15
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Plasma Trimethylamine N-Oxide and Its Precursors: Population Epidemiology, Parent–Child Concordance, and Associations with Reported Dietary Intake in 11- to 12-Year-Old Children and Their Parents

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          Trimethylamine N-oxide (TMAO) is a microbiome- and diet-derived metabolite implicated in adverse cardiovascular outcomes. To date, studies of plasma TMAO concentrations have largely focused on individuals with metabolic disease. As such, data on TMAO concentrations in population settings and parent–child dyads are lacking.

          Objectives

          This study aimed to investigate parent–child concordance, age, and sex effects on plasma concentrations of TMAO and its precursors [ l-carnitine, choline, betaine, and dimethylglycine (DMG)]. Associations between concentrations of TMAO and its precursors and self-reported dietary intakes of animal protein (i.e., red meat, meat products, chicken, fish, milk products, and cheese) and fast-food meals were also investigated.

          Methods

          A total of 1166 children (mean ± SD age: 11 ± 0.5 y, 51% female) and 1324 parents (mean ± SD age: 44 ± 5.1 y, 87% female) had a biomedical assessment as part of Growing Up in Australia's Child Health Checkpoint. Plasma TMAO and precursor concentrations were quantified using ultra-high-pressure LC coupled with tandem MS.

          Results

          Familial dyads significantly contributed to plasma TMAO and precursor concentrations ( < 0.0001), explaining 37% of variance for TMAO concentrations. Least-square mean ± SE plasma TMAO was lower in children (0.79 ± 0.02 µM on the log-scale) than in adults (1.22 ± 0.02 µM). By contrast, children's betaine (40.30 ± 0.34 µM) and DMG concentrations (1.02 ± 0.01 µM on the log-scale) were higher than adults’ betaine (37.50 ± 0.32 µM) and DMG concentrations (0.80 ± 0.01 µM) ( < 0.0001). Mean values of all metabolites, except adult TMAO, were higher in males than in females ( < 0.001). Greater reported intake of red meat and fish was associated with higher TMAO concentrations in both children [estimates (95% CIs) for red meat: 0.06 (0.01, 0.10); fish: 0.11 (0.06, 0.17)] and adults [red meat: 0.13 (0.08, 0.17); meat products: 0.07 (0.03, 0.12); and fish: 0.09 (0.04, 0.14)].

          Conclusions

          Age, sex, and shared family factors, including diet, contribute to variation in plasma concentrations of TMAO and its precursors.

          Abstract

          Age, sex, and shared family factors characterize the concentrations of plasma TMAO and those of its precursors in Growing Up in Australia's CheckPoint Study of children and adults.

          Related collections

          Most cited references54

          • Record: found
          • Abstract: found
          • Article: not found

          Prognostic value of choline and betaine depends on intestinal microbiota-generated metabolite trimethylamine-N-oxide.

          Recent metabolomics and animal model studies show trimethylamine-N-oxide (TMAO), an intestinal microbiota-dependent metabolite formed from dietary trimethylamine-containing nutrients such as phosphatidylcholine (PC), choline, and carnitine, is linked to coronary artery disease pathogenesis. Our aim was to examine the prognostic value of systemic choline and betaine levels in stable cardiac patients. We examined the relationship between fasting plasma choline and betaine levels and risk of major adverse cardiac events (MACE = death, myocardial infraction, stroke) in relation to TMAO over 3 years of follow-up in 3903 sequential stable subjects undergoing elective diagnostic coronary angiography. In our study cohort, median (IQR) TMAO, choline, and betaine levels were 3.7 (2.4-6.2)μM, 9.8 (7.9-12.2)μM, and 41.1 (32.5-52.1)μM, respectively. Modest but statistically significant correlations were noted between TMAO and choline (r = 0.33, P < 0.001) and less between TMAO and betaine (r = 0.09, P < 0.001). Higher plasma choline and betaine levels were associated with a 1.9-fold and 1.4-fold increased risk of MACE, respectively (Quartiles 4 vs. 1; P < 0.01, each). Following adjustments for traditional cardiovascular risk factors and high-sensitivity C-reactive protein, elevated choline [1.34 (1.03-1.74), P < 0.05], and betaine levels [1.33 (1.03-1.73), P < 0.05] each predicted increased MACE risk. Neither choline nor betaine predicted MACE risk when TMAO was added to the adjustment model, and choline and betaine predicted future risk for MACE only when TMAO was elevated. Elevated plasma levels of choline and betaine are each associated with incident MACE risk independent of traditional risk factors. However, high choline and betaine levels are only associated with higher risk of future MACE with concomitant increase in TMAO.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Trimethylamine-N-oxide (TMAO) response to animal source foods varies among healthy young men and is influenced by their gut microbiota composition: A randomized controlled trial.

            Trimethylamine-N-oxide (TMAO), a metabolite linked to the gut microbiota, is associated with excess risk of heart disease. We hypothesized that (i) TMAO response to animal source foods would vary among healthy men and (ii) this response would be modified by their gut microbiome.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Trimethylamine and Trimethylamine N-Oxide, a Flavin-Containing Monooxygenase 3 (FMO3)-Mediated Host-Microbiome Metabolic Axis Implicated in Health and Disease

              Flavin-containing monooxygenase 3 (FMO3) is known primarily as an enzyme involved in the metabolism of therapeutic drugs. On a daily basis, however, we are exposed to one of the most abundant substrates of the enzyme trimethylamine (TMA), which is released from various dietary components by the action of gut bacteria. FMO3 converts the odorous TMA to nonodorous TMA N-oxide (TMAO), which is excreted in urine. Impaired FMO3 activity gives rise to the inherited disorder primary trimethylaminuria (TMAU). Affected individuals cannot produce TMAO and, consequently, excrete large amounts of TMA. A dysbiosis in gut bacteria can give rise to secondary TMAU. Recently, there has been much interest in FMO3 and its catalytic product, TMAO, because TMAO has been implicated in various conditions affecting health, including cardiovascular disease, reverse cholesterol transport, and glucose and lipid homeostasis. In this review, we consider the dietary components that can give rise to TMA, the gut bacteria involved in the production of TMA from dietary precursors, the metabolic reactions by which bacteria produce and use TMA, and the enzymes that catalyze the reactions. Also included is information on bacteria that produce TMA in the oral cavity and vagina, two key microbiome niches that can influence health. Finally, we discuss the importance of the TMA/TMAO microbiome-host axis in health and disease, considering factors that affect bacterial production and host metabolism of TMA, the involvement of TMAO and FMO3 in disease, and the implications of the host-microbiome axis for management of TMAU.
                Bookmark

                Author and article information

                Contributors
                Journal
                Curr Dev Nutr
                Curr Dev Nutr
                cdn
                Current Developments in Nutrition
                Oxford University Press
                2475-2991
                10 June 2020
                July 2020
                10 June 2020
                : 4
                : 7
                : nzaa103
                Affiliations
                Liggins Institute, The University of Auckland , Auckland, New Zealand
                The Murdoch Children's Research Institute , Parkville, Victoria, Australia
                Department of Paediatrics, University of Melbourne , Parkville, Victoria, Australia
                The Murdoch Children's Research Institute , Parkville, Victoria, Australia
                Department of Paediatrics, University of Melbourne , Parkville, Victoria, Australia
                Department of Statistics, Faculty of Science, The University of Auckland , Auckland, New Zealand
                Liggins Institute, The University of Auckland , Auckland, New Zealand
                The Murdoch Children's Research Institute , Parkville, Victoria, Australia
                Department of Paediatrics, University of Melbourne , Parkville, Victoria, Australia
                The Murdoch Children's Research Institute , Parkville, Victoria, Australia
                Department of Paediatrics, University of Melbourne , Parkville, Victoria, Australia
                The Murdoch Children's Research Institute , Parkville, Victoria, Australia
                Department of Paediatrics, University of Melbourne , Parkville, Victoria, Australia
                The Murdoch Children's Research Institute , Parkville, Victoria, Australia
                Department of Paediatrics, University of Melbourne , Parkville, Victoria, Australia
                Department of Paediatrics, Monash University , Clayton, Victoria, Australia
                Liggins Institute, The University of Auckland , Auckland, New Zealand
                Author notes
                Address correspondence to JMOS (e-mail: justin.osullivan@ 123456auckland.ac.nz )
                Author information
                http://orcid.org/0000-0001-8118-251X
                http://orcid.org/0000-0001-7501-9257
                http://orcid.org/0000-0002-9510-4181
                http://orcid.org/0000-0003-2927-450X
                Article
                nzaa103
                10.1093/cdn/nzaa103
                7335361
                32666035
                01363215-a045-49b4-ac7b-98f9d9ee58cc
                Copyright © The Author(s) on behalf of the American Society for Nutrition 2020.

                This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License ( http://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@ 123456oup.com .

                History
                : 04 March 2020
                : 18 May 2020
                : 02 June 2020
                Page count
                Pages: 11
                Funding
                Funded by: Ministry of Business, Innovation and Employment, DOI 10.13039/501100003524;
                Funded by: The New Zealand-Australia Life Course Collaboration on Genes, Environment, Nutrition and Obesity;
                Award ID: UOAX1611
                Funded by: National Health and Medical Research Council, DOI 10.13039/501100000925;
                Award ID: 1064629
                Award ID: GTN1175744
                Award ID: 1041352
                Award ID: 1109355
                Funded by: Children's Hospital Foundation;
                Award ID: 2014-241
                Funded by: Financial Markets Foundation for Children, DOI 10.13039/501100005381;
                Award ID: 2014-055
                Award ID: 2016-310
                Funded by: Victorian Deaf Education Institute;
                Funded by: Murdoch Children's Research Institute, DOI 10.13039/100014555;
                Funded by: Victorian Government's Operational Infrastructure Support Program;
                Funded by: New Zealand International Doctoral Research Scholarship 2017;
                Award ID: 1046518
                Award ID: 1160906
                Funded by: National Heart Foundation of Australia, DOI 10.13039/501100001030;
                Award ID: 100660
                Funded by: Honorary Future Leader Fellowship;
                Award ID: 100369
                Categories
                Original Research
                Nutritional Epidemiology and Public Health
                AcademicSubjects/MED00060

                trimethylamine n-oxide,l-carnitine,choline,betaine,dimethylglycine,epidemiology,children,adults,growing up in australia,longitudinal study of australian children

                Comments

                Comment on this article