51
|
Fan Z, Li Z, Guo A, Li Y. The association of low serum uric acid with mortality in older people is modified by kidney function: National Health and Nutrition Examination Survey (NHANES) 1999-2018. BMC Nephrol 2024; 25:108. [PMID: 38504168 PMCID: PMC10953214 DOI: 10.1186/s12882-024-03546-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 03/14/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND In older individuals, the role of low serum uric acid (SUA) as risk factor for mortality is debated. We therefore studied whether SUA levels, particularly low SUA concentrations, are associated with all-cause and cardiovascular (CV) mortality in older population, and to clarify potential effect modification of kidney function. METHODS We identified 14,005 older people in National Health and Nutrition Examination Survey (NHANES) data from 1999 to 2018. SUA was measured only at baseline. The relationship between SUA and mortality was assessed using Cox proportional hazards models and restricted cubic spline Cox regression stratified by the estimated glomerular filtration rate (eGFR). RESULTS During mean 8.3 years of follow-up, 4852 all-cause death and 1602 CV death were recorded. A significant U-shaped association was observed between SUA with all-cause mortality, with the lowest risk concentration of 5.5 mg/dL. Comparing to the reference group (5 to 7 mg/dL), the HR of 2 to < 5 mg/dL group was 1.11 (1.03-1.21) and 1.14 (1.00-1.30). This relationship was more pronounced in participants with an eGFR ≥ 60 ml/min/1.73m2 (HR, 1.16; 95%CI, 1.06-1.28). This situation similarly occurred in Urine protein negative group (HR, 1.14; 95%CI, 1.04-1.25). CONCLUSIONS Low SUA concentrations are associated with an increased risk in all-cause and CV mortality among older participants. Extremely low SUA concentrations are especially undesirable, especially in the older adults with normal kidney function.
Collapse
Affiliation(s)
- Zhongcheng Fan
- Department of Osteology, Haikou Municipal People's Hospital and Central South University Xiangya Medical College Affiliated Hospital, Haikou, China
| | - Zhongju Li
- Division of Ultrasonography, Haikou Municipal People's Hospital and Central South University Xiangya Medical College Affiliated Hospital, Haikou, China
| | | | - Yang Li
- Division of Nephrology and Rheumatology, Haikou Municipal People's Hospital and Central South University Xiangya Medical College Affiliated Hospital, 43 Renmin Ave, Haikou, 570208, China.
| |
Collapse
|
52
|
Shi K, Zhu Y, Lv J, Sun D, Pei P, Du H, Chen Y, Yang L, Han B, Stevens R, Chen J, Chen Z, Li L, Yu C. Association of physical activity with risk of chronic kidney disease in China: A population-based cohort study. JOURNAL OF SPORT AND HEALTH SCIENCE 2024; 13:204-211. [PMID: 37532222 PMCID: PMC10980896 DOI: 10.1016/j.jshs.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/29/2023] [Accepted: 07/06/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND Information on the association between physical activity (PA) and the risk of chronic kidney disease (CKD) is limited. We aimed to explore the associations of total, domain-specific, and intensity-specific PA with CKD and its subtypes in China. METHODS The study included 475,376 adults from the China Kadoorie Biobank aged 30-79 years during 2004-2008 at baseline. An interviewer-administered questionnaire was used to collect the information about PA, which was quantified as metabolic equivalent of task hours per day (MET-h/day) and categorized into 4 groups based on quartiles. Cox regression was used to analyze the association between PA and CKD risk. RESULTS During a median follow-up of 12.1 years, 5415 incident CKD cases were documented, including 1159 incident diabetic kidney disease (DKD) cases and 362 incident hypertensive nephropathy (HTN) cases. Total PA was inversely associated with CKD risk, with an adjusted hazard ratio (HR, 95% confidence interval (95%CI)) of 0.83 (0.75-0.92) for incident CKD in the highest quartile of total PA as compared with participants in the lowest quartile. Similar results were observed for risk of DKD and HTN, and the corresponding HRs (95%CIs) were 0.75 (0.58-0.97) for DKD risk and 0.56 (0.37-0.85) for HTN risk. Increased nonoccupational PA, low-intensity PA, and moderate-to-vigorous-intensity PA were significantly associated with a decreased risk of CKD, with HRs (95%CIs) of 0.80 (0.73-0.88), 0.85 (0.77-0.94), and 0.85 (0.76-0.95) in the highest quartile, respectively. CONCLUSION PA, including nonoccupational PA, low-intensity PA, and moderate-to-vigorous-intensity PA, was inversely associated with the risk of CKD, including DKD, HTN, and other CKD, and such associations were dose dependent.
Collapse
Affiliation(s)
- Kexiang Shi
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China
| | - Yunqing Zhu
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China
| | - Jun Lv
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China; Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing 100191, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, China
| | - Dianjianyi Sun
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China; Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing 100191, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, China
| | - Pei Pei
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing 100191, China
| | - Huaidong Du
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK; Medical Research Council Population Health Research Unit at the University of Oxford, Oxford OX3 7LF, UK
| | - Yiping Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK; Medical Research Council Population Health Research Unit at the University of Oxford, Oxford OX3 7LF, UK
| | - Ling Yang
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK; Medical Research Council Population Health Research Unit at the University of Oxford, Oxford OX3 7LF, UK
| | - Bing Han
- NCDs Prevention and Control Department, Henan Center for Disease Control and Prevention, Zhengzhou 450016, China
| | - Rebecca Stevens
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Junshi Chen
- China National Center for Food Safety Risk Assessment, Beijing 100022, China
| | - Zhengming Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Liming Li
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China; Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing 100191, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, China
| | - Canqing Yu
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China; Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing 100191, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, China.
| |
Collapse
|
53
|
Alkaff FF, Kremer D, Thaunat O, Berger SP, van den Born J, Genovese F, Karsdal MA, Bakker SJL, Rasmussen DGK, Tepel M. Urinary Endotrophin and Long-term Outcomes in Kidney Transplant Recipients. Transplant Direct 2024; 10:e1591. [PMID: 39877646 PMCID: PMC11774563 DOI: 10.1097/txd.0000000000001591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 12/23/2023] [Indexed: 01/31/2025] Open
Abstract
Background Kidney fibrosis is a suggested cause of kidney failure and premature mortality. Because collagen type VI is closely linked to kidney fibrosis, we aimed to evaluate whether urinary endotrophin, a collagen type VI fragment, is associated with graft failure and mortality among kidney transplant recipients (KTR). Methods In this prospective cohort study, KTR with a functioning graft ≥1-y posttransplantation were recruited; 24-h urinary endotrophin excretion was measured using an ELISA method. Multivariate Cox regression analyses were performed. Results A total of 621 KTR (mean age 53 y old, 43% female) at a median of 5.2 y posttransplantation were included. Median 24-h urinary endotrophin excretion was 5.6 (3.1-13.6) µg/24h. During a median follow-up of 7.5 y, 87 KTR (14%) developed graft failure and 185 KTR (30%) died; 24-h urinary endotrophin excretion was associated with increased risk of graft failure (hazard ratio [95% confidence interva] per doubling = 1.24 [1.08-1.42]) and all-cause mortality (hazard ratio [95% confidence intervals] per doubling = 1.14 [1.03-1.25]) independent of potential confounders including plasma endotrophin concentration. Twenty-four-hour urinary protein excretion was a significant effect modifier for the association with mortality (Pinteraction = 0.002). Twenty-four-hour urinary endotrophin excretion was only significantly associated with mortality in KTR with low levels of proteinuria. Conclusions Urinary endotrophin is independently associated with an increased risk of graft failure in all KTR and mortality only in KTR with low levels of proteinuria. Further studies with different KTR populations are needed to confirm these findings.
Collapse
Affiliation(s)
- Firas F. Alkaff
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Division of Pharmacology and Therapy, Department of Anatomy, Histology, and Pharmacology, Faculty of Medicine Universitas Airlangga, Surabaya, Indonesia
| | - Daan Kremer
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Olivier Thaunat
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Transplantation, Néphrologie et Immunologie Clinique, Lyon, France
| | - Stefan P. Berger
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jacob van den Born
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | - Stephan J. L. Bakker
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Martin Tepel
- Department of Nephrology, Odense University Hospital, Odense, Denmark
- Institute of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
54
|
Frischmuth T, Tøndel BG, Brækkan SK, Hansen JB, Morelli VM. The Risk of Incident Venous Thromboembolism Attributed to Overweight and Obesity: The Tromsø Study. Thromb Haemost 2024; 124:239-249. [PMID: 37549694 DOI: 10.1055/s-0043-1772212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
BACKGROUND Obesity is a well-established risk factor for venous thromboembolism (VTE). However, data on the proportion of incident VTEs attributed to overweight and obesity in the general population are limited. OBJECTIVE To investigate the population attributable fraction (PAF) of VTE due to overweight and obesity in a population-based cohort with repeated measurements of body mass index (BMI). METHODS Participants from the fourth to seventh surveys of the Tromsø Study (enrolment: 1994-2016) were followed through 2020, and all incident VTEs were recorded. In total, 36,341 unique participants were included, and BMI measurements were updated for those attending more than one survey. BMI was categorized as <25 kg/m2, 25-30 kg/m2 (overweight), and ≥30 kg/m2 (obesity). Time-varying Cox regression models were used to calculate hazard ratios (HRs) with 95% confidence intervals (CIs). The PAF was estimated based on age- and sex-adjusted HRs and the prevalence of BMI categories in VTE cases. RESULTS At baseline, the prevalence of overweight and obesity was 37.9 and 13.8%, respectively. During a median follow-up of 13.9 years, 1,051 VTEs occurred. The age- and sex-adjusted HRs of VTE were 1.40 (95% CI: 1.21-1.61) for overweight and 1.86 (95% CI: 1.58-2.20) for obesity compared with subjects with BMI <25 kg/m2. The PAF of VTE due to overweight and obesity was 24.6% (95% CI: 16.6-32.9), with 12.9% (95% CI: 6.6-19.0) being attributed to overweight and 11.7% (95% CI: 8.5-14.9) to obesity. Similar PAFs were obtained in analyses stratified by sex and VTE subtypes (provoked/unprovoked events, deep vein thrombosis, pulmonary embolism). CONCLUSION Our findings indicate that almost 25% of all VTE events can be attributed to overweight and obesity in a general population from Norway.
Collapse
Affiliation(s)
- Tobias Frischmuth
- Thrombosis Research Group, Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
- Division of Internal Medicine, Thrombosis Research Center, University Hospital of North Norway, Tromsø, Norway
| | - Birgitte G Tøndel
- Thrombosis Research Group, Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Sigrid K Brækkan
- Thrombosis Research Group, Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
- Division of Internal Medicine, Thrombosis Research Center, University Hospital of North Norway, Tromsø, Norway
| | - John-Bjarne Hansen
- Thrombosis Research Group, Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
- Division of Internal Medicine, Thrombosis Research Center, University Hospital of North Norway, Tromsø, Norway
| | - Vânia M Morelli
- Thrombosis Research Group, Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
- Division of Internal Medicine, Thrombosis Research Center, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
55
|
Si K, Cao Z, Liu Q, Yang Y, Dai Q, Yao Y, Qiao Y, Xu C, Wu G. Accelerometer-measured physical activity, sedentary behavior, and risk of incident pelvic organ prolapse: a prospective cohort study in the UK Biobank. Int J Behav Nutr Phys Act 2024; 21:12. [PMID: 38308373 PMCID: PMC10835866 DOI: 10.1186/s12966-024-01559-w] [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: 07/15/2023] [Accepted: 01/03/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Previous studies on physical activity (PA) and pelvic organ prolapse (POP) were largely limited to self-reported PA in athletes, soldiers, and women in postpartum. We aimed to investigate the association of accelerometer-measured PA and sedentary behavior with the risk of POP in middle-aged and elderly women. METHODS In this prospective cohort derived from the UK Biobank, the intensity and duration of PA and sedentary behavior were measured with wrist-worn accelerometers over a 7-day period in 2013-2015 for 47,674 participants (aged 42.8-77.9 years) without pre-existing POP. Participants were followed up until the end of 2022, during which incident POP was ascertained mainly by the electronic health records. Multivariable-adjusted Cox proportional hazards models and restricted cubic splines were used to assess the associations of interest. Isotemporal substitution models were applied to test the effects of substituting a type of activity with equivalent duration of others. RESULTS During a median follow-up of 8.0 years, 779 cases of POP were recorded. The duration of light-intensity PA (LPA) was positively whereas sedentary time was negatively associated with the risk of POP. Every additional 1 h/day of LPA elevated the risk of POP by 18% (95% confidence interval [CI], 10%-26%). In contrast, the risk decreased by 5% (95% CI, 0-8%) per 1 h/day increment in sedentary behavior. No associations were found between moderate-intensity PA (MPA) or vigorous-intensity PA (VPA) and POP, except that women who had a history of hysterectomy were more likely to develop POP when performing more VPA (53% higher risk for every additional 15 min/day). Substituting 1 h/day of LPA with equivalent sedentary time was associated with a 18% (95% CI, 11%-24%) lower risk of POP. The risk can also be reduced by 17% (95% CI, 7%-25%) through substituting 30 min/day of LPA with MPA. CONCLUSIONS More time spent in LPA or less sedentary time was linked to an elevated risk of POP in middle-aged and elderly women, while MPA or VPA was not. Substituting LPA with equivalent duration of sedentary behavior or MPA may lower the risk of POP.
Collapse
Affiliation(s)
- Keyi Si
- Department of Gynecology, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699 West Gaoke Road, Shanghai, 201204, China
- School of Public Health, Hangzhou Normal University, No.2318 Yuhangtang Road, Yuhang District, Hangzhou, 311121, China
| | - Zhi Cao
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Qianqian Liu
- Department of Gynecology, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699 West Gaoke Road, Shanghai, 201204, China
| | - Yingying Yang
- Clinical Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qingqiang Dai
- Department of Gynecology, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699 West Gaoke Road, Shanghai, 201204, China
| | - Yuting Yao
- Department of Gynecology, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699 West Gaoke Road, Shanghai, 201204, China
| | - Yingying Qiao
- Department of Gynecology, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699 West Gaoke Road, Shanghai, 201204, China
| | - Chenjie Xu
- School of Public Health, Hangzhou Normal University, No.2318 Yuhangtang Road, Yuhang District, Hangzhou, 311121, China.
| | - Guizhu Wu
- Department of Gynecology, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699 West Gaoke Road, Shanghai, 201204, China.
| |
Collapse
|
56
|
Chen Z, Liu M, Xu X, He L, Wang P, Cai X, Huang R, Zhang S, Xu X, Lai Y, Huang Y, Li M, Lin Y, Xie P, Liao X, Zhuang X, Guo Y. Serum Klotho Modifies the Associations of 25-Hydroxy Vitamin D With All-Cause and Cardiovascular Mortality. J Clin Endocrinol Metab 2024; 109:581-591. [PMID: 37579499 DOI: 10.1210/clinem/dgad480] [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: 04/01/2023] [Revised: 08/03/2023] [Accepted: 08/10/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND The association between 25-hydroxyvitamin D and mortality remains controversial. Klotho, a biomarker of vitamin D activation and metabolism, may play a key role in this association. However, it is unclear whether the association between vitamin D deficiency and mortality risk is modified by klotho levels. Therefore, this study investigated the joint association of serum 25-hydroxyvitamin D [25(OH)D] and klotho with mortality risk in American community-dwelling adults. METHODS A total of 9870 adults from the National Health and Nutrition Examination Survey (2007-2016) were included in our study. Mortality data were ascertained by linking participants to National Death Index records. Cox proportional hazards models were used to assess the association among serum 25(OH)D, serum klotho, and all-cause and cardiovascular disease (CVD) mortality. RESULTS We found a significant interaction between klotho and serum 25(OH)D in all-cause mortality (P = .028). With klotho > 848.4 pg/mL (risk threshold on mortality), no significant all-cause and CVD mortality risk was observed at any level of serum 25(OH)D. However, with klotho < 848.4 pg/mL, a significant all-cause and CVD mortality risk was observed with serum 25(OH)D < 50 nmol/L [hazards ratio (HR), 1.36; 95% confidence interval (CI), 1.10-1.69; HR, 1.78; 95% CI, 1.16-3.45) and serum 25(OH)D of continuous variable (HR, 0.98; 95% CI, .97-.99; HR, 0.98; 95% CI, .98-.99). In addition, vitamin D metabolism disruption accessed by the combination of decreasing serum 25(OH)D (<50 nmol/L) and klotho (<848.4 pg/mL) was associated with significant all-cause mortality (HR, 1.48; 95% CI, 1.11-1.96) and CVD mortality (HR, 2.36; 95% CI, 1.48-3.75). CONCLUSIONS Vitamin D-associated mortality risk is observed only with concurrently decreasing klotho, indicating that vitamin D metabolism dysfunction increases the risk of mortality. Klotho levels could help predict long-term mortality outcomes and thus may be useful concurrently for guiding vitamin D supplementation therapy decision-making in populations with vitamin D deficiency.
Collapse
Affiliation(s)
- Zhuohui Chen
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, P.R. China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong 510080, P. R. China
| | - Menghui Liu
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, P.R. China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong 510080, P. R. China
| | - Xingfeng Xu
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, P.R. China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong 510080, P. R. China
| | - Lixiang He
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, P.R. China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong 510080, P. R. China
| | - Peng Wang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, P.R. China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong 510080, P. R. China
| | - Xiaojie Cai
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, P.R. China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong 510080, P. R. China
| | - Rihua Huang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, P.R. China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong 510080, P. R. China
| | - Shaozhao Zhang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, P.R. China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong 510080, P. R. China
| | - Xinghao Xu
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, P.R. China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong 510080, P. R. China
| | - Yuhui Lai
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, P.R. China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong 510080, P. R. China
| | - Yiquan Huang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, P.R. China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong 510080, P. R. China
| | - Miaohong Li
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, P.R. China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong 510080, P. R. China
| | - Yifen Lin
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, P.R. China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong 510080, P. R. China
| | - Peihan Xie
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, P.R. China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong 510080, P. R. China
| | - Xinxue Liao
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, P.R. China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong 510080, P. R. China
| | - Xiaodong Zhuang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, P.R. China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong 510080, P. R. China
| | - Yue Guo
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, P.R. China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong 510080, P. R. China
| |
Collapse
|
57
|
Allen NE, Lacey B, Lawlor DA, Pell JP, Gallacher J, Smeeth L, Elliott P, Matthews PM, Lyons RA, Whetton AD, Lucassen A, Hurles ME, Chapman M, Roddam AW, Fitzpatrick NK, Hansell AL, Hardy R, Marioni RE, O’Donnell VB, Williams J, Lindgren CM, Effingham M, Sellors J, Danesh J, Collins R. Prospective study design and data analysis in UK Biobank. Sci Transl Med 2024; 16:eadf4428. [PMID: 38198570 PMCID: PMC11127744 DOI: 10.1126/scitranslmed.adf4428] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/13/2023] [Indexed: 01/12/2024]
Abstract
Population-based prospective studies, such as UK Biobank, are valuable for generating and testing hypotheses about the potential causes of human disease. We describe how UK Biobank's study design, data access policies, and approaches to statistical analysis can help to minimize error and improve the interpretability of research findings, with implications for other population-based prospective studies being established worldwide.
Collapse
Affiliation(s)
- Naomi E Allen
- UK Biobank Ltd, Stockport, UK
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Ben Lacey
- UK Biobank Ltd, Stockport, UK
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Deborah A Lawlor
- Population Health Science, Bristol Medical School University of Bristol, Bristol, UK
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
| | - Jill P Pell
- School of Health and Wellbeing, University of Glasgow, Scotland
| | - John Gallacher
- Department of Psychiatry, University of Oxford, Oxford, UK
- Dementias Platform UK, Department of Psychiatry, University of Oxford, Oxford, UK
| | - Liam Smeeth
- London School of Hygiene and Tropical Medicine, London, UK
| | - Paul Elliott
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- NIHR Health Protection Research Unit in Chemical Radiation Threats and Hazards, Imperial College London, UK
| | - Paul M Matthews
- UK Dementia Research Centre Institute and Department of Brain Sciences, Imperial College London, London, UK
| | - Ronan A Lyons
- Population Data Science, Swansea University Medical School, Swansea, Wales
| | - Anthony D Whetton
- Veterinary Health Innovation Engine, University of Surrey, Guildford, UK
| | - Anneke Lucassen
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Faculty of Medicine, Southampton University, Southampton, UK
| | - Matthew E Hurles
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | | | | | | | - Anna L Hansell
- Centre for Environmental Health and Sustainability, University of Leicester, Leicester, UK
| | - Rebecca Hardy
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland
| | | | - Julie Williams
- UK Dementia Research Institute, Cardiff University, Cardiff, Wales
| | - Cecilia M Lindgren
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | | | | | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
| | - Rory Collins
- UK Biobank Ltd, Stockport, UK
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| |
Collapse
|
58
|
Zhang Y, Sun Y, Yu Q, Song S, Brenna JT, Shen Y, Ye K. Higher ratio of plasma omega-6/omega-3 fatty acids is associated with greater risk of all-cause, cancer, and cardiovascular mortality: a population-based cohort study in UK Biobank. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.01.16.23284631. [PMID: 36711941 PMCID: PMC9882493 DOI: 10.1101/2023.01.16.23284631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Background Circulating omega-3 and omega-6 polyunsaturated fatty acids (PUFAs) have been associated with various chronic diseases and mortality, but results are conflicting. Few studies examined the role of omega-6/omega-3 ratio in mortality. Methods We investigated plasma omega-3 and omega-6 PUFAs and their ratio in relation to all-cause and cause-specific mortality in a large prospective cohort, the UK Biobank. Of 85,425 participants who had complete information on circulating PUFAs, 6,461 died during follow-up, including 2,794 from cancer and 1,668 from cardiovascular disease (CVD). Associations were estimated by multivariable Cox proportional hazards regression with adjustment for relevant risk factors. Results Risk for all three mortality outcomes increased as the ratio of omega-6/omega-3 PUFAs increased (all Ptrend < 0.05). Comparing the highest to the lowest quintiles, individuals had 26% (95% CI, 15-38%) higher total mortality, 14% (95% CI, 0-31%) higher cancer mortality, and 31% (95% CI, 10-55%) higher CVD mortality. Moreover, omega-3 and omega-6 PUFAs in plasma were all inversely associated with all-cause, cancer, and CVD mortality, with omega-3 showing stronger effects. Conclusions Using a population-based cohort in UK Biobank, our study revealed a strong association between the ratio of circulating omega-6/omega-3 PUFAs and the risk of all-cause, cancer, and CVD mortality.
Collapse
Affiliation(s)
- Yuchen Zhang
- Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens, Georgia, US
| | - Yitang Sun
- Department of Genetics, University of Georgia, Athens, Georgia, US
| | - Qi Yu
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, US
| | - Suhang Song
- Department of Health Policy and Management, College of Public Health, University of Georgia, Athens, Georgia, US
| | - J. Thomas Brenna
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, US
- Dell Pediatric Research Institute and the Depts of Pediatrics, of Nutrition, and of Chemistry, University of Texas at Austin, Austin, TX, US
| | - Ye Shen
- Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens, Georgia, US
| | - Kaixiong Ye
- Department of Genetics, University of Georgia, Athens, Georgia, US
- Institute of Bioinformatics, University of Georgia, Athens, Georgia, US
| |
Collapse
|
59
|
Hansen ES, Edvardsen MS, Aukrust P, Ueland T, Hansen JB, Brækkan SK, Morelli VM. Galectin-3-binding protein and future venous thromboembolism. Res Pract Thromb Haemost 2024; 8:102311. [PMID: 38327613 PMCID: PMC10847771 DOI: 10.1016/j.rpth.2023.102311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/18/2023] [Accepted: 12/23/2023] [Indexed: 02/09/2024] Open
Affiliation(s)
- Ellen-Sofie Hansen
- Thrombosis Research Group, Department of Clinical Medicine, UiT – The Arctic University of Norway, Tromsø, Norway
| | - Magnus S. Edvardsen
- Thrombosis Research Group, Department of Clinical Medicine, UiT – The Arctic University of Norway, Tromsø, Norway
| | - Pål Aukrust
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Thor Ueland
- Thrombosis Research Group, Department of Clinical Medicine, UiT – The Arctic University of Norway, Tromsø, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo, Norway
| | - John-Bjarne Hansen
- Thrombosis Research Group, Department of Clinical Medicine, UiT – The Arctic University of Norway, Tromsø, Norway
- Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Sigrid K. Brækkan
- Thrombosis Research Group, Department of Clinical Medicine, UiT – The Arctic University of Norway, Tromsø, Norway
- Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Vânia M. Morelli
- Thrombosis Research Group, Department of Clinical Medicine, UiT – The Arctic University of Norway, Tromsø, Norway
- Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
60
|
Trichia E, Malden DE, Jin D, Wright N, Taylor H, Karpe F, Sherliker P, Murgia F, Hopewell JC, Lacey B, Emberson J, Bennett D, Lewington S. Independent relevance of adiposity measures to coronary heart disease risk among 0.5 million adults in UK Biobank. Int J Epidemiol 2023; 52:1836-1844. [PMID: 37935988 PMCID: PMC10749766 DOI: 10.1093/ije/dyad143] [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: 10/26/2022] [Accepted: 11/02/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Evidence on body fat distribution shows opposing effects of waist circumference (WC) and hip circumference (HC) for coronary heart disease (CHD). We aimed to investigate the causality and the shape of such associations. METHODS UK Biobank is a prospective cohort study of 0.5 million adults aged 40-69 years recruited between 2006 and 2010. Adjusted hazard ratios (HRs) for the associations of measured and genetically predicted body mass index (BMI), WC, HC and waist-to-hip ratio with incident CHD were obtained from Cox models. Mendelian randomization (MR) was used to assess causality. The analysis included 456 495 participants (26 225 first-ever CHD events) without prior CHD. RESULTS All measures of adiposity demonstrated strong, positive and approximately log-linear associations with CHD risk over a median follow-up of 12.7 years. For HC, however, the association became inverse given the BMI and WC (HR per usual SD 0.95, 95% CI 0.93-0.97). Associations for BMI and WC remained independently positive after adjustment for other adiposity measures and were similar (1.14, 1.13-1.16 and 1.18, 1.15-1.20, respectively), with WC displaying stronger associations among women. Blood pressure, plasma lipids and dysglycaemia accounted for much of the observed excess risk. MR results were generally consistent with the observational, implying causality. CONCLUSIONS Body fat distribution measures displayed similar associations with CHD risk as BMI except for HC, which was inversely associated with CHD risk (given WC and BMI). These findings suggest that different measures of body fat distribution likely influence CHD risk through both overlapping and independent mechanisms.
Collapse
Affiliation(s)
- Eirini Trichia
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Debbie E Malden
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Danyao Jin
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Neil Wright
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Hannah Taylor
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals (OUH) Foundation Trust, Oxford, UK
| | - Fredrik Karpe
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals (OUH) Foundation Trust, Oxford, UK
- The Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Oxford, UK
| | - Paul Sherliker
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Federico Murgia
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Jemma C Hopewell
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Ben Lacey
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Jonathan Emberson
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Derrick Bennett
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals (OUH) Foundation Trust, Oxford, UK
| | - Sarah Lewington
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Health Data Research UK Oxford, University of Oxford, Oxford, UK
| |
Collapse
|
61
|
Lovegrove CE, Bešević J, Wiberg A, Lacey B, Littlejohns TJ, Allen NE, Goldsworthy M, Kim J, Hannan FM, Curhan GC, Turney BW, McCarthy MI, Mahajan A, Thakker RV, Holmes MV, Furniss D, Howles SA. Central Adiposity Increases Risk of Kidney Stone Disease through Effects on Serum Calcium Concentrations. J Am Soc Nephrol 2023; 34:1991-2011. [PMID: 37787550 PMCID: PMC10703081 DOI: 10.1681/asn.0000000000000238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/25/2023] [Accepted: 09/14/2023] [Indexed: 10/04/2023] Open
Abstract
SIGNIFICANCE STATEMENT Kidney stone disease is a common disorder with poorly understood pathophysiology. Observational and genetic studies indicate that adiposity is associated with an increased risk of kidney stone disease. However, the relative contribution of general and central adipose depots and the mechanisms by which effects of adiposity on kidney stone disease are mediated have not been defined. Using conventional and genetic epidemiological techniques, we demonstrate that general and central adiposity are independently associated with kidney stone disease. In addition, one mechanism by which central adiposity increases risk of kidney stone disease is by increasing serum calcium concentration. Therapies targeting adipose depots may affect calcium homeostasis and help to prevent kidney stone disease. BACKGROUND Kidney stone disease affects approximately 10% of individuals in their lifetime and is frequently recurrent. The disease is linked to obesity, but the mechanisms mediating this association are uncertain. METHODS Associations of adiposity and incident kidney stone disease were assessed in the UK Biobank over a mean of 11.6 years/person. Genome-wide association studies and Mendelian randomization (MR) analyses were undertaken in the UK Biobank, FinnGen, and in meta-analyzed cohorts to identify factors that affect kidney stone disease risk. RESULTS Observational analyses on UK Biobank data demonstrated that increasing central and general adiposity is independently associated with incident kidney stone formation. Multivariable MR, using meta-analyzed UK Biobank and FinnGen data, established that risk of kidney stone disease increases by approximately 21% per one standard deviation increase in body mass index (BMI, a marker of general adiposity) independent of waist-to-hip ratio (WHR, a marker of central adiposity) and approximately 24% per one standard deviation increase of WHR independent of BMI. Genetic analyses indicate that higher WHR, but not higher BMI, increases risk of kidney stone disease by elevating adjusted serum calcium concentrations (β=0.12 mmol/L); WHR mediates 12%-15% of its effect on kidney stone risk in this way. CONCLUSIONS Our study indicates that visceral adipose depots elevate serum calcium concentrations, resulting in increased risk of kidney stone disease. These findings highlight the importance of weight loss in individuals with recurrent kidney stones and suggest that therapies targeting adipose depots may affect calcium homeostasis and contribute to prevention of kidney stone disease.
Collapse
Affiliation(s)
| | - Jelena Bešević
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Akira Wiberg
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Ben Lacey
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Thomas J. Littlejohns
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Naomi E. Allen
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Michelle Goldsworthy
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Jihye Kim
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Fadil M. Hannan
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Gary C. Curhan
- Channing Division of Network Medicine and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ben W. Turney
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Mark I. McCarthy
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Genentech, South San Francisco, Califirnia
| | - Anubha Mahajan
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Genentech, South San Francisco, Califirnia
| | - Rajesh V. Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Michael V. Holmes
- Medical Research Council, Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Dominic Furniss
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Sarah A. Howles
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
62
|
Orliacq J, Pérez-Cornago A, Parry SA, Kelly RK, Koutoukidis DA, Carter JL. Associations between types and sources of dietary carbohydrates and liver fat: a UK Biobank study. BMC Med 2023; 21:444. [PMID: 37968623 PMCID: PMC10652437 DOI: 10.1186/s12916-023-03135-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/26/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND AND AIMS Excess energy intake can lead to metabolic dysfunction-associated steatotic liver disease (MASLD), but the relationship between dietary carbohydrate intake and liver fat content remains unclear. This study aimed to examine the associations between types and sources of dietary carbohydrates and liver fat content. METHODS UK Biobank participants with no pre-existing diabetes, liver disease or cardiovascular disease reported dietary intake of types and sources of carbohydrates (total carbohydrates, free sugars, non-free sugars, starch from whole grains, starch from refined grains, and fibre) on at least two 24-h dietary assessments. In cross-sectional analyses, (n = 22,973), odds ratios (OR) of high liver fat content (defined as a score of ≥ 36 in the hepatic steatosis index) by quintiles of carbohydrate intakes were estimated using multivariable logistic regression models. In prospective analyses, a second sample (n = 9268) had liver proton density fat fraction (PDFF) measured by magnetic resonance imaging (2014-2020). Multivariable linear regression models estimated geometric means of PDFF (%) by quintiles of carbohydrate intakes. Models were adjusted for demographic and lifestyle confounders, including total energy intake. RESULTS In the cross-sectional analyses, 6894 cases of high liver fat content were identified. Inverse associations between intakes of fibre (OR of highest vs. lowest quintile 0.46 [95% CI: 0.41-0.52]), non-free sugars (0.63 [0.57-0.70]) and starch from whole grains (0.52 [0.47-0.57]) with liver fat were observed. There were positive associations between starch from refined grains and liver fat (1.33 [1.21-1.46]), but no association with free sugars (p=0.61). In prospective analyses, inverse associations with PDFF (%) were observed for intakes of fibre (- 0.48 geometric mean difference between highest and lowest quintile of intake [- 0.60 to - 0.35]), non-free sugars (- 0.37 [- 0.49 to - 0.25]) and starch from whole grains (- 0.31 [- 0.42 to - 0.19]). Free sugars, but not starch from refined grains, were positively associated with PDFF (0.17 [0.05 to 0.28]). CONCLUSION This study suggests that different carbohydrate types and sources have varying associations with liver fat, which may be important for MASLD prevention. Non-free sugars, fibre, and starch from whole grains could be protective, while associations with free sugars and starch from refined grains are less clear.
Collapse
Affiliation(s)
- Josefina Orliacq
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Cancer Epidemiology Unit (CEU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Aurora Pérez-Cornago
- Cancer Epidemiology Unit (CEU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Siôn A Parry
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Aston Medical School, Aston University, Birmingham, B4 7ET, UK
| | - Rebecca K Kelly
- Cancer Epidemiology Unit (CEU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- School of Medicine, College of Health and Medicine, The University of Tasmania, Hobart, Australia
| | | | - Jennifer L Carter
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| |
Collapse
|
63
|
Liao J, Xiao F, Yang L, Wei Y, Song C, Li J, Yu S, Lu Y, Zhang J, Dai L, Liang W, Li T, Xiong Z, Wu Y, Jardine MJ, Carrero JJ, Shan Y, Huang X. Cystatin C-based estimated glomerular filtration rate and risk of stroke in the general population: a prospective cohort study. Clin Kidney J 2023; 16:2059-2071. [PMID: 37915909 PMCID: PMC10616444 DOI: 10.1093/ckj/sfad188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Indexed: 11/03/2023] Open
Abstract
Background Previous results on the association between the estimated glomerular filtration rate (eGFR) and stroke are mixed. Most studies derived the eGFR from serum creatinine, which is affected by non-kidney determinants and thus has possibly biased the association with stroke risk. Methods In this cohort study, we included 429 566 UK Biobank participants (94.5% white, 54% women, age 56 ± 8 years) free of stroke at enrollment. The eGFRcys and eGFRcr were calculated with serum cystatin C and creatinine, respectively. Outcomes of interest were risk of total stroke and subtypes. We investigated the linear and nonlinear associations using Cox proportional hazards models and restricted cubic splines, corrected for regression dilution bias. Results During an average follow-up of 10.11 years, 4427 incident strokes occurred, among which 3447 were ischemic and 1163 were hemorrhagic. After adjustment for confounders, the regression dilution-corrected hazard ratios (95% confidence intervals) for every 10 mL/min/1.73 m2 decrement in eGFRcys were 1.10 (1.05-1.14) for total stroke and 1.11 (1.08-1.15) for ischemic stroke. A similar pattern was observed with eGFRcr, although the association was weaker. When either type of eGFR was below 75 mL/min/1.73 m2, the risks of total and ischemic stroke increased exponentially as eGFR decreased. A U-shaped relationship was witnessed if eGFRcr was used instead. There was a null association between eGFR and hemorrhagic stroke. Conclusions The risks of total stroke and ischemic stroke increased exponentially when the eGFRcys fell below 75 mL/min/1.73 m2.
Collapse
Affiliation(s)
- Jinlan Liao
- Renal Division, Peking University Shenzhen Hospital, Peking University, Shenzhen, Guangdong Province, China
| | - Fei Xiao
- Renal Division, Peking University Shenzhen Hospital, Peking University, Shenzhen, Guangdong Province, China
- Shantou University Medical College, Shantou, Guangdong Province, China
| | - Liuqiao Yang
- BGI-Shenzhen, Shenzhen, Guangdong Province, China
| | - Yanling Wei
- Clinical Research Academy, Peking University Shenzhen Hospital, Peking University, Shenzhen, Guangdong Province, China
| | - Congying Song
- Clinical Research Academy, Peking University Shenzhen Hospital, Peking University, Shenzhen, Guangdong Province, China
| | - Jing Li
- Renal Division, Peking University Shenzhen Hospital, Peking University, Shenzhen, Guangdong Province, China
| | - Sike Yu
- BGI-Shenzhen, Shenzhen, Guangdong Province, China
| | - Yueqi Lu
- BGI-Shenzhen, Shenzhen, Guangdong Province, China
| | - Jingwen Zhang
- Renal Division, Peking University Shenzhen Hospital, Peking University, Shenzhen, Guangdong Province, China
| | - Liang Dai
- Clinical Research Academy, Peking University Shenzhen Hospital, Peking University, Shenzhen, Guangdong Province, China
| | - Wei Liang
- Renal Division, Peking University Shenzhen Hospital, Peking University, Shenzhen, Guangdong Province, China
| | - Tao Li
- BGI-Shenzhen, Shenzhen, Guangdong Province, China
| | - Zuying Xiong
- Renal Division, Peking University Shenzhen Hospital, Peking University, Shenzhen, Guangdong Province, China
| | - Yangfeng Wu
- Peking University Clinical Research Institute, Peking University, Beijing, China
| | - Meg J Jardine
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
- Concord Repatriation General Hospital, Sydney, New South Wales, Australia
- Department of Medicine, Stanford Centre for Clinical Research, Stanford University School of Medicine, Stanford, CA, USA
| | - Juan Jesus Carrero
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
- Division of Nephrology, Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm 182 88, Sweden
| | - Ying Shan
- BGI-Shenzhen, Shenzhen, Guangdong Province, China
- Clinical Research Academy, Peking University Shenzhen Hospital, Peking University, Shenzhen, Guangdong Province, China
| | - Xiaoyan Huang
- Renal Division, Peking University Shenzhen Hospital, Peking University, Shenzhen, Guangdong Province, China
- Clinical Research Academy, Peking University Shenzhen Hospital, Peking University, Shenzhen, Guangdong Province, China
| |
Collapse
|
64
|
Batty GD, Kivimäki M, Frank P, Gale CR, Wright L. Systemic inflammation and subsequent risk of amyotrophic lateral sclerosis: Prospective cohort study. Brain Behav Immun 2023; 114:46-51. [PMID: 37543248 PMCID: PMC10937260 DOI: 10.1016/j.bbi.2023.07.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/14/2023] [Accepted: 07/30/2023] [Indexed: 08/07/2023] Open
Abstract
BACKGROUND While systemic inflammation has been implicated in the etiology of selected neurodegenerative disorders, its role in the development of amyotrophic lateral sclerosis (ALS), a condition with high case-fatality, is untested. Accordingly, we quantified the relationship of C-reactive protein (CRP), an acute-phase reactant and marker of systemic inflammation, with subsequent ALS occurrence. METHODS We used data from UK Biobank, a prospective cohort study of 502,649 participants who were aged 37 to 73 years when examined at research centers between 2006 and 2010. Venous blood was collected at baseline in the full cohort and assayed for CRP, and repeat measurement was made 3-7 years later in a representative subgroup (N = 14,514) enabling correction for regression dilution. ALS was ascertained via national hospitalization and mortality registries until 2021. We computed multivariable hazard ratios with accompanying 95% confidence intervals for log-transformed CRP expressed as standard deviation and tertiles. RESULTS In an analytical sample of 400,884 initially ALS-free individuals (218,203 women), a mean follow-up of 12 years gave rise to 231 hospitalizations and 223 deaths ascribed to ALS. After adjustment for covariates which included health behaviors, comorbidity, and socio-economic status, a one standard deviation higher log-CRP was associated with elevated rates of both ALS mortality (hazard ratios; 95% confidence intervals: 1.32; 1.13, 1.53) and hospitalizations (1.20; 1.00, 1.39). There was evidence of dose-response effects across tertiles of CRP for both outcomes (p for trend ≤ 0.05). Correction for regression dilution led to a strengthening of the relationship with CRP for both mortality (1.62; 1.27, 2.08) and hospitalizations (1.37; 1.05, 1.76). CONCLUSIONS Higher levels of CRP, a blood-based biomarker widely captured in clinical practice, is associated with moderately increased future risk of amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- G David Batty
- Department of Epidemiology and Public Health, University College London, UK.
| | - Mika Kivimäki
- UCL Brain Sciences, University College London, UK; Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Philipp Frank
- Department of Epidemiology and Public Health, University College London, UK.
| | - Catharine R Gale
- MRC Lifecourse Epidemiology Unit, University of Southampton, UK; Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, UK.
| | - Liam Wright
- Centre for Longitudinal Studies, University College London, UK.
| |
Collapse
|
65
|
Johansen MØ, Afzal S, Vedel-Krogh S, Nielsen SF, Smith GD, Nordestgaard BG. From plasma triglycerides to triglyceride metabolism: effects on mortality in the Copenhagen General Population Study. Eur Heart J 2023; 44:4174-4182. [PMID: 37575001 DOI: 10.1093/eurheartj/ehad330] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 03/06/2023] [Accepted: 05/15/2023] [Indexed: 08/15/2023] Open
Abstract
AIMS It is unclear whether higher triglyceride metabolism per se contributes to mortality separate from elevated triglyceride-rich lipoproteins and body mass index. This study tested the hypotheses that higher triglyceride metabolism, measured as higher plasma glycerol and β-hydroxybutyrate, is associated with increased all-cause, cardiovascular, cancer, and other mortality. METHODS AND RESULTS This study included 30 000 individuals nested within 109 751 individuals from the Copenhagen General Population Study. During a median follow-up of 10.7 years, 9897 individuals died (2204 from cardiovascular, 3366 from cancer, and 2745 from other causes), while none were lost to follow-up. In individuals with glycerol >80 µmol/L (highest fourth) vs. individuals with glycerol <52 µmol/L (lowest fourth), the multivariable adjusted hazard ratio for all-cause mortality was 1.31 (95% confidence interval 1.22-1.40). In individuals with β-hydroxybutyrate >154 µmol/L (highest fourth) vs. individuals with β-hydroxybutyrate <91 µmol/L (lowest fourth), the multivariable adjusted hazard ratio for all-cause mortality was 1.18 (1.11-1.26). Corresponding values for higher plasma glycerol and β-hydroxybutyrate were 1.37 (1.18-1.59) and 1.18 (1.03-1.35) for cardiovascular mortality, 1.24 (1.11-1.39) and 1.16 (1.05-1.29) for cancer mortality, and 1.45 (1.28-1.66) and 1.23 (1.09-1.39) for other mortality, respectively. Results were robust to exclusion of first years of follow-up, to stratification for covariates including plasma triglycerides and body mass index, and to further adjustments. CONCLUSION This study observed an increased risk of all-cause, cardiovascular, cancer, and other mortality with higher triglyceride metabolism. This was not explained by higher plasma triglycerides and body mass index. The hypothesis studied in the present paper should be further validated by isotope flux studies.
Collapse
Affiliation(s)
- Mia Ø Johansen
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Elevator 7, 4th Floor, N5, Herlev DK-2730, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Herlev DK-2730, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen N DK-2200, Denmark
| | - Shoaib Afzal
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Elevator 7, 4th Floor, N5, Herlev DK-2730, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Herlev DK-2730, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen N DK-2200, Denmark
| | - Signe Vedel-Krogh
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Elevator 7, 4th Floor, N5, Herlev DK-2730, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Herlev DK-2730, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen N DK-2200, Denmark
| | - Sune F Nielsen
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Elevator 7, 4th Floor, N5, Herlev DK-2730, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Herlev DK-2730, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen N DK-2200, Denmark
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Elevator 7, 4th Floor, N5, Herlev DK-2730, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Herlev DK-2730, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen N DK-2200, Denmark
| |
Collapse
|
66
|
Hansen ES, Edvardsen MS, Aukrust P, Ueland T, Hansen JB, Brækkan SK, Morelli VM. Combined effect of high factor VIII levels and high mean platelet volume on the risk of future incident venous thromboembolism. J Thromb Haemost 2023; 21:2844-2853. [PMID: 37393000 DOI: 10.1016/j.jtha.2023.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/10/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023]
Abstract
BACKGROUND High factor VIII (FVIII) levels and large platelets, as reflected by a high mean platelet volume (MPV), are separately associated with increased risk of venous thromboembolism (VTE). Whether the combination of high FVIII levels and large platelets has a supra-additive effect on VTE risk is unknown. OBJECTIVES We aimed to investigate the joint effect of high FVIII levels and large platelets, as reflected by high MPV, on the risk of future incident VTE. METHODS A population-based nested case-control study with 365 incident VTE cases and 710 controls was derived from the Tromsø study. FVIII antigen levels and MPV were measured in blood samples drawn at baseline. Odds ratios with 95% CIs were estimated across FVIII tertiles (<85%, 85%-108%, and ≥108%) and within predefined MPV strata (<8.5, 8.5-9.5, and ≥9.5 fL). RESULTS VTE risk increased linearly across FVIII tertiles (Ptrend < .001) in models adjusted for age, sex, body mass index, and C-reactive protein. In the combined analysis, participants with FVIII levels in the highest tertile and an MPV of ≥9.5 fL (ie, joint exposure) had an odds ratio for VTE of 2.71 (95% CI, 1.44-5.11) compared with those with FVIII levels in the lowest tertile and an MPV of <8.5 fL (reference). In the joint exposure group, 52% (95% CI, 17%-88%) of VTEs were attributable to the biological interaction between FVIII and MPV. CONCLUSION Our results suggest that large platelets, as reflected by high MPV, might play a role in the mechanism by which high FVIII level increases the risk of incident VTE.
Collapse
Affiliation(s)
- Ellen-Sofie Hansen
- Thrombosis Research Group, Department of Clinical Medicine, UiT-the Arctic University of Norway, Tromsø, Norway.
| | - Magnus S Edvardsen
- Thrombosis Research Group, Department of Clinical Medicine, UiT-the Arctic University of Norway, Tromsø, Norway
| | - Pål Aukrust
- Faculty of Medicine, University of Oslo, Oslo, Norway; Research Institute of Internal Medicine, Oslo, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Thor Ueland
- Thrombosis Research Group, Department of Clinical Medicine, UiT-the Arctic University of Norway, Tromsø, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway; Research Institute of Internal Medicine, Oslo, Norway
| | - John-Bjarne Hansen
- Thrombosis Research Group, Department of Clinical Medicine, UiT-the Arctic University of Norway, Tromsø, Norway; Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Sigrid K Brækkan
- Thrombosis Research Group, Department of Clinical Medicine, UiT-the Arctic University of Norway, Tromsø, Norway; Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Vânia M Morelli
- Thrombosis Research Group, Department of Clinical Medicine, UiT-the Arctic University of Norway, Tromsø, Norway; Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
67
|
Grover SP, Brækkan SK, Mackman N, Hansen JB. "High plasma levels of C1-inhibitor are associated with lower risk of future venous thromboembolism": reply. J Thromb Haemost 2023; 21:2993-2995. [PMID: 37739598 DOI: 10.1016/j.jtha.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 09/24/2023]
Affiliation(s)
- Steven P Grover
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, North Carolina. https://twitter.com/StevenPGrover
| | - Sigrid K Brækkan
- Thrombosis Research Group, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway; Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway.
| | - Nigel Mackman
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, North Carolina. https://twitter.com/NMackman
| | - John-Bjarne Hansen
- Thrombosis Research Group, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway; Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway. https://twitter.com/TREC_UiT
| |
Collapse
|
68
|
Wang S, Rao Z, Cao R, Blaes AH, Coresh J, Joshu CE, Lehallier B, Lutsey PL, Pankow JS, Sedaghat S, Tang W, Thyagarajan B, Walker KA, Ganz P, Platz EA, Guan W, Prizment A. Development and Characterization of Proteomic Aging Clocks in the Atherosclerosis Risk in Communities (ARIC) Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.06.23295174. [PMID: 37732184 PMCID: PMC10508816 DOI: 10.1101/2023.09.06.23295174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Biological age may be estimated by proteomic aging clocks (PACs). Previous published PACs were constructed either in smaller studies or mainly in White individuals, and they used proteomic measures from only one-time point. In the Atherosclerosis Risk in Communities (ARIC) study of about 12,000 persons followed for 30 years (around 75% White, 25% Black), we created de novo PACs and compared their performance to published PACs at two different time points. We measured 4,712 plasma proteins by SomaScan in 11,761 midlife participants, aged 46-70 years (1990-92), and 5,183 late-life pariticpants, aged 66-90 years (2011-13). All proteins were log2-transformed to correct for skewness. We created de novo PACs by training them against chronological age using elastic net regression in two-thirds of healthy participants in midlife and late life and compared their performance to three published PACs. We estimated age acceleration (by regressing each PAC on chronological age) and its change from midlife to late life. We examined their associations with mortality from all-cause, cardiovascular disease (CVD), cancer, and lower respiratory disease (LRD) using Cox proportional hazards regression in all remaining participants irrespective of health. The model was adjusted for chronological age, smoking, body mass index (BMI), and other confounders. The ARIC PACs had a slightly stronger correlation with chronological age than published PACs in healthy participants at each time point. Associations with mortality were similar for the ARIC and published PACs. For late-life and midlife age acceleration for the ARIC PACs, respectively, hazard ratios (HRs) per one standard deviation were 1.65 and 1.38 (both p<0.001) for all-cause mortality, 1.37 and 1.20 (both p<0.001) for CVD mortality, 1.21 (p=0.03) and 1.04 (p=0.19) for cancer mortality, and 1.46 and 1.68 (both p<0.001) for LRD mortality. For the change in age acceleration, HRs for all-cause, CVD, and LRD mortality were comparable to those observed for late-life age acceleration. The association between the change in age acceleration and cancer mortality was insignificant. In this prospective study, the ARIC and published PACs were similarly associated with an increased risk of mortality and advanced testing in relation to various age-related conditions in future studies is suggested.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN
| | - Zexi Rao
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Rui Cao
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Anne H. Blaes
- Division of Hematology, Oncology and Transplantation, Medical School, University of Minnesota, Minneapolis, MN
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Corinne E. Joshu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Benoit Lehallier
- Alkahest Inc, San Carlos, CA, United States, Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA
| | - Pamela L. Lutsey
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Sanaz Sedaghat
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, Intramural Research Program, National Institute on Aging, Baltimore, MD
| | - Peter Ganz
- Division of Cardiology, Zuckerberg San Francisco General Hospital and Department of Medicine, University of California, San Francisco, CA
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Weihua Guan
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Anna Prizment
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN
| |
Collapse
|
69
|
Cao Z, Min J, Tan Q, Si K, Yang H, Xu C. Circulating insulin-like growth factor-1 and brain health: Evidence from 369,711 participants in the UK Biobank. Alzheimers Res Ther 2023; 15:140. [PMID: 37608387 PMCID: PMC10463341 DOI: 10.1186/s13195-023-01288-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 08/13/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND The effects of insulin-like growth factor-1 (IGF-1) deficiency on cognitive decline have been consistently reported in animal studies, but the relationship between IGF-1 and human brain health remains controversial. Our study aimed to investigate the associations of serum IGF-1 concentrations with some brain-related disorders and neuroimaging features. METHODS This prospective study included 369,711 participants (55.8 ± 8.1 years) from the UK biobank who had serum IGF-1 measured and were free from brain-related disorders of interest - dementia, stroke, and Parkinson's disease (PD) - at enrollment (2006-2010). Restricted cubic splines and Cox proportional hazards models were used to detect the associations between IGF-1 concentrations and brain-related diseases. In addition, general linear regressions were applied to explore the relationship between IGF-1 concentrations and neuroimaging features (volumes of white matter, grey matter, and hippocampus and white matter hyperintensity) among a sub-sample of 36,458 participants with magnetic resonance imaging data collected since 2014. RESULTS During a median follow-up of 12.6 years, a total of 4,857 dementia, 6,240 stroke, and 2,116 PD cases were documented. The dose-response analyses yielded U-shaped relationships between IGF-1 concentrations and risks of dementia and stroke (P < 0.001 for non-linearity), with the lowest risks at 18 nmol/L and 26 nmol/L, respectively. A positive linear relationship was observed between IGF-1 concentrations and risk of PD (P = 0.163 for non-linearity). Moreover, neuroimaging analyses showed that higher IGF-1 concentrations were associated with greater volumes of white matter (β = 2.98 × 10-4, P < 0.001) and hippocampus (β = 3.37 × 10-4, P = 0.002) and smaller white matter hyperintensity (β = -3.12 × 10-3, P < 0.001). CONCLUSIONS Apart from the diverse associations with neuroimaging features, both low and high IGF-1 concentrations are associated with increased risks of dementia and stroke and higher IGF-1 concentrations are linked to a higher risk of PD, highlighting the potential of IGF-1 as a biomarker for risk stratification of brain health.
Collapse
Affiliation(s)
- Zhi Cao
- School of Public Health, Hangzhou Normal University, NO.2318, Yuhangtang Road, Yuhang District, Hangzhou, 311121, China
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiahao Min
- School of Public Health, Hangzhou Normal University, NO.2318, Yuhangtang Road, Yuhang District, Hangzhou, 311121, China
| | - Qilong Tan
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
- School of Public Health, Harbin Medical University, Harbin, China
| | - Keyi Si
- Department of Health Statistics, Naval Medical University, Shanghai, China
| | - Hongxi Yang
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Chenjie Xu
- School of Public Health, Hangzhou Normal University, NO.2318, Yuhangtang Road, Yuhang District, Hangzhou, 311121, China.
| |
Collapse
|
70
|
Jin D, Trichia E, Islam N, Lewington S, Lacey B. Associations of circulating fatty acids with incident coronary heart disease: a prospective study of 89,242 individuals in UK Biobank. BMC Cardiovasc Disord 2023; 23:365. [PMID: 37480048 PMCID: PMC10362581 DOI: 10.1186/s12872-023-03394-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/12/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND The role of fatty acids in coronary heart disease (CHD) remains uncertain. There is little evidence from large-scale epidemiological studies on the relevance of circulating fatty acids levels to CHD risk. This study aims to examine the independent associations of the major circulating types of fatty acids with CHD risk. METHODS UK Biobank is a prospective study of adults aged 40-69 in 2006-2010; in 2012-2013, a subset of the participants were resurveyed. Analyses were restricted to 89,242 participants with baseline plasma fatty acids (measured using nuclear magnetic resonance spectroscopy) and without prior CHD. Cox proportional hazards models were used to estimate hazard ratios (HRs) for the associations with incidence CHD, defined as the first-ever myocardial infarction, unstable angina pectoris, coronary-related death, or relevant procedure. And the major types of fatty acids were mutually adjusted to examine the independent associations. Hazard ratios were corrected for regression dilution using the correlation of baseline and resurvey fatty acids measures. RESULTS During a median follow-up of 11.8 years, 3,815 incident cases of CHD occurred. Independently of other fatty acids, CHD risk was positively associated with saturated fatty acids (SFA) and monounsaturated fatty acids (MUFA), inversely associated with omega-3 polyunsaturated fatty acids (PUFA), but there was no strong evidence of an association with omega-6 PUFA: HR per standard deviation higher were 1.14 (95% CI, 1.09-1.20), 1.15 (1.10-1.21), 0.91 (0.87-0.94), and 1.04 (0.99-1.09) respectively. Independently of triglycerides and cholesterol, the inverse association with omega-3 PUFA was not materially changed, but the positive associations with SFA and MUFA attenuated to null after adjusting for triglycerides levels. CONCLUSIONS This large-scale study has quantitated the independent associations of circulating fatty acids with CHD risk. Omega-3 PUFA was inversely related to CHD risk, independently of other fatty acids and major lipid fractions. By contrast, independently of other fatty acids, the positive associations of circulating SFA and MUFA with CHD risk were mostly attributed to their relationship with triglycerides.
Collapse
Affiliation(s)
- Danyao Jin
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| | - Eirini Trichia
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, University of Oxford, Oxford, UK
| | - Nazrul Islam
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - Sarah Lewington
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, University of Oxford, Oxford, UK
| | - Ben Lacey
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- UK Biobank, Stockport, Greater Manchester, UK
| |
Collapse
|
71
|
Levesque C, Sanger N, Edalati H, Sohi I, Shield KD, Sherk A, Stockwell T, Butt PR, Paradis C. A systematic review of relative risks for the relationships between chronic alcohol use and the occurrence of disease. Alcohol Clin Exp Res 2023. [PMID: 37422765 DOI: 10.1111/acer.15121] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 05/10/2023] [Accepted: 05/14/2023] [Indexed: 07/11/2023]
Abstract
Alcohol use is causally linked to the development of and mortality from numerous diseases. The aim of this study is to provide an update to a previous systematic review of meta-analyses that quantify the sex-specific dose-response risk relationships between chronic alcohol use and disease occurrence and/or mortality. An updated systematic search of multiple databases was performed in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses criteria to identify meta-analyses published from January 1, 2017, to March 8, 2021, which quantified the risk relationships between chronic alcohol use and the risk of disease occurrence and/or mortality. This systematic review was not preregistered. The comparator was people who have never consumed at least one standard drink of alcohol. Measurements included relative risks, odds ratios, and hazard ratios of disease occurrence and/or mortality based on long-term alcohol intake measured in grams per day. The systematic search yielded 5953 articles, of which 14 were included in the narrative review. All diseases showed an increased risk of occurrence as alcohol use increased. At all doses examined, alcohol had a significant detrimental effect on tuberculosis, lower respiratory infections, oral cavity and pharyngeal cancers, esophageal cancer, colorectal cancer, liver cancer, laryngeal cancer, epilepsy, hypertension, liver cirrhosis, and pancreatitis (among men). For ischemic heart disease, ischemic stroke, and intracerebral hemorrhage, protective effects from low-dose chronic alcohol use among both men and women were observed. Low-dose alcohol consumption also had a protective effect for diabetes mellitus and pancreatitis among women (approximately to 50 g/day and 30 g/day, respectively). Alcohol use increases the risk of numerous infectious and noncommunicable diseases in a dose-response manner. Higher levels of alcohol use have a clear detrimental impact on health; however, at lower levels of use, alcohol can have both disease-specific protective and detrimental effects.
Collapse
Affiliation(s)
| | - Nitika Sanger
- Canadian Centre on Substance Use and Addiction, Ottawa, Ontario, Canada
| | - Hanie Edalati
- Canadian Centre on Substance Use and Addiction, Ottawa, Ontario, Canada
| | - Ivneet Sohi
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Kevin D Shield
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Adam Sherk
- Canadian Institute for Substance Use Research, University of Victoria, Victoria, British Columbia, Canada
| | - Tim Stockwell
- Canadian Institute for Substance Use Research, University of Victoria, Victoria, British Columbia, Canada
- Department of Psychology, University of Victoria, Victoria, British Columbia, Canada
| | - Peter R Butt
- College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Catherine Paradis
- Canadian Centre on Substance Use and Addiction, Ottawa, Ontario, Canada
| |
Collapse
|
72
|
Grover SP, Snir O, Hindberg K, Englebert TM, Braekkan SK, Morelli VM, Jensen SB, Wolberg AS, Mollnes TE, Ueland T, Mackman N, Hansen JB. High plasma levels of C1-inhibitor are associated with lower risk of future venous thromboembolism. J Thromb Haemost 2023; 21:1849-1860. [PMID: 37003465 PMCID: PMC11112258 DOI: 10.1016/j.jtha.2023.03.024] [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: 01/10/2023] [Revised: 03/02/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023]
Abstract
BACKGROUND C1-inhibitor (C1INH) is a broad-acting serine protease inhibitor with anticoagulant activity. The impact of C1INH plasma levels within the normal physiological range on risk of venous thromboembolism (VTE) is unknown. We assessed the association of plasma C1INH levels and VTE risk and evaluated the impact of C1INH on thrombin and plasmin generation in ex vivo assays. METHODS A nested case-control study with 405 patients with VTE and 829 age- and sex-matched controls was derived from the Tromsø Study. Odds ratios (ORs) with 95% confidence intervals (95% CI) for VTE were estimated across plasma C1INH quartiles. Genetic regulation of C1INH was explored using quantitative trait loci analysis of whole exome sequencing data. The effect of plasma C1INH levels on coagulation was evaluated ex vivo by calibrated automated thrombography. RESULTS Individuals with C1INH levels in the highest quartile had a lower risk of VTE (OR 0.68, 95% CI: 0.49-0.96) compared with those with C1INH in the lowest quartile. In subgroup analysis, the corresponding ORs were 0.60 (95% CI: 0.39-0.89) for deep vein thrombosis and 0.85 (95% CI: 0.52-1.38) for pulmonary embolism, respectively. No significant genetic determinants of plasma C1INH levels were identified. Addition of exogenous C1INH to normal human plasma reduced thrombin generation triggered by an activator of the intrinsic coagulation pathway, but not when triggered by an activator of the extrinsic coagulation pathway. CONCLUSIONS High plasma levels of C1INH were associated with lower risk of VTE, and C1INH inhibited thrombin generation initiated by the intrinsic coagulation pathway ex vivo.
Collapse
Affiliation(s)
- Steven P Grover
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, North Carolina, USA. https://twitter.com/StevenPGrover
| | - Omri Snir
- Thrombosis Research Center, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway; Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Kristian Hindberg
- Thrombosis Research Center, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway. https://twitter.com/KristianHindbe1
| | - Tatianna M Englebert
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, North Carolina, USA. https://twitter.com/OlsonTatianna
| | - Sigrid K Braekkan
- Thrombosis Research Center, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway; Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway.
| | - Vânia M Morelli
- Thrombosis Research Center, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway; Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Søren B Jensen
- Thrombosis Research Center, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway; Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Alisa S Wolberg
- Department of Pathology and Laboratory Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, North Carolina, USA. https://twitter.com/aswolberg
| | - Tom Eirik Mollnes
- Research Laboratory, Nordland Hospital, Bodø, Norway; Department of Immunology, Oslo University Hospital and University of Oslo, Norway; Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Thor Ueland
- Thrombosis Research Center, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway; Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway. https://twitter.com/ThorUeland
| | - Nigel Mackman
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, North Carolina, USA. https://twitter.com/NMackman
| | - John-Bjarne Hansen
- Thrombosis Research Center, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway; Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
73
|
Li R, Li Y, Fan Z, Liu Z, Lin J, He M. L-shaped association of serum 25-hydroxyvitamin D with all-cause and cardiovascular mortality in older people with chronic kidney disease: results from the NHANES database prospective cohort study. BMC Public Health 2023; 23:1260. [PMID: 37380964 DOI: 10.1186/s12889-023-16165-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND This study was conducted to assess the association of serum 25-hydroxyvitamin D [25(OH)D] concentrations with all-cause and cardiovascular disease (CVD) mortality in older people with chronic kidney disease (CKD) in the United States. METHODS We identified 3230 CKD participants aged ≥ 60 years from the National Health and Nutrition Examination Survey (2001-2018). CKD was defined as an estimated glomerular filtration rate (eGFR) < 60 ml/min/1.73 m2. Mortality outcomes were determined by linkage to National Death Index (NDI) records through December 31, 2019. Restricted cubic spline based on Cox regression models were utilized to elucidate the nonlinear relationship between serum 25(OH)D concentrations and mortality in patients with CKD. RESULTS During median 74 months of follow-up, 1615 all-cause death and 580 CVD death were recorded. We found an L-shaped association between serum 25(OH)D concentrations and all-cause and CVD mortality, reaching a plateau at 90 nmol/L. Accordingly, per one-unit increment in natural log-transformed 25(OH)D was associated with a 32% and 33% reduced risk of all-cause mortality (hazard ratio [HR] 0.68; 95%CI, 0.56 to 0.83) and CV mortality (HR 0.69; 95%CI, 0.49 to 0.97) in participants with serum 25(OH)D < 90 nmol/L, but no considerable difference was observed in participants with serum 25(OH)D ≥ 90 nmol/L. Compared with those in the deficiency group (< 50 nmol/L), insufficient (50 to < 75 nmol/L) and sufficient group (≥ 75 nmol/L) were significantly associated with lower all-cause mortality (HR,0.83; 95%CI, 0.71 to 0.97 and HR, 0.75; 95%CI, 0.64 to 0.89) and CV mortality (HR,0.87; 95%CI, 0.68 to 1.10 and HR, 0.77; 95%CI, 0.59 to < 1.0), respectively. CONCLUSION An L-shaped relationship between serum 25(OH)D levels with all-cause and CVD mortality was observed in elderly CKD patients in the United States. A 25(OH)D concentration of 90 nmol/L may be the target to reduce the risk of premature death.
Collapse
Affiliation(s)
- Rugang Li
- Department of Nephrology, Affiliated Yuebei People's Hospital of Shantou University Medical College, No. 133 South Huimin Road, Shaoguan, 512026, Guangdong, China
| | - Yang Li
- Department of Nephrology, Haikou Municipal People's Hospital and Central South University Xiangya Medical College Affiliated Hospital, Haikou, China
| | - Zhongcheng Fan
- Department of Osteology, Haikou Municipal People's Hospital and Central South University Xiangya Medical College Affiliated Hospital, Haikou, China
| | - Zhaoqi Liu
- Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Juhua Lin
- Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Min He
- Department of Nephrology, Affiliated Yuebei People's Hospital of Shantou University Medical College, No. 133 South Huimin Road, Shaoguan, 512026, Guangdong, China.
- Guangdong Medical University, Zhanjiang, 524001, Guangdong, China.
| |
Collapse
|
74
|
Im PK, Wright N, Yang L, Chan KH, Chen Y, Guo Y, Du H, Yang X, Avery D, Wang S, Yu C, Lv J, Clarke R, Chen J, Collins R, Walters RG, Peto R, Li L, Chen Z, Millwood IY. Alcohol consumption and risks of more than 200 diseases in Chinese men. Nat Med 2023; 29:1476-1486. [PMID: 37291211 PMCID: PMC10287564 DOI: 10.1038/s41591-023-02383-8] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/02/2023] [Indexed: 06/10/2023]
Abstract
Alcohol consumption accounts for ~3 million annual deaths worldwide, but uncertainty persists about its relationships with many diseases. We investigated the associations of alcohol consumption with 207 diseases in the 12-year China Kadoorie Biobank of >512,000 adults (41% men), including 168,050 genotyped for ALDH2- rs671 and ADH1B- rs1229984 , with >1.1 million ICD-10 coded hospitalized events. At baseline, 33% of men drank alcohol regularly. Among men, alcohol intake was positively associated with 61 diseases, including 33 not defined by the World Health Organization as alcohol-related, such as cataract (n = 2,028; hazard ratio 1.21; 95% confidence interval 1.09-1.33, per 280 g per week) and gout (n = 402; 1.57, 1.33-1.86). Genotype-predicted mean alcohol intake was positively associated with established (n = 28,564; 1.14, 1.09-1.20) and new alcohol-associated (n = 16,138; 1.06, 1.01-1.12) diseases, and with specific diseases such as liver cirrhosis (n = 499; 2.30, 1.58-3.35), stroke (n = 12,176; 1.38, 1.27-1.49) and gout (n = 338; 2.33, 1.49-3.62), but not ischemic heart disease (n = 8,408; 1.04, 0.94-1.14). Among women, 2% drank alcohol resulting in low power to assess associations of self-reported alcohol intake with disease risks, but genetic findings in women suggested the excess male risks were not due to pleiotropic genotypic effects. Among Chinese men, alcohol consumption increased multiple disease risks, highlighting the need to strengthen preventive measures to reduce alcohol intake.
Collapse
Affiliation(s)
- Pek Kei Im
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Neil Wright
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Ling Yang
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Ka Hung Chan
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Oxford British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Yiping Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Yu Guo
- Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Huaidong Du
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Xiaoming Yang
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Daniel Avery
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Shaojie Wang
- NCD Prevention and Control Department, Qingdao CDC, Qingdao, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
| | - Robert Clarke
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Junshi Chen
- China National Center for Food Safety Risk Assessment, Beijing, China
| | - Rory Collins
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Robin G Walters
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Richard Peto
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
| | - Zhengming Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK.
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| | - Iona Y Millwood
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK.
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| |
Collapse
|
75
|
Edvardsen MS, Hansen ES, Ueland T, Aukrust P, Brækkan SK, Morelli VM, Hansen JB. Impact of the von Willebrand factor-ADAMTS-13 axis on the risk of future venous thromboembolism. J Thromb Haemost 2023; 21:1227-1237. [PMID: 36736832 DOI: 10.1016/j.jtha.2023.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/30/2022] [Accepted: 01/23/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND von Willebrand factor (VWF) and its cleaving protease, ADAMTS-13, form a pivotal axis that regulates hemostasis. However, the role of the VWF-ADAMTS-13 axis in the risk of future venous thromboembolism (VTE) is unknown. OBJECTIVES To investigate whether plasma ADAMTS-13 levels and an imbalance with VWF levels, assessed as the VWF/ADAMTS-13 ratio, are associated with the risk of future VTE. PATIENTS/METHODS A population-based nested case-control study, comprising 383 incident VTE cases and 780 age- and sex-matched controls, was derived from the Tromsø study cohort (1994-2007). Antigen levels of ADAMTS-13 and VWF were measured in plasma samples obtained at cohort baseline. Odds ratios (ORs) with 95% CIs were estimated according to quartile cutoffs of ADAMTS-13 and VWF/ADAMTS-13 ratio determined in controls. RESULTS In age- and sex-adjusted analysis, ADAMTS-13 levels were inversely associated with the VTE risk, with an OR of 1.40 (95% CI, 0.99-1.99) for the lowest vs highest quartiles. The VWF/ADAMTS-13 ratio was linearly associated with the VTE risk (P for trend = .001), with an OR of 1.70 (95% CI, 1.19-2.43) for the highest vs lowest quartiles, and the association was particularly pronounced for unprovoked VTE (OR, 2.81; 95% CI, 1.65-4.81). The ORs were only slightly attenuated after additional adjustments for body mass index and C-reactive protein. CONCLUSIONS Lowered ADAMTS-13 levels and an imbalance between ADAMTS-13 and VWF levels, reflected by an increased VWF/ADAMTS-13 ratio, were associated with an increased risk of future VTE. Our findings suggest that the VWF-ADAMTS-13 axis is involved in the pathogenesis of VTE.
Collapse
Affiliation(s)
- Magnus S Edvardsen
- Thrombosis Research Center (TREC), Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Ellen-Sofie Hansen
- Thrombosis Research Center (TREC), Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Thor Ueland
- Thrombosis Research Center (TREC), Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway; Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Sigrid K Brækkan
- Thrombosis Research Center (TREC), Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway; Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Vânia M Morelli
- Thrombosis Research Center (TREC), Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway; Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway.
| | - John-Bjarne Hansen
- Thrombosis Research Center (TREC), Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway; Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
76
|
Liang T, Xie C, Lv B, Su L, Long J, Liu S, Huang X, Pei P, Pan R, Lan J. Age at smoking initiation and smoking cessation influence the incidence of stroke in China: a 10-year follow-up study. J Thromb Thrombolysis 2023:10.1007/s11239-023-02812-y. [PMID: 37099076 DOI: 10.1007/s11239-023-02812-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 04/27/2023]
Abstract
Our study aimed to explore the correlation between age at smoking initiation and smoking cessation for the risk for stroke in China. We investigated 50,174 participants from one of the urban areas of China Kadoorie Biobank (CKB) Study. Hazard ratios (HRs) and 95% confidence intervals (95% CIs) for association between smoking and incidence of stroke were estimated using Cox regression model. During a median of 10.7 years of follow-up, 4370 total stroke cases were documented. Among men, comparing current smokers to never smokers, the HR of total stroke for current smokers was 1.279 (95% CI, 1.134-1.443) for total stroke. The HRs of total stroke were 1.344 (1.151-1.570) for those started smoking at age less than 20 years, 1.254 (1.090-1.443) for those started smoking at age 20-30 years, and 1.205 (1.012-1.435) for those started smoking at age 30 year and above, with a dose-response relation (P for trend, 0.004). Comparing former smokers to current smokers, in the low pack-year group, those stopped smoking at age less than 65 years had a 18.2% decreased risk for total stroke (0.818; 0.673-0.994). The decreased risk was not found in those stopped smoking at age 65 years and above. Similar results were observed in the high pack-year group. In conclusion, we found that current smokers had a higher stroke risk than never smokers, and the risk increased with a younger age at smoking initiation. Smoking cessation can reduce the risk for stroke, especially could benefit from cessation at a younger age.
Collapse
Affiliation(s)
- Tian Liang
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Changping Xie
- Liuzhou Center for Disease Control and Prevention, Liuzhou, Guangxi, 545007, China
| | - Bangjun Lv
- Liuzhou Center for Disease Control and Prevention, Liuzhou, Guangxi, 545007, China
| | - Li Su
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jianxiong Long
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Shengying Liu
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xiaolan Huang
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Pei Pei
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, 100191, China
| | - Rong Pan
- Liuzhou Center for Disease Control and Prevention, Liuzhou, Guangxi, 545007, China.
| | - Jian Lan
- Liuzhou Center for Disease Control and Prevention, Liuzhou, Guangxi, 545007, China.
| |
Collapse
|
77
|
Skjeflo EW, Evensen LH, Jensen SB, Latysheva N, Michelsen A, Ueland T, Brækkan SK, Hindberg K, Snir O, Mollnes TE, Hansen JB. Complement factors B, D, C3bBbP and risk of future venous thromboembolism. Clin Immunol 2023; 249:109278. [PMID: 36894046 DOI: 10.1016/j.clim.2023.109278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 02/19/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023]
Abstract
The complement system appears to be involved in the pathogenesis of venous thromboembolism (VTE). We investigated the association of complement factors (CF) B, D, and the alternative pathway convertase, C3bBbP, measured at inclusion, with the risk of future VTE in a nested case-control study; 380 VTE patients and 804 age- and sex-matched controls derived from the Tromsø study. Odds ratios (ORs) with 95% confidence intervals (95% CI) for VTE across tertiles of CF concentrations were estimated using logistic regression. There was no association between CFB or CFD and risk of future VTE. Higher levels of C3bBbP gave an increased risk of provoked VTE; subjects in Q4 had a 1.68-fold higher OR compared with Q1 in the age-, sex- and BMI-adjusted model (OR 1.68; 95% CI 1.08-2.64). There was no increased risk of future VTE in individuals with higher levels of complement factors B or D of the alternative pathway. Increased levels of the alternative pathway activation product, C3bBbP, showed an association with future risk of provoked VTE.
Collapse
Affiliation(s)
- Espen W Skjeflo
- K.G. Jebsen - Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, University of Tromsø - The Arctic University of Norway, Tromsø, Norway; Research Laboratory, Nordland Hospital, Bodø, Norway.
| | - Line H Evensen
- K.G. Jebsen - Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Søren B Jensen
- K.G. Jebsen - Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Nadezhda Latysheva
- K.G. Jebsen - Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Annika Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Thor Ueland
- K.G. Jebsen - Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, University of Tromsø - The Arctic University of Norway, Tromsø, Norway; Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sigrid K Brækkan
- K.G. Jebsen - Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, University of Tromsø - The Arctic University of Norway, Tromsø, Norway; Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Kristian Hindberg
- K.G. Jebsen - Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Omri Snir
- K.G. Jebsen - Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Tom Eirik Mollnes
- K.G. Jebsen - Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, University of Tromsø - The Arctic University of Norway, Tromsø, Norway; Research Laboratory, Nordland Hospital, Bodø, Norway; Department of Immunology, Oslo University Hospital and University of Oslo, Norway; Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - John-Bjarne Hansen
- K.G. Jebsen - Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, University of Tromsø - The Arctic University of Norway, Tromsø, Norway; Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
78
|
Clarke R, Wright N, Walters R, Gan W, Guo Y, Millwood IY, Yang L, Chen Y, Lewington S, Lv J, Yu C, Avery D, Lin K, Wang K, Peto R, Collins R, Li L, Bennett DA, Parish S, Chen Z. Genetically Predicted Differences in Systolic Blood Pressure and Risk of Cardiovascular and Noncardiovascular Diseases: A Mendelian Randomization Study in Chinese Adults. Hypertension 2023; 80:566-576. [PMID: 36601918 PMCID: PMC7614188 DOI: 10.1161/hypertensionaha.122.20120] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Mendelian randomization studies of systolic blood pressure (SBP) can assess the shape and strength of the associations of genetically predicted differences in SBP with major disease outcomes and are less constrained by biases in observational analyses. This study aimed to compare the associations of usual and genetically predicted SBP with major cardiovascular disease (CVD) outcomes, overall and by levels of SBP, age, and sex. METHODS The China Kadoorie Biobank involved a 12-year follow-up of a prospective study of 489 495 adults aged 40 to 79 years with no prior CVD and 86 060 with genetic data. Outcomes included major vascular events (59 490/23 151 in observational/genetic analyses), and its components (ischemic stroke [n=39 513/12 043], intracerebral hemorrhage [7336/5243], and major coronary events [7871/4187]). Genetically predicted SBP used 460 variants obtained from European ancestry genome-wide studies. Cox regression estimated adjusted hazard ratios for incident CVD outcomes down to usual SBP levels of 120 mm Hg. RESULTS Both observational and genetic analyses demonstrated log-linear positive associations of SBP with major vascular event and other major CVD types in the range of 120 to 170 mm Hg. Consistent with the observational analyses, the hazard ratios per 10 mm Hg higher genetically predicted SBP were 2-fold greater for intracerebral hemorrhage (1.71 [95% CI, 1.58-1.87]) than for ischemic stroke (1.37 [1.30-1.45]) or major coronary event (1.29 [1.18-1.42]). Genetic analyses also demonstrated 2-fold greater hazard ratios for major vascular event in younger (1.69 [95% CI, 1.54-1.86]) than in older people (1.28 [1.18-1.38]). CONCLUSIONS The findings provide support for initiation of blood pressure-lowering treatment at younger ages and below the conventional cut-offs for hypertension to maximize CVD prevention, albeit the absolute risks of CVD are far greater in older people.
Collapse
Affiliation(s)
- Robert Clarke
- Clinical Trial Service Unit and Epidemiological Studies, Nuffield Department of Population Health, University of Oxford, United Kingdom (R. Clarke, N.W., R.W., I.Y.M., L.Y., Y.C., S.L., D.A., K.L., R.P., R. Collins, D.A.B., S.P., Z.C.).,Medical Research Council, Population Health Research Unit, University of Oxford, United Kingdom (R. Clarke, R.W., I.Y.M., L.Y., Y.C., S.L., D.A.B., S.P., Z.C.)
| | - Neil Wright
- Clinical Trial Service Unit and Epidemiological Studies, Nuffield Department of Population Health, University of Oxford, United Kingdom (R. Clarke, N.W., R.W., I.Y.M., L.Y., Y.C., S.L., D.A., K.L., R.P., R. Collins, D.A.B., S.P., Z.C.).,Medical Research Council, Population Health Research Unit, University of Oxford, United Kingdom (R. Clarke, R.W., I.Y.M., L.Y., Y.C., S.L., D.A.B., S.P., Z.C.)
| | - Robin Walters
- Clinical Trial Service Unit and Epidemiological Studies, Nuffield Department of Population Health, University of Oxford, United Kingdom (R. Clarke, N.W., R.W., I.Y.M., L.Y., Y.C., S.L., D.A., K.L., R.P., R. Collins, D.A.B., S.P., Z.C.).,Medical Research Council, Population Health Research Unit, University of Oxford, United Kingdom (R. Clarke, R.W., I.Y.M., L.Y., Y.C., S.L., D.A.B., S.P., Z.C.)
| | - Wei Gan
- Novo Nordisk Research Centre Oxford, Novo Nordisk Ltd, Oxford, United Kingdom (W.G.)
| | - Yu Guo
- Fuwai Hospital Chinese Academy of Medical Sciences, National Center for Cardiovascular Diseases, Beijing, China (Y.G.)
| | - Iona Y Millwood
- Clinical Trial Service Unit and Epidemiological Studies, Nuffield Department of Population Health, University of Oxford, United Kingdom (R. Clarke, N.W., R.W., I.Y.M., L.Y., Y.C., S.L., D.A., K.L., R.P., R. Collins, D.A.B., S.P., Z.C.).,Medical Research Council, Population Health Research Unit, University of Oxford, United Kingdom (R. Clarke, R.W., I.Y.M., L.Y., Y.C., S.L., D.A.B., S.P., Z.C.)
| | - Ling Yang
- Clinical Trial Service Unit and Epidemiological Studies, Nuffield Department of Population Health, University of Oxford, United Kingdom (R. Clarke, N.W., R.W., I.Y.M., L.Y., Y.C., S.L., D.A., K.L., R.P., R. Collins, D.A.B., S.P., Z.C.).,Medical Research Council, Population Health Research Unit, University of Oxford, United Kingdom (R. Clarke, R.W., I.Y.M., L.Y., Y.C., S.L., D.A.B., S.P., Z.C.)
| | - Yiping Chen
- Clinical Trial Service Unit and Epidemiological Studies, Nuffield Department of Population Health, University of Oxford, United Kingdom (R. Clarke, N.W., R.W., I.Y.M., L.Y., Y.C., S.L., D.A., K.L., R.P., R. Collins, D.A.B., S.P., Z.C.).,Medical Research Council, Population Health Research Unit, University of Oxford, United Kingdom (R. Clarke, R.W., I.Y.M., L.Y., Y.C., S.L., D.A.B., S.P., Z.C.)
| | - Sarah Lewington
- Clinical Trial Service Unit and Epidemiological Studies, Nuffield Department of Population Health, University of Oxford, United Kingdom (R. Clarke, N.W., R.W., I.Y.M., L.Y., Y.C., S.L., D.A., K.L., R.P., R. Collins, D.A.B., S.P., Z.C.).,Medical Research Council, Population Health Research Unit, University of Oxford, United Kingdom (R. Clarke, R.W., I.Y.M., L.Y., Y.C., S.L., D.A.B., S.P., Z.C.)
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Sciences Center, Beijing, China (J.L., C.Y., L.L.).,Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China (J.L., C.Y., L.L.)
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Sciences Center, Beijing, China (J.L., C.Y., L.L.).,Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China (J.L., C.Y., L.L.)
| | - Daniel Avery
- Clinical Trial Service Unit and Epidemiological Studies, Nuffield Department of Population Health, University of Oxford, United Kingdom (R. Clarke, N.W., R.W., I.Y.M., L.Y., Y.C., S.L., D.A., K.L., R.P., R. Collins, D.A.B., S.P., Z.C.)
| | - Kuang Lin
- Clinical Trial Service Unit and Epidemiological Studies, Nuffield Department of Population Health, University of Oxford, United Kingdom (R. Clarke, N.W., R.W., I.Y.M., L.Y., Y.C., S.L., D.A., K.L., R.P., R. Collins, D.A.B., S.P., Z.C.)
| | - Kang Wang
- NCDs Prevention and Control Department, Shibei CDC, China (K.W.)
| | - Richard Peto
- Clinical Trial Service Unit and Epidemiological Studies, Nuffield Department of Population Health, University of Oxford, United Kingdom (R. Clarke, N.W., R.W., I.Y.M., L.Y., Y.C., S.L., D.A., K.L., R.P., R. Collins, D.A.B., S.P., Z.C.)
| | - Rory Collins
- Clinical Trial Service Unit and Epidemiological Studies, Nuffield Department of Population Health, University of Oxford, United Kingdom (R. Clarke, N.W., R.W., I.Y.M., L.Y., Y.C., S.L., D.A., K.L., R.P., R. Collins, D.A.B., S.P., Z.C.)
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Sciences Center, Beijing, China (J.L., C.Y., L.L.).,Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China (J.L., C.Y., L.L.)
| | - Derrick A Bennett
- Clinical Trial Service Unit and Epidemiological Studies, Nuffield Department of Population Health, University of Oxford, United Kingdom (R. Clarke, N.W., R.W., I.Y.M., L.Y., Y.C., S.L., D.A., K.L., R.P., R. Collins, D.A.B., S.P., Z.C.).,Medical Research Council, Population Health Research Unit, University of Oxford, United Kingdom (R. Clarke, R.W., I.Y.M., L.Y., Y.C., S.L., D.A.B., S.P., Z.C.)
| | - Sarah Parish
- Clinical Trial Service Unit and Epidemiological Studies, Nuffield Department of Population Health, University of Oxford, United Kingdom (R. Clarke, N.W., R.W., I.Y.M., L.Y., Y.C., S.L., D.A., K.L., R.P., R. Collins, D.A.B., S.P., Z.C.).,Medical Research Council, Population Health Research Unit, University of Oxford, United Kingdom (R. Clarke, R.W., I.Y.M., L.Y., Y.C., S.L., D.A.B., S.P., Z.C.)
| | - Zhengming Chen
- Clinical Trial Service Unit and Epidemiological Studies, Nuffield Department of Population Health, University of Oxford, United Kingdom (R. Clarke, N.W., R.W., I.Y.M., L.Y., Y.C., S.L., D.A., K.L., R.P., R. Collins, D.A.B., S.P., Z.C.).,Medical Research Council, Population Health Research Unit, University of Oxford, United Kingdom (R. Clarke, R.W., I.Y.M., L.Y., Y.C., S.L., D.A.B., S.P., Z.C.)
| | | |
Collapse
|
79
|
Watts EL, Perez-Cornago A, Fensom GK, Smith-Byrne K, Noor U, Andrews CD, Gunter MJ, Holmes MV, Martin RM, Tsilidis KK, Albanes D, Barricarte A, Bueno-de-Mesquita HB, Cohn BA, Deschasaux-Tanguy M, Dimou NL, Ferrucci L, Flicker L, Freedman ND, Giles GG, Giovannucci EL, Haiman CA, Hankey GJ, Holly JMP, Huang J, Huang WY, Hurwitz LM, Kaaks R, Kubo T, Le Marchand L, MacInnis RJ, Männistö S, Metter EJ, Mikami K, Mucci LA, Olsen AW, Ozasa K, Palli D, Penney KL, Platz EA, Pollak MN, Roobol MJ, Schaefer CA, Schenk JM, Stattin P, Tamakoshi A, Thysell E, Tsai CJ, Touvier M, Van Den Eeden SK, Weiderpass E, Weinstein SJ, Wilkens LR, Yeap BB. Circulating insulin-like growth factors and risks of overall, aggressive and early-onset prostate cancer: a collaborative analysis of 20 prospective studies and Mendelian randomization analysis. Int J Epidemiol 2023; 52:71-86. [PMID: 35726641 PMCID: PMC9908067 DOI: 10.1093/ije/dyac124] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Previous studies had limited power to assess the associations of circulating insulin-like growth factors (IGFs) and IGF-binding proteins (IGFBPs) with clinically relevant prostate cancer as a primary endpoint, and the association of genetically predicted IGF-I with aggressive prostate cancer is not known. We aimed to investigate the associations of IGF-I, IGF-II, IGFBP-1, IGFBP-2 and IGFBP-3 concentrations with overall, aggressive and early-onset prostate cancer. METHODS Prospective analysis of biomarkers using the Endogenous Hormones, Nutritional Biomarkers and Prostate Cancer Collaborative Group dataset (up to 20 studies, 17 009 prostate cancer cases, including 2332 aggressive cases). Odds ratios (OR) and 95% confidence intervals (CI) for prostate cancer were estimated using conditional logistic regression. For IGF-I, two-sample Mendelian randomization (MR) analysis was undertaken using instruments identified using UK Biobank (158 444 men) and outcome data from PRACTICAL (up to 85 554 cases, including 15 167 aggressive cases). Additionally, we used colocalization to rule out confounding by linkage disequilibrium. RESULTS In observational analyses, IGF-I was positively associated with risks of overall (OR per 1 SD = 1.09: 95% CI 1.07, 1.11), aggressive (1.09: 1.03, 1.16) and possibly early-onset disease (1.11: 1.00, 1.24); associations were similar in MR analyses (OR per 1 SD = 1.07: 1.00, 1.15; 1.10: 1.01, 1.20; and 1.13; 0.98, 1.30, respectively). Colocalization also indicated a shared signal for IGF-I and prostate cancer (PP4: 99%). Men with higher IGF-II (1.06: 1.02, 1.11) and IGFBP-3 (1.08: 1.04, 1.11) had higher risks of overall prostate cancer, whereas higher IGFBP-1 was associated with a lower risk (0.95: 0.91, 0.99); these associations were attenuated following adjustment for IGF-I. CONCLUSIONS These findings support the role of IGF-I in the development of prostate cancer, including for aggressive disease.
Collapse
Affiliation(s)
- Eleanor L Watts
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Aurora Perez-Cornago
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Georgina K Fensom
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Karl Smith-Byrne
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Urwah Noor
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Colm D Andrews
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Marc J Gunter
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | - Michael V Holmes
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, UK
| | - Richard M Martin
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Bristol, UK
- National Institute for Health Research (NIHR) Bristol Biomedical Research Centre, University Hospitals Bristol NHS Foundation Trust and Weston NHS Foundation Trust and University of Bristol, Bristol, UK
| | - Konstantinos K Tsilidis
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Aurelio Barricarte
- Group of Epidemiology of Cancer and Other Chronic Diseases, Navarra Public Health Institute, Pamplona, Spain
- Group of Epidemiology of Cancer and Other Chronic Diseases, Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- CIBER Epidemiology and Public Health CIBERESP, Madrid, Spain
| | - H Bas Bueno-de-Mesquita
- Centre for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment (RIVM), Utrecht, The Netherlands
| | - Barbara A Cohn
- Child Health and Development Studies, Public Health Institute, Berkeley, CA, USA
| | - Melanie Deschasaux-Tanguy
- Sorbonne Paris Nord University, Nutritional Epidemiology Research Team, Epidemiology and Statistics Research Center, University of Paris, Bobigny, France
| | - Niki L Dimou
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | | | - Leon Flicker
- WA Centre for Health & Ageing, Medical School, University of Western Australia, Perth, WA, Australia
- Western Australian Centre for Health and Ageing, University of Western Australia, Perth, WA, Australia
| | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Graham G Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Melbourne, VIC, Australia
| | - Edward L Giovannucci
- Department of Epidemiology, 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 Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Christopher A Haiman
- Department of Preventive Medicine, Center for Genetic Epidemiology, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Graham J Hankey
- WA Centre for Health & Ageing, Medical School, University of Western Australia, Perth, WA, Australia
| | - Jeffrey M P Holly
- IGFs & Metabolic Endocrinology Group, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Jiaqi Huang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Wen-Yi Huang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lauren M Hurwitz
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tatsuhiko Kubo
- Department of Public Health and Health Policy, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | | | - Robert J MacInnis
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Melbourne, VIC, Australia
| | - Satu Männistö
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - E Jeffrey Metter
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kazuya Mikami
- Departmemt of Urology, Japanese Red Cross Kyoto Daiichi Hospital, Kyoto, Japan
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Anja W Olsen
- Department of Public Health, Aarhus University, Aarhus, Denmark
- Danish Cancer Society, Research Center, Copenhagen, Denmark
| | - Kotaro Ozasa
- Departmemt of Epidemiology, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Domenico Palli
- Cancer Risk Factors and Life-Style Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network, Florence, Italy
| | - Kathryn L Penney
- Department of Epidemiology, 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
| | - Elizabeth A Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Michael N Pollak
- Departments of Medicine and Oncology, McGill University, Montreal, QC, Canada
| | - Monique J Roobol
- Department of Urology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Jeannette M Schenk
- Cancer Prevention Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Pär Stattin
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Akiko Tamakoshi
- Department of Public Health, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Elin Thysell
- Department of Medical Biosciences, Umeå University, Umeå, Sweden
| | - Chiaojung Jillian Tsai
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mathilde Touvier
- Sorbonne Paris Nord University, Nutritional Epidemiology Research Team, Epidemiology and Statistics Research Center, University of Paris, Bobigny, France
| | - Stephen K Van Den Eeden
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
- Department of Urology, University of CaliforniaSan Francisco, San Francisco, CA, USA
| | - Elisabete Weiderpass
- Director’s Office, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Stephanie J Weinstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Bu B Yeap
- WA Centre for Health & Ageing, Medical School, University of Western Australia, Perth, WA, Australia
- Department of Endocrinology and Diabetes, Fiona Stanley Hospital, Perth, WA, Australia
| |
Collapse
|
80
|
Watts EL, Saint-Maurice PF, Doherty A, Fensom GK, Freeman JR, Gorzelitz JS, Jin D, McClain KM, Papier K, Patel S, Shiroma EJ, Moore SC, Matthews CE. Association of Accelerometer-Measured Physical Activity Level With Risks of Hospitalization for 25 Common Health Conditions in UK Adults. JAMA Netw Open 2023; 6:e2256186. [PMID: 36795414 PMCID: PMC9936337 DOI: 10.1001/jamanetworkopen.2022.56186] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/29/2022] [Indexed: 02/17/2023] Open
Abstract
Importance Higher physical activity levels are associated with lower risks of cancer, cardiovascular disease, and diabetes, but associations with many common and less severe health conditions are not known. These conditions impose large health care burdens and reduce quality of life. Objectives To investigate the association between accelerometer-measured physical activity and the subsequent risk of hospitalization for 25 common reasons for hospitalization and to estimate the proportion of these hospitalizations that might have been prevented if participants had higher levels of physical activity. Design, Setting, and Participants This prospective cohort study used data from a subset of 81 717 UK Biobank participants aged 42 to 78 years. Participants wore an accelerometer for 1 week (between June 1, 2013, and December 23, 2015) and were followed up over a median (IQR) of 6.8 (6.2-7.3) years; follow-up for the current study ended in 2021 (exact date varied by location). Exposures Mean total and intensity-specific accelerometer-measured physical activity. Main Outcomes and Measures Hospitalization for the most common health conditions. Cox proportional hazards regression analysis was used to estimate hazard ratios (HRs) and 95% CIs for mean accelerometer-measured physical activity (per 1-SD increment) and risks of hospitalization for 25 conditions. Population-attributable risks were used to estimate the proportion of hospitalizations for each condition that might be prevented if participants increased their moderate to vigorous physical activity (MVPA) by 20 minutes per day. Results Among 81 717 participants, the mean (SD) age at accelerometer assessment was 61.5 (7.9) years; 56.4% were female, and 97.0% self-identified as White. Higher levels of accelerometer-measured physical activity were associated with lower risks of hospitalization for 9 conditions: gallbladder disease (HR per 1 SD, 0.74; 95% CI, 0.69-0.79), urinary tract infections (HR per 1 SD, 0.76; 95% CI, 0.69-0.84), diabetes (HR per 1 SD, 0.79; 95% CI, 0.74-0.84), venous thromboembolism (HR per 1 SD, 0.82; 95% CI, 0.75-0.90), pneumonia (HR per 1 SD, 0.83; 95% CI, 0.77-0.89), ischemic stroke (HR per 1 SD, 0.85; 95% CI, 0.76-0.95), iron deficiency anemia (HR per 1 SD, 0.91; 95% CI, 0.84-0.98), diverticular disease (HR per 1 SD, 0.94; 95% CI, 0.90-0.99), and colon polyps (HR per 1 SD, 0.96; 95% CI, 0.94-0.99). Positive associations were observed between overall physical activity and carpal tunnel syndrome (HR per 1 SD, 1.28; 95% CI, 1.18-1.40), osteoarthritis (HR per 1 SD, 1.15; 95% CI, 1.10-1.19), and inguinal hernia (HR per 1 SD, 1.13; 95% CI, 1.07-1.19), which were primarily induced by light physical activity. Increasing MVPA by 20 minutes per day was associated with reductions in hospitalization ranging from 3.8% (95% CI, 1.8%-5.7%) for colon polyps to 23.0% (95% CI, 17.1%-28.9%) for diabetes. Conclusions and Relevance In this cohort study of UK Biobank participants, those with higher physical activity levels had lower risks of hospitalization across a broad range of health conditions. These findings suggest that aiming to increase MVPA by 20 minutes per day may be a useful nonpharmaceutical intervention to reduce health care burdens and improve quality of life.
Collapse
Affiliation(s)
- Eleanor L. Watts
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Pedro F. Saint-Maurice
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Aiden Doherty
- Big Data Institute, Nuffield Department of Population Health, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Georgina K. Fensom
- Cancer Epidemiology Unit, Nuffield Department of Population Health, Oxford, United Kingdom
| | - Joshua R. Freeman
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | | | - David Jin
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Kathleen M. McClain
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Keren Papier
- Cancer Epidemiology Unit, Nuffield Department of Population Health, Oxford, United Kingdom
| | - Shreya Patel
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Eric J. Shiroma
- Laboratory of Epidemiology and Population Science, National Institute on Aging, Bethesda, Maryland
| | - Steven C. Moore
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Charles E. Matthews
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| |
Collapse
|
81
|
Metabolic dysfunction-associated fatty liver disease and liver function markers are associated with Crohn's disease but not Ulcerative Colitis: a prospective cohort study. Hepatol Int 2023; 17:202-214. [PMID: 36194337 PMCID: PMC9895026 DOI: 10.1007/s12072-022-10424-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/15/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Metabolic dysfunction-associated fatty liver disease (MAFLD) is recently recognized as a condition featured with metabolic dysfunctions in liver. It has been supposed that MAFLD might contribute to the development of IBD, but evidence from prospective cohort studies is lacking and inconclusive. METHODS A total of 221,546 females and 183,867 males from the UK Biobank cohort enrolled in 2006-2010 were included to examine whether MAFLD and liver function markers were related to incident IBD. MAFLD was identified based on hepatic steatosis defined by fatty liver index plus the prevalence of overweight, type 2 diabetes mellitus, or at least two metabolic abnormalities. Biomarker related to liver function (albumin [ALB], alkaline phosphatase [ALP], alanine transaminase [ALT], aspartate transaminase [AST]; gamma-glutamyl transferase [GGT], total bilirubin [TB], total protein [TP]) was measured using colorimetric or enzymatic assays. The incidence of IBD was ascertained based on primary care and inpatient records. Cox proportional hazard model was used to estimate hazard ratios (HRs) with 95% confidence intervals (CI) for the magnitude of their associations. RESULTS With a mean follow-up of 12.1 years, 2228 incident IBD cases were documented. We identified 150,385 individuals with MAFLD at baseline and 86% participants' circulating liver function markers were within the normal range. Participants with MAFLD were associated with a 12% (HR 1.12, 95% CI 1.03, 1.23, p = 0.012) increased risk of IBD compared with those without MAFLD at baseline; the association was stronger (p-Heterogeneity = 0.006) with Crohn's disease (HR 1.35, 95% CI 1.15, 1.59, p < 0.001) than ulcerative colitis (HR 1.03, 95% CI 0.93, 1.15, p = 0.57). As for the serum liver function markers, the HRs of IBD for per 1-SD increment in ALB, ALP, AST, and TB concentration were 0.86 (95% CI 0.83, 0.90, p < 0.001), 1.18 (95% CI 1.13, 1.24, p < 0.001), 0.95 (95% CI 0.91, 0.99, p = 0.027), 0.92 (95% CI 0.87, 0.96, p < 0.001), respectively. We did not observe significant associations of GGT and TP with IBD. CONCLUSIONS Individuals with MAFLD were at increased risk of developing IBD, especially CD, but not UC. Circulating levels of liver function biomarkers as the surrogate indicators of MAFLD were also associated with IBD risk.
Collapse
|
82
|
Conroy MC, Lacey B, Bešević J, Omiyale W, Feng Q, Effingham M, Sellers J, Sheard S, Pancholi M, Gregory G, Busby J, Collins R, Allen NE. UK Biobank: a globally important resource for cancer research. Br J Cancer 2023; 128:519-527. [PMID: 36402876 PMCID: PMC9938115 DOI: 10.1038/s41416-022-02053-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/21/2022] Open
Abstract
UK Biobank is a large-scale prospective study with deep phenotyping and genomic data. Its open-access policy allows researchers worldwide, from academia or industry, to perform health research in the public interest. Between 2006 and 2010, the study recruited 502,000 adults aged 40-69 years from the general population of the United Kingdom. At enrolment, participants provided information on a wide range of factors, physical measurements were taken, and biological samples (blood, urine and saliva) were collected for long-term storage. Participants have now been followed up for over a decade with more than 52,000 incident cancer cases recorded. The study continues to be enhanced with repeat assessments, web-based questionnaires, multi-modal imaging, and conversion of the stored biological samples to genomic and other '-omic' data. The study has already demonstrated its value in enabling research into the determinants of cancer, and future planned enhancements will make the resource even more valuable to cancer researchers. Over 26,000 researchers worldwide are currently using the data, performing a wide range of cancer research. UK Biobank is uniquely placed to transform our understanding of the causes of cancer development and progression, and drive improvements in cancer treatment and prevention over the coming decades.
Collapse
Affiliation(s)
- Megan C Conroy
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK.
| | - Ben Lacey
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | - Jelena Bešević
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | - Wemimo Omiyale
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | - Qi Feng
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | | | | | | | | | | | - John Busby
- UK Biobank, Stockport, Greater Manchester, UK
| | - Rory Collins
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
- UK Biobank, Stockport, Greater Manchester, UK
| | - Naomi E Allen
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| |
Collapse
|
83
|
Gomes Gonçalves N, Vidal Ferreira N, Khandpur N, Martinez Steele E, Bertazzi Levy R, Andrade Lotufo P, Bensenor IM, Caramelli P, Alvim de Matos SM, Marchioni DM, Suemoto CK. Association Between Consumption of Ultraprocessed Foods and Cognitive Decline. JAMA Neurol 2023; 80:142-150. [PMID: 36469335 PMCID: PMC9857155 DOI: 10.1001/jamaneurol.2022.4397] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/07/2022] [Indexed: 12/12/2022]
Abstract
Importance Although consumption of ultraprocessed food has been linked to higher risk of cardiovascular disease, metabolic syndrome, and obesity, little is known about the association of consumption of ultraprocessed foods with cognitive decline. Objective To investigate the association between ultraprocessed food consumption and cognitive decline in the Brazilian Longitudinal Study of Adult Health. Design, Setting, and Participants This was a multicenter, prospective cohort study with 3 waves, approximately 4 years apart, from 2008 to 2017. Data were analyzed from December 2021 to May 2022. Participants were public servants aged 35 to 74 years old recruited in 6 Brazilian cities. Participants who, at baseline, had incomplete food frequency questionnaire, cognitive, or covariate data were excluded. Participants who reported extreme calorie intake (<600 kcal/day or >6000 kcal/day) and those taking medication that could negatively interfere with cognitive performance were also excluded. Exposures Daily ultraprocessed food consumption as a percentage of total energy divided into quartiles. Main Outcomes and Measures Changes in cognitive performance over time evaluated by the immediate and delayed word recall, word recognition, phonemic and semantic verbal fluency tests, and Trail-Making Test B version. Results A total of 15 105 individuals were recruited and 4330 were excluded, leaving 10 775 participants whose data were analyzed. The mean (SD) age at the baseline was 51.6 (8.9) years, 5880 participants (54.6%) were women, 5723 (53.1%) were White, and 6106 (56.6%) had at least a college degree. During a median (range) follow-up of 8 (6-10) years, individuals with ultraprocessed food consumption above the first quartile showed a 28% faster rate of global cognitive decline (β = -0.004; 95% CI, -0.006 to -0.001; P = .003) and a 25% faster rate of executive function decline (β = -0.003, 95% CI, -0.005 to 0.000; P = .01) compared with those in the first quartile. Conclusions and Relevance A higher percentage of daily energy consumption of ultraprocessed foods was associated with cognitive decline among adults from an ethnically diverse sample. These findings support current public health recommendations on limiting ultraprocessed food consumption because of their potential harm to cognitive function.
Collapse
Affiliation(s)
| | - Naomi Vidal Ferreira
- Adventist University of São Paulo, Engenheiro Coelho, Brazil
- Division of Geriatrics, University of São Paulo Medical School, São Paulo, Brazil
| | - Neha Khandpur
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | | | - Renata Bertazzi Levy
- Department of Preventive Medicine, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Paulo Andrade Lotufo
- Center for Clinical and Epidemiological Research, Hospital Universitário, University of São Paulo, São Paulo, Brazil
| | - Isabela M. Bensenor
- Center for Clinical and Epidemiological Research, Hospital Universitário, University of São Paulo, São Paulo, Brazil
| | - Paulo Caramelli
- Behavioral and Cognitive Neurology Research Unit, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Dirce M. Marchioni
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
84
|
Balling M, Afzal S, Davey Smith G, Varbo A, Langsted A, Kamstrup PR, Nordestgaard BG. Elevated LDL Triglycerides and Atherosclerotic Risk. J Am Coll Cardiol 2023; 81:136-152. [PMID: 36631208 DOI: 10.1016/j.jacc.2022.10.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND It is unclear whether elevated low-density lipoprotein (LDL) triglycerides are associated with an increased risk of atherosclerotic cardiovascular disease (ASCVD). OBJECTIVES This study tested the hypothesis that elevated LDL triglycerides are associated with an increased risk of ASCVD and of each ASCVD component individually. METHODS The study investigators used the Copenhagen General Population Study, which measured LDL triglycerides in 38,081 individuals with a direct automated assay (direct LDL triglycerides) and in another 30,208 individuals with nuclear magnetic resonance (NMR) spectroscopy (NMR LDL triglycerides). Meta-analyses aggregated the present findings with previously reported results. RESULTS During a median follow-up of 3.0 and 9.2 years, respectively, 872 and 5,766 individuals in the 2 cohorts received a diagnosis of ASCVD. Per 0.1 mmol/L (9 mg/dL) higher direct LDL triglycerides, HRs were 1.26 (95% CI: 1.17-1.35) for ASCVD, 1.27 (95% CI: 1.16-1.39) for ischemic heart disease, 1.28 (95% CI: 1.11-1.48) for myocardial infarction, 1.22 (95% CI: 1.08-1.38) for ischemic stroke, and 1.38 (95% CI: 1.21-1.58) for peripheral artery disease. Corresponding HRs for NMR LDL triglycerides were 1.26 (95% CI: 1.20-1.33), 1.33 (95% CI: 1.25-1.41), 1.41 (95% CI: 1.31-1.52), 1.13 (95% CI: 1.05-1.23), and 1.26 (95% CI: 1.10-1.43), respectively. The foregoing results were not entirely statistically explained by apolipoprotein B levels. In meta-analyses for the highest quartile vs the lowest quartile of LDL triglycerides, random-effects risk ratios were 1.50 (95% CI: 1.35-1.66) for ASCVD (4 studies; 71,526 individuals; 8,576 events), 1.62 (95% CI: 1.37-1.93) for ischemic heart disease (6 studies; 107,538 individuals; 9,734 events), 1.30 (95% CI: 1.13-1.49) for ischemic stroke (4 studies; 78,026 individuals; 4,273 events), and 1.53 (95% CI: 1.29-1.81) for peripheral artery disease (4 studies; 107,511 individuals; 1,848 events). CONCLUSIONS Elevated LDL triglycerides were robustly associated with an increased risk of ASCVD and of each ASCVD component individually in 2 prospective cohort studies and in meta-analyses of previous and present studies combined.
Collapse
Affiliation(s)
- Mie Balling
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev and Gentofte Hospital, Herlev, Denmark; The Copenhagen General Population Study, Copenhagen University Hospital, Herlev and Gentofte Hospital, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Shoaib Afzal
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev and Gentofte Hospital, Herlev, Denmark; The Copenhagen General Population Study, Copenhagen University Hospital, Herlev and Gentofte Hospital, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, United Kingdom; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Anette Varbo
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev and Gentofte Hospital, Herlev, Denmark; The Copenhagen General Population Study, Copenhagen University Hospital, Herlev and Gentofte Hospital, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Langsted
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev and Gentofte Hospital, Herlev, Denmark; The Copenhagen General Population Study, Copenhagen University Hospital, Herlev and Gentofte Hospital, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pia R Kamstrup
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev and Gentofte Hospital, Herlev, Denmark; The Copenhagen General Population Study, Copenhagen University Hospital, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev and Gentofte Hospital, Herlev, Denmark; The Copenhagen General Population Study, Copenhagen University Hospital, Herlev and Gentofte Hospital, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
85
|
Karavasiloglou N, Hughes DJ, Murphy N, Schomburg L, Sun Q, Seher V, Rohrmann S, Weiderpass E, Tjønneland A, Olsen A, Overvad K, Boutron-Ruault MC, Mancini FR, Mahamat-Saleh Y, Kaaks R, Kuhn T, Schulze MB, Tumino R, Panico S, Masala G, Pala V, Sacerdote C, Derksen JWG, Skeie G, Hjartåker A, Lasheras C, Agudo A, Sánchez MJ, Chirlaque MD, Ardanaz E, Amiano P, Van Guelpen B, Gylling B, Bradbury KE, Papier K, Freisling H, Aglago EK, Cross AJ, Riboli E, Aune D, Gunter MJ, Jenab M. Prediagnostic serum calcium concentrations and risk of colorectal cancer development in 2 large European prospective cohorts. Am J Clin Nutr 2023; 117:33-45. [PMID: 36789942 PMCID: PMC10131343 DOI: 10.1016/j.ajcnut.2022.10.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/11/2022] [Accepted: 10/28/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Higher dietary calcium consumption is associated with lower colorectal cancer (CRC) risk. However, little data are available on the association between circulating calcium concentrations and CRC risk. OBJECTIVES To explore the association between circulating calcium concentrations and CRC risk using data from 2 large European prospective cohort studies. METHODS Conditional logistic regression models were used to calculate multivariable-adjusted ORs and 95% CIs in case-control studies nested within the European Prospective Investigation into Cancer and Nutrition (EPIC; n-cases = 947, n-controls = 947) and the UK Biobank (UK-BB; n-cases = 2759, n-controls = 12,021) cohorts. RESULTS In EPIC, nonalbumin-adjusted total serum calcium (a proxy of free calcium) was not associated with CRC (OR: 0.94; 95% CI: 0.85, 1.03; modeled as continuous variable, per 1 mg/dL increase), colon cancer (OR: 0.93; 95% CI: 0.82, 1.05) or rectal cancer (OR: 1.01; 95% CI: 0.84, 1.20) risk in the multivariable adjusted model. In the UK-BB, serum ionized calcium (free calcium, most active form) was inversely associated with the risk of CRC (OR: 0.85; 95% CI: 0.76, 0.95; per 1 mg/dL) and colon cancer (OR: 0.78; 95% CI: 0.68, 0.90), but not rectal cancer (OR: 1.02; 95% CI: 0.83, 1.24) in multivariable adjusted models. Meta-analysis of EPIC and UK-BB CRC risk estimates showed an inverse risk association for CRC in the multivariable adjusted model (OR: 0.90; 95%CI: 0.84, 0.97). In analyses by quintiles, in both cohorts, higher levels of serum calcium were associated with reduced CRC risk (EPIC: ORQ5vs.Q1: 0.69; 95% CI: 0.47, 1.00; P-trend = 0.03; UK-BB: ORQ5vs.Q1: 0.82; 95% CI: 0.72, 0.94; P-trend < 0.01). Analyses by anatomical subsite showed an inverse cancer risk association in the colon (EPIC: ORQ5vs.Q1: 0.63, 95% CI: 0.39, 1.02; P-trend = 0.05; UK-BB: ORQ5vs.Q1: 0.75; 95% CI: 0.64, 0.88; P-trend < 0.01) but not the rectum. CONCLUSIONS In UK-BB, higher serum ionized calcium levels were inversely associated with CRC, but the risk was restricted to the colon. Total serum calcium showed a null association in EPIC. Additional prospective studies in other populations are needed to better investigate these associations.
Collapse
Affiliation(s)
- Nena Karavasiloglou
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France; Division of Chronic Disease Epidemiology, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - David J Hughes
- Cancer Biology and Therapeutics Group, UCD Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Neil Murphy
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Lutz Schomburg
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Qian Sun
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Vartiter Seher
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sabine Rohrmann
- Division of Chronic Disease Epidemiology, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Elisabete Weiderpass
- Office of the Director, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Anne Tjønneland
- Danish Cancer Society Research Center, Diet, Genes and Environment, Copenhagen, Denmark; University of Copenhagen, Department of Public Health, Copenhagen, Denmark
| | - Anja Olsen
- Danish Cancer Society Research Center, Diet, Genes and Environment, Copenhagen, Denmark; University of Århus, Department of Public Health, Århus, Denmark
| | - Kim Overvad
- University of Århus, Department of Public Health, Århus, Denmark
| | - Marie-Christine Boutron-Ruault
- CESP, Fac. de médecine - Univ. Paris-Sud, Fac. de médecine - UVSQ, INSERM, Université Paris-Saclay, Villejuif, France; Gustave Roussy, Villejuif, France
| | - Francesca Romana Mancini
- CESP, Fac. de médecine - Univ. Paris-Sud, Fac. de médecine - UVSQ, INSERM, Université Paris-Saclay, Villejuif, France; Gustave Roussy, Villejuif, France
| | - Yahya Mahamat-Saleh
- CESP, Fac. de médecine - Univ. Paris-Sud, Fac. de médecine - UVSQ, INSERM, Université Paris-Saclay, Villejuif, France; Gustave Roussy, Villejuif, France
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tilman Kuhn
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany; School of Biological Sciences, Queens University of Belfast, Belfast, Northern Ireland
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Rosario Tumino
- Hyblean Association for Epidemiology Research, AIRE-ONLUS Ragusa, Italy
| | - Salvatore Panico
- Dipartimento Di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | - Giovanna Masala
- Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Valeria Pala
- Epidemiology and Prevention Unit Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Città della Salute e della Scienza University-Hospital, Turin, Italy
| | - Jeroen W G Derksen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Guri Skeie
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, The Arctic University of Norway, Norway
| | - Anette Hjartåker
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Cristina Lasheras
- Department of Functional Biology, Faculty of Medicine, University of Oviedo, Asturias, Spain
| | - Antonio Agudo
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Maria-José Sánchez
- Escuela Andaluza de Salud Pública (EASP), Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain; Department of Preventive Medicine and Public Health, University of Granada, Granada, Spain
| | - Maria-Dolores Chirlaque
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain; Department of Epidemiology, Regional Health Council, IMIB-Arrixaca, Murcia University, Spain
| | - Eva Ardanaz
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain; Navarra Public Health Institute, Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Pilar Amiano
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain; Ministry of Health of the Basque Government, Sub Directorate for Public Health and Addictions of Gipuzkoa; Biodonostia Health Research Institute, Epidemiology of Chronic and Communicable Diseases Group, San Sebastián, Spain
| | - Bethany Van Guelpen
- Department of Radiation Sciences, Oncology Umeå University, Umeå, Sweden; Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Björn Gylling
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Kathryn E Bradbury
- National Institute for Health Innovation, The University of Auckland, New Zealand
| | - Keren Papier
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Heinz Freisling
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Elom K Aglago
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France; Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
| | - Amanda J Cross
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
| | - Elio Riboli
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
| | - Dagfinn Aune
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
| | - Marc J Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Mazda Jenab
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France.
| |
Collapse
|
86
|
Pillay P, Lewington S, Taylor H, Lacey B, Carter J. Adiposity, Body Fat Distribution, and Risk of Major Stroke Types Among Adults in the United Kingdom. JAMA Netw Open 2022; 5:e2246613. [PMID: 36515951 PMCID: PMC9856404 DOI: 10.1001/jamanetworkopen.2022.46613] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
IMPORTANCE Uncertainty persists regarding the independent association of general and central adiposity with major stroke types. OBJECTIVE To determine the independent associations of general and central adiposity with risk of ischemic stroke, intracerebral hemorrhage, and subarachnoid hemorrhage. DESIGN, SETTING, AND PARTICIPANTS Between 2006 and 2010, adults without prior stroke at recruitment in England, Scotland, and Wales were recruited into the UK Biobank prospective cohort study and were followed up through linkage with electronic medical records. Data were analyzed from September 2021 to September 2022. EXPOSURES General adiposity (body mass index [BMI] calculated as weight in kilograms divided by height in meters squared) and central adiposity (waist circumference). MAIN OUTCOMES AND MEASURES Incident ischemic stroke, intracerebral hemorrhage, and subarachnoid hemorrhage as recorded through record linkage with electronic hospital records. Cox regression estimated adjusted hazard ratios (HRs) by stroke type. RESULTS Among 490 071 participants, the mean (SD) age was 56.5 (8.1) years, 267 579 (54.6%) were female, and 461 647 (94.2%) were White. During a median (IQR) of 12 (11.2-12.7) years follow-up, 7117 incident ischemic strokes, 1391 intracerebral hemorrhages, and 834 subarachnoid hemorrhages were identified. After mutual adjustment for waist circumference, BMI had no independent association with ischemic stroke (HR per 5-unit higher BMI, 1.04; 95% CI, 0.97-1.11), but was inversely associated with intracerebral hemorrhage (HR, 0.85; 95% CI, 0.74-0.96) and subarachnoid hemorrhage (HR, 0.82; 95% CI, 0.69-0.96). Waist circumference (adjusted for BMI) was positively associated with ischemic stroke (HR per 10-cm higher waist circumference, 1.19; 95% CI, 1.13-1.25) and intracerebral hemorrhage (HR, 1.17; 95% CI, 1.05-1.30), but was not associated with subarachnoid hemorrhage (HR, 1.07; 95% CI, 0.93-1.22). CONCLUSIONS AND RELEVANCE In this large-scale prospective study, the independent and contrasting associations of BMI and waist circumference with stroke types suggests the importance of considering body fat distribution to stroke risk. Waist circumference was positively associated with the risk of ischemic stroke and intracerebral hemorrhage, while BMI was inversely associated with the risk of subarachnoid hemorrhage and intracerebral hemorrhage. This study also suggests that some adverse correlate of low BMI may be associated with an increased risk of intracerebral hemorrhage and subarachnoid hemorrhage, which warrants further investigation.
Collapse
Affiliation(s)
- Preyanka Pillay
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, Big Data Institute, University of Oxford, Oxford, United Kingdom
| | - Sarah Lewington
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, Big Data Institute, University of Oxford, Oxford, United Kingdom
- Medical Research Council Population Health Research Unit, Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Hannah Taylor
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, Big Data Institute, University of Oxford, Oxford, United Kingdom
| | - Ben Lacey
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, Big Data Institute, University of Oxford, Oxford, United Kingdom
| | - Jennifer Carter
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, Big Data Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
87
|
Staplin N, Herrington WG, Murgia F, Ibrahim M, Bull KR, Judge PK, Ng SYA, Turner M, Zhu D, Emberson J, Landray MJ, Baigent C, Haynes R, Hopewell JC. Determining the Relationship Between Blood Pressure, Kidney Function, and Chronic Kidney Disease: Insights From Genetic Epidemiology. Hypertension 2022; 79:2671-2681. [PMID: 36082669 PMCID: PMC9640248 DOI: 10.1161/hypertensionaha.122.19354] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND It is well established that decreased kidney function can increase blood pressure (BP), but it is unproven whether moderately elevated BP causes chronic kidney disease (CKD) or glomerular hyperfiltration. METHODS 311 119 White British UK Biobank participants were included in logistic regression analyses to estimate the odds of CKD (defined as long-term kidney replacement therapy, estimated glomerular filtration rate [eGFR]< 60mL/min/1.73m2, or urinary albumin:creatinine ratio ≥3 mg/mmol) associated with higher genetically predicted BP using genetic risk scores comprising 219 systolic and 223 diastolic BP loci. Analyses estimating associations with clinical categories of eGFR and urinary albumin:creatinine ratio were also conducted, with an eGFR ≥120 mL (min·1.73m2) considered evidence of glomerular hyperfiltration. RESULTS 21 623 participants had CKD: 7781 with reduced eGFR and 15 500 with albuminuria. 1828 participants had an eGFR ≥120 mL/min/1.73m2. Each genetically predicted 10 mmHg higher systolic BP and 5 mmHg higher diastolic BP were associated with a 37% (95% CI, 1.29-1.45) and 19% (1.14-1.25) higher odds of CKD, respectively. Associations were evident for both the reduced eGFR and albuminuria components of the CKD outcome. The odds of hyperfiltration (versus an eGFR ≥60 and <90 mL/min/1.73m2 were 49% higher (95% CI, 1.21-1.84) for each genetically predicted 10 mmHg higher systolic BP. Associations with CKD and hyperfiltration were similar irrespective of preexisting diabetes, vascular disease, or different levels of adiposity. CONCLUSIONS In this general population, genetic epidemiological evidence supports a causal role of life-long differences in BP for decreased kidney function, glomerular hyperfiltration, and albuminuria. Physiological autoregulation may not afford complete renal protection against the moderate BP elevations.
Collapse
Affiliation(s)
- Natalie Staplin
- Medical Research Council Population Health Research Unit at the University of Oxford, Nuffield Department of Population Health (NDPH), United Kingdom (N.S., W.G.H., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.).,Clinical Trial Service Unit and Epidemiological Studies Unit, NDPH (N.S., W.G.H., F.M., M.I., P.J., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.), University of Oxford, Oxford, United Kingdom.,Big Data Institute, Li Ka Shing Centre for Health Information and Discovery (N.S., F.M., M.I., J.E., M.J.L., J.C.H.), University of Oxford, Oxford, United Kingdom
| | - William G Herrington
- Medical Research Council Population Health Research Unit at the University of Oxford, Nuffield Department of Population Health (NDPH), United Kingdom (N.S., W.G.H., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.).,Clinical Trial Service Unit and Epidemiological Studies Unit, NDPH (N.S., W.G.H., F.M., M.I., P.J., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.), University of Oxford, Oxford, United Kingdom.,Health Data Research UK (W.G.H., M.J.L.), University of Oxford, Oxford, United Kingdom.,Oxford Kidney Unit, Churchill Hospital, Oxford, United Kingdom (W.G.H., K.R.B., P.K.J., M.T., D.Z., R.H.)
| | - Federico Murgia
- Clinical Trial Service Unit and Epidemiological Studies Unit, NDPH (N.S., W.G.H., F.M., M.I., P.J., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.), University of Oxford, Oxford, United Kingdom.,Big Data Institute, Li Ka Shing Centre for Health Information and Discovery (N.S., F.M., M.I., J.E., M.J.L., J.C.H.), University of Oxford, Oxford, United Kingdom
| | - Maysson Ibrahim
- Clinical Trial Service Unit and Epidemiological Studies Unit, NDPH (N.S., W.G.H., F.M., M.I., P.J., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.), University of Oxford, Oxford, United Kingdom.,Big Data Institute, Li Ka Shing Centre for Health Information and Discovery (N.S., F.M., M.I., J.E., M.J.L., J.C.H.), University of Oxford, Oxford, United Kingdom
| | - Katherine R Bull
- Nuffield Department of Medicine (K.R.B.), University of Oxford, Oxford, United Kingdom.,Oxford Kidney Unit, Churchill Hospital, Oxford, United Kingdom (W.G.H., K.R.B., P.K.J., M.T., D.Z., R.H.)
| | - Parminder K Judge
- Clinical Trial Service Unit and Epidemiological Studies Unit, NDPH (N.S., W.G.H., F.M., M.I., P.J., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.), University of Oxford, Oxford, United Kingdom.,Oxford Kidney Unit, Churchill Hospital, Oxford, United Kingdom (W.G.H., K.R.B., P.K.J., M.T., D.Z., R.H.)
| | - Sarah Y A Ng
- Medical Research Council Population Health Research Unit at the University of Oxford, Nuffield Department of Population Health (NDPH), United Kingdom (N.S., W.G.H., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.).,Clinical Trial Service Unit and Epidemiological Studies Unit, NDPH (N.S., W.G.H., F.M., M.I., P.J., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.), University of Oxford, Oxford, United Kingdom
| | - Michael Turner
- Medical Research Council Population Health Research Unit at the University of Oxford, Nuffield Department of Population Health (NDPH), United Kingdom (N.S., W.G.H., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.).,Clinical Trial Service Unit and Epidemiological Studies Unit, NDPH (N.S., W.G.H., F.M., M.I., P.J., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.), University of Oxford, Oxford, United Kingdom.,Oxford Kidney Unit, Churchill Hospital, Oxford, United Kingdom (W.G.H., K.R.B., P.K.J., M.T., D.Z., R.H.)
| | - Doreen Zhu
- Medical Research Council Population Health Research Unit at the University of Oxford, Nuffield Department of Population Health (NDPH), United Kingdom (N.S., W.G.H., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.).,Clinical Trial Service Unit and Epidemiological Studies Unit, NDPH (N.S., W.G.H., F.M., M.I., P.J., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.), University of Oxford, Oxford, United Kingdom.,Oxford Kidney Unit, Churchill Hospital, Oxford, United Kingdom (W.G.H., K.R.B., P.K.J., M.T., D.Z., R.H.)
| | - Jonathan Emberson
- Medical Research Council Population Health Research Unit at the University of Oxford, Nuffield Department of Population Health (NDPH), United Kingdom (N.S., W.G.H., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.).,Clinical Trial Service Unit and Epidemiological Studies Unit, NDPH (N.S., W.G.H., F.M., M.I., P.J., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.), University of Oxford, Oxford, United Kingdom.,Big Data Institute, Li Ka Shing Centre for Health Information and Discovery (N.S., F.M., M.I., J.E., M.J.L., J.C.H.), University of Oxford, Oxford, United Kingdom
| | - Martin J Landray
- Medical Research Council Population Health Research Unit at the University of Oxford, Nuffield Department of Population Health (NDPH), United Kingdom (N.S., W.G.H., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.).,Clinical Trial Service Unit and Epidemiological Studies Unit, NDPH (N.S., W.G.H., F.M., M.I., P.J., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.), University of Oxford, Oxford, United Kingdom.,Big Data Institute, Li Ka Shing Centre for Health Information and Discovery (N.S., F.M., M.I., J.E., M.J.L., J.C.H.), University of Oxford, Oxford, United Kingdom.,Health Data Research UK (W.G.H., M.J.L.), University of Oxford, Oxford, United Kingdom.,National Institute for Health Research Oxford Biomedical Research Centre (M.J.L.), University of Oxford, Oxford, United Kingdom
| | - Colin Baigent
- Medical Research Council Population Health Research Unit at the University of Oxford, Nuffield Department of Population Health (NDPH), United Kingdom (N.S., W.G.H., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.).,Clinical Trial Service Unit and Epidemiological Studies Unit, NDPH (N.S., W.G.H., F.M., M.I., P.J., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.), University of Oxford, Oxford, United Kingdom
| | - Richard Haynes
- Medical Research Council Population Health Research Unit at the University of Oxford, Nuffield Department of Population Health (NDPH), United Kingdom (N.S., W.G.H., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.).,Clinical Trial Service Unit and Epidemiological Studies Unit, NDPH (N.S., W.G.H., F.M., M.I., P.J., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.), University of Oxford, Oxford, United Kingdom.,Oxford Kidney Unit, Churchill Hospital, Oxford, United Kingdom (W.G.H., K.R.B., P.K.J., M.T., D.Z., R.H.)
| | - Jemma C Hopewell
- Medical Research Council Population Health Research Unit at the University of Oxford, Nuffield Department of Population Health (NDPH), United Kingdom (N.S., W.G.H., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.).,Clinical Trial Service Unit and Epidemiological Studies Unit, NDPH (N.S., W.G.H., F.M., M.I., P.J., S.Y.A.N., M.T., D.Z., J.E., M.J.L., C.B., R.H., J.C.H.), University of Oxford, Oxford, United Kingdom.,Big Data Institute, Li Ka Shing Centre for Health Information and Discovery (N.S., F.M., M.I., J.E., M.J.L., J.C.H.), University of Oxford, Oxford, United Kingdom
| |
Collapse
|
88
|
Bjørnestad EØ, Dhar I, Svingen GFT, Pedersen ER, Ørn S, Svenningsson MM, Tell GS, Ueland PM, Sulo G, Laaksonen R, Nygård O. Circulating trimethylamine N-oxide levels do not predict 10-year survival in patients with or without coronary heart disease. J Intern Med 2022; 292:915-924. [PMID: 35916742 PMCID: PMC9804190 DOI: 10.1111/joim.13550] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Trimethylamine N-oxide (TMAO) is an amine oxide generated by gut microbial metabolism. TMAO may contribute to atherothrombosis and systemic inflammation. However, the prognostic value of circulating TMAO for risk stratification is uncertain. METHODS We assessed prospective relationships of plasma TMAO with long-term risk of all-cause, cardiovascular (CV), and non-CV mortality in the Western Norway Coronary Angiography Cohort (WECAC; 4132 patients with suspected coronary artery disease) and the Hordaland Health Study (HUSK; 6393 community-based subjects). Risk associations were examined using Cox regression analyses. RESULTS Mean follow-up was 9.8 and 10.5 years in WECAC and HUSK, respectively. Following adjustments for established CV risk factors and indices of renal function in WECAC, the hazard ratios (HRs) (95% confidence intervals [CIs]) per one standard deviation increase in log-transformed plasma TMAO were 1.04 (0.97-1.12), 1.06 (0.95-1.18), and 1.03 (0.93-1.13) for all-cause, CV, and non-CV mortality, respectively. Essentially similar results were obtained in patients with angiographically significant coronary artery disease and patients with reduced left ventricular ejection fraction. Corresponding HRs (95% CIs) in the HUSK cohort were 1.03 (0.96-1.10), 1.01 (0.89-1.13), and 1.03 (0.95-1.12) for all-cause-, CV, and non-CV mortality, respectively. CONCLUSIONS Circulating TMAO did not predict long-term all-cause, CV, or non-CV mortality in patients with coronary heart disease or in community-based adults. This large study does not support a role of TMAO for patient risk stratification in primary or secondary prevention.
Collapse
Affiliation(s)
- Espen Ø Bjørnestad
- Department of Cardiology, Stavanger University Hospital, Stavanger, Norway
| | - Indu Dhar
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Gard F T Svingen
- Department of Cardiology, Haukeland University Hospital, Bergen, Norway
| | - Eva R Pedersen
- Department of Cardiology, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Stein Ørn
- Department of Cardiology, Stavanger University Hospital, Stavanger, Norway
| | | | - Grethe S Tell
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Per M Ueland
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Gerhard Sulo
- Centre for Disease Burden, Norwegian Institute of Public Health, Bergen, Norway
| | - Reijo Laaksonen
- Finnish Cardiovascular Research Center, University of Tampere, Tampere, Finland
| | - Ottar Nygård
- Department of Cardiology, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
89
|
Li Y, Gao R, Zhao B, Zhang Y. Low Serum Bicarbonate Levels Increase the Risk of All-Cause, Cardiovascular Disease, and Cancer Mortality in Type 2 Diabetes. J Clin Endocrinol Metab 2022; 107:3055-3065. [PMID: 36066477 PMCID: PMC9681608 DOI: 10.1210/clinem/dgac504] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT The evidence regarding bicarbonate status and mortality among diabetes is scarce. OBJECTIVE The purpose of this study was to investigate the associations of bicarbonate concentrations with risk of all-cause, cardiovascular disease (CVD), and cancer mortality among patients with type 2 diabetes (T2D). METHODS This study included 8163 adult diabetic patients from the National Health and Nutrition Examination Survey (NHANES), 1999 to 2018. Death outcomes were ascertained by linkage to National Death Index records through 31 December 2019. The Cox proportional-risk model was used to estimate hazard ratios (HR) and 95% CIs for mortality from all causes, CVD, and cancer. The mediating effects of 11 metabolic, cardiovascular, and renal biomarkers were evaluated using a logistic regression model within a counterfactual framework. RESULTS During 8163 person-years of follow-up, 2310 deaths were documented, including 659 CVD deaths and 399 cancer deaths. After multivariate adjustment, lower serum bicarbonate levels were significantly linearly correlated with higher all-cause, CVD, and cancer mortality: The risk of all-cause death increased by 40%, the risk of CVD death increased by 48%, and the risk of cancer death increased by 84% compared with the normal group (all P < .05). Altered levels of estimated glomerular filtration rate explained 12.10% and 16.94% of the relation between serum bicarbonate with all-cause and CVD mortality, respectively. Total cholesterol mediated 4.70% and 10.51% of the associations of all-cause and CVD mortality, respectively. CONCLUSION Lower serum bicarbonate concentrations were significantly associated with higher all-cause, CVD, and cancer mortality. These findings suggest that maintaining adequate bicarbonate status may lower mortality risk in individuals with T2D.
Collapse
Affiliation(s)
- Yilan Li
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Nangang, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, Nangang, China
| | - Rong Gao
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Nangang, China
| | - Bing Zhao
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Nangang, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, Nangang, China
| | - Yao Zhang
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Nangang, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, Nangang, China
| |
Collapse
|
90
|
Matthews K, Dawes P, Elliot R, Maharani A, Pendleton N, Tampubolon G. Allostatic load and risk of hearing impairment. Brain Behav Immun Health 2022; 25:100496. [PMID: 36061925 PMCID: PMC9429496 DOI: 10.1016/j.bbih.2022.100496] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/28/2022] [Accepted: 08/08/2022] [Indexed: 11/26/2022] Open
Abstract
Background Methods Results Conclusions Allostatic load captures cumulative physiological stress which has consequence for subsequent hearing function. Prolonged high allostatic load was associated with risk of objectively measured hearing impairment. This association is shown in nationally representative longitudinal ageing study.
Collapse
|
91
|
Liu Z, Suo C, Jiang Y, Zhao R, Zhang T, Jin L, Chen X. Phenome-Wide Association Analysis Reveals Novel Links Between Genetically Determined Levels of Liver Enzymes and Disease Phenotypes. PHENOMICS (CHAM, SWITZERLAND) 2022; 2:295-311. [PMID: 36939802 PMCID: PMC9590558 DOI: 10.1007/s43657-021-00033-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/10/2021] [Accepted: 10/15/2021] [Indexed: 11/26/2022]
Abstract
Serum liver enzymes (alanine aminotransferase [ALT], aspartate aminotransferase [AST], λ-glutamyl transferase [GGT] and alkaline phosphatase [ALP]) are the leading biomarkers to measure liver injury, and they have been reported to be associated with several intrahepatic and extrahepatic diseases in observational studies. We conducted a phenome-wide association study (PheWAS) to identify disease phenotypes associated with genetically predicted liver enzymes based on the UK Biobank cohort. Univariable and multivariable Mendelian randomization (MR) analyses were performed to obtain the causal estimates of associations that detected in PheWAS. Our PheWAS identified 40 out of 1,376 pairs (16, 17, three and four pairs for ALT, AST, GGT and ALP, respectively) of genotype-phenotype associations reaching statistical significance at the 5% false discovery rate threshold. A total of 34 links were further validated in Mendelian randomization analyses. Most of the disease phenotypes that associated with genetically determined ALT level were liver-related, including primary liver cancer and alcoholic liver damage. The disease outcomes associated with genetically determined AST involved a wide range of phenotypic categories including endocrine/metabolic diseases, digestive diseases, and neurological disorder. Genetically predicted GGT level was associated with the risk of other chronic non-alcoholic liver disease, abnormal results of function study of liver, and cholelithiasis. Genetically determined ALP level was associated with pulmonary heart disease, phlebitis and thrombophlebitis of lower extremities, and hypercholesterolemia. Our findings reveal novel links between liver enzymes and disease phenotypes providing insights into the full understanding of the biological roles of liver enzymes. Supplementary Information The online version contains supplementary material available at 10.1007/s43657-021-00033-y.
Collapse
Affiliation(s)
- Zhenqiu Liu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, 200438 China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, 225316 China
| | - Chen Suo
- Fudan University Taizhou Institute of Health Sciences, Taizhou, 225316 China
- Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, 200032 China
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032 China
| | - Yanfeng Jiang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, 200438 China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, 225316 China
| | - Renjia Zhao
- Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, 200032 China
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032 China
| | - Tiejun Zhang
- Fudan University Taizhou Institute of Health Sciences, Taizhou, 225316 China
- Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, 200032 China
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032 China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, 200438 China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, 225316 China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, 200438 China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, 225316 China
| |
Collapse
|
92
|
Watts EL, Perez‐Cornago A, Fensom GK, Smith‐Byrne K, Noor U, Andrews CD, Gunter MJ, Holmes MV, Martin RM, Tsilidis KK, Albanes D, Barricarte A, Bueno‐de‐Mesquita B, Chen C, Cohn BA, Dimou NL, Ferrucci L, Flicker L, Freedman ND, Giles GG, Giovannucci EL, Goodman GE, Haiman CA, Hankey GJ, Huang J, Huang W, Hurwitz LM, Kaaks R, Knekt P, Kubo T, Langseth H, Laughlin G, Le Marchand L, Luostarinen T, MacInnis RJ, Mäenpää HO, Männistö S, Metter EJ, Mikami K, Mucci LA, Olsen AW, Ozasa K, Palli D, Penney KL, Platz EA, Rissanen H, Sawada N, Schenk JM, Stattin P, Tamakoshi A, Thysell E, Tsai CJ, Tsugane S, Vatten L, Weiderpass E, Weinstein SJ, Wilkens LR, Yeap BB, Allen NE, Key TJ, Travis RC. Circulating free testosterone and risk of aggressive prostate cancer: Prospective and Mendelian randomisation analyses in international consortia. Int J Cancer 2022; 151:1033-1046. [PMID: 35579976 PMCID: PMC7613289 DOI: 10.1002/ijc.34116] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/18/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022]
Abstract
Previous studies had limited power to assess the associations of testosterone with aggressive disease as a primary endpoint. Further, the association of genetically predicted testosterone with aggressive disease is not known. We investigated the associations of calculated free and measured total testosterone and sex hormone-binding globulin (SHBG) with aggressive, overall and early-onset prostate cancer. In blood-based analyses, odds ratios (OR) and 95% confidence intervals (CI) for prostate cancer were estimated using conditional logistic regression from prospective analysis of biomarker concentrations in the Endogenous Hormones, Nutritional Biomarkers and Prostate Cancer Collaborative Group (up to 25 studies, 14 944 cases and 36 752 controls, including 1870 aggressive prostate cancers). In Mendelian randomisation (MR) analyses, using instruments identified using UK Biobank (up to 194 453 men) and outcome data from PRACTICAL (up to 79 148 cases and 61 106 controls, including 15 167 aggressive cancers), ORs were estimated using the inverse-variance weighted method. Free testosterone was associated with aggressive disease in MR analyses (OR per 1 SD = 1.23, 95% CI = 1.08-1.40). In blood-based analyses there was no association with aggressive disease overall, but there was heterogeneity by age at blood collection (OR for men aged <60 years 1.14, CI = 1.02-1.28; Phet = .0003: inverse association for older ages). Associations for free testosterone were positive for overall prostate cancer (MR: 1.20, 1.08-1.34; blood-based: 1.03, 1.01-1.05) and early-onset prostate cancer (MR: 1.37, 1.09-1.73; blood-based: 1.08, 0.98-1.19). SHBG and total testosterone were inversely associated with overall prostate cancer in blood-based analyses, with null associations in MR analysis. Our results support free testosterone, rather than total testosterone, in the development of prostate cancer, including aggressive subgroups.
Collapse
Affiliation(s)
- Eleanor L. Watts
- Cancer Epidemiology Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Aurora Perez‐Cornago
- Cancer Epidemiology Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Georgina K. Fensom
- Cancer Epidemiology Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Karl Smith‐Byrne
- Genomic Epidemiology BranchInternational Agency for Research on CancerLyonFrance
| | - Urwah Noor
- Cancer Epidemiology Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Colm D. Andrews
- Cancer Epidemiology Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Marc J. Gunter
- Section of Nutrition and MetabolismInternational Agency for Research on CancerLyonFrance
| | - Michael V. Holmes
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population HealthUniversity of OxfordOxfordUK
- Medical Research Council Population Health Research Unit at the University of OxfordOxfordUK
| | - Richard M. Martin
- Department of Population Health Sciences, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUK
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUK
- National Institute for Health Research (NIHR) Bristol Biomedical Research CentreUniversity Hospitals Bristol NHS Foundation Trust and Weston NHS Foundation Trust and the University of BristolBristolUK
| | - Konstantinos K. Tsilidis
- Department of Epidemiology and Biostatistics, School of Public HealthImperial College LondonLondonUK
- Department of Hygiene and EpidemiologyUniversity of Ioannina School of MedicineIoanninaGreece
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Aurelio Barricarte
- Navarra Public Health InstitutePamplonaSpain
- Navarra Institute for Health Research (IdiSNA)PamplonaSpain
- CIBER Epidemiology and Public Health CIBERESPMadridSpain
| | - Bas Bueno‐de‐Mesquita
- Centre for Nutrition, Prevention and Health ServicesNational Institute for Public Health and the Environment (RIVM)The Netherlands
| | - Chu Chen
- Program in Epidemiology, Division of Public Health SciencesFred Hutchinson Cancer Research CenterSeattleWashingtonUSA
- Department of Epidemiology, School of Public HealthUniversity of WashingtonSeattleWashingtonUSA
- Department of Otolaryngology: Head and Neck Surgery, School of MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Barbara A. Cohn
- Child Health and Development StudiesPublic Health InstituteBerkeleyCaliforniaUSA
| | - Niki L. Dimou
- Section of Nutrition and MetabolismInternational Agency for Research on CancerLyonFrance
| | | | - Leon Flicker
- Medical SchoolUniversity of Western AustraliaPerthWestern AustraliaAustralia
- Western Australian Centre for Health and AgeingUniversity of Western AustraliaPerthWestern AustraliaAustralia
| | - Neal D. Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Graham G. Giles
- Cancer Epidemiology DivisionCancer Council VictoriaMelbourneVictoriaAustralia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global HealthThe University of MelbourneMelbourneVictoriaAustralia
- Precision Medicine, School of Clinical Sciences at Monash HealthMonash UniversityMelbourneVictoriaAustralia
| | - Edward L. Giovannucci
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
- Channing Division of Network MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
- Department of NutritionHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Gary E. Goodman
- Program in Epidemiology, Division of Public Health SciencesFred Hutchinson Cancer Research CenterSeattleWashingtonUSA
| | - Christopher A. Haiman
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of MedicineUniversity of Southern California/Norris Comprehensive Cancer CenterLos AngelesCaliforniaUSA
| | - Graeme J. Hankey
- Medical SchoolUniversity of Western AustraliaPerthWestern AustraliaAustralia
| | - Jiaqi Huang
- Division of Cancer Epidemiology and Genetics, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and EndocrinologyThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Wen‐Yi Huang
- Division of Cancer Epidemiology and Genetics, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Lauren M. Hurwitz
- Division of Cancer Epidemiology and Genetics, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Rudolf Kaaks
- Division of Cancer EpidemiologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Paul Knekt
- Department of Public Health and WelfareNational Institute for Health and WelfareHelsinkiFinland
| | - Tatsuhiko Kubo
- Department of Public Health and Health Policy, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Hilde Langseth
- Department of Epidemiology and Biostatistics, School of Public HealthImperial College LondonLondonUK
- Department of ResearchCancer Registry of NorwayOsloNorway
| | - Gail Laughlin
- Herbert Wertheim School of Public Health and Human Longevity ScienceUniversity of California San DiegoSan DiegoCaliforniaUSA
| | | | - Tapio Luostarinen
- Finnish Cancer RegistryInstitute for Statistical and Epidemiological Cancer ResearchHelsinkiFinland
| | - Robert J. MacInnis
- Cancer Epidemiology DivisionCancer Council VictoriaMelbourneVictoriaAustralia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global HealthThe University of MelbourneMelbourneVictoriaAustralia
| | - Hanna O. Mäenpää
- Department of OncologyHelsinki University Central HospitalHelsinkiFinland
| | - Satu Männistö
- Department of Public Health and WelfareFinnish Institute for Health and WelfareHelsinkiFinland
| | - E. Jeffrey Metter
- Department of NeurologyThe University of Tennessee Health Science Center, College of MedicineMemphisTennesseeUSA
| | - Kazuya Mikami
- Departmemt of UrologyJapanese Red Cross Kyoto Daiichi HospitalKyotoJapan
| | - Lorelei A. Mucci
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Anja W. Olsen
- Department of Public HealthAarhus UniversityAarhusDenmark
- Danish Cancer SocietyResearch CenterCopenhagenDenmark
| | - Kotaro Ozasa
- Departmemt of EpidemiologyRadiation Effects Research FoundationHiroshimaJapan
| | - Domenico Palli
- Cancer Risk Factors and Life‐Style Epidemiology Unit, Institute for Cancer ResearchPrevention and Clinical Network – ISPROFlorenceItaly
| | - Kathryn L. Penney
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
- Channing Division of Network MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Elizabeth A. Platz
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Harri Rissanen
- Department of Public Health and WelfareNational Institute for Health and WelfareHelsinkiFinland
| | - Norie Sawada
- Epidemiology and Prevention Group, Center for Public Health SciencesNational Cancer CenterTokyoJapan
| | - Jeannette M. Schenk
- Cancer Prevention Program, Public Health Sciences DivisionFred Hutchinson Cancer Research CenterSeattleWashingtonUSA
| | - Pär Stattin
- Department of Surgical SciencesUppsala UniversityUppsalaSweden
| | | | - Elin Thysell
- Department of Medical BiosciencesUmeå UniversityUmeåSweden
| | - Chiaojung Jillian Tsai
- Department of Radiation OncologyMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Shoichiro Tsugane
- Epidemiology and Prevention Group, Center for Public Health SciencesNational Cancer CenterTokyoJapan
| | - Lars Vatten
- Department of Public Health and Nursing, Faculty of MedicineNorwegian University of Science and TechnologyTrondheimNorway
| | - Elisabete Weiderpass
- Director Office, International Agency for Research on CancerWorld Health OrganizationLyonFrance
| | - Stephanie J. Weinstein
- Division of Cancer Epidemiology and Genetics, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | | | - Bu B. Yeap
- Medical SchoolUniversity of Western AustraliaPerthWestern AustraliaAustralia
- Department of Endocrinology and DiabetesFiona Stanley HospitalPerthWestern AustraliaAustralia
| | | | | | | | | | | | - Naomi E. Allen
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population HealthUniversity of OxfordOxfordUK
- UK Biobank LtdStockportUK
| | - Timothy J. Key
- Cancer Epidemiology Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Ruth C. Travis
- Cancer Epidemiology Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| |
Collapse
|
93
|
Xiao Q, Cai B, Yin A, Huo H, Lan K, Zhou G, Shen L, He B. L-shaped association of serum 25-hydroxyvitamin D concentrations with cardiovascular and all-cause mortality in individuals with osteoarthritis: results from the NHANES database prospective cohort study. BMC Med 2022; 20:308. [PMID: 36127705 PMCID: PMC9490951 DOI: 10.1186/s12916-022-02510-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/02/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The relationship between vitamin D status and mortality in patients with osteoarthritis (OA) is unknown. This study investigated the associations of serum 25-hydroxyvitamin D [25(OH)D] concentrations with all-cause and cause-specific mortality among American adults with OA. METHODS This study included 2556 adults with OA from the National Health and Nutrition Examination Survey (2001-2014). Death outcomes were ascertained by linkage to National Death Index (NDI) records through 31 December 2015. Cox proportional hazards model and two-piecewise Cox proportional hazards model were used to elucidate the nonlinear relationship between serum 25(OH)D concentrations and mortality in OA patients, and stratified analyses were performed to identify patients with higher mortality risk. RESULTS During 16,606 person-years of follow-up, 438 all-cause deaths occurred, including 74 cardiovascular disease (CVD)-related and 78 cancer deaths. After multivariable adjustment, lower serum 25(OH)D levels were significantly and nonlinearly associated with higher risks of all-cause and CVD mortality among participants with OA. Furthermore, we discovered L-shaped associations between serum 25(OH)D levels and all-cause and CVD mortality, with mortality plateauing at 54.40 nmol/L for all-cause mortality and 27.70 nmol/L for CVD mortality. Compared to participants with 25(OH)D levels below the inflection points, those with higher levels had a 2% lower risk for all-cause mortality (hazard ratio [HR] 0.98, 95% confidence interval [CI] 0.96-0.99) and 17% lower risk for CVD mortality (HR 0.83, 95% CI 0.72-0.95). CONCLUSIONS Nonlinear associations of serum 25(OH)D levels with all-cause and CVD mortality were observed in American patients with OA. The thresholds of 27.70 and 54.40 nmol/L for CVD and all-cause mortality, respectively, may represent intervention targets for lowering the risk of premature death and cardiovascular disease, but this needs to be confirmed in large clinical trials.
Collapse
Affiliation(s)
- Qingqing Xiao
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Cai
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Anwen Yin
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huanhuan Huo
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Keke Lan
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guo Zhou
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
94
|
Li H, Li S, Yang H, Zhang Y, Zhang S, Ma Y, Hou Y, Zhang X, Niu K, Borné Y, Wang Y. Association of Ultraprocessed Food Consumption With Risk of Dementia: A Prospective Cohort Study. Neurology 2022; 99:e1056-e1066. [PMID: 36219796 DOI: 10.1212/wnl.0000000000200871] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 05/05/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES There has been a growing body of evidence associating consumption of ultraprocessed foods (UPF) with adverse health outcomes including depression, cardiovascular disease, and all-cause mortality. However, whether UPF are associated with dementia is unknown. The authors investigated the associations between UPF and dementia incidence in the UK Biobank. METHODS We included 72,083 participants (55 years or older) who were free from dementia at baseline and provided at least 2 times 24-hour dietary assessments from the UK Biobank study. Follow-up occurred through March 2021. UPF were defined according to the NOVA classification. Incident all-cause dementia comprising Alzheimer disease (AD) and vascular dementia was ascertained through electronic linkages to hospital and mortality records. Cox proportional hazards were used to estimate the association between the proportion (%) of UPF in the diet and the subsequent risk of dementia. In addition, substitution analysis was used to estimate the risk of dementia when substituting UPF with an equivalent proportion of unprocessed or minimally processed foods. RESULTS During a total of 717,333 person-years of follow-up (median 10.0 years), 518 participants developed dementia, of whom 287 developed AD and 119 developed vascular dementia. In the fully adjusted model, consumption of UPF was associated with higher risk of dementia (hazard ratio [HR] for 10% increase in UPF 1.25; 95% CI 1.14-1.37), AD (HR 1.14; 95% CI 1.00-1.30), and vascular dementia (HR 1.28; 95% CI 1.06-1.55), respectively. In addition, replacing 10% of UPF weight in diet with an equivalent proportion of unprocessed or minimally processed foods was estimated to be associated with a 19% lower risk of dementia (HR 0.81; 95% CI 0.74-0.89). DISCUSSION In this prospective cohort study, higher consumption of UPF was associated with higher risk of dementia, whereas substituting unprocessed or minimally processed foods for UPF was associated with lower risk of dementia.
Collapse
Affiliation(s)
- Huiping Li
- From the School of Public Health (H.L., H.Y., Y.Z., S.Z., Y.M., Y.H., X.Z., K.N., Y.W.), Tianjin Medical University; School of Management (S.L.), Tianjin University of Traditional Chinese Medicine; and Department of Clinical Sciences in Malmö (Y.B.), Lund University, Sweden
| | - Shu Li
- From the School of Public Health (H.L., H.Y., Y.Z., S.Z., Y.M., Y.H., X.Z., K.N., Y.W.), Tianjin Medical University; School of Management (S.L.), Tianjin University of Traditional Chinese Medicine; and Department of Clinical Sciences in Malmö (Y.B.), Lund University, Sweden
| | - Hongxi Yang
- From the School of Public Health (H.L., H.Y., Y.Z., S.Z., Y.M., Y.H., X.Z., K.N., Y.W.), Tianjin Medical University; School of Management (S.L.), Tianjin University of Traditional Chinese Medicine; and Department of Clinical Sciences in Malmö (Y.B.), Lund University, Sweden
| | - Yuan Zhang
- From the School of Public Health (H.L., H.Y., Y.Z., S.Z., Y.M., Y.H., X.Z., K.N., Y.W.), Tianjin Medical University; School of Management (S.L.), Tianjin University of Traditional Chinese Medicine; and Department of Clinical Sciences in Malmö (Y.B.), Lund University, Sweden
| | - Shunming Zhang
- From the School of Public Health (H.L., H.Y., Y.Z., S.Z., Y.M., Y.H., X.Z., K.N., Y.W.), Tianjin Medical University; School of Management (S.L.), Tianjin University of Traditional Chinese Medicine; and Department of Clinical Sciences in Malmö (Y.B.), Lund University, Sweden
| | - Yue Ma
- From the School of Public Health (H.L., H.Y., Y.Z., S.Z., Y.M., Y.H., X.Z., K.N., Y.W.), Tianjin Medical University; School of Management (S.L.), Tianjin University of Traditional Chinese Medicine; and Department of Clinical Sciences in Malmö (Y.B.), Lund University, Sweden
| | - Yabing Hou
- From the School of Public Health (H.L., H.Y., Y.Z., S.Z., Y.M., Y.H., X.Z., K.N., Y.W.), Tianjin Medical University; School of Management (S.L.), Tianjin University of Traditional Chinese Medicine; and Department of Clinical Sciences in Malmö (Y.B.), Lund University, Sweden
| | - Xinyu Zhang
- From the School of Public Health (H.L., H.Y., Y.Z., S.Z., Y.M., Y.H., X.Z., K.N., Y.W.), Tianjin Medical University; School of Management (S.L.), Tianjin University of Traditional Chinese Medicine; and Department of Clinical Sciences in Malmö (Y.B.), Lund University, Sweden
| | - Kaijun Niu
- From the School of Public Health (H.L., H.Y., Y.Z., S.Z., Y.M., Y.H., X.Z., K.N., Y.W.), Tianjin Medical University; School of Management (S.L.), Tianjin University of Traditional Chinese Medicine; and Department of Clinical Sciences in Malmö (Y.B.), Lund University, Sweden
| | - Yan Borné
- From the School of Public Health (H.L., H.Y., Y.Z., S.Z., Y.M., Y.H., X.Z., K.N., Y.W.), Tianjin Medical University; School of Management (S.L.), Tianjin University of Traditional Chinese Medicine; and Department of Clinical Sciences in Malmö (Y.B.), Lund University, Sweden
| | - Yaogang Wang
- From the School of Public Health (H.L., H.Y., Y.Z., S.Z., Y.M., Y.H., X.Z., K.N., Y.W.), Tianjin Medical University; School of Management (S.L.), Tianjin University of Traditional Chinese Medicine; and Department of Clinical Sciences in Malmö (Y.B.), Lund University, Sweden.
| |
Collapse
|
95
|
Jensen SB, Latysheva N, Hindberg K, Ueland T. Plasma lipopolysaccharide-binding protein is a biomarker for future venous thromboembolism: Results from discovery and validation studies. J Intern Med 2022; 292:523-535. [PMID: 35426199 PMCID: PMC9539954 DOI: 10.1111/joim.13502] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
BACKGROUND Effect-size underestimation impedes biomarker identification. Long follow-up time in prospective studies attenuates effect-size estimates for transient biomarkers, while disease category-specific biomarkers are affected by merging of categories. Venous thromboembolism (VTE) encompasses deep vein thrombosis (DVT) and pulmonary embolism (PE). OBJECTIVES (i) To re-analyze untargeted proteomic data to identify biomarker candidates for future VTE that differ between DVT and PE and are attenuated by extended time between sampling and VTE. (ii) To perform targeted candidate validation. PATIENTS/METHODS A VTE case-control discovery study and a nested case-control validation study were derived from the general population surveyed in 1994-95. Plasma was obtained at study enrollment, and VTE events were registered until 2007. Untargeted proteomic data were re-analyzed for candidate discovery. Lipopolysaccharide-binding protein (LBP) was validated by enzyme-linked immunosorbent assay. RESULTS Elevated LBP was discovered as a candidate DVT biomarker in women with less than 3 years between blood sampling and DVT. In the validation study, the odds ratio (OR) for DVT was 2.03 (95% confidence intervals [CI]: 1.53-2.74) per standard deviation (SD) increase in LBP for women with less than 3 years between blood sampling and DVT. Adjustment for age, body mass index, and C-reactive protein attenuated the OR to 1.79 (95% CI: 1.25-2.62) per SD. In the validation study, we observed an OR for VTE of 0.47 (95% CI: 0.28-0.77) for men in the 25th to 50th percentiles when compared to the lowest quartile. CONCLUSIONS We discovered and validated increased LBP as a predictive biomarker for DVT in women. We found an increased VTE risk for men in the lowest quartile of LBP.
Collapse
Affiliation(s)
- Søren Beck Jensen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Nadezhda Latysheva
- K. G. Jebsen Thrombosis Research and Expertise Center, Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Kristian Hindberg
- K. G. Jebsen Thrombosis Research and Expertise Center, Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,K. G. Jebsen Thrombosis Research and Expertise Center, Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
96
|
Bešević J, Lacey B, Conroy M, Omiyale W, Feng Q, Collins R, Allen N. New Horizons: the value of UK Biobank to research on endocrine and metabolic disorders. J Clin Endocrinol Metab 2022; 107:2403-2410. [PMID: 35793237 PMCID: PMC9387695 DOI: 10.1210/clinem/dgac407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Indexed: 11/24/2022]
Abstract
UK Biobank is an intensively characterized prospective study of 500 000 men and women, aged 40 to 69 years when recruited, between 2006 and 2010, from the general population of the United Kingdom. Established as an open-access resource for researchers worldwide to perform health research that is in the public interest, UK Biobank has collected (and continues to collect) a vast amount of data on genetic, physiological, lifestyle, and environmental factors, with prolonged follow-up of heath conditions through linkage to administrative electronic health records. The study has already demonstrated its unique value in enabling research into the determinants of common endocrine and metabolic diseases. The importance of UK Biobank, heralded as a flagship project for UK health research, will only increase over time as the number of incident disease events accrue, and the study is enhanced with additional data from blood assays (such as whole-genome sequencing, metabolomics, and proteomics), wearable technologies (including physical activity and cardiac monitors), and body imaging (magnetic resonance imaging and dual-energy X-ray absorptiometry). This unique research resource is likely to transform our understanding of the causes, diagnosis, and treatment of many endocrine and metabolic disorders.
Collapse
Affiliation(s)
- Jelena Bešević
- Oxford Population Health (Nuffield Department of Population Health), University of Oxford
| | - Ben Lacey
- Oxford Population Health (Nuffield Department of Population Health), University of Oxford
| | - Megan Conroy
- Oxford Population Health (Nuffield Department of Population Health), University of Oxford
| | - Wemimo Omiyale
- Oxford Population Health (Nuffield Department of Population Health), University of Oxford
| | - Qi Feng
- Oxford Population Health (Nuffield Department of Population Health), University of Oxford
| | - Rory Collins
- Oxford Population Health (Nuffield Department of Population Health), University of Oxford
- UK Biobank, Stockport, Greater Manchester, UK
| | - Naomi Allen
- Oxford Population Health (Nuffield Department of Population Health), University of Oxford
- UK Biobank, Stockport, Greater Manchester, UK
| |
Collapse
|
97
|
Ahmadi MN, Lee IM, Hamer M, del Pozo Cruz B, Chen LJ, Eroglu E, Lai YJ, Ku PW, Stamatakis E. Changes in physical activity and adiposity with all-cause, cardiovascular disease, and cancer mortality. Int J Obes (Lond) 2022; 46:1849-1858. [PMID: 35915134 PMCID: PMC9492547 DOI: 10.1038/s41366-022-01195-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 12/31/2022]
Abstract
Background The relationship between joint changes in physical activity and adiposity with mortality is not well understood. We examined the association of changes in these two established risk factors with all-cause (ACM), cardiovascular disease (CVD), and cancer mortality. Methods We used longitudinal data from Taiwan’s MJ Cohort, comprising 116,228 general population adults recruited from 1998-2013 with repeated measures 4.6 y (2.5) apart and followed up for mortality for 11.9 y (3.5). Physical activity, body mass index (BMI), waist circumference (WC), and body fat percentage (BF%) groups and changes were based on public health and clinical guidelines. Results Compared to stable-insufficient physical activity, increasing physical activity from any baseline level was associated with lower ACM (HR [95%CI]): 0.85 [0.74, 0.96]) and CVD mortality (0.72 [0.55, 0.93]) risk. This was approximately equal to meeting physical activity guidelines at both timepoints (eg: 0.71 [0.58, 0.88] for CVD mortality). Compared to stable-overweight/moderate adiposity, decreasing adiposity level attenuated but did not offset mortality risk for all three outcomes (eg: BMI = 0.95 [0.76, 1.16] for CVD mortality). Only maintaining a healthy adiposity level at both timepoints offset mortality risk (BMI = 0.75 [0.61, 0.89]) for CVD mortality). In the joint changes analyses, lower mortality risk was a consequence of increases in physical activity across adiposity change groups (eg: WC decrease = 0.57 [0.48, 0.67]; WC stability = 0.73 [0.66, 0.80], WC increase = 0.83 [0.72, 0.97] for ACM). Decreasing adiposity attenuated the negative associations of decreased physical activity (BF% = 1.13 [0.95, 1.35] for ACM). Conclusions We found a lower risk for ACM, CVD, and cancer mortality from increasing physical activity and an attenuation from decreasing adiposity regardless of baseline levels. The beneficial associations of joint changes were primarily driven by physical activity, suggesting lower mortality risk may be more immediate through physical activity improvements compared to adiposity improvements alone.
Collapse
|
98
|
Matsumura T, Sankai T, Yamagishi K, Tanaka M, Kubota Y, Hayama-Terada M, Shimizu Y, Muraki I, Umesawa M, Cui R, Imano H, Ohira T, Kitamura A, Okada T, Kiyama M, Iso H. Trends for the Association between Body Mass Index and Risk of Cardiovascular Disease among the Japanese Population: The Circulatory Risk in Communities Study (CIRCS). J Atheroscler Thromb 2022; 30:335-347. [PMID: 35896353 PMCID: PMC10067340 DOI: 10.5551/jat.63415] [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] [Indexed: 01/01/2023] Open
Abstract
AIM This study aimed to investigate whether the impact of body mass index (BMI) on the risk of cardiovascular disease (CVD) has changed among the 1960s, 1970s, 1980s, 1990s, and early 2000s in Japan. METHODS The study population consisted of residents in Japan aged 40-69 years who had no history of CVD. The baseline surveys have been conducted every year since 1963. We defined the first, second, third, fourth, and fifth cohorts as 1963-1969 (n=4,248), 1970-1979 (n=6,742), 1980-1989 (n=12,789), 1990-1999 (n=12,537), and 2000-2005 (n=9,140) respectively. The participants were followed up for a median of 15 years for each cohort to determine the incidence of CVD. We classified them into four categories (BMI <21.0, 21.0-<23.0, 23.0-<25.0, and ≥ 25.0 kg/m 2). RESULTS From 1963-1969 to 2000-2005, the prevalence of BMI ≥ 25.0 increased over time. Compared with BMI 23.0-<25.0, the age-, sex- and community-adjusted hazard ratios (95% confidence interval [CIs]) of CVD for BMI ≥ 25.0 were 1.10 (0.77-1.57), 0.89 (0.68-1.18), 1.03 (0.85-1.26), 1.28 (1.04-1.58), and 1.36 (1.04-1.78)in the first, second, third, fourth, and fifth cohorts, respectively. The corresponding population attributable fractions were 2.0% (nonsignificant), -2.6% (nonsignificant), 0.9% (nonsignificant), 7.6%, and 10.9%. Further adjustment for systolic blood pressure and antihypertensive medication use in the fourth and fifth cohorts attenuated the associations, which may reflect that blood pressure may mediate the BMI-CVD association. CONCLUSION The proportion of CVD attributable to overweight/obesity has increased during the periods between 1963-1969 and 2000-2005. The significant associations between overweight/obesity and risk of CVD after the 1990s were mediated by blood pressure levels.
Collapse
Affiliation(s)
- Takumi Matsumura
- Public Health, Department of Social Medicine, Osaka University Graduate School of Medicine.,Osaka Center for Cancer and Cardiovascular Disease Prevention
| | - Tomoko Sankai
- Department of Public Health and Nursing, Faculty of Medicine, University of Tsukuba
| | - Kazumasa Yamagishi
- Department of Public Health Medicine, Faculty of Medicine, and Health Services Research and Development Center, University of Tsukuba.,Ibaraki Western Medical Center
| | - Mari Tanaka
- Public Health, Department of Social Medicine, Osaka University Graduate School of Medicine
| | - Yasuhiko Kubota
- Osaka Center for Cancer and Cardiovascular Disease Prevention
| | - Mina Hayama-Terada
- Osaka Center for Cancer and Cardiovascular Disease Prevention.,Yao City Public Health Center
| | - Yuji Shimizu
- Osaka Center for Cancer and Cardiovascular Disease Prevention
| | - Isao Muraki
- Public Health, Department of Social Medicine, Osaka University Graduate School of Medicine
| | - Mitsumasa Umesawa
- Department of Public Health Medicine, Faculty of Medicine, and Health Services Research and Development Center, University of Tsukuba.,School of Medicine, Dokkyo Medical University
| | - Renzhe Cui
- Department of Internal Medicine, Okanami General Hospital
| | - Hironori Imano
- Public Health, Department of Social Medicine, Osaka University Graduate School of Medicine.,Osaka Center for Cancer and Cardiovascular Disease Prevention.,Department of Public Health, Kindai University Faculty of Medicine
| | - Tetsuya Ohira
- Department of Epidemiology, Fukushima Medical University School of Medicine
| | | | - Takeo Okada
- Osaka Center for Cancer and Cardiovascular Disease Prevention
| | - Masahiko Kiyama
- Osaka Center for Cancer and Cardiovascular Disease Prevention
| | - Hiroyasu Iso
- Public Health, Department of Social Medicine, Osaka University Graduate School of Medicine.,Department of Public Health Medicine, Faculty of Medicine, and Health Services Research and Development Center, University of Tsukuba
| |
Collapse
|
99
|
Damoah CE, Snir O, Hindberg K, Garred P, Ludviksen JK, Brækkan SK, Morelli VM, Eirik Mollnes T, Hansen JB. High Levels of Complement Activating Enzyme MASP-2 Are Associated With the Risk of Future Incident Venous Thromboembolism. Arterioscler Thromb Vasc Biol 2022; 42:1186-1197. [PMID: 35861070 DOI: 10.1161/atvbaha.122.317746] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Experimental studies have shown that the complement activating enzyme MASP-2 (mannose-binding lectin associated serine protease 2) exhibits a thrombin-like activity and that inhibition of MASP-2 protects against thrombosis. In this study, we investigated whether plasma MASP-2 levels were associated with risk of future venous thromboembolism (VTE) and whether genetic variants linked to MASP-2 levels were associated with VTE risk. METHODS We conducted a population-based nested case-control study involving 410 VTE patients and 842 age- and sex-matched controls derived from the Norwegian Tromsø Study. Logistic regression was used to estimate odds ratios (ORs) of VTE across MASP-2 quartiles. Whole-exome sequencing and protein quantitative trait loci analyses were performed to assess genetic variants associated with MASP-2 levels. A 2-sample Mendelian randomization study, also including data from the INVENT consortium (International Network of Venous Thrombosis), was performed to assess causality. RESULTS Subjects with plasma MASP-2 in the highest quartile had a 48% higher OR of VTE (OR, 1.48 [95% CI, 1.06-2.06]) and 83% higher OR of deep vein thrombosis (OR, 1.83 [95% CI, 1.23-2.73]) compared with those with MASP-2 levels in the lowest quartile. The protein quantitative trait loci analysis revealed that 3 previously described gene variants, rs12711521 (minor allele frequency, 0.153), rs72550870 (minor allele frequency, 0.045; missense variants in the MASP2 gene), and rs2275527 (minor allele frequency, 0.220; exon variant in the adjacent MTOR gene) explained 39% of the variation of MASP-2 plasma concentration. The OR of VTE per 1 SD increase in genetically predicted MASP-2 was 1.03 ([95% CI, 1.01-1.05] P=0.0011). CONCLUSIONS Our findings suggest that high plasma MASP-2 levels are causally associated with risk of future VTE.
Collapse
Affiliation(s)
- Christabel Esi Damoah
- Department of Clinical Medicine, Thrombosis Research Center, UiT The Arctic University of Norway, Tromsø (C.E.D., O.S., K.H., S.K.B., V.M.M., T.E.M., J.-B.H.)
| | - Omri Snir
- Department of Clinical Medicine, Thrombosis Research Center, UiT The Arctic University of Norway, Tromsø (C.E.D., O.S., K.H., S.K.B., V.M.M., T.E.M., J.-B.H.)
| | - Kristian Hindberg
- Department of Clinical Medicine, Thrombosis Research Center, UiT The Arctic University of Norway, Tromsø (C.E.D., O.S., K.H., S.K.B., V.M.M., T.E.M., J.-B.H.)
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark (P.G.)
| | | | - Sigrid K Brækkan
- Department of Clinical Medicine, Thrombosis Research Center, UiT The Arctic University of Norway, Tromsø (C.E.D., O.S., K.H., S.K.B., V.M.M., T.E.M., J.-B.H.).,Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway (S.K.B., V.M.M., J.-B.H.)
| | - Vânia M Morelli
- Department of Clinical Medicine, Thrombosis Research Center, UiT The Arctic University of Norway, Tromsø (C.E.D., O.S., K.H., S.K.B., V.M.M., T.E.M., J.-B.H.).,Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway (S.K.B., V.M.M., J.-B.H.)
| | - Tom Eirik Mollnes
- Department of Clinical Medicine, Thrombosis Research Center, UiT The Arctic University of Norway, Tromsø (C.E.D., O.S., K.H., S.K.B., V.M.M., T.E.M., J.-B.H.).,Research Laboratory, Nordland Hospital, Bodø, Norway (J.K.L., T.E.M.).,Department of Immunology, Oslo University Hospital and University of Oslo, Norway (T.E.M.).,Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway (T.E.M.)
| | - John-Bjarne Hansen
- Department of Clinical Medicine, Thrombosis Research Center, UiT The Arctic University of Norway, Tromsø (C.E.D., O.S., K.H., S.K.B., V.M.M., T.E.M., J.-B.H.).,Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway (S.K.B., V.M.M., J.-B.H.)
| | | |
Collapse
|
100
|
Frischmuth T, Hindberg K, Aukrust P, Ueland T, Brækkan SK, Hansen J, Morelli VM. Elevated plasma levels of plasminogen activator inhibitor-1 are associated with risk of future incident venous thromboembolism. J Thromb Haemost 2022; 20:1618-1626. [PMID: 35289062 PMCID: PMC9314992 DOI: 10.1111/jth.15701] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 03/07/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Plasminogen activator inhibitor-1 (PAI-1), the main inhibitor of fibrinolysis, is frequently elevated in obesity and could potentially mediate the risk of venous thromboembolism (VTE) in obese subjects. However, whether PAI-1 is associated with VTE remains uncertain. OBJECTIVE To investigate the association between plasma PAI-1 levels and risk of future incident VTE and whether PAI-1 could mediate the VTE risk in obesity. METHODS A population-based nested case-control study, comprising 383 VTE cases and 782 age- and sex-matched controls, was derived from the Tromsø Study cohort. PAI-1 antigen levels were measured in samples collected at cohort inclusion. Logistic regression was used to calculate odds ratios (ORs) with 95% confidence intervals (CIs) for VTE across PAI-1 tertiles. RESULTS The VTE risk increased dose-dependently across PAI-1 tertiles (P for trend <.001) in the age- and sex-adjusted model. The OR of VTE for the highest versus lowest tertile was 1.73 (95% CI 1.27-2.35), and risk estimates were only slightly attenuated with additional stepwise adjustment for body mass index (BMI; OR 1.59, 95% CI 1.16-2.17) and C-reactive protein (CRP; OR 1.54, 95% CI 1.13-2.11). Similar results were obtained for provoked/unprovoked events, deep vein thrombosis, and pulmonary embolism. In obese subjects (BMI of ≥30 kg/m2 vs. <25 kg/m2 ), PAI-1 mediated 14.9% (95% CI 4.1%-49.4%) of the VTE risk in analysis adjusted for age, sex, and CRP. CONCLUSION Our findings indicate that plasma PAI-1 is associated with increased risk of future incident VTE and has the potential to partially mediate the VTE risk in obesity.
Collapse
Affiliation(s)
- Tobias Frischmuth
- Thrombosis Research CenterDepartment of Clinical MedicineUiT—The Arctic University of NorwayTromsøNorway
- Division of Internal MedicineUniversity Hospital of North NorwayTromsøNorway
| | - Kristian Hindberg
- Thrombosis Research CenterDepartment of Clinical MedicineUiT—The Arctic University of NorwayTromsøNorway
| | - Pål Aukrust
- Thrombosis Research CenterDepartment of Clinical MedicineUiT—The Arctic University of NorwayTromsøNorway
- Faculty of MedicineUniversity of OsloOsloNorway
- Research Institute of Internal MedicineOslo University Hospital RikshospitaletOsloNorway
- Section of Clinical Immunology and Infectious DiseasesOslo University Hospital RikshospitaletOsloNorway
| | - Thor Ueland
- Thrombosis Research CenterDepartment of Clinical MedicineUiT—The Arctic University of NorwayTromsøNorway
- Faculty of MedicineUniversity of OsloOsloNorway
- Research Institute of Internal MedicineOslo University Hospital RikshospitaletOsloNorway
| | - Sigrid K. Brækkan
- Thrombosis Research CenterDepartment of Clinical MedicineUiT—The Arctic University of NorwayTromsøNorway
- Division of Internal MedicineUniversity Hospital of North NorwayTromsøNorway
| | - John‐Bjarne Hansen
- Thrombosis Research CenterDepartment of Clinical MedicineUiT—The Arctic University of NorwayTromsøNorway
- Division of Internal MedicineUniversity Hospital of North NorwayTromsøNorway
| | - Vânia M. Morelli
- Thrombosis Research CenterDepartment of Clinical MedicineUiT—The Arctic University of NorwayTromsøNorway
- Division of Internal MedicineUniversity Hospital of North NorwayTromsøNorway
| |
Collapse
|