A red wine intervention does not modify plasma trimethylamine N-oxide but is associated with broad shifts in the plasma metabolome and gut microbiota composition

Elisa A Haas, Mario J A Saad, Andrey Santos, Nicola Vitulo, Wilson J F Lemos, Aline M A Martins, Carolina R C Picossi, Desidério Favarato, Renato S Gaspar, Daniéla O Magro, Peter Libby, Francisco R M Laurindo, Protasio L Da Luz, WineFlora Study, Elisa A Haas, Mario J A Saad, Andrey Santos, Nicola Vitulo, Wilson J F Lemos, Aline M A Martins, Carolina R C Picossi, Desidério Favarato, Renato S Gaspar, Daniéla O Magro, Peter Libby, Francisco R M Laurindo, Protasio L Da Luz, WineFlora Study

Abstract

Background: Gut microbiota profiles are closely related to cardiovascular diseases through mechanisms that include the reported deleterious effects of metabolites, such as trimethylamine N-oxide (TMAO), which have been studied as diagnostic and therapeutic targets. Moderate red wine (RW) consumption is reportedly cardioprotective, possibly by affecting the gut microbiota.

Objectives: To investigate the effects of RW consumption on the gut microbiota, plasma TMAO, and the plasma metabolome in men with documented coronary artery disease (CAD) using a multiomics assessment in a crossover trial.

Methods: We conducted a randomized, crossover, controlled trial involving 42 men (average age, 60 y) with documented CAD comparing 3-wk RW consumption (250 mL/d, 5 d/wk) with an equal period of alcohol abstention, both preceded by a 2-wk washout period. The gut microbiota was analyzed via 16S rRNA high-throughput sequencing. Plasma TMAO was evaluated by LC-MS/MS. The plasma metabolome of 20 randomly selected participants was evaluated by ultra-high-performance LC-MS/MS. The effect of RW consumption was assessed by individual comparisons using paired tests during the abstention and RW periods.

Results: Plasma TMAO did not differ between RW intervention and alcohol abstention, and TMAO concentrations showed low intraindividual concordance over time, with an intraclass correlation coefficient of 0.049 during the control period. After RW consumption, there was significant remodeling of the gut microbiota, with a difference in β diversity and predominance of Parasutterella, Ruminococcaceae, several Bacteroides species, and Prevotella. Plasma metabolomic analysis revealed significant changes in metabolites after RW consumption, consistent with improved redox homeostasis.

Conclusions: Modulation of the gut microbiota may contribute to the putative cardiovascular benefits of moderate RW consumption. The low intraindividual concordance of TMAO presents challenges regarding its role as a cardiovascular risk biomarker at the individual level. This study was registered at clinical trials.gov as NCT03232099.

Keywords: coronary artery disease; gut microbiota; metabolomics; redox; trimethylamine N-oxide (TMAO); wine.

© The Author(s) 2022. Published by Oxford University Press on behalf of the American Society for Nutrition.

Figures

FIGURE 1
FIGURE 1
Study design. A total of 42 patients with established coronary artery disease were randomly selected for 1 of the 2 groups in the crossover study involving red wine (RW) consumption for 3 wk or alcohol abstention for 3 wk. All patients were evaluated over 5 in-hospital visits for anthropometrics and clinical and nutritional assessments. After an initial evaluation, there was a 2-wk washout period when patients were instructed not to consume alcoholic beverages, fermented foods (yogurt, kombucha, soy lecithin, kefir, sauerkraut, and other fermented veggies), synthetic prebiotics (insulin, fructooligosaccharides), fiber, dairy, food polyphenols (grapes, grape juice, cranberries, strawberries), and probiotics. Samples were collected after the first 2-wk washout period, and patients were randomly assigned for a 3-wk intervention with RW consumption (250 mL/d, 5 d/wk) or 3 wk of alcohol abstention. After these 3 wk, new blood and stool samples were collected, and another 2-wk washout period was implemented. Then, new samples were collected, and patients crossed over: the group that received RW was instructed to abstain from alcohol for 3 wk, and the group that abstained from alcohol in the first 3 wk received RW. After the second 3-wk period, stool and blood samples were collected in both groups. In 20 randomly selected patients (10 from each group), plasma metabolomics was analyzed after the 2 washout periods and after the intervention with RW and abstention.
FIGURE 2
FIGURE 2
Sparse partial least squares–discriminant analysis (sPLS-DA). (A) The plot of the 2-component sPLS-DA model showed stool sample clustering according to red wine consumption (RW) or not (Abs). The percentage of variance captured for each principal component (x-axis for the first component and y-axis for the second component) for each study period (RW compared with abstention). The sPLS-DA plot is based on the relative abundance of bacterial taxa in the gut microbiota from the Abs group (blue circle) or RW group (purple triangle) and their 95% confidence ellipses. (B) The contribution plot indicates each genus's contribution to the first component of sPLS-DA. Genus contribution ranked from the bottom (most important) to the top. The colors blue (Abs) and purple (RW) indicate the group in which the genus is most abundant. The horizontal graduation line represents the variance explained by the single genera.
FIGURE 3
FIGURE 3
Untargeted plasma metabolome alterations in 20 subjects. (A) Heatmap showing metabolites that were significantly different between the red wine (RW) and abstention (Abst) periods. According to the metabolic pathways and biochemical functions, the graph highlights the most clinically relevant metabolites that were significantly different between the 2 periods of the study (P < 0.05, without adjustments for multiple comparisons). (B) Box-and-whisker plot of the distribution of the discriminating metabolites that were significantly altered after RW consumption compared with the abstention period. (C) Pentose and glucoronate interconversions adapted from Kyoto Encyclopedia of Genes and Genomes pathway analysis (62). In red are the metabolites that were significantly increased after RW, and in blue are the putative pathways these metabolites are involved with. Arrows indicate the direction of the reaction and reversible and irreversible reactions, which are indicated by bidirectional and unidirectional arrows, respectively. Bold lines indicate activation or interaction. Dashed lines indicate an indirect link or unknown reaction.
FIGURE 4
FIGURE 4
Correlation matrix and multiomics data integration analysis. (A) In the arrow plot, the arrow origin indicates the centroid among all data sets for a given sample, and the tips of the arrows indicate the location of that sample in each block. These graphs highlight the agreement among all data sets at the sample level when modeled with DIABLO. The 2 omics (microbiota taxa and metabolomics) data sets performed well in separating the 2 interventions: red wine (RW) and abstention (Abst). (B) The line outside each circle indicates the group with which each feature is associated [taxa, on the right side of the circle or metabolites (MET), on the left side]. The purple line represents biomarkers associated with the RW period, whereas the blue lines represent those associated with the abstention period. The higher the line is, the higher the discrimination power of the feature. The line inside the circle represents the correlation between the taxa and the metabolites (an orange line indicates a positive correlation, and a black line indicates a negative correlation). The correlation cutoff was set to 0.5.

References

    1. Tang WHW, Bäckhed F, Landmesser U, Hazen SL. Intestinal microbiota in cardiovascular health and disease: JACC state-of-the-art review. J Am Coll Cardiol. 2019;73(16):2089–105.
    1. Chadaideh KS, Carmody RN. Host-microbial interactions in the metabolism of different dietary fats. Cell Metab. 2021;33(5):857–72.
    1. Alexander M, Turnbaugh PJ. Deconstructing mechanisms of diet-microbiome-immune interactions. Immunity. 2020;53(2):264–76.
    1. Senthong V, Li XS, Hudec T, Coughlin J, Wu Y, Levison Bet al. . Plasma trimethylamine N-oxide, a gut microbe–generated phosphatidylcholine metabolite, is associated with atherosclerotic burden. J Am Coll Cardiol. 2016;67(22):2620–8.
    1. Mitchell SM, Milan AM, Mitchell CJ, Gillies NA, D'Souza RF, Zeng Net al. . Protein intake at twice the RDA in older men increases circulatory concentrations of the microbiome metabolite trimethylamine-N-oxide (TMAO). Nutrients. 2019;11(9):2207.
    1. Baugh ME, Steele CN, Angiletta CJ, Mitchell CM, Neilson AP, Davy BMet al. . Inulin supplementation does not reduce plasma trimethylamine N-oxide concentrations in individuals at risk for type 2 diabetes. Nutrients. 2018;10(6):793.
    1. Maier TV, Lucio M, Lee LH, VerBerkmoes NC, Brislawn CJ, Bernhardt Jet al. . Impact of dietary resistant starch on the human gut microbiome, metaproteome, and metabolome. MBio. 2017;8(5):e01343–17.
    1. Wang Z, Roberts AB, Buffa JA, Levison BS, Zhu W, Org Eet al. . Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell. 2015;163(7):1585–95.
    1. Gupta N, Buffa JA, Roberts AB, Sangwan N, Skye SM, Li Let al. . Targeted inhibition of gut microbial trimethylamine N-oxide production reduces renal tubulointerstitial fibrosis and functional impairment in a murine model of chronic kidney disease. Arterioscler Thromb Vasc Biol. 2020;40(5):1239–55.
    1. Levantesi G, Marfisi R, Mozaffarian D, Franzosi MG, Maggioni A, Nicolosi GLet al. . Wine consumption and risk of cardiovascular events after myocardial infarction: results from the GISSI-Prevenzione trial. Int J Cardiol. 2013;; 163(3):282–7.
    1. Dolara P, Luceri C, De Filippo C, Pietro FA, Giovannelli L, Caderni Get al. . Red wine polyphenols influence carcinogenesis, intestinal microflora, oxidative damage and gene expression profiles of colonic mucosa in F344 rats. Mut Res. 2005;591(1–2):237–46.
    1. Gronbaek M, Deis A, Sorensen TIA, Becker U, Schnohr P, Jensen G. Mortality associated with moderate intakes of wine, beer, or spirits. BMJ. 1995;310:1165.
    1. Zorraquín I, Sánchez-Hernández E, Ayuda-Durán B, Silva M, González-Paramás AM, Santos-Buelga Cet al. . Current and future experimental approaches in the study of grape and wine polyphenols interacting gut microbiota. J Sci Food Agric. 2020;100(10):3789–802.
    1. Cueva C, Gil-Sánchez I, Ayuda-Durán B, González-Manzano S, González-Paramás AM, Santos-Buelga Cet al. . An integrated view of the effects of wine polyphenols and their relevant metabolites on gut and host health. Molecules. 2017;22(1):99.
    1. Queipo-Ortuño MI, Boto-Ordóñez M, Murri M, Gomez-Zumaquero JM, Clemente-Postigo M, Estruch Ret al. . Influence of red wine polyphenols and ethanol on the gut microbiota ecology and biochemical biomarkers. Am J Clin Nutr. 2012;95(6):1323–34.
    1. Muñoz-González I, Jiménez-Girón A, Martín-Álvarez PJ, Bartolomé B, Moreno-Arribas MV. Profiling of microbial-derived phenolic metabolites in human feces after moderate red wine intake. J Agric Food Chem. 2013;61(39):9470–9.
    1. Jacobs DM, Fuhrmann JC, Van Dorsten FA, Rein D, Peters S, Van Velzen EJJet al. . Impact of short-term intake of red wine and grape polyphenol extract on the human metabolome. J Agric Food Chem. 2012;60(12):3078–85.
    1. Zhang L, Carmody RN, Kalariya HM, Duran RM, Moskal K, Poulev Aet al. . Grape proanthocyanidin-induced intestinal bloom of Akkermansia muciniphila is dependent on its baseline abundance and precedes activation of host genes related to metabolic health. J Nutr Biochem. 2018;56:142.
    1. Le Roy CI, Wells PM, Si J, Raes J, Bell JT, Spector TD. Red wine consumption associated with increased gut microbiota α-diversity in 3 independent cohorts. Gastroenterology. 2020;158(1):270–272.e2.
    1. Chen M, Yi L, Zhang Y, Zhou X, Ran L, Yang Jet al. . Resveratrol attenuates trimethylamine-N-oxide (TMAO)-induced atherosclerosis by regulating TMAO synthesis and bile acid metabolism via remodeling of the gut microbiota. MBio. 2016;7(2):1–14.
    1. Frezza M, di Padova C, Pozzato G, Terpin M, Baraona E, Lieber CS. High blood alcohol levels in women. N Engl J Med. 1990;322(2):95–9.
    1. Fennema D, Phillips IR, Shephard EA. Trimethylamine and trimethylamine N-oxide, a flavin-containing monooxygenase 3 (FMO3)-mediated host-microbiome metabolic axis implicated in health and disease. Drug Metab Dispos. 2016;44(11):1839–50.
    1. Bohn MJ, Babor TF, Kranzler HR. The Alcohol Use Disorders Identification Test (AUDIT): validation of a screening instrument for use in medical settings. J Stud Alcohol. 1995;56(4):423–32.
    1. Tang WHW, Wang Z, Levison BS, Koeth RA, Britt EB, Fu Xet al. . Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med. 2013;368(17):1575–84.
    1. Kohn M, Senyak J. Sample size calculators [Internet]. [cited 2022 Jan 31]. Available from:
    1. Haseeb S, Alexander B, Baranchuk A. Wine and cardiovascular health: a comprehensive review. Circulation2017; ;136:1434–48.
    1. Jiménez-Girón A, Muñoz-González I, Martín-Alvarez PJ, Moreno-Arribas MV, Bartolomé B. Towards the fecal metabolome derived from moderate red wine intake. Metabolites. 2014;4(4):1101–18.
    1. Voskoboinik A, Prabhu S, Ling L-H, Kalman JM, Kistler PM. Alcohol and atrial fibrillation: a sobering review. J Am Coll Cardiol. 2016;68(23):2567–76.
    1. Barros Filho A A, Lima DM, Salay E, Siliprandi E, Veris LKM, Bagnato MHSet al. . Tabela brasileira de composição de alimentos—TACO 4a edição revisada e ampliada [Internet]. Universidade Estadual De Campinas—UNICAMP; 2011; [cited 2018 Sep 28]. Available from:
    1. Illumina. Illumina 16S metagenomic sequencing library preparation guide [Internet]. Illumina Technical Note 15044223. 2013; [cited 2022 May 27]. Available from:
    1. Magro DO, Santos A, Guadagnini D, Godoy FM, Silva SHM, Lemos WJFet al. . Remission in Crohn's disease is accompanied by alterations in the gut microbiota and mucins production. Sci Reports. 2019;9:1–10.
    1. Singh A, Shannon CP, Gautier B, Rohart F, Vacher M, Tebbutt SJet al. . DIABLO: an integrative approach for identifying key molecular drivers from multi-omics assays. Bioinformatics. 2019;35(17):3055–62.
    1. Wang Z, Levison BS, Hazen JE, Donahue L, Li XM, Hazen SL. Measurement of trimethylamine-N-oxide by stable isotope dilution liquid chromatography tandem mass spectrometry. Anal Biochem. 2014;455:35–40.
    1. Nyamundanda G, Gormley IC, Fan Y, Gallagher WM, Brennan L. MetSizeR: selecting the optimal sample size for metabolomic studies using an analysis based approach. BMC Bioinformatics2013;14:338.
    1. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BTet al. . Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19(5):576–85.
    1. Genoni A, Lo J, Lyons-Wall P, Boyce MC, Christophersen CT, Bird Aet al. . A paleolithic diet lowers resistant starch intake but does not affect serum trimethylamine-N-oxide concentrations in healthy women. Br J Nutr. 2019;121(3):322–9.
    1. Jonsson AL, Bäckhed F. Drug the bug! Cell. 2015;163:1565–6.
    1. Angiletta CJ, Griffin LE, Steele CN, Baer DJ, Novotny JA, Davy KPet al. . Impact of short-term flavanol supplementation on fasting plasma trimethylamine N-oxide concentrations in obese adults. Food Function. 2018;9(10):5350–61.
    1. Wu W-K, Chen C-C, Liu P-Y, Panyod S, Liao B-Y, Chen P-Cet al. . Identification of TMAO-producer phenotype and host–diet–gut dysbiosis by carnitine challenge test in human and germ-free mice. Gut. 2019;68(8):1439–49.
    1. Skye SM, Zhu W, Romano KA, Guo C-J, Wang Z, Jia Xet al. . Microbial transplantation with human gut commensals containing CutC is sufficient to transmit enhanced platelet reactivity and thrombosis potential. Circ Res. 2018;123(10):1164–76.
    1. Konop M, Radkowski M, Grochowska M, Perlejewski K, Samborowska E, Ufnal M. Enalapril decreases rat plasma concentration of TMAO, gut bacteria-derived cardiovascular marker. Biomarkers. 2018;23: 380–5.
    1. Latkovskis G, Makarova E, Mazule M, Bondare L, Hartmane D, Cirule Het al. . Loop diuretics decrease the renal elimination rate and increase the plasma levels of trimethylamine-N-oxide. Br J Clin Pharmacol. 2018;84(11):2634–44.
    1. Vandeputte D, De Commer L, Tito RY, Kathagen G, Sabino J, Vermeire Set al. . Temporal variability in quantitative human gut microbiome profiles and implications for clinical research. Nat Commun. 2021;12:6740.
    1. Frost F, Kacprowski T, Rühlemann M, Pietzner M, Bang C, Franke Aet al. . Long-term instability of the intestinal microbiome is associated with metabolic liver disease, low microbiota diversity, diabetes mellitus and impaired exocrine pancreatic function. Gut. 2021;70(3):522–30.
    1. Chadaideh KS, Eappen KE, Moore BE, Carmody RN. Common proanthocyanidin-rich foods modulate gastrointestinal blooms of Akkermansia muciniphila in a diet-dependent manner. 2021;1–32. Available from: 10.1101/2021.11.07.466338.
    1. Ju T, Kong JY, Stothard P, Willing BP. Defining the role of Parasutterella, a previously uncharacterized member of the core gut microbiota. ISME J2019;13:1520–34.
    1. Chang Y-L, Rossetti M, Vlamakis H, Casero D, Sunga G, Harre Net al. . A screen of Crohn's disease-associated microbial metabolites identifies ascorbate as a novel metabolic inhibitor of activated human T cells. Mucosal Immunol. 2019;12(2):457–67.
    1. Falony G, Joossens M, Vieira-Silva S, Wang J, Darzi Y, Faust Ket al. . Population-level analysis of gut microbiome variation. Science. 2016;352(6285):560–4.
    1. Nigdikar SV, Williams NR, Griffin BA, Howard AN. Consumption of red wine polyphenols reduces the susceptibility of low-density lipoproteins to oxidation in vivo. Am J Clin Nutr. 1998;68(2):258–65.
    1. Linster CL, Van Schaftingen E, Vitamin C. Biosynthesis, recycling and degradation in mammals. FEBS J. 2007;274(1):1–22.
    1. Ge T, Yang J, Zhou S, Wang Y, Li Y, Tong X. The role of the pentose phosphate pathway in diabetes and cancer. Front Endocrinol. 2020;11:1–11.
    1. Ashoori M, Saedisomeolia A. Riboflavin (vitamin b 2) and oxidative stress: a review. Br J Nutr. 2014;111(11):1985–91.
    1. Abedi F, Razavi BM, Hosseinzadeh H. A review on gentisic acid as a plant derived phenolic acid and metabolite of aspirin: comprehensive pharmacology, toxicology, and some pharmaceutical aspects. Phytother Res. 2020;34(4):729–41.
    1. Koh A, Bäckhed F. From association to causality: the role of the gut microbiota and its functional products on host metabolism. Mol Cell. 2020;78(4):584–96.
    1. Zhu W, Sun S, Yang F, Zhou K. UHPLC/MS identifying potent α-glucosidase inhibitors of grape pomace via enzyme immobilized method. J Food Sci. 2018;83(4):1131–9.
    1. Schooneman MG, Vaz FM, Houten SM, Soeters MR. Acylcarnitines. Diabetes. 2013;62(1):1–8.
    1. Friedman AN, Kim J, Kaiser S, Pedersen TL, Newman JW, Watkins BA. Association between plasma endocannabinoids and appetite in hemodialysis patients: a pilot study. Nutr Res. 2016;36(7):658–62.
    1. Muccioli GG, Naslain D, Bäckhed F, Reigstad CS, Lambert DM, Delzenne NMet al. . The endocannabinoid system links gut microbiota to adipogenesis. Mol Syst Biol. 2010;6(1):392.
    1. Rodionov DA, Arzamasov AA, Khoroshkin MS, Iablokov SN, Leyn SA, Peterson SNet al. . Micronutrient requirements and sharing capabilities of the human gut microbiome. Front Microbiol. 2019;10:1316.
    1. Jeon S, Carr R. Alcohol effects on hepatic lipid metabolism. J Lipid Res. 2020;61(4):470–9.
    1. You M, Arteel GE. Effect of ethanol on lipid metabolism. J Hepatol. 2019;70(2):237.
    1. Kanehisa M. KEGG: Kyoto Encyclopedia of Genes and Genomes. Nucleic Acids Res. 2000;28(1):27–30.

Source: PubMed

3
Subscribe