1
|
Križanac AM, Reimer C, Heise J, Liu Z, Pryce JE, Bennewitz J, Thaller G, Falker-Gieske C, Tetens J. Sequence-based GWAS in 180,000 German Holstein cattle reveals new candidate variants for milk production traits. Genet Sel Evol 2025; 57:3. [PMID: 39905301 PMCID: PMC11796172 DOI: 10.1186/s12711-025-00951-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Milk production traits are complex and influenced by many genetic and environmental factors. Although extensive research has been performed for these traits, with many associations unveiled thus far, due to their crucial economic importance, complex genetic architecture, and the fact that causal variants in cattle are still scarce, there is a need for a better understanding of their genetic background. In this study, we aimed to identify new candidate loci associated with milk production traits in German Holstein cattle, the most important dairy breed in Germany and worldwide. For that purpose, 180,217 cattle were imputed to the sequence level and large-scale genome-wide association study (GWAS) followed by fine-mapping and evolutionary and functional annotation were carried out to identify and prioritize new association signals. RESULTS Using the imputed sequence data of a large cattle dataset, we identified 50,876 significant variants, confirming many known and identifying previously unreported candidate variants for milk (MY), fat (FY), and protein yield (PY). Genome-wide significant signals were fine-mapped with the Bayesian approach that determines the credible variant sets and generates the probability of causality for each signal. The variants with the highest probabilities of being causal were further classified using external information about the function and evolution, making the prioritization for subsequent validation experiments easier. The top potential causal variants determined with fine-mapping explained a large percentage of genetic variance compared to random ones; 178 variants explained 11.5%, 104 explained 7.7%, and 68 variants explained 3.9% of the variance for MY, FY, and PY, respectively, demonstrating the potential for causality. CONCLUSIONS Our findings proved the power of large samples and sequence-based GWAS in detecting new association signals. In order to fully exploit the power of GWAS, one should aim at very large samples combined with whole-genome sequence data. These can also come with both computational and time burdens, as presented in our study. Although milk production traits in cattle are comprehensively investigated, the genetic background of these traits is still not fully understood, with the potential for many new associations to be revealed, as shown. With constantly growing sample sizes, we expect more insights into the genetic architecture of milk production traits in the future.
Collapse
Affiliation(s)
- Ana-Marija Križanac
- Department of Animal Sciences, University of Goettingen, Burckhardtweg 2, 37077, Göttingen, Germany.
- Center for Integrated Breeding Research, Department of Animal Sciences, University of Goettingen, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany.
| | - Christian Reimer
- Center for Integrated Breeding Research, Department of Animal Sciences, University of Goettingen, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535, Neustadt, Germany
| | - Johannes Heise
- Vereinigte Informationssysteme Tierhaltung w.V. (VIT), 27283, Verden, Germany
| | - Zengting Liu
- Vereinigte Informationssysteme Tierhaltung w.V. (VIT), 27283, Verden, Germany
| | - Jennie E Pryce
- Agriculture Victoria Research, AgriBio, Centre for AgriBioscience, Bundoora, VIC, 3083, Australia
- School of Applied Systems Biology, La Trobe University, Bundoora, VIC, 3083, Australia
| | - Jörn Bennewitz
- Institute of Animal Science, University of Hohenheim, 70599, Stuttgart, Germany
| | - Georg Thaller
- Institute of Animal Breeding and Husbandry, Christian-Albrechts-University, 24118, Kiel, Germany
| | - Clemens Falker-Gieske
- Department of Animal Sciences, University of Goettingen, Burckhardtweg 2, 37077, Göttingen, Germany
- Center for Integrated Breeding Research, Department of Animal Sciences, University of Goettingen, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany
| | - Jens Tetens
- Department of Animal Sciences, University of Goettingen, Burckhardtweg 2, 37077, Göttingen, Germany
- Center for Integrated Breeding Research, Department of Animal Sciences, University of Goettingen, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany
| |
Collapse
|
2
|
Yilmaz A, Akyol S, Ashrafi N, Saiyed N, Turkoglu O, Graham SF. Lipidomics of Huntington's Disease: A Comprehensive Review of Current Status and Future Directions. Metabolites 2025; 15:10. [PMID: 39852353 PMCID: PMC11766911 DOI: 10.3390/metabo15010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Huntington's disease (HD) is a multifaceted neurological disorder characterized by the progressive deterioration of motor, cognitive, and psychiatric functions. Despite a limited understanding of its pathogenesis, research has implicated abnormal trinucleotide cytosine-adenine-guanine CAG repeat expansion in the huntingtin gene (HTT) as a critical factor. The development of innovative strategies is imperative for the early detection of predictive biomarkers, enabling timely intervention and mitigating irreversible cellular damage. Lipidomics, a comprehensive analytical approach, has emerged as an indispensable tool for systematically characterizing lipid profiles and elucidating their role in disease pathology. METHOD A MedLine search was performed to identify studies that use lipidomics for the characterization of HD. Search terms included "Huntington disease"; "lipidomics"; "biomarker discovery"; "NMR"; and "Mass spectrometry". RESULTS This review highlights the significance of lipidomics in HD diagnosis and treatment, exploring changes in brain lipids and their functions. Recent breakthroughs in analytical techniques, particularly mass spectrometry and NMR spectroscopy, have revolutionized brain lipidomics research, enabling researchers to gain deeper insights into the complex lipidome of the brain. CONCLUSIONS A comprehensive understanding of the broad spectrum of lipidomics alterations in HD is vital for precise diagnostic evaluation and effective disease management. The integration of lipidomics with artificial intelligence and interdisciplinary collaboration holds promise for addressing the clinical variability of HD.
Collapse
Affiliation(s)
- Ali Yilmaz
- Department of Obstetrics and Gynecology, Oakland University-William Beaumont School of Medicine, Rochester, MI 48309, USA; (A.Y.); (N.A.); (O.T.)
- Metabolomics Division, Beaumont Research Institute, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA;
| | - Sumeyya Akyol
- NX Prenatal Inc., 4350 Brownsboro Rd, Louisville, KY 40207, USA;
| | - Nadia Ashrafi
- Department of Obstetrics and Gynecology, Oakland University-William Beaumont School of Medicine, Rochester, MI 48309, USA; (A.Y.); (N.A.); (O.T.)
| | - Nazia Saiyed
- Metabolomics Division, Beaumont Research Institute, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA;
| | - Onur Turkoglu
- Department of Obstetrics and Gynecology, Oakland University-William Beaumont School of Medicine, Rochester, MI 48309, USA; (A.Y.); (N.A.); (O.T.)
| | - Stewart F. Graham
- Department of Obstetrics and Gynecology, Oakland University-William Beaumont School of Medicine, Rochester, MI 48309, USA; (A.Y.); (N.A.); (O.T.)
- Metabolomics Division, Beaumont Research Institute, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA;
| |
Collapse
|
3
|
Wunderle C, Ciobanu C, Ritz J, Tribolet P, Neyer P, Bernasconi L, Stanga Z, Mueller B, Schuetz P. Association of leucine and other branched chain amino acids with clinical outcomes in malnourished inpatients: a secondary analysis of the randomized clinical trial EFFORT. Eur J Clin Nutr 2025; 79:42-49. [PMID: 39245679 DOI: 10.1038/s41430-024-01507-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND The essential branched-chain amino acids leucine, isoleucine and valine are considered anabolic and stimulate protein synthesis in the muscles as well in the liver. They also promote muscle recovery and contribute to glucose homeostasis. Recent studies in critically ill patients have demonstrated that depletion of plasma leucine is associated with increased mortality, but data in the non-critical care setting is lacking. METHODS This secondary analysis of the randomized controlled Effect of early nutritional support on Frailty, Functional Outcomes, and Recovery of malnourished medical inpatients Trial (EFFORT), investigated the impact of leucine, isoleucine, and valine metabolism on clinical outcomes. The primary endpoint was 180-day all-cause mortality. RESULTS Among 238 polymorbid patients with available metabolite measurements, low serum leucin levels were associated with a doubled risk of 180-day all-cause mortality in a fully adjusted regression model (adjusted HR 2.20 [95% CI 1.46-3.30], p < 0.001). There was also an association with mortality for isoleucine (1.56 [95% CI 1.03-2.35], p = 0.035) and valine (1.69 [95% CI 1.13-2.53], p = 0.011). When comparing effects of nutritional support on mortality in patients with high and low levels of leucine, there was no evidence of significant differences in effectiveness of the intervention. The same was true for isoleucine and valine. CONCLUSION Our data suggest that depletion of leucine, isoleucine, and valine among malnourished polymorbid patients is associated with increases in long-term mortality. However, patients with low metabolite levels did not show a pronounced benefit from nutritional support. Further research should focus on the clinical effects of nutritional support in patients with depleted stores of essential branched-chain amino acids. CLINICAL TRIAL REGISTRATION clinicaltrials.gov as NCT02517476 (registered 7 August 2015).
Collapse
Affiliation(s)
- Carla Wunderle
- Medical University Department, Division of General Internal and Emergency Medicine, Division of Endocrinology, Diabetes and Metabolism, Kantonsspital Aarau, Aarau, Switzerland
| | - Claudia Ciobanu
- Medical University Department, Division of General Internal and Emergency Medicine, Division of Endocrinology, Diabetes and Metabolism, Kantonsspital Aarau, Aarau, Switzerland
- Medical Faculty of the University of Basel, Basel, Switzerland
| | - Jacqueline Ritz
- Medical University Department, Division of General Internal and Emergency Medicine, Division of Endocrinology, Diabetes and Metabolism, Kantonsspital Aarau, Aarau, Switzerland
- Medical Faculty of the University of Basel, Basel, Switzerland
| | - Pascal Tribolet
- Medical University Department, Division of General Internal and Emergency Medicine, Division of Endocrinology, Diabetes and Metabolism, Kantonsspital Aarau, Aarau, Switzerland
- Department of Health Professions, Bern University of Applied Sciences, Bern, Switzerland
- Department of Nutritional Sciences and Research Platform Active Ageing, University of Vienna, Vienna, Austria
| | - Peter Neyer
- Institute of Laboratory Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Luca Bernasconi
- Institute of Laboratory Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Zeno Stanga
- Division of Diabetes, Endocrinology, Nutritional Medicine, and Metabolism, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Beat Mueller
- Medical University Department, Division of General Internal and Emergency Medicine, Division of Endocrinology, Diabetes and Metabolism, Kantonsspital Aarau, Aarau, Switzerland
- Medical Faculty of the University of Basel, Basel, Switzerland
| | - Philipp Schuetz
- Medical University Department, Division of General Internal and Emergency Medicine, Division of Endocrinology, Diabetes and Metabolism, Kantonsspital Aarau, Aarau, Switzerland.
- Medical Faculty of the University of Basel, Basel, Switzerland.
| |
Collapse
|
4
|
Li M, Zhan DD, Fan LL, Wang Y, Hu XH, Zhang M, Zhou Z. Unraveling the Causal Relationship Between Blood Metabolites and Acne: A Metabolomic Mendelian Randomization Study. J Cosmet Dermatol 2025; 24:e16763. [PMID: 39737744 DOI: 10.1111/jocd.16763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/23/2024] [Accepted: 12/19/2024] [Indexed: 01/01/2025]
Abstract
BACKGROUND Acne is a common skin disorder that may be linked to metabolic dysfunction. However, the causal impact of blood metabolites on acne has not been thoroughly investigated. METHODS We performed a metabolome-wide Mendelian randomization (MR) analysis on 486 blood metabolites and acne using a genome-wide association dataset. The study included preliminary inverse-variance weighted (IVW) analysis, multivariable MR analysis, linkage disequilibrium score (LDSC) analysis, and colocalization analysis, along with reverse MR to address potential reverse causation. RESULTS Our analysis identified 12 metabolites significantly associated with acne. LDSC analysis revealed a genetic correlation between nonanoylcarnitine and acne. Colocalization analysis confirmed shared genetic variants, and metabolic pathway analysis implicated the arginine biosynthesis pathway and the selenocompound metabolism pathway in the development of acne. CONCLUSION This study offers a comprehensive understanding of the causal relationships between plasma metabolites and acne. The findings provide insights into potential biomarkers and therapeutic targets for acne treatment, underscoring the need for further research.
Collapse
Affiliation(s)
- Min Li
- Department of Dermatology, Chinese People's Liberation Army Western Theater Command General Hospital, Chengdu, China
| | - Dan Dan Zhan
- Department of Dermatology, Chinese People's Liberation Army Western Theater Command General Hospital, Chengdu, China
| | - Li Li Fan
- Department of Dermatology, Chinese People's Liberation Army Western Theater Command General Hospital, Chengdu, China
| | - Yu Wang
- Department of Dermatology, Chinese People's Liberation Army Western Theater Command General Hospital, Chengdu, China
| | - Xiao Han Hu
- Department of Dermatology, Chinese People's Liberation Army Western Theater Command General Hospital, Chengdu, China
| | - Ming Zhang
- Department of Dermatology, Chinese People's Liberation Army Western Theater Command General Hospital, Chengdu, China
| | - Zhou Zhou
- Department of Dermatology, Chinese People's Liberation Army Western Theater Command General Hospital, Chengdu, China
| |
Collapse
|
5
|
Buchmueller LC, Wunderle C, Laager R, Bernasconi L, Neyer PJ, Tribolet P, Stanga Z, Mueller B, Schuetz P. Association of phenylalanine and tyrosine metabolism with mortality and response to nutritional support among patients at nutritional risk: a secondary analysis of the randomized clinical trial EFFORT. Front Nutr 2024; 11:1451081. [PMID: 39600719 PMCID: PMC11588475 DOI: 10.3389/fnut.2024.1451081] [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: 06/18/2024] [Accepted: 10/11/2024] [Indexed: 11/29/2024] Open
Abstract
Background Elevated phenylalanine serum level is a surrogate marker of whole-body proteolysis and has been associated with increased mortality in critically ill patients. Tyrosine is a metabolite of phenylalanine and serves as a precursor of thyroid hormones and catecholamines with important functions in the oxidative stress response among others. Herein, we examined the prognostic significance of phenylalanine, tyrosine, as well as its metabolites nitrotyrosine, L-3,4-dihydroxyphenylalanine (DOPA), and dopamine regarding clinical outcomes and response to nutritional therapy in patients at nutritional risk. Methods This is a secondary analysis of the Effect of Early Nutritional Support on Frailty, Functional Outcomes, and Recovery of Malnourished Medical Inpatients Trial (EFFORT), a randomized controlled trial investigating individualized nutritional support compared to standard care in patients at risk of malnutrition. The primary outcome was 30-day all-cause mortality. Results We analyzed data of 238 patients and found a significant association between low plasma levels of phenylalanine [adjusted HR 2.27 (95% CI 1.29 to 3.00)] and tyrosine [adjusted HR 1.91 (95% CI 1.11 to 3.28)] with increased 30-day mortality. This association persisted over a longer period, extending to 5 years. Additionally, trends indicated elevated mortality rates among patients with low nitrotyrosine and high DOPA and dopamine levels. Patients with high tyrosine levels showed a more pronounced response to nutritional support compared to patients with low tyrosine levels (HR 0.45 versus 1.46, p for interaction = 0.02). Conclusion In medical inpatients at nutritional risk, low phenylalanine and tyrosine levels were associated with increased short-and long-term mortality and patients with high tyrosine levels showed a more pronounced response to nutritional support. Further research is warranted to gain a deeper understanding of phenylalanine and tyrosine pathways, their association with clinical outcomes in patients at nutritional risk, as well as their response to nutritional therapy. Clinical trial registration www.clinicaltrials.gov, identifier NCT02517476.
Collapse
Affiliation(s)
- Lena C. Buchmueller
- Medical University Department, Division of General Internal and Emergency Medicine, Division of Endocrinology, Diabetes, and Metabolism, Kantonsspital Aarau, Aarau, Switzerland
- Medical Faculty, University of Basel, Basel, Switzerland
| | - Carla Wunderle
- Medical University Department, Division of General Internal and Emergency Medicine, Division of Endocrinology, Diabetes, and Metabolism, Kantonsspital Aarau, Aarau, Switzerland
| | - Rahel Laager
- Medical University Department, Division of General Internal and Emergency Medicine, Division of Endocrinology, Diabetes, and Metabolism, Kantonsspital Aarau, Aarau, Switzerland
- Medical Faculty, University of Basel, Basel, Switzerland
- University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Luca Bernasconi
- Institute of Laboratory Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Peter J. Neyer
- Institute of Laboratory Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Pascal Tribolet
- Medical University Department, Division of General Internal and Emergency Medicine, Division of Endocrinology, Diabetes, and Metabolism, Kantonsspital Aarau, Aarau, Switzerland
- Department of Health Professions, Bern University of Applied Sciences, Bern, Switzerland
- Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Zeno Stanga
- Division of Diabetes, Endocrinology, Nutritional Medicine, and Metabolism, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Beat Mueller
- Medical University Department, Division of General Internal and Emergency Medicine, Division of Endocrinology, Diabetes, and Metabolism, Kantonsspital Aarau, Aarau, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
| | - Philipp Schuetz
- Medical University Department, Division of General Internal and Emergency Medicine, Division of Endocrinology, Diabetes, and Metabolism, Kantonsspital Aarau, Aarau, Switzerland
- Medical Faculty, University of Basel, Basel, Switzerland
| |
Collapse
|
6
|
Weinisch P, Raffler J, Römisch-Margl W, Arnold M, Mohney RP, Rist MJ, Prehn C, Skurk T, Hauner H, Daniel H, Suhre K, Kastenmüller G. The HuMet Repository: Watching human metabolism at work. Cell Rep 2024; 43:114416. [PMID: 39033506 PMCID: PMC11513335 DOI: 10.1016/j.celrep.2024.114416] [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: 11/09/2023] [Revised: 05/11/2024] [Accepted: 06/13/2024] [Indexed: 07/23/2024] Open
Abstract
Metabolism oscillates between catabolic and anabolic states depending on food intake, exercise, or stresses that change a multitude of metabolic pathways simultaneously. We present the HuMet Repository for exploring dynamic metabolic responses to oral glucose/lipid loads, mixed meals, 36-h fasting, exercise, and cold stress in healthy subjects. Metabolomics data from blood, urine, and breath of 15 young, healthy men at up to 56 time points are integrated and embedded within an interactive web application, enabling researchers with and without computational expertise to search, visualize, analyze, and contextualize the dynamic metabolite profiles of 2,656 metabolites acquired on multiple platforms. With examples, we demonstrate the utility of the resource for research into the dynamics of human metabolism, highlighting differences and similarities in systemic metabolic responses across challenges and the complementarity of metabolomics platforms. The repository, providing a reference for healthy metabolite changes to six standardized physiological challenges, is freely accessible through a web portal.
Collapse
Affiliation(s)
- Patrick Weinisch
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Johannes Raffler
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; Digital Medicine, University Hospital of Augsburg, Augsburg, Germany
| | - Werner Römisch-Margl
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Matthias Arnold
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | | | - Manuela J Rist
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, Karlsruhe, Germany
| | - Cornelia Prehn
- Metabolomics and Proteomics Core, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Skurk
- ZIEL Institute for Food and Health, Core Facility Human Studies, Technical University of Munich, Freising, Germany; Else Kröner Fresenius Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Hans Hauner
- Else Kröner Fresenius Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising, Germany; Institute for Nutritional Medicine, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Hannelore Daniel
- School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Karsten Suhre
- Department of Biophysics and Physiology, Weill Cornell Medicine - Qatar, Doha, Qatar
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
7
|
Wunderle C, Haller L, Laager R, Bernasconi L, Neyer P, Stumpf F, Tribolet P, Stanga Z, Mueller B, Schuetz P. The Association of the Essential Amino Acids Lysine, Methionine, and Threonine with Clinical Outcomes in Patients at Nutritional Risk: Secondary Analysis of a Randomized Clinical Trial. Nutrients 2024; 16:2608. [PMID: 39203745 PMCID: PMC11357570 DOI: 10.3390/nu16162608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
Lysine, methionine, and threonine are essential amino acids with vital functions for muscle and connective tissue health, metabolic balance, and the immune system. During illness, the demand for these amino acids typically increases, which puts patients at risk for deficiencies with harmful clinical consequences. In a secondary analysis of the Effect of Early Nutritional Support on Frailty, Functional Outcomes, and Recovery of Malnourished Medical Inpatients Trial (EFFORT), which compared individualized nutritional support to usual care nutrition in patients at nutritional risk, we investigated the prognostic impact of the lysine, methionine, and threonine metabolism. We had complete clinical and amino acid data in 237 patients, 58 of whom reached the primary endpoint of death at 30 days. In a model adjusted for comorbidities, sex, nutritional risk, and trial intervention, low plasma methionine levels were associated with 30-day mortality (adjusted HR 1.98 [95% CI 1.16 to 3.36], p = 0.01) and with a decline in functional status (adjusted OR 2.06 [95% CI 1.06 to 4.01], p = 0.03). The results for lysine and threonine did not show statistically significant differences regarding clinical outcomes. These findings suggest that low levels of methionine may be critical during hospitalization among patients at nutritional risk. Further studies should investigate the effect of supplementation of methionine in this patient group to improve outcomes.
Collapse
Affiliation(s)
- Carla Wunderle
- University Department of Medicine, Internal and Emergency Medicine, Cantonal Hospital Aarau, 5001 Aarau, Switzerland; (C.W.); (F.S.)
| | - Luana Haller
- University Department of Medicine, Internal and Emergency Medicine, Cantonal Hospital Aarau, 5001 Aarau, Switzerland; (C.W.); (F.S.)
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Rahel Laager
- University Department of Medicine, Internal and Emergency Medicine, Cantonal Hospital Aarau, 5001 Aarau, Switzerland; (C.W.); (F.S.)
- Medical Faculty, University of Basel, 4056 Basel, Switzerland
- University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, 3010 Bern, Switzerland
| | - Luca Bernasconi
- Institute of Laboratory Medicine, Cantonal Hospital Aarau, 5001 Aarau, Switzerland
| | - Peter Neyer
- Institute of Laboratory Medicine, Cantonal Hospital Aarau, 5001 Aarau, Switzerland
| | - Franziska Stumpf
- University Department of Medicine, Internal and Emergency Medicine, Cantonal Hospital Aarau, 5001 Aarau, Switzerland; (C.W.); (F.S.)
| | - Pascal Tribolet
- University Department of Medicine, Internal and Emergency Medicine, Cantonal Hospital Aarau, 5001 Aarau, Switzerland; (C.W.); (F.S.)
- Department of Health Professions, Bern University of Applied Sciences, 3008 Bern, Switzerland
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sport Sciences, University of Vienna, 1030 Vienna, Austria
| | - Zeno Stanga
- Department of Diabetology, Endocrinology, Nutritional Medicine, and Metabolism, University Hospital and University of Bern, 3010 Bern, Switzerland
| | - Beat Mueller
- University Department of Medicine, Internal and Emergency Medicine, Cantonal Hospital Aarau, 5001 Aarau, Switzerland; (C.W.); (F.S.)
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), 6900 Lugano, Switzerland
| | - Philipp Schuetz
- University Department of Medicine, Internal and Emergency Medicine, Cantonal Hospital Aarau, 5001 Aarau, Switzerland; (C.W.); (F.S.)
- Medical Faculty, University of Basel, 4056 Basel, Switzerland
| |
Collapse
|
8
|
Stumpf F, Wunderle C, Ritz J, Bernasconi L, Neyer P, Tribolet P, Stanga Z, Mueller B, Bischoff SC, Schuetz P. Prognostic implications of the arginine metabolism in patients at nutritional risk: A secondary analysis of the randomized EFFORT trial. Clin Nutr 2024; 43:660-673. [PMID: 38309228 DOI: 10.1016/j.clnu.2024.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/26/2023] [Accepted: 01/14/2024] [Indexed: 02/05/2024]
Abstract
BACKGROUND Arginine, a conditionally essential amino acid, is key component in metabolic pathways including immune regulation and protein synthesis. Depletion of arginine contributes to worse outcomes in severely ill and surgical patient populations. We assessed prognostic implications of arginine levels and its metabolites and ratios in polymorbid medical inpatients at nutritional risk regarding clinical outcomes and treatment response. METHODS Within this secondary analysis of the randomized controlled Effect of early nutritional support on Frailty, Functional Outcomes, and Recovery of malnourished medical inpatients Trial (EFFORT), we investigated the association of arginine, its metabolites and ratios (i.e., ADMA and SDMA, ratios of arginine/ADMA, arginine/ornithine, and global arginine bioavailability ratio) measured on hospital admission with short-term and long-term mortality by means of regression analysis. RESULTS Among the 231 patients with available measurements, low arginine levels ≤90.05 μmol/l (n = 86; 37 %) were associated with higher all-cause mortality at 30 days (primary endpoint, adjusted HR 3.27, 95 % CI 1.86 to 5.75, p < 0.001) and at 5 years (adjusted HR 1.50, 95 % CI 1.07 to 2.12, p = 0.020). Arginine metabolites and ratios were also associated with adverse outcome, but had lower prognostic value. There was, however, no evidence that treatment response was influenced by admission arginine levels. CONCLUSION This secondary analysis focusing on medical inpatients at nutritional risk confirms a strong association of low plasma arginine levels and worse clinical courses. The potential effects of arginine-enriched nutritional supplements should be investigated in this population of patients. CLINICAL TRIAL REGISTRATION clinicaltrials.gov as NCT02517476 (registered 7 August 2015).
Collapse
Affiliation(s)
- Franziska Stumpf
- Medical University Department, Division of General Internal and Emergency Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland; Institute of Nutritional Medicine, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
| | - Carla Wunderle
- Medical University Department, Division of General Internal and Emergency Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland
| | - Jacqueline Ritz
- Medical University Department, Division of General Internal and Emergency Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland
| | - Luca Bernasconi
- Institute of Laboratory Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland
| | - Peter Neyer
- Institute of Laboratory Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland
| | - Pascal Tribolet
- Medical University Department, Division of General Internal and Emergency Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland; Department of Health Professions, Bern University of Applied Sciences, Falkenplatz 24, 3012 Bern, Switzerland; Faculty of Life Sciences University of Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - Zeno Stanga
- Division of Diabetes, Endocrinology, Nutritional Medicine, and Metabolism, Bern University Hospital and University of Bern, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Beat Mueller
- Medical University Department, Division of General Internal and Emergency Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland; Medical Faculty of the University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland
| | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
| | - Philipp Schuetz
- Medical University Department, Division of General Internal and Emergency Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland; Medical Faculty of the University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland.
| |
Collapse
|
9
|
Ritz J, Wunderle C, Stumpf F, Laager R, Tribolet P, Neyer P, Bernasconi L, Stanga Z, Mueller B, Schuetz P. Association of tryptophan pathway metabolites with mortality and effectiveness of nutritional support among patients at nutritional risk: secondary analysis of a randomized clinical trial. Front Nutr 2024; 11:1335242. [PMID: 38425485 PMCID: PMC10902466 DOI: 10.3389/fnut.2024.1335242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
Tryptophan is an essential amino acid and is the precursor of many important metabolites and neurotransmitters. In malnutrition, the availability of tryptophan is reduced, potentially putting patients at increased risks. Herein, we investigated the prognostic implications of the tryptophan metabolism in a secondary analysis of the Effect of Early Nutritional Support on Frailty, Functional Outcomes, and Recovery of Malnourished Medical Inpatients Trial (EFFORT), a randomized, controlled trial comparing individualized nutritional support to usual care in patients at risk for malnutrition. Among 238 patients with available measurements, low plasma levels of metabolites were independently associated with 30-day mortality with adjusted hazard ratios (HR) of 1.77 [95% CI 1.05-2.99, p 0.034] for tryptophan, 3.49 [95% CI 1.81-6.74, p < 0.001] for kynurenine and 2.51 [95% CI 1.37-4.63, p 0.003] for serotonin. Nutritional support had more beneficial effects on mortality in patients with high tryptophan compared to patients with low tryptophan levels (adjusted HR 0.61 [95% CI 0.29-1.29] vs. HR 1.72 [95% CI 0.79-3.70], p for interaction 0.047). These results suggest that sufficient circulating levels of tryptophan might be a metabolic prerequisite for the beneficial effect of nutritional interventions in this highly vulnerable patient population.
Collapse
Affiliation(s)
- Jacqueline Ritz
- Medical University Department, Division of General Internal and Emergency Medicine, Cantonal Hospital Aarau, Aarau, Switzerland
- Medical Faculty of the University of Basel, Basel, Switzerland
| | - Carla Wunderle
- Medical University Department, Division of General Internal and Emergency Medicine, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Franziska Stumpf
- Medical University Department, Division of General Internal and Emergency Medicine, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Rahel Laager
- Medical University Department, Division of General Internal and Emergency Medicine, Cantonal Hospital Aarau, Aarau, Switzerland
- Medical Faculty of the University of Basel, Basel, Switzerland
| | - Pascal Tribolet
- Medical University Department, Division of General Internal and Emergency Medicine, Cantonal Hospital Aarau, Aarau, Switzerland
- Department of Health Professions, Bern University of Applied Sciences, Bern, Switzerland
- Department of Nutritional Sciences and Research Platform Active Aging, University of Vienna, Vienna, Austria
| | - Peter Neyer
- Institute of Laboratory Medicine, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Luca Bernasconi
- Institute of Laboratory Medicine, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Zeno Stanga
- Division of Diabetes, Endocrinology, Nutritional Medicine, and Metabolism, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Beat Mueller
- Medical University Department, Division of General Internal and Emergency Medicine, Cantonal Hospital Aarau, Aarau, Switzerland
- Medical Faculty of the University of Basel, Basel, Switzerland
| | - Philipp Schuetz
- Medical University Department, Division of General Internal and Emergency Medicine, Cantonal Hospital Aarau, Aarau, Switzerland
- Medical Faculty of the University of Basel, Basel, Switzerland
| |
Collapse
|
10
|
Louck LE, Cara KC, Klatt K, Wallace TC, Chung M. The Relationship of Circulating Choline and Choline-Related Metabolite Levels with Health Outcomes: A Scoping Review of Genome-Wide Association Studies and Mendelian Randomization Studies. Adv Nutr 2024; 15:100164. [PMID: 38128611 PMCID: PMC10819410 DOI: 10.1016/j.advnut.2023.100164] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023] Open
Abstract
Choline is essential for proper liver, muscle, brain, lipid metabolism, cellular membrane composition, and repair. Understanding genetic determinants of circulating choline metabolites can help identify new determinants of choline metabolism, requirements, and their link to disease endpoints. We conducted a scoping review to identify studies assessing the association of genetic polymorphisms on circulating choline and choline-related metabolite concentrations and subsequent associations with health outcomes. This study follows the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement scoping review extension. Literature was searched to September 28, 2022, in 4 databases: Embase, MEDLINE, Web of Science, and the Biological Science Index. Studies of any duration in humans were considered. Any genome-wide association study (GWAS) investigating genetic variant associations with circulating choline and/or choline-related metabolites and any Mendelian randomization (MR) study investigating the association of genetically predicted circulating choline and/or choline-related metabolites with any health outcome were considered. Qualitative evidence is presented in summary tables. From 1248 total reviewed articles, 53 were included (GWAS = 27; MR = 26). Forty-two circulating choline-related metabolites were tested in association with genetic variants in GWAS studies, primarily trimethylamine N-oxide, betaine, sphingomyelins, lysophosphatidylcholines, and phosphatidylcholines. MR studies investigated associations between 52 total unique choline metabolites and 66 unique health outcomes. Of these, 47 significant associations were reported between 16 metabolites (primarily choline, lysophosphatidylcholines, phosphatidylcholines, betaine, and sphingomyelins) and 27 health outcomes including cancer, cardiovascular, metabolic, bone, and brain-related outcomes. Some articles reported significant associations between multiple choline types and the same health outcome. Genetically predicted circulating choline and choline-related metabolite concentrations are associated with a wide variety of health outcomes. Further research is needed to assess how genetic variability influences choline metabolism and whether individuals with lower genetically predicted circulating choline and choline-related metabolite concentrations would benefit from a dietary intervention or supplementation.
Collapse
Affiliation(s)
- Lauren E Louck
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, United States
| | - Kelly C Cara
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, United States
| | - Kevin Klatt
- Nutritional Sciences and Toxicology, University of California, Berkeley, CA, United States
| | - Taylor C Wallace
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, United States; Think Health Group, Inc, Washington, DC, United States
| | - Mei Chung
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, United States.
| |
Collapse
|
11
|
Wunderle C, von Arx D, Mueller SC, Bernasconi L, Neyer P, Tribolet P, Stanga Z, Mueller B, Schuetz P. Association of Glutamine and Glutamate Metabolism with Mortality among Patients at Nutritional Risk-A Secondary Analysis of the Randomized Clinical Trial EFFORT. Nutrients 2024; 16:222. [PMID: 38257115 PMCID: PMC10821126 DOI: 10.3390/nu16020222] [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: 12/07/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Glutamine and its metabolite glutamate serve as the main energy substrates for immune cells, and their plasma levels drop during severe illness. Therefore, glutamine supplementation in the critical care setting has been advocated. However, little is known about glutamine metabolism in severely but not critically ill medical patients. We investigated the prognostic impact of glutamine metabolism in a secondary analysis of the Effect of Early Nutritional Support on Frailty, Functional Outcomes, and Recovery of Malnourished Medical Inpatients Trial (EFFORT), a randomized controlled trial comparing individualized nutritional support to usual care in patients at nutritional risk. Among 234 patients with available measurements, low plasma levels of glutamate were independently associated with 30-day mortality (adjusted HR 2.35 [95% CI 1.18-4.67, p = 0.015]). The impact on mortality remained consistent long-term for up to 5 years. No significant association was found for circulating glutamine levels and short- or long-term mortality. There was no association of glutamate nor glutamine with malnutrition parameters or with the effectiveness of nutritional support. This secondary analysis found glutamate to be independently prognostic among medical inpatients at nutritional risk but poorly associated with the effectiveness of nutritional support. In contrast to ICU studies, we found no association between glutamine and clinical outcome.
Collapse
Affiliation(s)
- Carla Wunderle
- Division of General Internal and Emergency Medicine, University Department of Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland; (C.W.); (B.M.)
| | - Diana von Arx
- Medical Faculty, University of Basel, 4056 Basel, Switzerland
| | - Sydney Chiara Mueller
- Medical Faculty, University of Basel, 4056 Basel, Switzerland
- Faculty of Biomedical Scienes, Università della Svizzera italiana (USI), 6900 Lugano, Switzerland
| | - Luca Bernasconi
- Institute of Laboratory Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland (P.N.)
| | - Peter Neyer
- Institute of Laboratory Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland (P.N.)
| | - Pascal Tribolet
- Division of General Internal and Emergency Medicine, University Department of Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland; (C.W.); (B.M.)
- Department of Health Professions, Bern University of Applied Sciences, Murtenstrasse 10, 3008 Bern, Switzerland
- Faculty of Life Sciences, University of Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - Zeno Stanga
- Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism (UDEM), Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Beat Mueller
- Division of General Internal and Emergency Medicine, University Department of Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland; (C.W.); (B.M.)
- Faculty of Biomedical Scienes, Università della Svizzera italiana (USI), 6900 Lugano, Switzerland
| | - Philipp Schuetz
- Division of General Internal and Emergency Medicine, University Department of Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland; (C.W.); (B.M.)
- Medical Faculty, University of Basel, 4056 Basel, Switzerland
| |
Collapse
|
12
|
Akyol S, Ashrafi N, Yilmaz A, Turkoglu O, Graham SF. Metabolomics: An Emerging "Omics" Platform for Systems Biology and Its Implications for Huntington Disease Research. Metabolites 2023; 13:1203. [PMID: 38132886 PMCID: PMC10744751 DOI: 10.3390/metabo13121203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/29/2023] [Accepted: 12/02/2023] [Indexed: 12/23/2023] Open
Abstract
Huntington's disease (HD) is a progressive, fatal neurodegenerative disease characterized by motor, cognitive, and psychiatric symptoms. The precise mechanisms of HD progression are poorly understood; however, it is known that there is an expansion of the trinucleotide cytosine-adenine-guanine (CAG) repeat in the Huntingtin gene. Important new strategies are of paramount importance to identify early biomarkers with predictive value for intervening in disease progression at a stage when cellular dysfunction has not progressed irreversibly. Metabolomics is the study of global metabolite profiles in a system (cell, tissue, or organism) under certain conditions and is becoming an essential tool for the systemic characterization of metabolites to provide a snapshot of the functional and pathophysiological states of an organism and support disease diagnosis and biomarker discovery. This review briefly highlights the historical progress of metabolomic methodologies, followed by a more detailed review of the use of metabolomics in HD research to enable a greater understanding of the pathogenesis, its early prediction, and finally the main technical platforms in the field of metabolomics.
Collapse
Affiliation(s)
- Sumeyya Akyol
- NX Prenatal Inc., 4350 Brownsboro Road, Louisville KY 40207, USA;
| | - Nadia Ashrafi
- Department of Obstetrics and Gynecology, Oakland University-William Beaumont School of Medicine, 318 Meadow Brook Road, Rochester, MI 48309, USA; (N.A.); (A.Y.); (O.T.)
| | - Ali Yilmaz
- Department of Obstetrics and Gynecology, Oakland University-William Beaumont School of Medicine, 318 Meadow Brook Road, Rochester, MI 48309, USA; (N.A.); (A.Y.); (O.T.)
- Metabolomics Division, Beaumont Research Institute, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA
| | - Onur Turkoglu
- Department of Obstetrics and Gynecology, Oakland University-William Beaumont School of Medicine, 318 Meadow Brook Road, Rochester, MI 48309, USA; (N.A.); (A.Y.); (O.T.)
| | - Stewart F. Graham
- Department of Obstetrics and Gynecology, Oakland University-William Beaumont School of Medicine, 318 Meadow Brook Road, Rochester, MI 48309, USA; (N.A.); (A.Y.); (O.T.)
- Metabolomics Division, Beaumont Research Institute, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA
| |
Collapse
|
13
|
Weinisch P, Raffler J, Römisch-Margl W, Arnold M, Mohney RP, Rist MJ, Prehn C, Skurk T, Hauner H, Daniel H, Suhre K, Kastenmüller G. The HuMet Repository: Watching human metabolism at work. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.08.550079. [PMID: 37609175 PMCID: PMC10441358 DOI: 10.1101/2023.08.08.550079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
The human metabolism constantly responds to stimuli such as food intake, fasting, exercise, and stress, triggering adaptive biochemical processes across multiple metabolic pathways. To understand the role of these processes and disruptions thereof in health and disease, detailed documentation of healthy metabolic responses is needed but still scarce on a time-resolved metabolome-wide level. Here, we present the HuMet Repository, a web-based resource for exploring dynamic metabolic responses to six physiological challenges (exercise, 36 h fasting, oral glucose and lipid loads, mixed meal, cold stress) in healthy subjects. For building this resource, we integrated existing and newly derived metabolomics data measured in blood, urine, and breath samples of 15 young healthy men at up to 56 time points during the six highly standardized challenge tests conducted over four days. The data comprise 1.1 million data points acquired on multiple platforms with temporal profiles of 2,656 metabolites from a broad range of biochemical pathways. By embedding the dataset into an interactive web application, we enable users to easily access, search, filter, analyze, and visualize the time-resolved metabolomic readouts and derived results. Users can put metabolites into their larger context by identifying metabolites with similar trajectories or by visualizing metabolites within holistic metabolic networks to pinpoint pathways of interest. In three showcases, we outline the value of the repository for gaining biological insights and generating hypotheses by analyzing the wash-out of dietary markers, the complementarity of metabolomics platforms in dynamic versus cross-sectional data, and similarities and differences in systemic metabolic responses across challenges. With its comprehensive collection of time-resolved metabolomics data, the HuMet Repository, freely accessible at https://humet.org/, is a reference for normal, healthy responses to metabolic challenges in young males. It will enable researchers with and without computational expertise, to flexibly query the data for their own research into the dynamics of human metabolism.
Collapse
Affiliation(s)
- Patrick Weinisch
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Johannes Raffler
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Digital Medicine, University Hospital of Augsburg, Augsburg, Germany
| | - Werner Römisch-Margl
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Matthias Arnold
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | | | - Manuela J. Rist
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, Karlsruhe, Germany
| | - Cornelia Prehn
- Metabolomics and Proteomics Core, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Skurk
- ZIEL Institute for Food and Health, Core Facility Human Studies, Technical University of Munich, Freising, Germany
- Else Kröner Fresenius Center of Nutritional Medicine, Department of Food and Nutrition, Technical University of Munich, Freising, Germany
| | - Hans Hauner
- Else Kröner Fresenius Center of Nutritional Medicine, Department of Food and Nutrition, Technical University of Munich, Freising, Germany
- Institute for Nutritional Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Hannelore Daniel
- Department of Food and Nutrition, Technical University of Munich, Freising, Germany
| | - Karsten Suhre
- Department of Biophysics and Physiology, Weill Cornell Medicine - Qatar, Doha, Qatar
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
14
|
Rahman M, Schellhorn HE. Metabolomics of infectious diseases in the era of personalized medicine. Front Mol Biosci 2023; 10:1120376. [PMID: 37275959 PMCID: PMC10233009 DOI: 10.3389/fmolb.2023.1120376] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 05/08/2023] [Indexed: 06/07/2023] Open
Abstract
Infectious diseases continue to be a major cause of morbidity and mortality worldwide. Diseases cause perturbation of the host's immune system provoking a response that involves genes, proteins and metabolites. While genes are regulated by epigenetic or other host factors, proteins can undergo post-translational modification to enable/modify function. As a result, it is difficult to correlate the disease phenotype based solely on genetic and proteomic information only. Metabolites, however, can provide direct information on the biochemical activity during diseased state. Therefore, metabolites may, potentially, represent a phenotypic signature of a diseased state. Measuring and assessing metabolites in large scale falls under the omics technology known as "metabolomics". Comprehensive and/or specific metabolic profiling in biological fluids can be used as biomarkers of disease diagnosis. In addition, metabolomics together with genomics can be used to differentiate patients with differential treatment response and development of host targeted therapy instead of pathogen targeted therapy where pathogens are more prone to mutation and lead to antimicrobial resistance. Thus, metabolomics can be used for patient stratification, personalized drug formulation and disease control and management. Currently, several therapeutics and in vitro diagnostics kits have been approved by US Food and Drug Administration (FDA) for personalized treatment and diagnosis of infectious diseases. However, the actual number of therapeutics or diagnostics kits required for tailored treatment is limited as metabolomics and personalized medicine require the involvement of personnel from multidisciplinary fields ranging from technological development, bioscience, bioinformatics, biostatistics, clinicians, and biotechnology companies. Given the significance of metabolomics, in this review, we discussed different aspects of metabolomics particularly potentials of metabolomics as diagnostic biomarkers and use of small molecules for host targeted treatment for infectious diseases, and their scopes and challenges in personalized medicine.
Collapse
|
15
|
Hong KU, Walls KM, Hein DW. Non-coding and intergenic genetic variants of human arylamine N-acetyltransferase 2 (NAT2) gene are associated with differential plasma lipid and cholesterol levels and cardiometabolic disorders. Front Pharmacol 2023; 14:1091976. [PMID: 37077812 PMCID: PMC10106703 DOI: 10.3389/fphar.2023.1091976] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/02/2023] [Indexed: 04/05/2023] Open
Abstract
Arylamine N-acetyltransferase 2 (NAT2) is a phase II metabolic enzyme, best known for metabolism of aromatic amines and hydrazines. Genetic variants occurring in the NAT2 coding region have been well-defined and are known to affect the enzyme activity or protein stability. Individuals can be categorized into rapid, intermediate, and slow acetylator phenotypes that significantly alter their ability to metabolize arylamines, including drugs (e.g., isoniazid) and carcinogens (e.g., 4-aminobiphenyl). However, functional studies on non-coding or intergenic variants of NAT2 are lacking. Multiple, independent genome wide association studies (GWAS) have reported that non-coding or intergenic variants of NAT2 are associated with elevated plasma lipid and cholesterol levels, as well as cardiometabolic disorders, suggesting a novel cellular role of NAT2 in lipid and cholesterol homeostasis. The current review highlights and summarizes GWAS reports that are relevant to this association. We also present a new finding that seven, non-coding, intergenic NAT2 variants (i.e., rs4921913, rs4921914, rs4921915, rs146812806, rs35246381, rs35570672, and rs1495741), which have been associated with plasma lipid and cholesterol levels, are in linkage disequilibrium with one another, and thus form a novel haplotype. The dyslipidemia risk alleles of non-coding NAT2 variants are associated with rapid NAT2 acetylator phenotype, suggesting that differential systemic NAT2 activity might be a risk factor for developing dyslipidemia. The current review also discusses the findings of recent reports that are supportive of the role of NAT2 in lipid or cholesterol synthesis and transport. In summary, we review data suggesting that human NAT2 is a novel genetic factor that influences plasma lipid and cholesterol levels and alters the risk of cardiometabolic disorders. The proposed novel role of NAT2 merits further investigations.
Collapse
|
16
|
Nag A, Dhindsa RS, Middleton L, Jiang X, Vitsios D, Wigmore E, Allman EL, Reznichenko A, Carss K, Smith KR, Wang Q, Challis B, Paul DS, Harper AR, Petrovski S. Effects of protein-coding variants on blood metabolite measurements and clinical biomarkers in the UK Biobank. Am J Hum Genet 2023; 110:487-498. [PMID: 36809768 PMCID: PMC10027475 DOI: 10.1016/j.ajhg.2023.02.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023] Open
Abstract
Genome-wide association studies (GWASs) have established the contribution of common and low-frequency variants to metabolic blood measurements in the UK Biobank (UKB). To complement existing GWAS findings, we assessed the contribution of rare protein-coding variants in relation to 355 metabolic blood measurements-including 325 predominantly lipid-related nuclear magnetic resonance (NMR)-derived blood metabolite measurements (Nightingale Health Plc) and 30 clinical blood biomarkers-using 412,393 exome sequences from four genetically diverse ancestries in the UKB. Gene-level collapsing analyses were conducted to evaluate a diverse range of rare-variant architectures for the metabolic blood measurements. Altogether, we identified significant associations (p < 1 × 10-8) for 205 distinct genes that involved 1,968 significant relationships for the Nightingale blood metabolite measurements and 331 for the clinical blood biomarkers. These include associations for rare non-synonymous variants in PLIN1 and CREB3L3 with lipid metabolite measurements and SYT7 with creatinine, among others, which may not only provide insights into novel biology but also deepen our understanding of established disease mechanisms. Of the study-wide significant clinical biomarker associations, 40% were not previously detected on analyzing coding variants in a GWAS in the same cohort, reinforcing the importance of studying rare variation to fully understand the genetic architecture of metabolic blood measurements.
Collapse
Affiliation(s)
- Abhishek Nag
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ryan S Dhindsa
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Lawrence Middleton
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Xiao Jiang
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Dimitrios Vitsios
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Eleanor Wigmore
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Erik L Allman
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Anna Reznichenko
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Keren Carss
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Katherine R Smith
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Quanli Wang
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA, USA
| | - Benjamin Challis
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Dirk S Paul
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Andrew R Harper
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK; Early Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Slavé Petrovski
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK; Department of Medicine, University of Melbourne, Austin Health, Melbourne, VIC, Australia.
| |
Collapse
|
17
|
Lee IH, Smith MR, Yazdani A, Sandhu S, Walker DI, Mandl KD, Jones DP, Kong SW. Comprehensive characterization of putative genetic influences on plasma metabolome in a pediatric cohort. Hum Genomics 2022; 16:67. [PMID: 36482414 PMCID: PMC9730628 DOI: 10.1186/s40246-022-00440-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The human exposome is composed of diverse metabolites and small chemical compounds originated from endogenous and exogenous sources, respectively. Genetic and environmental factors influence metabolite levels, while the extent of genetic contributions across metabolic pathways is not yet known. Untargeted profiling of human metabolome using high-resolution mass spectrometry (HRMS) combined with genome-wide genotyping allows comprehensive identification of genetically influenced metabolites. As such previous studies of adults discovered and replicated genotype-metabotype associations. However, these associations have not been characterized in children. RESULTS We conducted the largest genome by metabolome-wide association study to date of children (N = 441) using 619,688 common genetic variants and 14,342 features measured by HRMS. Narrow-sense heritability (h2) estimates of plasma metabolite concentrations using genomic relatedness matrix restricted maximum likelihood (GREML) method showed a bimodal distribution with high h2 (> 0.8) for 15.9% of features and low h2 (< 0.2) for most of features (62.0%). The features with high h2 were enriched for amino acid and nucleic acid metabolism, while carbohydrate and lipid concentrations showed low h2. For each feature, a metabolite quantitative trait loci (mQTL) analysis was performed to identify genetic variants that were potentially associated with plasma levels. Fifty-four associations among 29 features and 43 genetic variants were identified at a genome-wide significance threshold p < 3.5 × 10-12 (= 5 × 10-8/14,342 features). Previously reported associations such as UGT1A1 and bilirubin; PYROXD2 and methyl lysine; and ACADS and butyrylcarnitine were successfully replicated in our pediatric cohort. We found potential candidates for novel associations including CSMD1 and a monostearyl alcohol triglyceride (m/z 781.7483, retention time (RT) 89.3 s); CALN1 and Tridecanol (m/z 283.2741, RT 27.6). A gene-level enrichment analysis using MAGMA revealed highly interconnected modules for dADP biosynthesis, sterol synthesis, and long-chain fatty acid transport in the gene-feature network. CONCLUSION Comprehensive profiling of plasma metabolome across age groups combined with genome-wide genotyping revealed a wide range of genetic influence on diverse chemical species and metabolic pathways. The developmental trajectory of a biological system is shaped by gene-environment interaction especially in early life. Therefore, continuous efforts on generating metabolomics data in diverse human tissue types across age groups are required to understand gene-environment interaction toward healthy aging trajectories.
Collapse
Affiliation(s)
- In-Hee Lee
- grid.2515.30000 0004 0378 8438Computational Health Informatics Program, Boston Children’s Hospital, 401 Park Drive, Boston, MA 02215 USA
| | - Matthew Ryan Smith
- grid.189967.80000 0001 0941 6502Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30602 USA ,grid.414026.50000 0004 0419 4084Atlanta Department of Veterans Affairs Medical Center, Decatur, GA 30033 USA
| | - Azam Yazdani
- grid.38142.3c000000041936754XCenter of Perioperative Genetics and Genomics, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Sumiti Sandhu
- grid.2515.30000 0004 0378 8438Computational Health Informatics Program, Boston Children’s Hospital, 401 Park Drive, Boston, MA 02215 USA
| | - Douglas I. Walker
- grid.59734.3c0000 0001 0670 2351Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Kenneth D. Mandl
- grid.2515.30000 0004 0378 8438Computational Health Informatics Program, Boston Children’s Hospital, 401 Park Drive, Boston, MA 02215 USA ,grid.38142.3c000000041936754XDepartment of Biomedical Informatics, Harvard Medical School, Boston, MA 02115 USA ,grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Boston, MA 02115 USA
| | - Dean P. Jones
- grid.189967.80000 0001 0941 6502Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30602 USA
| | - Sek Won Kong
- grid.2515.30000 0004 0378 8438Computational Health Informatics Program, Boston Children’s Hospital, 401 Park Drive, Boston, MA 02215 USA ,grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Boston, MA 02115 USA
| |
Collapse
|
18
|
Hoshi RA, Liu Y, Luttmann-Gibson H, Tiwari S, Giulianini F, Andres AM, Watrous JD, Cook NR, Costenbader KH, Okereke OI, Ridker PM, Manson JE, Lee IM, Vinayagamoorthy M, Cheng S, Copeland T, Jain M, Chasman DI, Demler OV, Mora S. Association of Physical Activity With Bioactive Lipids and Cardiovascular Events. Circ Res 2022; 131:e84-e99. [PMID: 35862024 PMCID: PMC9357171 DOI: 10.1161/circresaha.122.320952] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 07/07/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND To clarify the mechanisms underlying physical activity (PA)-related cardioprotection, we examined the association of PA with plasma bioactive lipids (BALs) and cardiovascular disease (CVD) events. We additionally performed genome-wide associations. METHODS PA-bioactive lipid associations were examined in VITAL (VITamin D and OmegA-3 TriaL)-clinical translational science center (REGISTRATION: URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT01169259; N=1032) and validated in JUPITER (Justification for the Use of statins in Prevention: an Intervention Trial Evaluating Rosuvastatin)-NC (REGISTRATION: URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT00239681; N=589), using linear models adjusted for age, sex, race, low-density lipoprotein-cholesterol, total-C, and smoking. Significant BALs were carried over to examine associations with incident CVD in 2 nested CVD case-control studies: VITAL-CVD (741 case-control pairs) and JUPITER-CVD (415 case-control pairs; validation). RESULTS We detected 145 PA-bioactive lipid validated associations (false discovery rate <0.1). Annotations were found for 6 of these BALs: 12,13-diHOME, 9,10-diHOME, lysoPC(15:0), oxymorphone-3b-D-glucuronide, cortisone, and oleoyl-glycerol. Genetic analysis within JUPITER-NC showed associations of 32 PA-related BALs with 22 single-nucleotide polymorphisms. From PA-related BALs, 12 are associated with CVD. CONCLUSIONS We identified a PA-related bioactive lipidome profile out of which 12 BALs also had opposite associations with incident CVD events.
Collapse
Affiliation(s)
- Rosangela A. Hoshi
- Center for Lipid Metabolomics, Division of Preventive Medicine, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yanyan Liu
- Center for Lipid Metabolomics, Division of Preventive Medicine, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Heike Luttmann-Gibson
- Center for Lipid Metabolomics, Division of Preventive Medicine, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Saumya Tiwari
- Department of Pharmacology, University of California San Diego, La Jolla, CA 92037, USA
| | - Franco Giulianini
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Allen M. Andres
- Department of Pharmacology, University of California San Diego, La Jolla, CA 92037, USA
| | - Jeramie D. Watrous
- Department of Pharmacology, University of California San Diego, La Jolla, CA 92037, USA
| | - Nancy R. Cook
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Karen H. Costenbader
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Olivia I. Okereke
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Paul M Ridker
- Center for Lipid Metabolomics, Division of Preventive Medicine, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - JoAnn E. Manson
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - I-Min Lee
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | | | - Susan Cheng
- Smidt Heart Institute, Cedars-Sinai Medical Ctr, Los Angeles, CA 90048, USA
| | - Trisha Copeland
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mohit Jain
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Daniel I. Chasman
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Olga V. Demler
- Center for Lipid Metabolomics, Division of Preventive Medicine, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Computer Science, ETH Zurich, Zurich 8092, Switzerland
| | - Samia Mora
- Center for Lipid Metabolomics, Division of Preventive Medicine, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
19
|
Liu C, Wang Z, Hui Q, Chiang Y, Chen J, Brijkumar J, Edwards JA, Ordonez CE, Dudgeon MR, Sunpath H, Pillay S, Moodley P, Kuritzkes DR, Moosa MYS, Jones DP, Marconi VC, Sun YV. Crosstalk between Host Genome and Metabolome among People with HIV in South Africa. Metabolites 2022; 12:metabo12070624. [PMID: 35888748 PMCID: PMC9316179 DOI: 10.3390/metabo12070624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/16/2022] [Accepted: 07/01/2022] [Indexed: 02/01/2023] Open
Abstract
Genome-wide association studies (GWAS) of circulating metabolites have revealed the role of genetic regulation on the human metabolome. Most previous investigations focused on European ancestry, and few studies have been conducted among populations of African descent living in Africa, where the infectious disease burden is high (e.g., human immunodeficiency virus (HIV)). It is important to understand the genetic associations of the metabolome in diverse at-risk populations including people with HIV (PWH) living in Africa. After a thorough literature review, the reported significant gene−metabolite associations were tested among 490 PWH in South Africa. Linear regression was used to test associations between the candidate metabolites and genetic variants. GWAS of 154 plasma metabolites were performed to identify novel genetic associations. Among the 29 gene−metabolite associations identified in the literature, we replicated 10 in South Africans with HIV. The UGT1A cluster was associated with plasma levels of biliverdin and bilirubin; SLC16A9 and CPS1 were associated with carnitine and creatine, respectively. We also identified 22 genetic associations with metabolites using a genome-wide significance threshold (p-value < 5 × 10−8). In a GWAS of plasma metabolites in South African PWH, we replicated reported genetic associations across ancestries, and identified novel genetic associations using a metabolomics approach.
Collapse
Affiliation(s)
- Chang Liu
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA; (C.L.); (Q.H.); (Y.C.); (J.C.)
| | - Zicheng Wang
- College of Arts and Sciences, Emory University, Atlanta, GA 30322, USA;
| | - Qin Hui
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA; (C.L.); (Q.H.); (Y.C.); (J.C.)
| | - Yiyun Chiang
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA; (C.L.); (Q.H.); (Y.C.); (J.C.)
| | - Junyu Chen
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA; (C.L.); (Q.H.); (Y.C.); (J.C.)
| | - Jaysingh Brijkumar
- Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban 4041, South Africa; (J.B.); (H.S.); (S.P.); (M.Y.S.M.)
| | - Johnathan A. Edwards
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA;
- School of Medicine, Emory University, Atlanta, GA 30322, USA; (M.R.D.); (D.P.J.); (V.C.M.)
- Lincoln International Institute for Rural Health, School of Health and Social Care, University of Lincoln, Lincoln LN6 7TS, UK
| | - Claudia E. Ordonez
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA;
| | - Mathew R. Dudgeon
- School of Medicine, Emory University, Atlanta, GA 30322, USA; (M.R.D.); (D.P.J.); (V.C.M.)
| | - Henry Sunpath
- Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban 4041, South Africa; (J.B.); (H.S.); (S.P.); (M.Y.S.M.)
| | - Selvan Pillay
- Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban 4041, South Africa; (J.B.); (H.S.); (S.P.); (M.Y.S.M.)
| | - Pravi Moodley
- National Health Laboratory Service, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4011, South Africa;
| | - Daniel R. Kuritzkes
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Mohamed Y. S. Moosa
- Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban 4041, South Africa; (J.B.); (H.S.); (S.P.); (M.Y.S.M.)
| | - Dean P. Jones
- School of Medicine, Emory University, Atlanta, GA 30322, USA; (M.R.D.); (D.P.J.); (V.C.M.)
| | - Vincent C. Marconi
- School of Medicine, Emory University, Atlanta, GA 30322, USA; (M.R.D.); (D.P.J.); (V.C.M.)
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA;
- Emory Vaccine Center, Atlanta, GA 30322, USA
| | - Yan V. Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA; (C.L.); (Q.H.); (Y.C.); (J.C.)
- Correspondence: ; Tel.: +1-404-727-9090
| |
Collapse
|
20
|
Heritability of Urinary Amines, Organic Acids, and Steroid Hormones in Children. Metabolites 2022; 12:metabo12060474. [PMID: 35736407 PMCID: PMC9228478 DOI: 10.3390/metabo12060474] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 02/01/2023] Open
Abstract
Variation in metabolite levels reflects individual differences in genetic and environmental factors. Here, we investigated the role of these factors in urinary metabolomics data in children. We examined the effects of sex and age on 86 metabolites, as measured on three metabolomics platforms that target amines, organic acids, and steroid hormones. Next, we estimated their heritability in a twin cohort of 1300 twins (age range: 5.7–12.9 years). We observed associations between age and 50 metabolites and between sex and 21 metabolites. The monozygotic (MZ) and dizygotic (DZ) correlations for the urinary metabolites indicated a role for non-additive genetic factors for 50 amines, 13 organic acids, and 6 steroids. The average broad-sense heritability for these amines, organic acids, and steroids was 0.49 (range: 0.25–0.64), 0.50 (range: 0.33–0.62), and 0.64 (range: 0.43–0.81), respectively. For 6 amines, 7 organic acids, and 4 steroids the twin correlations indicated a role for shared environmental factors and the average narrow-sense heritability was 0.50 (range: 0.37–0.68), 0.50 (range; 0.23–0.61), and 0.47 (range: 0.32–0.70) for these amines, organic acids, and steroids. We conclude that urinary metabolites in children have substantial heritability, with similar estimates for amines and organic acids, and higher estimates for steroid hormones.
Collapse
|
21
|
Haince JF, Joubert P, Bach H, Ahmed Bux R, Tappia PS, Ramjiawan B. Metabolomic Fingerprinting for the Detection of Early-Stage Lung Cancer: From the Genome to the Metabolome. Int J Mol Sci 2022; 23:ijms23031215. [PMID: 35163138 PMCID: PMC8835988 DOI: 10.3390/ijms23031215] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 12/19/2022] Open
Abstract
The five-year survival rate of lung cancer patients is very low, mainly because most newly diagnosed patients present with locally advanced or metastatic disease. Therefore, early diagnosis is key to the successful treatment and management of lung cancer. Unfortunately, early detection methods of lung cancer are not ideal. In this brief review, we described early detection methods such as chest X-rays followed by bronchoscopy, sputum analysis followed by cytological analysis, and low-dose computed tomography (LDCT). In addition, we discussed the potential of metabolomic fingerprinting, compared to that of other biomarkers, including molecular targets, as a low-cost, high-throughput blood-based test that is both feasible and affordable for early-stage lung cancer screening of at-risk populations. Accordingly, we proposed a paradigm shift to metabolomics as an alternative to molecular and proteomic-based markers in lung cancer screening, which will enable blood-based routine testing and be accessible to those patients at the highest risk for lung cancer.
Collapse
Affiliation(s)
| | - Philippe Joubert
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Pathology, Laval University, Quebec, QC G1V 4G5, Canada;
| | - Horacio Bach
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, BC V6H 3Z6, Canada;
| | - Rashid Ahmed Bux
- BioMark Diagnostics Inc., Richmond, BC V6X 2W8, Canada; (J.-F.H.); (R.A.B.)
| | - Paramjit S. Tappia
- Asper Clinical Research Institute, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada;
- Correspondence: ; Tel.: +1-204-258-1230
| | - Bram Ramjiawan
- Asper Clinical Research Institute, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada;
- Department of Pharmacology & Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
| |
Collapse
|
22
|
Mikaeloff F, Svensson Akusjärvi S, Ikomey GM, Krishnan S, Sperk M, Gupta S, Magdaleno GDV, Escós A, Lyonga E, Okomo MC, Tagne CT, Babu H, Lorson CL, Végvári Á, Banerjea AC, Kele J, Hanna LE, Singh K, de Magalhães JP, Benfeitas R, Neogi U. Trans cohort metabolic reprogramming towards glutaminolysis in long-term successfully treated HIV-infection. Commun Biol 2022; 5:27. [PMID: 35017663 PMCID: PMC8752762 DOI: 10.1038/s42003-021-02985-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022] Open
Abstract
Despite successful combination antiretroviral therapy (cART), persistent low-grade immune activation together with inflammation and toxic antiretroviral drugs can lead to long-lasting metabolic flexibility and adaptation in people living with HIV (PLWH). Our study investigated alterations in the plasma metabolic profiles by comparing PLWH on long-term cART(>5 years) and matched HIV-negative controls (HC) in two cohorts from low- and middle-income countries (LMIC), Cameroon, and India, respectively, to understand the system-level dysregulation in HIV-infection. Using untargeted and targeted LC-MS/MS-based metabolic profiling and applying advanced system biology methods, an altered amino acid metabolism, more specifically to glutaminolysis in PLWH than HC were reported. A significantly lower level of neurosteroids was observed in both cohorts and could potentiate neurological impairments in PLWH. Further, modulation of cellular glutaminolysis promoted increased cell death and latency reversal in pre-monocytic HIV-1 latent cell model U1, which may be essential for the clearance of the inducible reservoir in HIV-integrated cells.
Collapse
Affiliation(s)
- Flora Mikaeloff
- The Systems Virology Lab, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, ANA Futura, Campus Flemingsberg, Stockholm, Sweden
| | - Sara Svensson Akusjärvi
- The Systems Virology Lab, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, ANA Futura, Campus Flemingsberg, Stockholm, Sweden
| | - George Mondinde Ikomey
- Center for the Study and Control of Communicable Diseases (CSCCD), Faculty of Medicine and Biomedical Sciences, University of Yaoundé 1, P.O. Box. 8445, Yaoundé, Cameroon
- Department of Microbiology, Haematology, Parasitology and Infectious Disease, Faculty of Medicine and Biomedical Sciences, University of Yaoundé 1, Yaoundé, Cameroon
| | - Shuba Krishnan
- The Systems Virology Lab, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, ANA Futura, Campus Flemingsberg, Stockholm, Sweden
| | - Maike Sperk
- The Systems Virology Lab, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, ANA Futura, Campus Flemingsberg, Stockholm, Sweden
| | - Soham Gupta
- The Systems Virology Lab, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, ANA Futura, Campus Flemingsberg, Stockholm, Sweden
| | - Gustavo Daniel Vega Magdaleno
- Integrative Genomics of Ageing Group, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Alejandra Escós
- The Systems Virology Lab, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, ANA Futura, Campus Flemingsberg, Stockholm, Sweden
| | - Emilia Lyonga
- Center for the Study and Control of Communicable Diseases (CSCCD), Faculty of Medicine and Biomedical Sciences, University of Yaoundé 1, P.O. Box. 8445, Yaoundé, Cameroon
- Department of Microbiology, Haematology, Parasitology and Infectious Disease, Faculty of Medicine and Biomedical Sciences, University of Yaoundé 1, Yaoundé, Cameroon
| | - Marie Claire Okomo
- Center for the Study and Control of Communicable Diseases (CSCCD), Faculty of Medicine and Biomedical Sciences, University of Yaoundé 1, P.O. Box. 8445, Yaoundé, Cameroon
- Department of Microbiology, Haematology, Parasitology and Infectious Disease, Faculty of Medicine and Biomedical Sciences, University of Yaoundé 1, Yaoundé, Cameroon
| | - Claude Tayou Tagne
- Department of Microbiology, Haematology, Parasitology and Infectious Disease, Faculty of Medicine and Biomedical Sciences, University of Yaoundé 1, Yaoundé, Cameroon
| | - Hemalatha Babu
- Department of HIV/AIDS, National Institute for Research in Tuberculosis, ICMR, Chennai, 600031, India
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory Vaccine Center, Emory University, Atlanta, GA, 30329, USA
| | - Christian L Lorson
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, 65211, USA
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA
| | - Ákos Végvári
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Akhil C Banerjea
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Julianna Kele
- Department of Physiology and Pharmacology, Neurovascular Biology and Health, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Luke Elizabeth Hanna
- Department of HIV/AIDS, National Institute for Research in Tuberculosis, ICMR, Chennai, 600031, India
| | - Kamal Singh
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, 65211, USA
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Rui Benfeitas
- National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, S-10691, Stockholm, Sweden
| | - Ujjwal Neogi
- The Systems Virology Lab, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, ANA Futura, Campus Flemingsberg, Stockholm, Sweden.
- Manipal Institute of Virology (MIV), Manipal Academy of Higher Education, Manipal, Karnataka, India.
| |
Collapse
|
23
|
Loretz N, Becker C, Hochstrasser S, Metzger K, Beck K, Mueller J, Gross S, Vincent A, Amacher SA, Sutter R, Tisljar K, Schuetz P, Bernasconi L, Neyer P, Pargger H, Marsch S, Hunziker S. Activation of the kynurenine pathway predicts mortality and neurological outcome in cardiac arrest patients: A validation study. J Crit Care 2021; 67:57-65. [PMID: 34673332 DOI: 10.1016/j.jcrc.2021.09.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/03/2021] [Accepted: 09/01/2021] [Indexed: 01/25/2023]
Abstract
PURPOSE Activation of the kynurenine pathway (KP) has been shown to predict outcome in cardiac arrest (CA) patients. We validated these findings in a Swiss cohort. METHODS We measured admission tryptophan and kynurenine levels in 270 consecutive CA patients (38 in-hospital CA) and investigated associations with in-hospital mortality and neurological outcome at hospital discharge. RESULTS 120 of 270 (44%) patients died in the hospital. Compared to survivors, non-survivors showed higher median initial kynurenine levels (5.28 μmol/l [IQR 2.91 to 7.40] vs 3.58 μmol/l [IQR 2.47 to 5.46]; p < 0.001) and a higher median kynurenine/tryptophan ratio (0.10 μmol/l [IQR 0.07 to 0.17] vs 0.07 μmol/l [IQR 0.05 to 0.1]; p < 0.001). In a model adjusted for age, gender and comorbidities, kynurenine (OR 1.16, 95% CI 1.05 to 1.27; p = 0.001) and kynurenine/tryptophan ratio (OR 1.19, 95% CI 1.08 to 1.31; p = 0.003) were significantly associated with mortality. Results were similar for neurological outcome. CONCLUSIONS Our findings validate a previous study and show associations of the activation of the KP with unfavorable outcomes after CA. Future studies should evaluate whether therapeutic modulation of the KP may impact clinical outcomes after CA.
Collapse
Affiliation(s)
- Nina Loretz
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, Klingelbergstrasse 23, 4056 Basel, Switzerland.
| | - Christoph Becker
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, Klingelbergstrasse 23, 4056 Basel, Switzerland; Emergency Department, University Hospital Basel, Petersgraben 2, 4051 Basel, Switzerland.
| | - Seraina Hochstrasser
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, Klingelbergstrasse 23, 4056 Basel, Switzerland.
| | - Kerstin Metzger
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, Klingelbergstrasse 23, 4056 Basel, Switzerland.
| | - Katharina Beck
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, Klingelbergstrasse 23, 4056 Basel, Switzerland.
| | - Jonas Mueller
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, Klingelbergstrasse 23, 4056 Basel, Switzerland.
| | - Sebastian Gross
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, Klingelbergstrasse 23, 4056 Basel, Switzerland.
| | - Alessia Vincent
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, Klingelbergstrasse 23, 4056 Basel, Switzerland.
| | - Simon A Amacher
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, Klingelbergstrasse 23, 4056 Basel, Switzerland; Intensive Care Unit, University Hospital Basel, Petersgraben 4, 4051 Basel, Switzerland.
| | - Raoul Sutter
- Intensive Care Unit, University Hospital Basel, Petersgraben 4, 4051 Basel, Switzerland; Faculty of Medicine, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland.
| | - Kai Tisljar
- Intensive Care Unit, University Hospital Basel, Petersgraben 4, 4051 Basel, Switzerland.
| | - Philipp Schuetz
- Faculty of Medicine, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland; Division of Internal Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland.
| | - Luca Bernasconi
- Institute of Laboratory Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland.
| | - Peter Neyer
- Institute of Laboratory Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland.
| | - Hans Pargger
- Intensive Care Unit, University Hospital Basel, Petersgraben 4, 4051 Basel, Switzerland; Faculty of Medicine, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland.
| | - Stephan Marsch
- Intensive Care Unit, University Hospital Basel, Petersgraben 4, 4051 Basel, Switzerland; Faculty of Medicine, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland.
| | - Sabina Hunziker
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, Klingelbergstrasse 23, 4056 Basel, Switzerland; Faculty of Medicine, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland.
| |
Collapse
|
24
|
Bermingham KM, Brennan L, Segurado R, Gray IJ, Barron RE, Gibney ER, Ryan MF, Gibney MJ, Newman JW, O'Sullivan DAM. Genetic and environmental influences on serum oxylipins, endocannabinoids, bile acids and steroids. Prostaglandins Leukot Essent Fatty Acids 2021; 173:102338. [PMID: 34500309 DOI: 10.1016/j.plefa.2021.102338] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 08/06/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022]
Abstract
Lipid bioactivity is a result of direct action and the action of lipid mediators including oxylipins, endocannabinoids, bile acids and steroids. Understanding the factors contributing to biological variation in lipid mediators may inform future approaches to understand and treat complex metabolic diseases. This research aims to determine the contribution of genetic and environmental influences on lipid mediators involved in the regulation of inflammation and energy metabolism. This study recruited 138 monozygotic (MZ) and dizygotic (DZ) twins aged 18-65 years and measured serum oxylipins, endocannabinoids, bile acids and steroids using liquid chromatography mass-spectrometry (LC-MS). In this classic twin design, the similarities and differences between MZ and DZ twins are modelled to estimate the contribution of genetic and environmental influences to variation in lipid mediators. Heritable lipid mediators included the 12-lipoxygenase products 12-hydroxyeicosatetraenoic acid [0.70 (95% CI: 0.12,0.82)], 12-hydroxyeicosatetraenoic acid [0.73 (95% CI: 0.30,0.83)] and 14‑hydroxy-docosahexaenoic acid [0.51 (95% CI: 0.07,0.71)], along with the endocannabinoid docosahexaenoy-lethanolamide [0.52 (95% CI: 0.15,0.72)]. For others such as 13-hydroxyoctadecatrienoic acid and lithocholic acid the contribution of environment to variation was stronger. With increased understanding of lipid mediator functions in health, it is important to understand the factors contributing to their variance. This study provides a comprehensive analysis of lipid mediators and extends pre-existing knowledge of the genetic and environmental influences on the human lipidome.
Collapse
MESH Headings
- 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/blood
- 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/genetics
- Adolescent
- Adult
- Aged
- Bile Acids and Salts/blood
- Bile Acids and Salts/genetics
- Dehydroepiandrosterone/blood
- Dehydroepiandrosterone/genetics
- Docosahexaenoic Acids/blood
- Docosahexaenoic Acids/genetics
- Eicosapentaenoic Acid/analogs & derivatives
- Eicosapentaenoic Acid/blood
- Eicosapentaenoic Acid/genetics
- Endocannabinoids/blood
- Endocannabinoids/genetics
- Fatty Acids, Omega-3/blood
- Fatty Acids, Omega-3/genetics
- Female
- Gene-Environment Interaction
- Humans
- Lipid Metabolism/genetics
- Male
- Middle Aged
- Oxylipins/blood
- Steroids/blood
- Twins, Dizygotic/genetics
- Twins, Monozygotic/genetics
- Young Adult
Collapse
Affiliation(s)
- K M Bermingham
- UCD Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - L Brennan
- UCD Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland; UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - R Segurado
- UCD School of Public Health, Physiotherapy and Sports Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - I J Gray
- Obesity and Metabolism Research Unit, United States Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, USA; West Coast Metabolomics Center, UC Davis Genome Center, University of California Davis, Davis, CA, USA
| | - R E Barron
- UCD Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - E R Gibney
- UCD Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - M F Ryan
- UCD Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - M J Gibney
- UCD Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - J W Newman
- Obesity and Metabolism Research Unit, United States Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, USA; West Coast Metabolomics Center, UC Davis Genome Center, University of California Davis, Davis, CA, USA; Dept of Nutrition, University of California Davis, Davis, CA, USA
| | - Dr A M O'Sullivan
- UCD Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
25
|
Chaby LE, Lasseter HC, Contrepois K, Salek RM, Turck CW, Thompson A, Vaughan T, Haas M, Jeromin A. Cross-Platform Evaluation of Commercially Targeted and Untargeted Metabolomics Approaches to Optimize the Investigation of Psychiatric Disease. Metabolites 2021; 11:609. [PMID: 34564425 PMCID: PMC8466258 DOI: 10.3390/metabo11090609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 11/17/2022] Open
Abstract
Metabolomics methods often encounter trade-offs between quantification accuracy and coverage, with truly comprehensive coverage only attainable through a multitude of complementary assays. Due to the lack of standardization and the variety of metabolomics assays, it is difficult to integrate datasets across studies or assays. To inform metabolomics platform selection, with a focus on posttraumatic stress disorder (PTSD), we review platform use and sample sizes in psychiatric metabolomics studies and then evaluate five prominent metabolomics platforms for coverage and performance, including intra-/inter-assay precision, accuracy, and linearity. We found performance was variable between metabolite classes, but comparable across targeted and untargeted approaches. Within all platforms, precision and accuracy were highly variable across classes, ranging from 0.9-63.2% (coefficient of variation) and 0.6-99.1% for accuracy to reference plasma. Several classes had high inter-assay variance, potentially impeding dissociation of a biological signal, including glycerophospholipids, organooxygen compounds, and fatty acids. Coverage was platform-specific and ranged from 16-70% of PTSD-associated metabolites. Non-overlapping coverage is challenging; however, benefits of applying multiple metabolomics technologies must be weighed against cost, biospecimen availability, platform-specific normative levels, and challenges in merging datasets. Our findings and open-access cross-platform dataset can inform platform selection and dataset integration based on platform-specific coverage breadth/overlap and metabolite-specific performance.
Collapse
Affiliation(s)
- Lauren E. Chaby
- Cohen Veterans Bioscience, New York, NY 10018, USA; (L.E.C.); (H.C.L.); (A.T.); (T.V.); (M.H.)
| | - Heather C. Lasseter
- Cohen Veterans Bioscience, New York, NY 10018, USA; (L.E.C.); (H.C.L.); (A.T.); (T.V.); (M.H.)
| | - Kévin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Reza M. Salek
- International Agency for Research on Cancer, Nutrition and Metabolism Branch, World Health Organisation, 150 Cours Albert Thomas, CEDEX 08, 69372 Lyon, France;
| | - Christoph W. Turck
- Max Planck Institute of Psychiatry, Proteomics and Biomarkers, 80804 Munich, Germany;
| | - Andrew Thompson
- Cohen Veterans Bioscience, New York, NY 10018, USA; (L.E.C.); (H.C.L.); (A.T.); (T.V.); (M.H.)
| | - Timothy Vaughan
- Cohen Veterans Bioscience, New York, NY 10018, USA; (L.E.C.); (H.C.L.); (A.T.); (T.V.); (M.H.)
| | - Magali Haas
- Cohen Veterans Bioscience, New York, NY 10018, USA; (L.E.C.); (H.C.L.); (A.T.); (T.V.); (M.H.)
| | - Andreas Jeromin
- Cohen Veterans Bioscience, New York, NY 10018, USA; (L.E.C.); (H.C.L.); (A.T.); (T.V.); (M.H.)
| |
Collapse
|
26
|
Guida F, Tan VY, Corbin LJ, Smith-Byrne K, Alcala K, Langenberg C, Stewart ID, Butterworth AS, Surendran P, Achaintre D, Adamski J, Amiano P, Bergmann MM, Bull CJ, Dahm CC, Gicquiau A, Giles GG, Gunter MJ, Haller T, Langhammer A, Larose TL, Ljungberg B, Metspalu A, Milne RL, Muller DC, Nøst TH, Pettersen Sørgjerd E, Prehn C, Riboli E, Rinaldi S, Rothwell JA, Scalbert A, Schmidt JA, Severi G, Sieri S, Vermeulen R, Vincent EE, Waldenberger M, Timpson NJ, Johansson M. The blood metabolome of incident kidney cancer: A case-control study nested within the MetKid consortium. PLoS Med 2021; 18:e1003786. [PMID: 34543281 PMCID: PMC8496779 DOI: 10.1371/journal.pmed.1003786] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 10/07/2021] [Accepted: 08/27/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Excess bodyweight and related metabolic perturbations have been implicated in kidney cancer aetiology, but the specific molecular mechanisms underlying these relationships are poorly understood. In this study, we sought to identify circulating metabolites that predispose kidney cancer and to evaluate the extent to which they are influenced by body mass index (BMI). METHODS AND FINDINGS We assessed the association between circulating levels of 1,416 metabolites and incident kidney cancer using pre-diagnostic blood samples from up to 1,305 kidney cancer case-control pairs from 5 prospective cohort studies. Cases were diagnosed on average 8 years after blood collection. We found 25 metabolites robustly associated with kidney cancer risk. In particular, 14 glycerophospholipids (GPLs) were inversely associated with risk, including 8 phosphatidylcholines (PCs) and 2 plasmalogens. The PC with the strongest association was PC ae C34:3 with an odds ratio (OR) for 1 standard deviation (SD) increment of 0.75 (95% confidence interval [CI]: 0.68 to 0.83, p = 2.6 × 10-8). In contrast, 4 amino acids, including glutamate (OR for 1 SD = 1.39, 95% CI: 1.20 to 1.60, p = 1.6 × 10-5), were positively associated with risk. Adjusting for BMI partly attenuated the risk association for some-but not all-metabolites, whereas other known risk factors of kidney cancer, such as smoking and alcohol consumption, had minimal impact on the observed associations. A mendelian randomisation (MR) analysis of the influence of BMI on the blood metabolome highlighted that some metabolites associated with kidney cancer risk are influenced by BMI. Specifically, elevated BMI appeared to decrease levels of several GPLs that were also found inversely associated with kidney cancer risk (e.g., -0.17 SD change [ßBMI] in 1-(1-enyl-palmitoyl)-2-linoleoyl-GPC (P-16:0/18:2) levels per SD change in BMI, p = 3.4 × 10-5). BMI was also associated with increased levels of glutamate (ßBMI: 0.12, p = 1.5 × 10-3). While our results were robust across the participating studies, they were limited to study participants of European descent, and it will, therefore, be important to evaluate if our findings can be generalised to populations with different genetic backgrounds. CONCLUSIONS This study suggests a potentially important role of the blood metabolome in kidney cancer aetiology by highlighting a wide range of metabolites associated with the risk of developing kidney cancer and the extent to which changes in levels of these metabolites are driven by BMI-the principal modifiable risk factor of kidney cancer.
Collapse
Affiliation(s)
- Florence Guida
- Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Vanessa Y. Tan
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Laura J. Corbin
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Karl Smith-Byrne
- Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Karine Alcala
- Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Isobel D. Stewart
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Adam S. Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, United Kingdom
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, United Kingdom
| | - Praveen Surendran
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, United Kingdom
- Rutherford Fund Fellow, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - David Achaintre
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Chair of Experimental Genetics, School of Life Science, Weihenstephan, Technische Universität München, Freising, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Pilar Amiano
- Ministry of Health of the Basque Government, Sub Directorate for Public Health and Addictions of Gipuzkoa, San Sebastián, Spain
- Biodonostia Health Research Institute, Epidemiology of Chronic and Communicable Diseases Group, San Sebastián, Spain
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Caroline J. Bull
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
- Bristol Renal, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | | | - Audrey Gicquiau
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Graham G. Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Australia
| | - Marc J. Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Toomas Haller
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Arnulf Langhammer
- HUNT Research Centre, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Levanger, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Tricia L. Larose
- Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
- HUNT Research Centre, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Levanger, Norway
- Department of Community Medicine and Global Health, Institute of Health and Society, University of Oslo, Oslo, Norway
| | - Börje Ljungberg
- Department of Surgical and Perioperative Sciences, Urology and Andrology, Umeå University, Umeå, Sweden
| | | | - Roger L. Milne
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Australia
| | - David C. Muller
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Therese H. Nøst
- Department of Community Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Elin Pettersen Sørgjerd
- HUNT Research Centre, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Levanger, Norway
| | - Cornelia Prehn
- Metabolomics and Proteomics Core (MPC), Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Elio Riboli
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Sabina Rinaldi
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Joseph A. Rothwell
- Université Paris-Saclay, UVSQ, Inserm, Gustave Roussy, Équipe “Exposome et Hérédité”, CESP UMR1018, Inserm, Villejuif, France
| | - Augustin Scalbert
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Julie A. Schmidt
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Gianluca Severi
- Université Paris-Saclay, UVSQ, Inserm, Gustave Roussy, Équipe “Exposome et Hérédité”, CESP UMR1018, Inserm, Villejuif, France
- Department of Statistics, Computer Science and Applications (DISIA), University of Florence, Florence, Italy
| | - Sabina Sieri
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Roel Vermeulen
- Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands
| | - Emma E. Vincent
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
- Bristol Renal, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Melanie Waldenberger
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Nicholas J. Timpson
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Mattias Johansson
- Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| |
Collapse
|
27
|
Luo S, Feofanova EV, Tin A, Tung S, Rhee EP, Coresh J, Arking DE, Surapaneni A, Schlosser P, Li Y, Köttgen A, Yu B, Grams ME. Genome-wide association study of serum metabolites in the African American Study of Kidney Disease and Hypertension. Kidney Int 2021; 100:430-439. [PMID: 33838163 PMCID: PMC8583323 DOI: 10.1016/j.kint.2021.03.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 02/26/2021] [Accepted: 03/11/2021] [Indexed: 01/23/2023]
Abstract
The genome-wide association study (GWAS) is a powerful means to study genetic determinants of disease traits and generate insights into disease pathophysiology. To date, few GWAS of circulating metabolite levels have been performed in African Americans with chronic kidney disease. Hypothesizing that novel genetic-metabolite associations may be identified in a unique population of African Americans with a lower glomerular filtration rate (GFR), we conducted a GWAS of 652 serum metabolites in 619 participants (mean measured glomerular filtration rate 45 mL/min/1.73m2) in the African American Study of Kidney Disease and Hypertension, a clinical trial of blood pressure lowering and antihypertensive medication in African Americans with chronic kidney disease. We identified 42 significant variant metabolite associations. Twenty associations had been previously identified in published GWAS, and eleven novel associations were replicated in a separate cohort of 818 African Americans with genetic and metabolomic data from the Atherosclerosis Risk in Communities Study. The replicated novel variant-metabolite associations comprised eight metabolites and eleven distinct genomic loci. Nine of the replicated associations represented clear enzyme-metabolite interactions, with high expression in the kidneys as well as the liver. Three loci (ACY1, ACY3, and NAT8) were associated with a common pool of metabolites, acetylated amino acids, but with different individual affinities. Thus, extensive metabolite profiling in an African American population with chronic kidney disease aided identification of novel genome-wide metabolite associations, providing clues about substrate specificity and the key roles of enzymes in modulating systemic levels of metabolites.
Collapse
Affiliation(s)
- Shengyuan Luo
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA.
| | - Elena V Feofanova
- Human Genetics Center, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Adrienne Tin
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sarah Tung
- Johns Hopkins Whiting School of Engineering, Baltimore, Maryland, USA
| | - Eugene P Rhee
- Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Dan E Arking
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aditya Surapaneni
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Pascal Schlosser
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
| | - Yong Li
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
| | - Anna Köttgen
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA; Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
| | - Bing Yu
- Human Genetics Center, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Morgan E Grams
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA; Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
28
|
Schmidt JA, Fensom GK, Rinaldi S, Scalbert A, Gunter MJ, Holmes MV, Key TJ, Travis RC. NMR Metabolite Profiles in Male Meat-Eaters, Fish-Eaters, Vegetarians and Vegans, and Comparison with MS Metabolite Profiles. Metabolites 2021; 11:121. [PMID: 33672542 PMCID: PMC7923783 DOI: 10.3390/metabo11020121] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/12/2021] [Accepted: 02/14/2021] [Indexed: 12/23/2022] Open
Abstract
Metabolomics may help to elucidate mechanisms underlying diet-disease relationships and identify novel risk factors for disease. To inform the design and interpretation of such research, evidence on diet-metabolite associations and cross-assay comparisons is needed. We aimed to compare nuclear magnetic resonance (NMR) metabolite profiles between meat-eaters, fish-eaters, vegetarians and vegans, and to compare NMR measurements to those from mass spectrometry (MS), clinical chemistry and capillary gas-liquid chromatography (GC). We quantified 207 serum NMR metabolite measures in 286 male participants of the European Prospective Investigation into Cancer and Nutrition (EPIC)-Oxford cohort. Using univariate and multivariate analyses, we found that metabolite profiles varied by diet group, especially for vegans; the main differences compared to meat-eaters were lower levels of docosahexaenoic acid, total n-3 and saturated fatty acids, cholesterol and triglycerides in very-low-density lipoproteins, various lipid factions in high-density lipoprotein, sphingomyelins, tyrosine and creatinine, and higher levels of linoleic acid, total n-6, polyunsaturated fatty acids and alanine. Levels in fish-eaters and vegetarians differed by metabolite measure. Concentrations of 13 metabolites measured using both NMR and MS, clinical chemistry or GC were mostly similar. In summary, vegans' metabolite profiles were markedly different to those of men consuming animal products. The studied metabolomics platforms are complementary, with limited overlap between metabolite classes.
Collapse
Affiliation(s)
- Julie A. Schmidt
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK; (G.K.F.); (T.J.K.); (R.C.T.)
| | - Georgina K. Fensom
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK; (G.K.F.); (T.J.K.); (R.C.T.)
- Department of International Development, University of Oxford, Oxford OX1 3TB, UK
| | - Sabina Rinaldi
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, 69372 Lyon, France; (S.R.); (A.S.); (M.J.G.)
| | - Augustin Scalbert
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, 69372 Lyon, France; (S.R.); (A.S.); (M.J.G.)
| | - Marc J. Gunter
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, 69372 Lyon, France; (S.R.); (A.S.); (M.J.G.)
| | - Michael V. Holmes
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK;
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Timothy J. Key
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK; (G.K.F.); (T.J.K.); (R.C.T.)
| | - Ruth C. Travis
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK; (G.K.F.); (T.J.K.); (R.C.T.)
| |
Collapse
|
29
|
Lotta LA, Pietzner M, Stewart ID, Wittemans LB, Li C, Bonelli R, Raffler J, Biggs EK, Oliver-Williams C, Auyeung VP, Luan J, Wheeler E, Paige E, Surendran P, Michelotti GA, Scott RA, Burgess S, Zuber V, Sanderson E, Koulman A, Imamura F, Forouhi NG, Khaw KT, Griffin JL, Wood AM, Kastenmüller G, Danesh J, Butterworth AS, Gribble FM, Reimann F, Bahlo M, Fauman E, Wareham NJ, Langenberg C. A cross-platform approach identifies genetic regulators of human metabolism and health. Nat Genet 2021; 53:54-64. [PMID: 33414548 PMCID: PMC7612925 DOI: 10.1038/s41588-020-00751-5] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 11/20/2020] [Indexed: 02/02/2023]
Abstract
In cross-platform analyses of 174 metabolites, we identify 499 associations (P < 4.9 × 10-10) characterized by pleiotropy, allelic heterogeneity, large and nonlinear effects and enrichment for nonsynonymous variation. We identify a signal at GLP2R (p.Asp470Asn) shared among higher citrulline levels, body mass index, fasting glucose-dependent insulinotropic peptide and type 2 diabetes, with β-arrestin signaling as the underlying mechanism. Genetically higher serine levels are shown to reduce the likelihood (by 95%) and predict development of macular telangiectasia type 2, a rare degenerative retinal disease. Integration of genomic and small molecule data across platforms enables the discovery of regulators of human metabolism and translation into clinical insights.
Collapse
Affiliation(s)
- Luca A. Lotta
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Maik Pietzner
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | | | - Laura B.L. Wittemans
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK,The Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford
| | - Chen Li
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Roberto Bonelli
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia,Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Johannes Raffler
- Institute of Computational Biology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Emma K. Biggs
- Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom
| | - Clare Oliver-Williams
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK,Homerton College, University of Cambridge, Cambridge, UK
| | | | - Jian’an Luan
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Eleanor Wheeler
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Ellie Paige
- National Centre for Epidemiology and Population Health, The Australian National University, Canberra, Australia
| | - 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,Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK,Rutherford Fund Fellow, Department of Public Health and Primary Care, University of Cambridge, UK
| | | | - Robert A. Scott
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom,Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Verena Zuber
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom,Department of Epidemiology and Biostatistics, Imperial College London, UK
| | - Eleanor Sanderson
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, UK
| | - Albert Koulman
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK,Institute of Computational Biology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany,NIHR BRC Nutritional Biomarker Laboratory, University of Cambridge, UK
| | - Fumiaki Imamura
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Nita G. Forouhi
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | | | - Julian L. Griffin
- Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, UK
| | - Angela M. Wood
- 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,Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK,National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK,The Alan Turing Institute, London, UK
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - John Danesh
- 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,Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK,National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK,National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK,Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Adam S. Butterworth
- 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,Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK,National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK,National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK,Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Fiona M. Gribble
- Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom
| | - Frank Reimann
- Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom
| | - Melanie Bahlo
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia,Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Eric Fauman
- Internal Medicine Research Unit, Pfizer Worldwide Research, Cambridge, MA 02142, USA
| | | | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK. .,Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK. .,Computational Medicine, Berlin Institute of Health (BIH), Charité University Medicine, Berlin, Germany.
| |
Collapse
|
30
|
A metabolome-wide association study in the general population reveals decreased levels of serum laurylcarnitine in people with depression. Mol Psychiatry 2021; 26:7372-7383. [PMID: 34088979 PMCID: PMC8873015 DOI: 10.1038/s41380-021-01176-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 05/07/2021] [Accepted: 05/17/2021] [Indexed: 02/05/2023]
Abstract
Depression constitutes a leading cause of disability worldwide. Despite extensive research on its interaction with psychobiological factors, associated pathways are far from being elucidated. Metabolomics, assessing the final products of complex biochemical reactions, has emerged as a valuable tool for exploring molecular pathways. We conducted a metabolome-wide association analysis to investigate the link between the serum metabolome and depressed mood (DM) in 1411 participants of the KORA (Cooperative Health Research in the Augsburg Region) F4 study (discovery cohort). Serum metabolomics data comprised 353 unique metabolites measured by Metabolon. We identified 72 (5.1%) KORA participants with DM. Linear regression tests were conducted modeling each metabolite value by DM status, adjusted for age, sex, body-mass index, antihypertensive, cardiovascular, antidiabetic, and thyroid gland hormone drugs, corticoids and antidepressants. Sensitivity analyses were performed in subcohorts stratified for sex, suicidal ideation, and use of antidepressants. We replicated our results in an independent sample of 968 participants of the SHIP-Trend (Study of Health in Pomerania) study including 52 (5.4%) individuals with DM (replication cohort). We found significantly lower laurylcarnitine levels in KORA F4 participants with DM after multiple testing correction according to Benjamini/Hochberg. This finding was replicated in the independent SHIP-Trend study. Laurylcarnitine remained significantly associated (p value < 0.05) with depression in samples stratified for sex, suicidal ideation, and antidepressant medication. Decreased blood laurylcarnitine levels in depressed individuals may point to impaired fatty acid oxidation and/or mitochondrial function in depressive disorders, possibly representing a novel therapeutic target.
Collapse
|
31
|
Arginine and Arginine/ADMA Ratio Predict 90-Day Mortality in Patients with Out-of-Hospital Cardiac Arrest-Results from the Prospective, Observational COMMUNICATE Trial. J Clin Med 2020; 9:jcm9123815. [PMID: 33255752 PMCID: PMC7760544 DOI: 10.3390/jcm9123815] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/08/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023] Open
Abstract
(1) Background: In patients with shock, the L-arginine nitric oxide pathway is activated, causing an elevation of nitric oxide, asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA) levels. Whether these metabolites provide prognostic information in patients after out-of-hospital cardiac arrest (OHCA) remains unclear. (2) Methods: We prospectively included OHCA patients, recorded clinical parameters and measured plasma ADMA, SDMA and Arginine levels by liquid chromatography tandem mass spectrometry (LC-MS). The primary endpoint was 90-day mortality. (3) Results: Of 263 patients, 130 (49.4%) died within 90 days after OHCA. Compared to survivors, non-survivors had significantly higher levels of ADMA and lower Arginine and Arginine/ADMA ratios in univariable regression analyses. Arginine levels and Arginine/ADMA ratio were significantly associated with 90-day mortality (OR 0.51 (95%CI 0.34 to 0.76), p < 0.01 and OR 0.40 (95%CI 0.26 to 0.61), p < 0.001, respectively). These associations remained significant in several multivariable models. Arginine/ADMA ratio had the highest predictive value with an area under the curve (AUC) of 0.67 for 90-day mortality. Results for secondary outcomes were similar with significant associations with in-hospital mortality and neurological outcome. (4) Conclusion: Arginine and Arginine/ADMA ratio were independently associated with 90-day mortality and other adverse outcomes in patients after OHCA. Whether therapeutic modification of the L-arginine-nitric oxide pathway has the potential to improve outcome should be evaluated.
Collapse
|
32
|
Non-targeted urine metabolomics and associations with prevalent and incident type 2 diabetes. Sci Rep 2020; 10:16474. [PMID: 33020500 PMCID: PMC7536211 DOI: 10.1038/s41598-020-72456-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 08/26/2020] [Indexed: 12/11/2022] Open
Abstract
Better risk prediction and new molecular targets are key priorities in type 2 diabetes (T2D) research. Little is known about the role of the urine metabolome in predicting the risk of T2D. We aimed to use non-targeted urine metabolomics to discover biomarkers and improve risk prediction for T2D. Urine samples from two community cohorts of 1,424 adults were analyzed by ultra-performance liquid chromatography/mass spectrometry (UPLC-MS). In a discovery/replication design, three out of 62 annotated metabolites were associated with prevalent T2D, notably lower urine levels of 3-hydroxyundecanoyl-carnitine. In participants without diabetes at baseline, LASSO regression in the training set selected six metabolites that improved prediction of T2D beyond established risk factors risk over up to 12 years' follow-up in the test sample, from C-statistic 0.866 to 0.892. Our results in one of the largest non-targeted urinary metabolomics study to date demonstrate the role of the urine metabolome in identifying at-risk persons for T2D and suggest urine 3-hydroxyundecanoyl-carnitine as a biomarker candidate.
Collapse
|
33
|
Xue J, Hutchins EK, Elnagheeb M, Li Y, Valdar W, McRitchie S, Sumner S, Ideraabdullah FY. Maternal Liver Metabolic Response to Chronic Vitamin D Deficiency Is Determined by Mouse Strain Genetic Background. Curr Dev Nutr 2020; 4:nzaa106. [PMID: 32851199 PMCID: PMC7439094 DOI: 10.1093/cdn/nzaa106] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/08/2020] [Accepted: 06/16/2020] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Liver metabolite concentrations have the potential to be key biomarkers of systemic metabolic dysfunction and overall health. However, for most conditions we do not know the extent to which genetic differences regulate susceptibility to metabolic responses. This limits our ability to detect and diagnose effects in heterogeneous populations. OBJECTIVES Here, we investigated the extent to which naturally occurring genetic differences regulate maternal liver metabolic response to vitamin D deficiency (VDD), particularly during perinatal periods when such changes can adversely affect maternal and fetal health. METHODS We used a panel of 8 inbred Collaborative Cross (CC) mouse strains, each with a different genetic background (72 dams, 3-6/treatment group, per strain). We identified robust maternal liver metabolic responses to vitamin D depletion before and during gestation and lactation using a vitamin-D-deficient (VDD; 0 IU vitamin D3/kg) or -sufficient diet (1000 IU vitamin D3/kg). We then identified VDD-induced metabolite changes influenced by strain genetic background. RESULTS We detected a significant VDD effect by orthogonal partial least squares discriminant analysis (Q2 = 0.266, pQ2 = 0.002): primarily, altered concentrations of 78 metabolites involved in lipid, amino acid, and nucleotide metabolism (variable importance to projection score ≥1.5). Metabolites in unsaturated fatty acid and glycerophospholipid metabolism pathways were significantly enriched [False Discovery Rate (FDR) <0.05]. VDD also significantly altered concentrations of putative markers of uremic toxemia, acylglycerols, and dipeptides. The extent of the metabolic response to VDD was strongly dependent on genetic strain, ranging from robustly responsive to nonresponsive. Two strains (CC017/Unc and CC032/GeniUnc) were particularly sensitive to VDD; however, each strain altered different pathways. CONCLUSIONS These novel findings demonstrate that maternal VDD induces different liver metabolic effects in different genetic backgrounds. Strains with differing susceptibility and metabolic response to VDD represent unique tools to identify causal susceptibility factors and further elucidate the role of VDD-induced metabolic changes in maternal and/or fetal health for ultimately translating findings to human populations.
Collapse
Affiliation(s)
- Jing Xue
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, USA
| | - Elizabeth K Hutchins
- Department of Nutrition, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Marwa Elnagheeb
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, USA
| | - Yi Li
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - William Valdar
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Susan McRitchie
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, USA
- Department of Nutrition, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Susan Sumner
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, USA
- Department of Nutrition, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Folami Y Ideraabdullah
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, USA
- Department of Nutrition, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
34
|
Genetics and Not Shared Environment Explains Familial Resemblance in Adult Metabolomics Data. Twin Res Hum Genet 2020; 23:145-155. [PMID: 32635965 DOI: 10.1017/thg.2020.53] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Metabolites are small molecules involved in cellular metabolism where they act as reaction substrates or products. The term 'metabolomics' refers to the comprehensive study of these molecules. The concentrations of metabolites in biological tissues are under genetic control, but this is limited by environmental factors such as diet. In adult mono- and dizygotic twin pairs, we estimated the contribution of genetic and shared environmental influences on metabolite levels by structural equation modeling and tested whether the familial resemblance for metabolite levels is mainly explained by genetic or by environmental factors that are shared by family members. Metabolites were measured across three platforms: two based on proton nuclear magnetic resonance techniques and one employing mass spectrometry. These three platforms comprised 237 single metabolic traits of several chemical classes. For the three platforms, metabolites were assessed in 1407, 1037 and 1116 twin pairs, respectively. We carried out power calculations to establish what percentage of shared environmental variance could be detected given these sample sizes. Our study did not find evidence for a systematic contribution of shared environment, defined as the influence of growing up together in the same household, on metabolites assessed in adulthood. Significant heritability was observed for nearly all 237 metabolites; significant contribution of the shared environment was limited to 6 metabolites. The top quartile of the heritability distribution was populated by 5 of the 11 investigated chemical classes. In this quartile, metabolites of the class lipoprotein were significantly overrepresented, whereas metabolites of classes glycerophospholipids and glycerolipids were significantly underrepresented.
Collapse
|
35
|
Herzog N, Laager R, Thommen E, Widmer M, Vincent AM, Keller A, Becker C, Beck K, Perrig S, Bernasconi L, Neyer P, Marsch S, Schuetz P, Sutter R, Tisljar K, Hunziker S. Association of Taurine with In-Hospital Mortality in Patients after Out-of-Hospital Cardiac Arrest: Results from the Prospective, Observational COMMUNICATE Study. J Clin Med 2020; 9:jcm9051405. [PMID: 32397548 PMCID: PMC7290691 DOI: 10.3390/jcm9051405] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/01/2020] [Accepted: 05/05/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Studies have suggested that taurine may have neuro- and cardio-protective functions, but there is little research looking at taurine levels in patients after out-of-hospital cardiac arrest (OHCA). Our aim was to evaluate the association of taurine with mortality and neurological deficits in a well-defined cohort of OHCA patients. Methods: We prospectively measured serum taurine concentration in OHCA patients upon admission to the intensive care unit (ICU) of the University Hospital Basel (Switzerland). We analyzed the association of taurine levels and in-hospital mortality (primary endpoint). We further evaluated neurological outcomes assessed by the cerebral performance category scale. We calculated logistic regression analyses and report odds ratios (OR) and 95% confidence intervals (CI). We calculated different predefined multivariable regression models including demographic variables, comorbidities, initial vital signs, initial blood markers and resuscitation measures. We assessed discrimination by means of area under the receiver operating curve (ROC). Results: Of 240 included patients, 130 (54.2%) survived until hospital discharge and 110 (45.8%) had a favorable neurological outcome. Taurine levels were significantly associated with higher in-hospital mortality (adjusted OR 4.12 (95%CI 1.22 to 13.91), p = 0.02). In addition, a significant association between taurine concentration and a poor neurological outcome was observed (adjusted OR of 3.71 (95%CI 1.13 to 12.25), p = 0.03). Area under the curve (AUC) suggested only low discrimination for both endpoints (0.57 and 0.57, respectively). Conclusion: Admission taurine levels are associated with mortality and neurological outcomes in OHCA patients and may help in the risk assessment of this vulnerable population. Further studies are needed to assess whether therapeutic modulation of taurine may improve clinical outcomes after cardiac arrest.
Collapse
Affiliation(s)
- Naemi Herzog
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
| | - Rahel Laager
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
| | - Emanuel Thommen
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
| | - Madlaina Widmer
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
| | - Alessia M. Vincent
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
| | - Annalena Keller
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
| | - Christoph Becker
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
- Faculty of Medicine, University of Basel, 4031 Basel, Switzerland; (S.M.); (P.S.); (R.S.)
- Emergency Department, University Hospital Basel, 4031 Basel, Switzerland
| | - Katharina Beck
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
| | - Sebastian Perrig
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
| | - Luca Bernasconi
- Institute of Laboratory Medicine, Kantonsspital Aarau, 5000 Aarau, Switzerland; (L.B.); (P.N.)
| | - Peter Neyer
- Institute of Laboratory Medicine, Kantonsspital Aarau, 5000 Aarau, Switzerland; (L.B.); (P.N.)
| | - Stephan Marsch
- Faculty of Medicine, University of Basel, 4031 Basel, Switzerland; (S.M.); (P.S.); (R.S.)
- Department of Intensive Care, University Hospital Basel, 4031 Basel, Switzerland;
| | - Philipp Schuetz
- Faculty of Medicine, University of Basel, 4031 Basel, Switzerland; (S.M.); (P.S.); (R.S.)
- Department of Internal Medicine, Kantonsspital Aarau, 5000 Aarau, Switzerland
| | - Raoul Sutter
- Faculty of Medicine, University of Basel, 4031 Basel, Switzerland; (S.M.); (P.S.); (R.S.)
- Department of Intensive Care, University Hospital Basel, 4031 Basel, Switzerland;
- Department of Neurology, University Hospital Basel, 4031 Basel, Switzerland
| | - Kai Tisljar
- Department of Intensive Care, University Hospital Basel, 4031 Basel, Switzerland;
| | - Sabina Hunziker
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, 4031 Basel, Switzerland; (N.H.); (R.L.); (E.T.); (M.W.); (A.M.V.); (A.K.); (C.B.); (K.B.); (S.P.)
- Faculty of Medicine, University of Basel, 4031 Basel, Switzerland; (S.M.); (P.S.); (R.S.)
- Department of Intensive Care, University Hospital Basel, 4031 Basel, Switzerland;
- Correspondence: ; Tel.: +41-61-265-25-25
| |
Collapse
|
36
|
Widmer M, Thommen EB, Becker C, Beck K, Vincent AM, Perrig S, Keller A, Bernasconi L, Neyer P, Marsch S, Pargger H, Sutter R, Tisljar K, Hunziker S. Association of acyl carnitines and mortality in out-of-hospital-cardiac-arrest patients: Results of a prospective observational study. J Crit Care 2020; 58:20-26. [PMID: 32279017 DOI: 10.1016/j.jcrc.2020.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 03/23/2020] [Accepted: 03/23/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Out-of-hospital cardiac arrest (OHCA) is a leading cause of mortality, yet the prediction of its outcome remains challenging. Serum Acyl Carnitines (ACs), a biomarker of beta-oxidation, have been associated with cardiovascular events. We evaluated the association of different AC species with mortality and neurological outcome in a cohort of OHCA patients. MATERIAL AND METHODS We consecutively included OHCA patients in this prospective observational study upon admission to the intensive care unit. We studied the association of thirty-nine different ACs measured at admission and 30-day mortality (primary endpoint), as well as neurological outcome at hospital discharge (secondary endpoint) using the Cerebral Performance Category scale. Multivariate models were adjusted for age, gender, comorbidities and shock markers. RESULTS Of 281 included patients, 137 (48.8%) died within 30 days and of the 144 survivors (51.2%), 15 (10.4%) had poor neurological outcome. While several ACs were associated with mortality, AC C2 had the highest prognostic value for mortality (fully-adjusted odds ratio 4.85 (95%CI 1.8 to 13.06, p < .01), area under curve (AUC) 0.65) and neurological outcome (fully-adjusted odds ratio 3.96 (95%CI 1.47 to 10.66, p < .01), AUC 0.63). CONCLUSIONS ACs are interesting surrogate biomarkers that are associated with mortality and poor neurological outcome in patients after OHCA and may help to improve the understanding of pathophysiological mechanisms and risk stratification.
Collapse
Affiliation(s)
- Madlaina Widmer
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, Klingelbergstrasse 23, 4031 Basel, Switzerland
| | - Emanuel B Thommen
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, Klingelbergstrasse 23, 4031 Basel, Switzerland
| | - Christoph Becker
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, Klingelbergstrasse 23, 4031 Basel, Switzerland; Faculty of Medicine, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland; Emergency Department, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| | - Katharina Beck
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, Klingelbergstrasse 23, 4031 Basel, Switzerland
| | - Alessia M Vincent
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, Klingelbergstrasse 23, 4031 Basel, Switzerland
| | - Sebastian Perrig
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, Klingelbergstrasse 23, 4031 Basel, Switzerland
| | - Annalena Keller
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, Klingelbergstrasse 23, 4031 Basel, Switzerland
| | - Luca Bernasconi
- Institute of Laboratory Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland
| | - Peter Neyer
- Institute of Laboratory Medicine, Kantonsspital Aarau, Tellstrasse 25, 5001 Aarau, Switzerland
| | - Stephan Marsch
- Faculty of Medicine, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland; Departement of Intensive Care, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| | - Hans Pargger
- Faculty of Medicine, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland; Departement of Intensive Care, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| | - Raoul Sutter
- Faculty of Medicine, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland; Departement of Intensive Care, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland; Department of Neurology, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| | - Kai Tisljar
- Departement of Intensive Care, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| | - Sabina Hunziker
- Department of Medical Communication and Psychosomatic Medicine, University Hospital Basel, Klingelbergstrasse 23, 4031 Basel, Switzerland; Faculty of Medicine, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland; Departement of Intensive Care, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland.
| |
Collapse
|
37
|
Maruvada P, Lampe JW, Wishart DS, Barupal D, Chester DN, Dodd D, Djoumbou-Feunang Y, Dorrestein PC, Dragsted LO, Draper J, Duffy LC, Dwyer JT, Emenaker NJ, Fiehn O, Gerszten RE, B Hu F, Karp RW, Klurfeld DM, Laughlin MR, Little AR, Lynch CJ, Moore SC, Nicastro HL, O'Brien DM, Ordovás JM, Osganian SK, Playdon M, Prentice R, Raftery D, Reisdorph N, Roche HM, Ross SA, Sang S, Scalbert A, Srinivas PR, Zeisel SH. Perspective: Dietary Biomarkers of Intake and Exposure-Exploration with Omics Approaches. Adv Nutr 2020; 11:200-215. [PMID: 31386148 PMCID: PMC7442414 DOI: 10.1093/advances/nmz075] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
While conventional nutrition research has yielded biomarkers such as doubly labeled water for energy metabolism and 24-h urinary nitrogen for protein intake, a critical need exists for additional, equally robust biomarkers that allow for objective assessment of specific food intake and dietary exposure. Recent advances in high-throughput MS combined with improved metabolomics techniques and bioinformatic tools provide new opportunities for dietary biomarker development. In September 2018, the NIH organized a 2-d workshop to engage nutrition and omics researchers and explore the potential of multiomics approaches in nutritional biomarker research. The current Perspective summarizes key gaps and challenges identified, as well as the recommendations from the workshop that could serve as a guide for scientists interested in dietary biomarkers research. Topics addressed included study designs for biomarker development, analytical and bioinformatic considerations, and integration of dietary biomarkers with other omics techniques. Several clear needs were identified, including larger controlled feeding studies, testing a variety of foods and dietary patterns across diverse populations, improved reporting standards to support study replication, more chemical standards covering a broader range of food constituents and human metabolites, standardized approaches for biomarker validation, comprehensive and accessible food composition databases, a common ontology for dietary biomarker literature, and methodologic work on statistical procedures for intake biomarker discovery. Multidisciplinary research teams with appropriate expertise are critical to moving forward the field of dietary biomarkers and producing robust, reproducible biomarkers that can be used in public health and clinical research.
Collapse
Affiliation(s)
- Padma Maruvada
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Johanna W Lampe
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- School of Public Health, University of Washington, Seattle, WA, USA
| | - David S Wishart
- Departments of Biological Sciences and Computing Science, University of Alberta, Edmonton, Alberta, Canada
| | - Dinesh Barupal
- West Coast Metabolomics Center, UC Davis Genome Center, University of California, Davis, Davis, CA, USA
| | - Deirdra N Chester
- Division of Nutrition, Institute of Food Safety and Nutrition at the National Institute of Food and Agriculture, USDA, Washington, DC, USA
| | - Dylan Dodd
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yannick Djoumbou-Feunang
- Departments of Biological Sciences and Computing Science, University of Alberta, Edmonton, Alberta, Canada
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, USA
| | - Lars O Dragsted
- Department of Nutrition, Exercise, and Sports, Section of Preventive and Clinical Nutrition, University of Copenhagen, Copenhagen, Denmark
| | - John Draper
- Institute of Biological Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, Ceredigion, United Kingdom
| | - Linda C Duffy
- National Institutes of Health, National Center for Complementary and Integrative Health, Bethesda, MD, USA
| | - Johanna T Dwyer
- National Institutes of Health, Office of Dietary Supplements, Bethesda, MD, USA
| | - Nancy J Emenaker
- National Institutes of Health, National Cancer Institute, Rockville, MD, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, UC Davis Genome Center, University of California, Davis, Davis, CA, USA
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Frank B Hu
- Departments of Nutrition; Epidemiology and Statistics, Harvard TH Chan School of Public Health, Boston, MA, USA
- Channing Division for Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Robert W Karp
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - David M Klurfeld
- Department of Nutrition, Food Safety/Quality, USDA—Agricultural Research Service, Beltsville, MD, USA
| | - Maren R Laughlin
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - A Roger Little
- National Institutes of Health, National Institute on Drug Abuse, Bethesda, MD, USA
| | - Christopher J Lynch
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Steven C Moore
- National Institutes of Health, National Cancer Institute, Rockville, MD, USA
| | - Holly L Nicastro
- National Institutes of Health, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Diane M O'Brien
- Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK, USA
| | - José M Ordovás
- Nutrition and Genomics Laboratory, Jean Mayer–USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Stavroula K Osganian
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Mary Playdon
- Department of Nutrition and Integrative Physiology, University of Utah and Division of Cancer Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Ross Prentice
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- School of Public Health, University of Washington, Seattle, WA, USA
| | - Daniel Raftery
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- School of Medicine, University of Washington, Seattle, WA, USA
| | | | - Helen M Roche
- Nutrigenomics Research Group, School of Public Health, Physiotherapy and Sports Science, UCD Institute of Food and Health, Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland
| | - Sharon A Ross
- National Institutes of Health, National Cancer Institute, Rockville, MD, USA
| | - Shengmin Sang
- Laboratory for Functional Foods and Human Health, Center for Excellence in Post-Harvest Technologies, North Carolina A&T State University, North Carolina Research Campus, Nutrition Research Building, Kannapolis, NC, USA
| | - Augustin Scalbert
- International Agency for Research on Cancer, Nutrition and Metabolism Section, Biomarkers Group, Lyon, France
| | - Pothur R Srinivas
- National Institutes of Health, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Steven H Zeisel
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, USA
| |
Collapse
|
38
|
Long NP, Nghi TD, Kang YP, Anh NH, Kim HM, Park SK, Kwon SW. Toward a Standardized Strategy of Clinical Metabolomics for the Advancement of Precision Medicine. Metabolites 2020; 10:E51. [PMID: 32013105 PMCID: PMC7074059 DOI: 10.3390/metabo10020051] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/18/2022] Open
Abstract
Despite the tremendous success, pitfalls have been observed in every step of a clinical metabolomics workflow, which impedes the internal validity of the study. Furthermore, the demand for logistics, instrumentations, and computational resources for metabolic phenotyping studies has far exceeded our expectations. In this conceptual review, we will cover inclusive barriers of a metabolomics-based clinical study and suggest potential solutions in the hope of enhancing study robustness, usability, and transferability. The importance of quality assurance and quality control procedures is discussed, followed by a practical rule containing five phases, including two additional "pre-pre-" and "post-post-" analytical steps. Besides, we will elucidate the potential involvement of machine learning and demonstrate that the need for automated data mining algorithms to improve the quality of future research is undeniable. Consequently, we propose a comprehensive metabolomics framework, along with an appropriate checklist refined from current guidelines and our previously published assessment, in the attempt to accurately translate achievements in metabolomics into clinical and epidemiological research. Furthermore, the integration of multifaceted multi-omics approaches with metabolomics as the pillar member is in urgent need. When combining with other social or nutritional factors, we can gather complete omics profiles for a particular disease. Our discussion reflects the current obstacles and potential solutions toward the progressing trend of utilizing metabolomics in clinical research to create the next-generation healthcare system.
Collapse
Affiliation(s)
- Nguyen Phuoc Long
- College of Pharmacy, Seoul National University, Seoul 08826, Korea; (N.P.L.); (N.H.A.); (H.M.K.)
| | - Tran Diem Nghi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea; (T.D.N.); (S.K.P.)
| | - Yun Pyo Kang
- Department of Cancer Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA;
| | - Nguyen Hoang Anh
- College of Pharmacy, Seoul National University, Seoul 08826, Korea; (N.P.L.); (N.H.A.); (H.M.K.)
| | - Hyung Min Kim
- College of Pharmacy, Seoul National University, Seoul 08826, Korea; (N.P.L.); (N.H.A.); (H.M.K.)
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea; (T.D.N.); (S.K.P.)
| | - Sung Won Kwon
- College of Pharmacy, Seoul National University, Seoul 08826, Korea; (N.P.L.); (N.H.A.); (H.M.K.)
| |
Collapse
|
39
|
Hagenbeek FA, Pool R, van Dongen J, Draisma HHM, Jan Hottenga J, Willemsen G, Abdellaoui A, Fedko IO, den Braber A, Visser PJ, de Geus EJCN, Willems van Dijk K, Verhoeven A, Suchiman HE, Beekman M, Slagboom PE, van Duijn CM, Harms AC, Hankemeier T, Bartels M, Nivard MG, Boomsma DI. Heritability estimates for 361 blood metabolites across 40 genome-wide association studies. Nat Commun 2020; 11:39. [PMID: 31911595 PMCID: PMC6946682 DOI: 10.1038/s41467-019-13770-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/25/2019] [Indexed: 01/16/2023] Open
Abstract
Metabolomics examines the small molecules involved in cellular metabolism. Approximately 50% of total phenotypic differences in metabolite levels is due to genetic variance, but heritability estimates differ across metabolite classes. We perform a review of all genome-wide association and (exome-) sequencing studies published between November 2008 and October 2018, and identify >800 class-specific metabolite loci associated with metabolite levels. In a twin-family cohort (N = 5117), these metabolite loci are leveraged to simultaneously estimate total heritability (h2total), and the proportion of heritability captured by known metabolite loci (h2Metabolite-hits) for 309 lipids and 52 organic acids. Our study reveals significant differences in h2Metabolite-hits among different classes of lipids and organic acids. Furthermore, phosphatidylcholines with a high degree of unsaturation have higher h2Metabolite-hits estimates than phosphatidylcholines with low degrees of unsaturation. This study highlights the importance of common genetic variants for metabolite levels, and elucidates the genetic architecture of metabolite classes.
Collapse
Affiliation(s)
- Fiona A Hagenbeek
- 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
| | - Jenny van Dongen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Harmen H M Draisma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jouke Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Gonneke Willemsen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Abdel Abdellaoui
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Iryna O Fedko
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Anouk den Braber
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, VU Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Department of Neurology, VU Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Psychiatry and Neuropsychology, School of Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | - Eco J C N de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Ko Willems van Dijk
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
- 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
| | - Aswin Verhoeven
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - H Eka Suchiman
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marian Beekman
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, 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
| | | | - Amy C Harms
- Division of Analytical Biosciences, Leiden Academic Center for Drug Research, Leiden University and The Netherlands Metabolomics Centre, Leiden, The Netherlands
| | - Thomas Hankemeier
- Division of Analytical Biosciences, Leiden Academic Center for Drug Research, Leiden University and The Netherlands Metabolomics Centre, Leiden, The Netherlands
| | - Meike Bartels
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Michel G Nivard
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam, The Netherlands.
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
40
|
Transcriptomic and metabolomic adaptation of Nannochloropsis gaditana grown under different light regimes. ALGAL RES 2020. [DOI: 10.1016/j.algal.2019.101735] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
41
|
Baumgartner T, Zurauskaite G, Steuer C, Bernasconi L, Huber A, Mueller B, Schuetz P. Association of serum sphingomyelin profile with clinical outcomes in patients with lower respiratory tract infections: results of an observational, prospective 6-year follow-up study. Clin Chem Lab Med 2019; 57:679-689. [PMID: 30267624 DOI: 10.1515/cclm-2018-0509] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 08/21/2018] [Indexed: 01/11/2023]
Abstract
Background Sphingolipids - the structural cell membrane components - and their metabolites are involved in signal transduction and participate in the regulation of immunity. We investigated the prognostic implications of sphingolipid metabolic profiling on mortality in a large cohort of patients with lower respiratory tract infections (LRTIs). Methods We measured 15 different sphingomyelin (SM) types in patients with LRTIs from a previous Swiss multicenter trial that examined the impact of procalcitonin-guided antibiotic therapy on total antibiotic use and rates and duration of hospitalization. Primary and secondary end points were adverse outcomes - defined as death or intensive care unit admission within 30 days - and 6-year mortality. Results Of 360 patients, 8.9% experienced an adverse outcome within 30 days and 46% died within 6 years. Levels of all SM types were significantly lower in pneumonia patients vs. those with chronic obstructive pulmonary disease (COPD) exacerbation (p<0.0001 for all comparisons). Sphingomyelin subspecies SM (OH) C22:1 and SM (OH) C22:2 were associated with lower risk for short-term adverse outcomes (sex-, gender- and comorbidity-adjusted odds ratios [OR]: 0.036; 95% confidence interval [CI], 0.002-0.600; p=0.021 and 0.037; 95% CI, 0.001-0.848; p=0.039, respectively). We found no significant associations with 6-year mortality for any SM. Conclusions Circulating sphingolipid levels are lower in inflammatory conditions such as pneumonia and correlate with adverse short-term outcomes. Further characterization of the physiological, pathophysiological and metabolic roles of sphingolipids under inflammatory conditions may facilitate understanding of their roles in infectious disease.
Collapse
Affiliation(s)
- Thomas Baumgartner
- Division of Endocrinology, Diabetology and Metabolism, Medical University Department, Kantonsspital Aarau, Aarau, Switzerland, Phone: 0041 62 838 68 32, Fax: 0041 62 838 98 73.,University Department of Internal Medicine, Kantonsspital Aarau, Tellstr., 5001 Aarau, Switzerland
| | - Giedre Zurauskaite
- Division of Endocrinology, Diabetology and Metabolism, Medical University Department, Kantonsspital Aarau, Aarau, Switzerland
| | - Christian Steuer
- Department of Laboratory Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Luca Bernasconi
- Department of Laboratory Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Andreas Huber
- Department of Laboratory Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Beat Mueller
- Division of Endocrinology, Diabetology and Metabolism, Medical University Department, Kantonsspital Aarau, Aarau, Switzerland
| | - Philipp Schuetz
- Division of Endocrinology, Diabetology and Metabolism, Medical University Department, Kantonsspital Aarau, Aarau, Switzerland
| |
Collapse
|
42
|
Al-Khelaifi F, Diboun I, Donati F, Botrè F, Abraham D, Hingorani A, Albagha O, Georgakopoulos C, Suhre K, Yousri NA, Elrayess MA. Metabolic GWAS of elite athletes reveals novel genetically-influenced metabolites associated with athletic performance. Sci Rep 2019; 9:19889. [PMID: 31882771 PMCID: PMC6934758 DOI: 10.1038/s41598-019-56496-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 12/12/2019] [Indexed: 01/10/2023] Open
Abstract
Genetic research of elite athletic performance has been hindered by the complex phenotype and the relatively small effect size of the identified genetic variants. The aims of this study were to identify genetic predisposition to elite athletic performance by investigating genetically-influenced metabolites that discriminate elite athletes from non-elite athletes and to identify those associated with endurance sports. By conducting a genome wide association study with high-resolution metabolomics profiling in 490 elite athletes, common variant metabolic quantitative trait loci (mQTLs) were identified and compared with previously identified mQTLs in non-elite athletes. Among the identified mQTLs, those associated with endurance metabolites were determined. Two novel genetic loci in FOLH1 and VNN1 are reported in association with N-acetyl-aspartyl-glutamate and Linoleoyl ethanolamide, respectively. When focusing on endurance metabolites, one novel mQTL linking androstenediol (3alpha, 17alpha) monosulfate and SULT2A1 was identified. Potential interactions between the novel identified mQTLs and exercise are highlighted. This is the first report of common variant mQTLs linked to elite athletic performance and endurance sports with potential applications in biomarker discovery in elite athletic candidates, non-conventional anti-doping analytical approaches and therapeutic strategies.
Collapse
Affiliation(s)
- Fatima Al-Khelaifi
- Anti Doping Laboratory Qatar, Sports City, Doha, Qatar.,Division of Medicine, University College London, London, NW3 2PF, United Kingdom
| | - Ilhame Diboun
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Francesco Donati
- Laboratorio Antidoping, Federazione Medico Sportiva Italiana, Largo Giulio Onesti 1, 00197, Rome, Italy
| | - Francesco Botrè
- Laboratorio Antidoping, Federazione Medico Sportiva Italiana, Largo Giulio Onesti 1, 00197, Rome, Italy
| | - David Abraham
- Division of Medicine, University College London, London, NW3 2PF, United Kingdom
| | - Aroon Hingorani
- UCL Institute of Cardiovascular Science, University College London, London, WC1E 6BT, United Kingdom
| | - Omar Albagha
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.,Center for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, UK
| | | | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medical College in Qatar, Qatar-Foundation, P.O. Box 24144, Doha, Qatar
| | - Noha A Yousri
- Department of Genetic Medicine, Weill Cornell Medical College in Qatar, Qatar-Foundation, P.O. Box 24144, Doha, Qatar.,Computer and Systems Engineering, Alexandria University, Alexandria, Egypt
| | | |
Collapse
|
43
|
Characterization of Bulk Phosphatidylcholine Compositions in Human Plasma Using Side-Chain Resolving Lipidomics. Metabolites 2019; 9:metabo9060109. [PMID: 31181753 PMCID: PMC6631474 DOI: 10.3390/metabo9060109] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 01/05/2023] Open
Abstract
Kit-based assays, such as AbsoluteIDQTM p150, are widely used in large cohort studies and provide a standardized method to quantify blood concentrations of phosphatidylcholines (PCs). Many disease-relevant associations of PCs were reported using this method. However, their interpretation is hampered by lack of functionally-relevant information on the detailed fatty acid side-chain compositions as only the total number of carbon atoms and double bonds is identified by the kit. To enable more substantiated interpretations, we characterized these PC sums using the side-chain resolving LipidyzerTM platform, analyzing 223 samples in parallel to the AbsoluteIDQTM. Combining these datasets, we estimated the quantitative composition of PC sums and subsequently tested their replication in an independent cohort. We identified major constituents of 28 PC sums, revealing also various unexpected compositions. As an example, PC 16:0_22:5 accounted for more than 50% of the PC sum with in total 38 carbon atoms and 5 double bonds (PC aa 38:5). For 13 PC sums, we found relatively high abundances of odd-chain fatty acids. In conclusion, our study provides insights in PC compositions in human plasma, facilitating interpretation of existing epidemiological data sets and potentially enabling imputation of PC compositions for future meta-analyses of lipidomics data.
Collapse
|
44
|
Yu B, Zanetti KA, Temprosa M, Albanes D, Appel N, Barrera CB, Ben-Shlomo Y, Boerwinkle E, Casas JP, Clish C, Dale C, Dehghan A, Derkach A, Eliassen AH, Elliott P, Fahy E, Gieger C, Gunter MJ, Harada S, Harris T, Herr DR, Herrington D, Hirschhorn JN, Hoover E, Hsing AW, Johansson M, Kelly RS, Khoo CM, Kivimäki M, Kristal BS, Langenberg C, Lasky-Su J, Lawlor DA, Lotta LA, Mangino M, Le Marchand L, Mathé E, Matthews CE, Menni C, Mucci LA, Murphy R, Oresic M, Orwoll E, Ose J, Pereira AC, Playdon MC, Poston L, Price J, Qi Q, Rexrode K, Risch A, Sampson J, Seow WJ, Sesso HD, Shah SH, Shu XO, Smith GCS, Sovio U, Stevens VL, Stolzenberg-Solomon R, Takebayashi T, Tillin T, Travis R, Tzoulaki I, Ulrich CM, Vasan RS, Verma M, Wang Y, Wareham NJ, Wong A, Younes N, Zhao H, Zheng W, Moore SC. The Consortium of Metabolomics Studies (COMETS): Metabolomics in 47 Prospective Cohort Studies. Am J Epidemiol 2019; 188:991-1012. [PMID: 31155658 PMCID: PMC6545286 DOI: 10.1093/aje/kwz028] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 01/29/2019] [Accepted: 01/29/2019] [Indexed: 12/11/2022] Open
Abstract
The Consortium of Metabolomics Studies (COMETS) was established in 2014 to facilitate large-scale collaborative research on the human metabolome and its relationship with disease etiology, diagnosis, and prognosis. COMETS comprises 47 cohorts from Asia, Europe, North America, and South America that together include more than 136,000 participants with blood metabolomics data on samples collected from 1985 to 2017. Metabolomics data were provided by 17 different platforms, with the most frequently used labs being Metabolon, Inc. (14 cohorts), the Broad Institute (15 cohorts), and Nightingale Health (11 cohorts). Participants have been followed for a median of 23 years for health outcomes including death, cancer, cardiovascular disease, diabetes, and others; many of the studies are ongoing. Available exposure-related data include common clinical measurements and behavioral factors, as well as genome-wide genotype data. Two feasibility studies were conducted to evaluate the comparability of metabolomics platforms used by COMETS cohorts. The first study showed that the overlap between any 2 different laboratories ranged from 6 to 121 metabolites at 5 leading laboratories. The second study showed that the median Spearman correlation comparing 111 overlapping metabolites captured by Metabolon and the Broad Institute was 0.79 (interquartile range, 0.56-0.89).
Collapse
Affiliation(s)
- Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas
| | - Krista A Zanetti
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, Maryland
| | - Marinella Temprosa
- Department of Epidemiology and Biostatistics Milken Institute School of Public Health, George Washington University, Washington, DC
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Nathan Appel
- Information Management Services, Inc., Rockville, Maryland
| | - Clara Barrios Barrera
- Department of Nephrology, Hospital del Mar, Institut Mar d´Investigacions Mediques, Barcelona, Spain
| | - Yoav Ben-Shlomo
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas
| | - Juan P Casas
- Institute of Health Informatics Research, UCL Institute of Health Informatics, University College London, London, United Kingdom
| | - Clary Clish
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Caroline Dale
- Institute of Health Informatics Research, UCL Institute of Health Informatics, University College London, London, United Kingdom
| | - Abbas Dehghan
- Medical Research Council–Public Health England Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Andriy Derkach
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - A Heather Eliassen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston Massachusetts
| | - Paul Elliott
- Medical Research Council–Public Health England Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- National Institute for Health Research, Imperial College Biomedical Research Center, London, United Kingdom
- Health Data Research UK Center at Imperial College London, London, United Kingdom
| | - Eoin Fahy
- Department of Bioengineering, School of Engineering, University of California, San Diego, La Jolla, California
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research
| | - Marc J Gunter
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | - Sei Harada
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Tamara Harris
- Laboratory of Epidemiology and Population Science Laboratory
| | - Deron R Herr
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Biology, San Diego State University, San Diego, California
| | - David Herrington
- Department of Internal Medicine, Division of Cardiology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Joel N Hirschhorn
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Elise Hoover
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, Maryland
| | - Ann W Hsing
- Stanford Prevention Research Center, Stanford Cancer Institute, Stanford, California
| | | | - Rachel S Kelly
- Systems Genetics and Genomics Unit, Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Chin Meng Khoo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Medicine, National University Health System, Singapore
- Duke–National University of Singapore Graduate Medical School, Singapore
| | - Mika Kivimäki
- Department of Epidemiology and Public Health, University College London, London, United Kingdom
| | - Bruce S Kristal
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Division of Sleep Medicine, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Jessica Lasky-Su
- Channing Laboratory, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Deborah A Lawlor
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom
| | - Luca A Lotta
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, United Kingdom
| | - Loïc Le Marchand
- University of Hawaii Cancer Center, Epidemiology Program, Honolulu, Hawaii
| | - Ewy Mathé
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, Ohio
| | - Charles E Matthews
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, United Kingdom
| | - Lorelei A Mucci
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston Massachusetts
| | - Rachel Murphy
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Matej Oresic
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
- School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Eric Orwoll
- Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Jennifer Ose
- Division of Cancer Population Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Alexandre C Pereira
- Instituto de Pesquisas Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Mary C Playdon
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
- Division of Cancer Population Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| | - Lucilla Poston
- Department of Women and Children’s Health, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King’s College London, St. Thomas’ Hospital, London, United Kingdom
| | - Jackie Price
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Kathryn Rexrode
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Division of Women’s Health, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Adam Risch
- Information Management Services, Inc., Rockville, Maryland
| | - Joshua Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Wei Jie Seow
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Howard D Sesso
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston Massachusetts
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Svati H Shah
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
- Duke Clinical Research Institute, Durham, North Carolina
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt Epidemiology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Gordon C S Smith
- Department of Obstetrics and Gynaecology, National Institute for Health Research, Cambridge Comprehensive Biomedical Research Center, University of Cambridge, Cambridge, United Kingdom
| | - Ulla Sovio
- Center for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Victoria L Stevens
- Department of Obstetrics and Gynaecology, University of Cambridge, National Institute for Health Research Cambridge Comprehensive Biomedical Research Centre, Cambridge, United Kingdom
| | | | - Toru Takebayashi
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia
| | - Therese Tillin
- Institute of Cardiovascular Sciences, University College London, London, United Kingdom
| | - Ruth Travis
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Ioanna Tzoulaki
- Medical Research Council–Public Health England Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Cornelia M Ulrich
- Division of Cancer Population Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Ramachandran S Vasan
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Section of Cardiovascular Medicine, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts
- Framingham Heart Study, Framingham, Massachusetts
| | - Mukesh Verma
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, Maryland
| | - Ying Wang
- Department of Obstetrics and Gynaecology, University of Cambridge, National Institute for Health Research Cambridge Comprehensive Biomedical Research Centre, Cambridge, United Kingdom
| | - Nick J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Andrew Wong
- MRC Unit for Lifelong Health and Ageing at University College London, London, United Kingdom
| | - Naji Younes
- Department of Epidemiology and Biostatistics Milken Institute School of Public Health, George Washington University, Washington, DC
| | - Hua Zhao
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt Epidemiology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Steven C Moore
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| |
Collapse
|
45
|
Bermingham KM, Brennan L, Segurado R, Barron RE, Gibney ER, Ryan MF, Gibney MJ, O’Sullivan AM. Exploring Covariation between Traditional Markers of Metabolic Health and the Plasma Metabolomic Profile: A Classic Twin Design. J Proteome Res 2019; 18:2613-2623. [DOI: 10.1021/acs.jproteome.9b00126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Kate M. Bermingham
- UCD Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Lorraine Brennan
- UCD Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Ricardo Segurado
- School of Public Health, Physiotherapy, and Sports Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Rebecca E. Barron
- UCD Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Eileen R. Gibney
- UCD Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Miriam F. Ryan
- UCD Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Michael J. Gibney
- UCD Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Aifric M. O’Sullivan
- UCD Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
46
|
Teslovich TM, Kim DS, Yin X, Stancáková A, Jackson AU, Wielscher M, Naj A, Perry JRB, Huyghe JR, Stringham HM, Davis JP, Raulerson CK, Welch RP, Fuchsberger C, Locke AE, Sim X, Chines PS, Narisu N, Kangas AJ, Soininen P, Ala-Korpela M, Gudnason V, Musani SK, Jarvelin MR, Schellenberg GD, Speliotes EK, Kuusisto J, Collins FS, Boehnke M, Laakso M, Mohlke KL. Identification of seven novel loci associated with amino acid levels using single-variant and gene-based tests in 8545 Finnish men from the METSIM study. Hum Mol Genet 2019; 27:1664-1674. [PMID: 29481666 DOI: 10.1093/hmg/ddy067] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 02/15/2018] [Indexed: 12/13/2022] Open
Abstract
Comprehensive metabolite profiling captures many highly heritable traits, including amino acid levels, which are potentially sensitive biomarkers for disease pathogenesis. To better understand the contribution of genetic variation to amino acid levels, we performed single variant and gene-based tests of association between nine serum amino acids (alanine, glutamine, glycine, histidine, isoleucine, leucine, phenylalanine, tyrosine, and valine) and 16.6 million genotyped and imputed variants in 8545 non-diabetic Finnish men from the METabolic Syndrome In Men (METSIM) study with replication in Northern Finland Birth Cohort (NFBC1966). We identified five novel loci associated with amino acid levels (P = < 5×10-8): LOC157273/PPP1R3B with glycine (rs9987289, P = 2.3×10-26); ZFHX3 (chr16:73326579, minor allele frequency (MAF) = 0.42%, P = 3.6×10-9), LIPC (rs10468017, P = 1.5×10-8), and WWOX (rs9937914, P = 3.8×10-8) with alanine; and TRIB1 with tyrosine (rs28601761, P = 8×10-9). Gene-based tests identified two novel genes harboring missense variants of MAF <1% that show aggregate association with amino acid levels: PYCR1 with glycine (Pgene = 1.5×10-6) and BCAT2 with valine (Pgene = 7.4×10-7); neither gene was implicated by single variant association tests. These findings are among the first applications of gene-based tests to identify new loci for amino acid levels. In addition to the seven novel gene associations, we identified five independent signals at established amino acid loci, including two rare variant signals at GLDC (rs138640017, MAF=0.95%, Pconditional = 5.8×10-40) with glycine levels and HAL (rs141635447, MAF = 0.46%, Pconditional = 9.4×10-11) with histidine levels. Examination of all single variant association results in our data revealed a strong inverse relationship between effect size and MAF (Ptrend<0.001). These novel signals provide further insight into the molecular mechanisms of amino acid metabolism and potentially, their perturbations in disease.
Collapse
Affiliation(s)
- Tanya M Teslovich
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Daniel Seung Kim
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xianyong Yin
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alena Stancáková
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Matthias Wielscher
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | - Adam Naj
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, University of Pennsylvania, PA 19104, USA.,Departments of Biostatistics, and Epidemiology (DBE) and Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, PA 19104, USA
| | - John R B Perry
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Jeroen R Huyghe
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Heather M Stringham
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - James P Davis
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Chelsea K Raulerson
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ryan P Welch
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Christian Fuchsberger
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Adam E Locke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xueling Sim
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter S Chines
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Narisu Narisu
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Antti J Kangas
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
| | - Pasi Soininen
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland.,NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | | | - Mika Ala-Korpela
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland.,NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland.,Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK.,Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK.,Systems Epidemiology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Epidemiology and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, School of Public Health and Preventive Medicine, The Alfred Hospital, Monash University, Melbourne, VIC, Australia
| | - Vilmundur Gudnason
- Icelandic Heart Association and the Faculty of Medicine, University of Iceland, Kopavogur, Iceland
| | - Solomon K Musani
- University of Mississippi Medical Center, Jackson, MS 39213, USA
| | - Marjo-Riitta Jarvelin
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK.,Center for Life Course Health Research, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland.,Biocenter Oulu, University of Oulu, 90014 Oulu, Finland.,Unit of Primary Care, Oulu University Hospital, Oulu, Finland
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, University of Pennsylvania, PA 19104, USA
| | - Elizabeth K Speliotes
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Francis S Collins
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
47
|
Köttgen A, Raffler J, Sekula P, Kastenmüller G. Genome-Wide Association Studies of Metabolite Concentrations (mGWAS): Relevance for Nephrology. Semin Nephrol 2019; 38:151-174. [PMID: 29602398 DOI: 10.1016/j.semnephrol.2018.01.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Metabolites are small molecules that are intermediates or products of metabolism, many of which are freely filtered by the kidneys. In addition, the kidneys have a central role in metabolite anabolism and catabolism, as well as in active metabolite reabsorption and/or secretion during tubular passage. This review article illustrates how the coupling of genomics and metabolomics in genome-wide association analyses of metabolites can be used to illuminate mechanisms underlying human metabolism, with a special focus on insights relevant to nephrology. First, genetic susceptibility loci for reduced kidney function and chronic kidney disease (CKD) were reviewed systematically for their associations with metabolite concentrations in metabolomics studies of blood and urine. Second, kidney function and CKD-associated metabolites reported from observational studies were interrogated for metabolite-associated genetic variants to generate and discuss complementary insights. Finally, insights originating from the simultaneous study of both blood and urine or by modeling intermetabolite relationships are summarized. We also discuss methodologic questions related to the study of metabolite concentrations in urine as well as among CKD patients. In summary, genome-wide association analyses of metabolites using metabolite concentrations quantified from blood and/or urine are a promising avenue of research to illuminate physiological and pathophysiological functions of the kidney.
Collapse
Affiliation(s)
- Anna Köttgen
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany.
| | - Johannes Raffler
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Peggy Sekula
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| |
Collapse
|
48
|
Seldin MM, Lusis AJ. Systems-based approaches for investigation of inter-tissue communication. J Lipid Res 2019; 60:450-455. [PMID: 30617149 PMCID: PMC6399495 DOI: 10.1194/jlr.s090316] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/27/2018] [Indexed: 11/23/2022] Open
Abstract
Secreted proteins serve as crucial mediators of many physiology processes, and beginning with the discovery of insulin, studies have revealed numerous context-specific regulatory networks across various cell types. Here, we review “omics” approaches to deconvolute the complex milieu of proteins that are released from the cell. We emphasize a novel “systems genetics” approach our laboratory has developed to investigate mechanisms of tissue-tissue communication using population-based datasets. Finally, we highlight potential future directions for these studies, discuss several caveats, and propose new ways to investigate modes of endocrine communication.
Collapse
Affiliation(s)
- Marcus M Seldin
- Departments of Medicine, University of California, Los Angeles, Los Angeles, CA 90095
| | - Aldons J Lusis
- Departments of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 .,Human Genetics University of California, Los Angeles, Los Angeles, CA 90095.,Microbiology, Immunology, and Molecular Genetics University of California, Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
49
|
Rhee EP, Waikar SS, Rebholz CM, Zheng Z, Perichon R, Clish CB, Evans AM, Avila J, Denburg MR, Anderson AH, Vasan RS, Feldman HI, Kimmel PL, Coresh J. Variability of Two Metabolomic Platforms in CKD. Clin J Am Soc Nephrol 2018; 14:40-48. [PMID: 30573658 PMCID: PMC6364529 DOI: 10.2215/cjn.07070618] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 10/15/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND OBJECTIVES Nontargeted metabolomics can measure thousands of low-molecular-weight biochemicals, but important gaps limit its utility for biomarker discovery in CKD. These include the need to characterize technical and intraperson analyte variation, to pool data across platforms, and to outline analyte relationships with eGFR. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Plasma samples from 49 individuals with CKD (eGFR<60 ml/min per 1.73 m2 and/or ≥1 g proteinuria) were examined from two study visits; 20 samples were repeated as blind replicates. To enable comparison across two nontargeted platforms, samples were profiled at Metabolon and the Broad Institute. RESULTS The Metabolon platform reported 837 known metabolites and 483 unnamed compounds (selected from 44,953 unknown ion features). The Broad Institute platform reported 594 known metabolites and 26,106 unknown ion features. Median coefficients of variation (CVs) across blind replicates were 14.6% (Metabolon) and 6.3% (Broad Institute) for known metabolites, and 18.9% for (Metabolon) unnamed compounds and 24.5% for (Broad Institute) unknown ion features. Median CVs for day-to-day variability were 29.0% (Metabolon) and 24.9% (Broad Institute) for known metabolites, and 41.8% for (Metabolon) unnamed compounds and 40.9% for (Broad Institute) unknown ion features. A total of 381 known metabolites were shared across platforms (median correlation 0.89). Many metabolites were negatively correlated with eGFR at P<0.05, including 35.7% (Metabolon) and 18.9% (Broad Institute) of known metabolites. CONCLUSIONS Nontargeted metabolomics quantifies >1000 analytes with low technical CVs, and agreement for overlapping metabolites across two leading platforms is excellent. Many metabolites demonstrate substantial intraperson variation and correlation with eGFR.
Collapse
Affiliation(s)
- Eugene P Rhee
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts;
| | - Sushrut S Waikar
- Renal Division, Brigham and Women's Hospital, Boston, Massachusetts
| | - Casey M Rebholz
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland.,Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Zihe Zheng
- Department of Biostatistics, Epidemiology, and Informatics
| | | | - Clary B Clish
- Metabolite Profiling, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | | | - Julian Avila
- Metabolite Profiling, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | | | | | - Ramachandran S Vasan
- Sections of Preventive Medicine and Epidemiology and Cardiology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts; and
| | - Harold I Feldman
- Department of Biostatistics, Epidemiology, and Informatics.,Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Paul L Kimmel
- Division of Kidney Urologic and Hematologic Diseases, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Josef Coresh
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland; .,Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | |
Collapse
|
50
|
Contrepois K, Mahmoudi S, Ubhi BK, Papsdorf K, Hornburg D, Brunet A, Snyder M. Cross-Platform Comparison of Untargeted and Targeted Lipidomics Approaches on Aging Mouse Plasma. Sci Rep 2018; 8:17747. [PMID: 30532037 PMCID: PMC6288111 DOI: 10.1038/s41598-018-35807-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 11/06/2018] [Indexed: 11/18/2022] Open
Abstract
Lipidomics – the global assessment of lipids – can be performed using a variety of mass spectrometry (MS)-based approaches. However, choosing the optimal approach in terms of lipid coverage, robustness and throughput can be a challenging task. Here, we compare a novel targeted quantitative lipidomics platform known as the Lipidyzer to a conventional untargeted liquid chromatography (LC)-MS approach. We find that both platforms are efficient in profiling more than 300 lipids across 11 lipid classes in mouse plasma with precision and accuracy below 20% for most lipids. While the untargeted and targeted platforms detect similar numbers of lipids, the former identifies a broader range of lipid classes and can unambiguously identify all three fatty acids in triacylglycerols (TAG). Quantitative measurements from both approaches exhibit a median correlation coefficient (r) of 0.99 using a dilution series of deuterated internal standards and 0.71 using endogenous plasma lipids in the context of aging. Application of both platforms to plasma from aging mouse reveals similar changes in total lipid levels across all major lipid classes and in specific lipid species. Interestingly, TAG is the lipid class that exhibits the most changes with age, suggesting that TAG metabolism is particularly sensitive to the aging process in mice. Collectively, our data show that the Lipidyzer platform provides comprehensive profiling of the most prevalent lipids in plasma in a simple and automated manner.
Collapse
Affiliation(s)
- Kévin Contrepois
- Department of Genetics, Stanford University, 300 Pasteur Drive, Stanford, California, 94305, USA
| | - Salah Mahmoudi
- Department of Genetics, Stanford University, 300 Pasteur Drive, Stanford, California, 94305, USA
| | - Baljit K Ubhi
- SCIEX, 1201 Radio Rd, Redwood City, California, 94065, USA
| | - Katharina Papsdorf
- Department of Genetics, Stanford University, 300 Pasteur Drive, Stanford, California, 94305, USA
| | - Daniel Hornburg
- Department of Genetics, Stanford University, 300 Pasteur Drive, Stanford, California, 94305, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, 300 Pasteur Drive, Stanford, California, 94305, USA
| | - Michael Snyder
- Department of Genetics, Stanford University, 300 Pasteur Drive, Stanford, California, 94305, USA.
| |
Collapse
|