1
|
Rodriguez Franco G, Hsu CC. Uncovering the sex steroid hormone secrets in alcohol. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025; 49:95-98. [PMID: 39523479 DOI: 10.1111/acer.15479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Affiliation(s)
- Gian Rodriguez Franco
- Liver Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Christine C Hsu
- Liver Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| |
Collapse
|
2
|
Zeng K, Du J, Chen YZ, Wang DY, Sun ML, Li YZ, Wang DY, Liu SH, Zhu XM, Lv P, Du Z, Liu K, Yao J. Metabolomics efficiently discriminates monozygotic twins in peripheral blood. Int J Legal Med 2024; 138:2249-2258. [PMID: 38858273 DOI: 10.1007/s00414-024-03269-1] [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: 11/15/2023] [Accepted: 06/03/2024] [Indexed: 06/12/2024]
Abstract
Monozygotic (MZ) twins cannot be distinguished using conventional forensic STR typing because they present identical STR genotypings. However, MZ twins do not always live in the same environment and often have different dietary and other lifestyle habits. Metabolic profiles are deyermined by individual characteristics and are also influenced by the environment in which they live. Therefore, they are potential markers capable of identifying MZ twins. Moreover, the production of proteins varies from organism to organism and is influenced by both the physiological state of the body and the external environment. Hence, we used metabolomics and proteomics to identify metabolites and proteins in peripheral blood to discriminate MZ twins. We identified 1749 known metabolites and 622 proteins in proteomic analysis. The metabolic profiles of four pairs of MZ twins revealed minor differences in intra-MZ twins and major differences in inter-MZ twins. Each pair of MZ twins exhibited distinct characteristics, and four metabolites-methyl picolinate, acesulfame, paraxanthine, and phenylbenzimidazole sulfonic acid-were observed in all four MZ twin pairs. These four differential exogenous metabolites conincidently show that the different external environments and life styles can be well distinguished by metabolites, considering that twins do not all have the same eating habits and living environments. Moreover, MZ twins showed different protein profiles in serum but not in whole blood. Thus, our results indicate that differential metabolites provide potential biomarkers for the personal identification of MZ twins in forensic medicine.
Collapse
Affiliation(s)
- Kuo Zeng
- School of Forensic Medicine, China Medical University, No.77, Puhe Road, Shenbei New District, Shenyang, 110122, P.R. China
- Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, P.R. China
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China
- Institute of Evidence Law and Forensic Science, China University of Political Science and Law, Beijing, China
| | - Jiang Du
- Department of Pathology, School of Basic Medicine, China Medical University, Shenyang, P.R. China
| | - Yun-Zhou Chen
- School of Forensic Medicine, China Medical University, No.77, Puhe Road, Shenbei New District, Shenyang, 110122, P.R. China
- Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, P.R. China
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China
| | - Dan-Yang Wang
- School of Forensic Medicine, China Medical University, No.77, Puhe Road, Shenbei New District, Shenyang, 110122, P.R. China
- Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, P.R. China
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China
| | - Mao-Ling Sun
- School of Forensic Medicine, China Medical University, No.77, Puhe Road, Shenbei New District, Shenyang, 110122, P.R. China
- Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, P.R. China
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China
| | - Yu-Zhang Li
- School of Forensic Medicine, China Medical University, No.77, Puhe Road, Shenbei New District, Shenyang, 110122, P.R. China
- Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, P.R. China
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China
| | - Dong-Yi Wang
- School of Forensic Medicine, China Medical University, No.77, Puhe Road, Shenbei New District, Shenyang, 110122, P.R. China
- Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, P.R. China
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China
| | - Shu-Han Liu
- School of Forensic Medicine, China Medical University, No.77, Puhe Road, Shenbei New District, Shenyang, 110122, P.R. China
- Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, P.R. China
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China
| | - Xiu-Mei Zhu
- School of Forensic Medicine, China Medical University, No.77, Puhe Road, Shenbei New District, Shenyang, 110122, P.R. China
- Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, P.R. China
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China
| | - Peng Lv
- School of Forensic Medicine, China Medical University, No.77, Puhe Road, Shenbei New District, Shenyang, 110122, P.R. China
- Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, P.R. China
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China
| | - Zhe Du
- School of Forensic Medicine, China Medical University, No.77, Puhe Road, Shenbei New District, Shenyang, 110122, P.R. China
- Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, P.R. China
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China
| | - Kun Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, P.R. China.
| | - Jun Yao
- School of Forensic Medicine, China Medical University, No.77, Puhe Road, Shenbei New District, Shenyang, 110122, P.R. China.
- Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, P.R. China.
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China.
| |
Collapse
|
3
|
Yang J, Bernard L, Wong KE, Yu B, Steffen LM, Sullivan VK, Rebholz CM. Serum metabolite signature of the modified Mediterranean-DASH intervention for neurodegenerative delay (MIND) diet. Metabolomics 2024; 20:118. [PMID: 39432124 DOI: 10.1007/s11306-024-02184-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/10/2024] [Indexed: 10/22/2024]
Abstract
INTRODUCTION There is a lack of biomarkers of clinically important diets, such as the Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND) diet. OBJECTIVES Our study explored serum metabolites associated with adherence to the MIND diet. METHODS In 3,908 Atherosclerosis Risk in Communities (ARIC) study participants, we calculated a modified MIND diet score based on a 66-item self-reported food frequency questionnaire (FFQ). The modified score did not include berries and olive oil, as these items were not assessed in the FFQ. We used multivariable linear regression models in 2 subgroups of ARIC study participants and meta-analyzed results using fixed effects regression to identify significant metabolites after Bonferroni correction. We also examined associations between these metabolites and food components of the modified MIND diet. C-statistics evaluated the prediction of high modified MIND diet adherence using significant metabolites beyond participant characteristics. RESULTS Of 360 metabolites analyzed, 27 metabolites (15 positive, 12 negative) were significantly associated with the modified MIND diet score (lipids, n = 13; amino acids, n = 5; xenobiotics, n = 3; cofactors and vitamins, n = 3; carbohydrates n = 2; nucleotide n = 1). The top 4 metabolites that improved the prediction of high dietary adherence to the modified MIND diet were 7-methylxanthine, theobromine, docosahexaenoate (DHA), and 3-carboxy-4-methyl-5-propyl-2-furanpropanoate (CMPF). CONCLUSION Twenty-seven metabolomic markers were correlated with the modified MIND diet. The biomarkers, if further validated, could be useful to objectively assess adherence to the MIND diet.
Collapse
Affiliation(s)
- Jiaqi Yang
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD, USA
| | - Lauren Bernard
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD, USA
- School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Kari E Wong
- Metabolon, Research Triangle Park, Morrisville, NC, USA
| | - Bing Yu
- Department of Epidemiology, University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Lyn M Steffen
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Valerie K Sullivan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD, USA
| | - Casey M Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
4
|
de la O V, Fernández-Cruz E, Valdés A, Cifuentes A, Walton J, Martínez JA. Exhaustive Search of Dietary Intake Biomarkers as Objective Tools for Personalized Nutrimetabolomics and Precision Nutrition Implementation. Nutr Rev 2024:nuae133. [PMID: 39331531 DOI: 10.1093/nutrit/nuae133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2024] Open
Abstract
OBJECTIVE To conduct an exhaustive scoping search of existing literature, incorporating diverse bibliographic sources to elucidate the relationships between metabolite biomarkers in human fluids and dietary intake. BACKGROUND The search for biomarkers linked to specific dietary food intake holds immense significance for precision health and nutrition research. Using objective methods to track food consumption through metabolites offers a more accurate way to provide dietary advice and prescriptions on healthy dietary patterns by healthcare professionals. An extensive investigation was conducted on biomarkers associated with the consumption of several food groups and consumption patterns. Evidence is integrated from observational studies, systematic reviews, and meta-analyses to achieve precision nutrition and metabolism personalization. METHODS Tailored search strategies were applied across databases and gray literature, yielding 158 primary research articles that met strict inclusion criteria. The collected data underwent rigorous analysis using STATA and Python tools. Biomarker-food associations were categorized into 5 groups: cereals and grains, dairy products, protein-rich foods, plant-based foods, and a miscellaneous group. Specific cutoff points (≥3 or ≥4 bibliographic appearances) were established to identify reliable biomarkers indicative of dietary consumption. RESULTS Key metabolites in plasma, serum, and urine revealed intake from different food groups. For cereals and grains, 3-(3,5-dihydroxyphenyl) propanoic acid glucuronide and 3,5-dihydroxybenzoic acid were significant. Omega-3 fatty acids and specific amino acids showcased dairy and protein foods consumption. Nuts and seafood were linked to hypaphorine and trimethylamine N-oxide. The miscellaneous group featured compounds like theobromine, 7-methylxanthine, caffeine, quinic acid, paraxanthine, and theophylline associated with coffee intake. CONCLUSIONS Data collected from this research demonstrate potential for incorporating precision nutrition into clinical settings and nutritional advice based on accurate estimation of food intake. By customizing dietary recommendations based on individualized metabolic profiles, this approach could significantly improve personalized food consumption health prescriptions and support integrating multiple nutritional data.This article is part of a Nutrition Reviews special collection on Precision Nutrition.
Collapse
Affiliation(s)
- Victor de la O
- Nutrition Precision and Cardiometabolic Health Program of IMDEA-Food Institute (Madrid Institute for Advances Studies), 28040, Madrid, Spain
- Faculty of Health Sciences, International University of La Rioja, 26006, Logroño, Spain
| | - Edwin Fernández-Cruz
- Nutrition Precision and Cardiometabolic Health Program of IMDEA-Food Institute (Madrid Institute for Advances Studies), 28040, Madrid, Spain
- Faculty of Health Sciences, International University of La Rioja, 26006, Logroño, Spain
| | - Alberto Valdés
- Foodomics Lab, Institute of Food Science Research, Spanish National Research Council, 28049, Madrid, Spain
| | - Alejandro Cifuentes
- Foodomics Lab, Institute of Food Science Research, Spanish National Research Council, 28049, Madrid, Spain
| | - Janette Walton
- Department of Biological Sciences, Munster Technological University, Cork, Republic of Ireland
| | - J Alfredo Martínez
- Nutrition Precision and Cardiometabolic Health Program of IMDEA-Food Institute (Madrid Institute for Advances Studies), 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 28049, Madrid, Spain
- Department of Medicine and Endocrinology, Campus of Soria, University of Valladolid, Valladolid, Spain
| |
Collapse
|
5
|
Landberg R, Karra P, Hoobler R, Loftfield E, Huybrechts I, Rattner JI, Noerman S, Claeys L, Neveu V, Vidkjaer NH, Savolainen O, Playdon MC, Scalbert A. Dietary biomarkers-an update on their validity and applicability in epidemiological studies. Nutr Rev 2024; 82:1260-1280. [PMID: 37791499 PMCID: PMC11317775 DOI: 10.1093/nutrit/nuad119] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023] Open
Abstract
The aim of this literature review was to identify and provide a summary update on the validity and applicability of the most promising dietary biomarkers reflecting the intake of important foods in the Western diet for application in epidemiological studies. Many dietary biomarker candidates, reflecting intake of common foods and their specific constituents, have been discovered from intervention and observational studies in humans, but few have been validated. The literature search was targeted for biomarker candidates previously reported to reflect intakes of specific food groups or components that are of major importance in health and disease. Their validity was evaluated according to 8 predefined validation criteria and adapted to epidemiological studies; we summarized the findings and listed the most promising food intake biomarkers based on the evaluation. Biomarker candidates for alcohol, cereals, coffee, dairy, fats and oils, fruits, legumes, meat, seafood, sugar, tea, and vegetables were identified. Top candidates for all categories are specific to certain foods, have defined parent compounds, and their concentrations are unaffected by nonfood determinants. The correlations of candidate dietary biomarkers with habitual food intake were moderate to strong and their reproducibility over time ranged from low to high. For many biomarker candidates, critical information regarding dose response, correlation with habitual food intake, and reproducibility over time is yet unknown. The nutritional epidemiology field will benefit from the development of novel methods to combine single biomarkers to generate biomarker panels in combination with self-reported data. The most promising dietary biomarker candidates that reflect commonly consumed foods and food components for application in epidemiological studies were identified, and research required for their full validation was summarized.
Collapse
Affiliation(s)
- Rikard Landberg
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Prasoona Karra
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Cancer Control and Population Sciences Program, Huntsman Cancer Institute, University of Utah Salt Lake City, UT, USA
| | - Rachel Hoobler
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Cancer Control and Population Sciences Program, Huntsman Cancer Institute, University of Utah Salt Lake City, UT, USA
| | - Erikka Loftfield
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Inge Huybrechts
- International Agency for Research on Cancer, Nutrition and Metabolism Branch, Lyon, France
| | - Jodi I Rattner
- International Agency for Research on Cancer, Nutrition and Metabolism Branch, Lyon, France
| | - Stefania Noerman
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Liesel Claeys
- International Agency for Research on Cancer, Molecular Mechanisms and Biomarkers Group, Lyon, France
| | - Vanessa Neveu
- International Agency for Research on Cancer, Nutrition and Metabolism Branch, Lyon, France
| | - Nanna Hjort Vidkjaer
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Otto Savolainen
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Mary C Playdon
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Cancer Control and Population Sciences Program, Huntsman Cancer Institute, University of Utah Salt Lake City, UT, USA
| | - Augustin Scalbert
- International Agency for Research on Cancer, Nutrition and Metabolism Branch, Lyon, France
| |
Collapse
|
6
|
Ottosson F, Russo F, Abrahamsson A, MacSween N, Courraud J, Skogstrand K, Melander O, Ericson U, Orho-Melander M, Cohen AS, Grove J, Mortensen PB, Hougaard DM, Ernst M. Unraveling the metabolomic architecture of autism in a large Danish population-based cohort. BMC Med 2024; 22:302. [PMID: 39026322 PMCID: PMC11264881 DOI: 10.1186/s12916-024-03516-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 07/02/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND The prevalence of autism in Denmark has been increasing, reaching 1.65% among 10-year-old children, and similar trends are seen elsewhere. Although there are several factors associated with autism, including genetic, environmental, and prenatal factors, the molecular etiology of autism is largely unknown. Here, we use untargeted metabolomics to characterize the neonatal metabolome from dried blood spots collected shortly after birth. METHODS We analyze the metabolomic profiles of a subset of a large Danish population-based cohort (iPSYCH2015) consisting of over 1400 newborns, who later are diagnosed with autism and matching controls and in two Swedish population-based cohorts comprising over 7000 adult participants. Mass spectrometry analysis was performed by a timsTOF Pro operated in QTOF mode, using data-dependent acquisition. By applying an untargeted metabolomics approach, we could reproducibly measure over 800 metabolite features. RESULTS We detected underlying molecular perturbations across several metabolite classes that precede autism. In particular, the cyclic dipeptide cyclo-leucine-proline (FDR-adjusted p = 0.003) and the carnitine-related 5-aminovaleric acid betaine (5-AVAB) (FDR-adjusted p = 0.03), were associated with an increased probability for autism, independently of known prenatal and genetic risk factors. Analysis of genetic and dietary data in adults revealed that 5-AVAB was associated with increased habitual dietary intake of dairy (FDR-adjusted p < 0.05) and with variants near SLC22A4 and SLC22A5 (p < 5.0e - 8), coding for a transmembrane carnitine transporter protein involved in controlling intracellular carnitine levels. CONCLUSIONS Cyclo-leucine-proline and 5-AVAB are associated with future diagnosis of autism in Danish neonates, both representing novel early biomarkers for autism. 5-AVAB is potentially modifiable and may influence carnitine homeostasis.
Collapse
Affiliation(s)
- Filip Ottosson
- Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark.
| | - Francesco Russo
- Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
| | - Anna Abrahamsson
- Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Nadia MacSween
- Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Julie Courraud
- Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
- Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771, Panepistimiopolis, ZografouAthens, Greece
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Alexandra Hospital, 11528, Athens, Greece
| | - Kristin Skogstrand
- Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Ulrika Ericson
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | | - Arieh S Cohen
- Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
- Testcenter Denmark, Statens Serum Institut, Copenhagen, Denmark
| | - Jakob Grove
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
- Department of Biomedicine - Human Genetics, Aarhus University, Aarhus, Denmark
- Bioinformatics Research Center, Aarhus University, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, Aarhus, Denmark
| | - Preben Bo Mortensen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
- NCRR - National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- CIRRAU - Centre for Integrated Registerbased Research at Aarhus University, Aarhus, Denmark
| | - David M Hougaard
- Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
| | - Madeleine Ernst
- Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark.
| |
Collapse
|
7
|
Prentice RL. Intake Biomarkers for Nutrition and Health: Review and Discussion of Methodology Issues. Metabolites 2024; 14:276. [PMID: 38786753 PMCID: PMC11123464 DOI: 10.3390/metabo14050276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/23/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Metabolomics profiles from blood, urine, or other body fluids have the potential to assess intakes of foods and nutrients objectively, thereby strengthening nutritional epidemiology research. Metabolomics platforms may include targeted components that estimate the relative concentrations for individual metabolites in a predetermined set, or global components, typically involving mass spectrometry, that estimate relative concentrations more broadly. While a specific metabolite concentration usually correlates with the intake of a single food or food group, multiple metabolites may be correlated with the intake of certain foods or with specific nutrient intakes, each of which may be expressed in absolute terms or relative to total energy intake. Here, I briefly review the progress over the past 20 years on the development and application intake biomarkers for foods/food groups, nutrients, and dietary patterns, primarily by drawing from several recent reviews. In doing so, I emphasize the criteria and study designs for candidate biomarker identification, biomarker validation, and intake biomarker application. The use of intake biomarkers for diet and chronic disease association studies is still infrequent in nutritional epidemiology research. My comments here will derive primarily from our research group's recent contributions to the Women's Health Initiative cohorts. I will complete the contribution by describing some opportunities to build on the collective 20 years of effort, including opportunities related to the metabolomics profiling of blood and urine specimens from human feeding studies that approximate habitual diets.
Collapse
Affiliation(s)
- Ross L Prentice
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Department of Biostatistics, University of Washington, 1100 Fairview Avenue North, P.O. Box 19024, Seattle, WA 98109-1024, USA
| |
Collapse
|
8
|
Yao S, Colangelo LA, Perry AS, Marron MM, Yaffe K, Sedaghat S, Lima JAC, Tian Q, Clish CB, Newman AB, Shah RV, Murthy VL. Implications of metabolism on multi-systems healthy aging across the lifespan. Aging Cell 2024; 23:e14090. [PMID: 38287525 PMCID: PMC11019145 DOI: 10.1111/acel.14090] [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: 07/24/2023] [Revised: 12/30/2023] [Accepted: 01/11/2024] [Indexed: 01/31/2024] Open
Abstract
Aging is increasingly thought to involve dysregulation of metabolism in multiple organ systems that culminate in decreased functional capacity and morbidity. Here, we seek to understand complex interactions among metabolism, aging, and systems-wide phenotypes across the lifespan. Among 2469 adults (mean age 74.7 years; 38% Black) in the Health, Aging and Body Composition study we identified metabolic cross-sectionally correlates across 20 multi-dimensional aging-related phenotypes spanning seven domains. We used LASSO-PCA and bioinformatic techniques to summarize metabolome-phenome relationships and derive metabolic scores, which were subsequently linked to healthy aging, mortality, and incident outcomes (cardiovascular disease, disability, dementia, and cancer) over 9 years. To clarify the relationship of metabolism in early adulthood to aging, we tested association of these metabolic scores with aging phenotypes/outcomes in 2320 participants (mean age 32.1, 44% Black) of the Coronary Artery Risk Development in Young Adults (CARDIA) study. We observed significant overlap in metabolic correlates across the seven aging domains, specifying pathways of mitochondrial/cellular energetics, host-commensal metabolism, inflammation, and oxidative stress. Across four metabolic scores (body composition, mental-physical performance, muscle strength, and physical activity), we found strong associations with healthy aging and incident outcomes, robust to adjustment for risk factors. Metabolic scores for participants four decades younger in CARDIA were related to incident cardiovascular, metabolic, and neurocognitive performance, as well as long-term cardiovascular disease and mortality over three decades. Conserved metabolic states are strongly related to domain-specific aging and outcomes over the life-course relevant to energetics, host-commensal interactions, and mechanisms of innate immunity.
Collapse
Affiliation(s)
- Shanshan Yao
- University of PittsburgPittsburghPennsylvaniaUSA
| | | | | | | | | | | | | | - Qu Tian
- National Institute of AgingBaltimoreMarylandUSA
| | - Clary B. Clish
- Broad Institute of Harvard and MITCambridgeMassachusettsUSA
| | | | - Ravi V. Shah
- Vanderbilt University Medical CenterNashvilleTennesseeUSA
| | | |
Collapse
|
9
|
Bernard L, Chen J, Kim H, Wong KE, Steffen LM, Yu B, Boerwinkle E, Levey AS, Grams ME, Rhee EP, Rebholz CM. Serum Metabolomic Markers of Protein-Rich Foods and Incident CKD: Results From the Atherosclerosis Risk in Communities Study. Kidney Med 2024; 6:100793. [PMID: 38495599 PMCID: PMC10940775 DOI: 10.1016/j.xkme.2024.100793] [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] [Indexed: 03/19/2024] Open
Abstract
Rationale & Objective While urine excretion of nitrogen estimates the total protein intake, biomarkers of specific dietary protein sources have been sparsely studied. Using untargeted metabolomics, this study aimed to identify serum metabolomic markers of 6 protein-rich foods and to examine whether dietary protein-related metabolites are associated with incident chronic kidney disease (CKD). Study Design Prospective cohort study. Setting & Participants A total of 3,726 participants from the Atherosclerosis Risk in Communities study without CKD at baseline. Exposures Dietary intake of 6 protein-rich foods (fish, nuts, legumes, red and processed meat, eggs, and poultry), serum metabolites. Outcomes Incident CKD (estimated glomerular filtration rate < 60 mL/min/1.73 m2 with ≥25% estimated glomerular filtration rate decline relative to visit 1, hospitalization or death related to CKD, or end-stage kidney disease). Analytical Approach Multivariable linear regression models estimated cross-sectional associations between protein-rich foods and serum metabolites. C statistics assessed the ability of the metabolites to improve the discrimination of highest versus lower 3 quartiles of intake of protein-rich foods beyond covariates (demographics, clinical factors, health behaviors, and the intake of nonprotein food groups). Cox regression models identified prospective associations between protein-related metabolites and incident CKD. Results Thirty significant associations were identified between protein-rich foods and serum metabolites (fish, n = 8; nuts, n = 5; legumes, n = 0; red and processed meat, n = 5; eggs, n = 3; and poultry, n = 9). Metabolites collectively and significantly improved the discrimination of high intake of protein-rich foods compared with covariates alone (difference in C statistics = 0.033, 0.051, 0.003, 0.024, and 0.025 for fish, nuts, red and processed meat, eggs, and poultry-related metabolites, respectively; P < 1.00 × 10-16 for all). Dietary intake of fish was positively associated with 1-docosahexaenoylglycerophosphocholine (22:6n3), which was inversely associated with incident CKD (HR, 0.82; 95% CI, 0.75-0.89; P = 7.81 × 10-6). Limitations Residual confounding and sample-storage duration. Conclusions We identified candidate biomarkers of fish, nuts, red and processed meat, eggs, and poultry. A fish-related metabolite, 1-docosahexaenoylglycerophosphocholine (22:6n3), was associated with a lower risk of CKD.
Collapse
Affiliation(s)
- Lauren Bernard
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Jingsha Chen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Hyunju Kim
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Kari E. Wong
- Metabolon, Research Triangle Park, Morrisville, NC
| | - Lyn M. Steffen
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, TX
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, TX
| | | | - Morgan E. Grams
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Division of Precision of Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Eugene P. Rhee
- Nephrology Division and Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Casey M. Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
10
|
Sato M, Hishinuma E, Matsukawa N, Shima Y, Saigusa D, Motoike IN, Kogure M, Nakaya N, Hozawa A, Kuriyama S, Yamamoto M, Koshiba S, Kinoshita K. Dietary habits and plasma lipid concentrations in a general Japanese population. Metabolomics 2024; 20:34. [PMID: 38441752 PMCID: PMC10914877 DOI: 10.1007/s11306-024-02087-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 01/02/2024] [Indexed: 03/07/2024]
Abstract
INTRODUCTION Accumulating data on the associations between food consumption and lipid composition in the body is essential for understanding the effects of dietary habits on health. OBJECTIVES As part of omics research in the Tohoku Medical Megabank Community-Based Cohort Study, this study sought to reveal the dietary impact on plasma lipid concentration in a Japanese population. METHODS We conducted a correlation analysis of food consumption and plasma lipid concentrations measured using mass spectrometry, for 4032 participants in Miyagi Prefecture, Japan. RESULTS Our analysis revealed 83 marked correlations between six food categories and the concentrations of plasma lipids in nine subclasses. Previously reported associations, including those between seafood consumption and omega-3 fatty acids, were validated, while those between dairy product consumption and odd-carbon-number fatty acids (odd-FAs) were validated for the first time in an Asian population. Further analysis suggested that dairy product consumption is associated with odd-FAs via sphingomyelin (SM), which suggests that SM is a carrier of odd-FAs. These results are important for understanding odd-FA metabolism with regards to dairy product consumption. CONCLUSION This study provides insight into the dietary impact on plasma lipid concentration in a Japanese population.
Collapse
Affiliation(s)
- Mitsuharu Sato
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8573, Japan
| | - Eiji Hishinuma
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8573, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8573, Japan
| | - Naomi Matsukawa
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8573, Japan
| | - Yoshiko Shima
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8573, Japan
| | - Daisuke Saigusa
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8573, Japan
- Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma-Science, Teikyo University, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Ikuko N Motoike
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8573, Japan
- Graduate School of Information Sciences, Tohoku University, 6-3-09 Aramaki Aza-Aoba, Aoba-ku, Sendai, Miyagi, 980-8579, Japan
| | - Mana Kogure
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8573, Japan
| | - Naoki Nakaya
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8573, Japan
| | - Atsushi Hozawa
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Shinichi Kuriyama
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
- International Research Institute of Disaster Science, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8573, Japan
| | - Masayuki Yamamoto
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8573, Japan
| | - Seizo Koshiba
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8573, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8573, Japan
| | - Kengo Kinoshita
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8573, Japan.
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8573, Japan.
- Graduate School of Information Sciences, Tohoku University, 6-3-09 Aramaki Aza-Aoba, Aoba-ku, Sendai, Miyagi, 980-8579, Japan.
| |
Collapse
|
11
|
Playdon MC, Tinker LF, Prentice RL, Loftfield E, Hayden KM, Van Horn L, Sampson JN, Stolzenberg-Solomon R, Lampe JW, Neuhouser ML, Moore SC. Measuring diet by metabolomics: a 14-d controlled feeding study of weighed food intake. Am J Clin Nutr 2024; 119:511-526. [PMID: 38212160 PMCID: PMC10884612 DOI: 10.1016/j.ajcnut.2023.10.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/12/2023] [Accepted: 10/11/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Metabolomics has the potential to enhance dietary assessment by revealing objective measures of many aspects of human food intake. Although metabolomics studies indicate that hundreds of metabolites are associated with dietary intake, correlations have been modest (e.g., r < 0.50), and few have been evaluated in controlled feeding studies. OBJECTIVES The aim of this study was to evaluate associations between metabolites and weighed food and beverage intake in a controlled feeding study of habitual diet. METHODS Healthy postmenopausal females from the Women's Health Initiative (N = 153) were provided with a customized 2-wk controlled diet designed to emulate their usual diet. Metabolites were measured by liquid chromatography tandem mass spectrometry in end-of-study 24-h urine and fasting serum samples (1293 urine metabolites; 1113 serum metabolites). We calculated partial Pearson correlations between these metabolites and intake of 65 food groups, beverages, and supplements during the feeding study. The threshold for significance was Bonferroni-adjusted to account for multiple testing (5.94 × 10-07 for urine metabolites; 6.91 × 10-07 for serum metabolites). RESULTS Significant diet-metabolite correlations were identified for 23 distinct foods, beverages, and supplements (171 distinct metabolites). Among foods, strong metabolite correlations (r ≥ 0.60) were evident for citrus (highest r = 0.80), dairy (r = 0.65), and broccoli (r = 0.63). Among beverages and supplements, strong correlations were evident for coffee (r = 0.86), alcohol (r = 0.69), multivitamins (r = 0.69), and vitamin E supplements (r = 0.65). Moderate correlations (r = 0.50-0.60) were also observed for avocado, fish, garlic, grains, onion, poultry, and black tea. Correlations were specific; each metabolite correlated with one food, beverage, or supplement, except for metabolites correlated with juice or multivitamins. CONCLUSIONS Metabolite levels had moderate to strong correlations with weighed intake of habitually consumed foods, beverages, and supplements. These findings exceed in magnitude those previously observed in population studies and exemplify the strong potential of metabolomics to contribute to nutrition research. The Women's Health Initiative is registered at clinicaltrials.gov as NCT00000611.
Collapse
Affiliation(s)
- Mary C Playdon
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT; Department of Population Health Sciences, University of Utah, Salt Lake City, UT; Cancer Control and Population Sciences Division, Huntsman Cancer Institute, Salt Lake City, UT; Division of Cancer Epidemiology and Genetics, National Cancer institute, Rockville, MD
| | - Lesley F Tinker
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Center and University of Washington, Seattle, WA
| | - Ross L Prentice
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Center and University of Washington, Seattle, WA
| | - Erikka Loftfield
- Division of Cancer Epidemiology and Genetics, National Cancer institute, Rockville, MD
| | - Kathleen M Hayden
- School of Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC
| | - Linda Van Horn
- Feinberg School of Medicine, Northwestern University, Chicago IL
| | - Joshua N Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer institute, Rockville, MD
| | | | - Johanna W Lampe
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Center and University of Washington, Seattle, WA
| | - Marian L Neuhouser
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Center and University of Washington, Seattle, WA
| | - Steven C Moore
- Division of Cancer Epidemiology and Genetics, National Cancer institute, Rockville, MD.
| |
Collapse
|
12
|
Chen S, Lin Z, Shen X, Li L, Pan W. Inference of causal metabolite networks in the presence of invalid instrumental variables with GWAS summary data. Genet Epidemiol 2023; 47:585-599. [PMID: 37573486 PMCID: PMC10840616 DOI: 10.1002/gepi.22535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 06/19/2023] [Accepted: 08/01/2023] [Indexed: 08/14/2023]
Abstract
We propose structural equation models (SEMs) as a general framework to infer causal networks for metabolites and other complex traits. Traditionally SEMs are used only for individual-level data under the assumption that all instrumental variables (IVs) are valid. To overcome these limitations, we propose both one- and two-sample approaches for causal network inference based on SEMs that can: (1) perform causal analysis and discover causal relationships among multiple traits; (2) account for the possible presence of some invalid IVs; (3) allow for data analysis using only genome-wide association studies (GWAS) summary statistics when individual-level data are not available; (4) consider the possibility of bidirectional relationships between traits. Our method employs a simple stepwise selection to identify invalid IVs, thus avoiding false positives while possibly increasing true discoveries based on two-stage least squares (2SLS). We use both real GWAS data and simulated data to demonstrate the superior performance of our method over the standard 2SLS/SEMs. For real data analysis, our proposed approach is applied to a human blood metabolite GWAS summary data set to uncover putative causal relationships among the metabolites; we also identify some metabolites (putative) causal to Alzheimer's disease (AD), which, along with the inferred causal metabolite network, suggest some possible pathways of metabolites involved in AD.
Collapse
Affiliation(s)
- Siyi Chen
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN 55455
| | - Zhaotong Lin
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN 55455
| | - Xiaotong Shen
- School of Statistics, University of Minnesota, Minneapolis, MN 55455
| | - Ling Li
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455
| | - Wei Pan
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
13
|
Lekka P, Fragopoulou E, Terpou A, Dasenaki M. Exploring Human Metabolome after Wine Intake-A Review. Molecules 2023; 28:7616. [PMID: 38005338 PMCID: PMC10673339 DOI: 10.3390/molecules28227616] [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: 10/02/2023] [Revised: 10/21/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
Wine has a rich history dating back to 2200 BC, originally recognized for its medicinal properties. Today, with the aid of advanced technologies like metabolomics and sophisticated analytical techniques, we have gained remarkable insights into the molecular-level changes induced by wine consumption in the human organism. This review embarks on a comprehensive exploration of the alterations in human metabolome associated with wine consumption. A great number of 51 studies from the last 25 years were reviewed; these studies systematically investigated shifts in metabolic profiles within blood, urine, and feces samples, encompassing both short-term and long-term studies of the consumption of wine and wine derivatives. Significant metabolic alterations were observed in a wide variety of metabolites belonging to different compound classes, such as phenolic compounds, lipids, organic acids, and amino acids, among others. Within these classes, both endogenous metabolites as well as diet-related metabolites that exhibited up-regulation or down-regulation following wine consumption were included. The up-regulation of short-chain fatty acids and the down-regulation of sphingomyelins after wine intake, as well as the up-regulation of gut microbial fermentation metabolites like vanillic and syringic acid are some of the most important findings reported in the reviewed literature. Our results confirm the intact passage of certain wine compounds, such as tartaric acid and other wine acids, to the human organism. In an era where the health effects of wine consumption are of growing interest, this review offers a holistic perspective on the metabolic underpinnings of this centuries-old tradition.
Collapse
Affiliation(s)
- Pelagia Lekka
- Food Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zographou, 15771 Athens, Greece;
| | - Elizabeth Fragopoulou
- School of Health Science and Education, Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece;
| | - Antonia Terpou
- Department of Agricultural Development, Agrofood and Management of Natural Resources, School of Agricultural Development, Nutrition & Sustainability, National and Kapodistrian University of Athens, 34400 Psachna, Greece;
| | - Marilena Dasenaki
- Food Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zographou, 15771 Athens, Greece;
| |
Collapse
|
14
|
Bernard L, Chen J, Kim H, Huang Z, Bazzano L, Qi L, He J, Rao VS, Potts KS, Kelly TN, Wong KE, Steffen LM, Yu B, Rhee EP, Rebholz CM. Serum Metabolomic Markers of Dairy Consumption: Results from the Atherosclerosis Risk in Communities Study and the Bogalusa Heart Study. J Nutr 2023; 153:2994-3002. [PMID: 37541543 PMCID: PMC10613758 DOI: 10.1016/j.tjnut.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/14/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023] Open
Abstract
BACKGROUND Dairy consumption is related to chronic disease risk; however, the measurement of dairy consumption has largely relied upon self-report. Untargeted metabolomics allows for the identification of objective markers of dietary intake. OBJECTIVES We aimed to identify associations between dietary dairy intake (total dairy, low-fat dairy, and high-fat dairy) and serum metabolites in 2 independent study populations of United States adults. METHODS Dietary intake was assessed with food frequency questionnaires. Multivariable linear regression models were used to estimate cross-sectional associations between dietary intake of dairy and 360 serum metabolites analyzed in 2 subgroups of the Atherosclerosis Risk in Communities study (ARIC; n = 3776). Results from the 2 subgroups were meta-analyzed using fixed effects meta-analysis. Significant meta-analyzed associations in the ARIC study were then tested in the Bogalusa Heart Study (BHS; n = 785). RESULTS In the ARIC study and BHS, the mean age was 54 and 48 years, 61% and 29% were Black, and the mean dairy intake was 1.7 and 1.3 servings/day, respectively. Twenty-nine significant associations between dietary intake of dairy and serum metabolites were identified in the ARIC study (total dairy, n = 14; low-fat dairy, n = 10; high-fat dairy, n = 5). Three associations were also significant in BHS: myristate (14:0) was associated with high-fat dairy, and pantothenate was associated with total dairy and low-fat dairy, but 23 of the 27 associations significant in the ARIC study and tested in BHS were not associated with dairy in BHS. CONCLUSIONS We identified metabolomic associations with dietary intake of dairy, including 3 associations found in 2 independent cohort studies. These results suggest that myristate (14:0) and pantothenate (vitamin B5) are candidate biomarkers of dairy consumption.
Collapse
Affiliation(s)
- Lauren Bernard
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Jingsha Chen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Hyunju Kim
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Zhijie Huang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Lydia Bazzano
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Lu Qi
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Varun S Rao
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Kaitlin S Potts
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States; Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, United States
| | - Tanika N Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States; Division of Nephrology, Department of Medicine, University of Illinois Chicago, Chicago, IL, United States
| | - Kari E Wong
- Metabolon, Research Triangle Park, Morrisville, NC, United States
| | - Lyn M Steffen
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, United States
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston School of Public Health, Houston, TX, United States
| | - Eugene P Rhee
- Division of Nephrology and Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Casey M Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States; Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
15
|
O'Connor LE, Hall KD, Herrick KA, Reedy J, Chung ST, Stagliano M, Courville AB, Sinha R, Freedman ND, Hong HG, Albert PS, Loftfield E. Metabolomic Profiling of an Ultraprocessed Dietary Pattern in a Domiciled Randomized Controlled Crossover Feeding Trial. J Nutr 2023; 153:2181-2192. [PMID: 37276937 PMCID: PMC10447619 DOI: 10.1016/j.tjnut.2023.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 06/07/2023] Open
Abstract
BACKGROUND Objective markers of ultraprocessed foods (UPF) may improve the assessment of UPF intake and provide insight into how UPF influences health. OBJECTIVES To identify metabolites that differed between dietary patterns (DPs) high in or void of UPF according to Nova classification. METHODS In a randomized, crossover, controlled-feeding trial (clinicaltrials.govNCT03407053), 20 domiciled healthy participants (mean ± standard deviation: age 31 ± 7 y, body mass index [kg/m2] 22 ± 11.6) consumed ad libitum a UPF-DP (80% UPF) and an unprocessed DP (UN-DP; 0% UPF) for 2 wk each. Metabolites were measured using liquid chromatography with tandem mass spectrometry in ethylenediaminetetraacetic acid plasma, collected at week 2 and 24-h, and spot urine, collected at weeks 1 and 2, of each DP. Linear mixed models, adjusted for energy intake, were used to identify metabolites that differed between DPs. RESULTS After multiple comparisons correction, 257 out of 993 plasma and 606 out of 1279 24-h urine metabolites differed between UPF-DP and UN-DP. Overall, 21 known and 9 unknown metabolites differed between DPs across all time points and biospecimen types. Six metabolites were higher (4-hydroxy-L-glutamic acid, N-acetylaminooctanoic acid, 2-methoxyhydroquinone sulfate, 4-ethylphenylsulfate, 4-vinylphenol sulfate, and acesulfame) and 14 were lower following the UPF-DP; pimelic acid, was lower in plasma but higher in urine following the UPF-DP. CONCLUSIONS Consuming a DP high in, compared with 1 void of, UPF has a measurable impact on the short-term human metabolome. Observed differential metabolites could serve as candidate biomarkers of UPF intake or metabolic response in larger samples with varying UPF-DPs. This trial was registered at clinicaltrials.gov as NCT03407053 and NCT03878108.
Collapse
Affiliation(s)
- Lauren E O'Connor
- Food Components and Health Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, USDA, Beltsville, MD, USA; Division of Cancer Control and Population Sciences, Risk Factor Assessment Branch, NCI, Bethesda, MD, USA
| | - Kevin D Hall
- Laboratory of Biological Modeling, NIDDK, Bethesda, MD, USA
| | - Kirsten A Herrick
- Division of Cancer Control and Population Sciences, Risk Factor Assessment Branch, NCI, Bethesda, MD, USA
| | - Jill Reedy
- Division of Cancer Control and Population Sciences, Risk Factor Assessment Branch, NCI, Bethesda, MD, USA
| | - Stephanie T Chung
- Diabetes, Endocrinology, and Obesity Branch, NIDDK, Bethesda, MD, USA
| | - Michael Stagliano
- Diabetes, Endocrinology, and Obesity Branch, NIDDK, Bethesda, MD, USA
| | - Amber B Courville
- Diabetes, Endocrinology, and Obesity Branch, NIDDK, Bethesda, MD, USA
| | - Rashmi Sinha
- Division of Cancer Epidemiology and Genetics, Metabolic Epidemiology Branch, NCI, Bethesda, MD, USA
| | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics, Metabolic Epidemiology Branch, NCI, Bethesda, MD, USA
| | - Hyokyoung G Hong
- Division of Cancer Epidemiology and Genetics, Biostatistics Branch, NCI, Bethesda, MD, USA
| | - Paul S Albert
- Division of Cancer Epidemiology and Genetics, Biostatistics Branch, NCI, Bethesda, MD, USA
| | - Erikka Loftfield
- Division of Cancer Epidemiology and Genetics, Metabolic Epidemiology Branch, NCI, Bethesda, MD, USA.
| |
Collapse
|
16
|
Vahid F, Hajizadeghan K, Khodabakhshi A. Nutritional Metabolomics in Diet-Breast Cancer Relations: Current Research, Challenges, and Future Directions-A Review. Biomedicines 2023; 11:1845. [PMID: 37509485 PMCID: PMC10377267 DOI: 10.3390/biomedicines11071845] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/21/2023] [Accepted: 06/24/2023] [Indexed: 07/30/2023] Open
Abstract
Breast cancer is one of the most common types of cancer in women worldwide, and its incidence is increasing. Diet has been identified as a modifiable risk factor for breast cancer, but the complex interplay between diet, metabolism, and cancer development is not fully understood. Nutritional metabolomics is a rapidly evolving field that can provide insights into the metabolic changes associated with dietary factors and their impact on breast cancer risk. The review's objective is to provide a comprehensive overview of the current research on the application of nutritional metabolomics in understanding the relationship between diet and breast cancer. The search strategy involved querying several electronic databases, including PubMed, Scopus, Web of Science, and Google Scholar. The search terms included combinations of relevant keywords such as "nutritional metabolomics", "diet", "breast cancer", "metabolites", and "biomarkers". In this review, both in vivo and in vitro studies were included, and we summarize the current state of knowledge on the role of nutritional metabolomics in understanding the diet-breast cancer relationship, including identifying specific metabolites and metabolic pathways associated with breast cancer risk. We also discuss the challenges associated with nutritional metabolomics research, including standardization of analytical methods, interpretation of complex data, and integration of multiple-omics approaches. Finally, we highlight future directions for nutritional metabolomics research in studying diet-breast cancer relations, including investigating the role of gut microbiota and integrating multiple-omics approaches. The application of nutritional metabolomics in the study of diet-breast cancer relations, including 2-amino-4-cyano butanoic acid, piperine, caprate, rosten-3β,17β-diol-monosulfate, and γ-carboxyethyl hydrochroman, among others, holds great promise for advancing our understanding of the role of diet in breast cancer development and identifying personalized dietary recommendations for breast cancer prevention, control, and treatment.
Collapse
Affiliation(s)
- Farhad Vahid
- Nutrition and Health Research Group, Precision Health Department, Luxembourg Institute of Health, 1445 Strassen, Luxembourg
| | - Kimia Hajizadeghan
- Department of Nutrition, Faculty of Public Health, Kerman University of Medical Sciences, Kerman 7616913555, Iran
| | - Adeleh Khodabakhshi
- Department of Nutrition, Faculty of Public Health, Kerman University of Medical Sciences, Kerman 7616913555, Iran
| |
Collapse
|
17
|
Trius-Soler M, Praticò G, Gürdeniz G, Garcia-Aloy M, Canali R, Fausta N, Brouwer-Brolsma EM, Andrés-Lacueva C, Dragsted LO. Biomarkers of moderate alcohol intake and alcoholic beverages: a systematic literature review. GENES & NUTRITION 2023; 18:7. [PMID: 37076809 PMCID: PMC10114415 DOI: 10.1186/s12263-023-00726-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 04/04/2023] [Indexed: 04/21/2023]
Abstract
The predominant source of alcohol in the diet is alcoholic beverages, including beer, wine, spirits and liquors, sweet wine, and ciders. Self-reported alcohol intakes are likely to be influenced by measurement error, thus affecting the accuracy and precision of currently established epidemiological associations between alcohol itself, alcoholic beverage consumption, and health or disease. Therefore, a more objective assessment of alcohol intake would be very valuable, which may be established through biomarkers of food intake (BFIs). Several direct and indirect alcohol intake biomarkers have been proposed in forensic and clinical contexts to assess recent or longer-term intakes. Protocols for performing systematic reviews in this field, as well as for assessing the validity of candidate BFIs, have been developed within the Food Biomarker Alliance (FoodBAll) project. The aim of this systematic review is to list and validate biomarkers of ethanol intake per se excluding markers of abuse, but including biomarkers related to common categories of alcoholic beverages. Validation of the proposed candidate biomarker(s) for alcohol itself and for each alcoholic beverage was done according to the published guideline for biomarker reviews. In conclusion, common biomarkers of alcohol intake, e.g., as ethyl glucuronide, ethyl sulfate, fatty acid ethyl esters, and phosphatidyl ethanol, show considerable inter-individual response, especially at low to moderate intakes, and need further development and improved validation, while BFIs for beer and wine are highly promising and may help in more accurate intake assessments for these specific beverages.
Collapse
Affiliation(s)
- Marta Trius-Soler
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 1958, Frederiksberg C, Denmark
- Polyphenol Research Laboratory, Department of Nutrition, Food Sciences and Gastronomy, XIA School of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, 08921, Santa Coloma de Gramanet, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Giulia Praticò
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 1958, Frederiksberg C, Denmark
| | - Gözde Gürdeniz
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 1958, Frederiksberg C, Denmark
| | - Mar Garcia-Aloy
- Biomarker & Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain
- Metabolomics Unit, Research and Innovation Centre, Fondazione Edmund Mach, San Michele All'Adige, Italy
| | - Raffaella Canali
- Consiglio Per La Ricerca in Agricoltura E L'analisi Dell'economia Agraria (CREA) Research Centre for Food and Nutrition, Rome, Italy
| | - Natella Fausta
- Consiglio Per La Ricerca in Agricoltura E L'analisi Dell'economia Agraria (CREA) Research Centre for Food and Nutrition, Rome, Italy
| | - Elske M Brouwer-Brolsma
- Division of Human Nutrition and Health, Department Agrotechnology and Food Sciences, Wageningen University and Research, P.O. Box 17, 6700 AA, Wageningen, The Netherlands
| | - Cristina Andrés-Lacueva
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, 08921, Santa Coloma de Gramanet, Spain
- Biomarker & Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Lars Ove Dragsted
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 1958, Frederiksberg C, Denmark.
| |
Collapse
|
18
|
Bernard L, Chen J, Kim H, Wong KE, Steffen LM, Yu B, Boerwinkle E, Rebholz CM. Metabolomics of Dietary Intake of Total, Animal, and Plant Protein: Results from the Atherosclerosis Risk in Communities (ARIC) Study. Curr Dev Nutr 2023; 7:100067. [PMID: 37304852 PMCID: PMC10257224 DOI: 10.1016/j.cdnut.2023.100067] [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: 01/26/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 06/13/2023] Open
Abstract
Background Dietary consumption has traditionally been studied through food intake questionnaires. Metabolomics can be used to identify blood markers of dietary protein that may complement existing dietary assessment tools. Objectives We aimed to identify associations between 3 dietary protein sources (total protein, animal protein, and plant protein) and serum metabolites using data from the Atherosclerosis Risk in Communities Study. Methods Participants' dietary protein intake was derived from a food frequency questionnaire administered by an interviewer, and fasting serum samples were collected at study visit 1 (1987-1989). Untargeted metabolomic profiling was performed in 2 subgroups (subgroup 1: n = 1842; subgroup 2: n = 2072). Multivariable linear regression models were used to assess associations between 3 dietary protein sources and 360 metabolites, adjusting for demographic factors and other participant characteristics. Analyses were performed separately within each subgroup and meta-analyzed with fixed-effects models. Results In this study of 3914 middle-aged adults, the mean (SD) age was 54 (6) y, 60% were women, and 61% were Black. We identified 41 metabolites significantly associated with dietary protein intake. Twenty-six metabolite associations overlapped between total protein and animal protein, such as pyroglutamine, creatine, 3-methylhistidine, and 3-carboxy-4-methyl-5-propyl-2-furanpropanoic acid. Plant protein was uniquely associated with 11 metabolites, such as tryptophan betaine, 4-vinylphenol sulfate, N-δ-acetylornithine, and pipecolate. Conclusions The results of 17 of the 41 metabolites (41%) were consistent with those of previous nutritional metabolomic studies and specific protein-rich food items. We discovered 24 metabolites that had not been previously associated with dietary protein intake. These results enhance the validity of candidate markers of dietary protein intake and introduce novel metabolomic markers of dietary protein intake.
Collapse
Affiliation(s)
- Lauren Bernard
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jingsha Chen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Hyunju Kim
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kari E. Wong
- Metabolon, Research Triangle Park, Morrisville, NC, USA
| | - Lyn M. Steffen
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
- Human Genome Sequencing Center, Baylor Colleague of Medicine, Houston, TX, USA
| | - Casey M. Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
19
|
Li K, Burton-Pimentel KJ, Brouwer-Brolsma EM, Blaser C, Badertscher R, Pimentel G, Portmann R, Feskens EJM, Vergères G. Identifying Plasma and Urinary Biomarkers of Fermented Food Intake and Their Associations with Cardiometabolic Health in a Dutch Observational Cohort. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:4426-4439. [PMID: 36853956 PMCID: PMC10021015 DOI: 10.1021/acs.jafc.2c05669] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 02/12/2023] [Accepted: 02/13/2023] [Indexed: 06/18/2023]
Abstract
Identification of food intake biomarkers (FIBs) for fermented foods could help improve their dietary assessment and clarify their associations with cardiometabolic health. We aimed to identify novel FIBs for fermented foods in the plasma and urine metabolomes of 246 free-living Dutch adults using nontargeted LC-MS and GC-MS. Furthermore, associations between identified metabolites and several cardiometabolic risk factors were explored. In total, 37 metabolites were identified corresponding to the intakes of coffee, wine, and beer (none were identified for cocoa, bread, cheese, or yoghurt intake). While some of these metabolites appeared to originate from raw food (e.g., niacin and trigonelline for coffee), others overlapped different fermented foods (e.g., 4-hydroxybenzeneacetic acid for both wine and beer). In addition, several fermentation-dependent metabolites were identified (erythritol and citramalate). Associations between these identified metabolites with cardiometabolic parameters were weak and inconclusive. Further evaluation is warranted to confirm their relationships with cardiometabolic disease risk.
Collapse
Affiliation(s)
- Katherine
J. Li
- Division
of Human Nutrition and Health, Department of Agrotechnology and Food
Science, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
- Agroscope, Schwarzenburgstrasse 161, CH-3003 Bern, Switzerland
| | | | - Elske M. Brouwer-Brolsma
- Division
of Human Nutrition and Health, Department of Agrotechnology and Food
Science, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
| | - Carola Blaser
- Agroscope, Schwarzenburgstrasse 161, CH-3003 Bern, Switzerland
| | | | | | - Reto Portmann
- Agroscope, Schwarzenburgstrasse 161, CH-3003 Bern, Switzerland
| | - Edith J. M. Feskens
- Division
of Human Nutrition and Health, Department of Agrotechnology and Food
Science, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
| | - Guy Vergères
- Agroscope, Schwarzenburgstrasse 161, CH-3003 Bern, Switzerland
| |
Collapse
|
20
|
Shah RV, Steffen LM, Nayor M, Reis JP, Jacobs DR, Allen NB, Lloyd-Jones D, Meyer K, Cole J, Piaggi P, Vasan RS, Clish CB, Murthy VL. Dietary metabolic signatures and cardiometabolic risk. Eur Heart J 2023; 44:557-569. [PMID: 36424694 PMCID: PMC10169425 DOI: 10.1093/eurheartj/ehac446] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 06/23/2022] [Accepted: 07/28/2022] [Indexed: 11/27/2022] Open
Abstract
AIMS Observational studies of diet in cardiometabolic-cardiovascular disease (CM-CVD) focus on self-reported consumption of food or dietary pattern, with limited information on individual metabolic responses to dietary intake linked to CM-CVD. Here, machine learning approaches were used to identify individual metabolic patterns related to diet and relation to long-term CM-CVD in early adulthood. METHODS AND RESULTS In 2259 White and Black adults (age 32.1 ± 3.6 years, 45% women, 44% Black) in the Coronary Artery Risk Development in Young Adults (CARDIA) study, multivariate models were employed to identify metabolite signatures of food group and composite dietary intake across 17 food groups, 2 nutrient groups, and healthy eating index-2015 (HEI2015) diet quality score. A broad array of metabolites associated with diet were uncovered, reflecting food-related components/catabolites (e.g. fish and long-chain unsaturated triacylglycerols), interactions with host features (microbiome), or pathways broadly implicated in CM-CVD (e.g. ceramide/sphingomyelin lipid metabolism). To integrate diet with metabolism, penalized machine learning models were used to define a metabolite signature linked to a putative CM-CVD-adverse diet (e.g. high in red/processed meat, refined grains), which was subsequently associated with long-term diabetes and CVD risk numerically more strongly than HEI2015 in CARDIA [e.g. diabetes: standardized hazard ratio (HR): 1.62, 95% confidence interval (CI): 1.32-1.97, P < 0.0001; CVD: HR: 1.55, 95% CI: 1.12-2.14, P = 0.008], with associations replicated for diabetes (P < 0.0001) in the Framingham Heart Study. CONCLUSION Metabolic signatures of diet are associated with long-term CM-CVD independent of lifestyle and traditional risk factors. Metabolomics improves precision to identify adverse consequences and pathways of diet-related CM-CVD.
Collapse
Affiliation(s)
- Ravi V Shah
- Vanderbilt University Medical Center, Vanderbilt Clinical and Translational Research Center (VTRACC), Nashville, TN, USA
| | - Lyn M Steffen
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Matthew Nayor
- Cardiology Division, Boston University School of Medicine, Boston, MA, USA
| | - Jared P Reis
- Epidemiology Branch, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - David R Jacobs
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Norrina B Allen
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
| | - Donald Lloyd-Jones
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
| | - Katie Meyer
- Nutrition Department, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Joanne Cole
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Paolo Piaggi
- Department of Information Engineering, University of Pisa, Pisa, Italy
| | - Ramachandran S Vasan
- Sections of Preventive Medicine and Epidemiology and Cardiovascular Medicine, Department of Medicine, and Department of Epidemiology, Boston University Schools of Medicine and Public Health, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Clary B Clish
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Venkatesh L Murthy
- Department of Medicine and Radiology, University of Michigan, 1338 Cardiovascular Center, Ann Arbor, MI 48109-5873, USA
| |
Collapse
|
21
|
Chemotherapy-Induced Peripheral Neuropathy. Handb Exp Pharmacol 2023; 277:299-337. [PMID: 36253554 DOI: 10.1007/164_2022_609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a debilitating side effect of many common anti-cancer agents that can lead to dose reduction or treatment discontinuation, which decrease chemotherapy efficacy. Long-term CIPN can interfere with activities of daily living and diminish the quality of life. The mechanism of CIPN is not yet fully understood, and biomarkers are needed to identify patients at high risk and potential treatment targets. Metabolomics can capture the complex behavioral and pathophysiological processes involved in CIPN. This chapter is to review the CIPN metabolomics studies to find metabolic pathways potentially involved in CIPN. These potential CIPN metabolites are then investigated to determine whether there is evidence from studies of other neuropathy etiologies such as diabetic neuropathy and Leber hereditary optic neuropathy to support the importance of these pathways in peripheral neuropathy. Six potential biomarkers and their putative mechanisms in peripheral neuropathy were reviewed. Among these biomarkers, histidine and phenylalanine have clear roles in neurotransmission or neuroinflammation in peripheral neuropathy. Further research is needed to discover and validate CIPN metabolomics biomarkers in large clinical studies.
Collapse
|
22
|
Hasken JM, de Vries MM, Marais AS, May PA, Parry CDH, Seedat S, Mooney SM, Smith SM. Untargeted Metabolome Analysis of Alcohol-Exposed Pregnancies Reveals Metabolite Differences That Are Associated with Infant Birth Outcomes. Nutrients 2022; 14:nu14245367. [PMID: 36558526 PMCID: PMC9786146 DOI: 10.3390/nu14245367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/06/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Prenatal alcohol exposure can produce offspring growth deficits and is a leading cause of neurodevelopmental disability. We used untargeted metabolomics to generate mechanistic insight into how alcohol impairs fetal development. In the Western Cape Province of South Africa, 52 women between gestational weeks 5-36 (mean 18.5 ± 6.5) were recruited, and they provided a finger-prick fasting bloodspot that underwent mass spectrometry. Metabolomic data were analyzed using partial least squares-discriminant analyses (PLS-DA) to identify metabolites that correlated with alcohol exposure and infant birth outcomes. Women who consumed alcohol in the past seven days were distinguished by a metabolite profile that included reduced sphingomyelins, cholesterol, and pregnenolones, and elevated fatty acids, acyl and amino acyl carnitines, and androsterones. Using PLS-DA, 25 of the top 30 metabolites differentiating maternal groups were reduced by alcohol with medium-chain free fatty acids and oxidized sugar derivatives having the greatest influence. A separate ortho-PLS-DA analysis identified a common set of 13 metabolites that were associated with infant length, weight, and head circumference. These included monoacylglycerols, glycerol-3-phosphate, and unidentified metabolites, and most of their associations were negative, implying they represent processes having adverse consequences for fetal development.
Collapse
Affiliation(s)
- Julie M. Hasken
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
- Correspondence: ; Tel.: +1-(704)-250-5002
| | - Marlene M. de Vries
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7602, South Africa
| | - Anna-Susan Marais
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7602, South Africa
| | - Philip A. May
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7602, South Africa
- Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
- Center on Alcohol, Substance Abuse, and Addictions, University of New Mexico, Albuquerque, NM 87131, USA
| | - Charles D. H. Parry
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7602, South Africa
- Alcohol, Tobacco, and Other Drug Research Unit, South African Medical Research Council, Cape Town 7760, South Africa
| | - Soraya Seedat
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7602, South Africa
| | - Sandra M. Mooney
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
- Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| | - Susan M. Smith
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
- Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| |
Collapse
|
23
|
García‐Gavilán J, Nishi SK, Paz‐Graniel I, Guasch‐Ferré M, Razquin C, Clish CB, Toledo E, Ruiz‐Canela M, Corella D, Deik A, Drouin‐Chartier J, Wittenbecher C, Babio N, Estruch R, Ros E, Fitó M, Arós F, Fiol M, Serra‐Majem L, Liang L, Martínez‐González MA, Hu FB, Salas‐Salvadó J. Plasma Metabolite Profiles Associated with the Amount and Source of Meat and Fish Consumption and the Risk of Type 2 Diabetes. Mol Nutr Food Res 2022; 66:e2200145. [PMID: 36214069 PMCID: PMC9722604 DOI: 10.1002/mnfr.202200145] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 09/12/2022] [Indexed: 01/18/2023]
Abstract
SCOPE Consumption of meat has been associated with a higher risk of type 2 diabetes (T2D), but if plasma metabolite profiles associated with these foods reflect this relationship is unknown. The objective is to identify a metabolite signature of consumption of total meat (TM), red meat (RM), processed red meat (PRM), and fish and examine if they are associated with T2D risk. METHODS AND RESULTS The discovery population includes 1833 participants from the PREDIMED trial. The internal validation sample includes 1522 participants with available 1-year follow-up metabolomic data. Associations between metabolites and TM, RM, PRM, and fish are evaluated with elastic net regression. Associations between the profiles and incident T2D are estimated using Cox regressions. The profiles included 72 metabolites for TM, 69 for RM, 74 for PRM, and 66 for fish. After adjusting for T2D risk factors, only profiles of TM (Hazard Ratio (HR): 1.25, 95% CI: 1.06-1.49), RM (HR: 1.27, 95% CI: 1.07-1.52), and PRM (HR: 1.27, 95% CI: 1.07-1.51) are associated with T2D. CONCLUSIONS The consumption of TM, its subtypes, and fish is associated with different metabolites, some of which have been previously associated with T2D. Scores based on the identified metabolites for TM, RM, and PRM show a significant association with T2D risk.
Collapse
Affiliation(s)
- Jesús García‐Gavilán
- Departament de Bioquímica i BiotecnologiaUnitat de Nutrició Humana, Hospital Universitari San Joan de ReusUniversitat Rovira i VirgiliReus43202Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV)Reus43204Spain
- Consorcio CIBER, Fisiopatología de la Obesidad y Nutrición (CIBERObn)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| | - Stephanie K. Nishi
- Departament de Bioquímica i BiotecnologiaUnitat de Nutrició Humana, Hospital Universitari San Joan de ReusUniversitat Rovira i VirgiliReus43202Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV)Reus43204Spain
- Consorcio CIBER, Fisiopatología de la Obesidad y Nutrición (CIBERObn)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
- Toronto 3D (Diet, Digestive Tract and Disease) Knowledge Synthesis and Clinical Trials UnitTorontoONM5C 2T2Canada
- Clinical Nutrition and Risk Factor Modification CentreSt. Michael's Hospital, Unity Health TorontoTorontoONM5C 2T2Canada
| | - Indira Paz‐Graniel
- Departament de Bioquímica i BiotecnologiaUnitat de Nutrició Humana, Hospital Universitari San Joan de ReusUniversitat Rovira i VirgiliReus43202Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV)Reus43204Spain
- Consorcio CIBER, Fisiopatología de la Obesidad y Nutrición (CIBERObn)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| | - Marta Guasch‐Ferré
- Department of NutritionHarvard TH Chan School of Public HealthBostonMA02115USA
- Channing Division for Network Medicine, Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Cristina Razquin
- Consorcio CIBER, Fisiopatología de la Obesidad y Nutrición (CIBERObn)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
- Department of Preventive Medicine and Public Health, Navarra Health Research Institute (IDISNA)University of NavarraPamplona31008Spain
| | | | - Estefanía Toledo
- Consorcio CIBER, Fisiopatología de la Obesidad y Nutrición (CIBERObn)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
- Department of Preventive Medicine and Public Health, Navarra Health Research Institute (IDISNA)University of NavarraPamplona31008Spain
| | - Miguel Ruiz‐Canela
- Consorcio CIBER, Fisiopatología de la Obesidad y Nutrición (CIBERObn)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
- Department of Preventive Medicine and Public Health, Navarra Health Research Institute (IDISNA)University of NavarraPamplona31008Spain
| | - Dolores Corella
- Consorcio CIBER, Fisiopatología de la Obesidad y Nutrición (CIBERObn)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
- Department of Preventive MedicineUniversity of ValenciaValencia46020Spain
| | - Amy Deik
- The Broad Institute of Harvard and MITBostonMA02142USA
| | - Jean‐Philippe Drouin‐Chartier
- Centre Nutrition, Santé et Société, Institut sur la Nutrition et les Aliments FonctionnelsFaculté de Pharmacie, Université LavalQuébecG1V 0A6Canada
| | - Clemens Wittenbecher
- Department of NutritionHarvard TH Chan School of Public HealthBostonMA02115USA
- Department of Molecular EpidemiologyGerman Institute of Human Nutrition Potsdam‐Rehbruecke14558NuthetalGermany
- German Center for Diabetes Research85764NeuherbergGermany
| | - Nancy Babio
- Departament de Bioquímica i BiotecnologiaUnitat de Nutrició Humana, Hospital Universitari San Joan de ReusUniversitat Rovira i VirgiliReus43202Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV)Reus43204Spain
- Consorcio CIBER, Fisiopatología de la Obesidad y Nutrición (CIBERObn)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| | - Ramon Estruch
- Consorcio CIBER, Fisiopatología de la Obesidad y Nutrición (CIBERObn)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
- Department of Internal Medicine, Institut d'Investigacions Biomèdiques August Pi SunyerHospital ClinicUniversity of BarcelonaBarcelona08036Spain
| | - Emilio Ros
- Consorcio CIBER, Fisiopatología de la Obesidad y Nutrición (CIBERObn)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
- Lipid Clinic, Department of Endocrinology and Nutrition, Agust Pi i Sunyer Biomedical Research Institute (IDIBAPS)Hospital Clinic, University of BarcelonaBarcelona08036Spain
| | - Montserrat Fitó
- Consorcio CIBER, Fisiopatología de la Obesidad y Nutrición (CIBERObn)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
- Cardiovascular and Nutrition Research GroupInstitut de Recerca Hospital del MarBarcelona08003Spain
| | - Fernando Arós
- Consorcio CIBER, Fisiopatología de la Obesidad y Nutrición (CIBERObn)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
- Department of CardiologyUniversity Hospital of AlavaVitoria01009Spain
| | - Miquel Fiol
- Consorcio CIBER, Fisiopatología de la Obesidad y Nutrición (CIBERObn)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
- Health Research Institute of the Balearic Islands (Idisba)University of Balearic Islands and Hospital Son EspasesPalma de Mallorca07122Spain
| | - Lluís Serra‐Majem
- Consorcio CIBER, Fisiopatología de la Obesidad y Nutrición (CIBERObn)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
- Research Institute of Biomedical and Health Sciences IUIBSUniversity of Las Palmas de Gran CanariaLas Palmas35001Spain
| | - Liming Liang
- Department of EpidemiologyHarvard T. H. Chan School of Public HealthBostonMA02115USA
- Department of StatisticsHarvard T. H. Chan School of Public HealthBostonMA02115USA
| | - Miguel A. Martínez‐González
- Consorcio CIBER, Fisiopatología de la Obesidad y Nutrición (CIBERObn)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
- Department of NutritionHarvard TH Chan School of Public HealthBostonMA02115USA
- Department of Preventive Medicine and Public Health, Navarra Health Research Institute (IDISNA)University of NavarraPamplona31008Spain
| | - Frank B. Hu
- Department of NutritionHarvard TH Chan School of Public HealthBostonMA02115USA
- Channing Division for Network Medicine, Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
- Department of EpidemiologyHarvard T. H. Chan School of Public HealthBostonMA02115USA
| | - Jordi Salas‐Salvadó
- Departament de Bioquímica i BiotecnologiaUnitat de Nutrició Humana, Hospital Universitari San Joan de ReusUniversitat Rovira i VirgiliReus43202Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV)Reus43204Spain
- Consorcio CIBER, Fisiopatología de la Obesidad y Nutrición (CIBERObn)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| |
Collapse
|
24
|
Louca P, Tran TQB, Toit CD, Christofidou P, Spector TD, Mangino M, Suhre K, Padmanabhan S, Menni C. Machine learning integration of multimodal data identifies key features of blood pressure regulation. EBioMedicine 2022; 84:104243. [PMID: 36084617 PMCID: PMC9463529 DOI: 10.1016/j.ebiom.2022.104243] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/02/2022] [Accepted: 08/11/2022] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Association studies have identified several biomarkers for blood pressure and hypertension, but a thorough understanding of their mutual dependencies is lacking. By integrating two different high-throughput datasets, biochemical and dietary data, we aim to understand the multifactorial contributors of blood pressure (BP). METHODS We included 4,863 participants from TwinsUK with concurrent BP, metabolomics, genomics, biochemical measures, and dietary data. We used 5-fold cross-validation with the machine learning XGBoost algorithm to identify features of importance in context of one another in TwinsUK (80% training, 20% test). The features tested in TwinsUK were then probed using the same algorithm in an independent dataset of 2,807 individuals from the Qatari Biobank (QBB). FINDINGS Our model explained 39·2% [4·5%, MAE:11·32 mmHg (95%CI, +/- 0·65)] of the variance in systolic BP (SBP) in TwinsUK. Of the top 50 features, the most influential non-demographic variables were dihomo-linolenate, cis-4-decenoyl carnitine, lactate, chloride, urate, and creatinine along with dietary intakes of total, trans and saturated fat. We also highlight the incremental value of each included dimension. Furthermore, we replicated our model in the QBB [SBP variance explained = 45·2% (13·39%)] cohort and 30 of the top 50 features overlapped between cohorts. INTERPRETATION We show that an integrated analysis of omics, biochemical and dietary data improves our understanding of their in-between relationships and expands the range of potential biomarkers for blood pressure. Our results point to potentially key biological pathways to be prioritised for mechanistic studies. FUNDING Chronic Disease Research Foundation, Medical Research Council, Wellcome Trust, Qatar Foundation.
Collapse
Affiliation(s)
- Panayiotis Louca
- Department of Twin Research and Genetic Epidemiology, King's College London, London, England, SE1 7EH, United Kingdom
| | - Tran Quoc Bao Tran
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Clea du Toit
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Paraskevi Christofidou
- Department of Twin Research and Genetic Epidemiology, King's College London, London, England, SE1 7EH, United Kingdom
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, England, SE1 7EH, United Kingdom
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London, England, SE1 7EH, United Kingdom; NIHR Biomedical Research Centre at Guy's and St Thomas' Foundation Trust, London, SE1 9RT, United Kingdom
| | - Karsten Suhre
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Doha, Qatar; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Sandosh Padmanabhan
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom.
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King's College London, London, England, SE1 7EH, United Kingdom.
| |
Collapse
|
25
|
Identification of Single and Combined Serum Metabolites Associated with Food Intake. Metabolites 2022; 12:metabo12100908. [PMID: 36295810 PMCID: PMC9607433 DOI: 10.3390/metabo12100908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/16/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
Assessment of dietary intake is challenging. Traditional methods suffer from both random and systematic errors; thus objective measures are important complements in monitoring dietary exposure. The study presented here aims to identify serum metabolites associated with reported food intake and to explore whether combinations of metabolites may improve predictive models. Fasting blood samples and a 4-day weighed food diary were collected from healthy Swedish subjects (n = 119) self-defined as having habitual vegan, vegetarian, vegetarian + fish, or omnivore diets. Serum was analyzed for metabolites by 1H-nuclear magnetic resonance spectroscopy. Associations between single and combined metabolites and 39 foods and food groups were explored. Area under the curve (AUC) was calculated for prediction models. In total, 24 foods or food groups associated with serum metabolites using the criteria of rho > 0.2, p < 0.01 and AUC ≥ 0.7 were identified. For the consumption of soybeans, citrus fruits and marmalade, nuts and almonds, green tea, red meat, poultry, total fish and shellfish, dairy, fermented dairy, cheese, eggs, and beer the final models included two or more metabolites. Our results indicate that a combination of metabolites improve the possibilities to use metabolites to identify several foods included in the current diet. Combined metabolite models should be confirmed in dose−response intervention studies.
Collapse
|
26
|
Weinisch P, Fiamoncini J, Schranner D, Raffler J, Skurk T, Rist MJ, Römisch-Margl W, Prehn C, Adamski J, Hauner H, Daniel H, Suhre K, Kastenmüller G. Dynamic patterns of postprandial metabolic responses to three dietary challenges. Front Nutr 2022; 9:933526. [PMID: 36211489 PMCID: PMC9540193 DOI: 10.3389/fnut.2022.933526] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Food intake triggers extensive changes in the blood metabolome. The kinetics of these changes depend on meal composition and on intrinsic, health-related characteristics of each individual, making the assessment of changes in the postprandial metabolome an opportunity to assess someone's metabolic status. To enable the usage of dietary challenges as diagnostic tools, profound knowledge about changes that occur in the postprandial period in healthy individuals is needed. In this study, we characterize the time-resolved changes in plasma levels of 634 metabolites in response to an oral glucose tolerance test (OGTT), an oral lipid tolerance test (OLTT), and a mixed meal (SLD) in healthy young males (n = 15). Metabolite levels for samples taken at different time points (20 per individual) during the challenges were available from targeted (132 metabolites) and non-targeted (502 metabolites) metabolomics. Almost half of the profiled metabolites (n = 308) showed a significant change in at least one challenge, thereof 111 metabolites responded exclusively to one particular challenge. Examples include azelate, which is linked to ω-oxidation and increased only in OLTT, and a fibrinogen cleavage peptide that has been linked to a higher risk of cardiovascular events in diabetes patients and increased only in OGTT, making its postprandial dynamics a potential target for risk management. A pool of 89 metabolites changed their plasma levels during all three challenges and represents the core postprandial response to food intake regardless of macronutrient composition. We used fuzzy c-means clustering to group these metabolites into eight clusters based on commonalities of their dynamic response patterns, with each cluster following one of four primary response patterns: (i) “decrease-increase” (valley-like) with fatty acids and acylcarnitines indicating the suppression of lipolysis, (ii) “increase-decrease” (mountain-like) including a cluster of conjugated bile acids and the glucose/insulin cluster, (iii) “steady decrease” with metabolites reflecting a carryover from meals prior to the study, and (iv) “mixed” decreasing after the glucose challenge and increasing otherwise. Despite the small number of subjects, the diversity of the challenges and the wealth of metabolomic data make this study an important step toward the characterization of postprandial responses and the identification of markers of metabolic processes regulated by food intake.
Collapse
Affiliation(s)
- Patrick Weinisch
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jarlei Fiamoncini
- Food Research Center – FoRC, Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Daniela Schranner
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Johannes Raffler
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- Digital Medicine, University Hospital of Augsburg, Augsburg, Germany
| | - Thomas Skurk
- Core Facility Human Studies, ZIEL Institute for Food and Health, Technical University of Munich, Freising, Germany
- Else Kröner Fresenius Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Manuela J. Rist
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, Karlsruhe, Germany
| | - Werner Römisch-Margl
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Cornelia Prehn
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - 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, 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, Neuherberg, Germany
- *Correspondence: Gabi Kastenmüller
| |
Collapse
|
27
|
Levy J, Silva AM, De Carli E, Cacau LT, de Alvarenga JFR, Fiamoncini J, Benseñor IM, Lotufo PA, Marchioni DM. Biomarkers of Fruit Intake Using a Targeted Metabolomics Approach: an Observational Cross-Sectional Analysis of the ELSA-Brasil Study. J Nutr 2022; 152:2023-2030. [PMID: 35641174 DOI: 10.1093/jn/nxac115] [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: 12/06/2021] [Revised: 03/11/2022] [Accepted: 05/24/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Advances in technology have led to the identification of a greater number of metabolites related to diet. Although fruit intake biomarkers have been reported in some studies, these findings require further replication, considering the relevance of fruits for diet quality and health. OBJECTIVES The aim of this study was to explore the associations of a set of potential urinary biomarkers of diet, assessed using a targeted metabolomics approach, with self-reported fruit intake data in participants of a computer-assisted 24-h dietary recall (GloboDiet software) validation study. METHODS A total of 93 individuals aged 43-72 y, 54% female, participated in this study. The subjects were a subsample of the Longitudinal Study of Adult Health (ELSA-Brasil). A 24-h dietary recall was obtained with the aid of GloboDiet software matching a 24-h urine sample from each participant. Candidate biomarkers were selected in a literature search and identified in urine by LC coupled to high-resolution MS. Spearman correlation analyses were performed between fruit intake and each biomarker. RESULTS Spearman correlation analysis showed that total fruits intake was significantly correlated with citric acid (ρ = 0.213, P = 0.041), ferulic acid sulfate I (ρ = 0.240, P = 0.020), hesperetin glucuronide/homoeriodictyol glucuronide (ρ = 0.303, P = 0.003), hydroxyhippuric acid (ρ = 0.239, P = 0.021), homovanillic alcohol sulfate (ρ = 0.339, P = 0.001), methylgallic acid sulfate (ρ = 0.268, P = 0.009), naringenin glucuronide (NG; ρ = 0.278, P = 0.007), proline betaine (PB; ρ = 0.305, P = 0.003), syringic acid sulfate (ρ = 0.210, P = 0.044), and sinapic acid sulfate (ρ = 0.412, P < 0.001). Among them, 3 have been described in literature as promising biomarkers for intake of total fruit, oranges, and citrus fruit: NG, hesperetin glucuronide, and PB. CONCLUSIONS Associations of total fruits intake with urinary measurements indicate the potential usefulness of dietary biomarkers in the Brazilian population as a complement to self-reported dietary assessments.
Collapse
Affiliation(s)
- Jessica Levy
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Alexsandro Macedo Silva
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Eduardo De Carli
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Leandro Teixeira Cacau
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - José Fernando Rinaldi de Alvarenga
- Food Research Center (FoRC), Department of Food and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jarlei Fiamoncini
- Food Research Center (FoRC), Department of Food and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Isabela Martins Benseñor
- Clinical and Epidemiological Research Center, University Hospital, University of São Paulo, São Paulo, Brazil
| | - Paulo Andrade Lotufo
- Clinical and Epidemiological Research Center, University Hospital, University of São Paulo, São Paulo, Brazil
| | - Dirce Maria Marchioni
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
28
|
Grenville ZS, Noor U, His M, Viallon V, Rinaldi S, Aglago EK, Amiano P, Brunkwall L, Chirlaque MD, Drake I, Eichelmann F, Freisling H, Grioni S, Heath AK, Kaaks R, Katzke V, Mayén-Chacon AL, Milani L, Moreno-Iribas C, Pala V, Olsen A, Sánchez MJ, Schulze MB, Tjønneland A, Tsilidis KK, Weiderpass E, Winkvist A, Zamora-Ros R, Key TJ, Smith-Byrne K, Travis RC, Schmidt JA. Diet and BMI Correlate with Metabolite Patterns Associated with Aggressive Prostate Cancer. Nutrients 2022; 14:3306. [PMID: 36014812 PMCID: PMC9415102 DOI: 10.3390/nu14163306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
Three metabolite patterns have previously shown prospective inverse associations with the risk of aggressive prostate cancer within the European Prospective Investigation into Cancer and Nutrition (EPIC). Here, we investigated dietary and lifestyle correlates of these three prostate cancer-related metabolite patterns, which included: 64 phosphatidylcholines and three hydroxysphingomyelins (Pattern 1), acylcarnitines C18:1 and C18:2, glutamate, ornithine, and taurine (Pattern 2), and 8 lysophosphatidylcholines (Pattern 3). In a two-stage cross-sectional discovery (n = 2524) and validation (n = 518) design containing 3042 men free of cancer in EPIC, we estimated the associations of 24 dietary and lifestyle variables with each pattern and the contributing individual metabolites. Associations statistically significant after both correction for multiple testing (False Discovery Rate = 0.05) in the discovery set and at p < 0.05 in the validation set were considered robust. Intakes of alcohol, total fish products, and its subsets total fish and lean fish were positively associated with Pattern 1. Body mass index (BMI) was positively associated with Pattern 2, which appeared to be driven by a strong positive BMI-glutamate association. Finally, both BMI and fatty fish were inversely associated with Pattern 3. In conclusion, these results indicate associations of fish and its subtypes, alcohol, and BMI with metabolite patterns that are inversely associated with risk of aggressive prostate cancer.
Collapse
Affiliation(s)
- Zoe S. Grenville
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Urwah Noor
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Mathilde His
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, 69008 Lyon, France
| | - Vivian Viallon
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, 69008 Lyon, France
| | - Sabina Rinaldi
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, 69008 Lyon, France
| | - Elom K. Aglago
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK
| | - Pilar Amiano
- Ministry of Health of the Basque Government, Sub Directorate for Public Health and Addictions of Gipuzkoa, 20013 San Sebastian, Spain
- Biodonostia Health Research Institute, Epidemiology of Chronic and Communicable Diseases Group, 20014 San Sebastián, Spain
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Louise Brunkwall
- Department of Clinical Sciences, Lund University, 221 84 Malmö, Sweden
| | - María Dolores Chirlaque
- CIBER of Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
- Department of Epidemiology, Regional Health Council, IMIB-Arrixaca, Murcia University, 30008 Murcia, Spain
| | - Isabel Drake
- Department of Clinical Sciences, Lund University, 221 84 Malmö, Sweden
- Skåne University Hospital, 214 28 Malmö, Sweden
| | - Fabian Eichelmann
- Department of Molecular Epidemiology, German Institute of Human Nutrition, 14558 Nuthetal, Germany
| | - Heinz Freisling
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, 69008 Lyon, France
| | - Sara Grioni
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Alicia K. Heath
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK
| | - Rudolf Kaaks
- Department of Cancer Epidemiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Verena Katzke
- Department of Cancer Epidemiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Ana-Lucia Mayén-Chacon
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, 69008 Lyon, France
| | - Lorenzo Milani
- Cancer Epidemiology Unit, Department of Medical Sciences, University of Turin, 10124 Turin, Italy
| | - Conchi Moreno-Iribas
- CIBER of Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
- Navarra Public Health Institute, 31003 Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Valeria Pala
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Anja Olsen
- Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark
- Department of Public Health, Aarhus University, DK-8000 Aarhus, Denmark
| | - Maria-Jose Sánchez
- Escuela Andaluza de Salud Pública (EASP), 18011 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Granada, 18071 Granada, Spain
| | - Matthias B. Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition, 14558 Nuthetal, Germany
| | - Anne Tjønneland
- Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, DK-1353 Copenhagen, Denmark
| | - Konstantinos K. Tsilidis
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK
- Department of Hygiene and Epidemiology, School of Medicine, University of Ioannina, 45110 Ioannina, Greece
| | - Elisabete Weiderpass
- International Agency for Research on Cancer, World Health Organization, 69008 Lyon, France
| | - Anna Winkvist
- Sustainable Health, Department of Public Health and Clinical Medicine, Umeå University, 901 87 Umeå, Sweden
- Department of Internal Medicine and Clinical Nutrition, The Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Raul Zamora-Ros
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Timothy J. Key
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Karl Smith-Byrne
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Ruth C. Travis
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Julie A. Schmidt
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
- Department of Clinical Epidemiology, Department of Clinical Medicine, University Hospital, Aarhus University and Aarhus, DK-8200 Aarhus N, Denmark
| |
Collapse
|
29
|
Wedekind R, Rothwell JA, Viallon V, Keski-Rahkonen P, Schmidt JA, Chajes V, Katzke V, Johnson T, Santucci de Magistris M, Krogh V, Amiano P, Sacerdote C, Redondo-Sánchez D, Huerta JM, Tjønneland A, Pokharel P, Jakszyn P, Tumino R, Ardanaz E, Sandanger TM, Winkvist A, Hultdin J, Schulze MB, Weiderpass E, Gunter MJ, Huybrechts I, Scalbert A. Determinants of blood acylcarnitine concentrations in healthy individuals of the European Prospective Investigation into Cancer and Nutrition. Clin Nutr 2022; 41:1735-1745. [PMID: 35779425 PMCID: PMC9358353 DOI: 10.1016/j.clnu.2022.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 04/07/2022] [Accepted: 05/28/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND & AIMS Circulating levels of acylcarnitines (ACs) have been associated with the risk of various diseases such as cancer and type 2 diabetes. Diet and lifestyle factors have been shown to influence AC concentrations but a better understanding of their biological, lifestyle and metabolic determinants is needed. METHODS Circulating ACs were measured in blood by targeted (15 ACs) and untargeted metabolomics (50 ACs) in 7770 and 395 healthy participants of the European Prospective Investigation into Cancer and Nutrition (EPIC), respectively. Associations with biological and lifestyle characteristics, dietary patterns, self-reported intake of individual foods, estimated intake of carnitine and fatty acids, and fatty acids in plasma phospholipid fraction and amino acids in blood were assessed. RESULTS Age, sex and fasting status were associated with the largest proportion of AC variability (partial-r up to 0.19, 0.18 and 0.16, respectively). Some AC species of medium or long-chain fatty acid moiety were associated with the corresponding fatty acids in plasma (partial-r = 0.24) or with intake of specific foods such as dairy foods containing the same fatty acid. ACs of short-chain fatty acid moiety (propionylcarnitine and valerylcarnitine) were moderately associated with concentrations of branched-chain amino acids (partial-r = 0.5). Intake of most other foods and of carnitine showed little association with AC levels. CONCLUSIONS Our results show that determinants of ACs in blood vary according to their fatty acid moiety, and that their concentrations are related to age, sex, diet, and fasting status. Knowledge on their potential determinants may help interpret associations of ACs with disease risk and inform on potential dietary and lifestyle factors that might be modified for disease prevention.
Collapse
Affiliation(s)
- Roland Wedekind
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, 150 Cours Albert Thomas, Lyon, France.
| | - Joseph A Rothwell
- (CESP), Faculté de Medicine, Université Paris-Saclay, Inserm, Villejuif, France; Institut Gustave Roussy, Villejuif, France
| | - Vivian Viallon
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, 150 Cours Albert Thomas, Lyon, France
| | - Pekka Keski-Rahkonen
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, 150 Cours Albert Thomas, Lyon, France
| | - Julie A Schmidt
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, UK
| | - Veronique Chajes
- International Agency for Research on Cancer, 150 Cours Albert Thomas, Lyon, France
| | - Vna Katzke
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Theron Johnson
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Vittorio Krogh
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale Dei Tumori di Milano, Milan, Italy
| | - Pilar Amiano
- Ministry of Health of the Basque Government, Sub Directorate for Public Health and Addictions of Gipuzkoa, San Sebastian, Spain; Biodonostia Health Research Institute, Epidemiology of Chronic and Communicable Diseases Group, San Sebastián, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Città Della Salute e Della Scienza University-Hospital, Via Santena 7, 10126 Turin, Italy
| | - Daniel Redondo-Sánchez
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain; Escuela Andaluza de Salud Pública (EASP), 18011 Granada, Spain; Instituto de Investigación Biosanitaria Ibs.GRANADA, 18012 Granada, Spain
| | - José María Huerta
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain; Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Murcia, Spain
| | - Anne Tjønneland
- Danish Cancer Society Research Center, Copenhagen, Denmark; Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Pratik Pokharel
- Danish Cancer Society Research Center, Copenhagen, Denmark; Institute for Nutrition Research, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Paula Jakszyn
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain; Blanquerna School of Health Sciences, Ramon Llull University, Barcelona, Spain
| | - Rosario Tumino
- Hyblean Association for Epidemiological Research AIRE - ONLUS, Ragusa, Italy
| | - Eva Ardanaz
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain; Navarra Public Health Institute, Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Torkjel M Sandanger
- Department of Community Medicine, UiT - the Arctic University of Norway, Langnes, Tromsø, Norway
| | - Anna Winkvist
- Sustainable Health, Dept Epidemiology and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Johan Hultdin
- Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; University of Potsdam, Institute of Nutritional Science, Potsdam, Germany
| | - Elisabete Weiderpass
- International Agency for Research on Cancer, 150 Cours Albert Thomas, Lyon, France
| | - Marc J Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, 150 Cours Albert Thomas, Lyon, France
| | - Inge Huybrechts
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, 150 Cours Albert Thomas, Lyon, France
| | - Augustin Scalbert
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, 150 Cours Albert Thomas, Lyon, France
| |
Collapse
|
30
|
Brachem C, Oluwagbemigun K, Langenau J, Weinhold L, Alexy U, Schmid M, Nöthlings U. Exploring the association between habitual food intake and the urine and blood metabolome in adolescents and young adults: a cohort study. Mol Nutr Food Res 2022; 66:e2200023. [PMID: 35785518 DOI: 10.1002/mnfr.202200023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/08/2022] [Indexed: 11/07/2022]
Abstract
SCOPE Habitual diet may be reflected in metabolite profiles that can improve accurate assessment of dietary exposure and further enhance our understanding of their link to health conditions. We aimed to explore the relationship of habitual food intake with blood and urine metabolites in adolescents and young adults. METHODS The study population comprised 228 participants (94 male and 134 female) of the DONALD study. Dietary intake was assessed by yearly repeated 3d-food records. Habitual diet was estimated as the average consumption of 23 food groups in adolescence. Using an untargeted metabolomics approach, we quantified 2638 metabolites in plasma and 1407 metabolites in urine. In each sex, we determined unique diet-metabolite associations using orthogonal projection to latent structures (oPLS) and random forests (RF). RESULTS We observed 6 metabolites in agreement between oPLS and RF in urine, 1 in females (vanillylmandelate to processed/ other meat) and 5 in males (indole-3-acetamide, and N6-methyladenosine to eggs; hippurate, citraconate/glutaconate, and X - 12111 to vegetables). We observed no association in blood in agreement. CONCLUSION We observed a limited reflection of habitual food group intake by single metabolites in urine and not in blood. The explored biomarkers should be confirmed in additional studies. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Christian Brachem
- Nutritional Epidemiology, Department of Nutrition and Food Sciences, Rheinische Friedrich-Wilhelms-University Bonn, 53115, Bonn, Germany
| | - Kolade Oluwagbemigun
- Nutritional Epidemiology, Department of Nutrition and Food Sciences, Rheinische Friedrich-Wilhelms-University Bonn, 53115, Bonn, Germany
| | - Julia Langenau
- Nutritional Epidemiology, Department of Nutrition and Food Sciences, Rheinische Friedrich-Wilhelms-University Bonn, 53115, Bonn, Germany
| | - Leonie Weinhold
- Institute for Medical Biometry, Informatics and Epidemiology (IMBIE), University Hospital Bonn, 53127, Bonn, Germany
| | - Ute Alexy
- Nutritional Epidemiology, Department of Nutrition and Food Sciences, Rheinische Friedrich-Wilhelms-University Bonn, DONALD Study, Heinstück 11, 44225, Dortmund, Germany
| | - Matthias Schmid
- Institute for Medical Biometry, Informatics and Epidemiology (IMBIE), University Hospital Bonn, 53127, Bonn, Germany
| | - Ute Nöthlings
- Nutritional Epidemiology, Department of Nutrition and Food Sciences, Rheinische Friedrich-Wilhelms-University Bonn, 53115, Bonn, Germany.,Nutritional Epidemiology, Department of Nutrition and Food Sciences, Rheinische Friedrich-Wilhelms-University Bonn, DONALD Study, Heinstück 11, 44225, Dortmund, Germany
| |
Collapse
|
31
|
Haikonen R, Kärkkäinen O, Koistinen V, Hanhineva K. Diet- and microbiota-related metabolite, 5-aminovaleric acid betaine (5-AVAB), in health and disease. Trends Endocrinol Metab 2022; 33:463-480. [PMID: 35508517 DOI: 10.1016/j.tem.2022.04.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/23/2022] [Accepted: 04/05/2022] [Indexed: 12/01/2022]
Abstract
5-Aminovaleric acid betaine (5-AVAB) is a trimethylated compound associated with the gut microbiota, potentially produced endogenously, and related to the dietary intake of certain foods such as whole grains. 5-AVAB accumulates within the metabolically active tissues and has been typically found in higher concentrations in the heart, muscle, and brown adipose tissue. Furthermore, 5-AVAB has been associated with positive health effects such as fetal brain development, insulin secretion, and reduced cancer risk. However, it also has been linked with some negative health outcomes such as cardiovascular disease and fatty liver disease. At the cellular level, 5-AVAB can influence cellular energy metabolism by reducing β-oxidation of fatty acids. This review will focus on the metabolic role of 5-AVAB with respect to both physiology and pathology. Moreover, the analytics and origin of 5-AVAB and related compounds will be reviewed.
Collapse
Affiliation(s)
- Retu Haikonen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland.
| | - Olli Kärkkäinen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Ville Koistinen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Life Technologies, Food Chemistry and Food Development Unit, University of Turku, Turku, Finland
| | - Kati Hanhineva
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Life Technologies, Food Chemistry and Food Development Unit, University of Turku, Turku, Finland; Department of Biology and Biological Engineering, Division of Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
32
|
Qi Q, Li J, Yu B, Moon JY, Chai JC, Merino J, Hu J, Ruiz-Canela M, Rebholz C, Wang Z, Usyk M, Chen GC, Porneala BC, Wang W, Nguyen NQ, Feofanova EV, Grove ML, Wang TJ, Gerszten RE, Dupuis J, Salas-Salvadó J, Bao W, Perkins DL, Daviglus ML, Thyagarajan B, Cai J, Wang T, Manson JE, Martínez-González MA, Selvin E, Rexrode KM, Clish CB, Hu FB, Meigs JB, Knight R, Burk RD, Boerwinkle E, Kaplan RC. Host and gut microbial tryptophan metabolism and type 2 diabetes: an integrative analysis of host genetics, diet, gut microbiome and circulating metabolites in cohort studies. Gut 2022; 71:1095-1105. [PMID: 34127525 PMCID: PMC8697256 DOI: 10.1136/gutjnl-2021-324053] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 06/07/2021] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Tryptophan can be catabolised to various metabolites through host kynurenine and microbial indole pathways. We aimed to examine relationships of host and microbial tryptophan metabolites with incident type 2 diabetes (T2D), host genetics, diet and gut microbiota. METHOD We analysed associations between circulating levels of 11 tryptophan metabolites and incident T2D in 9180 participants of diverse racial/ethnic backgrounds from five cohorts. We examined host genome-wide variants, dietary intake and gut microbiome associated with these metabolites. RESULTS Tryptophan, four kynurenine-pathway metabolites (kynurenine, kynurenate, xanthurenate and quinolinate) and indolelactate were positively associated with T2D risk, while indolepropionate was inversely associated with T2D risk. We identified multiple host genetic variants, dietary factors, gut bacteria and their potential interplay associated with these T2D-relaetd metabolites. Intakes of fibre-rich foods, but not protein/tryptophan-rich foods, were the dietary factors most strongly associated with tryptophan metabolites. The fibre-indolepropionate association was partially explained by indolepropionate-associated gut bacteria, mostly fibre-using Firmicutes. We identified a novel association between a host functional LCT variant (determining lactase persistence) and serum indolepropionate, which might be related to a host gene-diet interaction on gut Bifidobacterium, a probiotic bacterium significantly associated with indolepropionate independent of other fibre-related bacteria. Higher milk intake was associated with higher levels of gut Bifidobacterium and serum indolepropionate only among genetically lactase non-persistent individuals. CONCLUSION Higher milk intake among lactase non-persistent individuals, and higher fibre intake were associated with a favourable profile of circulating tryptophan metabolites for T2D, potentially through the host-microbial cross-talk shifting tryptophan metabolism toward gut microbial indolepropionate production.
Collapse
Affiliation(s)
- Qibin Qi
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Nutrtion, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| | - Jun Li
- Department of Nutrtion, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Houston, Texas, USA
| | - Jee-Young Moon
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jin C Chai
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jordi Merino
- Diabetes Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Jie Hu
- Division of Women's Health, Department of Medicine at Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Miguel Ruiz-Canela
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain
- CIBER Fisiopatologıa de la Obesidad y Nutricion, Instituto de Salud Carlos III, Madrid, Spain
| | - Casey Rebholz
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Zheng Wang
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
| | - Mykhaylo Usyk
- Departments of Pediatrics, Microbiology and Immunology, and Gynecology and Women's Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
| | - Guo-Chong Chen
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
| | - Bianca C Porneala
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Wenshuang Wang
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Houston, Texas, USA
- Department of Mathematics, University of Houston, Houston, Texas, USA
| | - Ngoc Quynh Nguyen
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Houston, Texas, USA
| | - Elena V Feofanova
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Houston, Texas, USA
| | - Megan L Grove
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Houston, Texas, USA
| | - Thomas J Wang
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Robert E Gerszten
- Programs in Metabolism and Medical & Population Genetics, Eli and Edythe L. Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Jordi Salas-Salvadó
- CIBER Fisiopatologıa de la Obesidad y Nutricion, Instituto de Salud Carlos III, Madrid, Spain
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Universidad Rovira i Virgili Departamento de Medicina y Cirurgía, Reus, Spain
| | - Wei Bao
- Department of Epidemiology, The University of Iowa College of Public Health, Iowa City, Iowa, USA
| | - David L Perkins
- Institute of Minority Health Research, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Martha L Daviglus
- Institute of Minority Health Research, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center, Minneapolis, Minnesota, USA
| | - Jianwen Cai
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Tao Wang
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
| | - JoAnn E Manson
- Department of Epidemiology, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
- Division of Preventive Medicine, Department of Medicine at Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Miguel A Martínez-González
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain
- CIBER Fisiopatologıa de la Obesidad y Nutricion, Instituto de Salud Carlos III, Madrid, Spain
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Kathryn M Rexrode
- Division of Women's Health, Department of Medicine at Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Clary B Clish
- Metabolomics Platform, Eli and Edythe L. Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Frank B Hu
- Channing Division of Network Medicine, Department of Medicine at Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - James B Meigs
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Rob Knight
- Department of Pediatrics, School of Medicine; Center for Microbiome Innovation, Department of Computer Science and Engineering, Jacobs School of Engineering, University of California San Diego, La Jolla, California, USA
| | - Robert D Burk
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
- Departments of Pediatrics, Microbiology and Immunology, and Gynecology and Women's Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Houston, Texas, USA
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
33
|
Smith E, Ericson U, Hellstrand S, Orho-Melander M, Nilsson PM, Fernandez C, Melander O, Ottosson F. A healthy dietary metabolic signature is associated with a lower risk for type 2 diabetes and coronary artery disease. BMC Med 2022; 20:122. [PMID: 35443726 PMCID: PMC9022292 DOI: 10.1186/s12916-022-02326-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/08/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The global burden of cardiovascular disease and type 2 diabetes could be decreased by improving dietary factors, but identification of groups suitable for interventional approaches can be difficult. Reporting of dietary intake is prone to errors, and measuring of metabolites has shown promise in determining habitual dietary intake. Our aim is to create a metabolic signature that is associated with healthy eating and test if it associates with type 2 diabetes and coronary artery disease risk. METHODS Using plasma metabolite data consisting of 111 metabolites, partial least square (PLS) regression was used to identify a metabolic signature associated with a health conscious food pattern in the Malmö Offspring Study (MOS, n = 1538). The metabolic signature's association with dietary intake was validated in the Malmö Diet and Cancer study (MDC, n = 2521). The associations between the diet-associated metabolic signature and incident type 2 diabetes and coronary artery disease (CAD) were tested using Cox regression in MDC and logistic regression in Malmö Preventive Project (MPP, n = 1083). Modelling was conducted unadjusted (model 1), adjusted for potential confounders (model 2) and additionally for potential mediators (model 3). RESULTS The metabolic signature was associated with lower risk for type 2 diabetes in both MDC (hazard ratio: 0.58, 95% CI 0.52-0.66, per 1 SD increment of the metabolic signature) and MPP (odds ratio: 0.54, 95% CI 0.44-0.65 per 1 SD increment of the metabolic signature) in model 2. The results were attenuated but remained significant in model 3 in both MDC (hazard ratio 0.73, 95% CI 0.63-0.83) and MPP (odds ratio 0.70, 95% CI 0.55-0.88). The diet-associated metabolic signature was also inversely associated with lower risk of CAD in both MDC and MPP in model 1, but the association was non-significant in model 3. CONCLUSIONS In this proof-of-concept study, we identified a healthy diet-associated metabolic signature, which was inversely associated with future risk for type 2 diabetes and coronary artery disease in two different cohorts. The association with diabetes was independent of traditional risk factors and might illustrate an effect of health conscious dietary intake on cardiometabolic health.
Collapse
Affiliation(s)
- Einar Smith
- Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, 91-12-027, SE-214 28, Malmö, Sweden.
| | - Ulrika Ericson
- Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, 91-12-027, SE-214 28, Malmö, Sweden
| | - Sophie Hellstrand
- Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, 91-12-027, SE-214 28, Malmö, Sweden
| | - Marju Orho-Melander
- Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, 91-12-027, SE-214 28, Malmö, Sweden
| | - Peter M Nilsson
- Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, 91-12-027, SE-214 28, Malmö, Sweden.,Department of Emergency and Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Céline Fernandez
- Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, 91-12-027, SE-214 28, Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, 91-12-027, SE-214 28, Malmö, Sweden.,Department of Emergency and Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Filip Ottosson
- Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, 91-12-027, SE-214 28, Malmö, Sweden.,Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
34
|
Crowder SL, Playdon MC, Gudenkauf LM, Ose J, Gigic B, Greathouse L, Peoples AR, Sleight AG, Jim HSL, Figueiredo JC. A Molecular Approach to Understanding the Role of Diet in Cancer-Related Fatigue: Challenges and Future Opportunities. Nutrients 2022; 14:nu14071496. [PMID: 35406105 PMCID: PMC9003400 DOI: 10.3390/nu14071496] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/27/2022] [Accepted: 03/31/2022] [Indexed: 02/04/2023] Open
Abstract
Cancer-related fatigue (CRF) is considered one of the most frequent and distressing symptoms for cancer survivors. Despite its high prevalence, factors that predispose, precipitate, and perpetuate CRF are poorly understood. Emerging research focuses on cancer and treatment-related nutritional complications, changes in body composition, and nutritional deficiencies that can compound CRF. Nutritional metabolomics, the novel study of diet-related metabolites in cells, tissues, and biofluids, offers a promising tool to further address these research gaps. In this position paper, we examine CRF risk factors, summarize metabolomics studies of CRF, outline dietary recommendations for the prevention and management of CRF in cancer survivorship, and identify knowledge gaps and challenges in applying nutritional metabolomics to understand dietary contributions to CRF over the cancer survivorship trajectory.
Collapse
Affiliation(s)
- Sylvia L. Crowder
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL 33617, USA; (S.L.C.); (L.M.G.); (H.S.L.J.)
| | - Mary C. Playdon
- Cancer Control and Population Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA;
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Lisa M. Gudenkauf
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL 33617, USA; (S.L.C.); (L.M.G.); (H.S.L.J.)
| | - Jennifer Ose
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT 84112, USA; (J.O.); (A.R.P.)
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA
| | - Biljana Gigic
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, 69047 Heidelberg, Germany;
| | - Leigh Greathouse
- Human Science and Design, Robbins College of Health and Human Sciences, Baylor University, Waco, TX 76798, USA;
| | - Anita R. Peoples
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT 84112, USA; (J.O.); (A.R.P.)
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA
| | - Alix G. Sleight
- Department of Physical Medicine and Rehabilitation, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
- Center for Integrated Research in Cancer and Lifestyle, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Heather S. L. Jim
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL 33617, USA; (S.L.C.); (L.M.G.); (H.S.L.J.)
| | - Jane C. Figueiredo
- Center for Integrated Research in Cancer and Lifestyle, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Correspondence:
| |
Collapse
|
35
|
Ottosson F, Hultgren L, Fernandez C, Engström G, Orho‐Melander M, Kennbäck C, Persson M, Demmer RT, Melander O, Klinge B, Nilsson PM, Jönsson D. The inverse association between a fish consumption biomarker and gingival inflammation and periodontitis: A population-based study. J Clin Periodontol 2022; 49:353-361. [PMID: 35132662 PMCID: PMC9303516 DOI: 10.1111/jcpe.13602] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/30/2022]
Abstract
AIM The metabolite 3-carboxy-4-methyl-5-propyl-2-furanpropionic acid (CMPF) is a fatty fish-intake biomarker. We investigated the association between plasma levels of CMPF in relation to gingival inflammation and periodontitis case definition, as well as the extent and severity variables. MATERIALS AND METHODS The Malmö Offspring Study is a population-based study, and the Malmö Offspring Dental Study (MODS) is its dental arm, including periodontal charting. Plasma CMPF was measured using liquid chromatography-mass spectrometry and studied in relation to periodontal diagnosis and parameters using multivariable linear or logistic regression modelling adjusting for age, sex, education, body mass index, fasting glucose, and smoking. RESULTS Metabolite data were available for 922 MODS participants. Higher CMPF levels were associated with less gingival inflammation (β = -2.12, p = .002) and lower odds of severe periodontitis (odds ratio [OR] = 0.74, 95% confidence interval [CI]: 0.56 to 0.98). Higher CMPF levels were also associated with more teeth (β = 0.19, p = .001), lower number of periodontal pockets (≥4 mm) (β = -1.07, p = .007), and lower odds of having two or more periodontal pockets of ≥6 mm (OR = 0.80, 95% CI: 0.65 to 0.98) in fully adjusted models. CONCLUSIONS CMPF, a validated biomarker of fatty fish consumption, is associated with less periodontal inflammation and periodontitis. Residual confounding cannot be ruled out, and future studies are warranted.
Collapse
Affiliation(s)
- Filip Ottosson
- Department of Clinical Sciences MalmöLund UniversityMalmöSweden,Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital DisordersStatens Serum InstitutCopenhagenDenmark
| | - Lina Hultgren
- Public Dental Service of SkåneLundSweden,Faculty of OdontologyMalmö UniversityMalmöSweden
| | | | - Gunnar Engström
- Department of Clinical Sciences MalmöLund UniversityMalmöSweden,Department of Internal MedicineSkåne University HospitalMalmöSweden
| | | | - Cecilia Kennbäck
- Department of Clinical Sciences MalmöLund UniversityMalmöSweden,Department of Internal MedicineSkåne University HospitalMalmöSweden
| | - Margaretha Persson
- Department of Clinical Sciences MalmöLund UniversityMalmöSweden,Department of Internal MedicineSkåne University HospitalMalmöSweden
| | - Ryan T. Demmer
- School of Public HealthUniversity of MinnesotaMinneapolisMinnesotaUSA,Mailman School of Public HealthColumbia UniversityNew YorkUSA
| | - Olle Melander
- Department of Clinical Sciences MalmöLund UniversityMalmöSweden,Department of Internal MedicineSkåne University HospitalMalmöSweden
| | - Björn Klinge
- Faculty of OdontologyMalmö UniversityMalmöSweden,Department of Dental MedicineKarolinska InstitutetSolnaSweden
| | - Peter M. Nilsson
- Department of Clinical Sciences MalmöLund UniversityMalmöSweden,Department of Internal MedicineSkåne University HospitalMalmöSweden
| | - Daniel Jönsson
- Department of Clinical Sciences MalmöLund UniversityMalmöSweden,Public Dental Service of SkåneLundSweden,Faculty of OdontologyMalmö UniversityMalmöSweden
| |
Collapse
|
36
|
Verri Hernandes V, Dordevic N, Hantikainen EM, Sigurdsson BB, Smárason SV, Garcia-Larsen V, Gögele M, Caprioli G, Bozzolan I, Pramstaller PP, Rainer J. Age, Sex, Body Mass Index, Diet and Menopause Related Metabolites in a Large Homogeneous Alpine Cohort. Metabolites 2022; 12:metabo12030205. [PMID: 35323648 PMCID: PMC8955763 DOI: 10.3390/metabo12030205] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 12/19/2022] Open
Abstract
Metabolomics in human serum samples provide a snapshot of the current metabolic state of an individuum. Metabolite concentrations are influenced by both genetic and environmental factors. Concentrations of certain metabolites can further depend on age, sex, menopause, and diet of study participants. A better understanding of these relationships is pivotal for the planning of metabolomics studies involving human subjects and interpretation of their results. We generated one of the largest single-site targeted metabolomics data sets consisting of 175 quantified metabolites in 6872 study participants. We identified metabolites significantly associated with age, sex, body mass index, diet, and menopausal status. While most of our results agree with previous large-scale studies, we also found novel associations including serotonin as a sex and BMI-related metabolite and sarcosine and C2 carnitine showing significantly higher concentrations in post-menopausal women. Finally, we observed strong associations between higher consumption of food items and certain metabolites, mostly phosphatidylcholines and lysophosphatidylcholines. Most, and the strongest, relationships were found for habitual meat intake while no significant relationships were found for most fruits, vegetables, and grain products. Summarizing, our results reconfirm findings from previous population-based studies on an independent cohort. Together, these findings will ultimately enable the consolidation of sets of metabolites which are related to age, sex, BMI, and menopause as well as to participants’ diet.
Collapse
Affiliation(s)
- Vinicius Verri Hernandes
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
| | - Nikola Dordevic
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
| | - Essi Marjatta Hantikainen
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
| | - Baldur Bragi Sigurdsson
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
- Department of Clinical Biochemistry, Landspitali—University Hospital, 108 Reykjavik, Iceland
| | - Sigurður Vidir Smárason
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
- BASF SE, 67063 Ludwigshafen, Germany
| | - Vanessa Garcia-Larsen
- Program in Human Nutrition, Department of International Health, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA;
| | - Martin Gögele
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
| | - Giulia Caprioli
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
| | - Ilaria Bozzolan
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
| | - Peter P. Pramstaller
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
| | - Johannes Rainer
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
- Correspondence:
| |
Collapse
|
37
|
Beelman RB, Phillips AT, Richie JP, Ba DM, Duiker SW, Kalaras MD. Health Consequences of Improving the Content of Ergothioneine in the Food Supply. FEBS Lett 2021; 596:1231-1240. [PMID: 34954825 DOI: 10.1002/1873-3468.14268] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 11/10/2022]
Abstract
Ergothioneine (ERGO) is a potent antioxidant and anti-inflammatory amino acid that is highly bioavailable to humans from the diet. ERGO is now regarded by some as a "longevity vitamin" that has the potential to mitigate some chronic diseases of aging and thereby increase life expectancy when present in adequate amounts. However, only limited knowledge exists regarding ERGO content in the human diet. Since ERGO is produced primarily by fungi, mushrooms are known to be the leading dietary source, but ERGO is found in relatively low amounts throughout the food chain as a result of soil-borne fungi or bacteria passing it on to plants through their roots. Some conventional agricultural practices that negatively impact soil fungi, such as excessive soil disturbance (plowing), can significantly reduce ERGO content of food crops when compared to regenerative practices such as eliminating tillage of the soil (No-Till). This has led us to the concept that ERGO may be a definitive connection between soil health and human health.
Collapse
Affiliation(s)
- Robert B Beelman
- Department of Food Science, College of Agricultural Sciences, Penn State University, 202 Rodney A. Erickson Food Science Building University Park, State College, PA, 16802, USA
| | - Allen T Phillips
- Department of Biochemistry and Molecular Biology, Eberly College of Science, Penn State University, 203A South Frear Building University Park, State College, PA, 16802, USA
| | - John P Richie
- Department of Public Health Sciences, College of Medicine, Penn State University, 500 University Dr. Hershey, PA, 17033, USA
| | - Djibril M Ba
- Department of Public Health Sciences, College of Medicine, Penn State University, 500 University Dr. Hershey, PA, 17033, USA
| | - Sjoerd W Duiker
- Department of Plant Science, College of Agricultural Sciences, Penn State University, 408 ASI Building, University Park, State College, PA, 16802, USA
| | - Michael D Kalaras
- Department of Food Science, College of Agricultural Sciences, Penn State University, 202 Rodney A. Erickson Food Science Building University Park, State College, PA, 16802, USA
| |
Collapse
|
38
|
Rafiq T, Azab SM, Teo KK, Thabane L, Anand SS, Morrison KM, de Souza RJ, Britz-McKibbin P. Nutritional Metabolomics and the Classification of Dietary Biomarker Candidates: A Critical Review. Adv Nutr 2021; 12:2333-2357. [PMID: 34015815 PMCID: PMC8634495 DOI: 10.1093/advances/nmab054] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/20/2021] [Accepted: 04/06/2021] [Indexed: 02/06/2023] Open
Abstract
Recent advances in metabolomics allow for more objective assessment of contemporary food exposures, which have been proposed as an alternative or complement to self-reporting of food intake. However, the quality of evidence supporting the utility of dietary biomarkers as valid measures of habitual intake of foods or complex dietary patterns in diverse populations has not been systematically evaluated. We reviewed nutritional metabolomics studies reporting metabolites associated with specific foods or food groups; evaluated the interstudy repeatability of dietary biomarker candidates; and reported study design, metabolomic approach, analytical technique(s), and type of biofluid analyzed. A comprehensive literature search of 5 databases (PubMed, EMBASE, Web of Science, BIOSIS, and CINAHL) was conducted from inception through December 2020. This review included 244 studies, 169 (69%) of which were interventional studies (9 of these were replicated in free-living participants) and 151 (62%) of which measured the metabolomic profile of serum and/or plasma. Food-based metabolites identified in ≥1 study and/or biofluid were associated with 11 food-specific categories or dietary patterns: 1) fruits; 2) vegetables; 3) high-fiber foods (grain-rich); 4) meats; 5) seafood; 6) pulses, legumes, and nuts; 7) alcohol; 8) caffeinated beverages, teas, and cocoas; 9) dairy and soya; 10) sweet and sugary foods; and 11) complex dietary patterns and other foods. We conclude that 69 metabolites represent good candidate biomarkers of food intake. Quantitative measurement of these metabolites will advance our understanding of the relation between diet and chronic disease risk and support evidence-based dietary guidelines for global health.
Collapse
Affiliation(s)
- Talha Rafiq
- Medical Sciences Graduate Program, Faculty of Health Sciences, McMaster University, Hamilton, Canada
- Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, Canada
| | - Sandi M Azab
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Canada
- Department of Pharmacognosy, Alexandria University, Alexandria, Egypt
| | - Koon K Teo
- Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, Canada
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Canada
- Department of Medicine, McMaster University, Hamilton, Canada
| | - Lehana Thabane
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Canada
| | - Sonia S Anand
- Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, Canada
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Canada
- Department of Medicine, McMaster University, Hamilton, Canada
| | | | - Russell J de Souza
- Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, Canada
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Canada
| | | |
Collapse
|
39
|
Loftfield E, Stepien M, Viallon V, Trijsburg L, Rothwell JA, Robinot N, Biessy C, Bergdahl IA, Bodén S, Schulze MB, Bergman M, Weiderpass E, Schmidt JA, Zamora-Ros R, Nøst TH, Sandanger TM, Sonestedt E, Ohlsson B, Katzke V, Kaaks R, Ricceri F, Tjønneland A, Dahm CC, Sánchez MJ, Trichopoulou A, Tumino R, Chirlaque MD, Masala G, Ardanaz E, Vermeulen R, Brennan P, Albanes D, Weinstein SJ, Scalbert A, Freedman ND, Gunter MJ, Jenab M, Sinha R, Keski-Rahkonen P, Ferrari P. Novel Biomarkers of Habitual Alcohol Intake and Associations With Risk of Pancreatic and Liver Cancers and Liver Disease Mortality. J Natl Cancer Inst 2021; 113:1542-1550. [PMID: 34010397 PMCID: PMC8562969 DOI: 10.1093/jnci/djab078] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 02/24/2021] [Accepted: 04/09/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Alcohol is an established risk factor for several cancers, but modest alcohol-cancer associations may be missed because of measurement error in self-reported assessments. Biomarkers of habitual alcohol intake may provide novel insight into the relationship between alcohol and cancer risk. METHODS Untargeted metabolomics was used to identify metabolites correlated with self-reported habitual alcohol intake in a discovery dataset from the European Prospective Investigation into Cancer and Nutrition (EPIC; n = 454). Statistically significant correlations were tested in independent datasets of controls from case-control studies nested within EPIC (n = 280) and the Alpha-Tocopherol, Beta-Carotene Cancer Prevention (ATBC; n = 438) study. Conditional logistic regression was used to estimate odds ratios (ORs) and 95% confidence intervals (CIs) for associations of alcohol-associated metabolites and self-reported alcohol intake with risk of pancreatic cancer, hepatocellular carcinoma (HCC), liver cancer, and liver disease mortality in the contributing studies. RESULTS Two metabolites displayed a dose-response association with self-reported alcohol intake: 2-hydroxy-3-methylbutyric acid and an unidentified compound. A 1-SD (log2) increase in levels of 2-hydroxy-3-methylbutyric acid was associated with risk of HCC (OR = 2.54, 95% CI = 1.51 to 4.27) and pancreatic cancer (OR = 1.43, 95% CI = 1.03 to 1.99) in EPIC and liver cancer (OR = 2.00, 95% CI = 1.44 to 2.77) and liver disease mortality (OR = 2.16, 95% CI = 1.63 to 2.86) in ATBC. Conversely, a 1-SD (log2) increase in questionnaire-derived alcohol intake was not associated with HCC or pancreatic cancer in EPIC or liver cancer in ATBC but was associated with liver disease mortality (OR = 2.19, 95% CI = 1.60 to 2.98) in ATBC. CONCLUSIONS 2-hydroxy-3-methylbutyric acid is a candidate biomarker of habitual alcohol intake that may advance the study of alcohol and cancer risk in population-based studies.
Collapse
Affiliation(s)
- Erikka Loftfield
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute,National Institutes of Health, Bethesda, MD, USA
| | - Magdalena Stepien
- Nutritional Epidemiology Group, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Vivian Viallon
- Nutritional Methodology and Biostatistics Group, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Laura Trijsburg
- Nutritional Methodology and Biostatistics Group, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Joseph A Rothwell
- Nutritional Epidemiology Group, International Agency for Research on Cancer (IARC-WHO), Lyon, France
- Gustave Roussy, F-94805, Villejuif, France
- Biomarkers Group, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Nivonirina Robinot
- Centre for Epidemiology and Population Health (U1018), Generations and Health team, Faculté de Médecine, Université Paris-Saclay, UVSQ, INSERM, Villejuif, France
| | - Carine Biessy
- Nutritional Methodology and Biostatistics Group, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | | | - Stina Bodén
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Manuela Bergman
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | | | - Julie A Schmidt
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Raul Zamora-Ros
- Unit of Nutrition and Cancer, Epidemiology Research Program, Catalan Institute of Oncology, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Therese H Nøst
- Department of Community Medicine, UiT- The Arctic University of Norway, Tromsø, Norway
| | - Torkjel M Sandanger
- Department of Community Medicine, UiT- The Arctic University of Norway, Tromsø, Norway
| | - Emily Sonestedt
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Bodil Ohlsson
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Verena Katzke
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Fulvio Ricceri
- Department of Clinical and Biological Sciences, University of Turin, Italy; Unit of Epidemiology, Regional Health Service ASL TO3, Grugliasco, TO, Italy
| | - Anne Tjønneland
- Danish Cancer Society Research Center; University of Copenhagen, Department of Public Health
| | | | - Maria-Jose Sánchez
- Escuela Andaluza de Salud Pública (EASP), Granada, Spain; Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Granada, Granada, Spain
| | | | - Rosario Tumino
- Cancer Registry and Histopathology Department, Provincial Health Authority (ASP 7), Ragusa, Italy
| | - María-Dolores Chirlaque
- Department of Epidemiology, Regional Health Council, IMIB-Arrixaca, Murcia University, Murcia, Spain
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Giovanna Masala
- Cancer Risk Factors and Life-Style Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network—ISPRO, Florence, Italy
| | - Eva Ardanaz
- Navarra Public Health Institute, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- CIBER Epidemiology and Public Health CIBERESP, Madrid, Spain
| | - Roel Vermeulen
- Institute for Risk Assessment Sciences, Division of Environmental Epidemiology, Utrecht University, Utrecht, the Netherlands
| | - Paul Brennan
- Genetic Epidemiology Group, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Demetrius Albanes
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute,National Institutes of Health, Bethesda, MD, USA
| | - Stephanie J Weinstein
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute,National Institutes of Health, Bethesda, MD, USA
| | - Augustin Scalbert
- Centre for Epidemiology and Population Health (U1018), Generations and Health team, Faculté de Médecine, Université Paris-Saclay, UVSQ, INSERM, Villejuif, France
| | - Neal D Freedman
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute,National Institutes of Health, Bethesda, MD, USA
| | - Marc J Gunter
- Nutritional Epidemiology Group, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Mazda Jenab
- Nutritional Epidemiology Group, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Rashmi Sinha
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute,National Institutes of Health, Bethesda, MD, USA
| | - Pekka Keski-Rahkonen
- Centre for Epidemiology and Population Health (U1018), Generations and Health team, Faculté de Médecine, Université Paris-Saclay, UVSQ, INSERM, Villejuif, France
| | - Pietro Ferrari
- Nutritional Methodology and Biostatistics Group, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| |
Collapse
|
40
|
Sehgal R, Ilha M, Vaittinen M, Kaminska D, Männistö V, Kärjä V, Tuomainen M, Hanhineva K, Romeo S, Pajukanta P, Pihlajamäki J, de Mello VD. Indole-3-Propionic Acid, a Gut-Derived Tryptophan Metabolite, Associates with Hepatic Fibrosis. Nutrients 2021; 13:nu13103509. [PMID: 34684510 PMCID: PMC8538297 DOI: 10.3390/nu13103509] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
Background and Aims: Gut microbiota-derived metabolites play a vital role in maintenance of human health and progression of disorders, including obesity and type 2 diabetes (T2D). Indole-3-propionic acid (IPA), a gut-derived tryptophan metabolite, has been recently shown to be lower in individuals with obesity and T2D. IPA’s beneficial effect on liver health has been also explored in rodent and cell models. In this study, we investigated the association of IPA with human liver histology and transcriptomics, and the potential of IPA to reduce hepatic stellate cell activation in vitro. Methods: A total of 233 subjects (72% women; age 48.3 ± 9.3 years; BMI 43.1 ± 5.4 kg/m2) undergoing bariatric surgery with detailed liver histology were included. Circulating IPA levels were measured using LC-MS and liver transcriptomics with total RNA-sequencing. LX-2 cells were used to study hepatoprotective effect of IPA in cells activated by TGF-β1. Results: Circulating IPA levels were found to be lower in individuals with liver fibrosis compared to those without fibrosis (p = 0.039 for all participants; p = 0.013 for 153 individuals without T2D). Accordingly, levels of circulating IPA associated with expression of 278 liver transcripts (p < 0.01) that were enriched for the genes regulating hepatic stellate cells (HSCs) activation and hepatic fibrosis signaling. Our results suggest that IPA may have hepatoprotective potential because it is able to reduce cell adhesion, cell migration and mRNA gene expression of classical markers of HSCs activation in LX-2 cells (all p < 0.05). Conclusion: The association of circulating IPA with liver fibrosis and the ability of IPA to reduce activation of LX-2 cells suggests that IPA may have a therapeutic potential. Further molecular studies are needed to investigate the mechanisms how IPA can ameliorate hepatic fibrosis.
Collapse
Affiliation(s)
- Ratika Sehgal
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; (R.S.); (M.I.); (M.V.); (D.K.); (M.T.); (K.H.); (J.P.)
| | - Mariana Ilha
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; (R.S.); (M.I.); (M.V.); (D.K.); (M.T.); (K.H.); (J.P.)
| | - Maija Vaittinen
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; (R.S.); (M.I.); (M.V.); (D.K.); (M.T.); (K.H.); (J.P.)
| | - Dorota Kaminska
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; (R.S.); (M.I.); (M.V.); (D.K.); (M.T.); (K.H.); (J.P.)
| | - Ville Männistö
- Departments of Medicine, University of Eastern Finland and Kuopio University Hospital, 70211 Kuopio, Finland;
| | - Vesa Kärjä
- Department of Pathology, University of Eastern Finland and Kuopio University Hospital, 70211 Kuopio, Finland;
| | - Marjo Tuomainen
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; (R.S.); (M.I.); (M.V.); (D.K.); (M.T.); (K.H.); (J.P.)
| | - Kati Hanhineva
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; (R.S.); (M.I.); (M.V.); (D.K.); (M.T.); (K.H.); (J.P.)
- Department of Life Technologies, Food Chemistry and Food Development Unit, University of Turku, 20500 Turku, Finland
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, University of Gothenburg, 40530 Gothenburg, Sweden;
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA;
- Institute for Precision Health, School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Jussi Pihlajamäki
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; (R.S.); (M.I.); (M.V.); (D.K.); (M.T.); (K.H.); (J.P.)
- Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, 70211 Kuopio, Finland
| | - Vanessa D. de Mello
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; (R.S.); (M.I.); (M.V.); (D.K.); (M.T.); (K.H.); (J.P.)
- Correspondence:
| |
Collapse
|
41
|
Phenol Biological Metabolites as Food Intake Biomarkers, a Pending Signature for a Complete Understanding of the Beneficial Effects of the Mediterranean Diet. Nutrients 2021; 13:nu13093051. [PMID: 34578929 PMCID: PMC8471182 DOI: 10.3390/nu13093051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/11/2021] [Accepted: 08/20/2021] [Indexed: 01/14/2023] Open
Abstract
The Mediterranean diet (MD) has become a dietary pattern of reference due to its preventive effects against chronic diseases, especially relevant in cardiovascular diseases (CVD). Establishing an objective tool to determine the degree of adherence to the MD is a pending task and deserves consideration. The central axis that distinguishes the MD from other dietary patterns is the choice and modality of food consumption. Identification of intake biomarkers of commonly consumed foods is a key strategy for estimating the degree of adherence to the MD and understanding the protective mechanisms that lead to a positive impact on health. Throughout this review we propose potential candidates to be validated as MD adherence biomarkers, with particular focus on the metabolites derived from the phenolic compounds that are associated with the consumption of typical Mediterranean plant foods. Certain phenolic metabolites are good indicators of the intake of specific foods, but others denote the intake of a wide-range of foods. For this, it is important to emphasise the need to increase the number of dietary interventions with specific foods in order to validate the biomarkers of MD adherence. Moreover, the identification and quantification of food phenolic intake biomarkers encouraging scientific research focuses on the study of the biological mechanisms in which polyphenols are involved.
Collapse
|
42
|
Moslehi N, Marzbani R, Rezadoost H, Mirmiran P, Ramezani Tehrani F, Azizi F. Serum metabolomics study of the association between dairy intake and the anti-müllerian hormone annual decline rate. Nutr Metab (Lond) 2021; 18:66. [PMID: 34176512 PMCID: PMC8237474 DOI: 10.1186/s12986-021-00591-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/12/2021] [Indexed: 02/08/2023] Open
Abstract
Background Dairy intake has been implicated in later ovarian aging but mechanism underlying the association is unknown. This study aimed to investigate (1) associations between dairy intake and metabolites previously shown related to anti-müllerian hormone (AMH) decline rate; (2) mediating roles of these metabolites in the prospective association of total dairy consumption with odds of AMH fast decline rate.
Methods The participants comprised 186 reproductive-aged women randomly selected from the Tehran Lipid and Glucose Study. AMH was measured at baseline (1999–2001) and the 5th follow-up (2014–2017), and dietary data was collected at the second follow-up (2005–2008) using a food frequency questionnaire. Untargeted metabolomics was performed by gas chromatography–mass spectrometry using fasting-serum samples of the second follow-up. We analyzed dairy intake in association with the eight metabolites linked to the higher odds of AMH fast decline rate using linear regression with the Benjamini–Hochberg false discovery correction. Mediatory roles of the metabolites were assessed by bootstrapping. Results Mean age and BMI of the participants at metabolomics assessment were 44.7 ± 5.87 years and 28.8 ± 4.88 kg/m2, respectively. Phosphate, branched-chain amino acids (BCAAs), and proline decreased significantly from the first to the third tertile of total dairy intake. Total dairy as a continuous variable inversely associated with phosphate (beta = −0.166; p value = 0.018), valine (beta = −0.176; p value = 0.016), leucine (beta = −0.226; p value = 0.002), proline (beta = −0.219; p value = 0.003), and urea (beta = −0.156; p = 0.035) after accounting for all potential covariates and correction for multiplicity (q-value < 0.1). Fermented dairy showed similar results, but milk did not associate with any of the metabolites. Simple mediation showed significant indirect effects for phosphate, proline, and BCAAs but not urea. Entering the sum of phosphate, proline, and BCAAs as a mediator, the metabolites' total indirect effects were significant [β = −0.12 (95% CIs − 0.26, − 0.04)]. In contrast, the direct association of total dairy intake with the fast decline in AMH was non-significant [β = −0.28 (95% CIs − 0.67, 0.10)]. Conclusions Total dairy was inversely associated with AMH decline rate-related metabolites. Inverse association of dairy intakes with the odds of AMH fast decline rate was indirectly mediated by lower phosphate, proline, and BCAAs.
Collapse
Affiliation(s)
- Nazanin Moslehi
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rezvan Marzbani
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Hassan Rezadoost
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Parvin Mirmiran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Fahimeh Ramezani Tehrani
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
43
|
Hübel C, Herle M, Santos Ferreira DL, Abdulkadir M, Bryant-Waugh R, Loos RJF, Bulik CM, Lawlor DA, Micali N. Childhood overeating is associated with adverse cardiometabolic and inflammatory profiles in adolescence. Sci Rep 2021; 11:12478. [PMID: 34127697 PMCID: PMC8203659 DOI: 10.1038/s41598-021-90644-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 05/07/2021] [Indexed: 12/15/2022] Open
Abstract
Childhood eating behaviour contributes to the rise of obesity and related noncommunicable disease worldwide. However, we lack a deep understanding of biochemical alterations that can arise from aberrant eating behaviour. In this study, we prospectively associate longitudinal trajectories of childhood overeating, undereating, and fussy eating with metabolic markers at age 16 years to explore adolescent metabolic alterations related to specific eating patterns in the first 10 years of life. Data are from the Avon Longitudinal Study of Parents and Children (n = 3104). We measure 158 metabolic markers with a high-throughput (1H) NMR metabolomics platform. Increasing childhood overeating is prospectively associated with an adverse cardiometabolic profile (i.e., hyperlipidemia, hypercholesterolemia, hyperlipoproteinemia) in adolescence; whereas undereating and fussy eating are associated with lower concentrations of the amino acids glutamine and valine, suggesting a potential lack of micronutrients. Here, we show associations between early behavioural indicators of eating and metabolic markers.
Collapse
Affiliation(s)
- Christopher Hübel
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- UK National Institute for Health Research (NIHR) Biomedical Research Centre for Mental Health, South London and Maudsley Hospital, London, UK
- National Centre for Register-Based Research, Department of Economics and Business Economics, Aarhus University, Aarhus, Denmark
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Moritz Herle
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Diana L Santos Ferreira
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Mohamed Abdulkadir
- Department of Pediatrics Gynaecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Rachel Bryant-Waugh
- Maudsley Centre for Child and Adolescent Eating Disorders, Michael Rutter Centre for Children and Young People, Maudsley Hospital, London, UK
| | - Ruth J F Loos
- Icahn School of Medicine At Mount Sinai, New York, NY, USA
| | - Cynthia M Bulik
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry, University of North Carolina At Chapel Hill, Chapel Hill, NC, USA
- Department of Nutrition, University of North Carolina At Chapel Hill, Chapel Hill, NC, USA
| | - Deborah A Lawlor
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Bristol National Institute of Health Research Biomedical Research Centre, Bristol, UK
| | - Nadia Micali
- Department of Pediatrics Gynaecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
- Great Ormond Street Institute of Child Health, University College London, London, UK.
| |
Collapse
|
44
|
Solvik BS, Øyen J, Kvestad I, Markhus MW, Ueland PM, McCann A, Strand TA. Biomarkers and Fatty Fish Intake: A Randomized Controlled Trial in Norwegian Preschool Children. J Nutr 2021; 151:2134-2141. [PMID: 33978160 PMCID: PMC8349119 DOI: 10.1093/jn/nxab112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/25/2021] [Accepted: 03/29/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Biomarkers such as omega-3 (n-3) PUFAs, urinary iodine concentration (UIC), 1-methylhistidine (1-MH), and trimethylamine N-oxide (TMAO) have been associated with fish intake in observational studies, but data from children in randomized controlled trials are limited. OBJECTIVES The objective of this exploratory analysis was to investigate the effects of fatty fish intake compared with meat intake on various biomarkers in preschool children. METHODS We randomly allocated (1:1) 232 children, aged 4 to 6 y, from 13 kindergartens. The children received lunch meals of either fatty fish (herring/mackerel) or meat (chicken/lamb/beef) 3 times a week for 16 wk. We analyzed 86 biomarkers in plasma (n = 207), serum (n = 195), RBCs (n = 211), urine (n = 200), and hair samples (n = 210). We measured the effects of the intervention on the normalized biomarker concentrations in linear mixed-effect regression models taking the clustering within the kindergartens into account. The results are presented as standardized effect sizes. RESULTS We found significant effects of the intervention on the following biomarkers: RBC EPA (20:5n-3), 0.61 (95% CI: 0.36, 0.86); DHA (22:6n-3), 0.43 (95% CI: 0.21, 0.66); total n-3 PUFAs, 0.41 (95% CI: 0.20, 0.64); n-3/n-6 ratio, 0.48 (95% CI: 0.24, 0.71); adrenic acid (22:4n-6, -0.65 (95% CI: -0.91, -0.40), arachidonic acid (20:4n-6), -0.54 (95% CI: -0.79, -0.28); total n-6 PUFAs, -0.31 (95% CI: -0.56, -0.06); UIC, 0.32 (95% CI: 0.052, 0.59); hair mercury, 0.83 (95% CI: 0.05, 1.05); and plasma 1-MH, -0.35 (95% CI: -0.61, -0.094). CONCLUSIONS Of the 86 biomarkers, the strongest effect of fatty fish intake was on n-3 PUFAs, UIC, hair mercury, and plasma 1-MH. We observed no or limited effects on biomarkers related to micronutrient status, inflammation, or essential amino acid, choline oxidation, and tryptophan pathways.The trial was registered at clinicaltrials.gov (NCT02331667).
Collapse
Affiliation(s)
- Beate S Solvik
- Innlandet Hospital Trust, Lillehammer, Norway,Centre for International Health, University of Bergen, Bergen, Norway
| | | | - Ingrid Kvestad
- Regional Center for Child and Youth Mental Health and Child Welfare, NORCE Norwegian Research Centre, Bergen, Norway
| | | | | | | | - Tor A Strand
- Innlandet Hospital Trust, Lillehammer, Norway,Centre for International Health, University of Bergen, Bergen, Norway
| |
Collapse
|
45
|
Jobard E, Dossus L, Baglietto L, Fornili M, Lécuyer L, Mancini FR, Gunter MJ, Trédan O, Boutron-Ruault MC, Elena-Herrmann B, Severi G, Rothwell JA. Investigation of circulating metabolites associated with breast cancer risk by untargeted metabolomics: a case-control study nested within the French E3N cohort. Br J Cancer 2021; 124:1734-1743. [PMID: 33723391 PMCID: PMC8110540 DOI: 10.1038/s41416-021-01304-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Perturbations in circulating metabolites prior to a breast cancer diagnosis are not well characterised. We aimed to gain more detailed knowledge to help understand and prevent the disease. METHODS Baseline plasma samples from 791 breast cancer cases and 791 matched controls from the E3N (EPIC-France) cohort were profiled by nuclear magnetic resonance (NMR)-based untargeted metabolomics. Partial least-squares discriminant analysis (PLS-DA) models were built from NMR profiles to predict disease outcome, and odds ratios and false discovery rate (FDR)-adjusted CIs were calculated for 43 identified metabolites by conditional logistic regression. RESULTS Breast cancer onset was predicted in the premenopausal subgroup with modest accuracy (AUC 0.61, 95% CI: 0.49-0.73), and 10 metabolites associated with risk, particularly histidine (OR = 1.70 per SD increase, FDR-adjusted CI 1.19-2.41), N-acetyl glycoproteins (OR = 1.53, FDR-adjusted CI 1.18-1.97), glycerol (OR = 1.55, FDR-adjusted CI 1.11-2.18) and ethanol (OR = 1.44, FDR-adjusted CI 1.05-1.97). No predictive capacity or significant metabolites were found overall or for postmenopausal women. CONCLUSIONS Perturbed metabolism compared to controls was observed in premenopausal but not postmenopausal cases. Histidine and NAC have known involvement in inflammatory pathways, and the robust association of ethanol with risk suggests the involvement of alcohol intake.
Collapse
Affiliation(s)
- Elodie Jobard
- Univ Lyon, CNRS, Université Claude Bernard Lyon 1, ENS de Lyon, Institut des Sciences Analytiques, UMR 5280, Villeurbanne, France
- Université de Lyon, Centre Léon Bérard, Département d'Oncologie Médicale, Lyon, France
| | - Laure Dossus
- Nutrition and Metabolism Section, International Agency for Research on Cancer, Lyon, France
| | - Laura Baglietto
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Marco Fornili
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Lucie Lécuyer
- Université Paris-Saclay, UVSQ, Univ. Paris-Sud, Inserm, Gustave Roussy, Exposome and Heredity Team, Centre for Epidemiology and Population Health, Villejuif, France
| | - Francesca Romana Mancini
- Université Paris-Saclay, UVSQ, Univ. Paris-Sud, Inserm, Gustave Roussy, Exposome and Heredity Team, Centre for Epidemiology and Population Health, Villejuif, France
| | - Marc J Gunter
- Nutrition and Metabolism Section, International Agency for Research on Cancer, Lyon, France
| | - Olivier Trédan
- Université de Lyon, Centre Léon Bérard, Département d'Oncologie Médicale, Lyon, France
| | - Marie-Christine Boutron-Ruault
- Université Paris-Saclay, UVSQ, Univ. Paris-Sud, Inserm, Gustave Roussy, Exposome and Heredity Team, Centre for Epidemiology and Population Health, Villejuif, France
| | - Bénédicte Elena-Herrmann
- Univ Lyon, CNRS, Université Claude Bernard Lyon 1, ENS de Lyon, Institut des Sciences Analytiques, UMR 5280, Villeurbanne, France
- Univ Grenoble Alpes, CNRS, INSERM, IAB, Allée des Alpes, Grenoble, France
| | - Gianluca Severi
- Université Paris-Saclay, UVSQ, Univ. Paris-Sud, Inserm, Gustave Roussy, Exposome and Heredity Team, Centre for Epidemiology and Population Health, Villejuif, France
- Department of Statistics, Computer Science and Applications "G. Parenti" (DISIA), University of Florence, Firenze, Italy
| | - Joseph A Rothwell
- Université Paris-Saclay, UVSQ, Univ. Paris-Sud, Inserm, Gustave Roussy, Exposome and Heredity Team, Centre for Epidemiology and Population Health, Villejuif, France.
| |
Collapse
|
46
|
Identification and Reproducibility of Urinary Metabolomic Biomarkers of Habitual Food Intake in a Cross-Sectional Analysis of the Cancer Prevention Study-3 Diet Assessment Sub-Study. Metabolites 2021; 11:metabo11040248. [PMID: 33920694 PMCID: PMC8072637 DOI: 10.3390/metabo11040248] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/06/2021] [Accepted: 04/14/2021] [Indexed: 11/16/2022] Open
Abstract
Previous cross-sectional metabolomics studies have identified many potential dietary biomarkers, mostly in blood. Few studies examined urine samples although urine is preferred for dietary biomarker discovery. Furthermore, little is known regarding the reproducibility of urinary metabolomic biomarkers over time. We aimed to identify urinary metabolomic biomarkers of diet and assess their reproducibility over time. We conducted a metabolomics analysis among 648 racially/ethnically diverse men and women in the Diet Assessment Sub-study of the Cancer Prevention Study-3 cohort to examine the correlation between >100 food groups/items [101 by a food frequency questionnaire (FFQ), and 105 by repeated 24 h diet recalls (24HRs)] and 1391 metabolites measured in 24 h urine sample replicates, six months apart. Diet-metabolite associations were examined by Pearson's partial correlation analysis. Biomarkers were evaluated for prediction accuracy assessed using area under the curve (AUC) calculated from the receiver operating characteristic curve and for reproducibility assessed using intraclass correlation coefficients (ICCs). A total of 1708 diet-metabolite associations were identified after Bonferroni correction for multiple comparisons and restricting correlation coefficients to >0.2 or <-0.2 (1570 associations using the FFQ and 933 using 24HRs), 513 unique metabolites correlated with 79 food groups/items. The median ICCs of the 513 putative biomarkers was 0.53 (interquartile range 0.42-0.62). In this study, with comprehensive dietary data and repeated 24 h urinary metabolic profiles, we identified a large number of diet-metabolite correlations and replicated many found in previous studies. Our findings revealed the promise of urine samples for dietary biomarker discovery in a large cohort study and provide important information on biomarker reproducibility, which could facilitate their utilization in future clinical and epidemiological studies.
Collapse
|
47
|
Wedekind R, Keski-Rahkonen P, Robinot N, Viallon V, Rothwell JA, Boutron-Ruault MC, Aleksandrova K, Wittenbecher C, Schulze MB, Halkjaer J, Rostgaard-Hansen AL, Kaaks R, Katzke V, Masala G, Tumino R, Santucci de Magistris M, Krogh V, Sacerdote C, Jakszyn P, Weiderpass E, Gunter MJ, Huybrechts I, Scalbert A. Pepper Alkaloids and Processed Meat Intake: Results from a Randomized Trial and the European Prospective Investigation into Cancer and Nutrition (EPIC) Cohort. Mol Nutr Food Res 2021; 65:e2001141. [PMID: 33592132 DOI: 10.1002/mnfr.202001141] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/15/2021] [Indexed: 12/27/2022]
Abstract
SCOPE Processed meat intake has been associated with adverse health outcomes. However, little is known about the type of processed meat more particularly responsible for these effects. This study aims to identify novel biomarkers for processed meat intake. METHODS AND RESULTS In a controlled randomized cross-over dietary intervention study, 12 healthy volunteers consume different processed and non-processed meats for 3 consecutive days each. Metabolomics analyses are applied on post-intervention fasting blood and urine samples to identify discriminating molecular features of processed meat intake. Nine and five pepper alkaloid metabolites, including piperine, are identified as major discriminants of salami intake in urine and plasma, respectively. The associations with processed meat intake are tested for replication in a cross-sectional study (n = 418) embedded within the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort. Three of the serum metabolites including piperine are associated with habitual intake of sausages and to a lesser extent of total processed meat. CONCLUSION Pepper alkaloids are major discriminants of intake for sausages that contain high levels of pepper used as ingredient. Further work is needed to assess if pepper alkaloids in combination with other metabolites may serve as biomarkers of processed meat intake.
Collapse
Affiliation(s)
- Roland Wedekind
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, 150 cours Albert Thomas, Lyon, France
| | - Pekka Keski-Rahkonen
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, 150 cours Albert Thomas, Lyon, France
| | - Nivonirina Robinot
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, 150 cours Albert Thomas, Lyon, France
| | - Vivian Viallon
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, 150 cours Albert Thomas, Lyon, France
| | - Joseph A Rothwell
- CESP, Faculté de Medicine, Université Paris-Saclay, Inserm, Villejuif, France
- Institut Gustave Roussy, Villejuif, France
| | | | - Krasimira Aleksandrova
- Department of Nutrition and Gerontology, Nutrition, Immunity and Metabolism Senior Scientist Group, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Clemens Wittenbecher
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- German Center of Diabetes Research (DZD), Neuherberg, Germany
| | - Matthias B Schulze
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Jytte Halkjaer
- Danish Cancer Society Research Centre, Diet, Genes and Environment, Copenhagen, Denmark
| | | | - Rudolf Kaaks
- Department of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Verena Katzke
- Department of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Giovanna Masala
- Cancer Risk Factors and Life-Style Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network - ISPRO, Florence, Italy
| | - Rosario Tumino
- Cancer Registry and Histopathology Department, Provincial Health Authority (ASP 7), Ragusa, Italy
| | | | - Vittorio Krogh
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Città della Salute e della Scienza University-Hospital, Turin, Italy
| | - Paula Jakszyn
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology, Barcelona, Spain
- Blanquerna School of Health Sciences, Ramon Llull University, Barcelona, Spain
| | - Elisabete Weiderpass
- International Agency for Research on Cancer, 150 cours Albert Thomas, Lyon, France
| | - Marc J Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, 150 cours Albert Thomas, Lyon, France
| | - Inge Huybrechts
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, 150 cours Albert Thomas, Lyon, France
| | - Augustin Scalbert
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, 150 cours Albert Thomas, Lyon, France
| |
Collapse
|
48
|
Rebholz CM, Gao Y, Talegawkar S, Tucker KL, Colantonio LD, Muntner P, Ngo D, Chen ZZ, Cruz D, Katz D, Tahir UA, Clish C, Gerszten RE, Wilson JG. Metabolomic Markers of Southern Dietary Patterns in the Jackson Heart Study. Mol Nutr Food Res 2021; 65:e2000796. [PMID: 33629508 PMCID: PMC8192080 DOI: 10.1002/mnfr.202000796] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 01/07/2021] [Indexed: 02/02/2023]
Abstract
SCOPE New biomarkers are needed that are representative of dietary intake. METHODS AND RESULTS We assess metabolites associated with Southern dietary patterns in 1401 Jackson Heart Study participants. Three dietary patterns are empirically derived using principal component analysis: meat and fast food, fish and vegetables, and starchy foods. We randomly select two subsets of the study population: two-third sample for discovery (n = 934) and one-third sample for replication (n = 467). Among the 327 metabolites analyzed, 14 are significantly associated with the meat and fast food dietary pattern, four are significantly associated with the fish and vegetables dietary pattern, and none are associated with the starchy foods dietary pattern in the discovery sample. In the replication sample, nine remain associated with the meat and fast food dietary pattern [indole-3-propanoic acid, C24:0 lysophosphatidylcholine (LPC), N-methyl proline, proline betaine, C34:2 phosphatidylethanolamine (PE) plasmalogen, C36:5 PE plasmalogen, C38:5 PE plasmalogen, cotinine, hydroxyproline] and three remain associated with the fish and vegetables dietary pattern [1,7-dimethyluric acid, C22:6 lysophosphatidylethanolamine, docosahexaenoic acid (DHA)]. CONCLUSION Twelve metabolites are discovered and replicated in association with dietary patterns detected in a Southern U.S. African-American population, which could be useful as biomarkers of Southern dietary patterns.
Collapse
Affiliation(s)
- Casey M. Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Yan Gao
- The Jackson Heart Study, University of Mississippi Medical Center, Jackson, Mississippi
| | - Sameera Talegawkar
- Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, George Washington University, Washington, DC
| | - Katherine L. Tucker
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, Massachusetts
| | - Lisandro D. Colantonio
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Alabama
| | - Paul Muntner
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Alabama
| | - Debby Ngo
- Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Zsu Zsu Chen
- Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Daniel Cruz
- Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Daniel Katz
- Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Usman A. Tahir
- Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | - Robert E. Gerszten
- Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - James G. Wilson
- Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| |
Collapse
|
49
|
Lightning TA, Gesteira TF, Mueller JW. Steroid disulfates - Sulfation double trouble. Mol Cell Endocrinol 2021; 524:111161. [PMID: 33453296 DOI: 10.1016/j.mce.2021.111161] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/24/2020] [Accepted: 01/05/2021] [Indexed: 02/08/2023]
Abstract
Sulfation pathways have recently come into the focus of biomedical research. For steroid hormones and related compounds, sulfation represents an additional layer of regulation as sulfated steroids are more water-soluble and tend to be biologically less active. For steroid diols, an additional sulfation is possible, carried out by the same sulfotransferases that catalyze the first sulfation step. The steroid disulfates that are formed are the focus of this review. We discuss both their biochemical production as well as their putative biological function. Steroid disulfates have also been linked to various clinical conditions in numerous untargeted metabolomics studies. New analytical techniques exploring the biosynthetic routes of steroid disulfates have led to novel insights, changing our understanding of sulfation in human biology. They promise a bright future for research into sulfation pathways, hopefully too for the diagnosis and treatment of several associated diseases.
Collapse
Affiliation(s)
- Thomas Alec Lightning
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Tarsis F Gesteira
- College of Optometry, University of Houston, Houston, TX, USA; Optimvia, LLC, Batavia, OH, USA
| | - Jonathan Wolf Mueller
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK.
| |
Collapse
|
50
|
Borodina I, Kenny LC, McCarthy CM, Paramasivan K, Pretorius E, Roberts TJ, van der Hoek SA, Kell DB. The biology of ergothioneine, an antioxidant nutraceutical. Nutr Res Rev 2020; 33:190-217. [PMID: 32051057 PMCID: PMC7653990 DOI: 10.1017/s0954422419000301] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 11/20/2019] [Accepted: 11/25/2019] [Indexed: 02/07/2023]
Abstract
Ergothioneine (ERG) is an unusual thio-histidine betaine amino acid that has potent antioxidant activities. It is synthesised by a variety of microbes, especially fungi (including in mushroom fruiting bodies) and actinobacteria, but is not synthesised by plants and animals who acquire it via the soil and their diet, respectively. Animals have evolved a highly selective transporter for it, known as solute carrier family 22, member 4 (SLC22A4) in humans, signifying its importance, and ERG may even have the status of a vitamin. ERG accumulates differentially in various tissues, according to their expression of SLC22A4, favouring those such as erythrocytes that may be subject to oxidative stress. Mushroom or ERG consumption seems to provide significant prevention against oxidative stress in a large variety of systems. ERG seems to have strong cytoprotective status, and its concentration is lowered in a number of chronic inflammatory diseases. It has been passed as safe by regulatory agencies, and may have value as a nutraceutical and antioxidant more generally.
Collapse
Affiliation(s)
- Irina Borodina
- The Novo Nordisk Foundation Center for Biosustainability, Building 220, Chemitorvet 200, Technical University of Denmark, 2800Kongens Lyngby, Denmark
| | - Louise C. Kenny
- Department of Women’s and Children’s Health, Institute of Translational Medicine, University of Liverpool, Crown Street, LiverpoolL8 7SS, UK
| | - Cathal M. McCarthy
- Irish Centre for Fetal and Neonatal Translational Research (INFANT), Cork University Maternity Hospital, Cork, Republic of Ireland
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Republic of Ireland
| | - Kalaivani Paramasivan
- The Novo Nordisk Foundation Center for Biosustainability, Building 220, Chemitorvet 200, Technical University of Denmark, 2800Kongens Lyngby, Denmark
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland, 7602, South Africa
| | - Timothy J. Roberts
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland, 7602, South Africa
- Department of Biochemistry, Institute of Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown Street, LiverpoolL69 7ZB, UK
| | - Steven A. van der Hoek
- The Novo Nordisk Foundation Center for Biosustainability, Building 220, Chemitorvet 200, Technical University of Denmark, 2800Kongens Lyngby, Denmark
| | - Douglas B. Kell
- The Novo Nordisk Foundation Center for Biosustainability, Building 220, Chemitorvet 200, Technical University of Denmark, 2800Kongens Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland, 7602, South Africa
- Department of Biochemistry, Institute of Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown Street, LiverpoolL69 7ZB, UK
| |
Collapse
|