1
|
Wadström BN, Wulff AB, Pedersen KM, Nordestgaard BG. Small Remnants versus Large Triglyceride-Rich Lipoproteins in Risk of Atherosclerotic Cardiovascular Disease. Clin Chem 2025:hvae222. [PMID: 39882976 DOI: 10.1093/clinchem/hvae222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/18/2024] [Indexed: 01/31/2025]
Abstract
BACKGROUND Small remnants may penetrate the arterial intima more efficiently compared to large triglyceride-rich lipoproteins (TGRL). We tested the hypothesis that the importance of remnant cholesterol for the risk of atherosclerotic cardiovascular disease (ASCVD) may depend on the size of the remnants and TGRL carrying cholesterol. METHODS The cholesterol content of small remnants and large TGRL were measured in 25 572 individuals from the Copenhagen General Population Study (2003-2015) and in 222 721 individuals from the UK Biobank (2006-2010) using nuclear magnetic resonance spectroscopy. In the Copenhagen cohort during up to 15 years of follow-up and in the UK Biobank cohort during up to 16 years of follow-up, the numbers of individuals diagnosed with ASCVD (=myocardial infarction, ischemic stroke, and peripheral artery disease) in national health registries were 3869 and 11 424, respectively. RESULTS Compared to individuals with low cholesterol content in both small remnants and large TGRL (cutpoints were median cholesterol content), multivariable-adjusted hazard ratios for risk of ASCVD were 1.21 (95% confidence interval: 1.07-1.37) for individuals with high cholesterol content in small remnants only and 0.94 (0.83-1.07) for individuals with high cholesterol content in large TGRL only; the multivariable-adjusted hazard ratio for risk of ASCVD per 10 percentile-units higher cholesterol content in small remnants vs that in large TGRL was 1.04 (1.01-1.07). In the UK Biobank cohort, corresponding hazard ratios were 1.11 (1.03-1.20), 1.01 (0.93-1.09), and 1.05 (1.04-1.07), respectively. CONCLUSION The importance of remnant cholesterol for the risk of ASCVD may depend on the size of the TGRL and remnants carrying cholesterol.
Collapse
Affiliation(s)
- Benjamin N Wadström
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders B Wulff
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kasper M Pedersen
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Nijssen KMR, Chavez-Alfaro MA, Joris PJ, Plat J, Mensink RP. Effects of Longer-Term Mixed Nut Consumption on Lipoprotein Particle Concentrations in Older Adults with Overweight or Obesity. Nutrients 2024; 17:8. [PMID: 39796442 PMCID: PMC11723242 DOI: 10.3390/nu17010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Recently, we reported that longer-term mixed nut intake significantly reduced serum total and low-density lipoprotein (LDL)-cholesterol, but these markers may not fully capture lipoprotein-related cardiovascular disease (CVD) risk. OBJECTIVES This randomized, controlled, single-blinded, crossover trial in older adults with overweight or obesity examined the effects of longer-term mixed nut consumption on lipoprotein particle size, number, and lipid distribution. METHODS Twenty-eight participants (aged 65 ± 3 years; BMI 27.9 ± 2.3 kg/m2) completed two 16-week periods (control [no nuts] vs. mixed nuts (60 g/day: 15 g of walnuts, pistachios, cashews, and hazelnuts), separated by an 8-week washout. Plasma lipoprotein particle numbers, sizes, and lipid distributions across subclasses were analyzed using high-throughput nuclear magnetic resonance (NMR) spectroscopy. RESULTS Mixed nut consumption significantly reduced Apolipoprotein B (ApoB) concentrations (-0.07 g/L; p = 0.009), total cholesterol (-0.27 mmol/L; p = 0.047), non-HDL cholesterol (-0.28 mmol/L; p = 0.022), and total triacylglycerol (TAG) (-0.27 mmol/L; p = 0.008). Total very large-density lipoprotein (VLDL) particle numbers decreased by 24 nmol/L (p < 0.001), with reductions observed across all VLDL subclasses. Total LDL particle numbers (p = 0.044), specifically intermediate-density lipoprotein (IDL) (p = 0.002) and large LDL particles (p = 0.015), were also reduced, while HDL particle numbers and sizes were unaffected. The mixed nut intervention significantly reduced cholesterol concentrations across all VLDL subclasses and IDL (all p < 0.01), with no changes in LDL or HDL subclasses. TAG concentrations showed reductions across all lipoprotein subclasses (all p < 0.05). CONCLUSIONS Longer-term mixed nut consumption may lower CVD risk in older adults and favorable shifts in apoB-containing lipoprotein subclasses towards a less atherogenic profile.
Collapse
Affiliation(s)
- Kevin M. R. Nijssen
- Department of Nutrition and Movement Sciences, NUTRIM Institute of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (M.A.C.-A.); (P.J.J.); (J.P.); (R.P.M.)
| | | | | | | | | |
Collapse
|
3
|
Zhu J, Chen J, Zuo Y, Song K, Liao H, Wu X. Investigating the causal effect of various metabolites on postherpetic neuralgia: a Mendelian randomization study. Front Neurol 2024; 15:1421670. [PMID: 39650245 PMCID: PMC11621009 DOI: 10.3389/fneur.2024.1421670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/24/2024] [Indexed: 12/11/2024] Open
Abstract
Background Common side effect of Herpes Zoster, postherpetic neuralgia (PHN), causes persistent pain that seriously affects quality of life. Lack of dependable biomarkers makes the clinical diagnosis and treatment of PHN difficult, so complicating the assessment of therapeutic efficacy. Blood metabolites are becoming more and more well known as significant disease markers. With an aim to find possible biomarkers for diagnosis and treatment, this work investigates the causal link between blood metabolites and PHN using Mendelian randomization. Methods This work evaluated causal relationships between PHN and 1,091 plasma metabolites using Mendelian randomization (MR). Complementing MR-Egger and weighted median approaches, the main causality analysis was done using inverse variance weighted (IVW) and Wald ratio (WR) approaches. Robustness was checked using sensitivity analyses including CAUSE, Cochran's Q tests, leave-one-out analysis, MR-PRESSO, and MR-Egger intercept analysis. Reverse MR analysis and linkage disequilibrium score regression (LDSC) was used to assess significant correlations as well. Two-step MR analysis was also used to look at the mediating function of positively correlated metabolites in the causal pathway. Results The results of this study indicated a significant association between N-acetyl-aspartyl-glutamate (NAAG) and PHN, with an odds ratio (OR) of 0.83 (95% CI: 0.76-0.91, p = 2.68E-05). Moreover, five potential associated metabolites were identified: Gamma-glutamylthreonine (OR = 1.60, 95% CI: 1.16-2.20, p = 0.004), 3-hydroxyphenylacetoylglutamine (OR = 1.43, 95% CI: 1.00-2.05, p = 0.048), Caprate (10:0) (OR = 1.86, 95% CI: 1.11-3.12, p = 0.018), X-12013 (OR = 1.64, 95% CI: 1.03-2.60, p = 0.035), and X-17328 (OR = 1.50, 95% CI: 1.04-2.18, p = 0.032). Additionally, NAAG likely acts as a complete mediator between FOLH1(CGPII) and postherpetic neuralgia in the causal pathway. Conclusion The results of this study indicated a significant association between N-acetyl-aspartyl-glutamate (NAAG) and PHN, with an odds ratio (OR) of 0.83 (95% CI: 0.76-0.91, p = 2.68E-05). Furthermore five possible related metabolites were found: Glutamylthreonine gamma-wise (OR = 1.60, 95% CI: 1.16-2.20, p = 0.004), 3-hydroxyphenylacetoylglutamine (OR = 1.43, 95% CI: 1.00-2.05, p = 0.048), Caprate (10:0) (OR = 1.86, 95% CI: 1.11-3.12, p = 0.018), X-12013 (OR = 1.64, 95% CI: 1.03-2.60, p = 0.035), and X-17328 (OR = 1.50, 95% CI: 1.04-2.18, p = 0.032). Furthermore, in the causal pathway NAAG most certainly serves as a complete mediator between FOLH1(CGPII) and postherpetic neuralgia.
Collapse
Affiliation(s)
- Jianyu Zhu
- Department of Clinical Medical Laboratory, Shunde Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiahao Chen
- Department of Anesthesiology, Shunde Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuefen Zuo
- Department of Anesthesiology, Shunde Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kun Song
- Department of Anesthesiology, Shunde Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huilian Liao
- Department of Nursing, Shunde Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xianping Wu
- Department of Anesthesiology, Shunde Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
4
|
Wadström BN, Pedersen KM, Wulff AB, Nordestgaard BG. One in Five Atherosclerotic Cardiovascular Disease Events in Individuals With Diabetes Attributed to Elevated Remnant Cholesterol. Diabetes Metab Res Rev 2024; 40:e70005. [PMID: 39550770 DOI: 10.1002/dmrr.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 09/09/2024] [Accepted: 09/27/2024] [Indexed: 11/19/2024]
Abstract
AIMS Elevated remnant cholesterol (= the cholesterol carried in triglyceride-rich lipoproteins) is a causal risk factor for atherosclerotic cardiovascular disease (ASCVD) and is common in individuals with diabetes. We tested the hypothesis that ASCVD in individuals with diabetes can be partly attributed to elevated remnant cholesterol. MATERIALS AND METHODS We included 3806 individuals with diabetes identified among 107,243 individuals from the Copenhagen General Population Study and used multivariable adjusted Poisson regression to estimate the fraction of ASCVD attributable to elevated remnant cholesterol. Elevated remnant cholesterol was defined as levels higher than those observed in individuals with non-high-density lipoprotein (non-HDL) cholesterol < 2.6 mmol/L (100 mg/dL), the European guideline goal. Results were replicated in the UK Biobank. RESULTS During 15 years of follow-up, 498 patients were diagnosed with ASCVD, 172 with peripheral artery disease, 185 with myocardial infarction and 195 with ischaemic stroke. In individuals with non-HDL cholesterol < 2.6 mmol/L (100 mg/dL) and in all individuals with diabetes, median remnant cholesterol levels were 0.5 mmol/L (20 mg/dL) and 0.8 mmol/L (31 mg/dL). The fraction of events attributable to elevated remnant cholesterol was 19% (95% confidence interval: 10%-28%) for ASCVD, 21% (5%-37%) for peripheral artery disease, 24% (10%-37%) for myocardial infarction and 17% (1%-31%) for ischaemic stroke; in the UK Biobank, corresponding values were 16% (9%-22%), 25% (12%-36%), 17% (8%-25%) and 7% (0%-19%), respectively. CONCLUSIONS One in five ASCVD events in individuals with diabetes can be attributed to elevated remnant cholesterol. It remains to be determined in clinical trials if remnant cholesterol-lowering therapy may prevent ASCVD.
Collapse
Affiliation(s)
- Benjamin N Wadström
- Department of Clinical Biochemistry, Copenhagen University Hospital─Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital─Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kasper M Pedersen
- Department of Clinical Biochemistry, Copenhagen University Hospital─Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital─Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders B Wulff
- Department of Clinical Biochemistry, Copenhagen University Hospital─Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital─Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital─Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital─Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
5
|
Zonneveld MH, Al Kuhaili N, Mooijaart SP, Slagboom PE, Jukema JW, Noordam R, Trompet S. Increased 1H-NMR metabolomics-based health score associates with declined cognitive performance and functional independence in older adults at risk of cardiovascular disease. GeroScience 2024:10.1007/s11357-024-01391-x. [PMID: 39436550 DOI: 10.1007/s11357-024-01391-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/11/2024] [Indexed: 10/23/2024] Open
Abstract
The 1-HMR metabolomics-based MetaboHealth score, comprised of 14 serum metabolic markers, associates with disease-specific mortality, but it is unclear whether the score also reflects cognitive changes and functional impairment. We aimed to assess the associations between the MetaboHealth score with cognitive function and functional decline in older adults at increased cardiovascular risk. A total of 5292 older adults free of dementia at baseline with mean age 75.3 years (SD = 3.4) from the Prospective Study of Pravastatin in the Elderly (PROSPER). MetaboHealth score were measured at baseline, and cognitive function and functional independence were measured at baseline and every 3 months during up to 2.5 years follow-up. Cognitive function was assessed using the Stroop test (selective attention), the Letter Digit Coding test (LDCT) (processing speed), and the two versions of the Picture Learning test (delayed and immediate; memory). Two tests of functional independence were used: Barthel Index (BI) and instrumental activities at daily living (IADL). A higher MetaboHealth score was associated with worse cognitive function (in all domains) and with worse functional independence. For example, after full adjustments, a 1-SD higher MetaboHealth score was associated with 9.02 s (95%CI 7.29, 10.75) slower performance on the Stroop test and 2.79 (2.21, 3.26) less digits coded on the LDCT. During follow-up, 1-SD higher MetaboHealth score was associated with an additional decline of 0.53 s (0.23, 0.83) on the Stroop test and - 0.08 (- 0.11, - 0.06) points on the IADL. Metabolic disturbance, as reflected by an increased metabolomics-based health score, may mark future cognitive and functional decline.
Collapse
Affiliation(s)
- Michelle H Zonneveld
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nour Al Kuhaili
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Simon P Mooijaart
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
- LUMC Center for Medicine for Older People, Leiden University Medical Center, Leiden, The Netherlands
| | - P Eline Slagboom
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.
| | - Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| |
Collapse
|
6
|
Jury EC, Peng J, Van Vijfeijken A, Martin Gutierrez L, Woodridge L, Wincup C, Pineda-Torra I, Ciurtin C, Robinson GA. Systemic lupus erythematosus patients have unique changes in serum metabolic profiles across age associated with cardiometabolic risk. Rheumatology (Oxford) 2024; 63:2741-2753. [PMID: 38048621 PMCID: PMC11443078 DOI: 10.1093/rheumatology/kead646] [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: 08/18/2023] [Revised: 11/02/2023] [Accepted: 11/05/2023] [Indexed: 12/06/2023] Open
Abstract
OBJECTIVES Cardiovascular disease through accelerated atherosclerosis is a leading cause of mortality for patients with systemic lupus erythematosus (SLE), likely due to increased chronic inflammation and cardiometabolic defects over age. We investigated age-associated changes in metabolomic profiles of SLE patients and healthy controls (HCs). METHODS Serum NMR metabolomic profiles from female SLE patients (n = 164, age = 14-76) and HCs (n = 123, age = 13-72) were assessed across age by linear regression and by age group between patients/HCs (Group 1, age ≤ 25, n = 62/46; Group 2, age = 26-49, n = 50/46; Group 3, age ≥ 50, n = 52/31) using multiple t tests. The impact of inflammation, disease activity and treatments were assessed, and UK Biobank disease-wide association analysis of metabolites was performed. RESULTS Age-specific metabolomic profiles were identified in SLE patients vs HCs, including reduced amino acids (Group 1), increased very-low-density lipoproteins (Group 2), and increased low-density lipoproteins (Group 3). Twenty-five metabolites were significantly altered in all SLE age groups, dominated by decreased atheroprotective high-density lipoprotein (HDL) subsets, HDL-bound apolipoprotein (Apo)A1 and increased glycoprotein acetyls (GlycA). Furthermore, ApoA1 and GlycA were differentially associated with disease activity and serological measures, as well as atherosclerosis incidence and myocardial infarction mortality risk through disease-wide association. Separately, glycolysis pathway metabolites (acetone/citrate/creatinine/glycerol/lactate/pyruvate) uniquely increased with age in SLE, significantly influenced by prednisolone (increased pyruvate/lactate) and hydroxychloroquine (decreased citrate/creatinine) treatment and associated with type 1 and type 2 diabetes by disease-wide association. CONCLUSIONS Increasing HDL (ApoA1) levels through therapeutic/nutritional intervention, whilst maintaining low disease activity, in SLE patients from a young age could improve cardiometabolic disease outcomes. Biomarkers from the glycolytic pathway could indicate adverse metabolic effects of current therapies.
Collapse
Affiliation(s)
- Elizabeth C Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
| | - Junjie Peng
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | | | - Lucia Martin Gutierrez
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | - Laurel Woodridge
- Centre for Experimental & Translational Medicine, Division of Medicine, University College London, London, UK
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| | - Chris Wincup
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
| | - Ines Pineda-Torra
- Centre for Experimental & Translational Medicine, Division of Medicine, University College London, London, UK
| | - Coziana Ciurtin
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | - George A Robinson
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| |
Collapse
|
7
|
Cao Zhang AM, Ziogos E, Harb T, Gerstenblith G, Leucker TM. Emerging clinical role of proprotein convertase subtilisin/kexin type 9 inhibition-Part two: Current and emerging concepts in the clinical use of PCSK9 inhibition. Eur J Clin Invest 2024; 54:e14272. [PMID: 38924090 DOI: 10.1111/eci.14272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/20/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors have emerged as a novel class of drugs with cardioprotective effects through their lipid-lowering effects. OBJECTIVE This review aims to discuss existing and novel strategies of PCSK9 inhibition, providing an overview of established randomized controlled trials and ongoing outcome trials that assess the efficacy and long-term safety of PCSK9 inhibitors. It also explores the evolving role of PCSK9 beyond lipid metabolism and outlines the pleiotropic actions of PCSK9 inhibition in various disorders and future directions including novel strategies to target PCSK9. CONCLUSION PCSK9 inhibition shows promise not only in lipid metabolism but also in other disease processes, including atherosclerotic plaque remodeling, acute coronary syndrome, stroke, inflammation, and immune response.
Collapse
Affiliation(s)
- Alexander M Cao Zhang
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Efthymios Ziogos
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tarek Harb
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Gary Gerstenblith
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Thorsten M Leucker
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Li CL, Liu SF. Exploring Molecular Mechanisms and Biomarkers in COPD: An Overview of Current Advancements and Perspectives. Int J Mol Sci 2024; 25:7347. [PMID: 39000454 PMCID: PMC11242201 DOI: 10.3390/ijms25137347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) plays a significant role in global morbidity and mortality rates, typified by progressive airflow restriction and lingering respiratory symptoms. Recent explorations in molecular biology have illuminated the complex mechanisms underpinning COPD pathogenesis, providing critical insights into disease progression, exacerbations, and potential therapeutic interventions. This review delivers a thorough examination of the latest progress in molecular research related to COPD, involving fundamental molecular pathways, biomarkers, therapeutic targets, and cutting-edge technologies. Key areas of focus include the roles of inflammation, oxidative stress, and protease-antiprotease imbalances, alongside genetic and epigenetic factors contributing to COPD susceptibility and heterogeneity. Additionally, advancements in omics technologies-such as genomics, transcriptomics, proteomics, and metabolomics-offer new avenues for comprehensive molecular profiling, aiding in the discovery of novel biomarkers and therapeutic targets. Comprehending the molecular foundation of COPD carries substantial potential for the creation of tailored treatment strategies and the enhancement of patient outcomes. By integrating molecular insights into clinical practice, there is a promising pathway towards personalized medicine approaches that can improve the diagnosis, treatment, and overall management of COPD, ultimately reducing its global burden.
Collapse
Affiliation(s)
- Chin-Ling Li
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
| | - Shih-Feng Liu
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
9
|
Holmes MV, Kartsonaki C, Boxall R, Lin K, Reeve N, Yu C, Lv J, Bennett DA, Hill MR, Yang L, Chen Y, Du H, Turnbull I, Collins R, Clarke RJ, Tobin MD, Li L, Millwood IY, Chen Z, Walters RG. PCSK9 genetic variants and risk of vascular and non-vascular diseases in Chinese and UK populations. Eur J Prev Cardiol 2024; 31:1015-1025. [PMID: 38198221 PMCID: PMC11144468 DOI: 10.1093/eurjpc/zwae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024]
Abstract
AIMS Lowering low-density lipoprotein cholesterol (LDL-C) through PCSK9 inhibition represents a new therapeutic approach to preventing and treating cardiovascular disease (CVD). Phenome-wide analyses of PCSK9 genetic variants in large biobanks can help to identify unexpected effects of PCSK9 inhibition. METHODS AND RESULTS In the prospective China Kadoorie Biobank, we constructed a genetic score using three variants at the PCSK9 locus associated with directly measured LDL-C [PCSK9 genetic score (PCSK9-GS)]. Logistic regression gave estimated odds ratios (ORs) for PCSK9-GS associations with CVD and non-CVD outcomes, scaled to 1 SD lower LDL-C. PCSK9-GS was associated with lower risks of carotid plaque [n = 8340 cases; OR = 0.61 (95% confidence interval: 0.45-0.83); P = 0.0015], major occlusive vascular events [n = 15 752; 0.80 (0.67-0.95); P = 0.011], and ischaemic stroke [n = 11 467; 0.80 (0.66-0.98); P = 0.029]. However, PCSK9-GS was also associated with higher risk of hospitalization with chronic obstructive pulmonary disease [COPD: n = 6836; 1.38 (1.08-1.76); P = 0.0089] and with even higher risk of fatal exacerbations amongst individuals with pre-existing COPD [n = 730; 3.61 (1.71-7.60); P = 7.3 × 10-4]. We also replicated associations for a PCSK9 variant, reported in UK Biobank, with increased risks of acute upper respiratory tract infection (URTI) [pooled OR after meta-analysis of 1.87 (1.38-2.54); P = 5.4 × 10-5] and self-reported asthma [pooled OR of 1.17 (1.04-1.30); P = 0.0071]. There was no association of a polygenic LDL-C score with COPD hospitalization, COPD exacerbation, or URTI. CONCLUSION The LDL-C-lowering PCSK9 genetic variants are associated with lower risk of subclinical and clinical atherosclerotic vascular disease but higher risks of respiratory diseases. Pharmacovigilance studies may be required to monitor patients treated with therapeutic PCSK9 inhibitors for exacerbations of respiratory diseases or respiratory tract infections. LAY SUMMARY Genetic analyses of over 100 000 participants of the China Kadoorie Biobank, mimicking the effect of new drugs intended to reduce cholesterol by targeting the PCSK9 protein, have identified potential severe effects of lower PCSK9 activity in patients with existing respiratory disease.PCSK9 genetic variants that are associated with lower cholesterol and reduced rates of cardiovascular disease are also associated with increased risk of a range of respiratory diseases, including asthma, upper respiratory tract infections, and hospitalization with chronic obstructive pulmonary disease (COPD).These genetic variants are not associated with whether or not individuals have COPD; instead, they are specifically associated with an increase in the chance of those who already have COPD being hospitalized and even dying, suggesting that careful monitoring of such patients should be considered during development of and treatment with anti-PCSK9 medication.
Collapse
Affiliation(s)
- Michael V Holmes
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
| | - Christiana Kartsonaki
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
| | - Ruth Boxall
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
| | - Kuang Lin
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
| | - Nicola Reeve
- Department of Population Health Sciences, University of Leicester, Leicester, UK
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, 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 and Response, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Derrick A Bennett
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
| | - Michael R Hill
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
| | - Ling Yang
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
| | - Yiping Chen
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
| | - Huaidong Du
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
| | - Iain Turnbull
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
| | - Rory Collins
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
| | - Robert J Clarke
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
| | - Martin D Tobin
- Department of Population Health Sciences, University of Leicester, Leicester, UK
- National Institute for Health and Care Research, Leicester Biomedical Research Centre, University of Leicester, Leicester, 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 and Response, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Iona Y Millwood
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
| | - Zhengming Chen
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
| | - Robin G Walters
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
| |
Collapse
|
10
|
Karjalainen MK, Karthikeyan S, Oliver-Williams C, Sliz E, Allara E, Fung WT, Surendran P, Zhang W, Jousilahti P, Kristiansson K, Salomaa V, Goodwin M, Hughes DA, Boehnke M, Fernandes Silva L, Yin X, Mahajan A, Neville MJ, van Zuydam NR, de Mutsert R, Li-Gao R, Mook-Kanamori DO, Demirkan A, Liu J, Noordam R, Trompet S, Chen Z, Kartsonaki C, Li L, Lin K, Hagenbeek FA, Hottenga JJ, Pool R, Ikram MA, van Meurs J, Haller T, Milaneschi Y, Kähönen M, Mishra PP, Joshi PK, Macdonald-Dunlop E, Mangino M, Zierer J, Acar IE, Hoyng CB, Lechanteur YTE, Franke L, Kurilshikov A, Zhernakova A, Beekman M, van den Akker EB, Kolcic I, Polasek O, Rudan I, Gieger C, Waldenberger M, Asselbergs FW, Hayward C, Fu J, den Hollander AI, Menni C, Spector TD, Wilson JF, Lehtimäki T, Raitakari OT, Penninx BWJH, Esko T, Walters RG, Jukema JW, Sattar N, Ghanbari M, Willems van Dijk K, Karpe F, McCarthy MI, Laakso M, Järvelin MR, Timpson NJ, Perola M, Kooner JS, Chambers JC, van Duijn C, Slagboom PE, Boomsma DI, Danesh J, Ala-Korpela M, Butterworth AS, Kettunen J. Genome-wide characterization of circulating metabolic biomarkers. Nature 2024; 628:130-138. [PMID: 38448586 PMCID: PMC10990933 DOI: 10.1038/s41586-024-07148-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/01/2024] [Indexed: 03/08/2024]
Abstract
Genome-wide association analyses using high-throughput metabolomics platforms have led to novel insights into the biology of human metabolism1-7. This detailed knowledge of the genetic determinants of systemic metabolism has been pivotal for uncovering how genetic pathways influence biological mechanisms and complex diseases8-11. Here we present a genome-wide association study for 233 circulating metabolic traits quantified by nuclear magnetic resonance spectroscopy in up to 136,016 participants from 33 cohorts. We identify more than 400 independent loci and assign probable causal genes at two-thirds of these using manual curation of plausible biological candidates. We highlight the importance of sample and participant characteristics that can have significant effects on genetic associations. We use detailed metabolic profiling of lipoprotein- and lipid-associated variants to better characterize how known lipid loci and novel loci affect lipoprotein metabolism at a granular level. We demonstrate the translational utility of comprehensively phenotyped molecular data, characterizing the metabolic associations of intrahepatic cholestasis of pregnancy. Finally, we observe substantial genetic pleiotropy for multiple metabolic pathways and illustrate the importance of careful instrument selection in Mendelian randomization analysis, revealing a putative causal relationship between acetone and hypertension. Our publicly available results provide a foundational resource for the community to examine the role of metabolism across diverse diseases.
Collapse
Affiliation(s)
- Minna K Karjalainen
- Systems Epidemiology, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland.
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland.
- Northern Finland Birth Cohorts, Arctic Biobank, Infrastructure for Population Studies, Faculty of Medicine, University of Oulu, Oulu, Finland.
| | - Savita Karthikeyan
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Clare Oliver-Williams
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Public Health Specialty Training Programme, Cambridge, UK
| | - Eeva Sliz
- Systems Epidemiology, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Elias Allara
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Wing Tung Fung
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Praveen Surendran
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Rutherford Fund Fellow, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, London North West University Healthcare NHS Trust, London, UK
| | - Pekka Jousilahti
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Kati Kristiansson
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Veikko Salomaa
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Matt Goodwin
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - David A Hughes
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Lilian Fernandes Silva
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Xianyong Yin
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Jiangsu, China
| | - Anubha Mahajan
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Genentech, South San Francisco, CA, USA
| | - Matt J Neville
- NIHR Oxford Biomedical Research Centre, OUHFT Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Natalie R van Zuydam
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, The Netherlands
| | - Ayse Demirkan
- Surrey Institute for People-Centred AI, University of Surrey, Guildford, UK
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, University of Surrey, Guildford, UK
| | - Jun Liu
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Zhengming Chen
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, University of Oxford, Oxford, UK
| | - Christiana Kartsonaki
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, 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 and Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases, Peking University, Ministry of Education, Beijing, China
| | - Kuang Lin
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Fiona A Hagenbeek
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Jouke Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - René Pool
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Joyce van Meurs
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Toomas Haller
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Mika Kähönen
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
| | - Pashupati P Mishra
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Peter K Joshi
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, Scotland
| | - Erin Macdonald-Dunlop
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, Scotland
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- NIHR Biomedical Research Centre at Guy's and St Thomas' Foundation Trust, London, UK
| | - Jonas Zierer
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Ilhan E Acar
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carel B Hoyng
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Yara T E Lechanteur
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lude Franke
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alexander Kurilshikov
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marian Beekman
- Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik B van den Akker
- Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
- Center for Computational Biology, Leiden University Medical Center, Leiden, The Netherlands
- The Delft Bioinformatics Lab, Delft University of Technology, Delft, The Netherlands
| | - Ivana Kolcic
- Department of Public Health, School of Medicine, University of Split, Split, Croatia
| | - Ozren Polasek
- Department of Public Health, School of Medicine, University of Split, Split, Croatia
| | - Igor Rudan
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, Scotland
| | - Christian Gieger
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Melanie Waldenberger
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Folkert W Asselbergs
- Amsterdam University Medical Centers, Department of Cardiology, University of Amsterdam, Amsterdam, The Netherlands
- Health Data Research UK and Institute of Health Informatics, University College London, London, UK
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Jingyuan Fu
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Anneke I den Hollander
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
- Genomics Research Center, Abbvie, Cambridge, MA, USA
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - James F Wilson
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, Scotland
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Terho Lehtimäki
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Olli T Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Tonu Esko
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Robin G Walters
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, University of Oxford, Oxford, UK
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Internal Medicine, Division Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Fredrik Karpe
- NIHR Oxford Biomedical Research Centre, OUHFT Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Mark I McCarthy
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Genentech, South San Francisco, CA, USA
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Kuopio University Hospital, Kuopio, Finland
| | - Marjo-Riitta Järvelin
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UK
- Unit of Primary Health Care, Oulu University Hospital, OYS, Oulu, Finland
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Markus Perola
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Diabetes and Obesity Research Program, University of Helsinki, Helsinki, Finland
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Jaspal S Kooner
- Department of Cardiology, Ealing Hospital, London North West University Healthcare NHS Trust, London, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - John C Chambers
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, London North West University Healthcare NHS Trust, London, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Cornelia van Duijn
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - P Eline Slagboom
- Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development (AR&D) Research Institute, Amsterdam, The Netherlands
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Mika Ala-Korpela
- Systems Epidemiology, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | - Johannes Kettunen
- Systems Epidemiology, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| |
Collapse
|
11
|
Shahjahan, Dey JK, Dey SK. Translational bioinformatics approach to combat cardiovascular disease and cancers. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 139:221-261. [PMID: 38448136 DOI: 10.1016/bs.apcsb.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Bioinformatics is an interconnected subject of science dealing with diverse fields including biology, chemistry, physics, statistics, mathematics, and computer science as the key fields to answer complicated physiological problems. Key intention of bioinformatics is to store, analyze, organize, and retrieve essential information about genome, proteome, transcriptome, metabolome, as well as organisms to investigate the biological system along with its dynamics, if any. The outcome of bioinformatics depends on the type, quantity, and quality of the raw data provided and the algorithm employed to analyze the same. Despite several approved medicines available, cardiovascular disorders (CVDs) and cancers comprises of the two leading causes of human deaths. Understanding the unknown facts of both these non-communicable disorders is inevitable to discover new pathways, find new drug targets, and eventually newer drugs to combat them successfully. Since, all these goals involve complex investigation and handling of various types of macro- and small- molecules of the human body, bioinformatics plays a key role in such processes. Results from such investigation has direct human application and thus we call this filed as translational bioinformatics. Current book chapter thus deals with diverse scope and applications of this translational bioinformatics to find cure, diagnosis, and understanding the mechanisms of CVDs and cancers. Developing complex yet small or long algorithms to address such problems is very common in translational bioinformatics. Structure-based drug discovery or AI-guided invention of novel antibodies that too with super-high accuracy, speed, and involvement of considerably low amount of investment are some of the astonishing features of the translational bioinformatics and its applications in the fields of CVDs and cancers.
Collapse
Affiliation(s)
- Shahjahan
- Laboratory for Structural Biology of Membrane Proteins, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Joy Kumar Dey
- Central Council for Research in Homoeopathy, Ministry of Ayush, Govt. of India, New Delhi, Delhi, India
| | - Sanjay Kumar Dey
- Laboratory for Structural Biology of Membrane Proteins, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India.
| |
Collapse
|
12
|
Levy FO, Osnes JB. Can acetate via FFA receptors contribute to the diabetogenic effect of statins? NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1245-1248. [PMID: 37642662 PMCID: PMC10791749 DOI: 10.1007/s00210-023-02647-7] [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: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/31/2023]
Abstract
Despite the proven effects of statins in preventing cardiovascular disease, their diabetogenic effect has caused concern. The mechanism of this diabetogenic effect is unknown. We suggest a novel mechanism that may contribute to the diabetogenic effect of statins, through an effect of statins that has apparently escaped previous consideration. Briefly, by inhibiting HMG-CoA reductase, statins may cause accumulation of acetate, which through FFA2 and FFA3 stimulation may inhibit insulin secretion.
Collapse
Affiliation(s)
- Finn Olav Levy
- Department of Pharmacology, Division of Laboratory Medicine, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Sognsvannsveien 20, P.O. Box 1057 Blindern, N-0316, Oslo, Norway.
| | - Jan-Bjørn Osnes
- Department of Pharmacology, Division of Laboratory Medicine, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Sognsvannsveien 20, P.O. Box 1057 Blindern, N-0316, Oslo, Norway
| |
Collapse
|
13
|
Wang J, Zhang S, Hu L, Wang Y, Liu K, Le J, Tan Y, Li T, Xue H, Wei Y, Zhong O, He J, Zi D, Lei X, Deng R, Luo Y, Tang M, Su M, Cao Y, Liu Q, Tang Z, Lei X. Pyrroloquinoline quinone inhibits PCSK9-NLRP3 mediated pyroptosis of Leydig cells in obese mice. Cell Death Dis 2023; 14:723. [PMID: 37935689 PMCID: PMC10630350 DOI: 10.1038/s41419-023-06162-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 08/25/2023] [Accepted: 09/19/2023] [Indexed: 11/09/2023]
Abstract
Abnormal lipid metabolism and chronic low-grade inflammation are the main traits of obesity. Especially, the molecular mechanism of concomitant deficiency in steroidogenesis-associated enzymes related to testosterone (T) synthesis of obesity dominated a decline in male fertility is still poorly understood. Here, we found that in vivo, supplementation of pyrroloquinoline quinone (PQQ) efficaciously ameliorated the abnormal lipid metabolism and testicular spermatogenic function from high-fat-diet (HFD)-induced obese mice. Moreover, the transcriptome analysis of the liver and testicular showed that PQQ supplementation not only inhibited the high expression of proprotein convertase subtilisin/Kexin type 9 (PCSK9) but also weakened the NOD-like receptor family pyrin domain containing 3 (NLRP3)-mediated pyroptosis, which both played a negative role in T synthesis of Leydig Cells (LCs). Eventually, the function and the pyroptosis of LCs cultured with palmitic acid in vitro were simultaneously benefited by suppressing the expression of NLRP3 or PCSK9 respectively, as well the parallel effects of PQQ were affirmed. Collectively, our data revealed that PQQ supplementation is a feasible approach to protect T synthesis from PCSK9-NLRP3 crosstalk-induced LCs' pyroptosis in obese men.
Collapse
Affiliation(s)
- Jinyuan Wang
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Shun Zhang
- Department of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Linlin Hu
- Reproductive Medicine Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Yan Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530004, China
| | - Ke Liu
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jianghua Le
- Department of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Yongpeng Tan
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Tianlong Li
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Haoxuan Xue
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yanhong Wei
- Reproductive Medicine Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Ou Zhong
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Junhui He
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Dan Zi
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xin Lei
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Renhe Deng
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yafei Luo
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Masong Tang
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Mingxuan Su
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yichang Cao
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Qingyou Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530004, China
| | - Zhihan Tang
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Xiaocan Lei
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
14
|
Wadström BN, Wulff AB, Pedersen KM, Nordestgaard BG. Do Triglyceride-Rich Lipoproteins Equal Low-Density Lipoproteins in Risk of ASCVD? Curr Atheroscler Rep 2023; 25:795-803. [PMID: 37768410 DOI: 10.1007/s11883-023-01153-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2023] [Indexed: 09/29/2023]
Abstract
PURPOSE OF REVIEW Recent large clinical trials have failed to show that triglyceride-rich lipoprotein-lowering therapies decrease the risk of atherosclerotic cardiovascular disease (ASCVD). In this review, we reconcile these findings with evidence showing that elevated levels of triglyceride-rich lipoproteins and the cholesterol they contain, remnant cholesterol, cause ASCVD alongside low-density lipoprotein (LDL) cholesterol. RECENT FINDINGS Results from observational epidemiology, genetic epidemiology, and randomized controlled trials indicate that lowering of remnant cholesterol and LDL cholesterol decrease ASCVD risk by a similar magnitude per 1 mmol/L (39 mg/dL) lower non-high-density lipoprotein cholesterol (remnant cholesterol+LDL cholesterol). Indeed, recent guidelines for ASCVD prevention recommend the use of non-high-density lipoprotein cholesterol instead of LDL cholesterol. Current consensus is moving towards recognizing remnant cholesterol and LDL cholesterols as equals per 1 mmol/L (39 mg/dL) higher levels in the risk assessment of ASCVD; hence, triglyceride-rich lipoprotein-lowering therapies should also lower levels of non-HDL cholesterol to reduce ASCVD risk.
Collapse
Affiliation(s)
- Benjamin N Wadström
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev Gentofte, Borgmester Ib Juuls Vej 73, entrance 7, 4th floor, N5, DK-2730, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev Gentofte, Borgmester Ib Juuls Vej 73, entrance 7, 4th floor, M3, DK-2730, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b 33.5, DK-2200, Copenhagen, Denmark
| | - Anders B Wulff
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev Gentofte, Borgmester Ib Juuls Vej 73, entrance 7, 4th floor, N5, DK-2730, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev Gentofte, Borgmester Ib Juuls Vej 73, entrance 7, 4th floor, M3, DK-2730, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b 33.5, DK-2200, Copenhagen, Denmark
| | - Kasper M Pedersen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev Gentofte, Borgmester Ib Juuls Vej 73, entrance 7, 4th floor, N5, DK-2730, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev Gentofte, Borgmester Ib Juuls Vej 73, entrance 7, 4th floor, M3, DK-2730, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b 33.5, DK-2200, Copenhagen, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev Gentofte, Borgmester Ib Juuls Vej 73, entrance 7, 4th floor, N5, DK-2730, Herlev, Denmark.
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev Gentofte, Borgmester Ib Juuls Vej 73, entrance 7, 4th floor, M3, DK-2730, Herlev, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b 33.5, DK-2200, Copenhagen, Denmark.
| |
Collapse
|
15
|
Luo S, Lam HS, Chan YH, Tang CSM, He B, Kwok MK, Leung GM, Schooling CM, Au Yeung SL. Assessing the safety of lipid-modifying medications among Chinese adolescents: a drug-target Mendelian randomization study. BMC Med 2023; 21:410. [PMID: 37904165 PMCID: PMC10617134 DOI: 10.1186/s12916-023-03115-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/16/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND With increasing hypercholesterolemia prevalence in East Asian adolescents, pharmacologic interventions (e.g., HMGCR inhibitors (statins) and PCSK9 inhibitors) may have to be considered although their longer-term safety in the general adolescent population is unclear. This study aims to investigate the longer-term safety of HMGCR inhibitors and PCSK9 inhibitors among East Asian adolescents using genetics. METHODS A drug-target Mendelian randomization study leveraging the Global Lipid Genetics Consortium (East Asian, n = 146,492) and individual-level data from Chinese participants in the Biobank clinical follow-up of Hong Kong's "Children of 1997" birth cohort (n = 3443, aged ~ 17.6 years). Safety outcomes (n = 100) included anthropometric and hematological traits, renal, liver, lung function, and other nuclear magnetic resonance metabolomics. Positive control outcomes were cholesterol markers from the "Children of 1997" birth cohort and coronary artery disease from Biobank Japan. RESULTS Genetic inhibition of HMGCR and PCSK9 were associated with reduction in cholesterol-related NMR metabolomics, e.g., apolipoprotein B (HMGCR: beta [95% CI], - 1.06 [- 1.52 to - 0.60]; PCSK9: - 0.93 [- 1.56 to - 0.31]) and had the expected effect on the positive control outcomes. After correcting for multiple comparisons (p-value < 0.006), genetic inhibition of HMGCR was associated with lower linoleic acid - 0.79 [- 1.25 to - 0.35]. Genetic inhibition of PCSK9 was not associated with the safety outcomes assessed. CONCLUSIONS Statins and PCSK9 inhibitors in East Asian adolescents appeared to be safe based on the outcomes concerned. Larger studies were warranted to verify these findings. This study serves as a proof of principle study to inform the medication safety among adolescents via genetics.
Collapse
Affiliation(s)
- Shan Luo
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 1/F, Patrick Manson Building, 7 Sassoon Road, Hong Kong SAR, China
| | - Hugh Simon Lam
- Department of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Yap Hang Chan
- Division of Cardiology, Queen Mary Hospital, The University of Hong Kong, Hong Kong SAR, China
| | - Clara Sze Man Tang
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong SAR, China
| | - Baoting He
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 1/F, Patrick Manson Building, 7 Sassoon Road, Hong Kong SAR, China
| | - Man Ki Kwok
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 1/F, Patrick Manson Building, 7 Sassoon Road, Hong Kong SAR, China
| | - Gabriel M Leung
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 1/F, Patrick Manson Building, 7 Sassoon Road, Hong Kong SAR, China
| | - C Mary Schooling
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 1/F, Patrick Manson Building, 7 Sassoon Road, Hong Kong SAR, China
- School of Public Health and Health Policy, City University of New York, New York, NY, USA
| | - Shiu Lun Au Yeung
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 1/F, Patrick Manson Building, 7 Sassoon Road, Hong Kong SAR, China.
| |
Collapse
|
16
|
Hamilton F, Pedersen KM, Ghazal P, Nordestgaard BG, Smith GD. Low levels of small HDL particles predict but do not influence risk of sepsis. Crit Care 2023; 27:389. [PMID: 37814277 PMCID: PMC10563213 DOI: 10.1186/s13054-023-04589-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/24/2023] [Indexed: 10/11/2023] Open
Abstract
BACKGROUND Low levels of high-density lipoprotein (HDL) cholesterol have been associated with higher rates and severity of infection. Alterations in inflammatory mediators and infection are associated with alterations in HDL cholesterol. It is unknown whether the association between HDL and infection is present for all particle sizes, and whether the observed associations are confounded by IL-6 signalling. METHODS In the UK Biobank, ~ 270,000 individuals have data on HDL subclasses derived from nuclear magnetic resonance analysis. We estimated the association of particle count of total HDL and HDL subclasses (small, medium, large, and extra-large HDL) with sepsis, sepsis-related death, and critical care admission in a Cox regression model. We subsequently utilised genetic data from UK Biobank and FinnGen to perform Mendelian randomisation (MR) of each HDL subclass and sepsis to test for a causal relationship. Finally, we explored the role of IL-6 signalling as a potential causal driver of changes in HDL subclasses. RESULTS In observational analyses, higher particle count of small HDL was associated with protection from sepsis (Hazard ratio, HR 0.80; 95% CI 0.74-0.86, p = 4 × 10-9 comparing Quartile 4, highest quartile of HDL to Quartile 1, lowest quartile of HDL), sepsis-related death (HR 0.80; 95% CI 0.74-0.86, p = 2 × 10-4), and critical care admission with sepsis (HR 0.72 95% CI 0.60-0.85, p = 2 × 10-4). Parallel associations with other HDL subclasses were likely driven by changes in the small HDL compartment. MR analyses did not strongly support causality of small HDL particle count on sepsis incidence (Odds ratio, OR 0.98; 95% CI 0.89-1.07, p = 0.6) or death (OR 0.94, 95% CI 0.75-1.17, p = 0.56), although the estimate on critical care admission with sepsis supported protection (OR 0.73, 95% CI 0.57-0.95, p = 0.02). Bidirectional MR analyses suggested that increased IL-6 signalling was associated with reductions in both small (beta on small HDL particle count - 0.16, 95% CI - 0.10 to - 0.21 per natural log change in SD-scaled CRP, p = 9 × 10-8).and total HDL particle count (beta - 0.13, 95% CI - 0.09 to - 0.17, p = 7 × 10-10), but that the reverse effect of HDL on IL-6 signalling was largely null. CONCLUSIONS Low number of small HDL particles are associated with increased hazard of sepsis, sepsis-related death, and sepsis-related critical care admission. However, genetic analyses did not strongly support this as causal. Instead, we demonstrate that increased IL-6 signalling, which is known to alter infection risk, could confound associations with reduced HDL particle count, and suggest this may explain part of the observed association between (small) HDL particle count and sepsis.
Collapse
Affiliation(s)
- Fergus Hamilton
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Road, Bristol, BS8 2PS, UK.
- Infection Science, North Bristol NHS Trust, Bristol, UK.
| | - Kasper Mønsted Pedersen
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Børge Grønne Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Road, Bristol, BS8 2PS, UK
| |
Collapse
|
17
|
Fung E, Chan EYS, Ng KH, Yu KM, Li H, Wang Y. Towards clinical application of GlycA and GlycB for early detection of inflammation associated with (pre)diabetes and cardiovascular disease: recent evidence and updates. J Inflamm (Lond) 2023; 20:32. [PMID: 37814278 PMCID: PMC10563214 DOI: 10.1186/s12950-023-00358-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/18/2023] [Indexed: 10/11/2023] Open
Abstract
Cardiometabolic diseases are associated with low-grade inflammation early in life and persists into old age. The long latency period presents opportunities for early detection, lifestyle modification and intervention. However, the performance of conventional biomarker assays to detect low-grade inflammation has been variable, particularly for early-stage cardiometabolic disorder including prediabetes and subclinical atherosclerotic vascular inflammation. During the last decade, the application of nuclear magnetic resonance (NMR) spectroscopy for metabolic profiling of biofluids in translational and epidemiological research has advanced to a stage approaching clinical application. Proton (1H)-NMR profiling induces no destructible physical changes to specimens, and generates quantitative signals from deconvoluted spectra that are highly repeatable and reproducible. Apart from quantitative analysis of amino acids, lipids/lipoproteins, metabolic intermediates and small proteins, 1H-NMR technology is unique in being able to detect composite signals of acute-phase and low-grade inflammation indicated by glycosylated acetyls (GlycA) and N-acetylneuraminic acid (sialic acid) moieties (GlycB). Different from conventional immunoassays that target epitopes and are susceptible to conformational variation in protein structure and binding, GlycA and GlycB signals are stable over time, and maybe complementary as well as superior to high-sensitivity C-reactive protein and other inflammatory cytokines. Here we review the physicochemical principles behind 1H-NMR profiling of GlycA and GlycB, and the available evidence supporting their potential clinical application for the prediction of incident (pre)diabetes, cardiovascular disease, and adverse outcomes.
Collapse
Affiliation(s)
- Erik Fung
- Department of Medicine & Therapeutics, Laboratory for Heart Failure + Circulation Research, Li Ka Shing Institute of Health Sciences, and Centre for Cardiovascular Genomics & Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China.
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong Children's Hospital, Kowloon Bay, Kowloon, Hong Kong SAR, China.
- Neural, Vascular, and Metabolic Biology Programme, and Ministry of Education Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China.
- Department of Epidemiology & Biostatistics, School of Public Health, St Mary's Campus, Imperial College London, London, UK.
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China.
- Prince of Wales Hospital, Room 124010, 10/F, LCWCSB, 30-32 Ngan Shing Street, Shatin, New Territories, Hong Kong SAR, China.
| | - Eunice Y S Chan
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Kwan Hung Ng
- Department of Medicine & Therapeutics, Laboratory for Heart Failure + Circulation Research, Li Ka Shing Institute of Health Sciences, and Centre for Cardiovascular Genomics & Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong Children's Hospital, Kowloon Bay, Kowloon, Hong Kong SAR, China
| | - Ka Man Yu
- Department of Medicine & Therapeutics, Laboratory for Heart Failure + Circulation Research, Li Ka Shing Institute of Health Sciences, and Centre for Cardiovascular Genomics & Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong Children's Hospital, Kowloon Bay, Kowloon, Hong Kong SAR, China
| | - Huijun Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Yulan Wang
- Singapore Phenome Centre, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
18
|
Fuller H, Zhu Y, Nicholas J, Chatelaine HA, Drzymalla EM, Sarvestani AK, Julián-Serrano S, Tahir UA, Sinnott-Armstrong N, Raffield LM, Rahnavard A, Hua X, Shutta KH, Darst BF. Metabolomic epidemiology offers insights into disease aetiology. Nat Metab 2023; 5:1656-1672. [PMID: 37872285 PMCID: PMC11164316 DOI: 10.1038/s42255-023-00903-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 09/06/2023] [Indexed: 10/25/2023]
Abstract
Metabolomic epidemiology is the high-throughput study of the relationship between metabolites and health-related traits. This emerging and rapidly growing field has improved our understanding of disease aetiology and contributed to advances in precision medicine. As the field continues to develop, metabolomic epidemiology could lead to the discovery of diagnostic biomarkers predictive of disease risk, aiding in earlier disease detection and better prognosis. In this Review, we discuss key advances facilitated by the field of metabolomic epidemiology for a range of conditions, including cardiometabolic diseases, cancer, Alzheimer's disease and COVID-19, with a focus on potential clinical utility. Core principles in metabolomic epidemiology, including study design, causal inference methods and multi-omic integration, are briefly discussed. Future directions required for clinical translation of metabolomic epidemiology findings are summarized, emphasizing public health implications. Further work is needed to establish which metabolites reproducibly improve clinical risk prediction in diverse populations and are causally related to disease progression.
Collapse
Affiliation(s)
- Harriett Fuller
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Yiwen Zhu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jayna Nicholas
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Haley A Chatelaine
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Emily M Drzymalla
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Afrand K Sarvestani
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | | | - Usman A Tahir
- Department of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ali Rahnavard
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Xinwei Hua
- Department of Cardiology, Peking University Third Hospital, Beijing, China
| | - Katherine H Shutta
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Burcu F Darst
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
19
|
Walters RG, Millwood IY, Lin K, Schmidt Valle D, McDonnell P, Hacker A, Avery D, Edris A, Fry H, Cai N, Kretzschmar WW, Ansari MA, Lyons PA, Collins R, Donnelly P, Hill M, Peto R, Shen H, Jin X, Nie C, Xu X, Guo Y, Yu C, Lv J, Clarke RJ, Li L, Chen Z. Genotyping and population characteristics of the China Kadoorie Biobank. CELL GENOMICS 2023; 3:100361. [PMID: 37601966 PMCID: PMC10435379 DOI: 10.1016/j.xgen.2023.100361] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 02/09/2023] [Accepted: 06/24/2023] [Indexed: 08/22/2023]
Abstract
The China Kadoorie Biobank (CKB) is a population-based prospective cohort of >512,000 adults recruited from 2004 to 2008 from 10 geographically diverse regions across China. Detailed data from questionnaires and physical measurements were collected at baseline, with additional measurements at three resurveys involving ∼5% of surviving participants. Analyses of genome-wide genotyping, for >100,000 participants using custom-designed Axiom arrays, reveal extensive relatedness, recent consanguinity, and signatures reflecting large-scale population movements from recent Chinese history. Systematic genome-wide association studies of incident disease, captured through electronic linkage to death and disease registries and to the national health insurance system, replicate established disease loci and identify 14 novel disease associations. Together with studies of candidate drug targets and disease risk factors and contributions to international genetics consortia, these demonstrate the breadth, depth, and quality of the CKB data. Ongoing high-throughput omics assays of collected biosamples and planned whole-genome sequencing will further enhance the scientific value of this biobank.
Collapse
Affiliation(s)
- Robin G. Walters
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
- MRC Population Health Research Unit, University of Oxford, Oxford OX3 7LF, UK
| | - Iona Y. Millwood
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
- MRC Population Health Research Unit, University of Oxford, Oxford OX3 7LF, UK
| | - Kuang Lin
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Dan Schmidt Valle
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Pandora McDonnell
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Alex Hacker
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Daniel Avery
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Ahmed Edris
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Hannah Fry
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Na Cai
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | | | - M. Azim Ansari
- Nuffield Department of Medicine, Oxford University, Oxford OX1 3SY, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Paul A. Lyons
- Cambridge Institute for Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Rory Collins
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Peter Donnelly
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Michael Hill
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
- MRC Population Health Research Unit, University of Oxford, Oxford OX3 7LF, UK
| | - Richard Peto
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Hongbing Shen
- Department of Epidemiology, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing 211116, China
| | - Xin Jin
- BGI-Shenzhen, Shenzhen 518083, China
| | - Chao Nie
- BGI-Shenzhen, Shenzhen 518083, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Yu Guo
- Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing 100037, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
- Center for Public Health and Epidemic Preparedness and Response, Peking University, Beijing 100191, China
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
- Center for Public Health and Epidemic Preparedness and Response, Peking University, Beijing 100191, China
| | - Robert J. Clarke
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
- Center for Public Health and Epidemic Preparedness and Response, Peking University, Beijing 100191, China
| | - Zhengming Chen
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
- MRC Population Health Research Unit, University of Oxford, Oxford OX3 7LF, UK
| | - China Kadoorie Biobank Collaborative Group
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
- MRC Population Health Research Unit, University of Oxford, Oxford OX3 7LF, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Nuffield Department of Medicine, Oxford University, Oxford OX1 3SY, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
- Department of Epidemiology, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing 211116, China
- BGI-Shenzhen, Shenzhen 518083, China
- Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing 100037, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
- Center for Public Health and Epidemic Preparedness and Response, Peking University, Beijing 100191, China
| |
Collapse
|
20
|
Goldstein ED, Garg G, Navarro K, Wolcott Z, Yaghi S, Wong KH, McLean K, de Havenon AH. Statin Usage Increases White Matter Hyperintensities: A Post Hoc Analysis of SPRINT-MIND. Neurologist 2023; 28:94-98. [PMID: 35680399 DOI: 10.1097/nrl.0000000000000448] [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: 11/25/2022]
Abstract
BACKGROUND Progression of white matter hyperintensities (WMHs), a radiographic marker of cerebral small vessel disease, occurs with uncontrolled conventional cerebrovascular risk factors. Less certain, however, is the influence of dyslipidemia and the impact of 3-hydroxy-3-methylglutaryl-coenzyme-A reductase inhibitors (statins) on WMH progression. The goal of this study was to evaluate the influence of statins on the progression of WMH over a 4-year interval. METHODS We performed a post hoc analysis of the SPRINT-MIND database on those with serial volumetric WMH data. WMH progression was calculated as the difference in WMH volume between the 2 scans and then segmented into tertiles due to rightward skew. We defined statin usage as no therapy (0% of visits), partial therapy (1% to 99% of visits) or full therapy (100% of visits) as logged during study visits. Analysis of variance and χ 2 tests were used for continuous and categorical variables with adjustments made for variables known to influence WMH development. RESULTS A total of 425 individuals were included in this study: 53% without statins use, 27% partial use, and 20% full use. Demographic characteristics and baseline WMH volumes were similar among the cohort. Those with full statin use were significantly more likely to be in the top tertile of WMH progression (adjusted odds ratio: 2.30, 95% confidence interval: 1.11-4.77, P =0.025), despite improvement in dyslipidemia. CONCLUSIONS SPRINT-MIND participants prescribed a statin were nearly 2.5 times more likely to be within the top tertile of WMH progression over 4 years, despite adjustment for synergistic risk factors and improvement in low-density lipoprotein.
Collapse
Affiliation(s)
- Eric D Goldstein
- Division of Stroke and Cerebrovascular Diseases, Department of Neurology, The Warren Alpert Medical School of Brown University, Providence, RI
| | - Gauri Garg
- Division of Vascular Neurology, Department of Neurology, University of Utah, Salt Lake City
| | - Kayla Navarro
- Division of Vascular Neurology, Department of Neurology, University of Utah, Salt Lake City
| | - Zoe Wolcott
- Division of Vascular Neurology, Department of Neurology, University of Utah, Salt Lake City
| | - Shadi Yaghi
- Division of Stroke and Cerebrovascular Diseases, Department of Neurology, The Warren Alpert Medical School of Brown University, Providence, RI
| | - Ka-Ho Wong
- Division of Vascular Neurology, Department of Neurology, University of Utah, Salt Lake City
| | - Kaitlin McLean
- Division of Vascular Neurology, Department of Neurology, University of Utah, Salt Lake City
| | | |
Collapse
|
21
|
Pharmacometabolomics for the Study of Lipid-Lowering Therapies: Opportunities and Challenges. Int J Mol Sci 2023; 24:ijms24043291. [PMID: 36834701 PMCID: PMC9960554 DOI: 10.3390/ijms24043291] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Lipid-lowering therapies are widely used to prevent the development of atherosclerotic cardiovascular disease (ASCVD) and related mortality worldwide. "Omics" technologies have been successfully applied in recent decades to investigate the mechanisms of action of these drugs, their pleiotropic effects, and their side effects, aiming to identify novel targets for future personalized medicine with an improvement of the efficacy and safety associated with the treatment. Pharmacometabolomics is a branch of metabolomics that is focused on the study of drug effects on metabolic pathways that are implicated in the variation of response to the treatment considering also the influences from a specific disease, environment, and concomitant pharmacological therapies. In this review, we summarized the most significant metabolomic studies on the effects of lipid-lowering therapies, including the most commonly used statins and fibrates to novel drugs or nutraceutical approaches. The integration of pharmacometabolomics data with the information obtained from the other "omics" approaches could help in the comprehension of the biological mechanisms underlying the use of lipid-lowering drugs in view of defining a precision medicine to improve the efficacy and reduce the side effects associated with the treatment.
Collapse
|
22
|
Øyri LKL, Christensen JJ, Sebert S, Thoresen M, Michelsen TM, Ulven SM, Brekke HK, Retterstøl K, Brantsæter AL, Magnus P, Bogsrud MP, Holven KB. Maternal prenatal cholesterol levels predict offspring weight trajectories during childhood in the Norwegian Mother, Father and Child Cohort Study. BMC Med 2023; 21:43. [PMID: 36747215 PMCID: PMC9903496 DOI: 10.1186/s12916-023-02742-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 01/18/2023] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Numerous intrauterine factors may affect the offspring's growth during childhood. We aimed to explore if maternal and paternal prenatal lipid, apolipoprotein (apo)B and apoA1 levels are associated with offspring weight, length, and body mass index from 6 weeks to eight years of age. This has previously been studied to a limited extent. METHODS This parental negative control study is based on the Norwegian Mother, Father and Child Cohort Study and uses data from the Medical Birth Registry of Norway. We included 713 mothers and fathers with or without self-reported hypercholesterolemia and their offspring. Seven parental metabolites were measured by nuclear magnetic resonance spectroscopy, and offspring weight and length were measured at 12 time points. Data were analyzed by linear spline mixed models, and the results are presented as the interaction between parental metabolite levels and offspring spline (age). RESULTS Higher maternal total cholesterol (TC) level was associated with a larger increase in offspring body weight up to 8 years of age (0.03 ≤ Pinteraction ≤ 0.04). Paternal TC level was not associated with change in offspring body weight (0.17 ≤ Pinteraction ≤ 0.25). Higher maternal high-density lipoprotein cholesterol (HDL-C) and apoA1 levels were associated with a lower increase in offspring body weight up to 8 years of age (0.001 ≤ Pinteraction ≤ 0.005). Higher paternal HDL-C and apoA1 levels were associated with a lower increase in offspring body weight up to 5 years of age but a larger increase in offspring body weight from 5 to 8 years of age (0.01 ≤ Pinteraction ≤ 0.03). Parental metabolites were not associated with change in offspring height or body mass index up to 8 years of age (0.07 ≤ Pinteraction ≤ 0.99). CONCLUSIONS Maternal compared to paternal TC, HDL-C, and apoA1 levels were more strongly and consistently associated with offspring body weight during childhood, supporting a direct intrauterine effect.
Collapse
Affiliation(s)
- Linn K L Øyri
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, PO Box 1046, Blindern, 0317, Oslo, Norway
| | - Jacob J Christensen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, PO Box 1046, Blindern, 0317, Oslo, Norway
| | - Sylvain Sebert
- Research Unit of Population Health, Faculty of Medicine, PO Box 5000, FI-90014 University of Oulu, Oulu, Finland
| | - Magne Thoresen
- Oslo Centre for Biostatistics and Epidemiology, Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, PO Box 1122, Blindern, 0317, Oslo, Norway
| | - Trond M Michelsen
- Department of Obstetrics, Oslo University Hospital Rikshospitalet, PO Box 4956, Nydalen, 0424, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, PO Box 1171, Blindern, 0318, Oslo, Norway
| | - Stine M Ulven
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, PO Box 1046, Blindern, 0317, Oslo, Norway
| | - Hilde K Brekke
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, PO Box 1046, Blindern, 0317, Oslo, Norway
| | - Kjetil Retterstøl
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, PO Box 1046, Blindern, 0317, Oslo, Norway.,The Lipid Clinic, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital Aker, PO Box 4959, Nydalen, 0424, Oslo, Norway
| | - Anne Lise Brantsæter
- Division of Climate and Environmental Health, Department of Food Safety, Norwegian Institute of Public Health, PO Box 222, Skøyen, 0213, Oslo, Norway
| | - Per Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, PO Box 222, Skøyen, 0213, Oslo, Norway
| | - Martin P Bogsrud
- Unit for Cardiac and Cardiovascular Genetics, Department of Medical Genetics, Oslo University Hospital Ullevål, PO Box 4956, Nydalen, 0424, Oslo, Norway
| | - Kirsten B Holven
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, PO Box 1046, Blindern, 0317, Oslo, Norway. .,Norwegian National Advisory Unit on Familial Hypercholesterolemia, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital Aker, PO Box 4959, Nydalen, 0424, Oslo, Norway.
| |
Collapse
|
23
|
Atlas of plasma NMR biomarkers for health and disease in 118,461 individuals from the UK Biobank. Nat Commun 2023; 14:604. [PMID: 36737450 PMCID: PMC9898515 DOI: 10.1038/s41467-023-36231-7] [Citation(s) in RCA: 141] [Impact Index Per Article: 70.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
Blood lipids and metabolites are markers of current health and future disease risk. Here, we describe plasma nuclear magnetic resonance (NMR) biomarker data for 118,461 participants in the UK Biobank. The biomarkers cover 249 measures of lipoprotein lipids, fatty acids, and small molecules such as amino acids, ketones, and glycolysis metabolites. We provide an atlas of associations of these biomarkers to prevalence, incidence, and mortality of over 700 common diseases ( nightingalehealth.com/atlas ). The results reveal a plethora of biomarker associations, including susceptibility to infectious diseases and risk of various cancers, joint disorders, and mental health outcomes, indicating that abundant circulating lipids and metabolites are risk markers beyond cardiometabolic diseases. Clustering analyses indicate similar biomarker association patterns across different disease types, suggesting latent systemic connectivity in the susceptibility to a diverse set of diseases. This work highlights the value of NMR based metabolic biomarker profiling in large biobanks for public health research and translation.
Collapse
|
24
|
Ritchie SC, Surendran P, Karthikeyan S, Lambert SA, Bolton T, Pennells L, Danesh J, Di Angelantonio E, Butterworth AS, Inouye M. Quality control and removal of technical variation of NMR metabolic biomarker data in ~120,000 UK Biobank participants. Sci Data 2023; 10:64. [PMID: 36720882 PMCID: PMC9887579 DOI: 10.1038/s41597-023-01949-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 01/10/2023] [Indexed: 02/01/2023] Open
Abstract
Metabolic biomarker data quantified by nuclear magnetic resonance (NMR) spectroscopy in approximately 121,000 UK Biobank participants has recently been released as a community resource, comprising absolute concentrations and ratios of 249 circulating metabolites, lipids, and lipoprotein sub-fractions. Here we identify and characterise additional sources of unwanted technical variation influencing individual biomarkers in the data available to download from UK Biobank. These included sample preparation time, shipping plate well, spectrometer batch effects, drift over time within spectrometer, and outlier shipping plates. We developed a procedure for removing this unwanted technical variation, and demonstrate that it increases signal for genetic and epidemiological studies of the NMR metabolic biomarker data in UK Biobank. We subsequently developed an R package, ukbnmr, which we make available to the wider research community to enhance the utility of the UK Biobank NMR metabolic biomarker data and to facilitate rapid analysis.
Collapse
Affiliation(s)
- Scott C Ritchie
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Cambridge Baker Systems Genomics Initiative, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia.
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Heart and Lung Research Institute, University of Cambridge, Cambridge, UK.
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK.
| | - Praveen Surendran
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Rutherford Fund Fellow, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Savita Karthikeyan
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Samuel A Lambert
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | - Thomas Bolton
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
| | - Lisa Pennells
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Emanuele Di Angelantonio
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Department of Clinical Pathology, University of Melbourne, Parkville, Victoria, Australia
- The Alan Turing Institute, London, UK
| |
Collapse
|
25
|
Rehues P, Girona J, Guardiola M, Plana N, Scicali R, Piro S, Muñiz-Grijalvo O, Díaz-Díaz JL, Recasens L, Pinyol M, Rosales R, Esteban Y, Amigó N, Masana L, Ibarretxe D, Ribalta J. PCSK9 Inhibitors Have Apolipoprotein C-III-Related Anti-Inflammatory Activity, Assessed by 1H-NMR Glycoprotein Profile in Subjects at High or very High Cardiovascular Risk. Int J Mol Sci 2023; 24:2319. [PMID: 36768645 PMCID: PMC9917120 DOI: 10.3390/ijms24032319] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/26/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease caused by the accumulation of cholesterol in the intima. Proprotein convertase subtilisin/kexin type 9 inhibitors (iPCSK9) can reduce low-density lipoprotein (LDL) cholesterol levels by 60%, but there is still no evidence that they can lower markers of systemic inflammation such as high-sensitivity C-reactive protein (hsCRP). Acute-phase serum glycoproteins are upregulated in the liver during systemic inflammation, and their role as inflammatory biomarkers is under clinical evaluation. In this observational study, we evaluate the effects of iPCSK9 on glycoproteins (Glyc) A, B and F. Thirty-nine patients eligible for iPCSK9 therapy were enrolled. One sample before and after one to six months of iPCSK9 therapy with alirocumab was obtained from each patient. Lipids, apolipoproteins, hsCRP and PCSK9 levels were measured by biochemical analyses, and the lipoprotein and glycoprotein profiles were measured by 1H nuclear magnetic resonance (1H-NMR). The PCSK9 inhibitor reduced total (36.27%, p < 0.001), LDL (55.05%, p < 0.001) and non-high-density lipoprotein (HDL) (45.11%, p < 0.001) cholesterol, apolipoprotein (apo) C-III (10%, p < 0.001), triglycerides (9.92%, p < 0.001) and glycoprotein signals GlycA (11.97%, p < 0.001), GlycB (3.83%, p = 0.017) and GlycF (7.26%, p < 0.001). It also increased apoA-I (2.05%, p = 0.043) and HDL cholesterol levels (11.58%, p < 0.001). Circulating PCSK9 levels increased six-fold (626.28%, p < 0.001). The decrease in Glyc signals positively correlated with the decrease in triglycerides and apoC-III. In conclusion, in addition to LDL cholesterol, iPCSK9 therapy also induces a reduction in systemic inflammation measured by 1H-NMR glycoprotein signals, which correlates with a decrease in triglycerides and apoC-III.
Collapse
Affiliation(s)
- Pere Rehues
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, 43201 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Josefa Girona
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, 43201 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Montse Guardiola
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, 43201 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Núria Plana
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
- Unitat de Medicina Vascular i Metabolisme, Servei de Medicina Interna, Hospital Universitari Sant Joan de Reus, 43204 Reus, Spain
| | - Roberto Scicali
- Department of Clinical and Experimental Medicine, University of Catania, 95131 Catania, Italy
| | - Salvatore Piro
- Department of Clinical and Experimental Medicine, University of Catania, 95131 Catania, Italy
| | - Ovidio Muñiz-Grijalvo
- Unidad Clinico-Experimental de Riesgo Vascular, Hospital Virgen del Rocío, 41013 Sevilla, Spain
| | - José Luis Díaz-Díaz
- Department of Internal Medicine, Complejo Hospitalario Universitario A Coruña, 15006 A Coruña, Spain
| | - Lluís Recasens
- Heart Diseases Biomedical Research Group, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain
- Cardiac Rehabilitation Unit, Department of Cardiology, Hospital del Mar, 08003 Barcelona, Spain
| | - Marta Pinyol
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, 43201 Reus, Spain
| | - Roser Rosales
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, 43201 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Yaiza Esteban
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, 43201 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Núria Amigó
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
- Biosfer Teslab, 43201 Reus, Spain
| | - Lluís Masana
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, 43201 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
- Unitat de Medicina Vascular i Metabolisme, Servei de Medicina Interna, Hospital Universitari Sant Joan de Reus, 43204 Reus, Spain
| | - Daiana Ibarretxe
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, 43201 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
- Unitat de Medicina Vascular i Metabolisme, Servei de Medicina Interna, Hospital Universitari Sant Joan de Reus, 43204 Reus, Spain
| | - Josep Ribalta
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, 43201 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| |
Collapse
|
26
|
Landfors F, Chorell E, Kersten S. Genetic Mimicry Analysis Reveals the Specific Lipases Targeted by the ANGPTL3-ANGPTL8 Complex and ANGPTL4. J Lipid Res 2023; 64:100313. [PMID: 36372100 PMCID: PMC9852701 DOI: 10.1016/j.jlr.2022.100313] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 10/14/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022] Open
Abstract
Angiopoietin-like proteins, ANGPTL3, ANGPTL4, and ANGPTL8, are involved in regulating plasma lipids. In vitro and animal-based studies point to LPL and endothelial lipase (EL, LIPG) as key targets of ANGPTLs. To examine the ANGPTL mechanisms for plasma lipid modulation in humans, we pursued a genetic mimicry analysis of enhancing or suppressing variants in the LPL, LIPG, lipase C hepatic type (LIPC), ANGPTL3, ANGPTL4, and ANGPTL8 genes using data on 248 metabolic parameters derived from over 110,000 nonfasted individuals in the UK Biobank and validated in over 13,000 overnight fasted individuals from 11 other European populations. ANGPTL4 suppression was highly concordant with LPL enhancement but not HL or EL, suggesting ANGPTL4 impacts plasma metabolic parameters exclusively via LPL. The LPL-independent effects of ANGPTL3 suppression on plasma metabolic parameters showed a striking inverse resemblance with EL suppression, suggesting ANGPTL3 not only targets LPL but also targets EL. Investigation of the impact of the ANGPTL3-ANGPTL8 complex on plasma metabolite traits via the ANGPTL8 R59W substitution as an instrumental variable showed a much higher concordance between R59W and EL activity than between R59W and LPL activity, suggesting the R59W substitution more strongly affects EL inhibition than LPL inhibition. Meanwhile, when using a rare and deleterious protein-truncating ANGPTL8 variant as an instrumental variable, the ANGPTL3-ANGPTL8 complex was very LPL specific. In conclusion, our analysis provides strong human genetic evidence that the ANGPTL3-ANGPTL8 complex regulates plasma metabolic parameters, which is achieved by impacting LPL and EL. By contrast, ANGPTL4 influences plasma metabolic parameters exclusively via LPL.
Collapse
Affiliation(s)
- Fredrik Landfors
- Department of Public Health and Clinical Medicine, Section of Medicine, Umeå University, Umeå, Sweden.
| | - Elin Chorell
- Department of Public Health and Clinical Medicine, Section of Medicine, Umeå University, Umeå, Sweden
| | - Sander Kersten
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| |
Collapse
|
27
|
Wishart DS, Cheng LL, Copié V, Edison AS, Eghbalnia HR, Hoch JC, Gouveia GJ, Pathmasiri W, Powers R, Schock TB, Sumner LW, Uchimiya M. NMR and Metabolomics-A Roadmap for the Future. Metabolites 2022; 12:678. [PMID: 35893244 PMCID: PMC9394421 DOI: 10.3390/metabo12080678] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 12/03/2022] Open
Abstract
Metabolomics investigates global metabolic alterations associated with chemical, biological, physiological, or pathological processes. These metabolic changes are measured with various analytical platforms including liquid chromatography-mass spectrometry (LC-MS), gas chromatography-mass spectrometry (GC-MS) and nuclear magnetic resonance spectroscopy (NMR). While LC-MS methods are becoming increasingly popular in the field of metabolomics (accounting for more than 70% of published metabolomics studies to date), there are considerable benefits and advantages to NMR-based methods for metabolomic studies. In fact, according to PubMed, more than 926 papers on NMR-based metabolomics were published in 2021-the most ever published in a given year. This suggests that NMR-based metabolomics continues to grow and has plenty to offer to the scientific community. This perspective outlines the growing applications of NMR in metabolomics, highlights several recent advances in NMR technologies for metabolomics, and provides a roadmap for future advancements.
Collapse
Affiliation(s)
- David S. Wishart
- Departments of Biological Sciences and Computing Science, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - Leo L. Cheng
- Department of Pathology, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA;
| | - Valérie Copié
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59715, USA;
| | - Arthur S. Edison
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA; (A.S.E.); (G.J.G.); (M.U.)
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602-0001, USA
| | - Hamid R. Eghbalnia
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT 06030-3305, USA; (H.R.E.); (J.C.H.)
| | - Jeffrey C. Hoch
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT 06030-3305, USA; (H.R.E.); (J.C.H.)
| | - Goncalo J. Gouveia
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA; (A.S.E.); (G.J.G.); (M.U.)
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602-0001, USA
| | - Wimal Pathmasiri
- Nutrition Research Institute, Department of Nutrition, School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Robert Powers
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588-0304, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE 68588-0304, USA
| | - Tracey B. Schock
- National Institute of Standards and Technology (NIST), Chemical Sciences Division, Charleston, SC 29412, USA;
| | - Lloyd W. Sumner
- Interdisciplinary Plant Group, MU Metabolomics Center, Bond Life Sciences Center, Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | - Mario Uchimiya
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA; (A.S.E.); (G.J.G.); (M.U.)
| |
Collapse
|
28
|
Ala-Korpela M, Zhao S, Järvelin MR, Mäkinen VP, Ohukainen P. Apt interpretation of comprehensive lipoprotein data in large-scale epidemiology: disclosure of fundamental structural and metabolic relationships. Int J Epidemiol 2022; 51:996-1011. [PMID: 34405869 PMCID: PMC9189959 DOI: 10.1093/ije/dyab156] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 07/09/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Quantitative lipoprotein analytics using nuclear magnetic resonance (NMR) spectroscopy is currently commonplace in large-scale studies. One methodology has become widespread and is currently being utilized also in large biobanks. It allows the comprehensive characterization of 14 lipoprotein subclasses, clinical lipids, apolipoprotein A-I and B. The details of these data are conceptualized here in relation to lipoprotein metabolism with particular attention on the fundamental characteristics of subclass particle numbers, lipid concentrations and compositional measures. METHODS AND RESULTS The NMR methodology was applied to fasting serum samples from Northern Finland Birth Cohorts 1966 and 1986 with 5651 and 5605 participants, respectively. All results were highly consistent between the cohorts. Circulating lipid concentrations in a particular lipoprotein subclass arise predominantly as the result of the circulating number of those subclass particles. The spherical lipoprotein particle shape, with a radially oriented surface monolayer, imposes size-dependent biophysical constraints for the lipid composition of individual subclass particles and inherently restricts the accommodation of metabolic changes via compositional modifications. The new finding that the relationship between lipoprotein subclass particle concentrations and the particle size is log-linear reveals that circulating lipoprotein particles are also under rather strict metabolic constraints for both their absolute and relative concentrations. CONCLUSIONS The fundamental structural and metabolic relationships between lipoprotein subclasses elucidated in this study empower detailed interpretation of lipoprotein metabolism. Understanding the intricate details of these extensive data is important for the precise interpretation of novel therapeutic opportunities and for fully utilizing the potential of forthcoming analyses of genetic and metabolic data in large biobanks.
Collapse
Affiliation(s)
- Mika Ala-Korpela
- Corresponding author. Computational Medicine, Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland. E-mail:
| | - Siyu Zhao
- Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Marjo-Riitta Järvelin
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Unit of Primary Health Care, Oulu University Hospital, OYS, Oulu, Finland
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, UK
| | - Ville-Petteri Mäkinen
- Australian Centre for Precision Health, University of South Australia, Adelaide, Australia
- Computational and Systems Biology Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Pauli Ohukainen
- Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
29
|
Richardson TG, Leyden GM, Wang Q, Bell JA, Elsworth B, Davey Smith G, Holmes MV. Characterising metabolomic signatures of lipid-modifying therapies through drug target mendelian randomisation. PLoS Biol 2022; 20:e3001547. [PMID: 35213538 PMCID: PMC8906647 DOI: 10.1371/journal.pbio.3001547] [Citation(s) in RCA: 128] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/09/2022] [Accepted: 01/19/2022] [Indexed: 12/02/2022] Open
Abstract
Large-scale molecular profiling and genotyping provide a unique opportunity to systematically compare the genetically predicted effects of therapeutic targets on the human metabolome. We firstly constructed genetic risk scores for 8 drug targets on the basis that they primarily modify low-density lipoprotein (LDL) cholesterol (HMGCR, PCKS9, and NPC1L1), high-density lipoprotein (HDL) cholesterol (CETP), or triglycerides (APOC3, ANGPTL3, ANGPTL4, and LPL). Conducting mendelian randomisation (MR) provided strong evidence of an effect of drug-based genetic scores on coronary artery disease (CAD) risk with the exception of ANGPTL3. We then systematically estimated the effects of each score on 249 metabolic traits derived using blood samples from an unprecedented sample size of up to 115,082 UK Biobank participants. Genetically predicted effects were generally consistent among drug targets, which were intended to modify the same lipoprotein lipid trait. For example, the linear fit for the MR estimates on all 249 metabolic traits for genetically predicted inhibition of LDL cholesterol lowering targets HMGCR and PCSK9 was r2 = 0.91. In contrast, comparisons between drug classes that were designed to modify discrete lipoprotein traits typically had very different effects on metabolic signatures (for instance, HMGCR versus each of the 4 triglyceride targets all had r2 < 0.02). Furthermore, we highlight this discrepancy for specific metabolic traits, for example, finding that LDL cholesterol lowering therapies typically had a weak effect on glycoprotein acetyls, a marker of inflammation, whereas triglyceride modifying therapies assessed provided evidence of a strong effect on lowering levels of this inflammatory biomarker. Our findings indicate that genetically predicted perturbations of these drug targets on the blood metabolome can drastically differ, despite largely consistent effects on risk of CAD, with potential implications for biomarkers in clinical development and measuring treatment response.
Collapse
Affiliation(s)
- Tom G. Richardson
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, United Kingdom
- Novo Nordisk Research Centre, Headington, Oxford, United Kingdom
| | - Genevieve M. Leyden
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, United Kingdom
- Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, United Kingdom
| | - Qin Wang
- MRC Population Health Research Unit (PHRU), Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Joshua A. Bell
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, United Kingdom
| | - Benjamin Elsworth
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, United Kingdom
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, United Kingdom
| | - Michael V. Holmes
- MRC Population Health Research Unit (PHRU), Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
30
|
Wei D, Marrachelli VG, Melgarejo JD, Liao CT, Janssens S, Verhamme P, Vanassche T, Van Aelst L, Monleon D, Redón J, Zhang ZY. Lipoprotein profiles of fat distribution and its association with insulin sensitivity. Front Endocrinol (Lausanne) 2022; 13:978745. [PMID: 36387872 PMCID: PMC9640977 DOI: 10.3389/fendo.2022.978745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Fat deposition is associated with adverse outcomes. Waist-to-hip (WHR) ratio is a simple feasible index to assess fat distribution. Lipoprotein particle composition in relation to WHR and to what extent their association is mediated by insulin sensitivity are less investigated. METHODS In 504 randomly recruited Flemish (mean age: 48.9 years; women: 51.6%), we analyzed the lipoprotein particle constitutions using nuclear magnetic resonance spectroscopy. WHR obesity described a WHR of ≥ 0.85 for women or 0.9 for men. Insulin sensitivity was evaluated by the homeostasis model assessment-estimated insulin resistance (HOMA-IR). SCORE-2 risk algorithm was applied to estimate 10-year cardiovascular risk. Statistical methods included multivariable-adjusted linear regression analysis, logistic regression analysis, and mediation analysis. RESULTS The prevalence of WHR obesity was 54.6%, approximately 3 times of BMI-determined obesity (19.1%). Individuals with WHR obesity had significantly higher metabolic complications, such as hypertension (57.1%), dyslipidemia (61.8%), and insulin resistance (14.2%). WHR and WHR obesity were positively associated with total very-low-density lipoprotein (VLDL) particle concentration, remnant cholesterol, and triglycerides, but were negatively associated with VLDL particle size (P ≤ 0.027), independent of body mass index and other covariates. WHR was inversely associated with total high-density lipoprotein (HDL) particle concentration, whereas WHR obesity was inversely associated with HDL cholesterol (P ≤ 0.039). Neither WHR nor WHR obesity was associated with the concentration of total low-density lipoprotein (LDL) particles, LDL particle size, and LDL cholesterol (P ≥ 0.089). In the mediation analysis, insulin sensitivity significantly mediated the effect of WHR on total VLDL particle concentration (mediation percentage: 37.0%), remnant cholesterol (47.7%), and HDL cholesterol (41.1%). Individuals with WHR obesity were at increased cardiovascular risk, regardless of LDL cholesterol (P ≤0.028). In WHR obesity, higher total VLDL particle concent36ration and remnant cholesterol, and lower HDL cholesterol were associated with an increased cardiovascular risk (P≤ 0.002). CONCLUSIONS Upper-body fat deposition was independently associated with an unfavorable lipoprotein profile, and insulin sensitivity significantly mediated this association. LDL cholesterol might underestimate lipid abnormality for people with upper-body obesity and lowering VLDL particles and remnant cholesterol might potentially reduce the residual cardiovascular risk.
Collapse
Affiliation(s)
- Dongmei Wei
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Vannina González Marrachelli
- Department of Physiology, Faculty of Medicine, University of Valencia, Valencia, Spain
- INCLIVA Research Institute, University of Valencia, Valencia, Spain
| | - Jesus D. Melgarejo
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Chia-Te Liao
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Stefan Janssens
- Division of Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Peter Verhamme
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Thomas Vanassche
- Division of Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Lucas Van Aelst
- Division of Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Daniel Monleon
- INCLIVA Research Institute, University of Valencia, Valencia, Spain
- Department of Pathology, University of Valencia, Valencia, Spain
| | - Josep Redón
- INCLIVA Research Institute, University of Valencia, Valencia, Spain
| | - Zhen-Yu Zhang
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
- *Correspondence: Zhen-Yu Zhang,
| |
Collapse
|
31
|
Martins AMA, Paiva MUB, Paiva DVN, de Oliveira RM, Machado HL, Alves LJSR, Picossi CRC, Faccio AT, Tavares MFM, Barbas C, Giraldez VZR, Santos RD, Monte GU, Atik FA. Innovative Approaches to Assess Intermediate Cardiovascular Risk Subjects: A Review From Clinical to Metabolomics Strategies. Front Cardiovasc Med 2021; 8:788062. [PMID: 35004898 PMCID: PMC8727773 DOI: 10.3389/fcvm.2021.788062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/22/2021] [Indexed: 12/31/2022] Open
Abstract
Current risk stratification strategies for coronary artery disease (CAD) have low predictive value in asymptomatic subjects classified as intermediate cardiovascular risk. This is relevant because not all coronary events occur in individuals with traditional multiple risk factors. Most importantly, the first manifestation of the disease may be either sudden cardiac death or acute coronary syndrome, after rupture and thrombosis of an unstable non-obstructive atherosclerotic plaque, which was previously silent. The inaccurate stratification using the current models may ultimately subject the individual to excessive or insufficient preventive therapies. A breakthrough in the comprehension of the molecular mechanisms governing the atherosclerosis pathology has driven many researches toward the necessity for a better risk stratification. In this Review, we discuss how metabolomics screening integrated with traditional risk assessments becomes a powerful approach to improve non-invasive CAD subclinical diagnostics. In addition, this Review highlights the findings of metabolomics studies performed by two relevant analytical platforms in current use-mass spectrometry (MS) hyphenated to separation techniques and nuclear magnetic resonance spectroscopy (NMR) -and evaluates critically the challenges for further clinical implementation of metabolomics data. We also discuss the modern understanding of the pathophysiology of atherosclerosis and the limitations of traditional analytical methods. Our aim is to show how discriminant metabolites originated from metabolomics approaches may become promising candidate molecules to aid intermediate risk patient stratification for cardiovascular events and how these tools could successfully meet the demands to translate cardiovascular metabolic biomarkers into clinical settings.
Collapse
Affiliation(s)
- Aline M. A. Martins
- Centre of Metabolomics and Bioanalysis (CEMBIO), San Pablo CEU University, Madrid, Spain
- School of Medicine, University of Brasilia, Brasilia, Brazil
- School of Medicine, University Center of Brasilia (UniCeub), Brasilia, Brazil
| | | | | | | | - Henrique L. Machado
- School of Medicine, University Center of Brasilia (UniCeub), Brasilia, Brazil
| | | | - Carolina R. C. Picossi
- Centre of Metabolomics and Bioanalysis (CEMBIO), San Pablo CEU University, Madrid, Spain
- Center for Multiplatform Metabolomics Studies (CEMM), University of Sao Paulo, São Paulo, Brazil
| | - Andréa T. Faccio
- Center for Multiplatform Metabolomics Studies (CEMM), University of Sao Paulo, São Paulo, Brazil
| | - Marina F. M. Tavares
- Center for Multiplatform Metabolomics Studies (CEMM), University of Sao Paulo, São Paulo, Brazil
| | - Coral Barbas
- Centre of Metabolomics and Bioanalysis (CEMBIO), San Pablo CEU University, Madrid, Spain
| | - Viviane Z. R. Giraldez
- Lipid Clinic, Heart Institute (InCor), University of Sao Paulo Medical School, São Paulo, Brazil
| | - Raul D. Santos
- Lipid Clinic, Heart Institute (InCor), University of Sao Paulo Medical School, São Paulo, Brazil
| | - Guilherme U. Monte
- Department of Heart Transplant, Federal District Institute of Cardiology (ICDF), Brasilia, Brazil
| | - Fernando A. Atik
- School of Medicine, University of Brasilia, Brasilia, Brazil
- Department of Heart Transplant, Federal District Institute of Cardiology (ICDF), Brasilia, Brazil
| |
Collapse
|
32
|
Kivelä J, Sormunen-Harju H, Girchenko PV, Huvinen E, Stach-Lempinen B, Kajantie E, Villa PM, Reynolds RM, Hämäläinen EK, Lahti-Pulkkinen M, Murtoniemi KK, Laivuori H, Eriksson JG, Räikkönen K, Koivusalo SB. Longitudinal Metabolic Profiling of Maternal Obesity, Gestational Diabetes, and Hypertensive Pregnancy Disorders. J Clin Endocrinol Metab 2021; 106:e4372-e4388. [PMID: 34185058 PMCID: PMC8530734 DOI: 10.1210/clinem/dgab475] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Indexed: 12/24/2022]
Abstract
CONTEXT Comprehensive assessment of metabolism in maternal obesity and pregnancy disorders can provide information about the shared maternal-fetal milieu and give insight into both maternal long-term health and intergenerational transmission of disease burden. OBJECTIVE To assess levels, profiles, and change in the levels of metabolic measures during pregnancies complicated by obesity, gestational diabetes (GDM), or hypertensive disorders. DESIGN, SETTING AND PARTICIPANTS A secondary analysis of 2 study cohorts, PREDO and RADIEL, including 741 pregnant women. MAIN OUTCOME MEASURES We assessed 225 metabolic measures by nuclear magnetic resonance in blood samples collected at median 13 [interquartile range (IQR) 12.4-13.7], 20 (IQR 19.3-23.0), and 28 (27.0-35.0) weeks of gestation. RESULTS Across all 3 time points women with obesity [body mass index (BMI) ≥ 30kg/m2] in comparison to normal weight (BMI 18.5-24.99 kg/m2) had significantly higher levels of most very-low-density lipoprotein-related measures, many fatty and most amino acids, and more adverse metabolic profiles. The change in the levels of most metabolic measures during pregnancy was smaller in obese than in normal weight women. GDM, preeclampsia, and chronic hypertension were associated with metabolic alterations similar to obesity. The associations of obesity held after adjustment for GDM and hypertensive disorders, but many of the associations with GDM and hypertensive disorders were rendered nonsignificant after adjustment for BMI and the other pregnancy disorders. CONCLUSIONS This study shows that the pregnancy-related metabolic change is smaller in women with obesity, who display metabolic perturbations already in early pregnancy. Metabolic alterations of obesity and pregnancy disorders resembled each other suggesting a shared metabolic origin.
Collapse
Affiliation(s)
- Jemina Kivelä
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Heidi Sormunen-Harju
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Polina V Girchenko
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Emilia Huvinen
- Teratology Information Service, Emergency Medicine, Department of Prehospital Emergency Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Beata Stach-Lempinen
- Department of Obstetrics and Gynecology, South Karelia Central Hospital, Lappeenranta, Finland
| | - Eero Kajantie
- PEDEGO Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Public Health Promotion Unit, Finnish Institute for Health and Welfare, Helsinki and Oulu, Finland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Children’s Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Pia M Villa
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Hyvinkää Hospital at Helsinki and Uusimaa Hospital District, Hyvinkää, Finland
| | - Rebecca M Reynolds
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Esa K Hämäläinen
- Department of Clinical Chemistry, University of Eastern Finland, Kuopio, Finland
| | - Marius Lahti-Pulkkinen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Finnish National Institute for Health and Welfare, Helsinki, Finland
| | - Katja K Murtoniemi
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Obstetrics and Gynaecology, University of Turku and Turku University Hospital, Turku, Finland
| | - Hannele Laivuori
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital and Tampere University, Faculty of Medicine and Health Technology, Tampere, Finland
| | - Johan G Eriksson
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Saila B Koivusalo
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
33
|
Letertre MPM, Giraudeau P, de Tullio P. Nuclear Magnetic Resonance Spectroscopy in Clinical Metabolomics and Personalized Medicine: Current Challenges and Perspectives. Front Mol Biosci 2021; 8:698337. [PMID: 34616770 PMCID: PMC8488110 DOI: 10.3389/fmolb.2021.698337] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022] Open
Abstract
Personalized medicine is probably the most promising area being developed in modern medicine. This approach attempts to optimize the therapies and the patient care based on the individual patient characteristics. Its success highly depends on the way the characterization of the disease and its evolution, the patient’s classification, its follow-up and the treatment could be optimized. Thus, personalized medicine must combine innovative tools to measure, integrate and model data. Towards this goal, clinical metabolomics appears as ideally suited to obtain relevant information. Indeed, the metabolomics signature brings crucial insight to stratify patients according to their responses to a pathology and/or a treatment, to provide prognostic and diagnostic biomarkers, and to improve therapeutic outcomes. However, the translation of metabolomics from laboratory studies to clinical practice remains a subsequent challenge. Nuclear magnetic resonance spectroscopy (NMR) and mass spectrometry (MS) are the two key platforms for the measurement of the metabolome. NMR has several advantages and features that are essential in clinical metabolomics. Indeed, NMR spectroscopy is inherently very robust, reproducible, unbiased, quantitative, informative at the structural molecular level, requires little sample preparation and reduced data processing. NMR is also well adapted to the measurement of large cohorts, to multi-sites and to longitudinal studies. This review focus on the potential of NMR in the context of clinical metabolomics and personalized medicine. Starting with the current status of NMR-based metabolomics at the clinical level and highlighting its strengths, weaknesses and challenges, this article also explores how, far from the initial “opposition” or “competition”, NMR and MS have been integrated and have demonstrated a great complementarity, in terms of sample classification and biomarker identification. Finally, a perspective discussion provides insight into the current methodological developments that could significantly raise NMR as a more resolutive, sensitive and accessible tool for clinical applications and point-of-care diagnosis. Thanks to these advances, NMR has a strong potential to join the other analytical tools currently used in clinical settings.
Collapse
Affiliation(s)
| | | | - Pascal de Tullio
- Metabolomics Group, Center for Interdisciplinary Research of Medicine (CIRM), Department of Pharmacy, Université de Liège, Liège, Belgique
| |
Collapse
|
34
|
Effects of dexmedetomidine, propofol, sevoflurane and S-ketamine on the human metabolome: A randomised trial using nuclear magnetic resonance spectroscopy. Eur J Anaesthesiol 2021; 39:521-532. [PMID: 34534172 DOI: 10.1097/eja.0000000000001591] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Pharmacometabolomics uses large-scale data capturing methods to uncover drug-induced shifts in the metabolic profile. The specific effects of anaesthetics on the human metabolome are largely unknown. OBJECTIVE We aimed to discover whether exposure to routinely used anaesthetics have an acute effect on the human metabolic profile. DESIGN Randomised, open-label, controlled, parallel group, phase IV clinical drug trial. SETTING The study was conducted at Turku PET Centre, University of Turku, Finland, 2016 to 2017. PARTICIPANTS One hundred and sixty healthy male volunteers were recruited. The metabolomic data of 159 were evaluable. INTERVENTIONS Volunteers were randomised to receive a 1-h exposure to equipotent doses (EC50 for verbal command) of dexmedetomidine (1.5 ng ml-1; n = 40), propofol (1.7 μg ml-1; n = 40), sevoflurane (0.9% end-tidal; n = 39), S-ketamine (0.75 μg ml-1; n = 20) or placebo (n = 20). MAIN OUTCOME MEASURES Metabolite subgroups of apolipoproteins and lipoproteins, cholesterol, glycerides and phospholipids, fatty acids, glycolysis, amino acids, ketone bodies, creatinine and albumin and the inflammatory marker GlycA, were analysed with nuclear magnetic resonance spectroscopy from arterial blood samples collected at baseline, after anaesthetic administration and 70 min postanaesthesia. RESULTS All metabolite subgroups were affected. Statistically significant changes vs. placebo were observed in 11.0, 41.3, 0.65 and 3.9% of the 155 analytes in the dexmedetomidine, propofol, sevoflurane and S-ketamine groups, respectively. Dexmedetomidine increased glucose, decreased ketone bodies and affected lipoproteins and apolipoproteins. Propofol altered lipoproteins, fatty acids, glycerides and phospholipids and slightly increased inflammatory marker glycoprotein acetylation. Sevoflurane was relatively inert. S-ketamine increased glucose and lactate, whereas branched chain amino acids and tyrosine decreased. CONCLUSION A 1-h exposure to moderate doses of routinely used anaesthetics led to significant and characteristic alterations in the metabolic profile. Dexmedetomidine-induced alterations mirror α2-adrenoceptor agonism. Propofol emulsion altered the lipid profile. The inertness of sevoflurane might prove useful in vulnerable patients. S-ketamine induced amino acid alterations might be linked to its suggested antidepressive properties. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT02624401. URL: https://clinicaltrials.gov/ct2/show/NCT02624401.
Collapse
|
35
|
Glycosylation and Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1325:307-319. [PMID: 34495542 DOI: 10.1007/978-3-030-70115-4_15] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide, accounting for approximately 18 million deaths in 2017. Coronary artery disease is the predominant cause of death from CVD, followed by stroke. Owing to recent technological advancements, glycans and glycosylation patterns of proteins have been investigated in association with CVD risk factors and clinical events. These studies have found significant associations of glycans as biomarkers of systemic inflammation and major CVD risk factors and events. While more limited, studies have also shown that glycans may be useful for monitoring response to anti-inflammatory therapies and may be responsive to changes in lifestyle, particularly in patients with chronic inflammatory diseases. Glycans capture summative risk information related to inflammatory, immune, and signaling pathways and are promising biomarkers for CVD risk prediction and therapeutic monitoring.
Collapse
|
36
|
Abstract
BACKGROUND Beyond their success in cardiovascular disease prevention, statins are increasingly recognized to have sex-specific pleiotropic effects. To gain additional insight, we characterized associations of genetically mimicked statins across the phenotype sex-specifically. We also assessed whether any apparently non-lipid effects identified extended to genetically mimicking other widely used lipid modifiers (proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors and ezetimibe) or were a consequence of low-density lipoprotein cholesterol (LDL-c). METHODS We performed a sex-specific phenome-wide association study assessing the association of genetic variants in HMGCR, mimicking statins, with 1701 phenotypes. We used Mendelian randomization (MR) to assess if any non-lipid effects found were evident for genetically mimicked PCSK9 inhibitors and ezetimibe or for LDL-c. RESULTS As expected, genetically mimicking statins was inversely associated with LDL-c, apolipoprotein B (ApoB), and total cholesterol (TC) and positively associated with glycated hemoglobin (HbA1c) and was related to body composition. Genetically mimicking statins was also inversely associated with serum calcium, sex hormone-binding globulin (SHBG), and platelet count and positively associated with basal metabolic rate (BMR) and mean platelet volume. Stronger associations with genetically mimicked statins were evident for women than men for lipid traits (LDL-c, ApoB, and TC), calcium, and SHBG, but not for platelet attributes, body composition, or BMR. Genetically mimicking PCSK9 inhibitors or ezetimibe was also associated with lower lipids, but was not related to calcium, SHBG, BMR, or body composition. Genetically higher LDL-c increased lipids and decreased BMR, but did not affect calcium, HbA1c, platelet attributes, or SHBG with minor effects on body composition. CONCLUSIONS Similar inverse associations were found for genetically mimicking statins on lipid traits in men and women as for other lipid modifiers. Besides the positive associations with HbA1c, BMI (which may explain the higher BMR), and aspects of body composition in men and women, genetically mimicking statins was additionally associated with platelet attributes in both sexes and was inversely associated with serum calcium and SHBG in women. This genetic evidence suggests potential pathways that contribute to the effects of statins particularly in women. Further investigation is needed to confirm these findings and their implications for clinical practice.
Collapse
|
37
|
Hübel C, Herle M, Santos Ferreira DL, Abdulkadir M, Bryant-Waugh R, Loos RJF, Bulik CM, Lawlor DA, Micali N. Childhood overeating is associated with adverse cardiometabolic and inflammatory profiles in adolescence. Sci Rep 2021; 11:12478. [PMID: 34127697 PMCID: PMC8203659 DOI: 10.1038/s41598-021-90644-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 05/07/2021] [Indexed: 12/15/2022] Open
Abstract
Childhood eating behaviour contributes to the rise of obesity and related noncommunicable disease worldwide. However, we lack a deep understanding of biochemical alterations that can arise from aberrant eating behaviour. In this study, we prospectively associate longitudinal trajectories of childhood overeating, undereating, and fussy eating with metabolic markers at age 16 years to explore adolescent metabolic alterations related to specific eating patterns in the first 10 years of life. Data are from the Avon Longitudinal Study of Parents and Children (n = 3104). We measure 158 metabolic markers with a high-throughput (1H) NMR metabolomics platform. Increasing childhood overeating is prospectively associated with an adverse cardiometabolic profile (i.e., hyperlipidemia, hypercholesterolemia, hyperlipoproteinemia) in adolescence; whereas undereating and fussy eating are associated with lower concentrations of the amino acids glutamine and valine, suggesting a potential lack of micronutrients. Here, we show associations between early behavioural indicators of eating and metabolic markers.
Collapse
Affiliation(s)
- Christopher Hübel
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- UK National Institute for Health Research (NIHR) Biomedical Research Centre for Mental Health, South London and Maudsley Hospital, London, UK
- National Centre for Register-Based Research, Department of Economics and Business Economics, Aarhus University, Aarhus, Denmark
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Moritz Herle
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Diana L Santos Ferreira
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Mohamed Abdulkadir
- Department of Pediatrics Gynaecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Rachel Bryant-Waugh
- Maudsley Centre for Child and Adolescent Eating Disorders, Michael Rutter Centre for Children and Young People, Maudsley Hospital, London, UK
| | - Ruth J F Loos
- Icahn School of Medicine At Mount Sinai, New York, NY, USA
| | - Cynthia M Bulik
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry, University of North Carolina At Chapel Hill, Chapel Hill, NC, USA
- Department of Nutrition, University of North Carolina At Chapel Hill, Chapel Hill, NC, USA
| | - Deborah A Lawlor
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Bristol National Institute of Health Research Biomedical Research Centre, Bristol, UK
| | - Nadia Micali
- Department of Pediatrics Gynaecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
- Great Ormond Street Institute of Child Health, University College London, London, UK.
| |
Collapse
|
38
|
Holmes MV, Richardson TG, Ference BA, Davies NM, Davey Smith G. Integrating genomics with biomarkers and therapeutic targets to invigorate cardiovascular drug development. Nat Rev Cardiol 2021; 18:435-453. [PMID: 33707768 DOI: 10.1038/s41569-020-00493-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/07/2020] [Indexed: 01/30/2023]
Abstract
Drug development in cardiovascular disease is stagnating, with lack of efficacy and adverse effects being barriers to innovation. Human genetics can provide compelling evidence of causation through approaches such as Mendelian randomization, with genetic support for causation increasing the probability of a clinical trial succeeding. Mendelian randomization applied to quantitative traits can identify risk factors for disease that are both causal and amenable to therapeutic modification. However, important differences exist between genetic investigations of a biomarker (such as HDL cholesterol) and a drug target aimed at modifying the same biomarker of interest (such as cholesteryl ester transfer protein), with implications for the methodology, interpretation and application of Mendelian randomization to drug development. Differences include the comparative nature of the genetic architecture - that is, biomarkers are typically polygenic, whereas protein drug targets are influenced by either cis-acting or trans-acting genetic variants - and the potential for drug targets to show disease associations that might differ from those of the biomarker that they are intended to modify (target-mediated pleiotropy). In this Review, we compare and contrast the use of Mendelian randomization to evaluate potential drug targets versus quantitative traits. We explain how genetic epidemiological studies can be used to assess the aetiological roles of biomarkers in disease and to prioritize drug targets, including designing their evaluation in clinical trials.
Collapse
Affiliation(s)
- Michael V Holmes
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, UK.
| | - Tom G Richardson
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Brian A Ference
- Centre for Naturally Randomised Trials, University of Cambridge, Cambridge, UK
| | - Neil M Davies
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| |
Collapse
|
39
|
Joshi A, Rienks M, Theofilatos K, Mayr M. Systems biology in cardiovascular disease: a multiomics approach. Nat Rev Cardiol 2021; 18:313-330. [PMID: 33340009 DOI: 10.1038/s41569-020-00477-1] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/02/2020] [Indexed: 12/13/2022]
Abstract
Omics techniques generate large, multidimensional data that are amenable to analysis by new informatics approaches alongside conventional statistical methods. Systems theories, including network analysis and machine learning, are well placed for analysing these data but must be applied with an understanding of the relevant biological and computational theories. Through applying these techniques to omics data, systems biology addresses the problems posed by the complex organization of biological processes. In this Review, we describe the techniques and sources of omics data, outline network theory, and highlight exemplars of novel approaches that combine gene regulatory and co-expression networks, proteomics, metabolomics, lipidomics and phenomics with informatics techniques to provide new insights into cardiovascular disease. The use of systems approaches will become necessary to integrate data from more than one omic technique. Although understanding the interactions between different omics data requires increasingly complex concepts and methods, we argue that hypothesis-driven investigations and independent validation must still accompany these novel systems biology approaches to realize their full potential.
Collapse
Affiliation(s)
- Abhishek Joshi
- King's British Heart Foundation Centre, King's College London, London, UK
- Bart's Heart Centre, St. Bartholomew's Hospital, London, UK
| | - Marieke Rienks
- King's British Heart Foundation Centre, King's College London, London, UK
| | | | - Manuel Mayr
- King's British Heart Foundation Centre, King's College London, London, UK.
| |
Collapse
|
40
|
Øyri LKL, Bogsrud MP, Christensen JJ, Ulven SM, Brantsæter AL, Retterstøl K, Brekke HK, Michelsen TM, Henriksen T, Roeters van Lennep JE, Magnus P, Veierød MB, Holven KB. Novel associations between parental and newborn cord blood metabolic profiles in the Norwegian Mother, Father and Child Cohort Study. BMC Med 2021; 19:91. [PMID: 33849542 PMCID: PMC8045233 DOI: 10.1186/s12916-021-01959-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND More than one third of Norwegian women and men between 20 and 40 years of age have elevated cholesterol concentration. Parental metabolic health around conception or during pregnancy may affect the offspring's cardiovascular disease risk. Lipids are important for fetal development, but the determinants of cord blood lipids have scarcely been studied. We therefore aimed to describe the associations between maternal and paternal peri-pregnancy lipid and metabolic profile and newborn cord blood lipid and metabolic profile. METHODS This study is based on 710 mother-father-newborn trios from the Norwegian Mother, Father and Child Cohort Study (MoBa) and uses data from the Medical Birth Registry of Norway (MBRN). The sample included in this study consisted of parents with and without self-reported hypercholesterolemia the last 6 months before pregnancy and their partners and newborns. Sixty-four cord blood metabolites detected by nuclear magnetic resonance spectroscopy were analyzed by linear mixed model analyses. The false discovery rate procedure was used to correct for multiple testing. RESULTS Among mothers with hypercholesterolemia, maternal and newborn plasma high-density lipoprotein cholesterol, apolipoprotein A1, linoleic acid, docosahexaenoic acid, alanine, glutamine, isoleucine, leucine, valine, creatinine, and particle concentration of medium high-density lipoprotein were significantly positively associated (0.001 ≤ q ≤ 0.09). Among mothers without hypercholesterolemia, maternal and newborn linoleic acid, valine, tyrosine, citrate, creatinine, high-density lipoprotein size, and particle concentration of small high-density lipoprotein were significantly positively associated (0.02 ≤ q ≤ 0.08). Among fathers with hypercholesterolemia, paternal and newborn ratio of apolipoprotein B to apolipoprotein A1 were significantly positively associated (q = 0.04). Among fathers without hypercholesterolemia, no significant associations were found between paternal and newborn metabolites. Sex differences were found for many cord blood lipids. CONCLUSIONS Maternal and paternal metabolites and newborn sex were associated with several cord blood metabolites. This may potentially affect the offspring's long-term cardiovascular disease risk.
Collapse
Affiliation(s)
- Linn K L Øyri
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1046, Blindern, 0317, Oslo, Norway
| | - Martin P Bogsrud
- Unit for Cardiac and Cardiovascular Genetics, Department of Medical Genetics, Oslo University Hospital Ullevål, PO Box 4956, Nydalen, 0424, Oslo, Norway.,Norwegian National Advisory Unit on Familial Hypercholesterolemia, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital Aker, PO Box 4959, Nydalen, 0424, Oslo, Norway
| | - Jacob J Christensen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1046, Blindern, 0317, Oslo, Norway.,Norwegian National Advisory Unit on Familial Hypercholesterolemia, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital Aker, PO Box 4959, Nydalen, 0424, Oslo, Norway
| | - Stine M Ulven
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1046, Blindern, 0317, Oslo, Norway
| | - Anne Lise Brantsæter
- Division of Infection Control and Environmental Health, Section of Environmental Exposure and Epidemiology, Norwegian Institute of Public Health, PO Box 222, Skøyen, 0213, Oslo, Norway
| | - Kjetil Retterstøl
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1046, Blindern, 0317, Oslo, Norway.,The Lipid Clinic, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital Aker, PO Box 4959, Nydalen, 0424, Oslo, Norway
| | - Hilde K Brekke
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1046, Blindern, 0317, Oslo, Norway
| | - Trond M Michelsen
- Department of Obstetrics, Oslo University Hospital Rikshospitalet, PO Box 4956, Nydalen, 0424, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, PO Box 1171, Blindern, 0318, Oslo, Norway
| | - Tore Henriksen
- Department of Obstetrics, Oslo University Hospital Rikshospitalet, PO Box 4956, Nydalen, 0424, Oslo, Norway
| | - Jeanine E Roeters van Lennep
- Department of Internal Medicine, Erasmus University Medical Center, Erasmus MC, Dr Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Per Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, PO Box 222, Skøyen, 0213, Oslo, Norway
| | - Marit B Veierød
- Oslo Centre for Biostatistics and Epidemiology, Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, PO Box 1122, Blindern, 0317, Oslo, Norway
| | - Kirsten B Holven
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1046, Blindern, 0317, Oslo, Norway. .,Norwegian National Advisory Unit on Familial Hypercholesterolemia, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital Aker, PO Box 4959, Nydalen, 0424, Oslo, Norway.
| |
Collapse
|
41
|
Cesario A, D’Oria M, Bove F, Privitera G, Boškoski I, Pedicino D, Boldrini L, Erra C, Loreti C, Liuzzo G, Crea F, Armuzzi A, Gasbarrini A, Calabresi P, Padua L, Costamagna G, Antonelli M, Valentini V, Auffray C, Scambia G. Personalized Clinical Phenotyping through Systems Medicine and Artificial Intelligence. J Pers Med 2021; 11:jpm11040265. [PMID: 33918214 PMCID: PMC8065854 DOI: 10.3390/jpm11040265] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
Personalized Medicine (PM) has shifted the traditional top-down approach to medicine based on the identification of single etiological factors to explain diseases, which was not suitable for explaining complex conditions. The concept of PM assumes several interpretations in the literature, with particular regards to Genetic and Genomic Medicine. Despite the fact that some disease-modifying genes affect disease expression and progression, many complex conditions cannot be understood through only this lens, especially when other lifestyle factors can play a crucial role (such as the environment, emotions, nutrition, etc.). Personalizing clinical phenotyping becomes a challenge when different pathophysiological mechanisms underlie the same manifestation. Brain disorders, cardiovascular and gastroenterological diseases can be paradigmatic examples. Experiences on the field of Fondazione Policlinico Gemelli in Rome (a research hospital recognized by the Italian Ministry of Health as national leader in "Personalized Medicine" and "Innovative Biomedical Technologies") could help understanding which techniques and tools are the most performing to develop potential clinical phenotypes personalization. The connection between practical experiences and scientific literature highlights how this potential can be reached towards Systems Medicine using Artificial Intelligence tools.
Collapse
Affiliation(s)
- Alfredo Cesario
- Open Innovation Unit, Scientific Directorate, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Marika D’Oria
- Open Innovation Unit, Scientific Directorate, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- Correspondence:
| | - Francesco Bove
- Neurology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.B.); (P.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giuseppe Privitera
- CEMAD—IBD Unit—Internal Medicine and Gastroenterology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (A.A.); (A.G.)
- Department of Medicine and Translational Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Ivo Boškoski
- Surgical Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (I.B.); (G.C.)
| | - Daniela Pedicino
- Cardiology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (D.P.); (G.L.); (F.C.)
| | - Luca Boldrini
- Radiation Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (L.B.); (V.V.)
| | - Carmen Erra
- High Intensity Neurorehabilitation Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (C.E.); (C.L.); (L.P.)
| | - Claudia Loreti
- High Intensity Neurorehabilitation Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (C.E.); (C.L.); (L.P.)
| | - Giovanna Liuzzo
- Cardiology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (D.P.); (G.L.); (F.C.)
| | - Filippo Crea
- Cardiology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (D.P.); (G.L.); (F.C.)
| | - Alessandro Armuzzi
- CEMAD—IBD Unit—Internal Medicine and Gastroenterology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (A.A.); (A.G.)
- Department of Medicine and Translational Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonio Gasbarrini
- CEMAD—IBD Unit—Internal Medicine and Gastroenterology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (A.A.); (A.G.)
- Department of Medicine and Translational Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Paolo Calabresi
- Neurology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.B.); (P.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Luca Padua
- High Intensity Neurorehabilitation Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (C.E.); (C.L.); (L.P.)
| | - Guido Costamagna
- Surgical Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (I.B.); (G.C.)
| | - Massimo Antonelli
- Anesthesia, Resuscitation, Intensive Care and Clinical Toxicology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Vincenzo Valentini
- Radiation Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (L.B.); (V.V.)
| | - Charles Auffray
- European Institute for Systems Biology and Medicine (EISBM), 69390 Vourles, France;
| | - Giovanni Scambia
- Scientific Directorate, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- Gynecological Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
42
|
Coelewij L, Waddington KE, Robinson GA, Chocano E, McDonnell T, Farinha F, Peng J, Dönnes P, Smith E, Croca S, Bakshi J, Griffin M, Nicolaides A, Rahman A, Jury EC, Pineda-Torra I. Serum Metabolomic Signatures Can Predict Subclinical Atherosclerosis in Patients With Systemic Lupus Erythematosus. Arterioscler Thromb Vasc Biol 2021; 41:1446-1458. [PMID: 33535791 PMCID: PMC7610443 DOI: 10.1161/atvbaha.120.315321] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/12/2020] [Indexed: 12/19/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Leda Coelewij
- Centre for Cardiometabolic and Vascular Science, Department of Medicine, University College London, London W1CE 6JF, U.K
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Kirsty E Waddington
- Centre for Cardiometabolic and Vascular Science, Department of Medicine, University College London, London W1CE 6JF, U.K
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - George A Robinson
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
- Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Elvira Chocano
- Centre for Cardiometabolic and Vascular Science, Department of Medicine, University College London, London W1CE 6JF, U.K
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Thomas McDonnell
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Filipa Farinha
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Junjie Peng
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
- Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Pierre Dönnes
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
- Scicross AB, Skövde, Sweden
| | - Edward Smith
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Sara Croca
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Jyoti Bakshi
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Maura Griffin
- Vascular Screening and Diagnostic Centre, Weymouth Street, London, UK
| | - Andrew Nicolaides
- Vascular Screening and Diagnostic Centre, Weymouth Street, London, UK
- St Georges London/Nicosia Medical School, University of Nicosia, Cyprus
| | - Anisur Rahman
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Elizabeth C Jury
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Ines Pineda-Torra
- Centre for Cardiometabolic and Vascular Science, Department of Medicine, University College London, London W1CE 6JF, U.K
| |
Collapse
|
43
|
Koh AS, Kovalik JP. Metabolomics and cardiovascular imaging: a combined approach for cardiovascular ageing. ESC Heart Fail 2021; 8:1738-1750. [PMID: 33783981 PMCID: PMC8120371 DOI: 10.1002/ehf2.13274] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 01/14/2021] [Accepted: 02/11/2021] [Indexed: 12/18/2022] Open
Abstract
The purpose of this review is to explore how metabolomics can help uncover new biomarkers and mechanisms for cardiovascular ageing. Cardiovascular ageing refers to cardiovascular structural and functional alterations that occur with chronological ageing and that can lead to the development of cardiovascular disease. These alterations, which were previously only detectable on tissue histology or corroborated on blood samples, are now detectable with modern imaging techniques. Despite the emergence of powerful new imaging tools, clinical investigation into cardiovascular ageing is challenging because ageing is a life course phenomenon involving known and unknown risk factors that play out in a dynamic fashion. Metabolomic profiling measures large numbers of metabolites with diverse chemical properties. Metabolomics has the potential to capture changes in biochemistry brought about by pathophysiologic processes as well as by normal ageing. When combined with non-invasive cardiovascular imaging tools, metabolomics can be used to understand pathological consequences of cardiovascular ageing. This review will summarize previous metabolomics and imaging studies in cardiovascular ageing. These methods may be a clinically relevant and novel approach to identify mechanisms of cardiovascular ageing and formulate or personalize treatment strategies.
Collapse
Affiliation(s)
- Angela S Koh
- National Heart Centre Singapore, 5 Hospital Drive, Singapore, 169609, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Jean-Paul Kovalik
- Duke-NUS Medical School, Singapore, Singapore.,Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
44
|
Bos MM, Goulding NJ, Lee MA, Hofman A, Bot M, Pool R, Vijfhuizen LS, Zhang X, Li C, Mustafa R, Neville MJ, Li-Gao R, Trompet S, Beekman M, Biermasz NR, Boomsma DI, de Boer I, Christodoulides C, Dehghan A, van Dijk KW, Ford I, Ghanbari M, Heijmans BT, Ikram MA, Jukema JW, Mook-Kanamori DO, Karpe F, Luik AI, Lumey LH, van den Maagdenberg AMJM, Mooijaart SP, de Mutsert R, Penninx BWJH, Rensen PCN, Richmond RC, Rosendaal FR, Sattar N, Schoevers RA, Slagboom PE, Terwindt GM, Thesing CS, Wade KH, Wijsman CA, Willemsen G, Zwinderman AH, van Heemst D, Noordam R, Lawlor DA. Investigating the relationships between unfavourable habitual sleep and metabolomic traits: evidence from multi-cohort multivariable regression and Mendelian randomization analyses. BMC Med 2021; 19:69. [PMID: 33731105 PMCID: PMC7971964 DOI: 10.1186/s12916-021-01939-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/11/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Sleep traits are associated with cardiometabolic disease risk, with evidence from Mendelian randomization (MR) suggesting that insomnia symptoms and shorter sleep duration increase coronary artery disease risk. We combined adjusted multivariable regression (AMV) and MR analyses of phenotypes of unfavourable sleep on 113 metabolomic traits to investigate possible biochemical mechanisms linking sleep to cardiovascular disease. METHODS We used AMV (N = 17,368) combined with two-sample MR (N = 38,618) to examine effects of self-reported insomnia symptoms, total habitual sleep duration, and chronotype on 113 metabolomic traits. The AMV analyses were conducted on data from 10 cohorts of mostly Europeans, adjusted for age, sex, and body mass index. For the MR analyses, we used summary results from published European-ancestry genome-wide association studies of self-reported sleep traits and of nuclear magnetic resonance (NMR) serum metabolites. We used the inverse-variance weighted (IVW) method and complemented this with sensitivity analyses to assess MR assumptions. RESULTS We found consistent evidence from AMV and MR analyses for associations of usual vs. sometimes/rare/never insomnia symptoms with lower citrate (- 0.08 standard deviation (SD)[95% confidence interval (CI) - 0.12, - 0.03] in AMV and - 0.03SD [- 0.07, - 0.003] in MR), higher glycoprotein acetyls (0.08SD [95% CI 0.03, 0.12] in AMV and 0.06SD [0.03, 0.10) in MR]), lower total very large HDL particles (- 0.04SD [- 0.08, 0.00] in AMV and - 0.05SD [- 0.09, - 0.02] in MR), and lower phospholipids in very large HDL particles (- 0.04SD [- 0.08, 0.002] in AMV and - 0.05SD [- 0.08, - 0.02] in MR). Longer total sleep duration associated with higher creatinine concentrations using both methods (0.02SD per 1 h [0.01, 0.03] in AMV and 0.15SD [0.02, 0.29] in MR) and with isoleucine in MR analyses (0.22SD [0.08, 0.35]). No consistent evidence was observed for effects of chronotype on metabolomic measures. CONCLUSIONS Whilst our results suggested that unfavourable sleep traits may not cause widespread metabolic disruption, some notable effects were observed. The evidence for possible effects of insomnia symptoms on glycoprotein acetyls and citrate and longer total sleep duration on creatinine and isoleucine might explain some of the effects, found in MR analyses of these sleep traits on coronary heart disease, which warrant further investigation.
Collapse
Affiliation(s)
- Maxime M Bos
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Neil J Goulding
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Matthew A Lee
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Amy Hofman
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Mariska Bot
- Amsterdam UMC, Vrije Universiteit, Psychiatry, Amsterdam Public Health research institute, Amsterdam, The Netherlands
| | - René Pool
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Lisanne S Vijfhuizen
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Xiang Zhang
- Department of Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Chihua Li
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, USA
| | - Rima Mustafa
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Matt J Neville
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Foundation Trust, Oxford, UK
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Oxford, UK
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Marian Beekman
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Nienke R Biermasz
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dorret I Boomsma
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Irene de Boer
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Constantinos Christodoulides
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Oxford, UK
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Dementia Research Institute at Imperial College London, London, W2 1PG, UK
- MRC Centre for Environment and Health, School of Public Health, Imperial College, London, UK
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Ian Ford
- Robertson Center for Biostatistics, University of Glasgow, Glasgow, UK
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Bastiaan T Heijmans
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, The Netherlands
| | - Fredrik Karpe
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Foundation Trust, Oxford, UK
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Oxford, UK
| | - Annemarie I Luik
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - L H Lumey
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, USA
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Arn M J M van den Maagdenberg
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Simon P Mooijaart
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Brenda W J H Penninx
- Amsterdam UMC, Vrije Universiteit, Psychiatry, Amsterdam Public Health research institute, Amsterdam, The Netherlands
| | - Patrick C N Rensen
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Rebecca C Richmond
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, Faculty of Medicine, Glasgow, UK
| | - Robert A Schoevers
- Department of Psychiatry, Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - P Eline Slagboom
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Gisela M Terwindt
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Carisha S Thesing
- Amsterdam UMC, Vrije Universiteit, Psychiatry, Amsterdam Public Health research institute, Amsterdam, The Netherlands
| | - Kaitlin H Wade
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Carolien A Wijsman
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Gonneke Willemsen
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Aeilko H Zwinderman
- Department of Clinical Epidemiology, Biostatistics, and Bioinformatics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK.
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
- NIHR Bristol Biomedical Research Centre, Bristol, UK.
| |
Collapse
|
45
|
PCSK9 Levels and Metabolic Profiles in Elderly Subjects with Different Glucose Tolerance under Statin Therapy. J Clin Med 2021; 10:jcm10050994. [PMID: 33801208 PMCID: PMC7957894 DOI: 10.3390/jcm10050994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/15/2021] [Accepted: 02/22/2021] [Indexed: 12/15/2022] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) degrades low-density lipoprotein cholesterol (LDL-C) receptors, and thus regulates the LDL-C levels in the circulation. Type 2 diabetics often have elevated LDL-C levels. However, the functions of PCSK9 in patients with alterations of glu-cose metabolism and statin therapy are still unclear. Method: we investigated a large cohort of 608 subjects, born in 1945 in Oulu, Finland (Oulu Cohort 1945). We studied the effects of PSCK9 lev-els with different glucose tolerances (normal glucose tolerance (NGT), prediabetes (PreDM) or type 2 diabetes (T2D)) with and without statin medication, and analyzed clinical data, NMR metabolomics and PCSK9 plasma levels. Results: PCSK9 plasma levels did not significantly differ between the three groups. Statin therapy significantly increased the PCSK9 levels in NGT, PreDM and T2D groups compared with subjects with no statins. In the NGT group, negative associations between PCSK9 and LDL-C, intermediate-density lipoprotein cholesterol (IDL-C), very low-density lipoprotein cholesterol (VLDL-C), total cholesterol and LDL and IDL triglycerides were observed under statin medication. In contrast, in the PreDM and T2D groups, these associa-tions were lost. Conclusions: our data suggest that in subjects with abnormal glucose metabolism and statin therapy, the significant PCSK9-mediated effects on the lipid metabolites are lost com-pared to NGT subjects, but statins reduced the LDL-C and VLDL-C levels.
Collapse
|
46
|
Robinson GA, Waddington KE, Coelewij L, Peng J, Naja M, Wincup C, Radziszewska A, Peckham H, Isenberg DA, Ioannou Y, Ciurtin C, Pineda-Torra I, Jury EC. Increased apolipoprotein-B:A1 ratio predicts cardiometabolic risk in patients with juvenile onset SLE. EBioMedicine 2021; 65:103243. [PMID: 33640328 PMCID: PMC7992074 DOI: 10.1016/j.ebiom.2021.103243] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/29/2021] [Accepted: 01/29/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cardiovascular disease is a leading cause of mortality in patients with juvenile-onset systemic lupus erythematosus (JSLE). Traditional factors for cardiovascular risk (CVR) prediction are less robust in younger patients. More reliable CVR biomarkers are needed for JSLE patient stratification and to identify therapeutic approaches to reduce cardiovascular morbidity and mortality in JSLE. METHODS Serum metabolomic analysis (including >200 lipoprotein measures) was performed on a discovery (n=31, median age 19) and validation (n=31, median age 19) cohort of JSLE patients. Data was analysed using cluster, receiver operating characteristic analysis and logistic regression. RNA-sequencing assessed gene expression in matched patient samples. FINDINGS Hierarchical clustering of lipoprotein measures identified and validated two unique JSLE groups. Group-1 had an atherogenic and Group-2 had an atheroprotective lipoprotien profile. Apolipoprotein(Apo)B:ApoA1 distinguished the two groups with high specificity (96.2%) and sensitivity (96.7%). JSLE patients with high ApoB:ApoA1 ratio had increased CD8+ T-cell frequencies and a CD8+ T-cell transcriptomic profile enriched in genes associated with atherogenic processes including interferon signaling. These metabolic and immune signatures overlapped statistically significantly with lipid biomarkers associated with sub-clinical atherosclerosis in adult SLE patients and with genes overexpressed in T-cells from human atherosclerotic plaque respectively. Finally, baseline ApoB:ApoA1 ratio correlated positively with SLE disease activity index (r=0.43, p=0.0009) and negatively with Lupus Low Disease Activity State (r=-0.43, p=0.0009) over 5-year follow-up. INTERPRETATION Multi-omic analysis identified high ApoB:ApoA1 as a potential biomarker of increased cardiometabolic risk and worse clinical outcomes in JSLE. ApoB:ApoA1 could help identify patients that require increased disease monitoring, lipid modification or lifestyle changes. FUNDING Lupus UK, The Rosetrees Trust, British Heart Foundation, UCL & Birkbeck MRC Doctoral Training Programme and Versus Arthritis.
Collapse
Affiliation(s)
- George A Robinson
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK; Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK.
| | - Kirsty E Waddington
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK; Centre for Cardiometabolic and Vascular Science, Department of Medicine, University College London, London W1CE 6JF, UK
| | - Leda Coelewij
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK; Centre for Cardiometabolic and Vascular Science, Department of Medicine, University College London, London W1CE 6JF, UK
| | - Junjie Peng
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK; Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK
| | - Meena Naja
- Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK
| | - Chris Wincup
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK
| | - Anna Radziszewska
- Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK
| | - Hannah Peckham
- Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK
| | - David A Isenberg
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK; Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK
| | - Yiannis Ioannou
- Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK
| | - Coziana Ciurtin
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK; Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK.
| | - Ines Pineda-Torra
- Centre for Cardiometabolic and Vascular Science, Department of Medicine, University College London, London W1CE 6JF, UK.
| | - Elizabeth C Jury
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK.
| |
Collapse
|
47
|
Vojinovic D, Kalaoja M, Trompet S, Fischer K, Shipley MJ, Li S, Havulinna AS, Perola M, Salomaa V, Yang Q, Sattar N, Jousilahti P, Amin N, Satizabal CL, Taba N, Sabayan B, Vasan RS, Ikram MA, Stott DJ, Ala-Korpela M, Jukema JW, Seshadri S, Kettunen J, Kivimaki M, Esko T, van Duijn CM. Association of Circulating Metabolites in Plasma or Serum and Risk of Stroke: Meta-analysis From 7 Prospective Cohorts. Neurology 2021; 96:e1110-e1123. [PMID: 33268560 PMCID: PMC8055347 DOI: 10.1212/wnl.0000000000011236] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 10/28/2020] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE To conduct a comprehensive analysis of circulating metabolites and incident stroke in large prospective population-based settings. METHODS We investigated the association of metabolites with risk of stroke in 7 prospective cohort studies including 1,791 incident stroke events among 38,797 participants in whom circulating metabolites were measured by nuclear magnetic resonance technology. The relationship between metabolites and stroke was assessed with Cox proportional hazards regression models. The analyses were performed considering all incident stroke events and ischemic and hemorrhagic events separately. RESULTS The analyses revealed 10 significant metabolite associations. Amino acid histidine (hazard ratio [HR] per SD 0.90, 95% confidence interval [CI] 0.85, 0.94; p = 4.45 × 10-5), glycolysis-related metabolite pyruvate (HR per SD 1.09, 95% CI 1.04, 1.14; p = 7.45 × 10-4), acute-phase reaction marker glycoprotein acetyls (HR per SD 1.09, 95% CI 1.03, 1.15; p = 1.27 × 10-3), cholesterol in high-density lipoprotein (HDL) 2, and several other lipoprotein particles were associated with risk of stroke. When focused on incident ischemic stroke, a significant association was observed with phenylalanine (HR per SD 1.12, 95% CI 1.05, 1.19; p = 4.13 × 10-4) and total and free cholesterol in large HDL particles. CONCLUSIONS We found association of amino acids, glycolysis-related metabolites, acute-phase reaction markers, and several lipoprotein subfractions with the risk of stroke. These findings support the potential of metabolomics to provide new insights into the metabolic changes preceding stroke.
Collapse
Affiliation(s)
- Dina Vojinovic
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Marita Kalaoja
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Stella Trompet
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Krista Fischer
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Martin J Shipley
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Shuo Li
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia.
| | - Aki S Havulinna
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Markus Perola
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Veikko Salomaa
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Qiong Yang
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Naveed Sattar
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Pekka Jousilahti
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Najaf Amin
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Claudia L Satizabal
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Nele Taba
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Behnam Sabayan
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Ramachandran S Vasan
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - M Arfan Ikram
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - David J Stott
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Mika Ala-Korpela
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - J Wouter Jukema
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Sudha Seshadri
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Johannes Kettunen
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Mika Kivimaki
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Tonu Esko
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia
| | - Cornelia M van Duijn
- From the Department of Epidemiology (D.V., N.A., M.A.I., C.M.v.D.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Computational Medicine (M. Kalaoja, M.A.-K., J.K.), Faculty of Medicine, University of Oulu and Biocenter Oulu, Finland; Departments of Gerontology and Geriatrics (S.T.), and Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands; Estonian Genome Centre (K.F., N.T., T.E.), Institute of Genomics, and Institute of Molecular and Cell Biology (N.T.), University of Tartu, Estonia; Department of Epidemiology and Public Health (M.J.S., M. Kivimaki), UCL, London, UK; Department of Biostatistics (S.L., O.Y.), School of Public Health, Boston University, MA; Department of Public Health Solutions (A.S.H., M.P., V.S., P.J., J.K.), Finnish Institute for Health and Welfare; Institute for Molecular Medicine Finland (A.S.H., M.P.), University of Helsinki; BHF Glasgow Cardiovascular Research Centre (N.S.), Faculty of Medicine, UK; Department of Neurology (B.S.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Framingham Heart Study (C.L.S., R.S.V., S.S.), MA; Department of Radiology and Nuclear Medicine (M.A.I.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Institute of Cardiovascular and Medical Sciences (D.J.S.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; Systems Epidemiology (M.A.-K.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; NMR Metabolomics Laboratory (M.A.-K.), School of Pharmacy, University of Eastern Finland, Kuopio; Population Health Science (M.A.-K.), Bristol Medical School, and Medical Research Council Integrative Epidemiology Unit (M.A.-K.), University of Bristol, UK; Department of Epidemiology and Preventive Medicine (M.A.-K.), School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia; Netherlands Heart Institute (J.W.J.), Utrecht, the Netherlands; Department of Neurology (C.L.S., S.S.), Boston University School of Medicine; Broad Institute of MIT and Harvard (T.E.), Boston, MA; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (C.L.S., S.S.), UT Health San Antonio, TX; Nuffield Department of Population Health (C.M.v.D.), University of Oxford, UK. D.V. is currently at the Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands, and K.F. is currently at the Institute of Mathematics and Statistics, University of Tartu, Estonia.
| |
Collapse
|
48
|
Bartoli-Leonard F, Aikawa E. Heart Valve Disease: Challenges and New Opportunities. Front Cardiovasc Med 2020; 7:602271. [PMID: 33195488 PMCID: PMC7642276 DOI: 10.3389/fcvm.2020.602271] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 09/09/2020] [Indexed: 01/23/2023] Open
Affiliation(s)
- Francesca Bartoli-Leonard
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Division of Cardiovascular Medicine, Department of Medicine, Center for Excellence in Vascular Biology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Department of Human Pathology, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
49
|
Baul PB, Deepak AD, Kakkar M, Modi S. Effect of Atorvastatin on blood ketone levels and glycemic control in patients with type 2 diabetes mellitus: A single arm pilot study. Diabetes Metab Syndr 2020; 14:1333-1337. [PMID: 32755832 DOI: 10.1016/j.dsx.2020.07.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 07/10/2020] [Accepted: 07/12/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND AND AIMS Cholesterol and ketone bodies are synthesized in liver from a common precursor acetyl coenzyme A (acetyl-CoA). Statins by inhibiting cholesterol synthesis may lead to accumulation of acetyl-CoA in hepatocytes and its diversion towards ketogenesis. Ketone bodies may act as alternative energy source thus sparing blood glucose and contributing to hyperglycemia. The present study aims to assess the effect of Atorvastatin therapy on blood ketone levels and glycemic control in patients with T2DM. METHODS Study included 24 statin naïve subjects with T2DM. They were prescribed tablet Atorvastatin at dose of 10 mg once daily at bedtime. Ongoing anti-diabetic medications were not changed. Estimation of blood ketones, urine ketones, fasting plasma glucose (FPG), post-prandial plasma glucose (PPG), glycated hemoglobin (HbA1c) and lipid parameters was carried out at baseline and at 3 months after starting Atorvastatin. RESULTS There was moderate but significant increase in blood ketones (0.16 ± 0.08 mmol/L vs. 0.26 ± 0.07 mmol/L; p-value = 0.0000), FPG (133.8 ± 17.91 mg/dL vs. 143.3 ± 22.99 mg/dL; p-value = 0.0016) and PPG (193.0 ± 36.54 mg/dL vs. 211.0 ± 49.51 mg/dL; p-value = 0.0344) after 3 months of Atorvastatin therapy. This was associated with significant reduction in serum total cholesterol and low density lipoprotein cholesterol. CONCLUSION Three months therapy with Atorvastatin at the dose of 10 mg once daily at bedtime in patients with T2DM resulted in moderate rise in blood ketone levels, FPG and PPG in addition to improvement in lipid parameters.
Collapse
Affiliation(s)
- Priyanka Basu Baul
- Department of Biochemistry, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun, Uttarakhand, 248106, India
| | - Arya Desh Deepak
- Department of Biochemistry, Autonomous State Medical College, Shahjahanpur, Uttar Pradesh, 242226, India
| | - Monica Kakkar
- Department of Biochemistry, NRI Academy of Medical Sciences, Mangalagiri Road, Chinakakani, Guntur, Andhra Pradesh, 522503, India
| | - Sagar Modi
- Division of Endocrinology, Department of General Medicine, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun, Uttarakhand, 248106, India.
| |
Collapse
|
50
|
Schooling CM, Zhao JV, Au Yeung SL, Leung GM. Investigating pleiotropic effects of statins on ischemic heart disease in the UK Biobank using Mendelian randomisation. eLife 2020; 9:e58567. [PMID: 32838838 PMCID: PMC7449694 DOI: 10.7554/elife.58567] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 08/13/2020] [Indexed: 12/11/2022] Open
Abstract
We examined whether specifically statins, of the major lipid modifiers (statins, proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors and ezetimibe) have pleiotropic effects on ischemic heart disease (IHD) via testosterone in men or women. As a validation, we similarly assessed whether a drug that unexpectedly likely increases IHD also operates via testosterone. Using previously published genetic instruments we conducted a sex-specific univariable and multivariable Mendelian randomization study in the UK Biobank, including 179918 men with 25410 IHD cases and 212080 women with 12511 IHD cases. Of these three lipid modifiers, only genetically mimicking the effects of statins in men affected testosterone, which partly mediated effects on IHD. Correspondingly, genetically mimicking effects of anakinra on testosterone and IHD presented a reverse pattern to that for statins. These insights may facilitate the development of new interventions for cardiovascular diseases as well as highlighting the importance of sex-specific explanations, investigations, prevention and treatment.
Collapse
Affiliation(s)
- CM Schooling
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong KongHong KongChina
- City University of New York, Graduate School of Public Health and Health PolicyNew YorkUnited States
| | - JV Zhao
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong KongHong KongChina
| | - SL Au Yeung
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong KongHong KongChina
| | - GM Leung
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong KongHong KongChina
| |
Collapse
|