheng Yuan
- From the Channing Division of Network Medicine (C.M., Z.M., J.H.K., A.A., M.J.S., D.D.W.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Nutrition (C.M.), University of Massachusetts Amherst; Departments of Nutrition (Y.L., A.A., W.C.W., M.J.S., D.D.W.), Epidemiology (A.A., W.C.W., A.T.C., M.J.S.), and Biostatistics (C.H.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Medicine (C.Y.), Zhejiang University, Hangzhou, China; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Gastroenterology (A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Broad Institute of MIT and Harvard (A.T.C., C.H., D.D.W), Cambridge, MA
| | - Jae H Kang
- From the Channing Division of Network Medicine (C.M., Z.M., J.H.K., A.A., M.J.S., D.D.W.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Nutrition (C.M.), University of Massachusetts Amherst; Departments of Nutrition (Y.L., A.A., W.C.W., M.J.S., D.D.W.), Epidemiology (A.A., W.C.W., A.T.C., M.J.S.), and Biostatistics (C.H.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Medicine (C.Y.), Zhejiang University, Hangzhou, China; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Gastroenterology (A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Broad Institute of MIT and Harvard (A.T.C., C.H., D.D.W), Cambridge, MA
| | - Francine Grodstein
- From the Channing Division of Network Medicine (C.M., Z.M., J.H.K., A.A., M.J.S., D.D.W.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Nutrition (C.M.), University of Massachusetts Amherst; Departments of Nutrition (Y.L., A.A., W.C.W., M.J.S., D.D.W.), Epidemiology (A.A., W.C.W., A.T.C., M.J.S.), and Biostatistics (C.H.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Medicine (C.Y.), Zhejiang University, Hangzhou, China; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Gastroenterology (A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Broad Institute of MIT and Harvard (A.T.C., C.H., D.D.W), Cambridge, MA
| | - Alberto Ascherio
- From the Channing Division of Network Medicine (C.M., Z.M., J.H.K., A.A., M.J.S., D.D.W.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Nutrition (C.M.), University of Massachusetts Amherst; Departments of Nutrition (Y.L., A.A., W.C.W., M.J.S., D.D.W.), Epidemiology (A.A., W.C.W., A.T.C., M.J.S.), and Biostatistics (C.H.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Medicine (C.Y.), Zhejiang University, Hangzhou, China; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Gastroenterology (A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Broad Institute of MIT and Harvard (A.T.C., C.H., D.D.W), Cambridge, MA
| | - Walter C Willett
- From the Channing Division of Network Medicine (C.M., Z.M., J.H.K., A.A., M.J.S., D.D.W.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Nutrition (C.M.), University of Massachusetts Amherst; Departments of Nutrition (Y.L., A.A., W.C.W., M.J.S., D.D.W.), Epidemiology (A.A., W.C.W., A.T.C., M.J.S.), and Biostatistics (C.H.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Medicine (C.Y.), Zhejiang University, Hangzhou, China; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Gastroenterology (A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Broad Institute of MIT and Harvard (A.T.C., C.H., D.D.W), Cambridge, MA
| | - Andrew T Chan
- From the Channing Division of Network Medicine (C.M., Z.M., J.H.K., A.A., M.J.S., D.D.W.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Nutrition (C.M.), University of Massachusetts Amherst; Departments of Nutrition (Y.L., A.A., W.C.W., M.J.S., D.D.W.), Epidemiology (A.A., W.C.W., A.T.C., M.J.S.), and Biostatistics (C.H.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Medicine (C.Y.), Zhejiang University, Hangzhou, China; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Gastroenterology (A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Broad Institute of MIT and Harvard (A.T.C., C.H., D.D.W), Cambridge, MA
| | - Curtis Huttenhower
- From the Channing Division of Network Medicine (C.M., Z.M., J.H.K., A.A., M.J.S., D.D.W.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Nutrition (C.M.), University of Massachusetts Amherst; Departments of Nutrition (Y.L., A.A., W.C.W., M.J.S., D.D.W.), Epidemiology (A.A., W.C.W., A.T.C., M.J.S.), and Biostatistics (C.H.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Medicine (C.Y.), Zhejiang University, Hangzhou, China; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Gastroenterology (A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Broad Institute of MIT and Harvard (A.T.C., C.H., D.D.W), Cambridge, MA
| | - Meir J Stampfer
- From the Channing Division of Network Medicine (C.M., Z.M., J.H.K., A.A., M.J.S., D.D.W.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Nutrition (C.M.), University of Massachusetts Amherst; Departments of Nutrition (Y.L., A.A., W.C.W., M.J.S., D.D.W.), Epidemiology (A.A., W.C.W., A.T.C., M.J.S.), and Biostatistics (C.H.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Medicine (C.Y.), Zhejiang University, Hangzhou, China; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Gastroenterology (A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Broad Institute of MIT and Harvard (A.T.C., C.H., D.D.W), Cambridge, MA
| | - Dong D Wang
- From the Channing Division of Network Medicine (C.M., Z.M., J.H.K., A.A., M.J.S., D.D.W.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Nutrition (C.M.), University of Massachusetts Amherst; Departments of Nutrition (Y.L., A.A., W.C.W., M.J.S., D.D.W.), Epidemiology (A.A., W.C.W., A.T.C., M.J.S.), and Biostatistics (C.H.), Harvard T.H. Chan School of Public Health, Boston, MA; School of Medicine (C.Y.), Zhejiang University, Hangzhou, China; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Gastroenterology (A.T.C.), Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Broad Institute of MIT and Harvard (A.T.C., C.H., D.D.W), Cambridge, MA..
| |
Collapse
|
19
|
Han X, Lains I, Li J, Li J, Chen Y, Yu B, Qi Q, Boerwinkle E, Kaplan R, Thyagarajan B, Daviglus M, Joslin CE, Cai J, Guasch-Ferré M, Tobias DK, Rimm E, Ascherio A, Costenbader K, Karlson E, Mucci L, Eliassen AH, Zeleznik O, Miller J, Vavvas DG, Kim IK, Silva R, Miller J, Hu F, Willett W, Lasky-Su J, Kraft P, Richards JB, MacGregor S, Husain D, Liang L. Integrating genetics and metabolomics from multi-ethnic and multi-fluid data reveals putative mechanisms for age-related macular degeneration. Cell Rep Med 2023; 4:101085. [PMID: 37348500 PMCID: PMC10394104 DOI: 10.1016/j.xcrm.2023.101085] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 02/22/2023] [Accepted: 05/22/2023] [Indexed: 06/24/2023]
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness in older adults. Investigating shared genetic components between metabolites and AMD can enhance our understanding of its pathogenesis. We conduct metabolite genome-wide association studies (mGWASs) using multi-ethnic genetic and metabolomic data from up to 28,000 participants. With bidirectional Mendelian randomization analysis involving 16,144 advanced AMD cases and 17,832 controls, we identify 108 putatively causal relationships between plasma metabolites and advanced AMD. These metabolites are enriched in glycerophospholipid metabolism, lysophospholipid, triradylcglycerol, and long chain polyunsaturated fatty acid pathways. Bayesian genetic colocalization analysis and a customized metabolome-wide association approach prioritize putative causal AMD-associated metabolites. We find limited evidence linking urine metabolites to AMD risk. Our study emphasizes the contribution of plasma metabolites, particularly lipid-related pathways and genes, to AMD risk and uncovers numerous putative causal associations between metabolites and AMD risk.
Collapse
Affiliation(s)
- Xikun Han
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Ines Lains
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Jun Li
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jinglun Li
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Yiheng Chen
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC, Canada; Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Robert Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA; Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center, Minneapolis, MN, USA
| | - Martha Daviglus
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Charlotte E Joslin
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Jianwen Cai
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Deirdre K Tobias
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Eric Rimm
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alberto Ascherio
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Karen Costenbader
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Elizabeth Karlson
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Lorelei Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - A Heather Eliassen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Oana Zeleznik
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - John Miller
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Demetrios G Vavvas
- Retina Service, Ines and Fredrick Yeatts Retinal Research Laboratory, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Ivana K Kim
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Rufino Silva
- Ophthalmology Unit, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal; University Clinic of Ophthalmology, Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Joan Miller
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Frank Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Walter Willett
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jessica Lasky-Su
- Systems Genetics and Genomics Unit, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - J Brent Richards
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC, Canada; Departments of Medicine, Human Genetics, Epidemiology and Biostatistics, McGill University, Montréal, QC, Canada; Department of Twin Research, King's College London, London, UK; Five Prime Sciences Inc, Montréal, QC, Canada
| | - Stuart MacGregor
- Statistical Genetics Lab, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Deeba Husain
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA.
| | - Liming Liang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
20
|
Jones SK, Alberg AJ, Wallace K, Froeliger B, Carpenter MJ, Wolf BJ. CHRNA5-A3-B4 and DRD2 Genes and Smoking Cessation Throughout Adulthood: A Longitudinal Study of Women. Nicotine Tob Res 2023; 25:1164-1173. [PMID: 36794842 PMCID: PMC10413434 DOI: 10.1093/ntr/ntad026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/22/2022] [Accepted: 02/14/2023] [Indexed: 02/17/2023]
Abstract
INTRODUCTION Smoking cessation is more than 50% heritable. Genetic studies of smoking cessation have been limited by short-term follow-up or cross-sectional design. AIMS AND METHODS This study tests single nucleotide polymorphism (SNP) associations with cessation during long-term follow-up throughout adulthood in women. The secondary aim tests whether genetic associations differ by smoking intensity. Associations between 10 SNPs in CHRNA5, CHRNA3, CHRNB2, CHRNB4, DRD2, and COMT and the probability of smoking cessation over time were evaluated in two longitudinal cohort studies of female nurses, the Nurses' Health Study (NHS) (n = 10 017) and NHS-2 (n = 2793). Participant follow-up ranged from 2 to 38 years with data collected every 2 years. RESULTS Women with the minor allele of either CHRNA5 SNP rs16969968 or CHRNA3 SNP rs1051730 had lower odds of cessation throughout adulthood [OR = 0.93, p-value = .003]. Women had increased odds of cessation if they had the minor allele of CHRNA3 SNP rs578776 [OR = 1.17, p-value = .002]. The minor allele of DRD2 SNP rs1800497 was associated with lower odds of cessation in moderate-to-heavy smokers [OR = 0.92, p-value = .0183] but increased odds in light smokers [OR = 1.24, p-value = .096]. CONCLUSIONS Some SNP associations with short-term smoking abstinence observed in prior studies were shown in the present study to persist throughout adulthood over decades of follow-up. Other SNP associations with short-term abstinence did not persist long-term. The secondary aim findings suggest genetic associations may differ by smoking intensity. IMPLICATIONS The results of the present study expand on previous studies of SNP associations in relation to short-term smoking cessation to demonstrate some of these SNPs were associated with smoking cessation throughout decades of follow-up, whereas other SNP associations with short-term abstinence did not persist long-term. The rate of relapse to smoking remains high for several years after quitting smoking, and many smokers experience multiple quit attempts and relapse episodes throughout adulthood. Understanding genetic associations with long-term cessation has potential importance for precision medicine approaches to long-term cessation management.
Collapse
Affiliation(s)
- Stephanie K Jones
- Department of Public Health, Baylor University, Waco, TX, USA
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Anthony J Alberg
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Kristin Wallace
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Brett Froeliger
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Matthew J Carpenter
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Bethany J Wolf
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
21
|
De Angelis F, Zeleznik OA, Wendt FR, Pathak GA, Tylee DS, De Lillo A, Koller D, Cabrera-Mendoza B, Clifford RE, Maihofer AX, Nievergelt CM, Curhan GC, Curhan SG, Polimanti R. Sex differences in the polygenic architecture of hearing problems in adults. Genome Med 2023; 15:36. [PMID: 37165447 PMCID: PMC10173489 DOI: 10.1186/s13073-023-01186-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/28/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Hearing problems (HP) in adults are common and are associated with several comorbid conditions. Its prevalence increases with age, reflecting the cumulative effect of environmental factors and genetic predisposition. Although several risk loci have been already identified, HP biology and epidemiology are still insufficiently investigated by large-scale genetic studies. METHODS Leveraging the UK Biobank, the Nurses' Health Studies (I and II), the Health Professionals Follow-up Study, and the Million Veteran Program, we conducted a comprehensive genome-wide investigation of HP in 748,668 adult participants (discovery N = 501,825; replication N = 226,043; cross-ancestry replication N = 20,800). We leveraged the GWAS findings to characterize HP polygenic architecture, exploring sex differences, polygenic risk across ancestries, tissue-specific transcriptomic regulation, cause-effect relationships with genetically correlated traits, and gene interactions with HP environmental risk factors. RESULTS We identified 54 risk loci and demonstrated that HP polygenic risk is shared across ancestry groups. Our transcriptomic regulation analysis highlighted the potential role of the central nervous system in HP pathogenesis. The sex-stratified analyses showed several additional associations related to peripheral hormonally regulated tissues reflecting a potential role of estrogen in hearing function. This evidence was supported by the multivariate interaction analysis that showed how genes involved in brain development interact with sex, noise pollution, and tobacco smoking in relation to their HP associations. Additionally, the genetically informed causal inference analysis showed that HP is linked to many physical and mental health outcomes. CONCLUSIONS The results provide many novel insights into the biology and epidemiology of HP in adults. Our sex-specific analyses and transcriptomic associations highlighted molecular pathways that may be targeted for drug development or repurposing. Additionally, the potential causal relationships identified may support novel preventive screening programs to identify individuals at risk.
Collapse
Affiliation(s)
- Flavio De Angelis
- Department of Psychiatry, Yale University School of Medicine, 60 Temple, Suite 7A, New Haven, CT, USA
- Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Oana A Zeleznik
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Frank R Wendt
- Department of Psychiatry, Yale University School of Medicine, 60 Temple, Suite 7A, New Haven, CT, USA
- Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Gita A Pathak
- Department of Psychiatry, Yale University School of Medicine, 60 Temple, Suite 7A, New Haven, CT, USA
- Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Daniel S Tylee
- Department of Psychiatry, Yale University School of Medicine, 60 Temple, Suite 7A, New Haven, CT, USA
- Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Antonella De Lillo
- Department of Psychiatry, Yale University School of Medicine, 60 Temple, Suite 7A, New Haven, CT, USA
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Dora Koller
- Department of Psychiatry, Yale University School of Medicine, 60 Temple, Suite 7A, New Haven, CT, USA
- Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Brenda Cabrera-Mendoza
- Department of Psychiatry, Yale University School of Medicine, 60 Temple, Suite 7A, New Haven, CT, USA
- Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Royce E Clifford
- Division of Otolaryngology, Department of Surgery, University of California, San Diego, La Jolla, CA, USA
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Adam X Maihofer
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Caroline M Nievergelt
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Gary C Curhan
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Sharon G Curhan
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Renato Polimanti
- Department of Psychiatry, Yale University School of Medicine, 60 Temple, Suite 7A, New Haven, CT, USA.
- Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA.
| |
Collapse
|
22
|
Shi J, Kraft P, Rosner BA, Benavente Y, Black A, Brinton LA, Chen C, Clarke MA, Cook LS, Costas L, Dal Maso L, Freudenheim JL, Frias-Gomez J, Friedenreich CM, Garcia-Closas M, Goodman MT, Johnson L, La Vecchia C, Levi F, Lissowska J, Lu L, McCann SE, Moysich KB, Negri E, O'Connell K, Parazzini F, Petruzella S, Polesel J, Ponte J, Rebbeck TR, Reynolds P, Ricceri F, Risch HA, Sacerdote C, Setiawan VW, Shu XO, Spurdle AB, Trabert B, Webb PM, Wentzensen N, Wilkens LR, Xu WH, Yang HP, Yu H, Du M, De Vivo I. Risk prediction models for endometrial cancer: development and validation in an international consortium. J Natl Cancer Inst 2023; 115:552-559. [PMID: 36688725 PMCID: PMC10165481 DOI: 10.1093/jnci/djad014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 09/01/2022] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Endometrial cancer risk stratification may help target interventions, screening, or prophylactic hysterectomy to mitigate the rising burden of this cancer. However, existing prediction models have been developed in select cohorts and have not considered genetic factors. METHODS We developed endometrial cancer risk prediction models using data on postmenopausal White women aged 45-85 years from 19 case-control studies in the Epidemiology of Endometrial Cancer Consortium (E2C2). Relative risk estimates for predictors were combined with age-specific endometrial cancer incidence rates and estimates for the underlying risk factor distribution. We externally validated the models in 3 cohorts: Nurses' Health Study (NHS), NHS II, and the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial. RESULTS Area under the receiver operating characteristic curves for the epidemiologic model ranged from 0.64 (95% confidence interval [CI] = 0.62 to 0.67) to 0.69 (95% CI = 0.66 to 0.72). Improvements in discrimination from the addition of genetic factors were modest (no change in area under the receiver operating characteristic curves in NHS; PLCO = 0.64 to 0.66). The epidemiologic model was well calibrated in NHS II (overall expected-to-observed ratio [E/O] = 1.09, 95% CI = 0.98 to 1.22) and PLCO (overall E/O = 1.04, 95% CI = 0.95 to 1.13) but poorly calibrated in NHS (overall E/O = 0.55, 95% CI = 0.51 to 0.59). CONCLUSIONS Using data from the largest, most heterogeneous study population to date (to our knowledge), prediction models based on epidemiologic factors alone successfully identified women at high risk of endometrial cancer. Genetic factors offered limited improvements in discrimination. Further work is needed to refine this tool for clinical or public health practice and expand these models to multiethnic populations.
Collapse
Affiliation(s)
- Joy Shi
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Bernard A Rosner
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yolanda Benavente
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, Bellvitge Biomedical Research Institute, Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER Epidemiología y Salud Pública, CIBERESP), Madrid, Spain
| | - Amanda Black
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Louise A Brinton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Chu Chen
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Megan A Clarke
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Linda S Cook
- Department of Epidemiology, Colorado School of Public Heath, University of Colorado-Anschutz, Aurora, CO, USA
- Department of Cancer Epidemiology and Prevention Research, Alberta Health Services, Calgary, AB, Canada
| | - Laura Costas
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, Bellvitge Biomedical Research Institute, Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER Epidemiología y Salud Pública, CIBERESP), Madrid, Spain
| | - Luigino Dal Maso
- Cancer Epidemiology Unit, Centro di Riferimento Oncologico di Aviano (CRO), Aviano, Italy
| | - Jo L Freudenheim
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Jon Frias-Gomez
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, Bellvitge Biomedical Research Institute, Barcelona, Spain
- Faculty of Medicine, University of Barcelona (UB), Barcelona, Spain
| | - Christine M Friedenreich
- Department of Cancer Epidemiology and Prevention Research, Alberta Health Services, Calgary, AB, Canada
| | | | - Marc T Goodman
- Community and Population Health Research Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lisa Johnson
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Carlo La Vecchia
- Department of Clinical Medicine and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Fabio Levi
- Department of Epidemiology and Health Services Research, Centre for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Jolanta Lissowska
- Department of Cancer Epidemiology and Prevention, M. Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, USA
| | - Susan E McCann
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kirsten B Moysich
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Eva Negri
- Department of Clinical Medicine and Community Health, Università degli Studi di Milano, Milan, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Kelli O'Connell
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fabio Parazzini
- Department of Clinical Medicine and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Stacey Petruzella
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jerry Polesel
- Cancer Epidemiology Unit, Centro di Riferimento Oncologico di Aviano (CRO), Aviano, Italy
| | - Jeanette Ponte
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Timothy R Rebbeck
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Population Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Peggy Reynolds
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Fulvio Ricceri
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | - Harvey A Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, USA
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Città della Salute e della Scienza University-Hospital and Center for Cancer Prevention (CPO), Turin, Italy
| | - Veronica W Setiawan
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Amanda B Spurdle
- Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Genetics and Computational Biology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Britton Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, University of Utah, Salt Lake City, UT, USA
| | - Penelope M Webb
- Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | | | - Wang Hong Xu
- Department of Epidemiology, Fudan University School of Public Health, Shanghai, China
| | - Hannah P Yang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Herbert Yu
- University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Mengmeng Du
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Immaculata De Vivo
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Radcliffe Institute for Advanced Study, Harvard University, Cambridge, MA, USA
| |
Collapse
|
23
|
Jones SK, Alberg AJ, Wallace K, Froeliger B, Carpenter MJ, Wolf B. Genetic Associations with Smoking Relapse and Proportion of Follow-up in Smoking Relapse throughout Adulthood in Pre- and Postmenopausal Women. Cancer Prev Res (Phila) 2023; 16:269-279. [PMID: 37070666 PMCID: PMC10159950 DOI: 10.1158/1940-6207.capr-22-0421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/10/2023] [Accepted: 03/08/2023] [Indexed: 04/19/2023]
Abstract
PREVENTION RELEVANCE This study is the first to quantify genetic associations with smoking relapse among female smokers throughout adulthood. These findings could inform precision medicine approaches to improve long-term smoking relapse prevention to reduce smoking attributable cancer morbidity and mortality.
Collapse
Affiliation(s)
| | - Anthony J. Alberg
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia SC 29208
| | - Kristin Wallace
- Department of Public Health Sciences, Medical University of South Carolina, Charleston SC 29425
- Hollings Cancer Center, 86 Jonathan Lucas Street, Medical University of South Carolina, Charleston SC 29425
| | - Brett Froeliger
- Department of Psychological Sciences, University of Missouri, Columbia MO 65211
| | - Matthew J. Carpenter
- Hollings Cancer Center, 86 Jonathan Lucas Street, Medical University of South Carolina, Charleston SC 29425
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston SC 29425
| | - Bethany Wolf
- Department of Public Health Sciences, Medical University of South Carolina, Charleston SC 29425
| |
Collapse
|
24
|
Zhang Z, Li H, Weng H, Zhou G, Chen H, Yang G, Zhang P, Zhang X, Ji Y, Ying K, Liu B, Xu Q, Tang Y, Zhu G, Liu Z, Xia S, Yang X, Dong L, Zhu L, Zeng M, Yuan Y, Yang Y, Zhang N, Xu X, Pang W, Zhang M, Zhang Y, Zhen K, Wang D, Lei J, Wu S, Shu S, Zhang Y, Zhang S, Gao Q, Huang Q, Deng C, Fu X, Chen G, Duan W, Wan J, Xie W, Zhang P, Wang S, Yang P, Zuo X, Zhai Z, Wang C. Genome-wide association analyses identified novel susceptibility loci for pulmonary embolism among Han Chinese population. BMC Med 2023; 21:153. [PMID: 37076872 PMCID: PMC10116678 DOI: 10.1186/s12916-023-02844-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 03/22/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND A large proportion of pulmonary embolism (PE) heritability remains unexplained, particularly among the East Asian (EAS) population. Our study aims to expand the genetic architecture of PE and reveal more genetic determinants in Han Chinese. METHODS We conducted the first genome-wide association study (GWAS) of PE in Han Chinese, then performed the GWAS meta-analysis based on the discovery and replication stages. To validate the effect of the risk allele, qPCR and Western blotting experiments were used to investigate possible changes in gene expression. Mendelian randomization (MR) analysis was employed to implicate pathogenic mechanisms, and a polygenic risk score (PRS) for PE risk prediction was generated. RESULTS After meta-analysis of the discovery dataset (622 cases, 8853 controls) and replication dataset (646 cases, 8810 controls), GWAS identified 3 independent loci associated with PE, including the reported loci FGG rs2066865 (p-value = 3.81 × 10-14), ABO rs582094 (p-value = 1.16 × 10-10) and newly reported locus FABP2 rs1799883 (p-value = 7.59 × 10-17). Previously reported 10 variants were successfully replicated in our cohort. Functional experiments confirmed that FABP2-A163G(rs1799883) promoted the transcription and protein expression of FABP2. Meanwhile, MR analysis revealed that high LDL-C and TC levels were associated with an increased risk of PE. Individuals with the top 10% of PRS had over a fivefold increased risk for PE compared to the general population. CONCLUSIONS We identified FABP2, related to the transport of long-chain fatty acids, contributing to the risk of PE and provided more evidence for the essential role of metabolic pathways in PE development.
Collapse
Affiliation(s)
- Zhu Zhang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital; National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; National Clinical Research Center for Respiratory Diseases, Beijing, 100029, China
| | - Haobo Li
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital; National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; National Clinical Research Center for Respiratory Diseases, Beijing, 100029, China
| | - Haoyi Weng
- Shenzhen WeGene Clinical Laboratory; WeGene, Shenzhen Zaozhidao Technology Co. Ltd; Hunan Provincial Key Lab On Bioinformatics, School of Computer Science and Engineering, Central South University, Shenzhen, 518042, China
| | - Geyu Zhou
- Department of Bioinformatics and Biostatistics, SJTU-Yale Joint Center for Biostatistics, College of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hong Chen
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Guoru Yang
- Department of Pulmonary and Critical Care Medicine, Weifang No.2 People's Hospital, Weifang, 261021, China
| | - Ping Zhang
- Department of Pulmonary and Critical Care Medicine, Dongguan People's Hospital, Dongguan, 523059, China
| | - Xiangyan Zhang
- Department of Pulmonary and Critical Care Medicine, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Yingqun Ji
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital Affiliated by Tongji University, Shanghai, 200120, China
| | - Kejing Ying
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310020, China
| | - Bo Liu
- Department of Pulmonary and Critical Care Medicine, Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Linzi District People's Hospital, Zibo, 255400, China
| | - Qixia Xu
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of University of Science and Technology of China, Hefei, 230001, China
| | - Yongjun Tang
- Department of Pulmonary and Critical Care Medicine, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Guangfa Zhu
- Department of Pulmonary and Critical Care, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Zhihong Liu
- Fuwai Hospital, Chinese Academy of Medical Science; National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Shuyue Xia
- Department of Pulmonary and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang, 110001, China
| | - Xiaohong Yang
- Department of Pulmonary and Critical Care Medicine, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, 830001, China
| | - Lixia Dong
- Department of Pulmonary and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, 300050, China
| | - Ling Zhu
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital, Jinan, 250021, China
| | - Mian Zeng
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yadong Yuan
- Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050004, China
| | - Yuanhua Yang
- Department of Pulmonary and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100026, China
| | - Nuofu Zhang
- State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510230, China
| | - Xiaomao Xu
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, Beijing, 100080, China
| | - Wenyi Pang
- Department of Pulmonary and Critical Care Medicine, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Meng Zhang
- Department of Pulmonary and Critical Care, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Yu Zhang
- China-Japan Friendship Hospital, Capital Medical University; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital; National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; National Clinical Research Center for Respiratory Diseases, Beijing, 100029, China
| | - Kaiyuan Zhen
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital; National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; National Clinical Research Center for Respiratory Diseases, Beijing, 100029, China
| | - Dingyi Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital; National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; National Clinical Research Center for Respiratory Diseases, Beijing, China, 100029
| | - Jieping Lei
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital; National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; National Clinical Research Center for Respiratory Diseases, Beijing, China, 100029
| | - Sinan Wu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital; National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; National Clinical Research Center for Respiratory Diseases, Beijing, China, 100029
| | - Shi Shu
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital; National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; National Clinical Research Center for Respiratory Diseases, Beijing, 100029, China
| | - Yunxia Zhang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital; National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; National Clinical Research Center for Respiratory Diseases, Beijing, 100029, China
| | - Shuai Zhang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital; National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; National Clinical Research Center for Respiratory Diseases, Beijing, 100029, China
| | - Qian Gao
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital; National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; National Clinical Research Center for Respiratory Diseases, Beijing, 100029, China
| | - Qiang Huang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital; National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; National Clinical Research Center for Respiratory Diseases, Beijing, 100029, China
| | - Chao Deng
- Department of Bioinformatics and Biostatistics, SJTU-Yale Joint Center for Biostatistics, College of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xi Fu
- Shenzhen WeGene Clinical Laboratory; WeGene, Shenzhen Zaozhidao Technology Co. Ltd; Hunan Provincial Key Lab On Bioinformatics, School of Computer Science and Engineering, Central South University, Shenzhen, 518042, China
| | - Gang Chen
- Shenzhen WeGene Clinical Laboratory; WeGene, Shenzhen Zaozhidao Technology Co. Ltd; Hunan Provincial Key Lab On Bioinformatics, School of Computer Science and Engineering, Central South University, Shenzhen, 518042, China
| | - Wenxin Duan
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Jun Wan
- Department of Pulmonary and Critical Care, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Wanmu Xie
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital; National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; National Clinical Research Center for Respiratory Diseases, Beijing, 100029, China
| | - Peng Zhang
- Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Shengfeng Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Peiran Yang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Xianbo Zuo
- Department of Dermatology, China-Japan Friendship Hospital, Beijing, China; Department of Pharmacy, China-Japan Friendship Hospital, No. 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China.
| | - Zhenguo Zhai
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital; National Center for Respiratory Medicine; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; National Clinical Research Center for Respiratory Diseases, Beijing, 100029, China.
| | - Chen Wang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.
- National Center for Respiratory Medicine, Beijing, China.
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China.
- National Clinical Research Center for Respiratory Diseases, Beijing, China.
- Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.
- Department of Respiratory Medicine, Capital Medical University, Beijing, China.
| |
Collapse
|
25
|
Plym A, Zhang Y, Stopsack KH, Delcoigne B, Wiklund F, Haiman C, Kenfield SA, Kibel AS, Giovannucci E, Penney KL, Mucci LA. A Healthy Lifestyle in Men at Increased Genetic Risk for Prostate Cancer. Eur Urol 2023; 83:343-351. [PMID: 35637041 DOI: 10.1016/j.eururo.2022.05.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/20/2022] [Accepted: 05/10/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Prostate cancer is the most heritable cancer. There is a need to identify possible modifiable factors for men at an increased risk of prostate cancer due to genetic factors. OBJECTIVE To examine whether men at an increased genetic risk of prostate cancer can offset their risk of disease or disease progression by adhering to a healthy lifestyle. DESIGN, SETTING, AND PARTICIPANTS We prospectively followed 12 411 genotyped men in the Health Professionals Follow-up Study (1993-2019) and the Physicians' Health Study (1983-2010). Genetic risk of prostate cancer was quantified using a polygenic risk score (PRS). A healthy lifestyle was defined by healthy weight, vigorous physical activity, not smoking, and a healthy diet. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Overall and lethal prostate cancer events (metastatic disease/prostate cancer-specific death) were analyzed using time-to-event analyses estimating hazard ratios (HRs) and lifetime risks. RESULTS AND LIMITATIONS During 27 yr of follow-up, 3005 overall prostate cancer and 435 lethal prostate cancer events were observed. The PRS enabled risk stratification not only for overall prostate cancer, but also for lethal disease with a four-fold difference between men in the highest and lowest quartiles (HR, 4.32; 95% confidence interval [CI], 3.16-5.89). Among men in the highest PRS quartile, adhering to a healthy lifestyle was associated with a decreased rate of lethal prostate cancer (HR, 0.55; 95% CI, 0.36-0.86) compared with having an unhealthy lifestyle, translating to a lifetime risk of 1.6% (95% CI, 0.8-3.1%) among the healthy and 5.3% (95% CI, 3.6-7.8%) among the unhealthy. Adhering to a healthy lifestyle was not associated with a decreased risk of overall prostate cancer. CONCLUSIONS Our findings suggest that a genetic predisposition for prostate cancer is not deterministic for a poor cancer outcome. Maintaining a healthy lifestyle may provide a way to offset the genetic risk of lethal prostate cancer. PATIENT SUMMARY This study examined whether the genetic risk of prostate cancer can be attenuated by a healthy lifestyle including a healthy weight, regular exercise, not smoking, and a healthy diet. We observed that adherence to a healthy lifestyle reduced the risk of metastatic disease and prostate cancer death among men at the highest genetic risk. We conclude that men at a high genetic risk of prostate cancer may benefit from adhering to a healthy lifestyle.
Collapse
Affiliation(s)
- Anna Plym
- Urology Division, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| | - Yiwen Zhang
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Konrad H Stopsack
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bénédicte Delcoigne
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Wiklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Christopher Haiman
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Stacey A Kenfield
- Departments of Urology and Epidemiology & Biostatistics, University of California, San Francisco, CA, USA
| | - Adam S Kibel
- Urology Division, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Edward Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kathryn L Penney
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
26
|
Powell M, Fuller S, Gunderson E, Benz C. A common IGF1R gene variant predicts later life breast cancer risk in women with preeclampsia. Breast Cancer Res Treat 2023; 197:149-159. [PMID: 36331687 PMCID: PMC9823040 DOI: 10.1007/s10549-022-06789-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE Preeclampsia has been inconsistently associated with altered later life risk of cancer. This study utilizes the Nurses' Health Study 2 (NHS2) to determine if the future risk of breast and non-breast cancers in women who experience preeclampsia is modified by carrying a protective variant of rs2016347, a functional insulin-like growth factor receptor-1 (IGF1R) single nucleotide polymorphism. METHODS This retrospective cohort study completed within the NHS2 evaluated participants enrolled in 1989 and followed them through 2015, with a study population of 86,751 after exclusions. Cox proportional hazards models both with and without the impact of rs2016347 genotype were used to assess the risk of invasive breast cancer, hormone receptor-positive (HR+) breast cancer, and non-breast cancers. RESULTS Women with preeclampsia had no change in risk of all breast, HR+ breast, or non-breast cancers when not considering genotype. However, women carrying at least one T allele of rs2016347 had a lower risk of HR+ breast cancer, HR 0.67, 95% CI: 0.47-0.97, P = 0.04, with interaction term P = 0.06. For non-breast cancers as a group, women carrying a T allele had an HR 0.76, 95% CI: 0.53-1.08, P = 0.12, with interaction term P = 0.26. CONCLUSIONS This retrospective cohort study found that women with preeclampsia who carry a T allele of IGF1R rs2016347 had a reduced future risk of developing HR+ breast cancer, and a reduced but not statistically significant decreased risk of non-breast cancers suggesting a possible role for the IGF-1 axis in the development of cancer in these women.
Collapse
Affiliation(s)
- Mark Powell
- grid.272799.00000 0000 8687 5377Buck Institute for Research On Aging, 8001 Redwood Blvd, Novato, CA 94945 USA
| | - Sophia Fuller
- grid.47840.3f0000 0001 2181 7878Graduate Group in Biostatistics, School of Public Health, University of California, Berkeley, CA USA
| | - Erica Gunderson
- grid.280062.e0000 0000 9957 7758Division of Research, Kaiser Permanente Northern California, Oakland, CA USA
| | - Christopher Benz
- grid.272799.00000 0000 8687 5377Buck Institute for Research On Aging, 8001 Redwood Blvd, Novato, CA 94945 USA
| |
Collapse
|
27
|
Plym A, Zhang Y, Stopsack KH, Jee YH, Wiklund F, Kibel AS, Kraft P, Giovannucci E, Penney KL, Mucci LA. Family History of Prostate and Breast Cancer Integrated with a Polygenic Risk Score Identifies Men at Highest Risk of Dying from Prostate Cancer before Age 75 Years. Clin Cancer Res 2022; 28:4926-4933. [PMID: 36103261 PMCID: PMC9660541 DOI: 10.1158/1078-0432.ccr-22-1723] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/04/2022] [Accepted: 09/12/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE Family history of prostate cancer is one of the few universally accepted risk factors for prostate cancer. How much an assessment of inherited polygenic risk for prostate cancer adds to lifetime risk stratification beyond family history is unknown. EXPERIMENTAL DESIGN We followed 10,120 men in the Health Professionals Follow-up Study with existing genotype data for risk of prostate cancer and prostate cancer-specific death. We assessed to what extent family history of prostate or breast cancer, combined with a validated polygenic risk score (PRS) including 269 prostate cancer risk variants, identifies men at risk of prostate cancer and prostate cancer death across the age span. RESULTS During 20 years of follow-up, 1,915 prostate cancer and 166 fatal prostate cancer events were observed. Men in the top PRS quartile with a family history of prostate or breast cancer had the highest rate of both prostate cancer and prostate cancer-specific death. Compared with men at lowest genetic risk (bottom PRS quartile and no family history), the HR was 6.95 [95% confidence interval (CI), 5.57-8.66] for prostate cancer and 4.84 (95% CI, 2.59-9.03) for prostate cancer death. Men in the two upper PRS quartiles (50%-100%) or with a family history of prostate or breast cancer (61.8% of the population) accounted for 97.5% of prostate cancer deaths by age 75 years. CONCLUSIONS Our study shows that prostate cancer risk stratification on the basis of family history and inherited polygenic risk can identify men at highest risk of dying from prostate cancer before age 75 years.
Collapse
Affiliation(s)
- Anna Plym
- Urology Division, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Corresponding Author: Anna Plym, Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, PO Box 281, Stockholm SE-171 77, Sweden. Phone: 468-5248-0000; Fax: 468-314-975; E-mail:
| | - Yiwen Zhang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Konrad H. Stopsack
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yon Ho Jee
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Fredrik Wiklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Adam S. Kibel
- Urology Division, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Edward Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Kathryn L. Penney
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lorelei A. Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
28
|
Liu Y, Chen H, Heine J, Lindstrom S, Turman C, Warner ET, Winham SJ, Vachon CM, Tamimi RM, Kraft P, Jiang X. A genome-wide association study of mammographic texture variation. Breast Cancer Res 2022; 24:76. [PMID: 36344993 PMCID: PMC9639267 DOI: 10.1186/s13058-022-01570-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Breast parenchymal texture features, including grayscale variation (V), capture the patterns of texture variation on a mammogram and are associated with breast cancer risk, independent of mammographic density (MD). However, our knowledge on the genetic basis of these texture features is limited. METHODS We conducted a genome-wide association study of V in 7040 European-ancestry women. V assessments were generated from digitized film mammograms. We used linear regression to test the single-nucleotide polymorphism (SNP)-phenotype associations adjusting for age, body mass index (BMI), MD phenotypes, and the top four genetic principal components. We further calculated genetic correlations and performed SNP-set tests of V with MD, breast cancer risk, and other breast cancer risk factors. RESULTS We identified three genome-wide significant loci associated with V: rs138141444 (6q24.1) in ECT2L, rs79670367 (8q24.22) in LINC01591, and rs113174754 (12q22) near PGAM1P5. 6q24.1 and 8q24.22 have not previously been associated with MD phenotypes or breast cancer risk, while 12q22 is a known locus for both MD and breast cancer risk. Among known MD and breast cancer risk SNPs, we identified four variants that were associated with V at the Bonferroni-corrected thresholds accounting for the number of SNPs tested: rs335189 (5q23.2) in PRDM6, rs13256025 (8p21.2) in EBF2, rs11836164 (12p12.1) near SSPN, and rs17817449 (16q12.2) in FTO. We observed significant genetic correlations between V and mammographic dense area (rg = 0.79, P = 5.91 × 10-5), percent density (rg = 0.73, P = 1.00 × 10-4), and adult BMI (rg = - 0.36, P = 3.88 × 10-7). Additional significant relationships were observed for non-dense area (z = - 4.14, P = 3.42 × 10-5), estrogen receptor-positive breast cancer (z = 3.41, P = 6.41 × 10-4), and childhood body fatness (z = - 4.91, P = 9.05 × 10-7) from the SNP-set tests. CONCLUSIONS These findings provide new insights into the genetic basis of mammographic texture variation and their associations with MD, breast cancer risk, and other breast cancer risk factors.
Collapse
Affiliation(s)
- Yuxi Liu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building 2-249A, Boston, MA, 02115, USA
| | - Hongjie Chen
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - John Heine
- Division of Population Sciences, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Sara Lindstrom
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Constance Turman
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Erica T Warner
- Clinical and Translational Epidemiology Unit, Department of Medicine, Mongan Institute, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Stacey J Winham
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Celine M Vachon
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Rulla M Tamimi
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building 2-249A, Boston, MA, 02115, USA.
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Xia Jiang
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Visionsgatan 18, 171 77, Solna, Stockholm, Sweden.
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
29
|
Mathur R, Fang F, Gaddis N, Hancock DB, Cho MH, Hokanson JE, Bierut LJ, Lutz SM, Young K, Smith AV, Silverman EK, Page GP, Johnson EO. GAWMerge expands GWAS sample size and diversity by combining array-based genotyping and whole-genome sequencing. Commun Biol 2022; 5:806. [PMID: 35953715 PMCID: PMC9372058 DOI: 10.1038/s42003-022-03738-6] [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: 12/21/2021] [Accepted: 07/18/2022] [Indexed: 11/09/2022] Open
Abstract
Genome-wide association studies (GWAS) have made impactful discoveries for complex diseases, often by amassing very large sample sizes. Yet, GWAS of many diseases remain underpowered, especially for non-European ancestries. One cost-effective approach to increase sample size is to combine existing cohorts, which may have limited sample size or be case-only, with public controls, but this approach is limited by the need for a large overlap in variants across genotyping arrays and the scarcity of non-European controls. We developed and validated a protocol, Genotyping Array-WGS Merge (GAWMerge), for combining genotypes from arrays and whole-genome sequencing, ensuring complete variant overlap, and allowing for diverse samples like Trans-Omics for Precision Medicine to be used. Our protocol involves phasing, imputation, and filtering. We illustrated its ability to control technology driven artifacts and type-I error, as well as recover known disease-associated signals across technologies, independent datasets, and ancestries in smoking-related cohorts. GAWMerge enables genetic studies to leverage existing cohorts to validly increase sample size and enhance discovery for understudied traits and ancestries.
Collapse
Affiliation(s)
- Ravi Mathur
- GenOmics, Bioinformatics, and Translational Research Center, RTI International, Research Triangle Park, NC, USA
| | - Fang Fang
- GenOmics, Bioinformatics, and Translational Research Center, RTI International, Research Triangle Park, NC, USA
| | - Nathan Gaddis
- GenOmics, Bioinformatics, and Translational Research Center, RTI International, Research Triangle Park, NC, USA
| | - Dana B Hancock
- GenOmics, Bioinformatics, and Translational Research Center, RTI International, Research Triangle Park, NC, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - John E Hokanson
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, Aurora, CO, USA
| | - Laura J Bierut
- Department of Psychiatry, Washington University, St. Louis, MO, USA
| | - Sharon M Lutz
- PRecisiOn Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care, Boston, MA, USA
| | - Kendra Young
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, Aurora, CO, USA
| | - Albert V Smith
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Grier P Page
- GenOmics, Bioinformatics, and Translational Research Center, RTI International, Research Triangle Park, NC, USA
- Fellow Program, RTI International, Research Triangle Park, NC, USA
| | - Eric O Johnson
- GenOmics, Bioinformatics, and Translational Research Center, RTI International, Research Triangle Park, NC, USA.
- Fellow Program, RTI International, Research Triangle Park, NC, USA.
| |
Collapse
|
30
|
Powell MJ, Fuller S, Gunderson EP, Benz CC. Reduced cardiovascular risks in women with endometriosis or polycystic ovary syndrome carrying a common functional IGF1R variant. Hum Reprod 2022; 37:1083-1094. [PMID: 35362533 PMCID: PMC9071223 DOI: 10.1093/humrep/deac059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 03/03/2022] [Indexed: 11/28/2022] Open
Abstract
STUDY QUESTION Is the increased future cardiovascular risk seen in women with endometriosis or polycystic ovary syndrome (PCOS) mitigated by functional insulin-like growth factor-1 receptor (IGF1R) single-nucleotide polymorphism (SNP) rs2016347 as previously shown in women with hypertensive disorders of pregnancy? SUMMARY ANSWER This cohort study found that women with endometriosis or PCOS who carry a T allele of IGF1R SNP rs2016347 had a reduced future risk of developing cardiovascular disease (CVD) and associated risk factors, with risk reduction dependent on cohort era. WHAT IS KNOWN ALREADY Women with endometriosis or PCOS have been shown to have an increased future risk of CVD and associated risk factors with limited predictive ability. STUDY DESIGN, SIZE, DURATION This retrospective cohort study took place in the Nurses' Health Study 2 (NHS2), which enrolled 116 430 participants in 1989 who were followed through 2015. The study population was analyzed in its entirety, and subdivided into entry (pre-1989) and after entry (post-1989) exposure cohorts. All NHS2 participants were eligible for inclusion in the study, 9599 (8.2%) were excluded for missing covariates. PARTICIPANTS/MATERIALS, SETTING, METHODS The NHS2 enrolled female registered nurses from 14 different states who ranged in age from 25 to 42 years at study entry. Data were collected from entry and biennial questionnaires, and analysis conducted from November 2020 to June 2021. Cox proportional hazard models were used to assess risk of CVD, hypertension (HTN), hypercholesterolemia (HC) and type 2 diabetes, both with and without genotyping for rs2016347. MAIN RESULTS AND THE ROLE OF CHANCE While women without endometriosis or PCOS, as a whole, demonstrated no impact of genotype on risk in either cohort, women with endometriosis carrying a T allele had a lower risk of CVD (hazard ratio (HR), 0.48; 95% CI, 0.27-0.86, P = 0.02) and HTN (HR, 0.80; 95% CI, 0.66-0.97, P = 0.03) in the pre-1989 cohort, while those in the post-1989 cohort had a decrease in risk for HC (HR, 0.76; 95% CI, 0.62-0.94, P = 0.01). Women with PCOS in the post-1989 cohort showed a significant protective impact of the T allele on HTN (HR, 0.44; 95% CI, 0.27-0.73, P = 0.002) and HC (HR, 0.62; 95% CI, 0.40-0.95, P = 0.03). LIMITATIONS, REASONS FOR CAUTION Data on specific endometriosis lesion locations or disease stage, as well as on PCOS phenotypes were lacking. In addition, data on systemic medical treatments beyond the use of oral contraceptives were missing, and these treatments may have confounded the results. WIDER IMPLICATIONS OF THE FINDINGS These findings implicate systemic dysregulation of the insulin-like growth factor-1 axis in the development of HTN, HC and clinical CVD in endometriosis and PCOS, suggesting a common underlying pathogenetic mechanism. STUDY FUNDING/COMPETING INTEREST(S) The NHS2 infrastructure for questionnaire data collection was supported by National Institute of Health (NIH) grant U01CA176726. This work was also supported in part by NIH and National Cancer Institute grant U24CA210990; as well, research effort and publication costs were supported by the Elizabeth MA Stevens donor funds provided to the Buck Institute for Research on Aging. The authors declare they have no conflicts of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Mark J Powell
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Sophia Fuller
- Graduate Group in Biostatistics, University of California, Berkeley, School of Public Health, Berkeley, CA, USA
| | - Erica P Gunderson
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
- Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA, USA
| | | |
Collapse
|
31
|
Lim J, Wirth J, Wu K, Giovannucci E, Kraft P, Turman C, Song M, Jovani M, Chan AT, Joshi AD. Obesity, Adiposity, and Risk of Symptomatic Gallstone Disease According to Genetic Susceptibility. Clin Gastroenterol Hepatol 2022; 20:e1083-e1120. [PMID: 34217876 PMCID: PMC8720320 DOI: 10.1016/j.cgh.2021.06.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/12/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Adiposity has been consistently associated with gallstone disease risk. We aimed to characterize associations of anthropometric measures (body mass index [BMI], recent weight change, long-term weight change, waist circumference, and waist-to-hip ratio) with symptomatic gallstone disease according to strata of gallstone disease polygenic risk score (PRS). METHODS We conducted analysis among 34,626 participants with available genome-wide genetic data within 3 large, prospective, U.S. cohorts-the Nurses' Health Study (NHS), Health Professionals Follow-Up Study, and NHS II. We characterized joint associations of PRS and anthropometric measures and tested for interactions on the relative and absolute risk scales. RESULTS Women in the highest BMI and PRS categories (BMI ≥30 kg/m2 and PRS ≥1 SD above mean) had odds ratio for gallstone disease of 5.55 (95% confidence interval, 5.29 to 5.81) compared with those in the lowest BMI and PRS categories (BMI <25 kg/m2 and PRS <1 SD below the mean). The corresponding odds ratio among men was 1.65 (95% confidence interval, 1.02 to 2.29). Associations for BMI did not vary within strata of PRS on the relative risk scale. On the absolute risk scale, the incidence rate difference between obese and normal-weight individuals was 1086 per 100,000 person-years within the highest PRS category, compared with 666 per 100,000 person-years in the lowest PRS category, with strong evidence for interaction with the ABCG8 locus. CONCLUSIONS While maintenance of a healthy body weight reduces gallstone disease risk among all individuals, risk reduction is higher among the subset with greater genetic susceptibility to gallstone disease.
Collapse
Affiliation(s)
- Junghyun Lim
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Janine Wirth
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; UCD Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Kana Wu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Edward Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Constance Turman
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Mingyang Song
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Manol Jovani
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Amit D Joshi
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
32
|
Fassier P, Kang JH, Lee IM, Grodstein F, Vercambre MN. Vigorous Physical Activity and Cognitive Trajectory Later in Life: Prospective Association and Interaction by Apolipoprotein E e4 in the Nurses' Health Study. J Gerontol A Biol Sci Med Sci 2022; 77:817-825. [PMID: 34125204 PMCID: PMC8974346 DOI: 10.1093/gerona/glab169] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The apolipoprotein E (APOE) e4 allele is a well-established genetic risk factor of brain aging. Vigorous physical activity may be particularly important in APOE-e4 carriers, but data have been inconsistent, likely due to differences in the timing of the physical activity assessment, definition of cognitive decline, and/or sample size. METHODS We prospectively evaluated the association between vigorous physical activity and cognition assessed at least 9 years later, according to APOE-e4 carrier status. Biennially from 1986, Nurses' Health Study participants reported their leisure-time physical activities. Starting in 1995-2001 and through 2008, participants (aged 70+ years) underwent up to 4 repeated cognitive telephone assessments (6 tasks averaged together using z-scores). RESULTS Among 7252 women, latent process mixed models identified 3 major patterns of cognitive change over 6 years: high-stable, medium-stable, and decline. Taking the high-stable cognitive trajectory as the outcome reference in multinomial logistic regressions, highest tertile of vigorous physical activity (≥5.9 metabolic-equivalent [MET]-hours/wk) compared to lowest tertile (≤0.9 MET-hours/wk) was significantly associated with subsequent lower likelihood of the medium-stable trajectory in the global score (odds ratio [OR] [95% CI] = 0.72 [0.63, 0.82]), verbal memory (OR [95% CI] = 0.78 [0.68-0.89]), and telephone interview of cognitive status score (OR [95% CI] = 0.81 [0.70-0.94]). Vigorous physical activity was also associated with lower likelihood of decline in category fluency (OR [95% CI] = 0.72 [0.56, 0.92]). We observed some evidence (p-interaction = .07 for the global score) that the association was stronger among APOE-e4 carriers than noncarriers (OR [95% CI] = 0.60 [0.39, 0.92] vs 0.82 [0.59, 1.16]). CONCLUSION Midlife vigorous physical activity was associated with better cognitive trajectories in women in their seventies, with suggestions of stronger associations among APOE-e4 carriers.
Collapse
Affiliation(s)
| | - Jae Hee Kang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - I-Min Lee
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Francine Grodstein
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois, USA
| | | |
Collapse
|
33
|
Polygenic scores, diet quality, and type 2 diabetes risk: An observational study among 35,759 adults from 3 US cohorts. PLoS Med 2022; 19:e1003972. [PMID: 35472203 PMCID: PMC9041832 DOI: 10.1371/journal.pmed.1003972] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 03/21/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Both genetic and lifestyle factors contribute to the risk of type 2 diabetes, but the extent to which there is a synergistic effect of the 2 factors is unclear. The aim of this study was to examine the joint associations of genetic risk and diet quality with incident type 2 diabetes. METHODS AND FINDINGS We analyzed data from 35,759 men and women in the United States participating in the Nurses' Health Study (NHS) I (1986 to 2016) and II (1991 to 2017) and the Health Professionals Follow-up Study (HPFS; 1986 to 2016) with available genetic data and who did not have diabetes, cardiovascular disease, or cancer at baseline. Genetic risk was characterized using both a global polygenic score capturing overall genetic risk and pathway-specific polygenic scores denoting distinct pathophysiological mechanisms. Diet quality was assessed using the Alternate Healthy Eating Index (AHEI). Cox models were used to calculate hazard ratios (HRs) for type 2 diabetes after adjusting for potential confounders. With over 902,386 person-years of follow-up, 4,433 participants were diagnosed with type 2 diabetes. The relative risk of type 2 diabetes was 1.29 (95% confidence interval [CI] 1.25, 1.32; P < 0.001) per standard deviation (SD) increase in global polygenic score and 1.13 (1.09, 1.17; P < 0.001) per 10-unit decrease in AHEI. Irrespective of genetic risk, low diet quality, as compared to high diet quality, was associated with approximately 30% increased risk of type 2 diabetes (Pinteraction = 0.69). The joint association of low diet quality and increased genetic risk was similar to the sum of the risk associated with each factor alone (Pinteraction = 0.30). Limitations of this study include the self-report of diet information and possible bias resulting from inclusion of highly educated participants with available genetic data. CONCLUSIONS These data provide evidence for the independent associations of genetic risk and diet quality with incident type 2 diabetes and suggest that a healthy diet is associated with lower diabetes risk across all levels of genetic risk.
Collapse
|
34
|
Oral postmenopausal hormone therapy and genetic risk on venous thromboembolism: gene-hormone interaction results from a large prospective cohort study. Menopause 2022; 29:293-303. [PMID: 35013060 PMCID: PMC8881382 DOI: 10.1097/gme.0000000000001924] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Oral postmenopausal hormone therapy (HT) has been shown to be associated with venous thromboembolism (VTE), but whether this association is modified by VTE-associated genetic susceptibility is unknown. We examined interactions between oral HT use and a genetic risk score (GRS) of VTE. METHOD Eligible women were postmenopausal women who had data on oral HT use, VTE incidence between 1990 and 2012, and genetic data in the Nurses' Health Study. We built a GRS aggregating 16 VTE-related genetic variants. We used Cox regression to estimate associations of HT use with incident VTE and assessed interactions between HT use and VTE GRS. We also estimated incidence of VTE between age 50 and 79 years for groups of women defined by HT use and VTE GRS. RESULTS We identified 432 incident VTE cases. Current HT users were at higher risk of VTE than never users (HR: 1.9, 95% CI: 1.5-2.6), with slightly higher risk for estrogen plus progestin HT than estrogen only (HR: 2.4 vs 1.9). The GRS was associated with VTE risk (HR comparing 4th quartile to 1st: 2.0, 95% CI: 1.2-3.4). We did not observe significant multiplicative interactions between HT use and GRS. The estimated VTE risk difference (per 10,000 person-years) comparing 50-year-old current HT users to never users was 22.5 for women in the highest GRS quartile and 9.8 for women in the lowest GRS quartile. CONCLUSION The VTE GRS might inform clinical guidance regarding the balance of risks and benefits of HT use, especially among younger women.
Collapse
|
35
|
Cornelis MC, van Dam RM. Genetic determinants of liking and intake of coffee and other bitter foods and beverages. Sci Rep 2021; 11:23845. [PMID: 34903748 PMCID: PMC8669025 DOI: 10.1038/s41598-021-03153-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/25/2021] [Indexed: 11/16/2022] Open
Abstract
Coffee is a widely consumed beverage that is naturally bitter and contains caffeine. Genome-wide association studies (GWAS) of coffee drinking have identified genetic variants involved in caffeine-related pathways but not in taste perception. The taste of coffee can be altered by addition of milk/sweetener, which has not been accounted for in GWAS. Using UK and US cohorts, we test the hypotheses that genetic variants related to taste are more strongly associated with consumption of black coffee than with consumption of coffee with milk or sweetener and that genetic variants related to caffeine pathways are not differentially associated with the type of coffee consumed independent of caffeine content. Contrary to our hypotheses, genetically inferred caffeine sensitivity was more strongly associated with coffee taste preferences than with genetically inferred bitter taste perception. These findings extended to tea and dark chocolate. Taste preferences and physiological caffeine effects intertwine in a way that is difficult to distinguish for individuals which may represent conditioned taste preferences.
Collapse
Affiliation(s)
- Marilyn C Cornelis
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 680 North Lake Shore Drive, Suite 1400, Chicago, IL, 60611, USA.
| | - Rob M van Dam
- Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
- Department of Epidemiology, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
36
|
Shi J, Swanson SA, Kraft P, Rosner B, De Vivo I, Hernán MA. Instrumental variable estimation for a time-varying treatment and a time-to-event outcome via structural nested cumulative failure time models. BMC Med Res Methodol 2021; 21:258. [PMID: 34823502 PMCID: PMC8620657 DOI: 10.1186/s12874-021-01449-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/21/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND In many applications of instrumental variable (IV) methods, the treatments of interest are intrinsically time-varying and outcomes of interest are failure time outcomes. A common example is Mendelian randomization (MR), which uses genetic variants as proposed IVs. In this article, we present a novel application of g-estimation of structural nested cumulative failure models (SNCFTMs), which can accommodate multiple measures of a time-varying treatment when modelling a failure time outcome in an IV analysis. METHODS A SNCFTM models the ratio of two conditional mean counterfactual outcomes at time k under two treatment strategies which differ only at an earlier time m. These models can be extended to accommodate inverse probability of censoring weights, and can be applied to case-control data. We also describe how the g-estimates of the SNCFTM parameters can be used to calculate marginal cumulative risks under nondynamic treatment strategies. We examine the performance of this method using simulated data, and present an application of these models by conducting an MR study of alcohol intake and endometrial cancer using longitudinal observational data from the Nurses' Health Study. RESULTS Our simulations found that estimates from SNCFTMs which used an IV approach were similar to those obtained from SNCFTMs which adjusted for confounders, and similar to those obtained from the g-formula approach when the outcome was rare. In our data application, the cumulative risk of endometrial cancer from age 45 to age 72 under the "never drink" strategy (4.0%) was similar to that under the "always ½ drink per day" strategy (4.3%). CONCLUSIONS SNCFTMs can be used to conduct MR and other IV analyses with time-varying treatments and failure time outcomes.
Collapse
Affiliation(s)
- Joy Shi
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- The CAUSALab, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Sonja A Swanson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- The CAUSALab, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Bernard Rosner
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Immaculata De Vivo
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Miguel A Hernán
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- The CAUSALab, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
37
|
Wooding SP, Ramirez VA, Behrens M. Bitter taste receptors: Genes, evolution and health. Evol Med Public Health 2021; 9:431-447. [PMID: 35154779 PMCID: PMC8830313 DOI: 10.1093/emph/eoab031] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/05/2021] [Indexed: 02/01/2023] Open
Abstract
Bitter taste perception plays vital roles in animal behavior and fitness. By signaling the presence of toxins in foods, particularly noxious defense compounds found in plants, it enables animals to avoid exposure. In vertebrates, bitter perception is initiated by TAS2Rs, a family of G protein-coupled receptors expressed on the surface of taste buds. There, oriented toward the interior of the mouth, they monitor the contents of foods, drinks and other substances as they are ingested. When bitter compounds are encountered, TAS2Rs respond by triggering neural pathways leading to sensation. The importance of this role placed TAS2Rs under selective pressures in the course of their evolution, leaving signatures in patterns of gene gain and loss, sequence polymorphism, and population structure consistent with vertebrates' diverse feeding ecologies. The protective value of bitter taste is reduced in modern humans because contemporary food supplies are safe and abundant. However, this is not always the case. Some crops, particularly in the developing world, retain surprisingly high toxicity and bitterness remains an important measure of safety. Bitter perception also shapes health through its influence on preference driven behaviors such as diet choice, alcohol intake and tobacco use. Further, allelic variation in TAS2Rs is extensive, leading to individual differences in taste sensitivity that drive these behaviors, shaping susceptibility to disease. Thus, bitter taste perception occupies a critical intersection between ancient evolutionary processes and modern human health.
Collapse
Affiliation(s)
- Stephen P Wooding
- Department of Anthropology and Health Sciences Research Institute, University of California, Merced, CA, USA
| | - Vicente A Ramirez
- Department of Public Health, University of California, Merced, CA, USA
| | - Maik Behrens
- Maik Behrens, Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| |
Collapse
|
38
|
Fang Z, Hang D, Wang K, Joshi A, Wu K, Chan AT, Ogino S, Giovannucci EL, Song M. Risk prediction models for colorectal cancer: Evaluating the discrimination due to added biomarkers. Int J Cancer 2021; 149:1021-1030. [PMID: 33948940 DOI: 10.1002/ijc.33621] [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: 02/05/2021] [Revised: 04/07/2021] [Accepted: 04/20/2021] [Indexed: 02/05/2023]
Abstract
Most risk prediction models for colorectal cancer (CRC) are based on questionnaires and show a modest discriminatory ability. Therefore, we aim to develop risk prediction models incorporating plasma biomarkers for CRC to improve discrimination. We assessed the predictivity of 11 biomarkers in 736 men in the Health Professionals Follow-up Study and 639 women in the Nurses' Health Study. We used stepwise logistic regression to examine whether a set of biomarkers improved the predictivity on the basis of predictors in the National Cancer Institute's (NCI) Colorectal Cancer Risk Assessment Tool. Model discrimination was assessed using C-statistics. Bootstrap with 500 randomly sampled replicates was used for internal validation. The models containing each biomarker generated a C-statistic ranging from 0.50 to 0.59 in men and 0.50 to 0.54 in women. The NCI model demonstrated a C-statistic (95% CI) of 0.67 (0.62-0.71) in men and 0.58 (0.54-0.63) in women. Through stepwise selection of biomarkers, the C-statistic increased to 0.70 (0.66-0.74) in men after adding growth/differentiation factor 15, total adiponectin, sex hormone binding globulin and tumor necrosis factor receptor superfamily member 1B (P for difference = 0.008); and increased to 0.62 (0.57-0.66) in women after further including insulin-like growth factor 1 and insulin-like growth factor-binding protein 3 (P for difference = .06). The NCI + selected biomarkers model was internally validated with a C-statistic (95% CI) of 0.73 (0.70-0.77) in men and 0.66 (0.61-0.70) in women. Circulating plasma biomarkers may improve the performance of risk factor-based prediction model for CRC.
Collapse
Affiliation(s)
- Zhe Fang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Dong Hang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Kai Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Amit Joshi
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kana Wu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Shuji Ogino
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Edward L Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mingyang Song
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
39
|
Huang M, Lyu C, Li X, Qureshi AA, Han J, Li M. Identifying Susceptibility Loci for Cutaneous Squamous Cell Carcinoma Using a Fast Sequence Kernel Association Test. Front Genet 2021; 12:657499. [PMID: 34040636 PMCID: PMC8141858 DOI: 10.3389/fgene.2021.657499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 04/09/2021] [Indexed: 11/13/2022] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) accounts for about 20% of all skin cancers, the most common type of malignancy in the United States. Genome-wide association studies (GWAS) have successfully identified multiple genetic variants associated with the risk of cSCC. Most of these studies were single-locus-based, testing genetic variants one-at-a-time. In this article, we performed gene-based association tests to evaluate the joint effect of multiple variants, especially rare variants, on the risk of cSCC by using a fast sequence kernel association test (fastSKAT). The study included 1,710 cSCC cases and 24,304 cancer-free controls from the Nurses' Health Study, the Nurses' Health Study II and the Health Professionals Follow-up Study. We used UCSC Genome Browser to define gene units as candidate loci, and further evaluated the association between all variants within each gene unit and disease outcome. Four genes HP1BP3, DAG1, SEPT7P2, and SLFN12 were identified using Bonferroni adjusted significance level. Our study is complementary to the existing GWASs, and our findings may provide additional insights into the etiology of cSCC. Further studies are needed to validate these findings.
Collapse
Affiliation(s)
- Manyan Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University at Bloomington, Bloomington, IN, United States
| | - Chen Lyu
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University at Bloomington, Bloomington, IN, United States
| | - Xin Li
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University - Purdue University Indianapolis, Indianapolis, IN, United States.,Melvin and Bren Simon Cancer Center, Indianapolis, IN, United States
| | - Abrar A Qureshi
- Department of Dermatology, Alpert Medical School, Brown University, Providence, RI, United States
| | - Jiali Han
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University - Purdue University Indianapolis, Indianapolis, IN, United States.,Melvin and Bren Simon Cancer Center, Indianapolis, IN, United States
| | - Ming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University at Bloomington, Bloomington, IN, United States
| |
Collapse
|
40
|
Warner ET, Jiang L, Adjei DN, Turman C, Gordon W, Wang L, Tamimi R, Kraft P, Lindström S. A Genome-Wide Association Study of Childhood Body Fatness. Obesity (Silver Spring) 2021; 29:446-453. [PMID: 33491310 PMCID: PMC7842657 DOI: 10.1002/oby.23070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 11/05/2022]
Abstract
OBJECTIVE This study aimed to uncover genetic contributors to adiposity in early life. METHODS A genome-wide association study of childhood body fatness in 34,401 individuals within the Nurses' Health Studies and the Health Professionals Follow-up Study was conducted. Data were imputed to the 1000 Genomes Phase 3 version 5 reference panel. RESULTS A total of 1,354 single-nucleotide polymorphisms (P < 10-4 ) were selected for replication in a previously published genome-wide association study of childhood BMI. Nineteen significant genome-wide (P < 5 × 10-8 ) regions were observed, fourteen of which were previously associated with childhood obesity and five were novel: BNDF (P = 7.58 × 10-13 ), PRKD1 (P = 1.43 × 10-10 ), 20p13 (P = 2.05 × 10-10 ), FHIT (P = 1.77 × 10-8 ), and LOC101927575 (P = 3.22 × 10-8 ). The BNDF, FHIT, and PRKD1 regions were previously associated with adult BMI. LOC101927575 and 20p13 regions have not previously been associated with adiposity phenotypes. In a transcriptome-wide analysis, associations for POMC at 2p23.3 (P = 3.36 × 10-6 ) and with TMEM18 at 2p25.3 (P = 3.53 × 10-7 ) were observed. Childhood body fatness was genetically correlated with hip (rg = 0.42, P = 4.44 × 10-16 ) and waist circumference (rg = 0.39, P = 5.56 × 10-16 ), as well as age at menarche (rg = -0.37, P = 7.96 × 10-19 ). CONCLUSIONS Additional loci that contribute to childhood adiposity were identified, further explicating its genetic architecture.
Collapse
Affiliation(s)
- Erica T. Warner
- Clinical Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Lai Jiang
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA
| | - David Nana Adjei
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA
| | - Constance Turman
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA
| | - William Gordon
- Department of Epidemiology, University of Washington, Seattle, WA
| | - Lu Wang
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA
| | - Rulla Tamimi
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Peter Kraft
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA
| | - Sara Lindström
- Department of Epidemiology, University of Washington, Seattle, WA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
41
|
Li J, Guasch-Ferré M, Chung W, Ruiz-Canela M, Toledo E, Corella D, Bhupathiraju SN, Tobias DK, Tabung FK, Hu J, Zhao T, Turman C, Feng YCA, Clish CB, Mucci L, Eliassen AH, Costenbader KH, Karlson EW, Wolpin BM, Ascherio A, Rimm EB, Manson JE, Qi L, Martínez-González MÁ, Salas-Salvadó J, Hu FB, Liang L. The Mediterranean diet, plasma metabolome, and cardiovascular disease risk. Eur Heart J 2020; 41:2645-2656. [PMID: 32406924 PMCID: PMC7377580 DOI: 10.1093/eurheartj/ehaa209] [Citation(s) in RCA: 197] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/10/2020] [Accepted: 03/18/2020] [Indexed: 12/16/2022] Open
Abstract
AIMS To investigate whether metabolic signature composed of multiple plasma metabolites can be used to characterize adherence and metabolic response to the Mediterranean diet and whether such a metabolic signature is associated with cardiovascular disease (CVD) risk. METHODS AND RESULTS Our primary study cohort included 1859 participants from the Spanish PREDIMED trial, and validation cohorts included 6868 participants from the US Nurses' Health Studies I and II, and Health Professionals Follow-up Study (NHS/HPFS). Adherence to the Mediterranean diet was assessed using a validated Mediterranean Diet Adherence Screener (MEDAS), and plasma metabolome was profiled by liquid chromatography-tandem mass spectrometry. We observed substantial metabolomic variation with respect to Mediterranean diet adherence, with nearly one-third of the assayed metabolites significantly associated with MEDAS (false discovery rate < 0.05). Using elastic net regularized regressions, we identified a metabolic signature, comprised of 67 metabolites, robustly correlated with Mediterranean diet adherence in both PREDIMED and NHS/HPFS (r = 0.28-0.37 between the signature and MEDAS; P = 3 × 10-35 to 4 × 10-118). In multivariable Cox regressions, the metabolic signature showed a significant inverse association with CVD incidence after adjusting for known risk factors (PREDIMED: hazard ratio [HR] per standard deviation increment in the signature = 0.71, P < 0.001; NHS/HPFS: HR = 0.85, P = 0.001), and the association persisted after further adjustment for MEDAS scores (PREDIMED: HR = 0.73, P = 0.004; NHS/HPFS: HR = 0.85, P = 0.004). Further genome-wide association analysis revealed that the metabolic signature was significantly associated with genetic loci involved in fatty acids and amino acids metabolism. Mendelian randomization analyses showed that the genetically inferred metabolic signature was significantly associated with risk of coronary heart disease (CHD) and stroke (odds ratios per SD increment in the genetically inferred metabolic signature = 0.92 for CHD and 0.91 for stroke; P < 0.001). CONCLUSIONS We identified a metabolic signature that robustly reflects adherence and metabolic response to a Mediterranean diet, and predicts future CVD risk independent of traditional risk factors, in Spanish and US cohorts.
Collapse
Affiliation(s)
- Jun Li
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 3rd Floor, Boston, MA 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 2nd Floor, Boston, MA 02115, USA
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 3rd Floor, Boston, MA 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA
| | - Wonil Chung
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 2nd Floor, Boston, MA 02115, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 4th Floor, Boston, MA 02115, USA
| | - Miguel Ruiz-Canela
- Department of Preventive Medicine and Public Health, University of Navarra, Irunlarrea 1, Pamplona 31008, Spain
- IdiSNA (Instituto de Investigación Sanitaria de Navarra), Edificio LUNA-Navarrabiomed, Irunlarrea 3, Pamplona 31008, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, C/Monforte de Lemos 3-5, Pabellón 11, Madrid 28029, Spain
| | - Estefanía Toledo
- Department of Preventive Medicine and Public Health, University of Navarra, Irunlarrea 1, Pamplona 31008, Spain
- IdiSNA (Instituto de Investigación Sanitaria de Navarra), Edificio LUNA-Navarrabiomed, Irunlarrea 3, Pamplona 31008, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, C/Monforte de Lemos 3-5, Pabellón 11, Madrid 28029, Spain
| | - Dolores Corella
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, C/Monforte de Lemos 3-5, Pabellón 11, Madrid 28029, Spain
- Department of Preventive Medicine, University of Valencia, Valencia 46010, Spain
| | - Shilpa N Bhupathiraju
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 3rd Floor, Boston, MA 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA
| | - Deirdre K Tobias
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 3rd Floor, Boston, MA 02115, USA
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 900 Commonwealth Avenue, Boston, MA 02115, USA
| | - Fred K Tabung
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 3rd Floor, Boston, MA 02115, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University College of Medicine and Comprehensive Cancer Center – James Cancer Hospital and Solove Research Institute, 410 W 12th Ave Columbus, OH 43210, USA
| | - Jie Hu
- Division of Women’s Health, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 1620 Tremont St, 3rd floor, Boston, MA 02120, USA
| | - Tong Zhao
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 4th Floor, Boston, MA 02115, USA
| | - Constance Turman
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 2nd Floor, Boston, MA 02115, USA
| | - Yen-Chen Anne Feng
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 2nd Floor, Boston, MA 02115, USA
- Metabolomics Platform,Broad Institute of Harvard and MIT, 415 Main St, Cambridge, MA 02142, USA
| | - Clary B Clish
- Metabolomics Platform,Broad Institute of Harvard and MIT, 415 Main St, Cambridge, MA 02142, USA
| | - Lorelei Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 2nd Floor, Boston, MA 02115, USA
| | - A Heather Eliassen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 2nd Floor, Boston, MA 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA
| | - Karen H Costenbader
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115, USA
| | - Elizabeth W Karlson
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115, USA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Alberto Ascherio
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 3rd Floor, Boston, MA 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 2nd Floor, Boston, MA 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA
| | - Eric B Rimm
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 3rd Floor, Boston, MA 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 2nd Floor, Boston, MA 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA
| | - JoAnn E Manson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 2nd Floor, Boston, MA 02115, USA
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 900 Commonwealth Avenue, Boston, MA 02115, USA
- Mary Horrigan Connors Center for Women’s Health and Gender Biology, Brigham and Women’s Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Lu Qi
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 3rd Floor, Boston, MA 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, 1440 Canal Street, New Orleans, LA 70112, USA
| | - Miguel Ángel Martínez-González
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 3rd Floor, Boston, MA 02115, USA
- Department of Preventive Medicine and Public Health, University of Navarra, Irunlarrea 1, Pamplona 31008, Spain
- IdiSNA (Instituto de Investigación Sanitaria de Navarra), Edificio LUNA-Navarrabiomed, Irunlarrea 3, Pamplona 31008, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, C/Monforte de Lemos 3-5, Pabellón 11, Madrid 28029, Spain
| | - Jordi Salas-Salvadó
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, C/Monforte de Lemos 3-5, Pabellón 11, Madrid 28029, Spain
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, C/Sant Llorenç 21, Reus 43201, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 3rd Floor, Boston, MA 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 2nd Floor, Boston, MA 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA
| | - Liming Liang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 2nd Floor, Boston, MA 02115, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II 4th Floor, Boston, MA 02115, USA
| |
Collapse
|
42
|
Ding M, Ahmad S, Qi L, Hu Y, Bhupathiraju SN, Guasch-Ferré M, Jensen MK, Chavarro JE, Ridker PM, Willett WC, Chasman DI, Hu FB, Kraft P. Additive and Multiplicative Interactions Between Genetic Risk Score and Family History and Lifestyle in Relation to Risk of Type 2 Diabetes. Am J Epidemiol 2020; 189:445-460. [PMID: 31647510 DOI: 10.1093/aje/kwz251] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 11/09/2018] [Accepted: 10/17/2019] [Indexed: 12/28/2022] Open
Abstract
We examined interactions between lifestyle factors and genetic risk of type 2 diabetes (T2D-GR), captured by genetic risk score (GRS) and family history (FH). Our initial study cohort included 20,524 European-ancestry participants, of whom 1,897 developed incident T2D, in the Nurses' Health Study (1984-2016), Nurses' Health Study II (1989-2016), and Health Professionals Follow-up Study (1986-2016). The analyses were replicated in 19,183 European-ancestry controls and 2,850 incident T2D cases in the Women's Genome Health Study (1992-2016). We defined 2 categories of T2D-GR: high GRS (upper one-third) with FH and low GRS or without FH. Compared with participants with the healthiest lifestyle and low T2D-GR, the relative risk of T2D for participants with the healthiest lifestyle and high T2D-GR was 2.24 (95% confidence interval (CI): 1.76, 2.86); for participants with the least healthy lifestyle and low T2D-GR, it was 4.05 (95% CI: 3.56, 4.62); and for participants with the least healthy lifestyle and high T2D-GR, it was 8.72 (95% CI: 7.46, 10.19). We found a significant departure from an additive risk difference model in both the initial and replication cohorts, suggesting that adherence to a healthy lifestyle could lead to greater absolute risk reduction among those with high T2D-GR. The public health implication is that a healthy lifestyle is important for diabetes prevention, especially for individuals with high GRS and FH of T2D.
Collapse
|
43
|
Hang D, Joshi AD, He X, Chan AT, Jovani M, Gala MK, Ogino S, Kraft P, Turman C, Peters U, Bien SA, Lin Y, Hu Z, Shen H, Wu K, Giovannucci EL, Song M. Colorectal cancer susceptibility variants and risk of conventional adenomas and serrated polyps: results from three cohort studies. Int J Epidemiol 2020; 49:259-269. [PMID: 31038671 PMCID: PMC7426026 DOI: 10.1093/ije/dyz096] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Increasing evidence suggests that conventional adenomas (CAs) and serrated polyps (SPs) represent two distinct groups of precursor lesions for colorectal cancer (CRC). The influence of common genetic variants on risk of CAs and SPs remain largely unknown. METHODS Among 27 426 participants within three prospective cohort studies, we created a weighted genetic risk score (GRS) based on 40 CRC-related single nucleotide polymorphisms (SNPs) identified in previous genome-wide association studies; and we examined the association of GRS (per one standard deviation increment) with risk of CAs, SPs and synchronous CAs and SPs, by multivariable logistic regression. We also analysed individual variants in the secondary analysis. RESULTS During 18-20 years of follow-up, we documented 2952 CAs, 1585 SPs and 794 synchronous CAs and SPs. Higher GRS was associated with increased risk of CAs [odds ratio (OR) = 1.17, 95% confidence interval (CI): 1.12-1.21] and SPs (OR = 1.09, 95% CI: 1.03-1.14), with a stronger association for CAs than SPs (Pheterogeneity=0.01). An even stronger association was found for patients with synchronous CAs and SPs (OR = 1.32), advanced CAs (OR = 1.22) and multiple CAs (OR = 1.25). Different sets of variants were associated with CAs and SPs, with a Spearman correlation coefficient of 0.02 between the ORs associating the 40 SNPs with the two lesions. After correcting for multiple testing, three variants were associated with CAs (rs3802842, rs6983267 and rs7136702) and two with SPs (rs16892766 and rs4779584). CONCLUSIONS Common genetic variants play a potential role in the conventional and serrated pathways of CRC. Different sets of variants are identified for the two pathways, further supporting the aetiological heterogeneity of CRC.
Collapse
Affiliation(s)
- Dong Hang
- Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, China
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Amit D Joshi
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Xiaosheng He
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Manol Jovani
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Manish K Gala
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shuji Ogino
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
- Program in MPE Molecular Pathological Epidemiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Constance Turman
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | - Stephanie A Bien
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Yi Lin
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Zhibin Hu
- Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, China
| | - Hongbing Shen
- Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, China
| | - Kana Wu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Edward L Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mingyang Song
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
44
|
Li M, Rahman ML, Wu J, Ding M, Chavarro JE, Lin Y, Ley SH, Bao W, Grunnet LG, Hinkle SN, Thuesen ACB, Yeung E, Gore-Langton RE, Sherman S, Hjort L, Kampmann FB, Bjerregaard AA, Damm P, Tekola-Ayele F, Liu A, Mills JL, Vaag A, Olsen SF, Hu FB, Zhang C. Genetic factors and risk of type 2 diabetes among women with a history of gestational diabetes: findings from two independent populations. BMJ Open Diabetes Res Care 2020; 8:8/1/e000850. [PMID: 31958311 PMCID: PMC7039588 DOI: 10.1136/bmjdrc-2019-000850] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/22/2019] [Accepted: 12/10/2019] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE Women with a history of gestational diabetes mellitus (GDM) have an exceptionally high risk for type 2 diabetes (T2D). Yet, little is known about genetic determinants for T2D in this population. We examined the association of a genetic risk score (GRS) with risk of T2D in two independent populations of women with a history of GDM and how this association might be modified by non-genetic determinants for T2D. RESEARCH DESIGN AND METHODS This cohort study included 2434 white women with a history of GDM from the Nurses' Health Study II (NHSII, n=1884) and the Danish National Birth Cohort (DNBC, n=550). A GRS for T2D was calculated using 59 candidate single nucleotide polymorphisms for T2D identified from genome-wide association studies in European populations. An alternate healthy eating index (AHEI) score was derived to reflect dietary quality after the pregnancy affected by GDM. RESULTS Women on average were followed for 21 years in NHSII and 13 years in DNBC, during which 446 (23.7%) and 155 (28.2%) developed T2D, respectively. The GRS was generally positively associated with T2D risk in both cohorts. In the pooled analysis, the relative risks (RRs) for increasing quartiles of GRS were 1.00, 0.97, 1.25 and 1.19 (p trend=0.02). In both cohorts, the association appeared to be stronger among women with poorer (AHEI <median) than better dietary quality (AHEI ≥median), although the interaction was not significant. For example, in NHSII, the RRs across increasing quartiles of GRS were 1.00, 0.99, 1.51 and 1.29 (p trend=0.06) among women with poorer dietary quality and 1.00, 0.83, 0.81 and 0.94 (p trend=0.79) among women with better dietary quality (p interaction=0.11). CONCLUSIONS Among white women with a history of GDM, higher GRS for T2D was associated with an increased risk of T2D.
Collapse
Affiliation(s)
- Mengying Li
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| | - Mohammad L Rahman
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
- Department of Population Medicine and Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Jing Wu
- Glotech, Rockville, Maryland, USA
| | - Ming Ding
- Department of Nutrition, Harvard University T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Jorge E Chavarro
- Department of Nutrition, Harvard University T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Yuan Lin
- Epidemiology Department, Richard M. Fairbanks School of Public Health, Indiana University, Bloomington, Indiana, USA
| | - Sylvia H Ley
- Department of Nutrition, Harvard University T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Wei Bao
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, Iowa, USA
| | - Louise G Grunnet
- Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
| | - Stefanie N Hinkle
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| | - Anne Cathrine B Thuesen
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Edwina Yeung
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| | | | - Seth Sherman
- The Emmes Company, LLC, Rockville, Maryland, USA
| | - Line Hjort
- Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
- Departments of Obstetrics, Center for Pregnant Women with Diabetes, Rigshospitalet, Copenhagen, Denmark
| | - Freja Bach Kampmann
- Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
- Division for Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Lyngby, Denmark
| | | | - Peter Damm
- Departments of Obstetrics, Center for Pregnant Women with Diabetes, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Fasil Tekola-Ayele
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| | - Aiyi Liu
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| | - James L Mills
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| | - Allan Vaag
- Early Clinical Development and Innovative Medicines, AstraZeneca, Mölndal, Sweden
| | - Sjurdur F Olsen
- Nutrition Group, Statens Serum Institut, Copenhagen, Denmark
| | - Frank B Hu
- Department of Nutrition, Harvard University T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Cuilin Zhang
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| |
Collapse
|
45
|
Abstract
PURPOSE Urinary incontinence and fecal incontinence are common disorders in women that negatively impact quality of life. In addition to known health and lifestyle risk factors, genetics may have a role in continence. Identification of genetic variants associated with urinary incontinence and fecal incontinence could result in a better understanding of etiologic pathways, and new interventions and treatments. MATERIALS AND METHODS We previously generated genome-wide single nucleotide polymorphism data from Nurses' Health Studies participants. The participants provided longitudinal urinary incontinence and fecal incontinence information via questionnaires. Cases of urinary incontinence (6,120) had at least weekly urinary incontinence reported on a majority of questionnaires (3 or 4 across 12 to 16 years) while controls (4,811) consistently had little to no urinary incontinence reported. We classified cases of urinary incontinence in women into stress (1,809), urgency (1,942) and mixed (2,036) subtypes. Cases of fecal incontinence (4,247) had at least monthly fecal incontinence reported on a majority of questionnaires while controls (11,634) consistently had no fecal incontinence reported. We performed a genome-wide association study for each incontinence outcome. RESULTS We identified 8 single nucleotide polymorphisms significantly associated (p <5×10-8) with urinary incontinence located in 2 loci, chromosomes 8q23.3 and 1p32.2. There were no genome-wide significant findings for the urinary incontinence subtype analyses. However, the significant associations for overall urinary incontinence were stronger for the urgency and mixed subtypes than for stress. While no single nucleotide polymorphism reached genome-wide significance for fecal incontinence, 4 single nucleotide polymorphisms had p <10-6. CONCLUSIONS Few studies have collected genetic data and detailed urinary incontinence and fecal incontinence information. This genome-wide association study provides initial evidence of genetic associations for urinary incontinence and merits further research to replicate our findings and identify additional risk variants.
Collapse
|
46
|
Lindström S, Wang L, Smith EN, Gordon W, van Hylckama Vlieg A, de Andrade M, Brody JA, Pattee JW, Haessler J, Brumpton BM, Chasman DI, Suchon P, Chen MH, Turman C, Germain M, Wiggins KL, MacDonald J, Braekkan SK, Armasu SM, Pankratz N, Jackson RD, Nielsen JB, Giulianini F, Puurunen MK, Ibrahim M, Heckbert SR, Damrauer SM, Natarajan P, Klarin D, de Vries PS, Sabater-Lleal M, Huffman JE, Bammler TK, Frazer KA, McCauley BM, Taylor K, Pankow JS, Reiner AP, Gabrielsen ME, Deleuze JF, O'Donnell CJ, Kim J, McKnight B, Kraft P, Hansen JB, Rosendaal FR, Heit JA, Psaty BM, Tang W, Kooperberg C, Hveem K, Ridker PM, Morange PE, Johnson AD, Kabrhel C, Trégouët DA, Smith NL. Genomic and transcriptomic association studies identify 16 novel susceptibility loci for venous thromboembolism. Blood 2019; 134:1645-1657. [PMID: 31420334 PMCID: PMC6871304 DOI: 10.1182/blood.2019000435] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 07/17/2019] [Indexed: 12/29/2022] Open
Abstract
Venous thromboembolism (VTE) is a significant contributor to morbidity and mortality. To advance our understanding of the biology contributing to VTE, we conducted a genome-wide association study (GWAS) of VTE and a transcriptome-wide association study (TWAS) based on imputed gene expression from whole blood and liver. We meta-analyzed GWAS data from 18 studies for 30 234 VTE cases and 172 122 controls and assessed the association between 12 923 718 genetic variants and VTE. We generated variant prediction scores of gene expression from whole blood and liver tissue and assessed them for association with VTE. Mendelian randomization analyses were conducted for traits genetically associated with novel VTE loci. We identified 34 independent genetic signals for VTE risk from GWAS meta-analysis, of which 14 are newly reported associations. This included 11 newly associated genetic loci (C1orf198, PLEK, OSMR-AS1, NUGGC/SCARA5, GRK5, MPHOSPH9, ARID4A, PLCG2, SMG6, EIF5A, and STX10) of which 6 replicated, and 3 new independent signals in 3 known genes. Further, TWAS identified 5 additional genetic loci with imputed gene expression levels differing between cases and controls in whole blood (SH2B3, SPSB1, RP11-747H7.3, RP4-737E23.2) and in liver (ERAP1). At some GWAS loci, we found suggestive evidence that the VTE association signal for novel and previously known regions colocalized with expression quantitative trait locus signals. Mendelian randomization analyses suggested that blood traits may contribute to the underlying risk of VTE. To conclude, we identified 16 novel susceptibility loci for VTE; for some loci, the association signals are likely mediated through gene expression of nearby genes.
Collapse
Affiliation(s)
- Sara Lindström
- Department of Epidemiology, University of Washington, Seattle, WA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Lu Wang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - Erin N Smith
- Department of Pediatrics and Rady Children's Hospital, University of California San Diego, La Jolla, CA
- K.G. Jebsen Thrombosis Research and Expertise Center, Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - William Gordon
- Department of Epidemiology, University of Washington, Seattle, WA
| | | | | | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | - Jack W Pattee
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Jeffrey Haessler
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Ben M Brumpton
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Clinic of Thoracic and Occupational Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Pierre Suchon
- Laboratory of Haematology, La Timone Hospital, Marseille, France
- Center for CardioVascular and Nutrition research (C2VN), Universite Aix-Marseille, Institut National de la Recherche Agronomique (INRA), INSERM, Marseille, France
| | - Ming-Huei Chen
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA
- The Framingham Heart Study, Framingham, MA
| | - Constance Turman
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Marine Germain
- INSERM UMR_S 1219, Bordeaux Population Health Research Center, University of Bordeaux, Bordeaux, France
| | - Kerri L Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - Sigrid K Braekkan
- K.G. Jebsen Thrombosis Research and Expertise Center, Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
- Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | | | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, MN
| | - Rebecca D Jackson
- Division of Endocrinology, Diabetes, and Metabolism, The Ohio State University, Columbus OH
| | - Jonas B Nielsen
- Division of Cardiology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| | - Franco Giulianini
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA
| | | | - Manal Ibrahim
- Laboratory of Haematology, La Timone Hospital, Marseille, France
| | - Susan R Heckbert
- Department of Epidemiology, University of Washington, Seattle, WA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA
| | - Scott M Damrauer
- Department of Surgery, Corporal Michael Crescenz VA Medical Center, Philadelphia, PA
- Department of Surgery, Perleman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Pradeep Natarajan
- Boston VA Healthcare System, Boston, MA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA
| | - Derek Klarin
- Boston VA Healthcare System, Boston, MA
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - 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
| | - Maria Sabater-Lleal
- Unit of Genomics of Complex Diseases, Institut de Recerca de l'Hospital de Sant Pau, IIB-Sant Pau, Barcelona, Spain
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Jennifer E Huffman
- Center for Population Genomics, MAVERIC, VA Boston Healthcare System, Boston, MA
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - Kelly A Frazer
- Department of Pediatrics and Rady Children's Hospital, University of California San Diego, La Jolla, CA
- K.G. Jebsen Thrombosis Research and Expertise Center, Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
- Institute of Genomic Medicine, University of California San Diego, La Jolla, CA
| | - Bryan M McCauley
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Kent Taylor
- Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-University of California Los Angeles Medical Center, Torrence CA
| | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, WA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Maiken E Gabrielsen
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine, Direction de la Recherche Fondamentale, Le Commissariat à l'énergie atomique et aux énergies alternatives, Evry, France
- The Centre d'Etude du Polymorphism Humain (CEPH), Fondation Jean Dausset, Paris, France
| | - Chris J O'Donnell
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA
- The Framingham Heart Study, Framingham, MA
- Million Veteran Program, Veteran's Administration, Boston, MA
| | - Jihye Kim
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Barbara McKnight
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Peter Kraft
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - John-Bjarne Hansen
- K.G. Jebsen Thrombosis Research and Expertise Center, Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
- Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - John A Heit
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Bruce M Psaty
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Services, University of Washington, Seattle, WA
| | - Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Charles Kooperberg
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Pierre-Emmanuel Morange
- Laboratory of Haematology, La Timone Hospital, Marseille, France
- Center for CardioVascular and Nutrition research (C2VN), Universite Aix-Marseille, Institut National de la Recherche Agronomique (INRA), INSERM, Marseille, France
- Centre de Ressources Biologiques Assistance Publique-Hôpitaux de Marseille, HemoVasc, Marseille, France
| | - Andrew D Johnson
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA
- The Framingham Heart Study, Framingham, MA
| | - Christopher Kabrhel
- Center for Vascular Emergencies, Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA
- Department of Emergency Medicine, Harvard Medical School, Boston, MA; and
| | - David-Alexandre Trégouët
- INSERM UMR_S 1219, Bordeaux Population Health Research Center, University of Bordeaux, Bordeaux, France
| | - Nicholas L Smith
- Department of Epidemiology, University of Washington, Seattle, WA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA
- Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle, WA
| |
Collapse
|
47
|
Kim IY, Grodstein F, Kraft P, Curhan GC, Hughes KC, Huang H, Kang JH, Hunter DJ. Interaction between apolipoprotein E genotype and hypertension on cognitive function in older women in the Nurses' Health Study. PLoS One 2019; 14:e0224975. [PMID: 31697783 PMCID: PMC6837309 DOI: 10.1371/journal.pone.0224975] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 10/25/2019] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE To examine the interaction between APOE genotypes and both treated and untreated hypertension on cognitive function in an updated analysis of Nurses' Health Study (NHS) data. DESIGN At baseline (1995-2001) and 3 biennial follow-up assessments over ~6 years, cognitive function was assessed. SETTING AND PARTICIPANTS 8300 NHS participants aged 70+ years underwent a cognitive battery, which comprised 6 tests including the Telephone Interview for Cognitive Status (TICS) and tests of verbal memory, category fluency, and working memory. MEASURES We estimated the mean differences in average cognitive scores across up to 4 assessments using multiple linear regression. We also tested for interaction between APOE e4 allele carrier status and hypertension overall, as well as for apparently untreated and treated hypertension. RESULTS We confirmed that, compared with those with APOE e3/3 genotype, APOE e4 allele carriers scored lower by 0.55 units on the average TICS score (95%CI:-0.67,-0.43). We also observed a significantly worse average TICS score among women with untreated hypertension compared with women without hypertension (difference = -0.23, 95%CI:-0.37,-0.09), while no significant difference was observed for women with treated hypertension. Significant interaction was detected between the APOE e4 allele and untreated hypertension (p-int = 0.02 for the TICS; p-int = 0.045 for global score), but not with treated hypertension. Specifically, compared with normotensive women with the APOE e3/3 genotype, APOE e4 allele carriers with treated hypertension scored lower by 0.50 units (95%CI:-0.69,-0.31); however, the APOE e4 allele carriers with untreated hypertension scored lower by 1.02 units on the TICS score (95%CI:-1.29, -0.76). This interaction of APOE e4 and untreated hypertension was also consistently observed for the global score. CONCLUSIONS Women with hypertension and at least one APOE e4 allele had worse average cognitive function compared with women without hypertension with the e3/3 genotype; this difference was amplified among APOE e4 allele carriers with untreated hypertension.
Collapse
Affiliation(s)
- Iris Y. Kim
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- * E-mail:
| | - Francine Grodstein
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Peter Kraft
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Gary C. Curhan
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Katherine C. Hughes
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Hongyan Huang
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Jae H. Kang
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - David J. Hunter
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
48
|
Chung W, Chen J, Turman C, Lindstrom S, Zhu Z, Loh PR, Kraft P, Liang L. Efficient cross-trait penalized regression increases prediction accuracy in large cohorts using secondary phenotypes. Nat Commun 2019; 10:569. [PMID: 30718517 PMCID: PMC6361917 DOI: 10.1038/s41467-019-08535-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 01/17/2019] [Indexed: 01/15/2023] Open
Abstract
We introduce cross-trait penalized regression (CTPR), a powerful and practical approach for multi-trait polygenic risk prediction in large cohorts. Specifically, we propose a novel cross-trait penalty function with the Lasso and the minimax concave penalty (MCP) to incorporate the shared genetic effects across multiple traits for large-sample GWAS data. Our approach extracts information from the secondary traits that is beneficial for predicting the primary trait based on individual-level genotypes and/or summary statistics. Our novel implementation of a parallel computing algorithm makes it feasible to apply our method to biobank-scale GWAS data. We illustrate our method using large-scale GWAS data (~1M SNPs) from the UK Biobank (N = 456,837). We show that our multi-trait method outperforms the recently proposed multi-trait analysis of GWAS (MTAG) for predictive performance. The prediction accuracy for height by the aid of BMI improves from R2 = 35.8% (MTAG) to 42.5% (MCP + CTPR) or 42.8% (Lasso + CTPR) with UK Biobank data. Information of genetic architectures of complex traits can be leveraged for predicting phenotypes. Here, the authors develop CTPR (Cross-Trait Penalized Regression), a method for multi-trait polygenic risk prediction using individual-level genotypes and/or summary statistics from large cohorts.
Collapse
Affiliation(s)
- Wonil Chung
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Jun Chen
- Division of Biomedical Statistics and Informatics and Center for Individualized Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Constance Turman
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Sara Lindstrom
- Department of Epidemiology, University of Washington, Seattle, WA, 98195, USA
| | - Zhaozhong Zhu
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.,Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Po-Ru Loh
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.,Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Peter Kraft
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Liming Liang
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA. .,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA. .,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.
| |
Collapse
|
49
|
Kim J, Ziyatdinov A, Laville V, Hu FB, Rimm E, Kraft P, Aschard H. Joint Analysis of Multiple Interaction Parameters in Genetic Association Studies. Genetics 2019; 211:483-494. [PMID: 30578273 PMCID: PMC6366922 DOI: 10.1534/genetics.118.301394] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 12/10/2018] [Indexed: 01/24/2023] Open
Abstract
With growing human genetic and epidemiologic data, there has been increased interest for the study of gene-by-environment (G-E) interaction effects. Still, major questions remain on how to test jointly a large number of interactions between multiple SNPs and multiple exposures. In this study, we first compared the relative performance of four fixed-effect joint analysis approaches using simulated data, considering up to 10 exposures and 300 SNPs: (1) omnibus test, (2) multi-exposure and genetic risk score (GRS) test, (3) multi-SNP and environmental risk score (ERS) test, and (4) GRS-ERS test. Our simulations explored both linear and logistic regression while considering three statistics: the Wald test, the Score test, and the likelihood ratio test (LRT). We further applied the approaches to three large sets of human cohort data (n = 37,664), focusing on type 2 diabetes (T2D), obesity, hypertension, and coronary heart disease with smoking, physical activity, diets, and total energy intake. Overall, GRS-based approaches were the most robust, and had the highest power, especially when the G-E interaction effects were correlated with the marginal genetic and environmental effects. We also observed severe miscalibration of joint statistics in logistic models when the number of events per variable was too low when using either the Wald test or LRT test. Finally, our real data application detected nominally significant interaction effects for three outcomes (T2D, obesity, and hypertension), mainly from the GRS-ERS approach. In conclusion, this study provides guidelines for testing multiple interaction parameters in modern human cohorts including extensive genetic and environmental data.
Collapse
Affiliation(s)
- Jihye Kim
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115
| | - Andrey Ziyatdinov
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115
| | - Vincent Laville
- Centre de Bioinformatique, Biostatistique et Biologie Intégrative (C3BI), Institut Pasteur, 75724 Paris, France
| | - Frank B Hu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115
| | - Eric Rimm
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115
| | - Peter Kraft
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115
| | - Hugues Aschard
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115
- Centre de Bioinformatique, Biostatistique et Biologie Intégrative (C3BI), Institut Pasteur, 75724 Paris, France
| |
Collapse
|
50
|
Day F, Karaderi T, Jones MR, Meun C, He C, Drong A, Kraft P, Lin N, Huang H, Broer L, Magi R, Saxena R, Laisk T, Urbanek M, Hayes MG, Thorleifsson G, Fernandez-Tajes J, Mahajan A, Mullin BH, Stuckey BGA, Spector TD, Wilson SG, Goodarzi MO, Davis L, Obermayer-Pietsch B, Uitterlinden AG, Anttila V, Neale BM, Jarvelin MR, Fauser B, Kowalska I, Visser JA, Andersen M, Ong K, Stener-Victorin E, Ehrmann D, Legro RS, Salumets A, McCarthy MI, Morin-Papunen L, Thorsteinsdottir U, Stefansson K, the 23andMe Research Team, Styrkarsdottir U, Perry JRB, Dunaif A, Laven J, Franks S, Lindgren CM, Welt CK. Large-scale genome-wide meta-analysis of polycystic ovary syndrome suggests shared genetic architecture for different diagnosis criteria. PLoS Genet 2018; 14:e1007813. [PMID: 30566500 PMCID: PMC6300389 DOI: 10.1371/journal.pgen.1007813] [Citation(s) in RCA: 321] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 11/06/2018] [Indexed: 11/19/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a disorder characterized by hyperandrogenism, ovulatory dysfunction and polycystic ovarian morphology. Affected women frequently have metabolic disturbances including insulin resistance and dysregulation of glucose homeostasis. PCOS is diagnosed with two different sets of diagnostic criteria, resulting in a phenotypic spectrum of PCOS cases. The genetic similarities between cases diagnosed based on the two criteria have been largely unknown. Previous studies in Chinese and European subjects have identified 16 loci associated with risk of PCOS. We report a fixed-effect, inverse-weighted-variance meta-analysis from 10,074 PCOS cases and 103,164 controls of European ancestry and characterisation of PCOS related traits. We identified 3 novel loci (near PLGRKT, ZBTB16 and MAPRE1), and provide replication of 11 previously reported loci. Only one locus differed significantly in its association by diagnostic criteria; otherwise the genetic architecture was similar between PCOS diagnosed by self-report and PCOS diagnosed by NIH or non-NIH Rotterdam criteria across common variants at 13 loci. Identified variants were associated with hyperandrogenism, gonadotropin regulation and testosterone levels in affected women. Linkage disequilibrium score regression analysis revealed genetic correlations with obesity, fasting insulin, type 2 diabetes, lipid levels and coronary artery disease, indicating shared genetic architecture between metabolic traits and PCOS. Mendelian randomization analyses suggested variants associated with body mass index, fasting insulin, menopause timing, depression and male-pattern balding play a causal role in PCOS. The data thus demonstrate 3 novel loci associated with PCOS and similar genetic architecture for all diagnostic criteria. The data also provide the first genetic evidence for a male phenotype for PCOS and a causal link to depression, a previously hypothesized comorbid disease. Thus, the genetics provide a comprehensive view of PCOS that encompasses multiple diagnostic criteria, gender, reproductive potential and mental health.
Collapse
Affiliation(s)
- Felix Day
- MRC Epidemiology Unit, Cambridge Biomedical Campus, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Tugce Karaderi
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Department of Biological Sciences, Faculty of Arts and Sciences, Eastern Mediterranean University, Famagusta, Cyprus
| | - Michelle R. Jones
- Center for Bioinformatics & Functional Genomics, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Cindy Meun
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Chunyan He
- Department of Internal Medicine, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- University of Kentucky Markey Cancer Center, Lexington, Kentucky, United States of America
| | - Alex Drong
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Peter Kraft
- Departments of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Nan Lin
- Department of Internal Medicine, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- University of Kentucky Markey Cancer Center, Lexington, Kentucky, United States of America
| | - Hongyan Huang
- Departments of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Linda Broer
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Reedik Magi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Richa Saxena
- Broad Institute of Harvard and MIT and Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Triin Laisk
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Margrit Urbanek
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - M. Geoffrey Hayes
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- Department of Anthropology, Northwestern University, Evanston, Illinois, United States of America
| | | | - Juan Fernandez-Tajes
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Anubha Mahajan
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Benjamin H. Mullin
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia
| | - Bronwyn G. A. Stuckey
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia
- Keogh Institute for Medical Research, Nedlands, Western Australia, Australia
| | - Timothy D. Spector
- Department of Twin Research & Genetic Epidemiology, King's College London, London, United Kingdom
| | - Scott G. Wilson
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia
- Department of Twin Research & Genetic Epidemiology, King's College London, London, United Kingdom
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Lea Davis
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Genomics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Barbara Obermayer-Pietsch
- Division of Endocrinology and Diabetology, Department of Internal Medicine Medical University of Graz, Graz, Austria
| | - André G. Uitterlinden
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Verneri Anttila
- Stanley Center for Psychiatric Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Benjamin M. Neale
- Stanley Center for Psychiatric Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Marjo-Riitta Jarvelin
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, United Kingdom
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Unit of Primary Care, Oulu University Hospital, Oulu, Finland
| | - Bart Fauser
- Department of Reproductive Medicine and Gynaecology, University Medical Center, Utrecht, The Netherlands
| | - Irina Kowalska
- Department of Internal Medicine and Metabolic Diseases, Medical University of Białystok, Białystok, Poland
| | - Jenny A. Visser
- Department of Internal Medicine, Section of Endocrinology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marianne Andersen
- Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Ken Ong
- MRC Epidemiology Unit, Cambridge Biomedical Campus, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | | | - David Ehrmann
- Department of Medicine, Section of Adult and Paediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago, Chicago, Illinois, United States of America
| | - Richard S. Legro
- Department of Obstetrics and Gynecology and Public Health Sciences, Penn State University College of Medicine, Hershey, Pennsylvania, United States of America
| | - Andres Salumets
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
- Institute of Bio- and Translational Medicine, University of Tartu, Tartu, Estonia
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mark I. McCarthy
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, United Kingdom
| | - Laure Morin-Papunen
- Department of Obstetrics and Gynecology, University of Oulu and Oulu University Hospital, Medical Research Center, PEDEGO Research Unit, Oulu, Finland
| | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Kari Stefansson
- deCODE genetics/Amgen, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | | | | | - John R. B. Perry
- MRC Epidemiology Unit, Cambridge Biomedical Campus, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Andrea Dunaif
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Joop Laven
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Steve Franks
- Institute of Reproductive & Developmental Biology, Department of Surgery & Cancer, Imperial College London, London, United Kingdom
| | - Cecilia M. Lindgren
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Broad Institute of Harvard and MIT and Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Corrine K. Welt
- Division of Endocrinology, Metabolism and Diabetes, University of Utah, Salt Lake City, Utah, United States of America
- Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| |
Collapse
|